Research article

Cheese starter cultures attenuate inflammation in the in vitro Caco-2 model

  • Received: 22 January 2025 Revised: 06 May 2025 Accepted: 15 May 2025 Published: 28 May 2025
  • Chronic inflammation is identified to be an underlying pathophysiology in different conditions including inflammatory bowel disease (IBD). Since the aberrant interaction of the mucosal immune system with the dysbiotic flora has been reported to contribute to IBD development, probiotics have been studied for potential prophylaxis and treatment. In this regard, fermented dairy foods are a rich source of probiotics and bioactive compounds. However, limited studies have determined the impact of fermented dairy products in the context of chronic inflammation. In particular, a potential role for dairy starter cultures is not well studied. Hence, in this study we evaluated the anti-inflammatory effect of two cheese starter cultures (Lactococcus lactis subsp. lactis M58 and Streptococcus thermophilus TA 61) in comparison with commercial probiotic strains (Bifidobacterium animalis subsp. lactis BB-12, Lactobacillus acidophilus LA-5) using the Cmax-induced Caco-2 inflammation model. Specifically, we characterized their ability to attenuate inflammatory response via modulation of IL-8 secretion, NF-κB activation, barrier integrity (TEER), and tight junction gene expression. Overall, pre-exposure to the starter cultures before Cmax treatment significantly reduced the activation and nuclear translocation of NF-κB, compared to cytokine control (P < 0.05). Further, the reduction in pNF-κB was found to be associated with a significant reduction in IL 8 secretion (P < 0.05). Moreover, the cultures protected the Caco-2 monolayer from inflammation-induced increase in permeability by upregulating the genes associated with ZO-1 and occludin production. Furthermore, the protective effect of the starter cultures was comparable to that of the commercial probiotics with known anti-inflammatory properties. Therefore, cheese starter cultures could be a potential strategy against chronic gut inflammation.

    Citation: Deepa Kuttappan, Sulthana Humayoon Muttathukonam, Mary Anne Amalaradjou. Cheese starter cultures attenuate inflammation in the in vitro Caco-2 model[J]. AIMS Microbiology, 2025, 11(2): 369-387. doi: 10.3934/microbiol.2025017

    Related Papers:

    [1] Dimitra Papadopoulou, Vasiliki Chrysikopoulou, Aikaterini Rampaouni, Alexandros Tsoupras . Antioxidant and anti-inflammatory properties of water kefir microbiota and its bioactive metabolites for health promoting bio-functional products and applications. AIMS Microbiology, 2024, 10(4): 756-811. doi: 10.3934/microbiol.2024034
    [2] Andrew S Day . The impact of exclusive enteral nutrition on the intestinal microbiota in inflammatory bowel disease. AIMS Microbiology, 2018, 4(4): 584-593. doi: 10.3934/microbiol.2018.4.584
    [3] John Samelis, Athanasia Kakouri . Hurdle factors minimizing growth of Listeria monocytogenes while counteracting in situ antilisterial effects of a novel nisin A-producing Lactococcus lactis subsp. cremoris costarter in thermized cheese milks. AIMS Microbiology, 2018, 4(1): 19-41. doi: 10.3934/microbiol.2018.1.19
    [4] Ellie Sayers, Alex MacGregor, Simon R. Carding . Drug-microbiota interactions and treatment response: Relevance to rheumatoid arthritis. AIMS Microbiology, 2018, 4(4): 642-654. doi: 10.3934/microbiol.2018.4.642
    [5] Lucy McMullen, Steven T Leach, Daniel A Lemberg, Andrew S Day . Current roles of specific bacteria in the pathogenesis of inflammatory bowel disease. AIMS Microbiology, 2015, 1(1): 82-91. doi: 10.3934/microbiol.2015.1.82
    [6] Jasmine S. Ritschard, Lea Amato, Yadhu Kumar, Britta Müller, Leo Meile, Markus Schuppler . The role of the surface smear microbiome in the development of defective smear on surface-ripened red-smear cheese. AIMS Microbiology, 2018, 4(4): 622-641. doi: 10.3934/microbiol.2018.4.622
    [7] Paola Cacchio, Marika Pellegrini, Beatrice Farda, Rihab Djebaili, Silvia Tabacchioni, Maddalena Del Gallo . Preliminary indication of the role of AHL-dependent quorum sensing systems in calcium carbonate precipitation in Gram-negative bacteria. AIMS Microbiology, 2023, 9(4): 692-711. doi: 10.3934/microbiol.2023035
    [8] Garmasheva I . Isolation and characterization of lactic acid bacteria from Ukrainiantraditional dairy products. AIMS Microbiology, 2016, 2(3): 372-387. doi: 10.3934/microbiol.2016.3.372
    [9] Tru Tran, Stephanie N. Dawrs, Grant J. Norton, Ravleen Virdi, Jennifer R. Honda . Brought to you courtesy of the red, white, and blue–pigments of nontuberculous mycobacteria. AIMS Microbiology, 2020, 6(4): 434-450. doi: 10.3934/microbiol.2020026
    [10] Farzad Rahmati . Characterization of Lactobacillus, Bacillus and Saccharomyces isolated from Iranian traditional dairy products for potential sources of starter cultures. AIMS Microbiology, 2017, 3(4): 815-825. doi: 10.3934/microbiol.2017.4.815
  • Chronic inflammation is identified to be an underlying pathophysiology in different conditions including inflammatory bowel disease (IBD). Since the aberrant interaction of the mucosal immune system with the dysbiotic flora has been reported to contribute to IBD development, probiotics have been studied for potential prophylaxis and treatment. In this regard, fermented dairy foods are a rich source of probiotics and bioactive compounds. However, limited studies have determined the impact of fermented dairy products in the context of chronic inflammation. In particular, a potential role for dairy starter cultures is not well studied. Hence, in this study we evaluated the anti-inflammatory effect of two cheese starter cultures (Lactococcus lactis subsp. lactis M58 and Streptococcus thermophilus TA 61) in comparison with commercial probiotic strains (Bifidobacterium animalis subsp. lactis BB-12, Lactobacillus acidophilus LA-5) using the Cmax-induced Caco-2 inflammation model. Specifically, we characterized their ability to attenuate inflammatory response via modulation of IL-8 secretion, NF-κB activation, barrier integrity (TEER), and tight junction gene expression. Overall, pre-exposure to the starter cultures before Cmax treatment significantly reduced the activation and nuclear translocation of NF-κB, compared to cytokine control (P < 0.05). Further, the reduction in pNF-κB was found to be associated with a significant reduction in IL 8 secretion (P < 0.05). Moreover, the cultures protected the Caco-2 monolayer from inflammation-induced increase in permeability by upregulating the genes associated with ZO-1 and occludin production. Furthermore, the protective effect of the starter cultures was comparable to that of the commercial probiotics with known anti-inflammatory properties. Therefore, cheese starter cultures could be a potential strategy against chronic gut inflammation.



    Inflammation is a physiological process directed at healing and maintenance of homeostasis [1],[2]. With multifactorial etiology, inflammation can be initiated by stimuli such as pathogens, chemical irritants, nutritional imbalance, and cell injury [2]. Although intended as a protective mechanism, when inflammation becomes chronic, it can cause severe and irreversible complications. In fact, chronic inflammation is identified to be an underlying pathophysiology in different conditions including inflammatory bowel disease (IBD) [3]. IBD is a progressive immune-mediated disease of the intestinal tract characterized by uncontrolled, relapsing inflammation leading to bowel damage [4]. Though the exact etiology of IBD is unclear, it is hypothesized that the disease originates from genetic susceptibility to dysregulated interaction between the immune system and the enteric commensal flora in the compromised gut. In fact, the two central features associated with IBD are a defective epithelial barrier and an exaggerated immune response [5][7].

    Although different cell types contribute to inflammation in IBD, the intestinal epithelial cells are known to play a critical role in pathogenesis. The intestinal epithelial cells (IECs) have a strategic position at the interface between the luminal environment and the internal milieu [8]. IECs institute bidirectional interactions with the underlying immune cells and contribute to the mucosal inflammatory response [9]. Besides this, the IECs form an impermeable polarized monolayer along the gut wall in the absence of specific transporters. The intercellular space is furthermore sealed by junctional protein complexes, of which the tight junctions are located at the most apical pole of the epithelial cells. Tight junctions are the main gatekeepers of paracellular space and can mediate the permeability of ions and small molecules up to 20 kDa. The adherens junctions and desmosomes, in contrast, form strong adhesive bonds and are primarily responsible for maintaining tissue cohesion and integrity [10],[11]. The intact tight junctions between the epithelial cells are responsible for maintaining selective epithelial permeability in the intestine. When this is challenged as in IBD, pathogens, intestinal contents, and toxins can gain access into the epithelial layers, leading to sustained inflammation [12].

    When inflamed, the mucosal immune cells are activated leading to IEC response and further barrier disruption [13][15]. The intestinal macrophages and dendritic cells sense pathogen-associated molecular patterns (PAMPs) and activate signal pathways, such as NF-kB, producing proinflammatory cytokines, chemokines, and anti-microbial peptides [16]. Increased production of inflammatory mediators including IFN-γ, IL-1β, TNF-α, IL-6, IL-8, IL-17A/F, IL-21, and IL-22 are observed in the intestine of IBD patients [17],[18]. Similarly, Caco2 cells exposed to a specific combination of inflammatory mediators, IL-1β, TNF-α, IFN-γ, and LPS, are found to mimic the gut inflammation [8].

    Since the aberrant interaction of the mucosal immune system with the dysbiotic flora has been reported to contribute to IBD development, probiotics have been studied for potential prophylaxis and treatment [19]. Probiotics belonging to the lactic acid bacteria (LAB) group and in particular to the genera Lactobacillus, Lactococcus, and Bifidobacterium are reported to have therapeutic properties in IBD [19][21]. Besides supplementation of live probiotics, foods supplemented with the strains or fermented using these cultures are also shown to exert a protective effect against IBD [22],[23]. Among the different fermented foods, dairy products constitute a significant portion of our daily diet. These products are a rich source of probiotics, and prebiotic and bioactive compounds [24]. However, limited studies have determined the impact of fermented dairy products in the context of chronic inflammation including IBD [23]. Among the common bacteria associated with fermented dairy products, starter cultures including Streptococcus thermophilus, Lactobacillus acidophilus, and Lactococcus lactis are reported to be ingested in high concentrations in fermented milk and cheese [25][28]. Beyond their role in food fermentation, select starter culture strains have been shown to exert a gut protective effect [29][31]. Along these lines, in this study, we determined the anti-inflammatory potential of commercial cheese starter cultures (Streptococcus thermophilus TA-61 and Lactococcus lactis subsp lactis M-58) in comparison to established probiotic strains (Bifidobacterium animalis subsp. lactis BB-12 and Lactobacillus acidophilus LA-5) using an in vitro model simulating active inflammation associated with IBD.

    Commercial probiotics, namely, Bifidobacterium animalis subsp. lactis BB-12 (BB) and Lactobacillus acidophilus LA-5 (LA), were kindly donated by Chr Hansen (Hoersholm, Denmark). The cheese starter cultures, Streptococcus thermophilus TA-61 (TA; Danisco A/S, Copenhagen, Denmark) and Lactococcus lactis subsp lactis M-58 (M; Danisco A/S, Copenhagen, Denmark) were obtained from Dairy Connection Inc. (Madison, WI, USA). All cultures except BB were grown in de Man, Rogosa, and Sharpe (MRS) broth (Difco, Sparks, MD, USA) under aerobic conditions at 37 °C for 24 h [32],[33]. Bifidobacterium animalis subsp. lactis (BB, Chr Hansen) was grown under anaerobic conditions at 37 °C in MRS containing 0.2 g/L lithium chloride and 0.3 g/L sodium propionate [34]. The bacterial count in these cultures was determined by plating 0.1 ml portions of appropriate dilutions on MRS agar (Difco, Sparks, MD, USA) with incubation at 37 °C for 24 h. The cultures were sedimented by centrifugation (3600 g, 15 min, 4 °C), and the pellets were washed twice with sterile PBS (pH 7.2). The pellets were resuspended in sterile Dulbecco's modified Eagle's medium supplemented with 20% fetal bovine serum to obtain the desired bacterial load (6 log CFU/mL).

    Caco-2 cells were obtained from ATCC (ATCC® HTB-37™) and were between passages 30–40 for all experiments (Manassas, VA, USA). Cells were cultured in Dulbecco's modified Eagle's medium supplemented with 20% fetal bovine serum. Cells were incubated at 37 °C and 7% CO2, and were subcultured at 80–90% confluence every 5–7 days. Once confluent, cells were detached with trypsin, counted, and seeded at a density of 1 × 105 cells per mL wither on 12-well plates or on polycarbonate membrane Transwell inserts with 0.4 µm pore size (Corning, Inc; Lowell, MA, USA) for further assays. Seeded cells were cultured for 21 days to reach differentiation, and growth media was refreshed every 2–3 days [35],[36].

    Caco-2 monolayer (Human intestinal epithelial colon carcinoma cell line, American Type Culture Collection) was cultured in Dulbecco's modified Eagle's medium supplemented with 20% fetal bovine serum in 12-well plates at 37 °C in the presence of 7% CO2 for 21 days. Following differentiation, Caco-2 cell monolayers (1 × 105 cells/well) were exposed to the different treatments at (~6 log CFU/well; Table 1) for 24 h [37]. In addition, uninoculated monolayers were set up as controls. The monolayers were then washed and treated with the cytokine cocktail (Cmax; IL-1β-25 ng/mL, TNFα-50 ng/mL, IFNγ-50 ng/mL, and LPS-10 µg/mL; Thermo Fisher Scientific, Waltham, MA, USA; [8]) for 24 h to stimulate an inflammatory response.

    Table 1.  Experimental groups. Monolayers were exposed to the different cultures for 24 h @ 6 log CFU/well followed by treatment with Cmax (IL-1β-25 ng/mL, TNFα-50 ng/mL, IFNγ-50 ng/mL, and LPS-10 µg/mL) for 24 h. Samples were then processed for further analysis.
    Group Treatment
    Control Untreated (no culture, no Cmax)
    Cmax Monolayer treated with cytokine cocktail
    BB Monolayer exposed to Bifidobacterium animalis subsp. lactis BB-12 (BB) alone
    LA Monolayer exposed to Lactobacillus acidophilus LA-5 (LA) alone
    M Monolayer exposed to Lactococcus lactis subsp. lactis M-58 (M) alone
    TA Monolayer exposed to Streptococcus thermophilus TA-61 (TA) alone
    BBCmax Monolayer exposed to BB and treated with Cmax
    LACmax Monolayer exposed to LA and treated with Cmax
    MCmax Monolayer exposed to M and treated with Cmax
    TACmax Monolayer exposed to TA and treated with Cmax

     | Show Table
    DownLoad: CSV

    Caco-2 cells (1 × 105 cells/mL) were grown in 12-well plates and treated as described above. Following stimulation with Cmax for 24 h, the cell culture supernatant was collected and stored at −80 °C until cytokines were analyzed. IL-8 estimation was performed using the IL-8/CXCL8 ELISA kit (R&D Systems Inc., Minneapolis, MN, USA; [38]).

    For this assay, Caco-2 cells (1 × 105 cells/well) were cultured in 6-well plates at 37 °C in the presence of 7% CO2 for 21 days. The cells were then exposed to the different cultures for 14 h followed by stimulation with Cmax for an additional 24 h as described earlier (treatment scheme as in Table 1). Following this, nuclear proteins were isolated using NE-PER Nuclear and Cytoplasmic Extraction kit according to the manufacturer's protocol (Thermo Fisher Scientific, Waltham, MA, USA). Protein concentrations were determined using the Bradford protein assay (Bio-Rad). Fifty micrograms of nuclear protein from each sample were then subject to NF-kB p65 [pS536] ELISA as per the manufacturer's protocol (Thermo Fisher Scientific, Waltham, MA, USA; [39][41]).

    Caco2 cells were seeded (1 × 105 cells/well) on Transwell inserts in 12-well culture plates and allowed to differentiate as previously described [35],[36]. The monolayers were exposed to the cultures and stimulated with Cmax as described earlier (Table 1). TEER of Caco-2 cells before and after treatment was measured using a Millicell ERS system (Millipore, Billerica, MA, USA). An insert without cells was used as a blank and its mean resistance was subtracted from all samples. For untreated, fully differentiated monolayers, TEER values were routinely 300–500 Ωcm2.

    To understand the effect of probiotics and starter cultures on tight junction genes, RNA was isolated from the Caco-2 cells following exposure to the different experimental groups (Table 1), using the Qiagen RNeasy according to the manufacturer's instructions (Qiagen). cDNA was synthesized using the Bio-Rad iScript cDNA synthesis kit (Bio-Rad, Hercules, CA, USA). RT-qPCR analysis of the genes associated with tight junction protein expression (ZO-1, Occludin) was performed [35] and normalized against GAPDH gene expression. The relative fold change in gene expression was calculated using the 2−ΔΔCt method [42].

    Each experiment was set up as a completely randomized design with three independent trials. All trials were performed in duplicate, and the results are presented as the mean ± standard error (SEM). The data were analyzed using the GraphPad Prism (v.10.1.1). One-way analysis of variance (ANOVA) was performed followed by Tukey-Kramer post hoc test for multiple comparisons amongst means. A p ≤ 0.05 was considered to be statistically significant.

    This study determined the potential anti-inflammatory properties of lactic cultures including starter cultures using an in vitro intestinal inflammation model. Since the IECs are reported to play a critical role in the pathophysiology of IBD, we employed the Caco-2 model. Further, the differentiated Caco-2 cell culture model is known to express tight junctions, cell surface receptors, transporters, and biotransformation enzymes making it the most commonly used in-vitro model of human enterocytes [43],[44]. To simulate the inflammatory environment in IBD, we used a cytokine cocktail (Cmax) consisting of TNFα, IFNγ, IL-1β, and LPS. These inflammatory mediators are reported to be associated with initiating, mediating, perpetuating, and controlling intestinal inflammation and tissue injury in IBD [45][48]. Specifically, these cytokines are known to play a role in barrier disruption, increased permeation of LPS and further precipitation of the inflammatory response [49],[50]. Moreover, the concerted effect of the cytokine cocktail serves to represent the endogenous (cytokines) and exogenous (LPS) inflammatory stimuli required for IBD development as seen in the acute phase of the condition [8].

    IL8 is a major chemokine active in IBD and involved in the chemotaxis of neutrophils and granulocytes to the inflammation site [51],[52]. Further, use of the cytokine cocktail (Cmax) has been shown to induce IL-8 secretion in a dose-dependent manner [8]. Further, literature demonstrates a key role for IL-8 in the pathophysiology of ulcerative colitis, a major type of IBD. Given this, we determined IL-8 levels in our inflammatory model following exposure to lactic cultures and administration of Cmax. As seen in Figure 1, in the Control samples (uninflamed, healthy cells, negative control), we observed a basal IL-8 concentration of 37.69 ± 12.20 pg/mL. Exposure of the Caco-2 cells to the different test strains (BBC, LCC, TAC, MC; Table 1) by themselves did not result in any significant change in IL-8 levels (15.4–37.3 pg/mL) when compared to the Control (37.69 pg/mL). On the other hand, as previously reported [8], administration of Cmax led to significant increase in IL-8 levels (p ≤ 0.0001). When compared to the basal level in the Control group, IL-8 levels were almost ten times higher (367.22 ± 29.82 pg/mL) in the Cmax group (positive control). On the other hand, pre-exposure to the test strains prior to Cmax administration significantly reduced IL-8 secretion in the Caco-2 monolayers (Figure 1). Specifically, IL-8 concentrations were 61.99 ± 15.62, 72.57 ± 22.69, 66.48 ± 25.13, and 78.28 ± 20.03 pg/mL in the BBCmax, LACmax, MCmax, and TACmax groups, respectively. Furthermore, the IL-8 levels in the treatment groups were not found to be significantly different from the control, indicating the potential anti-inflammatory effect of these strains. In addition, cheese starter cultures (TA, M) were equally as effective as the established probiotic strains (BB, LA) in protecting the monolayer from Cmax-mediated IL-8 production (Figure 1).

    Figure 1.  Pre-exposure to starter cultures (M, TA) and probiotics (BB, LA) decreases IL-8 production by differentiated Caco-2 cells exposed to inflammatory stimuli. Data is presented as means ± SD. Bars with different superscripts are significantly different from each other at p ≤ 0.05.

    IL-8 is one of the most extensively studied chemokines produced by different cell types including IECs, neutrophils, T lymphocytes, and macrophages [53][56]. More particularly, although barely detectable in unstimulated cells, IL-8 levels can increase by 10–100 times following inflammation such as activation by TNF-α, IL-6, IFN-γ, and LPS [57],[58]. This is in line with our results which demonstrate a ~100-fold increase in IL-8 levels in the in-vitro IEC model following administration of the cytokine cocktail (Figure 1). IL-8 is known to exert a pleiotropic role in inflammatory response via the recruitment and activation of neutrophils and granulocytes to the site of inflammation resulting in intense and chronic immune response and tissue damage [59]. Thus, it plays a key role in chronic inflammatory conditions including IBD and in particular ulcerative colitis [58],[60],[61]. Moreover, increased IL-8 secretion is well documented in the colonic mucosa of patients with active inflammation with a direct correlation to severity of colitis [62],[63].

    Given its critical role in inflammation, several studies have reported a protective effect following attenuation of proinflammatory cytokine production and downstream signaling [64][66]. Related to our results, application of probiotics has been proven to be beneficial in IBD when used alone or in combination with conventional drugs [67][69]. Specifically, regular consumption of kefir containing lactobacilli was seen to reduce inflammation and improve quality of life in patients with IBD [70]. Moreover, probiotic supplements containing Lactobacillus and Bifidobacterium strains were shown to be more effective in inducing remission in IBD [68]. For instance, administration of BB and LA was shown to suppress IL-8 secretion by TNF-α-stimulated HT-29 cells in vitro while improving colitis in a DNBS-induced mouse model [71]. Similarly, this anti-inflammatory effect was also observed following Salmonella infection in a gnotobiotic piglet model [72]. Our results align with these findings as seen from the significant reduction in IL-8 production in the inflamed IECs in vitro. More importantly, we observed that the starter cultures (TA and M) exerted a significant anti-inflammatory effect on the Caco-2 cells similar to the known probiotic strains (BB, LA; Figure 1). This is significant since previous studies using live cultures of related LAB strains derived from dairy products were shown to reduce Il-8 production in vitro and attenuate inflammation in vivo [73][75]. This highlights a potential role for dairy starter cultures in mediating the gut-health-promoting role of fermented dairy foods [23].

    NF-κB is activated by viral and bacterial infections, necrotic cell products, DNA damage, oxidative stress, and pro-inflammatory cytokines [76],[77]. When the stimulation occurs, the activated p65 subunit of NF-κB translocates to the nucleus and binds to the response elements transactivating the expression of pro-inflammatory cytokines including IL-8 [78]. In effect, NF-κB binding to the IL-8 promoter element is required for its transcriptional activation. Therefore, any stimuli that modulates NF-κB activity can also modify IL-8 induction. Specifically, inhibition of NF-κB activation can reduce transcriptional activation of IL-8 thereby attenuating the inflammatory response [53]. Thus, given our previous observation that exposure to starter cultures and probiotics significantly reduced IL-8 production in stimulated Caco-2 cells, as a next step we determined nuclear pNF-kB levels using ELISA.

    As seen with the IL-8 assay, exposure of the Caco-2 monolayers to the lactic cultures by themselves did not induce any significant activation of NF-kB when compared to the control (p > 0.05; Figure 2). Whereas treatment with Cmax led to a significant activation of NF-kB as evident from the increased pNF-kB levels in the nuclear fraction (4523 ± 628.6 pg/mL) when compared to the control (154.6 ± 31.03 pg/mL; p < 0.0001; Figure 2). However, pretreating the Caco-2 cells with probiotics and cheese cultures followed by Cmax stimulation led to significant attenuation of NF-kB activation as seen from the reduced pNF-kB levels in the nuclear fraction of the cell lysate in comparison to Cmax alone (p < 0.0001; Figure 2). For instance, pNF-kB levels in the inflamed IECs exposed to the cheese starter cultures (MCmax, TACmax) ranged from 572–994 pg/mL as opposed to 4523 pg/mL in the Cmax group (Figure 2). These results demonstrate that starter cultures potentially exert their anti-inflammatory effect by inhibiting NF-kB-mediated signaling and subsequent cytokine production including IL-8. Further, their anti-inflammatory effect was comparable to that of the established probiotics, namely BB and LA (Figure 2).

    Figure 2.  Pre-exposure to starter cultures (M, TA) and probiotics (BB, LA) attenuates NF-kB activation in differentiated Caco-2 cells exposed to inflammatory stimuli. Data is presented as means ± SD. Bars with different superscripts are significantly different from each other at p ≤ 0.05.

    NF-kB, as a regulator of gene transcription, is involved in the imbalance of activation of pro-inflammatory and anti-inflammatory signaling pathways in the gut [79]. In line with our findings, Heuvelin et al. [80] demonstrated that Bifidobacterium breve-conditioned medium inhibited IL-8 secretion by HT29-19A epithelial cells through the NF-kB pathway. Further, consumption of yogurt fermented with starter culture (YF-L702) containing Streptococcus thermophiles and Lactobacillus delbrueckii subsp. bulgaricus and co-inoculated with BB-12 was shown to reduce pro-inflammatory cytokines in cultured peripheral blood monocytes from healthy individuals following in-vitro LPS stimulation [81]. Similar reduction in inflammatory mediators in LPS-stimulated RAW264.7 macrophages by Bifidobacterium adolescentis was associated with reduced phosphorylation of I-κBα subunit of NF-kB [82]. Likewise, Lactobacillus casei and Bifidobacterium lactis NCC362 were seen to inhibit p65 nuclear translocation through a decrease in I-kBα ubiquitination and degradation, thereby attenuating NF-kB-mediated inflammatory signaling in HT-29 cells [83],[84]. Overall, the inhibition of NF-κB activity may explain the reduction in IL-8 production from stimulated IECs pre-exposed to probiotics and starter cultures in our study.

    A compromised intestinal epithelium is a feature observed in different intestinal inflammatory conditions including IBD and celiac disease [85]. Studies have revealed several defects in the specialized components of the mucosal barrier, from the mucus layer composition to the adhesion molecules that regulate paracellular permeability in IBD patients [86]. Given the critical role for barrier integrity in chronic inflammation, several studies have demonstrated the efficacy of different probiotics including Escherichia coli Nissle 1917, Bifidobacterium, Lactobacillus rhamnosus GG, and the multispecies VSL#3 in preventing leaky gut in IBD [86][88]. Along these lines, we evaluated the effect of our treatments on differentiated Caco-2 cell barrier integrity using TEER measurements. Except for LA, exposure to the starter cultures and/or the probiotics alone did not result in any significant reduction in TEER in the Caco-2 monolayers (p > 0.05). However, stimulation with the cytokine cocktail (Cmax) led to a 62% reduction (-275.33 ± 16.21 Ωcm2, Figure 3) in TEER from the initial value prior to treatment application. Van De Walle et al. [8] reported a similar reduction in TEER and loss in Caco-2 barrier integrity following treatment with Cmax.

    Figure 3.  Pre-exposure to starter cultures (M, TA) and probiotics (BB, LA) mitigates barrier permeability in differentiated Caco-2 monolayers exposed to inflammatory stimuli. Data is presented as means ± SD. Bars with different superscripts are significantly different from each other at p ≤ 0.05.

    On the other hand, pre-exposure to the starter cultures and probiotics protected the monolayer from Cmax-induced increase in permeability and reduction in TEER (p < 0.001, Figure 3). For example, in the TACmax and MCmax groups, we only observed a 25% reduction in TEER as opposed to the 62% reduction observed in the Cmax group. These data indicate that in addition to attenuating inflammation, cheese starter cultures can also protect the IECs from inflammation induced loss in barrier integrity. Further, their effect was comparable to that of the commercial probiotic strains (BB and LA) with proven anti-inflammatory and gut protective effects [71],[89]. A similar protective effect was reported following exposure of stimulated Caco-2 cells to lyophilized yogurt [35]. Also, it has been suggested that the protective effect of the dairy culture S. thermophilus NCIMB 41,856 is mediated by its ability to maintain the mucosal barrier thereby allowing healing of colitis [90]. Similar observations are also reported for other dairy-derived cultures including Lactobacillus helveticus, Lactobacillus delbrueckii, and Lactococcus lactis in in-vitro and in-vivo models [23].

    The intestinal epithelial barrier is comprised of a series of intercellular junctions made up of tight junctions, adherens junctions, and desmosomes [85]. Of these, the tight junctions primarily regulate paracellular permeability through a network of proteins including tight junction-associated marvel proteins such as occludin and intracellular scaffold proteins namely zonula occludens (ZO; [91][93]). Occludin is a transmembrane protein that is critical for localization of tight junctions [94]. Further, the carboxy terminal end of occludin contains the binding site for ZO-1. The ZO group of proteins (ZO-1, ZO-2, ZO-3) interacts with actin and help link the tight junction strands with the cytoskeleton [95]. This association of the cytoskeleton is critical for the maintenance of tight junction function and regulation of paracellular permeability. Toward this, abnormal tight junction structure and a down-regulation and redistribution of proteins including ZO-1 and occludin have been associated with loss in barrier permeability seen in conditions such as IBD [96][98]. Given the critical role of the tight junctions in barrier integrity and the results of our TEER assays, we performed gene expression assays to elucidate the effect of probiotics and starter cultures on the expression of tight junction protein-coding genes, namely OCLN and TJP-1 (ZO-1).

    Table 2.  Pre-exposure to starter cultures (M, TA) and probiotics (BB, LA) promotes epithelial barrier integrity by modulating tight junction protein gene expression. Data is presented as means ± SD. For each gene, different superscripts indicate a significant difference between treatments at p ≤ 0.05.
    Treatments Relative fold change in gene expression
    TJP1 OCLN
    Control 1.07 ± 0.03a 1.12 ± 0.05a
    BB 1.75 ± 0.14b 2.03 ± 0.24b
    LA 1.85 ± 0.22b 1.75 ± 0.35b
    M 2.11 ± 0.13b 1.80 ± 0.29b
    TA 2.00 ± 0.15b 1.63 ± 0.09a
    Cmax -1.82 ± 0.21c -2.16 ± 0.20c
    BBCmax 1.54 ± 0.21ab 1.66 ± 0.25ab
    LACmax 1.75 ± 0.17b 1.64 ± 0.39ab
    MCmax 1.80 ± 0.17b 1.36 ± 0.23a
    TACmax 1.66 ± 0.15b 1.34 ± 0.04a

     | Show Table
    DownLoad: CSV

    As seen from Table 2, treatment with Cmax significantly reduced TJP1 and OCLN expression by −1.82 ± 0.21 and −2.16 ± 0.19 fold, respectively, when compared to the Control (p ≤ 0.05). However, pre-exposure to the starter cultures and commercial probiotics helped protect the monolayer from Cmax induced downregulation in TJP-1 and OCLN expression (p ≤ 0.05). Specifically, we observed that pre-exposure to the starter cultures and probiotics significantly increased TJP-1 and OCLN expression in the healthy monolayer when compared to the Control (p ≤ 0.05; Table 2). Further, once these monolayers were stimulated using Cmax, the target strains continued to protect the monolayer from the inflammation-mediated attenuation of tight junction gene expression. Pre-exposure to the cheese starter cultures prior to Cmax treatment helped maintain TJP-1 and OCLN levels like that of the unstimulated cells exposed to TA or M. In addition, as seen with our previous assays, the improvement in tight junction gene expression was comparable to that of the commercial probiotic strains BB and LA. Moreover, these results could help explain the improvement in TEER measurements seen with the starter culture and probiotic-treated groups when compared to Cmax alone (Figure 3).

    Our findings are in agreement with increased mRNA expression of Caco-2 tight junction proteins and improved intestinal barrier function after exposure to Lactobacillus plantarum MB452 [99]. Similarly, treatment with lyophilized yogurt containing Lactobacillus bulgaricus and Streptococcus thermophilus was seen to increase claudin-1, ZO-1, and occludin mRNA levels in Cmax-stimulated Caco-2 cells [35]. Likewise, Bifidobacterium dentium N8 was shown to alleviate LPS-induced intestinal barrier injury in Caco-2 monolayers by regulating tight junction gene expression [100]. Besides these in-vitro studies, supplementation of dairy derived cultures including S. thermophiles MN-BM-A01 and Propionibacterium freudenreichii CIRM-BIA129 were seen to improve gut barrier integrity in the DSS-induced mice colitis model [90],[101]. Similarly, use of an E. coli Nissile strain engineered to carry zinc and indole-3-carbinol (ZI@EcN) on its surface was shown to significantly reduce inflammation in Caco-2 cells by restoring tight junction protein expression and restoring the epithelial barrier integrity [102]. In summary, none of our tested strains exerted any intrinsic pro-inflammatory effect on the Caco-2 monolayer. However, following the inflammatory stimulus, all tested strains exerted a significant protective effect on the IECs (p ≤ 0.05).

    Overall, our data indicate that cheese starter cultures (Streptococcus thermophilus TA-61 and Lactococcus lactis subsp. lactis M-58) exert a significant protective effect against cytokine-mediated inflammation on IECs in vitro. Specifically, by attenuating NF-κB-mediated inflammatory signaling and chemokine production (IL-8), and upregulating tight junction gene expression, the starter cultures (TA and M) protected the Caco-2 monolayer from inflammation and loss in barrier permeability. Moreover, we also observed that the anti-inflammatory effect of the starter cultures was comparable to that of commercial probiotics (Bifidobacterium animalis subsp. lactis BB-12 (BB) and Lactobacillus acidophilus LA-5) with demonstrated anti-inflammatory effects in vitro and in vivo. Therefore, the cheese starter cultures, Streptococcus thermophilus TA-61 and Lactococcus lactis subsp. lactis M-58, could be employed as an adjunct therapy for inflammation associated with IBD in humans. However, further validation of their anti-inflammatory and gut-protective effects in vivo is warranted.

    The authors declare they have not used Artificial Intelligence (AI) tools in the creation of this article.


    Acknowledgments



    This study was supported through the University of Connecticut's Research Excellence Program Award.

    Conflict of interest



    The authors declare no conflict of interest.

    Author contributions



    Conceptualization, M. A.; Writing—original draft preparation, all authors; writing—review and editing, M. A; visualization, M. A.; supervision, M. A.; project administration, M. A. All authors have read and agreed to the published version of the manuscript.

    [1] Netea MG, Balkwill F, Chonchol, et al. (2017) A guiding map for inflammation. Nat Immunol 18: 826-831. https://doi.org/10.1038/ni.3790
    [2] Agrawal M, Allin KH, Petralia F, et al. (2022) Multiomics to elucidate inflammatory bowel disease risk factors and pathways. Nat Rev Gastroenterol Hepatol 19: 399-409. https://doi.org/10.1038/s41575-022-00593-y
    [3] Furman D, Campisi J, Verdin E, et al. (2019) Chronic inflammation in the etiology of disease across the life span. Nat Med 25: 1822-1832. https://doi.org/10.1038/s41591-019-0675-0
    [4] Massimino L, Lamparelli LA, Houshyar Y, et al. (2021) The inflammatory bowel disease transcriptome and metatranscriptome meta-analysis (IBD TaMMA) framework. Nat Comput Sci 1: 511-515. https://doi.org/10.1038/s43588-021-00114-y
    [5] McCole DF (2014) IBD candidate genes and intestinal barrier regulation. Inflamm Bowel Dis 20: 1829-1849. https://doi.org/10.1097/MIB.0000000000000090
    [6] Ramos GP, Papadakis KA (2019) Mechanisms of disease: inflammatory bowel diseases. Mayo Clin Proceedings 94: 155-165. https://doi.org/10.1016/j.mayocp.2018.09.013
    [7] Vancamelbeke M, Vanuytsel T, Farré R, et al. (2017) Genetic and transcriptomic bases of intestinal epithelial barrier dysfunction in inflammatory bowel disease. Inflamm Bowel Dis 23: 1718-1729. https://doi.org/10.1097/MIB.0000000000001246
    [8] Van De Walle J, Hendrickx A, Romier B, et al. (2010) Inflammatory parameters in Caco-2 cells: Effect of stimuli nature, concentration, combination, and cell differentiation. In Vitro Toxicol 24: 1441-1449. https://doi.org/10.1016/j.tiv.2010.04.002
    [9] Danese S (2008) Nonimmune cells in inflammatory bowel disease: from victim to villain. Trends Immunol 29: 555-564. http://doi.org/10.1016/j.it.2008.07.009
    [10] Allaire JM, Morampudi V, Crowley SM, et al. (2018) Frontline defenders: goblet cell mediators dictate host-microbe interactions in the intestinal tract during health and disease. Am J Physiol Gastrointest Liver Physiol 314: G360-G377. https://doi.org/10.1152/ajpgi.00181.2017
    [11] Koch S, Nusrat A, Parkos CA (2013) The epithelial barrier. Molecular Genetics of Inflammatory Bowel Disease . New York, NY: Springer New York 265-280. https://doi.org/10.1007/978-1-4614-8256-7_13
    [12] Wallace KL, Zheng LB, Kanazawa Y, et al. (2014) Immunopathology of inflammatory bowel disease. WJG 20: 6. https://doi.org/10.3748/wjg.v20.i1.6
    [13] Barbara G, Barbaro MR, Fuschi D, et al. (2021) Inflammatory and microbiota-related regulation of the intestinal epithelial barrier. Front Nutr 8: 718356. https://doi.org/10.3389/fnut.2021.718356
    [14] Mahapatro M, Erkert L, Becker C (2021) Cytokine-mediated crosstalk between immune cells and epithelial cells in the gut. Cells 10: 111. https://doi.org/10.3390/cells10010111
    [15] Yao Y, Shang W, Bao L, et al. (2024) Epithelial-immune cell crosstalk for intestinal barrier homeostasis. Eur J Immunol 54: 2350631. https://doi.org/10.1002/eji.202350631
    [16] Akira S, Uematsu S, Takeuchi O (2006) Pathogen recognition and innate immunity. Cell 124: 783-801. https://doi.org/10.1016/j.cell.2006.02.015
    [17] Nakase H, Sato N, Mizuno N, et al. (2022) The influence of cytokines on the complex pathology of ulcerative colitis. Autoimmun Rev 21: 103017. https://doi.org/10.1016/j.autrev.2021.103017
    [18] Leppkes M, Neurath MF (2020) Cytokines in inflammatory bowel diseases–update 2020. Pharmacol Res 158: 104835. https://doi.org/10.1016/j.phrs.2020.104835
    [19] Ma Y, Yang D, Huang J, et al. (2024) Probiotics for inflammatory bowel disease: Is there sufficient evidence?. Open Life Sci 19: 20220821. https://doi.org/10.1515/biol-2022-0821
    [20] Chae JM, Heo W, Cho HT, et al. (2018) Effects of orally-administered Bifidobacterium animalis subsp. lactis strain BB12 on dextran sodium sulfate-induced colitis in mice. J Microbiol Biotechnol 28: 1800-1805. https://doi.org/10.4014/jmb.1805.05072
    [21] Wildt S, Nordgaard I, Hansen U, et al. (2011) A randomised double-blind placebo-controlled trial with Lactobacillus acidophilus La-5 and Bifidobacterium animalis subsp. lactis BB-12 for maintenance of remission in ulcerative colitis. JCC 5: 115-121. https://doi.org/10.1093/ecco-jcc/jjac104
    [22] Feathers A, Grigoryan Z, Falzon L, et al. (2023) S1004 The role of fermented food in inflammatory bowel disease treatment: A systematic review of randomized trials. ACG 118: S760-S761. https://doi.org/10.14309/01.ajg.0000953656.43074.2e
    [23] Illikoud N, Mantel M, Rolli-Derkinderen M, et al. (2022) Dairy starters and fermented dairy products modulate gut mucosal immunity. Immunol Lett 251: 91-102. https://doi.org/10.1016/j.imlet.2022.11.002
    [24] García-Burgos M, Moreno-Fernández J, Alférez MJ, et al. (2020) New perspectives in fermented dairy products and their health relevance. J Funct Foods 72: 104059. https://doi.org/10.1016/j.jff.2020.104059
    [25] Moreno Y, Collado MC, Ferrús MA, et al. (2006) Viability assessment of lactic acid bacteria in commercial dairy products stored at 4 C using LIVE/DEAD® BacLightTM staining and conventional plate counts. IJFST 41: 275-280. https://doi.org/10.1111/j.1365-2621.2005.01060.x
    [26] Moser A, Schafroth K, Meile L, et al. (2018) Population dynamics of Lactobacillus helveticus in Swiss Gruyère-type cheese manufactured with natural whey cultures. Front Microbiol 9: 637. https://doi.org/10.3389/fmicb.2018.00637
    [27] De Freitas I, Pinon N, Thierry A, et al. (2007) In depth dynamic characterization of French PDO Cantal cheese made from raw milk. Diary Sci Technol 87: 97-117. https://doi.org/10.1051/lait:2007007
    [28] Poveda JM, Sousa MJ, Cabezas L, et al. (2003) Preliminary observations on proteolysis in Manchego cheese made with a defined-strain starter culture and adjunct starter (Lactobacillus plantarum) or a commercial starter. Int Dairy J 13: 169-178. https://doi.org/10.1016/S0958-6946(02)00150-4
    [29] Ito M, Ohishi K, Yoshida Y, et al. (2008) Preventive effect of Streptococcus thermophilus YIT 2001 on dextran sulfate sodium-induced colitis in mice. BBBIEJ 72: 2543-2547. https://doi.org/10.1271/bbb.80240
    [30] Ogita T, Nakashima M, Morita H, et al. (2011) Streptococcus thermophilus ST28 ameliorates colitis in mice partially by suppression of inflammatory Th17 cells. Biomed Res Int 1: 378417. https://doi.org/10.1155/2011/378417
    [31] Berlec A, Perše M, Ravnikar M, et al. (2017) Dextran sulphate sodium colitis in C57BL/6J mice is alleviated by Lactococcus lactis and worsened by the neutralization of tumor necrosis factor α. Int Immunopharmacol 43: 219-226. https://doi.org/10.1016/j.intimp.2016.12.027
    [32] Ambalam P, Kondepudi KK, Balusupati P, et al. (2015) Prebiotic preferences of human lactobacilli strains in co-culture with bifidobacteria and antimicrobial activity against Clostridium difficile. J Appl Microbiol 119: 1672-1682. https://doi.org/10.1111/jam.12953
    [33] de Carvalho Lima KG, Kruger MF, Behrens J, et al. (2009) Evaluation of culture media for enumeration of Lactobacillus acidophilus, Lactobacillus casei and Bifidobacterium animalis in the presence of Lactobacillus delbrueckii subsp bulgaricus and Streptococcus thermophilus. LWT-Food Sci Technol 42: 491-495. https://doi.org/10.1016/j.lwt.2008.08.011
    [34] Fachin L, Moryia J, Gândara ALN, et al. (2008) Evaluation of culture media for counts of Bifidobacterium animalis subsp. lactis Bb 12in yoghurt after refrigerated storage. Braz J Microbiol 39: 357-361. https://doi.org/10.1590/S1517-83822008000200029
    [35] Putt KK, Pei R, White HM, et al. (2017) Yogurt inhibits intestinal barrier dysfunction in Caco-2 cells by increasing tight junctions. Food Funct 8: 406-414. https://doi.org/10.1039/C6FO01592A
    [36] Ciorba MA, Riehl TE, Rao MS, et al. (2012) Lactobacillus probiotic protects intestinal epithelium from radiation injury in a TLR-2/cyclo-oxygenase-2-dependent manner. Gut 61: 829-838. https://doi.org/10.1136/gutjnl-2011-300367
    [37] Dimitrov Z, Gotova I, Chorbadjiyska E (2014) In vitro characterization of the adhesive factors of selected probiotics to Caco-2 epithelium cell line. Biotechnol Biotechnol Equip 28: 1079-1083. https://doi.org/10.1080/13102818.2014.969948
    [38] Buagaew A, Poomipark N (2020) Protective effect of piperine from Piper chaba fruits on LPS-induced inflammation in human intestinal cell line. J Med Plants Res 14: 438-444. https://doi.org/10.5897/JMPR2020.6996
    [39] Simmons LJ, Surles-Zeigler MC, Li Y, et al. (2016) Regulation of inflammatory responses by neuregulin-1 in brain ischemia and microglial cells in vitro involves the NF-kappa B pathway. J Neuroinflammation 13: 1-15. https://doi.org/10.1186/s12974-016-0703-7
    [40] Van K, Burns JL, Monk JM (2024) Effect of short-chain fatty acids on inflammatory and metabolic function in an obese skeletal muscle cell culture model. Nutrients 16: 500. https://doi.org/10.3390/nu16040500
    [41] Matsumoto S, Hara T, Hori T, et al. (2005) Probiotic Lactobacillus-induced improvement in murine chronic inflammatory bowel disease is associated with the down-regulation of pro-inflammatory cytokines in lamina propria mononuclear cells. CEI 140: 417-426. https://doi.org/10.1111/j.1365-2249.2005.02790.x
    [42] 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. https://doi.org/10.1006/meth.2001.1262
    [43] Ferruzza S, Rossi C, Scarino ML, et al. (2012) A protocol for differentiation of human intestinal Caco-2 cells in asymmetric serum-containing medium. In Vitro Toxicol 26: 1252-1255. https://doi.org/10.1016/j.tiv.2012.01.008
    [44] Lea T (2015) Caco-2 cell line. The Impact of Food Bioactives on Health: in vitro and ex vivo models . Springer 103-111. https://doi.org/10.1007/978-3-319-16104-4
    [45] Guan Q, Zhang J (2017) Recent advances: the imbalance of cytokines in the pathogenesis of inflammatory bowel disease. Mediat Inflamm 1: 4810258. https://doi.org/10.1155/2017/4810258
    [46] Xu P, Elamin E, Elizalde M, et al. (2019) Modulation of intestinal epithelial permeability by plasma from patients with Crohn's disease in a three-dimensional cell culture model. Sci Rep 9: 2030. https://doi.org/10.1038/s41598-018-38322-8
    [47] Aggeletopoulou I, Kalafateli M, Tsounis EP, et al. (2024) Exploring the role of IL-1β in inflammatory bowel disease pathogenesis. Front Med 11: 1307394. https://doi.org/10.3389/fmed.2024.1307394
    [48] Kurumi H, Yokoyama Y, Hirano T, et al. (2024) Cytokine profile in predicting the effectiveness of advanced therapy for ulcerative colitis: A narrative review. Biomedicines 12: 952. https://doi.org/10.3390/biomedicines12050952
    [49] Kaminsky LW, Al-Sadi R, Ma TY (2021) IL-1β and the intestinal epithelial tight junction barrier. Front Immunol 12: 767456. https://doi.org/10.3389/fimmu.2021.767456
    [50] Meyer F, Wendling D, Demougeot C, et al. (2023) Cytokines and intestinal epithelial permeability: A systematic review. Autoimmun Rev 22: 103331. https://doi.org/10.1016/j.autrev.2023.103331
    [51] Subramanian S, Rhodes JM, Hart AC, et al. (2008) Characterization of epithelial IL-8 response to inflammatory bowel disease mucosal E. coli and its inhibition by mesalamine. Inflamm Bowel Dis 14: 162-175. https://doi.org/10.1002/ibd.20296
    [52] Hoffmann E, Dittrich-Breiholz O, Holtmann H, et al. (2002) Multiple control of interleukin-8 gene expression. J Leukoc Biol 72: 847-855. https://doi.org/10.1189/jlb.72.5.847
    [53] Zhu Y, Yang S, Zhao N, et al. (2021) CXCL8 chemokine in ulcerative colitis. Biomed Pharmacother 138: 111427. https://doi.org/10.1016/j.biopha.2021.111427
    [54] Yoshida N, Katada K, Handa O, et al. (2007) Interleukin-8 production via protease-activated receptor 2 in human esophageal epithelial cells. Int J Mol Med 19: 335-340. https://doi.org/10.3892/ijmm.19.2.335
    [55] Bie Y, Ge W, Yang Z, et al. (2019) The crucial role of CXCL8 and its receptors in colorectal liver metastasis. Dis Markers 1: 8023460. https://doi.org/10.1155/2019/8023460
    [56] Wanninger J, Neumeier M, Weigert J, et al. (2009) Adiponectin-stimulated CXCL8 release in primary human hepatocytes is regulated by ERK1/ERK2, p38 MAPK, NF-κB, and STAT3 signaling pathways. Am J Physiol Gastrointest Liver Physiol 297: G611-G618. https://doi.org/10.1152/ajpgi.90644.2008
    [57] Brat DJ, Bellail AC, Van Meir EG (2005) The role of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro-oncology 7: 122-133. https://doi.org/10.1215/S1152851704001061
    [58] Ha H, Debnath B, Neamati N (2017) Role of the CXCL8-CXCR1/2 axis in cancer and inflammatory diseases. Theranostics 7: 1543. https://doi.org/10.7150/thno.15625
    [59] Waugh DJ, Wilson C (2008) The interleukin-8 pathway in cancer. Clin Cancer Res 14: 6735-6741. https://doi.org/10.1158/1078-0432.CCR-07-4843
    [60] Russo RC, Garcia CC, Teixeira MM, et al. (2014) The CXCL8/IL-8 chemokine family and its receptors in inflammatory diseases. Expert Rev Clin Immunol 10: 593-619. https://doi.org/10.1586/1744666X.2014.894886
    [61] Ibusuki K, Sakiyama T, Kanmura S, et al. (2015) Human neutrophil peptides induce interleukin-8 in intestinal epithelial cells through the P2 receptor and ERK1/2 signaling pathways. Int J Mol Med 35: 1603-1609. https://doi.org/10.3892/ijmm.2015.2156
    [62] Bruno ME, Rogier EW, Arsenescu RI, et al. (2015) Correlation of biomarker expression in colonic mucosa with disease phenotype in Crohn's disease and ulcerative colitis. DDS 60: 2976-2984. https://doi.org/10.1007/s10620-015-3700-2
    [63] Zahn A, Giese T, Karner M, et al. (2009) Transcript levels of different cytokines and chemokines correlate with clinical and endoscopic activity in ulcerative colitis. BMC Gastroenterol 9: 1-7. https://doi.org/10.1186/1471-230X-9-13
    [64] Cai Z, Wang S, Li J (2021) Treatment of inflammatory bowel disease: a comprehensive review. Front Med 8: 765474. https://doi.org/10.3389/fmed.2021.765474
    [65] Shin JY, Wey M, Umutesi HG, et al. (2016) Thiopurine prodrugs mediate immunosuppressive effects by interfering with Rac1 protein function. JBC 291: 13699-13714. https://doi.org/10.1074/jbc.M115.694422
    [66] Van Dieren JM, Kuipers EJ, Samsom JN, et al. (2006) Revisiting the immunomodulators tacrolimus, methotrexate, and mycophenolate mofetil: their mechanisms of action and role in the treatment of IBD. Inflamm Bowel Dis 12: 311-327. https://doi.org/10.1097/01.MIB.0000209787.19952.53
    [67] Mardini HE, Grigorian AY (2014) Probiotic mix VSL# 3 is effective adjunctive therapy for mild to moderately active ulcerative colitis: a meta-analysis. Inflamm Bowel Dis 20: 1562-1567. https://doi.org/10.1097/MIB.0000000000000084
    [68] Zhang XF, Guan XX, Tang YJ, et al. (2021) Clinical effects and gut microbiota changes of using probiotics, prebiotics or synbiotics in inflammatory bowel disease: A systematic review and meta-analysis. Eur J Nutr 60: 2855-2875. https://doi.org/10.1007/s00394-021-02503-5
    [69] Ganji-Arjenaki M, Rafieian-Kopaei M (2018) Probiotics are a good choice in remission of inflammatory bowel diseases: A meta analysis and systematic review. J Cell Physiol 233: 2091-2103. https://doi.org/10.1002/jcp.25911
    [70] Yılmaz İ, Dolar ME, Özpınar H (2018) Effect of administering kefir on the changes in fecal microbiota and symptoms of inflammatory bowel disease: A randomized controlled trial. Turk J Gastroenterol 30: 242. https://doi.org/10.5152/tjg.2018.18227
    [71] Pápai G, Torres-Maravilla E, Chain F, et al. (2021) The administration matrix modifies the beneficial properties of a probiotic mix of Bifidobacterium animalis subsp. lactis BB-12 and Lactobacillus acidophilus LA-5. Probiotics Antimicrob Proteins 13: 484-494. https://doi.org/10.1007/s12602-020-09702-2
    [72] Splichal I, Donovan SM, Kindlova Z, et al. (2023) Release of HMGB1 and toll-like receptors 2, 4, and 9 signaling are modulated by Bifidobacterium animalis subsp. lactis BB-12 and Salmonella Typhimurium in a gnotobiotic piglet model of preterm infants. Int J Mol Sci 24: 2329. https://doi.org/10.3390/ijms24032329
    [73] Luerce TD, Gomes-Santos AC, Rocha CS, et al. (2014) Anti-inflammatory effects of Lactococcus lactis NCDO 2118 during the remission period of chemically induced colitis. Gut Pathog 6: 1-11. https://doi.org/10.1186/1757-4749-6-33
    [74] Kawahara M, Nemoto M, Nakata T, et al. (2015) Anti-inflammatory properties of fermented soy milk with Lactococcus lactis subsp. lactis S-SU2 in murine macrophage RAW264. 7 cells and DSS-induced IBD model mice. Int Immunopharmacol 26: 295-303. https://doi.org/10.1016/j.intimp.2015.04.004
    [75] Gotova I, Dimitrov Z, Najdenski H (2017) Selected Lactobacillus bulgaricus and Streptococcus thermophilus strains from bulgarian yogurt demonstrate significant anti-inflammatory potential. Acta Microbiol Bulg 33: 7.
    [76] Karin M, Greten FR (2005) NF-κB: linking inflammation and immunity to cancer development and progression. Nat Rev Immunol 5: 749-759. https://doi.org/10.1038/nri1703
    [77] Taniguchi K, Karin M (2018) NF-κB, inflammation, immunity, and cancer: coming of age. Nat Rev Immunol 18: 309-324. https://doi.org/10.1038/nri.2017.142
    [78] Hayden MS, Ghosh S (2008) Shared principles in NF-κB signaling. Cell 132: 344-362. https://doi.org/10.1016/j.cell.2008.01.020
    [79] McDaniel DK, Eden K, Ringel VM, et al. (2016) Emerging roles for noncanonical NF-κB signaling in the modulation of inflammatory bowel disease pathobiology. Inflamm Bowel Dis 22: 2265-2279. https://doi.org/10.1097/MIB.0000000000000858
    [80] Heuvelin E, Lebreton C, Grangett, C, et al. (2009) Mechanisms involved in alleviation of intestinal inflammation by Bifidobacterium breve soluble factors. PLoS One 4: e5184. https://doi.org/10.1371/journal.pone.0005184
    [81] Meng H, Ba Z, Lee Y, et al. (2017) Consumption of Bifidobacterium animalis subsp. lactis BB-12 in yogurt reduced expression of TLR-2 on peripheral blood-derived monocytes and pro-inflammatory cytokine secretion in young adults. Eur J Nutr 56: 649-661. https://doi.org/10.1007/s00394-015-1109-5
    [82] Okada Y, Tsuzuki Y, Hokari R, et al. (2009) Anti-inflammatory effects of the genus Bifidobacterium on macrophages by modification of phospho-IκB and SOCS gene expression. Int J Exp Pathol 90: 131-140. https://doi.org/10.1111/j.1365-2613.2008.00632.x
    [83] Riedel CU, Foata F, Philippe D, et al. (2006) Anti-inflammatory effects of bifidobacteria by inhibition of LPS-induced NF-κB activation. WJG 12: 3729. https://doi.org/10.3748/wjg.v12.i23.3729
    [84] Tien MT, Girardin SE, Regnault B, et al. (2006) Anti-inflammatory effect of Lactobacillus casei on Shigella-infected human intestinal epithelial cells. J Immunol 176: 1228-1237. https://doi.org/10.4049/jimmunol.176.2.1228
    [85] Barbara G (2006) Mucosal barrier defects in irritable bowel syndrome. Who left the door open?. ACG 101: 1295-1298. https://journals.lww.com/ajg/fulltext/2006/06000/Mucosal_Barrier_Defects_in_Irritable_Bowel.23.aspx
    [86] Michielan A, D'Incà R (2015) Intestinal permeability in inflammatory bowel disease: pathogenesis, clinical evaluation, and therapy of leaky gut. Mediat Inflamm 1: 628157. https://doi.org/10.1155/2015/628157
    [87] Wang W, Chen L, Zhou R, et al. (2014) Increased proportions of Bifidobacterium and the Lactobacillus group and loss of butyrate-producing bacteria in inflammatory bowel disease. J Clin Microbiol 52: 398-406. https://doi.org/10.1128/jcm.01500-13
    [88] Zakostelska Z, Kverka M, Klimesova K, et al. (2011) Lysate of probiotic Lactobacillus casei DN-114 001 ameliorates colitis by strengthening the gut barrier function and changing the gut microenvironment. PloS One 6: e27961. https://doi.org/10.1371/journal.pone.0027961
    [89] Wang H, Fan C, Zhao Z, et al. (2022) Anti-inflammatory effect of Bifidobacterium animalis subsp. lactis A6 on DSS-induced colitis in mice. J App Microbiol 133: 2063-2073. https://doi.org/10.1111/jam.15681
    [90] Bailey JR, Vince V, Williams NA, et al. (2017) Streptococcus thermophilus NCIMB 41856 ameliorates signs of colitis in an animal model of inflammatory bowel disease. Benef Microbes 8: 605-614. https://doi.org/10.3920/BM2016.0110
    [91] Anderson JM, Van Itallie CM (2009) Physiology and function of the tight junction. CSH Perspect Biol 1: a002584.
    [92] Furuse M, Hirase T, Itoh M, et al. (1993) Occludin: a novel integral membrane protein localizing at tight junctions. JCB 123: 1777-1788. https://doi.org/10.1083/jcb.123.6.1777
    [93] Ikenouchi J, Umeda K, Tsukita S, et al. (2007) Requirement of ZO-1 for the formation of belt-like adherens junctions during epithelial cell polarization. JCB 176: 779-786. https://doi.org/10.1083/jcb.200612080
    [94] Wong V (1997) Phosphorylation of occludin correlates with occludin localization and function at the tight junction. Am J Physiol Cell Physiol 273: C1859-C1867. https://doi.org/10.1152/ajpcell.1997.273.6.C1859
    [95] Fanning AS, Ma TY, Anderson JM (2002) Isolation and functional characterization of the actin-binding region in the tight junction protein ZO-1. FASEB J 16: 1-23. https://doi.org/10.1096/fj.02-0121fje
    [96] Martínez C, Vicario M, Ramos L, et al. (2012) The jejunum of diarrhea-predominant irritable bowel syndrome shows molecular alterations in the tight junction signaling pathway that are associated with mucosal pathobiology and clinical manifestations. Am J Gastroenterol 107: 736-746. https://doi.org/10.1038/ajg.2011.472
    [97] Wilcz-Villega E, McClean S, O'sullivan M (2014) Reduced E-cadherin expression is associated with abdominal pain and symptom duration in a study of alternating and diarrhea predominant IBS. Neurogastroenterol Motil 26: 316-325. https://doi.org/10.1111/nmo.12262
    [98] Drago S, El Asmar R, Di Pierro M, et al. (2006) Gliadin, zonulin and gut permeability: Effects on celiac and non-celiac intestinal mucosa and intestinal cell lines. Scand J Gastroenterol 41: 408-419. https://doi.org/10.1080/00365520500235334
    [99] Anderson RC, Cookson AL, McNabb WC, et al. (2010) Lactobacillus plantarum MB452 enhances the function of the intestinal barrier by increasing the expression levels of genes involved in tight junction formation. BMC Microbiol 10: 1-11. https://doi.org/10.1186/1471-2180-10-316
    [100] Zhao L, Xie Q, Evivie SE, et al. (2021) Bifidobacterium dentium N8 with potential probiotic characteristics prevents LPS-induced intestinal barrier injury by alleviating the inflammatory response and regulating the tight junction in Caco-2 cell monolayers. Food Funct 12: 7171-7184. https://doi.org/10.1039/D1FO01164B
    [101] Do Carmo FLR, Rabah H, Cordeiro BF, et al. (2019) Probiotic Propionibacterium freudenreichii requires SlpB protein to mitigate mucositis induced by chemotherapy. Oncotarget 10: 7198. https://doi.org/10.18632/oncotarget.27319
    [102] Chen Y, Bi S, Zhang X, et al. (2025) Engineered probiotics remodel the intestinal epithelial barrier and enhance bacteriotherapy for inflammatory bowel diseases. Acta Biomaterialia 198: 467-481. https://doi.org/10.1016/j.actbio.2025.04.016
  • Reader Comments
  • © 2025 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(92) PDF downloads(8) Cited by(0)

Figures and Tables

Figures(3)  /  Tables(2)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog