Pub Date : 2025-02-01DOI: 10.1016/j.jff.2025.106669
Alan Aitchison, Teagan S. Edwards, Andrew S. Day
Gastrointestinal digestion of bovine milk protein β-casein releases a biologically active peptide β-casomorphin (BCM)-7, which has been associated with increased incidence of non-communicable diseases and negative effects on digestive comfort. In this study, casein isolates containing both β-casein isoforms were enzymatically digested using a previously described method. The digestion products were incubated with intestinal epithelial cells with or without the presence of an inflammatory cytokine, interleukin (IL)-1β, and the effects on inflammatory markers studied. BCM-7 was released from digestion of both β-casein isoforms although higher concentrations were released from A1 β-casein. No difference was found in the effects of the digestion products of each β-casein on gene expression of inflammatory markers interleukin (IL)-8, carcinoembryonic antigen-related cell adhesion molecule (CEACAM)6 or intestinal alkaline phosphatase (IAP). Together, the results of this study do not provide insight for the reported health benefits of milk containing A2 β-casein over milk containing the A1 isoform.
{"title":"Simulated gastrointestinal digestion of milk caseins containing A1 β-casein does not lead to increased production of inflammatory proteins in intestinal epithelial cells in vitro when compared to milk caseins containing A2 β-casein.","authors":"Alan Aitchison, Teagan S. Edwards, Andrew S. Day","doi":"10.1016/j.jff.2025.106669","DOIUrl":"10.1016/j.jff.2025.106669","url":null,"abstract":"<div><div>Gastrointestinal digestion of bovine milk protein β-casein releases a biologically active peptide β-casomorphin (BCM)-7, which has been associated with increased incidence of non-communicable diseases and negative effects on digestive comfort. In this study, casein isolates containing both β-casein isoforms were enzymatically digested using a previously described method. The digestion products were incubated with intestinal epithelial cells with or without the presence of an inflammatory cytokine, interleukin (IL)-1β, and the effects on inflammatory markers studied. BCM-7 was released from digestion of both β-casein isoforms although higher concentrations were released from A1 β-casein. No difference was found in the effects of the digestion products of each β-casein on gene expression of inflammatory markers interleukin (IL)-8, carcinoembryonic antigen-related cell adhesion molecule (CEACAM)6 or intestinal alkaline phosphatase (IAP). Together, the results of this study do not provide insight for the reported health benefits of milk containing A2 β-casein over milk containing the A1 isoform.</div></div>","PeriodicalId":360,"journal":{"name":"Journal of Functional Foods","volume":"125 ","pages":"Article 106669"},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143182949","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01DOI: 10.1016/j.jff.2025.106691
Wenna Yao , Ruilin Du , Shuo Yan , Teligun Bao , Huimin Zhang , Fang Yang , Yue Xue , Yulong Zhao , Siqin Bao , Xihe Li , Yongli Song
<div><div>Inflammatory bowel disease (IBD) is a recurrent inflammatory disease affecting the gastrointestinal tract, including ulcerative colitis (UC) and Crohn's disease (CD). IBD mainly causes intestinal mucosal injury, intestinal flora and metabolite disturbance. An imbalance between pro-inflammatory and anti-inflammatory cytokines also is a hallmark of IBD. At present, IBD is treated with medicines. The latest studies have revealed the adverse effects of drugs on people's health. Numerous studies have shown that animal milk can effectively protect the intestinal mucosa and alleviate UC symptoms. It also exhibits antibacterial, antioxidant, anticancer and anti-inflammatory properties. Sheep milk (SM) contains a higher concentration of fatty acids, immunoglobulins, proteins, hormones, vitamins and minerals, which exhibits anti-inflammatory properties. The DSS-induced colitis mice are a chemically induced model that mimics UC-like pathology. However, it is unclear whether gut microbes and metabolites play a role in preventing and treating UC in sheep milk. In this study, 30 mice were randomly divided into 3 groups. Mice had free access to sheep milk for 14 days, and free access to 2.5 % DSS started on the 7th day of SM administration and sustained for 7 days. In this study, q-PCR, ELISA, IHC, microbiome, untargeted metabolome and transcriptome were employed to investigate the mechanisms of sheep milk treatment in colitis. ELISA and qPCR analysis showed that relative mRNA expression levels of <em>TNF-α, IL-1β, IL-6,</em> and <em>IL-17</em> were decreased in SM treatment compared to DSS-induced colitis mice. In the DSS group, LGR5, KI67, and MUC2 were significantly lower than in the CON group, but the SM treatment increased these protein expression levels. We also found that the tight junction proteins expression level was elevated in the SM group compared to the DSS group. Microbiome analysis showed that potentially harmful genera, such as <em>unidentified_Enterobacteriaceae</em> and <em>Romboutsia</em> decreased. In contrast, beneficial genera, such as <em>Alloprevotella</em> and <em>Muribaculum</em> increased in the SM group compared to the DSS group. The KEGG of non-targeted metabolome analysis showed that these metabolites were involved in amino acid metabolism, such as phenylalanine metabolism and propanoate metabolism in the SM group compared to the DSS group. Heatmap analysis showed that the purine and tryptophan metabolism signaling pathways changed, especially kynurenic acid and indole-3-glycol aldehyde were significantly downregulated in the SM group compared to the DSS group. Transcriptome analysis demonstrated that the expression of genes related to the IL-17 signaling and PI3K-Akt signaling pathways, such as <em>S100A8</em>, <em>S100A9</em>, <em>LNC2</em>, <em>CXCL3</em>, <em>CREB313</em>, <em>CREB314</em> and <em>CHRM1</em> were downregulated in the SM group compared to the DSS group. The results suggest that sheep milk can effectiv
{"title":"Integrated microbiome, transcriptome and metabolome insight into the alleviating mechanisms of sheep milk in a DSS-induced colitis mouse model","authors":"Wenna Yao , Ruilin Du , Shuo Yan , Teligun Bao , Huimin Zhang , Fang Yang , Yue Xue , Yulong Zhao , Siqin Bao , Xihe Li , Yongli Song","doi":"10.1016/j.jff.2025.106691","DOIUrl":"10.1016/j.jff.2025.106691","url":null,"abstract":"<div><div>Inflammatory bowel disease (IBD) is a recurrent inflammatory disease affecting the gastrointestinal tract, including ulcerative colitis (UC) and Crohn's disease (CD). IBD mainly causes intestinal mucosal injury, intestinal flora and metabolite disturbance. An imbalance between pro-inflammatory and anti-inflammatory cytokines also is a hallmark of IBD. At present, IBD is treated with medicines. The latest studies have revealed the adverse effects of drugs on people's health. Numerous studies have shown that animal milk can effectively protect the intestinal mucosa and alleviate UC symptoms. It also exhibits antibacterial, antioxidant, anticancer and anti-inflammatory properties. Sheep milk (SM) contains a higher concentration of fatty acids, immunoglobulins, proteins, hormones, vitamins and minerals, which exhibits anti-inflammatory properties. The DSS-induced colitis mice are a chemically induced model that mimics UC-like pathology. However, it is unclear whether gut microbes and metabolites play a role in preventing and treating UC in sheep milk. In this study, 30 mice were randomly divided into 3 groups. Mice had free access to sheep milk for 14 days, and free access to 2.5 % DSS started on the 7th day of SM administration and sustained for 7 days. In this study, q-PCR, ELISA, IHC, microbiome, untargeted metabolome and transcriptome were employed to investigate the mechanisms of sheep milk treatment in colitis. ELISA and qPCR analysis showed that relative mRNA expression levels of <em>TNF-α, IL-1β, IL-6,</em> and <em>IL-17</em> were decreased in SM treatment compared to DSS-induced colitis mice. In the DSS group, LGR5, KI67, and MUC2 were significantly lower than in the CON group, but the SM treatment increased these protein expression levels. We also found that the tight junction proteins expression level was elevated in the SM group compared to the DSS group. Microbiome analysis showed that potentially harmful genera, such as <em>unidentified_Enterobacteriaceae</em> and <em>Romboutsia</em> decreased. In contrast, beneficial genera, such as <em>Alloprevotella</em> and <em>Muribaculum</em> increased in the SM group compared to the DSS group. The KEGG of non-targeted metabolome analysis showed that these metabolites were involved in amino acid metabolism, such as phenylalanine metabolism and propanoate metabolism in the SM group compared to the DSS group. Heatmap analysis showed that the purine and tryptophan metabolism signaling pathways changed, especially kynurenic acid and indole-3-glycol aldehyde were significantly downregulated in the SM group compared to the DSS group. Transcriptome analysis demonstrated that the expression of genes related to the IL-17 signaling and PI3K-Akt signaling pathways, such as <em>S100A8</em>, <em>S100A9</em>, <em>LNC2</em>, <em>CXCL3</em>, <em>CREB313</em>, <em>CREB314</em> and <em>CHRM1</em> were downregulated in the SM group compared to the DSS group. The results suggest that sheep milk can effectiv","PeriodicalId":360,"journal":{"name":"Journal of Functional Foods","volume":"125 ","pages":"Article 106691"},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143182948","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01DOI: 10.1016/j.jff.2025.106678
Xiao-jun Yu , Ling-mei Kong , Bin Wang , Chang-qi Zhai , Yong-zhen Lao , Long-jian Zhang , Yan Gao , Bo-Nian Zhao
Asini Corii Colla (Ejiao) is recognized as an excellent source that bridges both medicinal and nutritional benefits. It is rich in protein, polypeptides, amino acids, fatty acids, polysaccharides, and other components. It offers a range of therapeutic benefits, including anti-anemia properties, immune regulation, hemostasis, anti-inflammatory effects, antioxidant activity, and antibacterial properties. Beyond its medicinal uses, the food industry uses Asini Corii Colla to produce foods like Ejiao cake and Ejiao porridge, which are both delicious and nutritious and promote health, making them popular among consumers. This review aims to provide comprehensive data on the chemical composition, pharmacological activity, quality control, and modern applications of Asini Corii Colla. Additionally, it addresses current research gaps and suggests future directions and methodologies for studying food and medicine derived from animal sources to guide subsequent research and applications.
{"title":"Asini Corii Colla (Ejiao) as a health-care food and traditional Chinese medicine: A review of its chemical composition, pharmacological activity, quality control, modern applications","authors":"Xiao-jun Yu , Ling-mei Kong , Bin Wang , Chang-qi Zhai , Yong-zhen Lao , Long-jian Zhang , Yan Gao , Bo-Nian Zhao","doi":"10.1016/j.jff.2025.106678","DOIUrl":"10.1016/j.jff.2025.106678","url":null,"abstract":"<div><div>Asini Corii Colla (Ejiao) is recognized as an excellent source that bridges both medicinal and nutritional benefits. It is rich in protein, polypeptides, amino acids, fatty acids, polysaccharides, and other components. It offers a range of therapeutic benefits, including anti-anemia properties, immune regulation, hemostasis, anti-inflammatory effects, antioxidant activity, and antibacterial properties. Beyond its medicinal uses, the food industry uses Asini Corii Colla to produce foods like Ejiao cake and Ejiao porridge, which are both delicious and nutritious and promote health, making them popular among consumers. This review aims to provide comprehensive data on the chemical composition, pharmacological activity, quality control, and modern applications of Asini Corii Colla. Additionally, it addresses current research gaps and suggests future directions and methodologies for studying food and medicine derived from animal sources to guide subsequent research and applications.</div></div>","PeriodicalId":360,"journal":{"name":"Journal of Functional Foods","volume":"125 ","pages":"Article 106678"},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143183670","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
In this study we aimed to explore the protective effects of a probiotic yeast Pichia kudriavzevii (Y1) and its cell-free supernatant (CFS-Y1) during infection simulated in vitro using Caco-2 cells. Results showed that both live P. kudriavzevii and CFS-Y1 effectively decreased Salmonella spp. adhesion and invasion, in competition, inhibition and displacement assays, displaying the most potent inhibitory effects when Caco-2 cells were preincubated with Y1/ CFS-Y1 at different concentrations and then bacteria were added for an additional time. Furthermore, the treatment of LPS-induced Caco-2 cells with both Y1 and its CFS-Y1 effectively relieved the oxidative stress and improved the intestinal TJ barrier function. In particular, Y1 and CFS-Y-1 prevented damage to the intestinal epithelial function in LPS-induced Caco-2 cells by upregulating TJ protein expression and reducing ROS accumulation, as well as overproducing anti-inflammatory cytokines. Therefore, the Y1 and its CFS-Y1 possess potential effects on the maintenance of human health and could be considered as a dietary supplementation for food and a promising approach to treat intestinal barrier dysfunction.
{"title":"Protective and immunomodulatory effects of the novel probiotic yeast Pichia kudriavzevii isolated from a home-made kefir during infection in human colon epithelial cells: An exploratory study","authors":"Angela Maione , Marianna Imparato , Annalisa Buonanno , Marilena Galdiero , Elisabetta de Alteriis , Marco Guida , Emilia Galdiero","doi":"10.1016/j.jff.2025.106666","DOIUrl":"10.1016/j.jff.2025.106666","url":null,"abstract":"<div><div>In this study we aimed to explore the protective effects of a probiotic yeast <em>Pichia kudriavzevii</em> (Y1) and its cell-free supernatant (CFS-Y1) during infection simulated in vitro using Caco-2 cells. Results showed that both live <em>P. kudriavzevii</em> and CFS-Y1 effectively decreased <em>Salmonella</em> spp. adhesion and invasion, in competition, inhibition and displacement assays, displaying the most potent inhibitory effects when Caco-2 cells were preincubated with Y1/ CFS-Y1 at different concentrations and then bacteria were added for an additional time. Furthermore, the treatment of LPS-induced Caco-2 cells with both Y1 and its CFS-Y1 effectively relieved the oxidative stress and improved the intestinal TJ barrier function. In particular, Y1 and CFS-Y-1 prevented damage to the intestinal epithelial function in LPS-induced Caco-2 cells by upregulating TJ protein expression and reducing ROS accumulation, as well as overproducing anti-inflammatory cytokines. Therefore, the Y1 and its CFS-Y1 possess potential effects on the maintenance of human health and could be considered as a dietary supplementation for food and a promising approach to treat intestinal barrier dysfunction.</div></div>","PeriodicalId":360,"journal":{"name":"Journal of Functional Foods","volume":"125 ","pages":"Article 106666"},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143183687","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<div><div>Soybean oil, rich in linoleic acid, plays a crucial role in lowering blood lipids and cholesterol, which helps prevent cardiovascular diseases. The lecithin content in soybeans is also essential for the development and growth of the nervous system, blood vessels, and brain. Canola oil, with a higher level of Omega-3 polyunsaturated fatty acids, helps soften blood vessels and can prevent atherosclerosis, offering benefits for both cardiovascular and cerebrovascular health. Blending these oils can create a synergistic effect, maximizing their health benefits. Furthermore, all three oils should meet the national standard GB 2716–2018 for vegetable oils. Peony seed oil, high in unsaturated fatty acids, particularly α-linolenic acid, has shown significant hypoglycemic effects. To enhance these effects, soybean and canola oils, which contain beneficial components like linoleic acid and oleic acid, were blended with peony seed oil. This study aimed to evaluate the combined hypoglycemic potential of these oils through in vitro and in vivo experiments, supported by network pharmacology and molecular docking analyses to explore the underlying mechanisms. The research provides valuable insights into the potential therapeutic use of this blended oil in diabetes management, offering a natural alternative for hypoglycemic treatments. <span><span>Zhao et al. (2021)</span></span>, <span><span>Suyun et al. (2024)</span></span> (<span><span>Suyun et al., 2024</span></span>; <span><span>Zhao et al., 2021</span></span>). In vitro and in vivo experiments, as well as network pharmacology and molecular docking analysis, were conducted to evaluate the hypoglycemic effects of peony seed oil, soybean oil, rapeseed oil, and their blends. GC–MS analysis identified nine major components, including γ-sitosterol, α-linolenic acid, and linoleic acid (<span><span><em>Journal of Food Biochemistry</em>, 2020</span></span>). The results showed that the optimal blend ratio of peony seed oil to soybean and rapeseed oils was 60:27:11, which significantly inhibited α-amylase and α-glucosidase activities. Network pharmacology and molecular docking revealed that α-linolenic acid and linoleic acid are key active compounds, targeting PPARG and MAPK3, primarily through the PPAR and IL17 signaling pathways. This multi-component, multi-target, and multi-pathway approach offers potential synergistic benefits for diabetes treatment. The study provides valuable insights for the development of novel hypoglycemic agents (<span><span>Yang, 2014</span></span> (<span><span>Yang, 2014</span></span>)).</div><div>The study evaluated the combined hypoglycemic effects of peony seed oil, soybean oil, rapeseed oil, and their blends through in vitro and in vivo experiments, supported by network pharmacology and molecular docking analyses. GC–MS analysis identified nine major components, including γ-sitosterol, α-linolenic acid, and linoleic acid (<span><span>Zhao et al., 2021</span></span>; <span><
{"title":"Active ingredients and mechanisms of peony seed blended oil in diabetes treatment using Kunming mice: Insights from GC–MS, network pharmacology, and experimental verification","authors":"Yuning Gu, Fengliang Zhong, Tiantian Sun, Zirui Chang","doi":"10.1016/j.jff.2024.106652","DOIUrl":"10.1016/j.jff.2024.106652","url":null,"abstract":"<div><div>Soybean oil, rich in linoleic acid, plays a crucial role in lowering blood lipids and cholesterol, which helps prevent cardiovascular diseases. The lecithin content in soybeans is also essential for the development and growth of the nervous system, blood vessels, and brain. Canola oil, with a higher level of Omega-3 polyunsaturated fatty acids, helps soften blood vessels and can prevent atherosclerosis, offering benefits for both cardiovascular and cerebrovascular health. Blending these oils can create a synergistic effect, maximizing their health benefits. Furthermore, all three oils should meet the national standard GB 2716–2018 for vegetable oils. Peony seed oil, high in unsaturated fatty acids, particularly α-linolenic acid, has shown significant hypoglycemic effects. To enhance these effects, soybean and canola oils, which contain beneficial components like linoleic acid and oleic acid, were blended with peony seed oil. This study aimed to evaluate the combined hypoglycemic potential of these oils through in vitro and in vivo experiments, supported by network pharmacology and molecular docking analyses to explore the underlying mechanisms. The research provides valuable insights into the potential therapeutic use of this blended oil in diabetes management, offering a natural alternative for hypoglycemic treatments. <span><span>Zhao et al. (2021)</span></span>, <span><span>Suyun et al. (2024)</span></span> (<span><span>Suyun et al., 2024</span></span>; <span><span>Zhao et al., 2021</span></span>). In vitro and in vivo experiments, as well as network pharmacology and molecular docking analysis, were conducted to evaluate the hypoglycemic effects of peony seed oil, soybean oil, rapeseed oil, and their blends. GC–MS analysis identified nine major components, including γ-sitosterol, α-linolenic acid, and linoleic acid (<span><span><em>Journal of Food Biochemistry</em>, 2020</span></span>). The results showed that the optimal blend ratio of peony seed oil to soybean and rapeseed oils was 60:27:11, which significantly inhibited α-amylase and α-glucosidase activities. Network pharmacology and molecular docking revealed that α-linolenic acid and linoleic acid are key active compounds, targeting PPARG and MAPK3, primarily through the PPAR and IL17 signaling pathways. This multi-component, multi-target, and multi-pathway approach offers potential synergistic benefits for diabetes treatment. The study provides valuable insights for the development of novel hypoglycemic agents (<span><span>Yang, 2014</span></span> (<span><span>Yang, 2014</span></span>)).</div><div>The study evaluated the combined hypoglycemic effects of peony seed oil, soybean oil, rapeseed oil, and their blends through in vitro and in vivo experiments, supported by network pharmacology and molecular docking analyses. GC–MS analysis identified nine major components, including γ-sitosterol, α-linolenic acid, and linoleic acid (<span><span>Zhao et al., 2021</span></span>; <span><","PeriodicalId":360,"journal":{"name":"Journal of Functional Foods","volume":"125 ","pages":"Article 106652"},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143183667","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ulcerative colitis is a type of inflammatory bowel disease (IBD) that has risen around the world in recent years. Diet, lifestyle, genetics, stress, are the risk factors of ulcerative colitis. Indeed, oxidative stress, impairment of the intestinal barrier, inflammation, and dysbiosis of gut microbiota are the main pathways involved in ulcerative colitis. Therefore, using the anti-inflammatory and antioxidant ingredients can modulate the ulcerative colitis symptoms. Recent investigations in functional foods have demonstrated that diet and fortified foods play major therapeutic effects in human health, thus, the natural remedies have become an attractive approach. Among various natural substances, algae and marine products are valuable sources and rich in a wide range of metabolites with biological activities that can ameliorate ulcerative colitis signs. It has been reported that algal sulfated polysaccharides, peptides, pigments, polyphenols, and marine oil are useful for controlling ulcerative colitis via different pathways including modulating of TNF-α and other related inflammatory cytokines secretion, increasing the antioxidant enzymes [superoxide dismutase (SOD) and catalase(CAT)] activities, reduction of myeloperoxidase (MPO) activity and malondialdehyde (MDA) production, improving the intestinal barrier function and repairing histological damage in the colon. This review focused on the pathology of ulcerative colitis, pathogenesis and key inflammatory pathways, and the role of algae and marine-based metabolites for reduction of ulcerative colitis signs via alleviating the inflammation cytokines secretion, and improving the barrier function and gut microbiota.
{"title":"Health benefits of algae and marine-derived bioactive metabolites for modulating ulcerative colitis symptoms","authors":"Najmeh Oliyaei , Saghar Zekri , Aida Iraji , Alireza Oliyaei , Romina Tanideh , Nadiar M. Mussin , Amin Tamadon , Nader Tanideh","doi":"10.1016/j.jff.2025.106690","DOIUrl":"10.1016/j.jff.2025.106690","url":null,"abstract":"<div><div>Ulcerative colitis is a type of inflammatory bowel disease (IBD) that has risen around the world in recent years. Diet, lifestyle, genetics, stress, are the risk factors of ulcerative colitis. Indeed, oxidative stress, impairment of the intestinal barrier, inflammation, and dysbiosis of gut microbiota are the main pathways involved in ulcerative colitis. Therefore, using the anti-inflammatory and antioxidant ingredients can modulate the ulcerative colitis symptoms. Recent investigations in functional foods have demonstrated that diet and fortified foods play major therapeutic effects in human health, thus, the natural remedies have become an attractive approach. Among various natural substances, algae and marine products are valuable sources and rich in a wide range of metabolites with biological activities that can ameliorate ulcerative colitis signs. It has been reported that algal sulfated polysaccharides, peptides, pigments, polyphenols, and marine oil are useful for controlling ulcerative colitis via different pathways including modulating of TNF-α and other related inflammatory cytokines secretion, increasing the antioxidant enzymes [superoxide dismutase (SOD) and catalase(CAT)] activities, reduction of myeloperoxidase (MPO) activity and malondialdehyde (MDA) production, improving the intestinal barrier function and repairing histological damage in the colon. This review focused on the pathology of ulcerative colitis, pathogenesis and key inflammatory pathways, and the role of algae and marine-based metabolites for reduction of ulcerative colitis signs via alleviating the inflammation cytokines secretion, and improving the barrier function and gut microbiota.</div></div>","PeriodicalId":360,"journal":{"name":"Journal of Functional Foods","volume":"125 ","pages":"Article 106690"},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143183683","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01DOI: 10.1016/j.jff.2025.106689
Xu Zhang , Yan Luo , Li Qin , Yage Ma , Dan Chen , Shenglan Zhao
{"title":"Corrigendum to “Effects of walnut shell extract on lipid metabolism and serum metabolomics in rats with high-fat diet-induced obesity” [Journal of Functional Foods, 122 (2024) 106558]","authors":"Xu Zhang , Yan Luo , Li Qin , Yage Ma , Dan Chen , Shenglan Zhao","doi":"10.1016/j.jff.2025.106689","DOIUrl":"10.1016/j.jff.2025.106689","url":null,"abstract":"","PeriodicalId":360,"journal":{"name":"Journal of Functional Foods","volume":"125 ","pages":"Article 106689"},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143347854","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01DOI: 10.1016/j.jff.2025.106668
Jinjia Liu , Songhao Tian , Jixiang Wang , Qing Sang , Wenting Su , Jinqiang Wu , Fenglei Zai
This study aimed to evaluate the hepatoprotective effects of Propolis ethanol extract (PEE) in D-galactose (D-gal)-induced senescence mice and explore its underlying mechanisms. Mice were administered PEE by gavage, and body weight, organ index, and serum biomarkers (SOD, MDA, AST, ALT) were measured. Liver histopathological changes were examined using HE staining. Additionally, transcriptomics and metabolomics analyses were employed to elucidate the molecular mechanisms. The results demonstrated that PEE ameliorated the reductions in body weight and liver index in aged mice. PEE significantly increased serum SOD levels and reduced MDA, AST, and ALT levels. Histological analysis revealed that PEE mitigated liver damage. Mechanistically, PEE appeared to exert hepatoprotective effects by modulating the insulin resistance pathway, particularly through the PI3K-AKT/PP1 signaling axis, thereby enhancing glycogen synthesis in the damaged liver. These findings provide a scientific foundation for the therapeutic potential of PEE in anti-aging and liver disease.
{"title":"Hepatoprotective effects of propolis ethanol extract on D-galactose-induced aging mice: A metabolomics and transcriptomics analysis focusing on insulin resistance pathways","authors":"Jinjia Liu , Songhao Tian , Jixiang Wang , Qing Sang , Wenting Su , Jinqiang Wu , Fenglei Zai","doi":"10.1016/j.jff.2025.106668","DOIUrl":"10.1016/j.jff.2025.106668","url":null,"abstract":"<div><div>This study aimed to evaluate the hepatoprotective effects of Propolis ethanol extract (PEE) in D-galactose (D-gal)-induced senescence mice and explore its underlying mechanisms. Mice were administered PEE by gavage, and body weight, organ index, and serum biomarkers (SOD, MDA, AST, ALT) were measured. Liver histopathological changes were examined using HE staining. Additionally, transcriptomics and metabolomics analyses were employed to elucidate the molecular mechanisms. The results demonstrated that PEE ameliorated the reductions in body weight and liver index in aged mice. PEE significantly increased serum SOD levels and reduced MDA, AST, and ALT levels. Histological analysis revealed that PEE mitigated liver damage. Mechanistically, PEE appeared to exert hepatoprotective effects by modulating the insulin resistance pathway, particularly through the PI3K-AKT/PP1 signaling axis, thereby enhancing glycogen synthesis in the damaged liver. These findings provide a scientific foundation for the therapeutic potential of PEE in anti-aging and liver disease.</div></div>","PeriodicalId":360,"journal":{"name":"Journal of Functional Foods","volume":"125 ","pages":"Article 106668"},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143183688","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01DOI: 10.1016/j.jff.2025.106676
Tan Wang , Rongrong Li , Ping Niu , Zhen Wei , Donge Xie , Huorong Huang , Jiayao Pan , Chunshu Rong
Astaxanthin exhibits promising anti-inflammatory effects in the treatment of neurodegenerative diseases. Our aim was to explore the mechanisms by which astaxanthin mitigates inflammation associated with these conditions. We treated HT22 cells with lipopolysaccharide (LPS), astaxanthin (ATX), and the TLR4 inhibitor TAK-242, employing CCK-8 assays, flow cytometry, and Western blotting to assess cell survival, oxidative stress, inflammation, and apoptosis. We found that LPS decreased the survival rate of HT22 cells, induced reactive oxygen species (ROS) accumulation and apoptosis, and increased the expression of inflammation-related proteins. Conversely, ATX diminished LPS-induced ROS accumulation and apoptosis, as well as the expression of inflammation-related proteins. The application of TLR4 inhibitors further enhanced the anti-inflammatory effects of ATX on LPS. In summary, we demonstrated that ATX can alleviate LPS-induced ROS accumulation, inflammation, and apoptosis, with its anti-inflammatory effects closely linked to the TLR4/MyD88/NFκB signaling pathway.
{"title":"Astaxanthin relieves HT22 cells from LPS-induced inflammation and apoptosis by inhibiting oxygen species and regulating the TLR4/MyD88/NFκB signaling pathway","authors":"Tan Wang , Rongrong Li , Ping Niu , Zhen Wei , Donge Xie , Huorong Huang , Jiayao Pan , Chunshu Rong","doi":"10.1016/j.jff.2025.106676","DOIUrl":"10.1016/j.jff.2025.106676","url":null,"abstract":"<div><div>Astaxanthin exhibits promising anti-inflammatory effects in the treatment of neurodegenerative diseases. Our aim was to explore the mechanisms by which astaxanthin mitigates inflammation associated with these conditions. We treated HT22 cells with lipopolysaccharide (LPS), astaxanthin (ATX), and the TLR4 inhibitor TAK-242, employing CCK-8 assays, flow cytometry, and Western blotting to assess cell survival, oxidative stress, inflammation, and apoptosis. We found that LPS decreased the survival rate of HT22 cells, induced reactive oxygen species (ROS) accumulation and apoptosis, and increased the expression of inflammation-related proteins. Conversely, ATX diminished LPS-induced ROS accumulation and apoptosis, as well as the expression of inflammation-related proteins. The application of TLR4 inhibitors further enhanced the anti-inflammatory effects of ATX on LPS. In summary, we demonstrated that ATX can alleviate LPS-induced ROS accumulation, inflammation, and apoptosis, with its anti-inflammatory effects closely linked to the TLR4/MyD88/NFκB signaling pathway.</div></div>","PeriodicalId":360,"journal":{"name":"Journal of Functional Foods","volume":"125 ","pages":"Article 106676"},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143183054","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-01DOI: 10.1016/j.jff.2025.106682
Shanshan Li , Meng Wang , Buyun Jia , Baikun Li , Hui Cheng , Shasha Tian , Qinglin Li
Aims
To investigate the effect of GAS in PD models by regulating ferroptosis and cell senescence.
Methods
MPTP or MPP+ was used to establish PD models in vivo or in vitro, and the effect of GAS was detected by behavioral analysis, immunohistochemistry and cell viability. At the same time, transmission electron microscopy, biochemical kits, Western blotting, immunofluorescence, and molecular docking were used.
Results
We found that gastrodin could inhibit cell senescence in PD models. In addition, GAS was found to inhibit ferroptosis, regulate oxidative stress level, and level of ACSL4, FTH1, SLC7A11, GPX4. We also found that Ferrostatin-1 and GAS could inhibit cell senescence. The results of molecular docking and immunofluorescence showed that p53 may be an important target of GAS. After treatment with Pifithrin-α, the protective effect of GAS was enhanced.
Conclusion
GAS inhibits cellular senescence in PD models and treats PD through a novel mechanism related to ferroptosis.
{"title":"Neuroprotective effects of gastrodin in both in vivo and in vitro models of Parkinson's disease: Mechanisms from the perspective of oxidative stress, ferroptosis, and cellular senescence","authors":"Shanshan Li , Meng Wang , Buyun Jia , Baikun Li , Hui Cheng , Shasha Tian , Qinglin Li","doi":"10.1016/j.jff.2025.106682","DOIUrl":"10.1016/j.jff.2025.106682","url":null,"abstract":"<div><h3>Aims</h3><div>To investigate the effect of GAS in PD models by regulating ferroptosis and cell senescence.</div></div><div><h3>Methods</h3><div>MPTP or MPP<sup>+</sup> was used to establish PD models in vivo or in vitro, and the effect of GAS was detected by behavioral analysis, immunohistochemistry and cell viability. At the same time, transmission electron microscopy, biochemical kits, Western blotting, immunofluorescence, and molecular docking were used.</div></div><div><h3>Results</h3><div>We found that gastrodin could inhibit cell senescence in PD models. In addition, GAS was found to inhibit ferroptosis, regulate oxidative stress level, and level of ACSL4, FTH1, SLC7A11, GPX4. We also found that Ferrostatin-1 and GAS could inhibit cell senescence. The results of molecular docking and immunofluorescence showed that p53 may be an important target of GAS. After treatment with Pifithrin-α, the protective effect of GAS was enhanced.</div></div><div><h3>Conclusion</h3><div>GAS inhibits cellular senescence in PD models and treats PD through a novel mechanism related to ferroptosis.</div></div>","PeriodicalId":360,"journal":{"name":"Journal of Functional Foods","volume":"125 ","pages":"Article 106682"},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143183057","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}