557
Views
1
CrossRef citations to date
0
Altmetric
Research Article

Influence of Porphyra-derived polysaccharides and oligosaccharides on attenuating food allergy and modulating enteric microflora in mice

, , , , , & show all
Article: 2248419 | Received 06 Jul 2023, Accepted 09 Aug 2023, Published online: 06 Sep 2023

ABSTRACT

Recently, there have been reports on the immunomodulatory properties of polysaccharides (PP), oligosaccharides (PO), and mixtures of PO/PP derived from Porphyra. To explore their potential in alleviating allergic responses and enteric dysbiosis, this study delved deeper into their effects using a murine model of food allergy. Daily administration of Porphyra samples mitigated allergic diarrhea, enteritis, and allergen-specific IgE production. Moreover, Porphyra samples exerted a significant reduction in the secretion of IL-2, IL-4, IFN-γ, and IL-17A, along with an elevation in the IL-10/IL-4 ratio in splenocytes upon allergen stimulation. Similarly, expression of IL-4 and IFN-γ in the duodenum was suppressed. Notably, allergic mice displayed a distinct fecal microflora profile compared to that of normal mice. Interestingly, PP and PO treatments exhibited different effects on improving enteric dysbiosis. These findings collectively suggest the potential of PP and PO as functional food candidates for the management of allergic enteritis and dysbiosis.

1. Introduction

The prevalence of food allergy, an immune disorder resulting from the consumption of food allergens, continues to rise, affecting approximately 3–10% of adults and 8% of children (Messina & Venter, Citation2020). One potential cause of food allergy is the failure of oral tolerance to food allergens (Vickery et al., Citation2011). Upon crossing the epithelial barrier, food allergens are recognised by dendritic cells, which subsequently present them to naïve T cells (Renz et al., Citation2018; Vickery et al., Citation2011). This interaction elicits the differentiation of naïve T helper (Th) cells into IL-4-secreting Th2 cells, promoting IgE secretion by B cells (Anvari et al., Citation2019). The allergen-specific IgE binds to the high-affinity immunoglobulin ϵ receptor on mast cells (Anvari et al., Citation2019; Kumar et al., Citation2012). When the gastrointestinal tract is exposed to the same allergen, IgE cross-linking induces mast cell degranulation and the secretion of inflammatory factors, leading to enteritis (Kumar et al., Citation2012; Ray, Citation2006; Vickery et al., Citation2011). Currently, there are no effective treatments for food allergy, highlighting the urgent need for the development of novel therapeutic and preventive strategies.

The composition of enteric microflora plays a crucial role in intestinal immunity and modulation of allergic responses. Various factors, including dietary choices, have been identified to influence the composition of enteric microflora (Hooper et al., Citation2012). Studies have shown that enteric dysbiosis can trigger and exacerbate intestinal inflammation and even lead to sepsis caused by opportunistic pathogens, primarily due to the disruption of the intestinal barrier (Gill et al., Citation2006; Huang et al., Citation2020). Therefore, maintaining a balanced enteric microflora is crucial for the development of immune tolerance. It has been proposed that preventing enteric dysbiosis could maintain the balance between Th1 and Th2 immune responses and preserve an adequate number of regulatory T (Treg) cells in the intestinal-associated lymphoid tissue. Additionally, treatment with probiotics has been shown to inhibit intestinal inflammation, stimulate Treg cell proliferation, and enhance the secretion of IL-10 (Atarashi et al., Citation2013; Rodriguez et al., Citation2012).

Dietary indigestible carbohydrates, such as oligosaccharides and polysaccharides, possess the unique ability to selectively enhance the growth or activity of beneficial enteric probiotics (Davani-Davari et al., Citation2019). In the intestinal tract, Bacteroides and Firmicutes are the predominant bacterial phyla. Bacteroides, in particular, has the capability to metabolise and utilise the polysaccharides present in the intestine (Cockburn & Koropatkin, Citation2016). Previous research suggests that natural polysaccharides can promote the growth of Bacteroidetes, reduce the abundance of Proteobacteria, and increase the population of microorganisms that produce short-chain fatty acids in the intestine. Consequently, polysaccharides contribute to maintaining a balanced enteric microflora and creating an unfavourable environment for opportunistic pathogens (Di et al., Citation2018).

Porphyra sp., a type of red seaweed commonly consumed in East Asia, serves as both an economic crop and a food source. It contains various bioactive components, especially polysaccharides, which constitute 20–40% of the dry weight of Porphyra (Bhatia et al., Citation2013; Cian et al., Citation2012; Mu & Wu, Citation2011; Wang et al., Citation2008). These polysaccharides share a structural similarity to agarose and are characterised by the presence of β-D-galactose, 6-O-methyl-β-D-galactose, α-L-galactose, and α-L-galactose-6-sulphate (Cian et al., Citation2015). Numerous studies have highlighted the diverse bioactivities of Porphyra polysaccharides. For instance, Shi et al. demonstrated that sulphated polysaccharides extracted from P. haitanensis can mitigate allergic reaction in mice (Shi et al., Citation2015). Oligosaccharides derived from the hydrolysis of Porphyra polysaccharides have also exhibited bioactivities. Osumi et al. used bacterial enzymes to hydrolyse P. yezoensis–derived polysaccharides, resulting in oligosaccharides that showed hypocholesterolemic potential (Osumi et al., Citation2002). Wu et al. employed Aeromonas salmonicida MAEF108 enzymes to hydrolyse Porphyra polysaccharides and obtain Porphyra oligosaccharides with antioxidant activity (S.-C. Wu et al., Citation2005). Our recent study has provided evidence supporting the effects of PP, PO, and their mixtures on regulating antigen-specific humoral responses and splenic cytokine production, highlighting their potential protective role against allergen-induced immune responses (Wei et al., Citation2022).

To assess the potential use of PP, PO, and their mixtures in managing allergy, the objective of this study was to investigate their anti-allergic effects in a murine model of food allergy. Furthermore, the profile of immune response and gut microflora was illustrated to unravel the mechanisms underlying the actions of PP and PO.

2. Materials and methods

2.1. Chemicals, reagents, detection kits and Porphyra-derived polysaccharides (PP) and oligosaccharides (PO)

Chemicals and reagents were procured from Sigma Chemical (St. Louis, MO, USA), unless specified otherwise. Cell culture reagents were obtained from GE Healthcare Life Sciences (Marlborough, MA, USA). ELISA kits for cytokine and immunoglobulin (Ig) measurements were sourced from eBioscience, Inc. (San Diego, CA, USA). The necessary reagents and antibodies for immunohistochemical (IHC) staining were acquired from Vector Laboratories, Inc. (Newark, CA, USA), and Thermo Fisher Scientific Inc. (Waltham, MA, USA). The source of dried Porphyra sp. and methods for PP and PO preparation were the same as described in the previous study (Wei et al., Citation2022). In addition, the characteristics of PP and PO were shown in the previous study (Wei et al., Citation2022).

2.2. Food allergy model and design of animal experiment

Female BALB/c mice at 5 weeks of age were procured from the National Laboratory Animal Center of Taiwan. The mice were housed in the laboratory animal facility of the National Taiwan Ocean University (NTOU) and allowed free access to water and food for 1 week prior to the conduction of experiment. As shown in (A), mice were divided into following groups (5 mice per group): the naïve group (NA), the vehicle group (VH), and the treatment groups. In the VH group, mice received a daily oral gavage of 0.1 mL phosphate-buffered saline (PBS). In the treatment groups, mice were administered a daily oral gavage of 150 mg/kg body weight of PP, PO, or PO/PP mixtures (PO:PP = 2:1, 1:1, and 1:2; referred to as O2P1, O1P1, and O1P2, respectively) dissolved in 0.1 mL PBS. The dosage regimen was according to the results in a previous study (Wei et al., Citation2022).

Figure 1. Protocol of the animal experiment, diarrhea scores and histopathological examination of duodenal tissues. (A) BALB/c mice were divided into the naïve group (NA), the vehicle group (VH), and the treatment groups (5 mice per group). In the VH group, mice received a daily oral gavage of 0.1 mL PBS. In the treatment groups, mice were administered a daily oral gavage of 150 mg/kg body weight of PP, PO, or PO/PP mixtures (PO:PP = 2:1, 1:1, and 1:2; referred to as O2P1, O1P1, and O1P2, respectively) dissolved in 0.1 mL PBS. Except for the mice in the NA group, all other mice were sensitised with ovalbumin (OVA) on day 4 and day 18. From day 32 to day 40, the sensitised mice were subjected to repeated challenges with OVA via oral gavage every other day. On day 41, the mice were euthanized, and the samples of feces, spleen and duodenal tissues were isolated for further experiments. (B) The severity of allergic diarrhea, occurring 30–60 min after the challenge, was assessed by scoring the fecal form on a scale of 0–3: 0 for normal feces, 1 for soft feces, 2 for sloppy feces, and 3 for diarrhea. (C) Representative images of H&E-stained duodenal sections are shown, highlighting villus edema (marked by red arrows) and crypt hyperplasia (marked by yellow arrows). (D) The crypt/villus ratio was quantified using ImageJ software. Data are presented as mean ± SEM (n = 5) and represent three independent experiments. Statistical analysis was performed to compare the different groups, with significance indicated by the symbols #(p < 0.05 compared to the NA group) and *(p < 0.05 compared to the VH group).

Figure 1. Protocol of the animal experiment, diarrhea scores and histopathological examination of duodenal tissues. (A) BALB/c mice were divided into the naïve group (NA), the vehicle group (VH), and the treatment groups (5 mice per group). In the VH group, mice received a daily oral gavage of 0.1 mL PBS. In the treatment groups, mice were administered a daily oral gavage of 150 mg/kg body weight of PP, PO, or PO/PP mixtures (PO:PP = 2:1, 1:1, and 1:2; referred to as O2P1, O1P1, and O1P2, respectively) dissolved in 0.1 mL PBS. Except for the mice in the NA group, all other mice were sensitised with ovalbumin (OVA) on day 4 and day 18. From day 32 to day 40, the sensitised mice were subjected to repeated challenges with OVA via oral gavage every other day. On day 41, the mice were euthanized, and the samples of feces, spleen and duodenal tissues were isolated for further experiments. (B) The severity of allergic diarrhea, occurring 30–60 min after the challenge, was assessed by scoring the fecal form on a scale of 0–3: 0 for normal feces, 1 for soft feces, 2 for sloppy feces, and 3 for diarrhea. (C) Representative images of H&E-stained duodenal sections are shown, highlighting villus edema (marked by red arrows) and crypt hyperplasia (marked by yellow arrows). (D) The crypt/villus ratio was quantified using ImageJ software. Data are presented as mean ± SEM (n = 5) and represent three independent experiments. Statistical analysis was performed to compare the different groups, with significance indicated by the symbols #(p < 0.05 compared to the NA group) and *(p < 0.05 compared to the VH group).

The protocol and methods for the induction of food allergy were the same as that reported in the previous study (Huang et al., Citation2020). Except for the mice in the NA group, all other mice were sensitised intraperitoneally with ovalbumin (OVA) and alum as adjuvant on day 4 and day 18. From day 32 to day 40, the sensitised mice were subjected to repeated challenges with OVA via oral gavage every other day. On the days of sensitisation and challenge, treatment of Porphyra samples was at least 4 h prior to the sensitisation or challenge. The severity of allergic diarrhea, occurring 30–60 min after the challenge, was assessed by scoring the fecal form on a scale of 0–3: 0 for normal feces, 1 for soft feces, 2 for sloppy feces, and 3 for diarrhea. On day 41, the mice were euthanized, and the duodenal tissues, which showed obvious histopathological changes in the employed model as described in the previous studies, were collected for histopathological examination (Huang et al., Citation2009, Citation2010, Citation2017). The protocol and methods of immunohistochemical (IHC) staining for IL-4 and IFN-γ detection were the same as that reported in the previous study (Huang et al., Citation2010). Hematoxylin and Eosin (H & E)-stained sections and IHC-stained sections were observed by an upright microscope (Olympus BX53) and analysed using ImageJ software to determine histopathological changes, the crypt/villus ratio, and the area ratios of IL-4 and IFN-γ positive signals to the total tissue area, following previously described methods (Huang et al., Citation2009, Citation2010, Citation2017). Spleen samples were individually isolated to prepare splenocyte suspensions, and fresh fecal samples were collected after the final challenge for next-generation sequencing (NGS) analysis of 16S rRNA gene, as described in previous studies (Huang et al., Citation2020).

2.3. Treatment of splenocytes and measurement of cytokine and Ig

Splenocytes prepared from the spleen isolated from each mouse were seeded into 24-well culture plates at a concentration of 5 × 106 cells/mL. These cells were then treated with OVA for a period of 24–72 h. After the designated treatment time, the supernatants from the culture plates were collected. The levels of IFN-γ, IL-2, IL-4, IL-10, and IL-17A were measured using ELISA. Furthermore, the levels of total IgG, IgE, OVA-specific IgG, and OVA-specific IgE in the serum samples were also measured using ELISA. The ELISA protocol was conducted following the instructions provided by the supplier.

2.4. Statistical analysis

Comparison between groups were conducted by use of ANOVA followed by a Student’s t-test (SigmaPlot V14, Systat Software Inc.). The differences were considered significant at p < 0.05.

3. Results and discussion

3.1. Influence of PP, PO and PO/PP mixtures on alleviating allergic diarrhea and enteritis in mice

In our previous study, it was found that the dosage of 150 mg/kg of PP, PO, and PO/PP mixtures had a modulatory effect on antigen-specific immune responses (Wei et al., Citation2022). Therefore, the same dosage of 150 mg/kg was employed to investigate and compare the anti-allergic effects of PP, PO, and PO/PP mixtures. During the experiment, no abnormal behaviour or activity were observed in any of the mice, revealing that the employed dosage did not cause any obvious adverse effects on the mice. The diarrhea scores of mice in the VH group increased with each challenge. However, scores of mice in PP, PO, and PO/PP mixtures groups were statistically lower than those in the VH group at the 4th and 5th challenges, indicating that Porphyra samples could reduce the severity of allergic diarrhea ((B)). Furthermore, the effects of PP, PO, and PO/PP mixtures on improving allergic enteritis were assessed through histopathological examination. In the VH group, significant tissue damage, villus edema, and crypt hyperplasia were observed ((C)). However, the treatment groups showed milder histopathological changes compared to the VH group ((C)). The ratio of crypt depth to villus height is a commonly used index for assessing gut health (Pluske et al., Citation1996). Treatment with Porphyra samples resulted in a reduced crypt/villus ratio compared to the VH control group ((D)), indicating the protective effects of PP, PO, and PO/PP mixtures against allergic enteritis.

3.2. Impact of PP, PO and PO/PP mixtures on serum antibodies production

The impact of Porphyra samples on IgG production was investigated to understand their effects on humoral immunity. The levels of total IgG were comparable between the VH group and treatment groups ((A)). This results reveals that PP, PO, and PO/PP mixtures had limited influence on the production of total IgG, indicating that their effects on humoral immunity for general pathogen protection were minimal (Vidarsson et al., Citation2014). Furthermore, the levels of total IgE, another type of antibody, were not significantly altered by the treatment of Porphyra samples ((B)). Since allergen-specific IgE and IgG play crucial roles in the initiation and inhibition of allergen-induced mast cell degranulation through the cross-linking of IgE receptors, the levels of OVA-specific IgE and IgG were specifically investigated (Finkelman, Citation2007). Compared to that in the NA group, the increased level of OVA-specific IgE in the VH group indicated a successful induction of type I hypersensitivity ((D)). Compared to the VH group, the levels of OVA-specific IgE were down-regulated, while the levels of OVA-specific IgG were slightly up-regulated in mice treated with Porphyra samples ((C,D)). These results indicate that the modulatory effects of Porphyra samples on humoral immunity were allergen-specific, specifically affecting the production of allergen-specific IgE in response to OVA. In line with the results of the current study, Persiyanova et al. have demonstrated that polysaccharides derived from the brown algae Fucus evanescens could raise the level of OVA-specific IgG (Persiyanova et al., Citation2020). As the binding of allergen-specific IgG to allergen blocks the binding of IgE and allergen (Finkelman, Citation2007), the up-regulation of OVA-specific IgG is suggested as one of the potential manifestations of effective treatment.

Figure 2. The production of antibodies in serum of mice. Serum sample was collected from each mouse, and the concentrations of immunoglobulins, including (A) total IgG, (B) total IgE, (C) OVA-specific IgG, and (D) OVA-specific IgE, were measured. Data are presented as mean ± SEM (n = 5) and represent three independent experiments. Statistical analysis was conducted to compare the different groups, and the symbols #(p < 0.05 compared to the NA group) and *(p < 0.05 compared to the VH group) were used to indicate significant differences.

Figure 2. The production of antibodies in serum of mice. Serum sample was collected from each mouse, and the concentrations of immunoglobulins, including (A) total IgG, (B) total IgE, (C) OVA-specific IgG, and (D) OVA-specific IgE, were measured. Data are presented as mean ± SEM (n = 5) and represent three independent experiments. Statistical analysis was conducted to compare the different groups, and the symbols #(p < 0.05 compared to the NA group) and *(p < 0.05 compared to the VH group) were used to indicate significant differences.

3.3. Effects of PP, PO and PO/PP mixtures on regulating splenic and duodenal cytokine production

To investigate the effects of Porphyra samples on regulating allergen-induced Th subset activation, the production of IL-2, IFN-γ, IL-17A, IL-4 and IL-10, representing the activation of Th1, Th17, and Th2 and Treg subsets, by OVA-stimulated splenocytes was measured. OVA sensitisation and challenge led to an increase in the secretion of these cytokines ((A–E)), indicating the activation of Th1, Th17, and Th2 subsets. Treatment of Porphyra samples mitigated the OVA-induced activation of Th1, Th17, and Th2 subsets to varying degrees, as evidenced by the decreased levels of these cytokines ((A–E)). IL-10 is predominantly generated by Th2 cells or Treg cells in splenocytes. To understand the impact of Porphyra samples on the activation of Treg cells, the production of TGF-β, another regulatory cytokine, was measured. However, the level of TGF-β was too low to detect by ELISA. Instead, the ratio of IL-10/IL-4 was calculated to evaluate the source of IL-10. Treatment with Porphyra samples significantly increased the IL-10/IL-4 ratio ((F)), suggesting a potential upregulation of Treg immunity as a mechanism for the anti-allergic effects of Porphyra samples. However, further studies will be required to address this issue.

Figure 3. Cytokine production from OVA-stimulated splenocytes. Splenocyte suspensions prepared from mice in each experimental group were cultured in the presence of OVA. The levels of (A) IL-2, (B) IFN-γ, (C) IL-17A, (D) IL-4 and (E) IL-10 in the supernatants were measured, and the ratio of (F) IL-10/IL-4 was calculated. Data are expressed as mean ± SEM (n = 5) and represent three independent experiments. Statistical analysis was performed to compare the different groups, and the symbols #(p < 0.05 compared to the NA group) and *(p < 0.05 compared to the VH group) were used to denote significant differences.

Figure 3. Cytokine production from OVA-stimulated splenocytes. Splenocyte suspensions prepared from mice in each experimental group were cultured in the presence of OVA. The levels of (A) IL-2, (B) IFN-γ, (C) IL-17A, (D) IL-4 and (E) IL-10 in the supernatants were measured, and the ratio of (F) IL-10/IL-4 was calculated. Data are expressed as mean ± SEM (n = 5) and represent three independent experiments. Statistical analysis was performed to compare the different groups, and the symbols #(p < 0.05 compared to the NA group) and *(p < 0.05 compared to the VH group) were used to denote significant differences.

Since systemic (splenic) and mucosal (duodenal) responses may differ, the duodenal expression of IL-4 and IFN-γ were measured to elucidate the intestinal Th1/Th2 immune balance. Consistent with the results of splenic cytokine production, OVA sensitisation and challenge elevated the duodenal expression of IL-4 and IFN-γ. However, treatment with Porphyra samples suppressed duodenal expression of IL-4 and IFN-γ (), suggesting the influence of Porphyra samples in alleviating allergen-induced Th1 and Th2 immune responses.

Figure 4. The expression of IL-4 and IFN-γ in the duodenal tissues of mice. Representative images of the IHC-stained sections are presented in (A, C), where the brown signals indicate the presence of IL-4 or IFN-γ. (B, D) The area occupied by IL-4 and IFN-γ positive signals was measured, and the ratio of positive area to the total tissue area was calculated using ImageJ software. Data are expressed as mean ± SEM (n = 5) and represent three independent experiments. Statistical analysis was conducted to compare the different groups, and the symbols #(p < 0.05 compared to the NA group) and *(p < 0.05 compared to the VH group) were used to indicate significant differences.

Figure 4. The expression of IL-4 and IFN-γ in the duodenal tissues of mice. Representative images of the IHC-stained sections are presented in (A, C), where the brown signals indicate the presence of IL-4 or IFN-γ. (B, D) The area occupied by IL-4 and IFN-γ positive signals was measured, and the ratio of positive area to the total tissue area was calculated using ImageJ software. Data are expressed as mean ± SEM (n = 5) and represent three independent experiments. Statistical analysis was conducted to compare the different groups, and the symbols #(p < 0.05 compared to the NA group) and *(p < 0.05 compared to the VH group) were used to indicate significant differences.

Although both IL-10 and TGF-β are classified as regulatory cytokines secreted from Foxp3+ Treg cells, it is primarily IL-10, not TGF-β, that inhibits mast cell activation and intestinal inflammatory responses in murine models of food allergy (Burrello et al., Citation2018; Huang et al., Citation2010). This phenomenon was also observed in allergic mice treated with Porphyra samples in this study. Furthermore, the balance between effector Th cell subsets plays an essential role in controlling inflammation and allergy (Chaplin, Citation2010). Cytokine production is a common indicator for Th subset activation, with each subset producing specific cytokines. Indeed, IL-2 is involved in clonal expansion of lymphocytes. IFN-γ and IL-17A contribute to different inflammatory responses in certain immune disorders. IL-4 induces IgE isotype switching and subsequent type I hypersensitivity. IL-10 serves to restrict the immune response of the host and preserve immune balance (Chaplin, Citation2010). Based on the results of the current study, the anti-allergic effects of Porphyra samples were closely related to the attenuation of allergen-specific Th subset activation through the upregulation of IL-10 production.

3.4. Impact of PP, PO and PO/PP mixtures on enteric microflora

In this study, the profile of enteric microflora was investigated using 16S rRNA gene sequencing. The main bacterial classes identified in the enteric microflora were Bacteroidia, Bacilli, and Clostridia. Compared to that in mice of the NA group. The relative abundance of Bacilli and Clostridia was decreased, while the relative abundance of Bacteroidia was increased in the VH group ((A)). However, treatment with PP and PO resulted in different impact on the enteric microflora. PP treatment decreased the relative abundance of Bacteroidia, and the predominant bacterial class was Bacilli. On the other hand, PO treatment decreased the relative abundance of Bacteroidia and increased the relative abundance of Clostridia, Lachnospiraceae, Actinobacteria, and Coriobacteriia ((A)). To further analyse the specific bacteria affected by PP and PO treatment, LEfSe analysis was performed. It was found that the relative abundance of Bifidobacterium was 4.8-fold higher in the PP group compared to that in the VH group ((B)). The relative abundance of Clostridia, Lachnospiraceae, Actinobacteria, and Coriobacteriia was at least 3.6-fold higher in the PO group compared to that in the VH group ((B)). These findings indicate that PP and PO exerted distinct effects on modulating enteric dysbiosis, which could be attributed to their different molecular weights and sulphate contents (Wei et al., Citation2022).

Figure 5. Full-length 16S rRNA gene analysis of fecal microbiota. (A) The microbial diversity and relative abundance of fecal bacterial classes in each group are represented using pie charts. These charts provide a snapshot of the composition and distribution of different bacterial classes in the fecal samples from each experimental group. (B) Linear discriminant analysis (LDA) effect size (LEfSe) graphics were generated to analyse the differences between the groups of mice. The horizontal bars in the graphics represent the effect size, while the length of the bar corresponds to the log10 transformed LDA score, indicated by vertical dotted lines. The colour scheme distinguishes between mice in the VH group (red) and mice in the PO or PP groups (green). To identify taxa of bacteria that exhibited statistically significant changes in their relative abundance (p < 0.05), the taxonomic names of these bacteria are labelled alongside the horizontal lines in the LEfSe graphics. A threshold of 2.0 on the logarithmic LDA score was applied to determine discriminative features.

Figure 5. Full-length 16S rRNA gene analysis of fecal microbiota. (A) The microbial diversity and relative abundance of fecal bacterial classes in each group are represented using pie charts. These charts provide a snapshot of the composition and distribution of different bacterial classes in the fecal samples from each experimental group. (B) Linear discriminant analysis (LDA) effect size (LEfSe) graphics were generated to analyse the differences between the groups of mice. The horizontal bars in the graphics represent the effect size, while the length of the bar corresponds to the log10 transformed LDA score, indicated by vertical dotted lines. The colour scheme distinguishes between mice in the VH group (red) and mice in the PO or PP groups (green). To identify taxa of bacteria that exhibited statistically significant changes in their relative abundance (p < 0.05), the taxonomic names of these bacteria are labelled alongside the horizontal lines in the LEfSe graphics. A threshold of 2.0 on the logarithmic LDA score was applied to determine discriminative features.

Previous studies have shown that Bifidobacterium has beneficial effects in ameliorating food allergy by modulating the populations and activities of Th subsets and Treg cells (Gu et al., Citation2023; Liu et al., Citation2018). Clostridia has been reported to limit allergen exposure, suppress sensitisation, and then prevent against the development of food allergy (Stefka et al., Citation2014). The role of Lachnospiraceae and Actinobacteria in the development or prevention of food allergy is still debated. While some studies have observed a higher prevalence of Lachnospiraceae in children with egg and milk allergy, others have demonstrated a potential protective association with milk allergy (Berni Canani et al., Citation2016; Fazlollahi et al., Citation2018; Feehley et al., Citation2019). Actinobacteria abundances have been found to be both higher and lower in allergic populations compared to healthy individuals in different studies (Dethlefsen et al., Citation2007; Ling et al., Citation2014). Limited information is available regarding the relationship between Coriobacteriia and food allergy, but recent studies suggest a potential negative effect of Coriobacteriia on allergic rhinitis and asthma (Jin et al., Citation2023; Zheng et al., Citation2022). Polysaccharides and oligosaccharides are known to be metabolised by enteric microbes, and their metabolites are important for maintaining intestinal barrier integrity and immune tolerance (Cheng et al., Citation2022; Iribarren et al., Citation2021; Wu et al., Citation2023). Since the fecal microflora profile differed between PP- and PO-treated mice, further metabolome analysis would be necessary to understand how PP and PO influence the enteric microflora and how the affected microbes modulate the immune system and ameliorate allergic responses.

4. Conclusion

This study provides valuable insights into the anti-allergic effects of PP and PO. The distinct effects observed between PP and PO on attenuating inflammation, modulating the production of allergen-specific antibodies and cytokines and enteric microflora could be attributed to their differences in molecular weight and chemical composition. These findings open up possibilities for the development of functional foods containing Porphyra-derived oligosaccharides and polysaccharides as therapeutic options for managing allergic enteritis and dysbiosis.

Credit authorship contribution statement

Y.J. Wei: Conceptualisation, Data curation, Investigation, Methodology, Writing. R.E. Fang and J.S. Liu: Formal analysis, Validation. Y.C. Chen: Funding acquisition, Writing – review & editing. H.T. Victor Lin: Writing – review & editing. C.L. Pan: Project administration, Supervision, Validation. C.H. Huang: Conceptualisation, Funding acquisition, Supervision, Writing – review & editing.

Ethics statement and animal experiments

All animal experiments were conducted in accordance with the guidelines of the National Research Council’s Guide for the Care and Use of Laboratory Animals and approved by the NTOU Institutional Animal Care and Use Committee (NTOU IACUC-108040).

Acknowledgements

The authors would like to thank the Animal Technology Research Center of Agricultural Technology Research Institute (Miaoli, Taiwan), the Pathology Core Laboratory of National Health Research Institutes (Miaoli, Taiwan) and the National Laboratory Animal Center of National Applied Research Laboratories (Taipei, Taiwan) for the assistance of histopathological examination, and Taiwan Genomic Industry Alliance Inc. (Taipei, Taiwan) for the assistance of NGS analysis.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

Data will be made available on request.

Additional information

Funding

This research is under grant support of the National Science and Technology Council, Taiwan (grant numbers: MOST 109-2320-B-019-007-MY3 and MOST 109-2221-E-019-032-MY3) and the University System of Taipei Joint Research Program (grant number: USTP-NTOU-TMU-112-05).

References

  • Anvari, S., Miller, J., Yeh, C.-Y., & Davis, C. M. (2019). IgE-mediated food allergy. Clinical Reviews in Allergy & Immunology, 57(2), 244–260. https://doi.org/10.1007/s12016-018-8710-3
  • Atarashi, K., Tanoue, T., Oshima, K., Suda, W., Nagano, Y., Nishikawa, H., Fukuda, S., Saito, T., Narushima, S., Hase, K., Kim, S., Fritz, J. V., Wilmes, P., Ueha, S., Matsushima, K., Ohno, H., Olle, B., Sakaguchi, S., Taniguchi, T., … Hase, K. (2013). Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature, 500(7461), 232–236. https://doi.org/10.1038/nature12331
  • Berni Canani, R., Sangwan, N., Stefka, A. T., Nocerino, R., Paparo, L., Aitoro, R., Calignano, A., Khan, A. A., Gilbert, J. A., & Nagler, C. R. (2016). Lactobacillus rhamnosus GG-supplemented formula expands butyrate-producing bacterial strains in food allergic infants. The ISME Journal, 10(3), 742–750. https://doi.org/10.1038/ismej.2015.151
  • Bhatia, S., Rathee, P., Sharma, K., Chaugule, B., Kar, N., & Bera, T. (2013). Immuno-modulation effect of sulphated polysaccharide (porphyran) from Porphyra vietnamensis. International Journal of Biological Macromolecules, 57, 50–56. https://doi.org/10.1016/j.ijbiomac.2013.03.012
  • Burrello, C., Garavaglia, F., Cribiù, F. M., Ercoli, G., Lopez, G., Troisi, J., Colucci, A., Guglietta, S., Carloni, S., Guglielmetti, S., Taverniti, V., Nizzoli, G., Bosari, S., Caprioli, F., Rescigno, M., & Facciotti, F. (2018). Therapeutic faecal microbiota transplantation controls intestinal inflammation through IL10 secretion by immune cells. Nature Communications, 9(1), 5184. https://doi.org/10.1038/s41467-018-07359-8
  • Chaplin, D. D. (2010). Overview of the immune response. Journal of Allergy and Clinical Immunology, 125(2), S3–S23. https://doi.org/10.1016/j.jaci.2009.12.980
  • Cheng, J., Hu, J., Geng, F., & Nie, S. (2022). Bacteroides utilization for dietary polysaccharides and their beneficial effects on gut health. Food Science and Human Wellness, 11(5), 1101–1110. https://doi.org/10.1016/j.fshw.2022.04.002
  • Cian, R. E., Drago, S. R., Sanchez de Medina, F., & Martínez-Augustin, O. (2015). Proteins and carbohydrates from red seaweeds: Evidence for beneficial effects on gut function and microbiota. Marine Drugs, 13(8), 5358–5383. https://doi.org/10.3390/md13085358
  • Cian, R. E., Martínez-Augustin, O., & Drago, S. R. (2012). Bioactive properties of peptides obtained by enzymatic hydrolysis from protein byproducts of Porphyra columbina. Food Research International, 49(1), 364–372. https://doi.org/10.1016/j.foodres.2012.07.003
  • Cockburn, D. W., & Koropatkin, N. M. (2016). Polysaccharide degradation by the intestinal microbiota and its influence on human health and disease. Journal of Molecular Biology, 428(16), 3230–3252. https://doi.org/10.1016/j.jmb.2016.06.021
  • Davani-Davari, D., Negahdaripour, M., Karimzadeh, I., Seifan, M., Mohkam, M., Masoumi, S. J., Berenjian, A., & Ghasemi, Y. (2019). Prebiotics: Definition, types, sources, mechanisms, and clinical applications. Foods (Basel, Switzerland), 8(3), 92. https://doi.org/10.3390/foods8030092
  • Dethlefsen, L., McFall-Ngai, M., & Relman, D. A. (2007). An ecological and evolutionary perspective on human–microbe mutualism and disease. Nature, 449(7164), 811–818. https://doi.org/10.1038/nature06245
  • Di, T., Chen, G., Sun, Y., Ou, S., Zeng, X., & Ye, H. (2018). In vitro digestion by saliva, simulated gastric and small intestinal juices and fermentation by human fecal microbiota of sulfated polysaccharides from Gracilaria rubra. Journal of Functional Foods, 40, 18–27. https://doi.org/10.1016/j.jff.2017.10.040
  • Fazlollahi, M., Chun, Y., Grishin, A., Wood, R. A., Burks, A. W., Dawson, P., Jones, S. M., Leung, D. Y. M., Sampson, H. A., Sicherer, S. H., & Bunyavanich, S. (2018). Early-life gut microbiome and egg allergy. Allergy, 73(7), 1515–1524. https://doi.org/10.1111/all.13389
  • Feehley, T., Plunkett, C. H., Bao, R., Choi Hong, S. M., Culleen, E., Belda-Ferre, P., Campbell, E., Aitoro, R., Nocerino, R., Paparo, L., Andrade, J., Antonopoulos, D. A., Berni Canani, R., & Nagler, C. R. (2019). Healthy infants harbor intestinal bacteria that protect against food allergy. Nature Medicine, 25(3), 448–453. https://doi.org/10.1038/s41591-018-0324-z
  • Finkelman, F. D. (2007). Anaphylaxis: Lessons from mouse models. Journal of Allergy and Clinical Immunology, 120(3), 506–515. https://doi.org/10.1016/j.jaci.2007.07.033
  • Gill, S. R., Pop, M., DeBoy, R. T., Eckburg, P. B., Turnbaugh, P. J., Samuel, B. S., Gordon, J. I., Relman, D. A., Fraser-Liggett, C. M., & Nelson, K. E. (2006). Metagenomic analysis of the human distal gut microbiome. Science, 312(5778), 1355–1359. https://doi.org/10.1126/science.1124234
  • Gu, S., Yang, D., Liu, C., & Xue, W. (2023). The role of probiotics in prevention and treatment of food allergy. Food Science and Human Wellness, 12(3), 681–690. https://doi.org/10.1016/j.fshw.2022.09.001
  • Hooper, L. V., Littman, D. R., & Macpherson, A. J. (2012). Interactions between the microbiota and the immune system. Science, 336(6086), 1268–1273. https://doi.org/10.1126/science.1223490
  • Huang, C.-H., Ku, C.-Y., & Jan, T.-R. (2009). Diosgenin attenuates allergen-induced intestinal inflammation and IgE production in a murine model of food allergy. Planta Medica, 75(12), 1300–1305. https://doi.org/10.1055/s-0029-1185578
  • Huang, C.-H., Lin, Y.-C., & Jan, T.-R. (2017). Lactobacillus reuteri induces intestinal immune tolerance against food allergy in mice. Journal of Functional Foods, 31, 44–51. https://doi.org/10.1016/j.jff.2017.01.034
  • Huang, C.-H., Liu, D.-Z., & Jan, T.-R. (2010). Diosgenin, a plant-derived sapogenin, enhances regulatory T-cell immunity in the intestine of mice with food allergy. Journal of Natural Products, 73(6), 1033–1037. https://doi.org/10.1021/np900690z
  • Huang, C.-H., Lu, S.-Y., & Tsai, W.-C. (2020). Relevant fecal microbes isolated from mice with food allergy elicited intestinal cytokine/chemokine network and T-cell immune responses. Bioscience of Microbiota, Food and Health, 39(4), 234–242. https://doi.org/10.12938/bmfh.2020-014
  • Iribarren, C., Magnusson, M. K., Vigsnæs, L. K., Aziz, I., Amundsen, I. D., Šuligoj, T., Juge, N., Patel, P., Sapnara, M., Johnsen, L., Sørensen, N., Sundin, J., Törnblom, H., Simrén, M., & Öhman, L. (2021). The effects of human milk oligosaccharides on gut microbiota, metabolite profiles and host mucosal response in patients with irritable bowel syndrome. Nutrients, 13(11), 3836. https://doi.org/10.3390/nu13113836
  • Jin, Q., Ren, F., Dai, D., Sun, N., Qian, Y., & Song, P. (2023). The causality between intestinal flora and allergic diseases: Insights from a bi-directional two-sample Mendelian randomization analysis. Frontiers in Immunology, 14, 1121273.
  • Kumar, S., Verma, A. K., Das, M., & Dwivedi, P. D. (2012). Molecular mechanisms of IgE mediated food allergy. International Immunopharmacology, 13(4), 432–439. https://doi.org/10.1016/j.intimp.2012.05.018
  • Ling, Z., Li, Z., Liu, X., Cheng, Y., Luo, Y., Tong, X., Yuan, L., Wang, Y., Sun, J., Li, L., & Xiang, C. (2014). Altered fecal microbiota composition associated with food allergy in infants. Applied and Environmental Microbiology, 80(8), 2546–2554. https://doi.org/10.1128/AEM.00003-14
  • Liu, Q., Jing, W., & Wang, W. (2018). Bifidobacterium lactis ameliorates the risk of food allergy in Chinese children by affecting relative percentage of Treg and Th17 cells. Canadian Journal of Infectious Diseases and Medical Microbiology, 2018, 4561038.
  • Messina, M., & Venter, C. (2020). Recent surveys on food allergy prevalence. Nutrition Today, 55(1), 22–29. https://doi.org/10.1097/NT.0000000000000389
  • Mu, K., & Wu, Y. (2011). Separation and purification of Porphyra haitanensis polysaccharide and determination of its constitution. Journal of Chinese Institute of Food Science and Technology, 11(1), 159.
  • Osumi, Y., Kawai, M., Amano, H., & Noda, H. (2002). Physiological activities of oligosaccharides derived from marine algae Porphyra yezoensis Porphyran. Fisheries Science, 68(Suppl. 2), 1441–1444. https://doi.org/10.2331/fishsci.68.sup2_1441
  • Persiyanova, E. V., Kuznetsova, T. A., & Silchenko, A. S. (2020). Effect of sulfated polysaccharides from marine hydrobionts on humoral immune response to ovalbumin in mice. Bulletin of Experimental Biology and Medicine, 169(2), 246–248. https://doi.org/10.1007/s10517-020-04860-3
  • Pluske, J. R., Thompson, M. J., Atwood, C. S., Bird, P. H., Williams, I. H., & Hartmann, P. E. (1996). Maintenance of villus height and crypt depth, and enhancement of disaccharide digestion and monosaccharide absorption, in piglets fed on cows’ whole milk after weaning. British Journal of Nutrition, 76(3), 409–422. https://doi.org/10.1079/BJN19960046
  • Ray, B. (2006). Polysaccharides from Enteromorpha compressa: Isolation, purification and structural features. Carbohydrate Polymers, 66(3), 408–416. https://doi.org/10.1016/j.carbpol.2006.03.027
  • Renz, H., Allen, K. J., Sicherer, S. H., Sampson, H. A., Lack, G., Beyer, K., & Oettgen, H. C. (2018). Food allergy. Nature Reviews Disease Primers, 4(1), 1–20. https://doi.org/10.1038/nrdp.2017.98
  • Rodriguez, B., Prioult, G., Hacini-Rachinel, F., Moine, D., Bruttin, A., Ngom-Bru, C., Labellie, C., Nicolis, I., Berger, B., Mercenier, A., Butel, M.-J., & Waligora-Dupriet, A.-J. (2012). Infant gut microbiota is protective against cow's milk allergy in mice despite immature ileal T-cell response. FEMS Microbiology Ecology, 79(1), 192–202. https://doi.org/10.1111/j.1574-6941.2011.01207.x
  • Shi, C., Pan, T., Cao, M., Liu, Q., Zhang, L., & Liu, G. (2015). Suppression of Th2 immune responses by the sulfated polysaccharide from Porphyra haitanensis in tropomyosin-sensitized mice. International Immunopharmacology, 24(2), 211–218. https://doi.org/10.1016/j.intimp.2014.11.019
  • Stefka, A. T., Feehley, T., Tripathi, P., Qiu, J., McCoy, K., Mazmanian, S. K., Tjota, M. Y., Seo, G.-Y., Cao, S., Theriault, B. R., Antonopoulos, D. A., Zhou, L., Chang, E. B., Fu, Y.-X., & Nagler, C. R. (2014). Commensal bacteria protect against food allergen sensitization. Proceedings of the National Academy of Sciences, 111(36), 13145–13150. https://doi.org/10.1073/pnas.1412008111
  • Vickery, B. P., Scurlock, A. M., Jones, S. M., & Burks, A. W. (2011). Mechanisms of immune tolerance relevant to food allergy. Journal of Allergy and Clinical Immunology, 127(3), 576–584. https://doi.org/10.1016/j.jaci.2010.12.1116
  • Vidarsson, G., Dekkers, G., & Rispens, T. (2014). IgG subclasses and allotypes: From structure to effector functions. Frontiers in Immunology, 5, 520. https://doi.org/10.3389/fimmu.2014.00520
  • Wang, J., Zhang, Q., Zhang, Z., & Li, Z. (2008). Antioxidant activity of sulfated polysaccharide fractions extracted from Laminaria japonica. International Journal of Biological Macromolecules, 42(2), 127–132. https://doi.org/10.1016/j.ijbiomac.2007.10.003
  • Wei, Y.-J., Fang, R.-E., Ou, J.-Y., Pan, C.-L., & Huang, C.-H. (2022). Modulatory effects of Porphyra-derived polysaccharides, oligosaccharides and their mixture on antigen-specific immune responses in ovalbumin-sensitized mice. Journal of Functional Foods, 96, 105209. https://doi.org/10.1016/j.jff.2022.105209
  • Wu, G., Gu, W., Chen, G., Cheng, H., Li, D., & Xie, Z. (2023). Interactions of tea polysaccharides with gut microbiota and their health-promoting effects to host: Advances and perspectives. Journal of Functional Foods, 102, 105468. https://doi.org/10.1016/j.jff.2023.105468
  • Wu, S.-C., Wen, T.-N., & Pan, C.-L. (2005). Algal-oligosaccharide-lysates prepared by two bacterial agarases stepwise hydrolyzed and their anti-oxidative properties. Fisheries Science, 71(5), 1149–1159. https://doi.org/10.1111/j.1444-2906.2005.01075.x
  • Zheng, P., Zhang, K., Lv, X., Liu, C., Wang, Q., & Bai, X. (2022). Gut microbiome and metabolomics profiles of allergic and non-allergic childhood asthma. Journal of Asthma and Allergy, 15, 419–435. https://doi.org/10.2147/JAA.S354870