1
|
Vissers E, Wellens J, Giorio L, Zadora W, Verstockt B, Ferrante M, Vermeire S, Matthys C, Arnauts K, Sabino J. Dietary Carrageenan Amplifies the Inflammatory Profile, but not Permeability, of Intestinal Epithelial Cells from Patients With Crohn's Disease. Inflamm Bowel Dis 2025; 31:1392-1403. [PMID: 39720875 PMCID: PMC12069985 DOI: 10.1093/ibd/izae306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND The consumption of ultra-processed foods has increased significantly worldwide and is associated with the rise in inflammatory bowel diseases. However, any causative factors and their underlying mechanisms are yet to be identified. This study aimed to further elucidate whether different types of the dietary emulsifier carrageenan (CGN) can alter the permeability and inflammatory state of the intestinal epithelium. METHODS Caco-2/HT29-MTX cocultures (n = 4) were exposed to either κ-, ι-, or λ-CGN (100 µg mL-1) for 24 hours. Organoid-derived monolayers from patients with Crohn's Disease (CD) were exposed to κ-CGN (100 µg mL-1) for 48 hours (n = 10). In both models, an inflamed condition was established by adding a mix of inflammatory stimuli. Changes in permeability were measured by transepithelial electrical resistance (TEER). In the organoid-derived monolayers, cytokines were quantified in the apical and basolateral supernatant and gene expression was analyzed with RT-qPCR. RESULTS None of the CGN subtypes altered permeability of non-inflamed or inflamed Caco-2/HT29-MTX cocultures. In organoid-derived monolayers, κ-CGN did not affect TEER, but induced alterations in the gene expression of tight junctions and mucus proteins. Expression of TNF, IL8, and IL1B increased upon κ-CGN stimulation, both in inflamed and non-inflamed monolayers. Cytokine release in the supernatant was increased by κ-CGN for IL-6, IL-13, IL-4, IL-2, and IL-10. CONCLUSIONS Dietary CGN caused upregulation of inflammatory markers and affected cytokine release of intestinal epithelial cells from CD patients, while permeability remained unaltered. When inflammation was already present, this pro-inflammatory effect was more pronounced, suggesting a role for dietary CGN during active CD.
Collapse
Affiliation(s)
- Eva Vissers
- Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Judith Wellens
- Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Lorenzo Giorio
- Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Ward Zadora
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Bram Verstockt
- Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Marc Ferrante
- Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Séverine Vermeire
- Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Christophe Matthys
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Department of Endocrinology, UZ Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Kaline Arnauts
- Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - João Sabino
- Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, UZ Leuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
2
|
Solanki S, Taranto J, Rebernick R, Castillo C, Ponnusamy V, Sykes MM, Leiser SF, Lee JH, Schmidt T, Shah YM. Low Protein Diet Exacerbates Experimental Mouse Models of Colitis through Epithelial Autonomous and Nonautonomous Mechanisms. J Nutr 2025:S0022-3166(25)00186-5. [PMID: 40216295 DOI: 10.1016/j.tjnut.2025.03.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/30/2025] [Accepted: 03/04/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Patients with inflammatory bowel diseases (IBDs) are at risk of protein malnutrition due to increased protein loss or reduced dietary intake. The consequences of protein malnutrition on intestinal epithelial metabolism and disease progression remain poorly understood. OBJECTIVES Given the critical role of the mechanistic target of rapamycin complex 1 (mTORC1) as an amino acid sensor and a key regulator of intestinal epithelial metabolism and homeostasis, along with the well-established influence of diet on the gut microbiota and IBD, we focused on accessing the role of dietary protein in modulating intestinal epithelial mTORC1, determine the contributions of specific amino acids such as leucine and arginine, and examine the interplay between protein malnutrition and gut microbiota driving IBD. METHODS C57BL/6 mice were assigned to a control (20% protein, n = 6), a low protein (4% protein, n = 7), or diets selectively deficient in leucine, arginine, and other essential amino acids (n = 5-6). Colitis was induced by administering 2.5% dextran sulfate sodium in drinking water for 6 d. Intestinal epithelial mTORC1 activity was assessed by immunoblotting. Gut microbiota composition was characterized using 16S sequencing, and the microbiota's role in colitis was evaluated through broad-spectrum antibiotic treatment. Disease severity was quantified by monitoring weight loss, colon shortening, histopathological damage, and inflammatory cytokine expression. RESULTS Protein restriction increased the severity of dextran sulfate sodium-induced colitis compared to the control diet (∗∗∗P < 0.001). Mice fed arginine-restricted diets exhibited increased colitis (∗P < 0.05). Protein restriction induced significant alterations in gut microbiota composition, and antibiotic-mediated microbiota depletion partially ameliorated colitis severity, revealing a microbiota-dependent mechanism underlying disease exacerbation. CONCLUSIONS Our study demonstrates a complex interplay between dietary protein, epithelial mTORC1 signaling, and gut microbiota in modulating IBD pathogenesis and highlights the potential for targeted dietary strategies, including amino acid supplementation, to improve disease management in patients with IBD.
Collapse
Affiliation(s)
- Sumeet Solanki
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Joseph Taranto
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Ryan Rebernick
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Cristina Castillo
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Varun Ponnusamy
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Madeline M Sykes
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Scott F Leiser
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Jun Hee Lee
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Thomas Schmidt
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States; Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, United States.
| |
Collapse
|
3
|
Han Y, Guo X, Thanuphol P, Ji R, Zhu Z, Wu Y, Du H, Xiao H. Gut Microbiota-Mediated Degradation of Food-Grade Lambda-Carrageenan by Bacteroides xylanisolvens and Its Role in Inflammation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:4288-4298. [PMID: 39920146 DOI: 10.1021/acs.jafc.4c10159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Concerns about the safety of food additives have intensified among consumers, scientists, and policymakers. Ensuring the safety of these additives is crucial to public health. Carrageenan (CGN), a common additive in the food industry, has become the subject of controversy, particularly regarding whether it can be degraded in the gastrointestinal tract, forming degraded carrageenans (dCGNs) that may pose health risks. This study is among the first to identify Bacteroides xylanisolvens C3 as a key gut bacterium involved in the degradation of food-grade lambda-CGN (L-CGN). Using high-performance liquid chromatography (HPLC), thin-layer chromatography (TLC), and metabolic analysis, we confirmed the ability of this bacterium to degrade L-CGN. Importantly, we found that the microbiota-generated dCGNs significantly increased nitric oxide (NO) and COX-2 production and upregulated pro-inflammatory genes, including IL-1β, TNF-α, and IL-6, in macrophages. This study also highlights how microbial degradation of L-CGN can drive inflammation, particularly through the activation of the Nrf2 and NLRP3 pathways. These results suggest that microbial degradation of L-CGN in the gut may contribute to inflammation, underscoring the need to better understand microbial interactions with food-grade L-CGN, particularly in the context of colon health and inflammation-related diseases such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Yanhui Han
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
- Engineering Research Center for High-Valued Utilization of Fruit Resources in Western China, Ministry of Education, Shaanxi Normal University, Xi'an 710119, China
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Xiaojing Guo
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Pongpol Thanuphol
- Department of Food Chemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Ruya Ji
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Zhengjun Zhu
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yanyan Wu
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Hengjun Du
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
4
|
Shang X, Guo J, Chen P. Effect of Food Matrix on Regulation of Intestinal Barrier and Microbiota Homeostasis by Polysaccharides Sulfated Carrageenan. Foods 2025; 14:635. [PMID: 40002079 PMCID: PMC11854102 DOI: 10.3390/foods14040635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 02/27/2025] Open
Abstract
Carrageenan (CGN) has side effects on the intestinal barrier. Damage to the intestinal barrier is associated with exposure to sulfate groups. Food matrix has significant influence on the exposure quantity of sulfate groups and conformation in κ-CGN, but the corresponding side effects are not reported specifically. This study aimed to explore the regulatory effect of κ-CGN dissolved in aqueous (κ-CGN) and in 3% casein (κ-carrageenan-casein, κ-CC) on the intestinal barrier and microbiota homeostasis. Research has shown that both κ-CGN and κ-CC can induce different extents of intestinal barrier damage through disrupting microbiota homeostasis. Importantly, κ-CGN in casein with lower sulfate groups content was found to repair the intestinal barrier injury induced by an equivalent dose of κ-CGN aqueous through increasing the abundance of Oscillibacter and decreasing Weissella. These alleviating effects were reflected in lower levels of tumor necrosis factor (TNF)-α and C-reaction protein (CRP), higher levels of interleukin (IL)-10, raised secretion of mucus and goblet cells, and improved expression of epithelial cell compact proteins zonula occluden (ZO)-1 and mucin protein 2 (MUC2). This study states that κ-CGN in casein has a positive regulatory effect on the intestinal barrier damage compared to in aqueous solution, which can provide guidance for processing and utilization of CGN.
Collapse
Affiliation(s)
- Xuke Shang
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou 362000, China; (X.S.); (P.C.)
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou 362000, China
- Key Laboratory of Inshore Resources Biotechnology, Quanzhou Normal University, Quanzhou 362000, China
| | - Juanjuan Guo
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou 362000, China; (X.S.); (P.C.)
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou 362000, China
- Key Laboratory of Inshore Resources Biotechnology, Quanzhou Normal University, Quanzhou 362000, China
| | - Peilin Chen
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou 362000, China; (X.S.); (P.C.)
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou 362000, China
- Key Laboratory of Inshore Resources Biotechnology, Quanzhou Normal University, Quanzhou 362000, China
| |
Collapse
|
5
|
Gao F, Cheng C, Li R, Chen Z, Tang K, Du G. The role of Akkermansia muciniphila in maintaining health: a bibliometric study. Front Med (Lausanne) 2025; 12:1484656. [PMID: 39967592 PMCID: PMC11833336 DOI: 10.3389/fmed.2025.1484656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 01/21/2025] [Indexed: 02/20/2025] Open
Abstract
Background Akkermansia muciniphila, as a probiotic, is negatively linked to IBD, obesity, and T2DM. The aim of this study was to comprehensively assess the research status of Akkermansia muciniphila over the past decade and explore the relationships between this bacterium and various health-related aspects. Methods Tools VOSviewer, Bibliometrix, and CiteSpace were used to analyze various aspects including publication metrics, contributors, institutions, geography, journals, funding, and keywords. Results Over the past decade, research on Akkermansia muciniphila has demonstrated a consistent annual growth in the number of publications, with a notable peak in 2021. China led in the number of publications, totaling 151, whereas the United States exhibited a higher centrality value. Among the 820 institutions involved in the research, the University of California (from the United States) and the Chinese Academy of Sciences (from China) occupied central positions. Willem M. De Vos ranked at the top, with 12 publications and 1,108 citations. The journal GUT, which had 5,125 citations and an Impact Factor of 23.0 in 2024, was the most highly cited. The most cited articles deepened the understanding of the bacterium's impact on human health, spanning from basic research to translational medicine. Thirty-nine high-frequency keywords were grouped into five clusters, illustrating Akkermansia muciniphila's associations with metabolic diseases, chronic kidney disease, the gut-brain axis, intestinal inflammation, and Bacteroidetes-Firmicutes shifts. Conclusion Given Akkermansia muciniphila's anti-inflammatory and gut-barrier-strengthening properties, it holds promise as a therapeutic for obesity, metabolic disorders, and inflammatory conditions. Therefore, future research should explore its potential further by conducting clinical trials, elucidating its mechanisms of action, and investigating its efficacy and safety in diverse patient populations.
Collapse
Affiliation(s)
- Fangfang Gao
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Canyu Cheng
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Runwei Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Zongcun Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
- Department of Endocrinology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ke Tang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Guankui Du
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
- Department of Biochemistry and Molecular Biology, Hainan Medical University, Haikou, China
| |
Collapse
|
6
|
Qin L, Lv W. Dietary content and eating behavior in ulcerative colitis: a narrative review and future perspective. Nutr J 2025; 24:12. [PMID: 39849464 PMCID: PMC11755847 DOI: 10.1186/s12937-025-01075-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/06/2025] [Indexed: 01/25/2025] Open
Abstract
Ulcerative colitis (UC) has experienced a steady increase in global incidence and prevalence recently. Current research into UC pathogenesis focuses on the complex interplay of genetic and environmental factors with the immune system and gut microbiome, leading to disruption of the intestinal barrier. Normally, the microbiome, intestinal epithelium, and immune system interact to maintain intestinal homeostasis. However, when this equilibrium is disturbed, a harmful cycle of dysbiosis, immune dysregulation, and inflammation emerges, resulting in intestinal barrier dysfunction and UC progression. Among various risk factors, diet significantly influences epithelial barrier integrity and architectural stability through both direct and indirect mechanisms, shaping the entire UC continuum from pre-clinical prevention to active phase treatment and remission maintenance. This review provides insights into the impact of dietary content and eating behaviors on UC, focusing on specific food, food groups, nutrients, and intermittent fasting, while providing a detailed explanation of why the gut microbiota may mediate the sustained effects of diet across all stages of UC. Additionally, it addresses the limitations of current studies, explores underexamined areas in UC dietary research and proposes potential directions for future research and expansion.
Collapse
Affiliation(s)
- Lingxi Qin
- Clinical College, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Wenliang Lv
- Clinical College, Hubei University of Chinese Medicine, Wuhan, Hubei, China.
| |
Collapse
|
7
|
Valitutti F, Mennini M, Monacelli G, Fagiolari G, Piccirillo M, Di Nardo G, Di Cara G. Intestinal permeability, food antigens and the microbiome: a multifaceted perspective. FRONTIERS IN ALLERGY 2025; 5:1505834. [PMID: 39850945 PMCID: PMC11754301 DOI: 10.3389/falgy.2024.1505834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
The gut barrier encompasses several interactive, physical, and functional components, such as the gut microbiota, the mucus layer, the epithelial layer and the gut mucosal immunity. All these contribute to homeostasis in a well-regulated manner. Nevertheless, this frail balance might be disrupted for instance by westernized dietary habits, infections, pollution or exposure to antibiotics, thus diminishing protective immunity and leading to the onset of chronic diseases. Several gaps of knowledge still exist as regards this multi-level interaction. In this review we aim to summarize current evidence linking food antigens, microbiota and gut permeability interference in diverse disease conditions such as celiac disease (CeD), non-celiac wheat sensitivity (NCWS), food allergies (FA), eosinophilic gastrointestinal disorder (EOGID) and irritable bowel syndrome (IBS). Specific food elimination diets are recommended for CeD, NCWS, FA and in some cases for EOGID. Undoubtfully, each of these conditions is very different and quite unique, albeit food antigens/compounds, intestinal permeability and specific microbiota signatures orchestrate immune response and decide clinical outcomes for all of them.
Collapse
Affiliation(s)
- Francesco Valitutti
- Department of Medicine and Surgery, Pediatric Unit, University of Perugia, Perugia, Italy
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| | - Maurizio Mennini
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Gianluca Monacelli
- Department of Medicine and Surgery, Pediatric Unit, University of Perugia, Perugia, Italy
| | - Giulia Fagiolari
- Department of Medicine and Surgery, Pediatric Unit, University of Perugia, Perugia, Italy
| | - Marisa Piccirillo
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Giovanni Di Nardo
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Giuseppe Di Cara
- Department of Medicine and Surgery, Pediatric Unit, University of Perugia, Perugia, Italy
| |
Collapse
|
8
|
Wahnou H, Chgari O, Ndayambaje M, Hba S, Ouadghiri Z, Limami Y, Oudghiri M. Carrageenan and TLR4 Crosstalk: A Comprehensive Review of Inflammatory Responses in Animal Models. RECENT ADVANCES IN INFLAMMATION & ALLERGY DRUG DISCOVERY 2025; 19:5-17. [PMID: 40195700 DOI: 10.2174/0127722708303188240708071523] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/24/2024] [Accepted: 06/12/2024] [Indexed: 04/09/2025]
Abstract
Carrageenan, a naturally occurring polysaccharide derived from red seaweed, has been utilized extensively in the food industry as a stabilizer, thickener, and emulsifier due to its unique gel-forming properties. This versatile compound exists in various forms, including kappa, iota, and lambda, each with distinct characteristics suitable for different applications. Its widespread use as a food additive has raised concerns regarding its safety, particularly its potential inflammatory effects on the gastrointestinal tract. While carrageenan has been deemed safe for consumption by regulatory agencies in small amounts, studies have suggested its association with intestinal inflammation and gastrointestinal disturbances, particularly in susceptible individuals. Animal models, including rodents and non-human primates, have been employed to investigate the inflammatory response induced by carrageenan ingestion. These models have provided valuable insights into the molecular mechanisms underlying its pro-inflammatory properties. At the molecular level, carrageenan is believed to trigger inflammation by activating toll-like receptor 4 (TLR4) signaling pathways, leading to the production of pro-inflammatory cytokines and the recruitment of immune cells to the site of exposure. Furthermore, carrageenan-induced inflammation may disrupt the intestinal barrier function, facilitating the translocation of luminal antigens and exacerbating immune responses. This review provides a comprehensive examination of the current understanding of carrageenan's role in inflammation, encompassing its diverse applications in the food industry, safety concerns, experimental findings from animal models, and molecular mechanisms underlying its pro-inflammatory effects.
Collapse
Affiliation(s)
- Hicham Wahnou
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University of Casablanca, Casablanca, Morocco
| | - Oumaima Chgari
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University of Casablanca, Casablanca, Morocco
| | - Martin Ndayambaje
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University of Casablanca, Casablanca, Morocco
| | - Soufyane Hba
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University of Casablanca, Casablanca, Morocco
- University of Limoges, LABCiS, UR 22722, F-87000, Limoges, France
| | - Zaynab Ouadghiri
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University of Casablanca, Casablanca, Morocco
| | - Youness Limami
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University of Casablanca, Casablanca, Morocco
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat, Morocco
| | - Mounia Oudghiri
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University of Casablanca, Casablanca, Morocco
| |
Collapse
|
9
|
Sandys O, Stokkers PCF, Te Velde AA. DAMP-ing IBD: Extinguish the Fire and Prevent Smoldering. Dig Dis Sci 2025; 70:49-73. [PMID: 38963463 PMCID: PMC11761125 DOI: 10.1007/s10620-024-08523-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024]
Abstract
In inflammatory bowel diseases (IBD), the most promising therapies targeting cytokines or immune cell trafficking demonstrate around 40% efficacy. As IBD is a multifactorial inflammation of the intestinal tract, a single-target approach is unlikely to solve this problem, necessitating an alternative strategy that addresses its variability. One approach often overlooked by the pharmaceutically driven therapeutic options is to address the impact of environmental factors. This is somewhat surprising considering that IBD is increasingly viewed as a condition heavily influenced by such factors, including diet, stress, and environmental pollution-often referred to as the "Western lifestyle". In IBD, intestinal responses result from a complex interplay among the genetic background of the patient, molecules, cells, and the local inflammatory microenvironment where danger- and microbe-associated molecular patterns (D/MAMPs) provide an adjuvant-rich environment. Through activating DAMP receptors, this array of pro-inflammatory factors can stimulate, for example, the NLRP3 inflammasome-a major amplifier of the inflammatory response in IBD, and various immune cells via non-specific bystander activation of myeloid cells (e.g., macrophages) and lymphocytes (e.g., tissue-resident memory T cells). Current single-target biological treatment approaches can dampen the immune response, but without reducing exposure to environmental factors of IBD, e.g., by changing diet (reducing ultra-processed foods), the adjuvant-rich landscape is never resolved and continues to drive intestinal mucosal dysregulation. Thus, such treatment approaches are not enough to put out the inflammatory fire. The resultant smoldering, low-grade inflammation diminishes physiological resilience of the intestinal (micro)environment, perpetuating the state of chronic disease. Therefore, our hypothesis posits that successful interventions for IBD must address the complexity of the disease by simultaneously targeting all modifiable aspects: innate immunity cytokines and microbiota, adaptive immunity cells and cytokines, and factors that relate to the (micro)environment. Thus the disease can be comprehensively treated across the nano-, meso-, and microscales, rather than with a focus on single targets. A broader perspective on IBD treatment that also includes options to adapt the DAMPing (micro)environment is warranted.
Collapse
Affiliation(s)
- Oliver Sandys
- Tytgat Institute for Liver and Intestinal Research, AmsterdamUMC, AGEM, University of Amsterdam, Amsterdam, The Netherlands
| | - Pieter C F Stokkers
- Department of Gastroenterology and Hepatology, OLVG West, Amsterdam, The Netherlands
| | - Anje A Te Velde
- Tytgat Institute for Liver and Intestinal Research, AmsterdamUMC, AGEM, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
10
|
Mei L, Wang J, Hao Y, Zeng X, Yang Y, Wu Z, Ji Y. A comprehensive update on the immunoregulatory mechanisms of Akkermansia muciniphila: insights into active ingredients, metabolites, and nutrient-driven modulation. Crit Rev Food Sci Nutr 2024:1-18. [PMID: 39413040 DOI: 10.1080/10408398.2024.2416481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Akkermansia muciniphila (A. muciniphila) has gained recognition as a pioneering probiotic, exhibiting considerable potential to enhance immune conditions across both humans and animals. The health benefits of A. muciniphila are attributed to its various components, including outer membrane proteins (PilQ and Amuc_1100), secreted proteins (P9 and AmTARS), extracellular vesicles, and metabolites such as SCFAs, ornithine lipids, γ-aminobutyric acid, cobalamin, and inosine. The dynamic control of the mucus layer by A. muciniphila plays a crucial role in regulating intestinal mucosal immunity. Furthermore, A. muciniphila modulates immune function by interacting with macrophages, dendritic cells, T lymphocytes, and Paneth cells. Increasing the abundance of A. muciniphila in the gut through nutritional strategies represents a safe and effective means to augment immune function. Various polyphenols, oligosaccharides, and polysaccharides have been shown to elevate the levels of this bacterium, thereby contributing to favorable immunoregulatory outcomes. This paper delves into the latest research advancements related to the probiotic mechanisms of A. muciniphila and provides an overview of the current understanding of how its abundance responds to nutrients. These insights offer a theoretical foundation for the utilization of A. muciniphila in immunoregulation.
Collapse
Affiliation(s)
- Lihua Mei
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Jiaxin Wang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Yanling Hao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Yun Ji
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| |
Collapse
|
11
|
Rampin A, Rossoni A, Chaniotaki L, Gkiatas IS, Tzora A, Skoufos I, Diakakis N, Prassinos N, Zeugolis DI. Xenogeneic versus allogeneic serum and macromolecular crowding in human tenocyte cultures. Eur J Cell Biol 2024; 103:151445. [PMID: 39024989 DOI: 10.1016/j.ejcb.2024.151445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024] Open
Abstract
Allogeneic serum and tissue-specific extracellular matrix have been shown to maintain permanently differentiated cell phenotype in culture. This is of particular importance for human tenocytes, a cell population that readily loses its function during ex vivo culture. With these in mind, herein we extracted human tenocytes using either foetal bovine serum or human serum, cultured them in the absence and presence of carrageenan and Ficoll®, the most widely used macromolecular crowding agents (to induce tissue-specific extracellular matrix deposition), and assessed cellular function, via metabolic activity, viability, proliferation and immunofluorescence for collagen related molecules, non-collagenous molecules and transmembrane molecules. At day 7, longest time point assessed, neither carrageenan nor Ficoll® significantly affected metabolic activity, viability and proliferation in either serum and human serum significantly increased metabolic activity and proliferation. At day 7, in the absence of macromolecular crowding, cells in human serum deposited significantly lower collagen type VI, biglycan, versican and tenomodulin than cells in foetal bovine serum. Interestingly, at day 7, in comparison to the no macromolecular crowding group, carrageenan in foetal bovine serum induced the highest effect, as judged by the highest number of significantly increased molecules (collagen type I, collagen type IV, collagen type V, collagen type VI, transforming growth factor β1, matrix metalloproteinase 14, lumican, versican, scleraxis and integrin α2β1). These data, although contradict previous observations where human serum outperformed foetal bovine serum, at the same time, support the use of foetal bovine serum in the development of cell-based medicines.
Collapse
Affiliation(s)
- Andrea Rampin
- Department of Agriculture, University of Ioannina, Arta, Greece; School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece; Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Andrea Rossoni
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Lefki Chaniotaki
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Ioannis S Gkiatas
- Department of Orthopaedic Surgery, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Athina Tzora
- Department of Agriculture, University of Ioannina, Arta, Greece
| | - Ioannis Skoufos
- Department of Agriculture, University of Ioannina, Arta, Greece
| | - Nikolaos Diakakis
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikitas Prassinos
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland.
| |
Collapse
|
12
|
Katsoudas N, Tavakoli P, Wu N, Shapiro A, Leach ST, Williams AJ, Paramsothy R, Ghaly S, Connor SJ, Samocha-Bonet D, Lambert K, Hold GL. Dietary Emulsifier Exposure in People With Inflammatory Bowel Disease Compared With Healthy Controls: Is There a Cause for Concern? Inflamm Bowel Dis 2024; 30:1241-1250. [PMID: 38244236 DOI: 10.1093/ibd/izad318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Emulsifiers are implicated in the pathogenesis of inflammatory bowel disease (IBD). Few studies have examined emulsifier intake in people with existing IBD. We aimed to describe the frequency of exposure to 6 selected emulsifiers in a contemporary cohort of people with IBD and compare intake with healthy controls (HCs). METHODS Baseline food records from participants in an Australian prospective cohort study examining the microbiome of IBD patients and HCs were analyzed. Exposure to inflammatory emulsifiers polysorbate-80 (P80); carboxymethylcellulose (CMC); carrageenan; xanthan gum (XG); lecithin (soy and sunflower) and mono- and diglycerides of fatty acids (MDGs) were determined by examining ingredient lists. Frequency of emulsifier exposure between groups (IBD vs HC, Crohn's disease [CD] vs ulcerative colitis [UC], IBD children vs adults, active disease vs remission) was examined after controlling for confounders. RESULTS Records from 367 participants were analyzed (n = 176 IBD, of which there were 101 CD, 75 UC, and 191 HC patients). In total, 5022 unique food items were examined, with 18% containing 1 or more emulsifier of interest. Inflammatory bowel disease participants had significantly higher total daily emulsifier exposure compared with HCs (2.7 ± 1.8 vs 2.3 ± 1.6, P = .02). In IBD participants, emulsifiers with the highest daily exposure were MDGs (1.2 ± 0.93), lecithin (0.85 ± 0.93), and XG (0.38 ± 0.42). There were no recorded exposures to P80. CONCLUSIONS Inflammatory bowel disease participants were exposed to more emulsifiers than HCs. Intake of inflammatory emulsifiers were low or nonexistent, suggesting their presence in the food supply are not as common as frequently stated.
Collapse
Affiliation(s)
- Natasha Katsoudas
- School of Medical, Indigenous and Health Science, University of Wollongong, New South Wales, Australia
| | - Paris Tavakoli
- Microbiome Research Centre, University of New South Wales, Kogarah, New South Wales, Australia
- St Vincent's Hospital, Department of Gastroenterology and Hepatology, Sydney, New South Wales, Australia
| | - Nan Wu
- Microbiome Research Centre, University of New South Wales, Kogarah, New South Wales, Australia
| | - Amanda Shapiro
- Department of Paediatrics, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Steven T Leach
- Department of Paediatrics, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Astrid-Jane Williams
- Liverpool Hospital Dept of Gastroenterology and Hepatology, Liverpool, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South West Sydney Clinical Campuses, University of New South Wales Medicine & Health, University of New South Wales, Sydney, Australia
| | - Ramesh Paramsothy
- Centre for Gastrointestinal Health Castle Hill, New South Wales, Australia
| | - Simon Ghaly
- St Vincent's Hospital, Department of Gastroenterology and Hepatology, Sydney, New South Wales, Australia
| | - Susan J Connor
- Liverpool Hospital Dept of Gastroenterology and Hepatology, Liverpool, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South West Sydney Clinical Campuses, University of New South Wales Medicine & Health, University of New South Wales, Sydney, Australia
| | - Dorit Samocha-Bonet
- Garvan Institute of Medical Research and School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, University of New South Wales Medicine & Health, University of New South Wales, Sydney, Australia
| | - Kelly Lambert
- School of Medical, Indigenous and Health Science, University of Wollongong, New South Wales, Australia
| | - Georgina L Hold
- Microbiome Research Centre, University of New South Wales, Kogarah, New South Wales, Australia
| |
Collapse
|
13
|
Su Y, Cai X, Fan X, Ning J, Shen M. Effect of Trace Element Selenium on the Intestinal Microbial Community in Nude Mice with Colorectal Cancer. Microorganisms 2024; 12:1336. [PMID: 39065104 PMCID: PMC11279152 DOI: 10.3390/microorganisms12071336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/31/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. The role of intestinal microbiota in carcinogenesis has also become an important research topic, and CRC is closely related to the intestinal microbiota. Selenium-containing compounds have attracted more attention as anticancer drugs as they can have minimal side effects. The purpose of this study was to determine and compare the effect of sodium selenite and selenomethionine on the microbial communities of nude mice with CRC. A CRC ectopic tumorigenesis model was established by subcutaneously injecting HCT116 cells into nude mice. The mice were then intraperitoneally injected with sodium selenite and selenomethionine for 24 days to regulate their intestinal microbiota. Compared with sodium selenite, selenomethionine resulted in a greater reduction in the richness and diversity of intestinal microbiota in nude mice with CRC, and the richness and diversity were closer to healthy levels. Selenomethionine also regulated a wider variety of flora. Additionally, sodium selenite and selenomethionine produced different microorganisms, changed function and metabolic pathways in the intestinal microbiota. Both sodium selenite and selenomethionine have certain effects on restoring the intestinal microbial diversity in nude mice with CRC, and the effect of selenomethionine is better than that of sodium selenite.
Collapse
Affiliation(s)
| | | | | | | | - Mei Shen
- Department of Hygiene Inspection & Quarantine Science, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (Y.S.); (X.C.); (X.F.); (J.N.)
| |
Collapse
|
14
|
Ayuso P, Quizhpe J, Yepes F, Miranzo D, Avellaneda A, Nieto G, Ros G. Improving the Nutritional Quality of Protein and Microbiota Effects in Additive- and Allergen-Free Cooked Meat Products. Foods 2024; 13:1792. [PMID: 38928734 PMCID: PMC11202710 DOI: 10.3390/foods13121792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/19/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
The primary objective of the meat industry is to enhance the quality and positive attributes of meat products, driven by an increasing consumer demand for healthier, less processed options. One common approach to achieving this goal is the replacement of additives and allergens with natural ingredients. Nevertheless, the nutritional impact of these changes has not been extensively studied. To address these gaps, two new meat products were developed: cooked turkey breast and cooked ham. The products in question exclude additives and allergens and instead incorporate a blend of natural extracts containing vitamin C, chlorogenic acids, hydroxytyrosol, catechins, epicatechins, vinegar, and inulin fibre. The objective of this study was to evaluate the impact of these reformulations on protein quality and gut microbiota. Protein quality was evaluated using the Digestible Indispensable Amino Acid Score (DIAAS) following in vitro digestion. The microbial composition and short-chain fatty acid (SCFA) production were analysed through in vitro colonic fermentations in both normal-weight and obese participants in order to gauge their effect on gut microbiota. The results demonstrated that the reformulation of cooked turkey breast increased its digestibility by 6.4%, while that of cooked ham exhibited a significant 17.9% improvement. Furthermore, protein quality was found to have improved significantly, by 19.5% for cooked turkey breast and 32.9% for cooked ham. Notwithstanding these alterations in protein digestibility, the microbial composition at the phylum and genus levels remained largely unaltered. Nevertheless, total SCFA production was observed to increase in both groups, with a more pronounced effect observed in the normal-weight group. In conclusion, the substitution of artificial additives with natural ingredients in reformulated cooked meat products has resulted in enhanced digestibility, improved protein quality, and increased production of short-chain fatty acids.
Collapse
Affiliation(s)
- Pablo Ayuso
- Department of Food Technology, Nutrition and Food Science, Veterinary Faculty, University of Murcia, Regional Campus of International Excellence “Campus Mare Nostrum”, Espinardo, 30100 Murcia, Spain; (P.A.); (J.Q.); (G.N.)
- Cátedra de Seguridad y Sostenibilidad Alimentaria Grupo Fuertes, Universidad de Murcia, 30003 Murcia, Spain (A.A.)
| | - Jhazmin Quizhpe
- Department of Food Technology, Nutrition and Food Science, Veterinary Faculty, University of Murcia, Regional Campus of International Excellence “Campus Mare Nostrum”, Espinardo, 30100 Murcia, Spain; (P.A.); (J.Q.); (G.N.)
- Cátedra de Seguridad y Sostenibilidad Alimentaria Grupo Fuertes, Universidad de Murcia, 30003 Murcia, Spain (A.A.)
| | - Fani Yepes
- Cátedra de Seguridad y Sostenibilidad Alimentaria Grupo Fuertes, Universidad de Murcia, 30003 Murcia, Spain (A.A.)
| | - Domingo Miranzo
- Cátedra de Seguridad y Sostenibilidad Alimentaria Grupo Fuertes, Universidad de Murcia, 30003 Murcia, Spain (A.A.)
| | - Antonio Avellaneda
- Cátedra de Seguridad y Sostenibilidad Alimentaria Grupo Fuertes, Universidad de Murcia, 30003 Murcia, Spain (A.A.)
| | - Gema Nieto
- Department of Food Technology, Nutrition and Food Science, Veterinary Faculty, University of Murcia, Regional Campus of International Excellence “Campus Mare Nostrum”, Espinardo, 30100 Murcia, Spain; (P.A.); (J.Q.); (G.N.)
| | - Gaspar Ros
- Department of Food Technology, Nutrition and Food Science, Veterinary Faculty, University of Murcia, Regional Campus of International Excellence “Campus Mare Nostrum”, Espinardo, 30100 Murcia, Spain; (P.A.); (J.Q.); (G.N.)
- Cátedra de Seguridad y Sostenibilidad Alimentaria Grupo Fuertes, Universidad de Murcia, 30003 Murcia, Spain (A.A.)
| |
Collapse
|
15
|
Kimilu N, Gładyś-Cieszyńska K, Pieszko M, Mańkowska-Wierzbicka D, Folwarski M. Carrageenan in the Diet: Friend or Foe for Inflammatory Bowel Disease? Nutrients 2024; 16:1780. [PMID: 38892712 PMCID: PMC11174395 DOI: 10.3390/nu16111780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
While the exact pathogenesis of IBD remains unclear, genetic, environmental and nutritional factors as well as the composition of the gut microbiome play crucial roles. Food additives, which are increasingly consumed in the Western diet, are being investigated for their potential effects on IBD. These additives can affect gut health by altering the composition of the microbiota, immune responses, and intestinal permeability, contributing to autoimmune diseases and inflammation. Despite the growing number of studies on food additives and IBD, the specific effects of carrageenan have not yet been sufficiently researched. This review addresses this gap by critically analyzing recent studies on the effects of carrageenan on the gut microbiota, intestinal permeability, and inflammatory processes. We searched the MEDLINE and SCOPUS databases using the following terms: carrageenan, carrageenan and inflammatory bowel disease, carrageenan and cancer, food additives and microbiome, food additives and intestinal permeability, and food additives and autoimmune diseases. In animal studies, degraded carrageenan has been shown to trigger intestinal ulceration and inflammation, highlighting its potential risk for exacerbating IBD. It can affect the gut microbiota, reduce bacterial diversity, and increase intestinal permeability, contributing to "leaky gut" syndrome. Some studies suggest that carrageenan may inhibit the growth of cancer cells by influencing the progression of the cell cycle, but the anti-cancer effect is still unclear. Carrageenan may also increase glucose intolerance and insulin resistance. Further research is needed to determine whether carrageenan should be excluded from the diet of individuals with IBD.
Collapse
Affiliation(s)
- Nina Kimilu
- Students’ Scientific Circle of Clinical Nutrition, Medical University of Gdansk, 80-211 Gdansk, Poland
| | | | - Magdalena Pieszko
- Department of Clinical Nutrition and Dietetics, Medical University of Gdansk, 80-210 Gdansk, Poland (M.P.)
| | - Dorota Mańkowska-Wierzbicka
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Marcin Folwarski
- Department of Clinical Nutrition and Dietetics, Medical University of Gdansk, 80-210 Gdansk, Poland (M.P.)
- Home Enteral and Parenteral Nutrition Unit, Nicolaus Copernicus Hospital, 80-803 Gdansk, Poland
| |
Collapse
|
16
|
Komisarska P, Pinyosinwat A, Saleem M, Szczuko M. Carrageenan as a Potential Factor of Inflammatory Bowel Diseases. Nutrients 2024; 16:1367. [PMID: 38732613 PMCID: PMC11085445 DOI: 10.3390/nu16091367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Carrageenan is a widely used food additive and is seen as a potential candidate in the pharmaceutical industry. However, there are two faces to carrageenan that allows it to be used positively for therapeutic purposes. Carrageenan can be used to create edible films and for encapsulating drugs, and there is also interest in the use of carrageenan for food printing. Carrageenan is a naturally occurring polysaccharide gum. Depending on the type of carrageenan, it is used in regulating the composition of intestinal microflora, including the increase in the population of Bifidobacterium bacteria. On the other hand, the studies have demonstrated the harmfulness of carrageenan in animal and human models, indicating a direct link between diet and intestinal inflammatory states. Carrageenan changes the intestinal microflora, especially Akkermansia muciniphilia, degrades the mucous barrier and breaks down the mucous barrier, causing an inflammatory reaction. It directly affects epithelial cells by activating the pro-inflammatory nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) pathway. The mechanism is based on activation of the TLR4 receptor, alterations in macrophage activity, production of proinflammatory cytokines and activation of innate immune pathways. Carrageenan increases the content of Bacteroidetes bacteria, also causing a reduction in the number of short chain fatty acid (SCFA)-producing bacteria. The result is damage to the integrity of the intestinal membrane and reduction of the mucin layer. The group most exposed to the harmful effects of carrageenan are people suffering from intestinal inflammation, including Crohn disease (CD) and ulcerative colitis (UC).
Collapse
Affiliation(s)
| | | | | | - Małgorzata Szczuko
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland (M.S.)
| |
Collapse
|
17
|
Limketkai BN, Rau S, Fasulo C. Preventative and therapeutic potential of nutrition for inflammatory bowel diseases: A narrative review. JPEN J Parenter Enteral Nutr 2024; 48:258-266. [PMID: 38357793 DOI: 10.1002/jpen.2606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/10/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
Diet strongly shapes the gut microbiome and metabolome, which in turn influence intestinal inflammation in patients with inflammatory bowel disease (IBD). Separate from inflammation and malnutrition, diet's direct interactions with the gastrointestinal system can also provoke or attenuate a host of nonspecific gastrointestinal symptoms. Given these multifaceted effects of diet on inflammation and symptoms, nutrition has been investigated for its potential roles in the prevention and treatment of IBD. This review presents epidemiological, observational cohort, and clinical trial evidence that underlie our current understanding of nutrition for prevention and treatment of IBD.
Collapse
Affiliation(s)
- Berkeley N Limketkai
- Vatche & Tamar Manoukian Division of Digestive Diseases, UCLA School of Medicine, Los Angeles, California, USA
| | - Sameeha Rau
- Vatche & Tamar Manoukian Division of Digestive Diseases, UCLA School of Medicine, Los Angeles, California, USA
| | - Christina Fasulo
- Vatche & Tamar Manoukian Division of Digestive Diseases, UCLA School of Medicine, Los Angeles, California, USA
| |
Collapse
|
18
|
Guillaumin S, Gurdal M, Zeugolis DI. Gums as Macromolecular Crowding Agents in Human Skin Fibroblast Cultures. Life (Basel) 2024; 14:435. [PMID: 38672707 PMCID: PMC11051389 DOI: 10.3390/life14040435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/10/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Even though tissue-engineered medicines are under intense academic, clinical, and commercial investigation, only a handful of products have been commercialised, primarily due to the costs associated with their prolonged manufacturing. While macromolecular crowding has been shown to enhance and accelerate extracellular matrix deposition in eukaryotic cell culture, possibly offering a solution in this procrastinating tissue-engineered medicine development, there is still no widely accepted macromolecular crowding agent. With these in mind, we herein assessed the potential of gum Arabic, gum gellan, gum karaya, and gum xanthan as macromolecular crowding agents in WS1 skin fibroblast cultures (no macromolecular crowding and carrageenan were used as a control). Dynamic light scattering analysis revealed that all macromolecules had negative charge and were polydispersed. None of the macromolecules affected basic cellular function. At day 7 (the longest time point assessed), gel electrophoresis analysis revealed that all macromolecules significantly increased collagen type I deposition in comparison to the non-macromolecular crowding group. Also at day 7, immunofluorescence analysis revealed that carrageenan; the 50 µg/mL, 75 µg/mL, and 100 µg/mL gum gellan; and the 500 µg/mL and 1000 µg/mL gum xanthan significantly increased both collagen type I and collagen type III deposition and only carrageenan significantly increased collagen type V deposition, all in comparison to the non-macromolecular crowding group at the respective time point. This preliminary study demonstrates the potential of gums as macromolecular crowding agents, but more detailed biological studies are needed to fully exploit their potential in the development of tissue-engineered medicines.
Collapse
Affiliation(s)
- Salome Guillaumin
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, H91 TK33 Galway, Ireland; (S.G.); (M.G.)
| | - Mehmet Gurdal
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, H91 TK33 Galway, Ireland; (S.G.); (M.G.)
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), D04 V1W8 Dublin, Ireland
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, H91 TK33 Galway, Ireland; (S.G.); (M.G.)
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), D04 V1W8 Dublin, Ireland
| |
Collapse
|
19
|
Bekar C, Ozmen O, Ozkul C, Ayaz A. Inulin protects against the harmful effects of dietary emulsifiers on mice gut microbiome. PeerJ 2024; 12:e17110. [PMID: 38525281 PMCID: PMC10961058 DOI: 10.7717/peerj.17110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/23/2024] [Indexed: 03/26/2024] Open
Abstract
Background The prevalence of inflammatory bowel diseases is increasing, especially in developing countries, with adoption of Western-style diet. This study aimed to investigate the effects of two emulsifiers including lecithin and carboxymethyl cellulose (CMC) on the gut microbiota, intestinal inflammation and the potential of inulin as a means to protect against the harmful effects of emulsifiers. Methods In this study, male C57Bl/6 mice were divided into five groups (n:6/group) (control, CMC, lecithin, CMC+inulin, and lecithin+inulin). Lecithin and CMC were diluted in drinking water (1% w/v) and inulin was administered daily at 5 g/kg for 12 weeks. Histological examination of the ileum and colon, serum IL-10, IL-6, and fecal lipocalin-2 levels were analyzed. 16S rRNA gene V3-V4 region amplicon sequencing was performed on stool samples. Results In the CMC and lecithin groups, shortening of the villus and a decrease in goblet cells were observed in the ileum and colon, whereas inulin reversed this effect. The lipocalin level, which was 9.7 ± 3.29 ng in the CMC group, decreased to 4.1 ± 2.98 ng with the administration of inulin. Bifidobacteria and Akkermansia were lower in the CMC group than the control, while they were higher in the CMC+inulin group. In conclusion, emulsifiers affect intestinal health negatively by disrupting the epithelial integrity and altering the composition of the microbiota. Inulin is protective on their harmful effects. In addition, it was found that CMC was more detrimental to microbiota composition than lecithin.
Collapse
Affiliation(s)
- Cansu Bekar
- Department of Nutrition and Dietetics, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Ozlem Ozmen
- Department of Pathology, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Ceren Ozkul
- Department of Pharmaceutical Microbiology, Hacettepe University, Ankara, Turkey
| | - Aylin Ayaz
- Department of Nutrition and Dietetics, Hacettepe University, Ankara, Turkey
| |
Collapse
|
20
|
Qin X, Nong K, Liu Z, Fang X, Zhang B, Chen W, Wang Z, Wu Y, Shi H, Wang X, Zhang H. Regulation of the intestinal flora using polysaccharides from Callicarpa nudiflora Hook to alleviate ulcerative colitis and the molecular mechanisms involved. Int J Biol Macromol 2024; 258:128887. [PMID: 38118262 DOI: 10.1016/j.ijbiomac.2023.128887] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/04/2023] [Accepted: 12/17/2023] [Indexed: 12/22/2023]
Abstract
Ulcerative colitis (UC) is a type of inflammatory bowel disease (IBD) that cannot be completely cured by current treatments. C. nudiflora Hook has antibacterial, anti-inflammatory, and hemostatic biological functions; however, the therapeutic role of C. nudiflora Hook or its extracts in IBD remains poorly understood. In this study, we extracted and purified three fractions of C. nudiflora Hook polysaccharides by hydroalcohol precipitation method, which were named as CNLP-1, CNLP-2 and CNLP-3, respectively. CNLP-2, the main component of the polysaccharides of C. nudiflora Hook is an pyranose type acidic polysaccharide composed of Fuc, Rha, Ara, Gal, Glc, Xyl, Man, Gal-UA and Glc-UA, with an Mn of 15.624 kDa; Mw of 31.375 kDa. CNLP-2 was found to have a smooth lamellar structure as observed by scanning electron microscopy. To investigate the effect of CNLP-2 (abbreviated to CNLP) on dextran sodium sulfate (DSS)-induced UC mice and its mechanism of action, we treated DSS-induced UC mice by administering CNLP at a dose of 100 mg/kg every other day. The results of the study showed that CNLP alleviated the clinical symptoms such as body weight (BW) loss, pathological damage, and systemic inflammation. The mechanism may be through the regulation of intestinal flora and its metabolism, which in turn affects the expression of NF-κB/MAPK pathway-related proteins through the metabolites of intestinal flora to further alleviate inflammation and ultimately improve the intestinal barrier function in UC mice. In conclusion, CNLP has great potential for the treatment of IBD.
Collapse
Affiliation(s)
- Xinyun Qin
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Keyi Nong
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Zhineng Liu
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Xin Fang
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Bin Zhang
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Wanyan Chen
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Zihan Wang
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Yijia Wu
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Huiyu Shi
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Xuemei Wang
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Haiwen Zhang
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China.
| |
Collapse
|
21
|
Grondin JA, Khan WI. Emerging Roles of Gut Serotonin in Regulation of Immune Response, Microbiota Composition and Intestinal Inflammation. J Can Assoc Gastroenterol 2024; 7:88-96. [PMID: 38314177 PMCID: PMC10836984 DOI: 10.1093/jcag/gwad020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2024] Open
Abstract
Although the exact etiology of inflammatory bowel diseases (IBD) is unknown, studies have shown that dysregulated immune responses, genetic factors, gut microbiota, and environmental factors contribute to their pathogenesis. Intriguingly, serotonin (5-hydroxytryptamine or 5-HT) seems to be a molecule with increasingly strong implications in the pathogenesis of intestinal inflammation, affecting host physiology, including autophagy and immune responses, as well as microbial composition and function. 5-HT may also play a role in mediating how environmental effects impact outcomes in IBD. In this review, we aim to explore the production and important functions of 5-HT, including its impact on the gut. In addition, we highlight the bidirectional impacts of 5-HT on the immune system, the gut microbiota, and the process of autophagy and how these effects contribute to the manifestation of intestinal inflammation. We also explore recent findings connecting 5-HT signalling and the influence of environmental factors, particularly diet, in the pathogenesis of IBD. Ultimately, we explore the pleiotropic effects of this ancient molecule on biology and health in the context of intestinal inflammation.
Collapse
Affiliation(s)
- Jensine A Grondin
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Waliul I Khan
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
22
|
Tahiri M, Johnsrud C, Steffensen IL. Evidence and hypotheses on adverse effects of the food additives carrageenan (E 407)/processed Eucheuma seaweed (E 407a) and carboxymethylcellulose (E 466) on the intestines: a scoping review. Crit Rev Toxicol 2023; 53:521-571. [PMID: 38032203 DOI: 10.1080/10408444.2023.2270574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023]
Abstract
This scoping review provides an overview of publications reporting adverse effects on the intestines of the food additives carrageenan (CGN) (E 407)/processed Eucheuma seaweed (PES) (E 407a) and carboxymethylcellulose (CMC) (E 466). It includes evidence from human, experimental mammal and in vitro research publications, and other evidence. The databases Medline, Embase, Scopus, Web of Science Core Collection, Cochrane Database of Systematic Reviews and Epistemonikos were searched without time limits, in addition to grey literature. The publications retrieved were screened against predefined criteria. From two literature searches, 2572 records were screened, of which 224 records were included, as well as 38 records from grey literature, making a total of 262 included publications, 196 on CGN and 101 on CMC. These publications were coded and analyzed in Eppi-Reviewer and data gaps presented in interactive maps. For CGN, five, 69 and 33 research publications on humans, experimental mammals and in vitro experiments were found, further separated as degraded or native (non-degraded) CGN. For CMC, three human, 20 animal and 14 in vitro research publications were obtained. The most studied adverse effects on the intestines were for both additives inflammation, the gut microbiome, including fermentation, intestinal permeability, and cancer and metabolic effects, and immune effects for CGN. Further studies should focus on native CGN, in the form and molecular weight used as food additive. For both additives, randomized controlled trials of sufficient power and with realistic dietary exposure levels of single additives, performed in persons of all ages, including potentially vulnerable groups, are needed.
Collapse
Affiliation(s)
- Mirlinda Tahiri
- Department of Food Safety, Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Celine Johnsrud
- Department of Food Safety, Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Inger-Lise Steffensen
- Department of Food Safety, Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
23
|
Yokomori R, Shirai T, Tsukiji N, Oishi S, Sasaki T, Takano K, Suzuki-Inoue K. C-type lectin-like receptor-2 (CLEC-2) is a key regulator of kappa-carrageenan-induced tail thrombosis model in mice. Platelets 2023; 34:2281941. [PMID: 38010137 DOI: 10.1080/09537104.2023.2281941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/03/2023] [Indexed: 11/29/2023]
Abstract
Kappa-carrageenan (KCG), which is used to induce thrombosis in laboratory animals for antithrombotic drug screening, can trigger platelet aggregation. However, the cell-surface receptor and related signaling pathways remain unclear. In this study, we investigated the molecular basis of KCG-induced platelet activation using light-transmittance aggregometry, flow cytometry, western blotting, and surface plasmon resonance assays using platelets from platelet receptor-deficient mice and recombinant proteins. KCG-induced tail thrombosis was also evaluated in mice lacking the platelet receptor. We found that KCG induces platelet aggregation with α-granule secretion, activated integrin αIIbβ3, and phosphatidylserine exposure. As this aggregation was significantly inhibited by the Src family kinase inhibitor and spleen tyrosine kinase (Syk) inhibitor, a tyrosine kinase-dependent pathway is required. Platelets exposed to KCG exhibited intracellular tyrosine phosphorylation of Syk, linker activated T cells, and phospholipase C gamma 2. KCG-induced platelet aggregation was abolished in platelets from C-type lectin-like receptor-2 (CLEC-2)-deficient mice, but not in platelets pre-treated with glycoprotein VI-blocking antibody, JAQ1. Surface plasmon resonance assays showed a direct association between murine/human recombinant CLEC-2 and KCG. KCG-induced thrombosis and thrombocytopenia were significantly inhibited in CLEC-2-deficient mice. Our findings show that KCG induces platelet activation via CLEC-2.
Collapse
Affiliation(s)
- Ryohei Yokomori
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Toshiaki Shirai
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Nagaharu Tsukiji
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Saori Oishi
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Tomoyuki Sasaki
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Katsuhiro Takano
- Department of Transfusion and Cell Therapy, University of Yamanashi Hospital, Chuo, Japan
| | - Katsue Suzuki-Inoue
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
- Department of Transfusion and Cell Therapy, University of Yamanashi Hospital, Chuo, Japan
| |
Collapse
|
24
|
He P, Zhang Y, Chen R, Tong Z, Zhang M, Wu H. The maca protein ameliorates DSS-induced colitis in mice by modulating the gut microbiota and production of SCFAs. Food Funct 2023; 14:10329-10346. [PMID: 37955225 DOI: 10.1039/d3fo03654e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Maca is a functional food with anti-inflammatory activity, and it is rich in protein. Currently, inflammatory bowel disease (IBD) is a common gastrointestinal disease. However, there is little research focusing on the effect of maca protein (MCP) on IBD. In this study, we extracted MCP from maca root and explored its effect and mechanism on improving dextran sodium sulfate (DSS)-induced IBD in mice. The results indicated that MCP intervention alleviated the clinical symptoms and colon tissue damage of mice with DSS-induced colitis and inhibited the expression of inflammatory factors. Moreover, it can modulate the gut microbiota composition in mice with DSS-induced colitis. The regulation is achieved by reducing the relative abundance of the IBD-exacerbating key bacterial genera: Lachnospiraceae_NK4A136_group, Bacteroides, Desulfovibrio, Prevotella, Helicobacter and Sutterella, while increasing the relative abundance of the IBD-alleviating key bacterial genera: norank_f_Muribaculaceae, Lactobacillus, Oscillospira, Akkermansia and Bifidobacterium. MCP can also promote the production of short-chain fatty acids (SCFAs). The further western blotting results indicated that MCP can regulate the Treg/Th17 immune balance in mice with colitis via the SCFAs-GPR41/43/HDAC1 signaling pathway. Overall, MCP can alleviate colitis by comprehensively regulating the gut microbiota and inflammatory response. It may be a promising functional component that reduces the risk of colitis by maintaining intestinal health.
Collapse
Affiliation(s)
- Ping He
- College of Food Sciences and Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Yizhe Zhang
- College of Food Sciences and Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Ruyang Chen
- College of Food Sciences and Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Zhaoli Tong
- College of Food Sciences and Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Mengmeng Zhang
- College of Chemistry and Chemical Engineering, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China.
| | - Hui Wu
- College of Food Sciences and Engineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
25
|
Lee ZJ, Xie C, Ng K, Suleria HAR. Unraveling the bioactive interplay: seaweed polysaccharide, polyphenol and their gut modulation effect. Crit Rev Food Sci Nutr 2023; 65:382-405. [PMID: 37991467 DOI: 10.1080/10408398.2023.2274453] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Seaweed is rich in many unique bioactive compounds such as polyphenols and sulfated polysaccharides that are not found in terrestrial plant. The discovery of numerous biological activities from seaweed has made seaweed an attractive functional food source with the potential to be exploited for human health benefits. During food processing and digestion, cell wall polysaccharide and polyphenols commonly interact, and this may influence the nutritional properties of food. Interactions between cell wall polysaccharide and polyphenols in plant-based system has been extensively studied. However, similar interactions in seaweed have received little attention despite the vast disparity between the structural and chemical composition of plant and seaweed cell wall. This poses a challenge in extracting seaweed bioactive compounds with intact biological properties. This review aims to summarize the cell wall polysaccharide and polyphenols present in brown, red and green seaweed, and current knowledge on their potential interactions. Moreover, this review gives an overview of the gut modulation effect of seaweed polysaccharide and polyphenol.
Collapse
Affiliation(s)
- Zu Jia Lee
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, Australia
| | - Cundong Xie
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, Australia
| | - Ken Ng
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, Australia
| | - Hafiz A R Suleria
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, Australia
| |
Collapse
|
26
|
Zhang Y, Zhou M, Zhou Y, Guan X. Dietary components regulate chronic diseases through gut microbiota: a review. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:6752-6766. [PMID: 37225671 DOI: 10.1002/jsfa.12732] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/27/2023] [Accepted: 05/16/2023] [Indexed: 05/26/2023]
Abstract
In recent years, gut microbiota as an immune organ has gradually become the mainstream of research. When the composition of the gut microbiota is changed significantly, this may affect human health. This review details the major microbiota composition and metabolites in the gut and discusses chronic diseases based on gut dysbiosis, including obesity, liver injury, colon cancer, atherosclerosis, and central nervous system diseases. We comprehensively summarize the changes in abundance of relevant gut microbiota by ingesting different diet components (such as food additives, dietary polyphenols, polysaccharides, fats, proteins) and their influence on the microbial quorum sensing system, thereby regulating related diseases. We believe that quorum sensing can be used as a new entry point to explain the mechanism of ingesting dietary components to improve gut microbiota and thereby regulate related diseases. This review hopes to provide a theoretical basis for future research on improving disease symptoms by ingesting functional foods containing dietary components. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ying Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai, China
| | - Ming Zhou
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yaqin Zhou
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Xiao Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai, China
| |
Collapse
|
27
|
Trewin I, Kathrani A. Pre-illness dietary risk factors in dogs with chronic enteropathy. J Vet Intern Med 2023; 37:2093-2101. [PMID: 37743693 PMCID: PMC10658591 DOI: 10.1111/jvim.16872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 09/08/2023] [Indexed: 09/26/2023] Open
Abstract
BACKGROUND Dietary factors have been extensively studied as potential triggers of inflammatory bowel disease in humans. Scant literature exists regarding diet as a pre-illness risk factor in dogs with chronic enteropathy (CE). HYPOTHESIS To evaluate possible pre-illness dietary risk factors in dogs with CE. ANIMALS Ninety-five client-owned dogs; 48 with CE (25 presumptive and 23 confirmed) and 47 without a history of signs of gastrointestinal disease. METHODS Retrospective case-control questionnaire-based study at a veterinary referral teaching hospital in the United Kingdom. Diet history was obtained relating to the onset of initial presenting signs for all dogs. The main diet consumed underwent ingredient analysis and caloric distribution calculation using a guaranteed analysis convertor software. Length of time the main diet was fed and adherence to the World Small Animal Veterinary Association Global Nutrition Committee guidelines was also recorded. RESULTS The frequency of the main diet containing no carbohydrate was greater for controls (5/47 dogs, 11%) vs the combined presumptive and confirmed CE dogs (0/48 dogs, 0%; P = .05). Fewer dogs with confirmed CE were fed a main diet containing red meat as the primary protein source (2/23 dogs, 9%) vs controls (15/47 dogs, 32%; P = .03). A main diet moisture percentage of ≤14% as fed was significantly associated with confirmed CE in logistic regression analysis (OR 5.71 [95% CI: 1.18-27.69]; P = .03). CONCLUSIONS AND CLINICAL IMPORTANCE The presence of dietary carbohydrate, protein source, and dietary moisture content, or factors related to moisture content such as preservatives, might play a role as potential pre-illness dietary risk factors in dogs with CE.
Collapse
Affiliation(s)
- Isla Trewin
- Royal Veterinary CollegeUniversity of LondonLondonEngland
| | - Aarti Kathrani
- Royal Veterinary CollegeUniversity of LondonLondonEngland
| |
Collapse
|
28
|
Garnica-Galvez S, Skoufos I, Tzora A, Diakakis N, Prassinos N, Zeugolis DI. Macromolecular crowding in equine bone marrow mesenchymal stromal cell cultures using single and double hyaluronic acid macromolecules. Acta Biomater 2023; 170:111-123. [PMID: 37634833 DOI: 10.1016/j.actbio.2023.08.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 08/29/2023]
Abstract
Macromolecular crowding (MMC) enhances and accelerates extracellular matrix (ECM) deposition in eukaryotic cell culture. Single hyaluronic acid (HA) molecules have not induced a notable increase in the amount and rate of deposited ECM. Thus, herein we assessed the physicochemical properties and biological consequences in equine bone marrow mesenchymal stromal cell cultures of single and mixed HA molecules and correlated them to the most widely used MMC agents, the FicollⓇ cocktail (FC) and carrageenan (CR). Dynamic light scattering analysis revealed that all HA cocktails had significantly higher hydrodynamic radius than the FC and CR; the FC and the 0.5 mg/ml 100 kDa and 500 kDa single HA molecules had the highest charge; and, in general, all molecules had high polydispersity index. Biological analyses revealed that none of the MMC agents affected cell morphology and basic cell functions; in general, CR outperformed all other macromolecules in collagen type I and V deposition; FC, the individual HA molecules and the HA cocktails outperformed CR in collagen type III deposition; FC outperformed CR and the individual HA molecules and the HA cocktails outperformed their constituent HA molecules in collagen type IV deposition; FC and certain HA cocktails outperformed CR and constituent HA molecules in collagen type VI deposition; and all individual HA molecules outperformed FC and CR and the HA cocktails outperformed their constituent HA molecules in laminin deposition. With respect to tri-lineage analysis, CR and HA enhanced chondrogenesis and osteogenesis, whilst FC enhanced adipogenesis. This work opens new avenues in mixed MMC in eukaryotic cell culture. STATEMENT OF SIGNIFICANCE: Mixed macromolecular crowding (MMC) in eukaryotic cell culture is still under-investigated. Herein, single and double hyaluronic acid (HA) macromolecules, along with the traditional MMC agents FicollⓇ cocktail (FC) and carrageenan (CR), were used as MMC agents in equine mesenchymal stromal cell cultures. Biological analysis showed that none of the MMC agents affected cell morphology and basic cell functions. Protein deposition analysis made apparent that CR outperformed all other macromolecules in collagen type I and collagen type V deposition, whilst FC, the individual HA macromolecules and the HA cocktails outperformed CR in collagen type III deposition. Tri-lineage analysis revealed that CR and HA enhanced chondrogenesis and osteogenesis, whilst FC enhanced adipogenesis. These data illustrate that MMC agents are not inert macromolecules.
Collapse
Affiliation(s)
- Sergio Garnica-Galvez
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, Arta, Greece; School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Skoufos
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, Arta, Greece
| | - Athina Tzora
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, Arta, Greece
| | - Nikolaos Diakakis
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikitas Prassinos
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland.
| |
Collapse
|
29
|
Khatib MA, Saleemani HH, Kurdi NB, Alhibshi HN, Jastaniah MA, Ajabnoor SM. Low Emulsifier Diet in Healthy Female Adults: A Feasibility Study of Nutrition Education and Counseling Intervention. Healthcare (Basel) 2023; 11:2644. [PMID: 37830680 PMCID: PMC10572653 DOI: 10.3390/healthcare11192644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/14/2023] Open
Abstract
Emulsifiers are food additives commonly found in processed foods to improve texture stabilization and food preservation. Dietary emulsifier intake can potentially damage the gut mucosal lining resulting in chronic inflammation such as Crohn's disease. This study investigates the feasibility of a low-emulsifier diet among healthy female adults, as no previous reports have studied the feasibility of such a diet on healthy participants. A quasi-experimental study for a nutrition education and counseling intervention was conducted over 14 days among healthy Saudi participants aged 18 years and over. Assessment of dietary intake using 3-day food records was conducted at the baseline and 2-week follow-up. Participants attended an online educational session using the Zoom application illustrating instructions for a low-emulsifier diet. Daily exposure to emulsifiers was evaluated and nutrient intake was measured. A total of 30 participants completed the study. At baseline, 38 emulsifiers were identified, with a mean ± SD exposure of 12.23 ± 10.07 emulsifiers consumed per day. A significant reduction in the mean frequency of dietary emulsifier intake was observed at the end of the intervention (12.23 ± 10.07 vs. 6.30 ± 7.59, p < 0.01). However, intake of macronutrients and micronutrients was significantly reduced (p < 0.05). Good adherence to the diet was achieved by 40% of the participants, and 16.66% attained a 50% reduction of emulsifier intake. The study demonstrates that a low-emulsifier diet provided via dietary advice is feasible to follow and tolerable by healthy participants. However, the diet still needs further investigation and assessment of it is nutritional intake and quality before implementing it in patients with inflammatory bowel disease who are at high risk of poor nutritional intake.
Collapse
Affiliation(s)
- Mai A. Khatib
- Clinical Nutrition Department, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80215, Jeddah 21589, Saudi Arabia
| | | | | | | | | | | |
Collapse
|
30
|
Sellem L, Srour B, Javaux G, Chazelas E, Chassaing B, Viennois E, Debras C, Salamé C, Druesne-Pecollo N, Esseddik Y, de Edelenyi FS, Agaësse C, De Sa A, Lutchia R, Louveau E, Huybrechts I, Pierre F, Coumoul X, Fezeu LK, Julia C, Kesse-Guyot E, Allès B, Galan P, Hercberg S, Deschasaux-Tanguy M, Touvier M. Food additive emulsifiers and risk of cardiovascular disease in the NutriNet-Santé cohort: prospective cohort study. BMJ 2023; 382:e076058. [PMID: 37673430 PMCID: PMC10480690 DOI: 10.1136/bmj-2023-076058] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 09/08/2023]
Abstract
OBJECTIVE To assess the associations between exposure to food additive emulsifiers and risk of cardiovascular disease (CVD). DESIGN Prospective cohort study. SETTING French NutriNet-Santé study, 2009-21. PARTICIPANTS 95 442 adults (>18 years) without prevalent CVD who completed at least three 24 hour dietary records during the first two years of follow-up. MAIN OUTCOME MEASURES Associations between intake of food additive emulsifiers (continuous (mg/day)) and risk of CVD, coronary heart disease, and cerebrovascular disease characterised using multivariable proportional hazard Cox models to compute hazard ratios for each additional standard deviation (SD) of emulsifier intake, along with 95% confidence intervals. RESULTS Mean age was 43.1 (SD 14.5) years, and 79.0% (n=75 390) of participants were women. During follow-up (median 7.4 years), 1995 incident CVD, 1044 coronary heart disease, and 974 cerebrovascular disease events were diagnosed. Higher intake of celluloses (E460-E468) was found to be positively associated with higher risks of CVD (hazard ratio for an increase of 1 standard deviation 1.05, 95% confidence interval 1.02 to 1.09, P=0.003) and coronary heart disease (1.07, 1.02 to 1.12, P=0.004). Specifically, higher cellulose E460 intake was linked to higher risks of CVD (1.05, 1.01 to 1.09, P=0.007) and coronary heart disease (1.07, 1.02 to 1.12, P=0.005), and higher intake of carboxymethylcellulose (E466) was associated with higher risks of CVD (1.03, 1.01 to 1.05, P=0.004) and coronary heart disease (1.04, 1.02 to 1.06, P=0.001). Additionally, higher intakes of monoglycerides and diglycerides of fatty acids (E471 and E472) were associated with higher risks of all outcomes. Among these emulsifiers, lactic ester of monoglycerides and diglycerides of fatty acids (E472b) was associated with higher risks of CVD (1.06, 1.02 to 1.10, P=0.002) and cerebrovascular disease (1.11, 1.06 to 1.16, P<0.001), and citric acid ester of monoglycerides and diglycerides of fatty acids (E472c) was associated with higher risks of CVD (1.04, 1.02 to 1.07, P=0.004) and coronary heart disease (1.06, 1.03 to 1.09, P<0.001). High intake of trisodium phosphate (E339) was associated with an increased risk of coronary heart disease (1.06, 1.00 to 1.12, P=0.03). Sensitivity analyses showed consistent associations. CONCLUSION This study found positive associations between risk of CVD and intake of five individual and two groups of food additive emulsifiers widely used in industrial foods. TRIAL REGISTRATION ClinicalTrials.gov NCT03335644.
Collapse
Affiliation(s)
- Laury Sellem
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Bernard Srour
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Guillaume Javaux
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Eloi Chazelas
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Benoit Chassaing
- INSERM U1016, team "Mucosal microbiota in chronic inflammatory diseases," Université Paris Cité, Paris, France
| | - Emilie Viennois
- INSERM U1149, Centre for Research on Inflammation, Université de Paris, Paris, France
| | - Charlotte Debras
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Clara Salamé
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Nathalie Druesne-Pecollo
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Younes Esseddik
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Fabien Szabo de Edelenyi
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Cédric Agaësse
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Alexandre De Sa
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Rebecca Lutchia
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Erwan Louveau
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Inge Huybrechts
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Fabrice Pierre
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | | | - Léopold K Fezeu
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Chantal Julia
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
- Public Health Department, Groupe Hospitalier Paris-Seine-Saint-Denis, Assistance Publique-Hôpitaux de Paris (AP-HP), Bobigny, France
| | - Emmanuelle Kesse-Guyot
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Benjamin Allès
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Pilar Galan
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Serge Hercberg
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
- Public Health Department, Groupe Hospitalier Paris-Seine-Saint-Denis, Assistance Publique-Hôpitaux de Paris (AP-HP), Bobigny, France
| | - Mélanie Deschasaux-Tanguy
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| | - Mathilde Touvier
- Université Sorbonne Paris Nord and Université Paris Cité, INSERM, INRAE, CNAM, Center of Research in Epidemiology and StatisticS (CRESS), Nutritional Epidemiology Research Team (EREN), Bobigny, France
| |
Collapse
|
31
|
Han M, Liao W, Dong Y, Fei T, Gai Z. Sustained ameliorative effect of Lactobacillus acidophilus LA85 on dextran sulfate sodium-induced colitis in mice. J Food Sci 2023; 88:3893-3904. [PMID: 37548631 DOI: 10.1111/1750-3841.16723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/28/2023] [Accepted: 07/14/2023] [Indexed: 08/08/2023]
Abstract
Ulcerative colitis (UC) is a type of inflammatory bowel disease associated with immune system dysfunction caused by gut dysbiosis. This study aimed to investigate the alleviating effect of Lactobacillus acidophilus LA85 on colitis and its underlying mechanism using mouse models of dextran sulfate sodium (DSS)-induced UC. The UC mouse models were established by treating C57BL/6J male mice with 2.5% (w/v) DSS in drinking water for 7 days. These mice received supplementation with either L. acidophilus LA85 (1 × 109 colony-forming units/day) or 200 µL of sterile water once daily (LA85-treated and UC model mice, respectively). The disease activity index (DAI), colon length, and histological changes in the colons of mice were then analyzed at Day 21, and the effects of L. acidophilus LA85 on the gut microbiota and serum inflammatory cytokines were also investigated. Compared with the UC model mice, L. acidophilus LA85-treated UC mice showed significant reductions in a variety of colitis symptoms, including weight loss, the DAI score, colon shortening, and colon tissue damage. Lactobacillus acidophilus LA85 supplementation also significantly decreased the serum concentrations of tumor necrosis factor α and interleukin-6 while increasing the serum concentration of IL-10. Furthermore, LA85 supplementation improved the diversity and composition of the gut microbiota, both of which had been decreased by DSS. In particular, L. acidophilus LA85-treated UC mice showed higher relative abundances of Akkermansia and Romboutsia than the UC model mice. These results demonstrate that L. acidophilus LA85 can alleviate inflammatory diseases of the intestine, such as inflammatory bowel disease, by regulating immune responses and restoring the gut microbiota. PRACTICAL APPLICATION: Ulcerative colitis is a type of inflammatory bowel disease caused by imbalance of gut microbiota. This study showed that L. acidophilus LA85 can alleviate DSS-induced colitis in mice through regulation of inflammatory cytokines, protection of intestinal barrier, and regulation of specific gut microbiota. L. acidophilus LA85 is a promising probiotic candidate for the treatment of UC.
Collapse
Affiliation(s)
- Mei Han
- Shanghai Business School, Shanghai, China
| | - Wenyan Liao
- State Key Laboratory of Dairy Biotechnology, Technology Center Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Yao Dong
- Department of Research and Development, Wecare Probiotics Co., Ltd., Suzhou, China
| | - Teng Fei
- Department of Research and Development, Wecare Probiotics Co., Ltd., Suzhou, China
| | - Zhonghui Gai
- Department of Research and Development, Wecare Probiotics Co., Ltd., Suzhou, China
| |
Collapse
|
32
|
Narula N, Chang NH, Mohammad D, Wong ECL, Ananthakrishnan AN, Chan SSM, Carbonnel F, Meyer A. Food Processing and Risk of Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. Clin Gastroenterol Hepatol 2023; 21:2483-2495.e1. [PMID: 36731590 DOI: 10.1016/j.cgh.2023.01.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/29/2022] [Accepted: 01/12/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND & AIMS Several studies have been published on the association between food processing and risks of Crohn's disease (CD) and ulcerative colitis (UC), with some variability in results. We performed a systematic literature review and meta-analysis to study this association. METHODS From PubMed, Medline, and Embase until October 2022, we identified cohort studies that studied the association between food processing and the risk of CD or UC. Risk of bias of the included studies was assessed by the Newcastle-Ottawa scale. We computed pooled hazard ratios (HRs) and 95% confidence intervals (CIs) using random-effects meta-analysis based on estimates and standard errors. RESULTS A total of 1,068,425 participants were included (13,594,422 person-years) among 5 cohort studies published between 2020 and 2022. Four of the 5 included studies were scored as high quality. The average age of participants ranged from 43 to 56 years; 55%-83% were female. During follow-up, 916 participants developed CD, and 1934 developed UC. There was an increased risk for development of CD for participants with higher consumption of ultra-processed foods compared with those with lower consumption (HR, 1.71; 95% CI, 1.37-2.14; I2 = 0%) and a lower risk of CD for participants with higher consumption of unprocessed/minimally processed foods compared with those with lower consumption (HR, 0.71; 95% CI, 0.53-0.94; I2 = 11%). There was no association between risk of UC and ultra-processed foods (HR, 1.17; 95% CI, 0.86-1.61; I2 = 74%) or unprocessed/minimally processed foods (HR, 0.84; 95% CI, 0.68-1.02; I2 = 0%). CONCLUSIONS Higher ultra-processed food and lower unprocessed/minimally processed food intakes are associated with higher risk of CD but not UC.
Collapse
Affiliation(s)
- Neeraj Narula
- Department of Medicine (Division of Gastroenterology) and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada.
| | - Nicole H Chang
- Department of Medicine (Division of Gastroenterology) and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Danah Mohammad
- Department of Medicine (Division of Gastroenterology) and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Emily C L Wong
- Department of Medicine (Division of Gastroenterology) and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Simon S M Chan
- Department of Gastroenterology, Norfolk and Norwich University Hospital and Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Franck Carbonnel
- Department of Gastroenterology, University Hospital of Bicêtre, Assistance Publique-Hôpitaux de Paris and Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Antoine Meyer
- Department of Gastroenterology, University Hospital of Bicêtre, Assistance Publique-Hôpitaux de Paris and Université Paris-Saclay, Le Kremlin Bicêtre, France
| |
Collapse
|
33
|
Stolfi C, Pacifico T, Monteleone G, Laudisi F. Impact of Western Diet and Ultra-Processed Food on the Intestinal Mucus Barrier. Biomedicines 2023; 11:2015. [PMID: 37509654 PMCID: PMC10377275 DOI: 10.3390/biomedicines11072015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/19/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
The intestinal epithelial barrier plays a key role in the absorption of nutrients and water, in the regulation of the interactions between luminal contents and the underlying immune cells, and in the defense against enteric pathogens. Additionally, the intestinal mucus layer provides further protection due to mucin secretion and maturation by goblet cells, thus representing a crucial player in maintaining intestinal homeostasis. However, environmental factors, such as dietary products, can disrupt this equilibrium, leading to the development of inflammatory intestinal disorders. In particular, ultra-processed food, which is broadly present in the Western diet and includes dietary components containing food additives and/or undergoing multiple industrial processes (such as dry heating cooking), was shown to negatively impact intestinal health. In this review, we summarize and discuss current knowledge on the impact of a Western diet and, in particular, ultra-processed food on the mucus barrier and goblet cell function, as well as potential therapeutic approaches to maintain and restore the mucus layer under pathological conditions.
Collapse
Affiliation(s)
- Carmine Stolfi
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Teresa Pacifico
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
- Gastroenterology Unit, Policlinico Universitario Tor Vergata, 00133 Rome, Italy
| | - Federica Laudisi
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| |
Collapse
|
34
|
Ren R, Zhao AQ, Chen L, Wu S, Hung WL, Wang B. Therapeutic effect of Lactobacillus plantarum JS19 on mice with dextran sulfate sodium induced acute and chronic ulcerative colitis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:4143-4156. [PMID: 36573836 DOI: 10.1002/jsfa.12414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 05/03/2023]
Abstract
BACKGROUND Ulcerative colitis is associated with intestinal inflammation and dysbiosis. Previous studies have shown that probiotics are potential agents for treatment of inflammatory bowel disease (IBD). Jiang-shui is a traditional fermented vegetable that is rich in lactic acid bacteria (LABs), but the preventive effect of LABs in jiang-shui on IBD is not yet fully understood. RESULTS We isolated 38 LAB strains from jiang-shui, and Lactobacillus plantarum JS19 exhibited the strongest antioxidant activity among them. Our data indicate that oral administration of L. plantarum JS19 significantly inhibited body weight loss, colon shortening and damage, and reduced the disease activity index score in the mice with dextran sulfate sodium (DSS)-induced colitis. In addition, L. plantarum JS19 also alleviated inflammatory responses and oxidative stress through reducing lipid peroxidation, tumor necrosis factor-α expression, and myeloperoxidase activity and enhancing the antioxidant enzyme activity. Importantly, L. plantarum JS19 significantly rebalanced DSS-induced dysbiosis of gut microbiota. CONCLUSION L. plantarum JS19 may be used as a potential probiotic to prevent IBD, particularly ulcerative colitis. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Rong Ren
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Ai-Qing Zhao
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Li Chen
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Shan Wu
- Research and Development Center, Xi'an Yinqiao Dairy (Group) Co., Ltd, Xi'an, China
| | - Wei-Lun Hung
- School of Food Safety, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Bini Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
35
|
Song Z, Song R, Liu Y, Wu Z, Zhang X. Effects of ultra-processed foods on the microbiota-gut-brain axis: The bread-and-butter issue. Food Res Int 2023; 167:112730. [PMID: 37087282 DOI: 10.1016/j.foodres.2023.112730] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/11/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023]
Abstract
The topic of gut microbiota and the microbiota-gut-brain (MGB) axis has become the forefront of research and reports in the past few years. The gut microbiota is a dynamic interface between the environment, food, and the host, reflecting the health status as well as maintaining normal physiological metabolism. Modern ultra-processed foods (UPF) contain large quantities of saturated and trans fat, added sugar, salt, and food additives that seriously affect the gut and physical health. In addition, these unhealthy components directly cause changes in gut microbiota functions and microbial metabolism, subsequently having the potential to impact the neural network. This paper reviews an overview of the link between UPF ingredients and the MGB axis. Considerable studies have examined that high intake of trans fat, added sugar and salt have deleterious effects on gut and brain functions, but relatively less focus has been placed on the impact of food additives on the MGB axis. Data from several studies suggest that food additives might be linked to metabolic diseases and inflammation. They may also alter the gut microbiota composition and microbial metabolites, which potentially affect cognition and behavior. Therefore, we emphasize that food additives including emulsifiers, artificial sweeteners, colorants, and preservatives interact with the gut microbiota and their possible effects on altering the brain and behavior based on the latest research. Future studies should further investigate whether gut dysbiosis mediates the effect of UPF on brain diseases and behavior. This thesis here sheds new light on future research pointing to the potentially detrimental effects of processed food consumption on brain health.
Collapse
|
36
|
Xue C, Li G, Gu X, Su Y, Zheng Q, Yuan X, Bao Z, Lu J, Li L. Health and Disease: Akkermansia muciniphila, the Shining Star of the Gut Flora. RESEARCH (WASHINGTON, D.C.) 2023; 6:0107. [PMID: 37040299 PMCID: PMC10079265 DOI: 10.34133/research.0107] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/15/2023] [Indexed: 04/05/2023]
Abstract
Akkermansia muciniphila (A. muciniphila) has drawn much attention as an important gut microbe strain in recent years. A. muciniphila can influence the occurrence and development of diseases of the endocrine, nervous, digestive, musculoskeletal, and respiratory systems and other diseases. It can also improve immunotherapy for some cancers. A. muciniphila is expected to become a new probiotic in addition to Lactobacillus and Bifidobacterium. An increase in A. muciniphila abundance through direct or indirect A. muciniphila supplementation may inhibit or even reverse disease progression. However, some contrary findings are found in type 2 diabetes mellitus and neurodegenerative diseases, where increased A. muciniphila abundance may aggravate the diseases. To enable a more comprehensive understanding of the role of A. muciniphila in diseases, we summarize the relevant information on A. muciniphila in different systemic diseases and introduce regulators of A. muciniphila abundance to promote the clinical transformation of A. muciniphila research.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ganglei Li
- Department of Neurosurgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuanshuai Su
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiuxian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhengyi Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
37
|
Rheological behavior and molecular dynamics simulation of κ-carrageenan/casein under simulated gastrointestinal electrolyte conditions. Food Hydrocoll 2023. [DOI: 10.1016/j.foodhyd.2022.108240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
38
|
Guo J, Shang X, Chen P, Huang X. How does carrageenan cause colitis? A review. Carbohydr Polym 2023; 302:120374. [PMID: 36604052 DOI: 10.1016/j.carbpol.2022.120374] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022]
Abstract
Carrageenan is a common additive, but mounting studies have reported that it may cause or aggravate inflammation in the intestines. The safety of carrageenan remains controversial and its inflammatory mechanisms are unclear. In this review, the pathogenesis of colitis by carrageenans was discussed. We analyzed the pathogenesis of inflammatory bowel disease, followed that line of thought, the existing evidence of carrageenans causing colitis in cellular and animal models was summarized to draw its colitis pathogenesis. Two pathways were described including: 1) carrageenan changed the composition of intestinal microbiota, especially Akkermansia muciniphila, which destroyed the mucosal barrier and triggered the inflammatory immune response; and 2) carrageenan directly contacted with receptors on epithelial cells and activated the NF-κB inflammatory pathway. This review aim to provide guidance for exploring the treatment of colitis caused by carrageenan, and safe processing and utilization of carrageenan in food industry, which is worthy of study in the future.
Collapse
Affiliation(s)
- Juanjuan Guo
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou, Fujian 362000, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China.
| | - Xuke Shang
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou, Fujian 362000, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Peilin Chen
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou, Fujian 362000, China
| | - Xiaozhou Huang
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou, Fujian 362000, China
| |
Collapse
|
39
|
Saha S, Patel N. What Should I Eat? Dietary Recommendations for Patients with Inflammatory Bowel Disease. Nutrients 2023; 15:nu15040896. [PMID: 36839254 PMCID: PMC9966256 DOI: 10.3390/nu15040896] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disorder thought to be caused by enteric inflammation in a genetically susceptible host. Although the pathogenesis of IBD is largely unknown, it is widely accepted that dietary components play an important role. Human and animal-based studies have explored the role of various dietary components such as meat, artificial sweeteners and food additives in causing enteric inflammation. Several diets have also been studied in patients with IBD, specifically their role in the induction or maintenance of remission. The most well-studied of these include exclusive enteral nutrition and specific carbohydrate diet. A diet low in FODMAPs (fermentable oligosaccharides, disaccharides, monosaccharides and polyols), typically prescribed for patients with irritable bowel syndrome, has also been studied in a specific subgroup of patients with IBD. In this review, we describe the current evidence on how various dietary components can induce enteric and colonic inflammation, and the clinical-epidemiological evidence exploring their role in predisposing to or protecting against the development of IBD. We also discuss several special diets and how they affect clinical outcomes in IBD patients. Based on the available evidence, we provide guidance for patients and clinicians managing IBD regarding the best practice in dietary modifications.
Collapse
Affiliation(s)
- Srishti Saha
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Neha Patel
- Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence: ; Tel.: +1-469-776-0671
| |
Collapse
|
40
|
Wang Y, Wang K, Du M, Khandpur N, Rossato SL, Lo CH, VanEvery H, Kim DY, Zhang FF, Chavarro JE, Sun Q, Huttenhower C, Song M, Nguyen LH, Chan AT. Maternal consumption of ultra-processed foods and subsequent risk of offspring overweight or obesity: results from three prospective cohort studies. BMJ 2022; 379:e071767. [PMID: 36198411 PMCID: PMC9533299 DOI: 10.1136/bmj-2022-071767] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/18/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To assess whether maternal ultra-processed food intake during peripregnancy and during the child rearing period is associated with offspring risk of overweight or obesity during childhood and adolescence. DESIGN Population based prospective cohort study. SETTING The Nurses' Health Study II (NHSII) and the Growing Up Today Study (GUTS I and II) in the United States. PARTICIPANTS 19 958 mother-child (45% boys, aged 7-17 years at study enrollment) pairs with a median follow-up of 4 years (interquartile range 2-5 years) until age 18 or the onset of overweight or obesity, including a subsample of 2925 mother-child pairs with information on peripregnancy diet. MAIN OUTCOME MEASURES Multivariable adjusted, log binomial models with generalized estimating equations and an exchangeable correlation structure were used to account for correlations between siblings and to estimate the relative risk of offspring overweight or obesity defined by the International Obesity Task Force. RESULTS 2471 (12.4%) offspring developed overweight or obesity in the full analytic cohort. After adjusting for established maternal risk factors and offspring's ultra-processed food intake, physical activity, and sedentary time, maternal consumption of ultra-processed foods during the child rearing period was associated with overweight or obesity in offspring, with a 26% higher risk in the group with the highest maternal ultra-processed food consumption (group 5) versus the lowest consumption group (group 1; relative risk 1.26, 95% confidence interval 1.08 to 1.47, P for trend<0.001). In the subsample with information on peripregnancy diet, while rates were higher, peripregnancy ultra-processed food intake was not significantly associated with an increased risk of offspring overweight or obesity (n=845 (28.9%); group 5 v group 1: relative risk 1.17, 95% confidence interval 0.89 to 1.53, P fortrend=0.07). These associations were not modified by age, sex, birth weight, and gestational age of offspring or maternal body weight. CONCLUSIONS Maternal consumption of ultra-processed food during the child rearing period was associated with an increased risk of overweight or obesity in offspring, independent of maternal and offspring lifestyle risk factors. Further study is needed to confirm these findings and to understand the underlying biological mechanisms and environmental determinants. These data support the importance of refining dietary recommendations and the development of programs to improve nutrition for women of reproductive age to promote offspring health.
Collapse
Affiliation(s)
- Yiqing Wang
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kai Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Mengxi Du
- Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Neha Khandpur
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
- Center for Epidemiological Studies in Health and Nutrition, Faculty of Public Health, University of São Paulo, São Paulo, Brazil
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sinara Laurini Rossato
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Institute of Geography, Graduation course of Collective Health, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Chun-Han Lo
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Hannah VanEvery
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Daniel Y Kim
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Fang Fang Zhang
- Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
- Department of Public Health and Community Medicine, School of Medicine, Tufts University, Boston, MA, USA
| | - Jorge E Chavarro
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Qi Sun
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mingyang Song
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Long H Nguyen
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
41
|
Chelliah R, Park SJ, Oh S, Lee E, Daliri EBM, Elahi F, Park CR, Sultan G, Madar IH, Oh DH. Unveiling the potentials of bioactive oligosaccharide1-kestose (GF2) from Musa paradisiaca Linn peel with an anxiolytic effect based on gut microbiota modulation in stressed mice model. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Chelliah R, Park SJ, Oh S, Lee E, Daliri EBM, Elahi F, Park CR, Sultan G, Madar IH, Oh DH. Unveiling the potentials of bioactive oligosaccharide1-kestose (GF2) from Musa paradisiaca Linn peel with an anxiolytic effect based on gut microbiota modulation in stressed mice model. FOOD BIOSCI 2022; 49:101881. [DOI: https:/doi.10.1016/j.fbio.2022.101881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
|
43
|
Djouina M, Vignal C, Dehaut A, Caboche S, Hirt N, Waxin C, Himber C, Beury D, Hot D, Dubuquoy L, Launay D, Duflos G, Body-Malapel M. Oral exposure to polyethylene microplastics alters gut morphology, immune response, and microbiota composition in mice. ENVIRONMENTAL RESEARCH 2022; 212:113230. [PMID: 35398082 DOI: 10.1016/j.envres.2022.113230] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 06/14/2023]
Abstract
The ubiquitous and growing presence of microplastics (MPs) in all compartments of the environment raises concerns about their possible harmful effects on human health. Human exposure to MPs occurs largely through ingestion. Polyethylene (PE) is widely employed for reusable bags and food packaging and found to be present in drinking water and food. It is also one of the major polymers detected in human stool. The aim of this study was to characterize the effects of intestinal exposure to PE MPs on gut homeostasis. Mice were orally exposed for 6 weeks to PE microbeads of 2 different sizes, 36 and 116 μm, that correspond to those found in human stool. They were administrated either individually or as a mixture at a dose of 100 μg/g of food. Both PE microbead sizes were detected in mouse stool. Different parameters related to major intestinal functions were compared between control mice, mice exposed to each type of microbead, or co-exposed to the 2 types of microbeads. Intestinal disturbances were observed after individual exposure to each size of PE microbead, and the most marked deleterious effects were found in co-exposed mice. At the histomorphological level, crypt depth was increased throughout the intestinal tissues. Significant variations of gene expression related to epithelial, permeability, and inflammatory biomarkers were quantified. Defective recruitment of some intestinal immune cells was observed from the proximal portion of the small intestine to the colon. Several bacterial taxa at the order level were found to be affected by exposure to the MPs by metagenomic analysis of cecal microbiota. These results show that ingestion of PE microbeads induces significant alterations of crucial intestinal markers in mice and underscores the need to further study the health impact of MP exposure in humans.
Collapse
Affiliation(s)
- Madjid Djouina
- Univ. Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - Cécile Vignal
- Univ. Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - Alexandre Dehaut
- ANSES - Laboratoire de Sécurité des Aliments, 6 Boulevard Du Bassin Napoléon, 62200, Boulogne-sur-Mer, France
| | - Ségolène Caboche
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR2014 - US41 - PLBS-Plateformes Lilloises de Biologie & Santé, F-59000, Lille, France
| | - Nell Hirt
- Univ. Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - Christophe Waxin
- Univ. Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - Charlotte Himber
- ANSES - Laboratoire de Sécurité des Aliments, 6 Boulevard Du Bassin Napoléon, 62200, Boulogne-sur-Mer, France
| | - Delphine Beury
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR2014 - US41 - PLBS-Plateformes Lilloises de Biologie & Santé, F-59000, Lille, France
| | - David Hot
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR2014 - US41 - PLBS-Plateformes Lilloises de Biologie & Santé, F-59000, Lille, France
| | - Laurent Dubuquoy
- Univ. Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - David Launay
- Univ. Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - Guillaume Duflos
- ANSES - Laboratoire de Sécurité des Aliments, 6 Boulevard Du Bassin Napoléon, 62200, Boulogne-sur-Mer, France
| | - Mathilde Body-Malapel
- ANSES - Laboratoire de Sécurité des Aliments, 6 Boulevard Du Bassin Napoléon, 62200, Boulogne-sur-Mer, France.
| |
Collapse
|
44
|
Liu C, Zhan S, Tian Z, Li N, Li T, Wu D, Zeng Z, Zhuang X. Food Additives Associated with Gut Microbiota Alterations in Inflammatory Bowel Disease: Friends or Enemies? Nutrients 2022; 14:nu14153049. [PMID: 35893902 PMCID: PMC9330785 DOI: 10.3390/nu14153049] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/08/2022] [Accepted: 07/21/2022] [Indexed: 12/13/2022] Open
Abstract
During the 21st century, the incidence and prevalence of inflammatory bowel disease (IBD) is rising globally. Despite the pathogenesis of IBD remaining largely unclear, the interactions between environmental exposure, host genetics and immune response contribute to the occurrence and development of this disease. Growing evidence implicates that food additives might be closely related to IBD, but the involved molecular mechanisms are still poorly understood. Food additives may be categorized as distinct types in accordance with their function and property, including artificial sweeteners, preservatives, food colorant, emulsifiers, stabilizers, thickeners and so on. Various kinds of food additives play a role in modifying the interaction between gut microbiota and intestinal inflammation. Therefore, this review comprehensively synthesizes the current evidence on the interplay between different food additives and gut microbiome alterations, and further elucidates the potential mechanisms of food additives–associated microbiota changes involved in IBD.
Collapse
Affiliation(s)
- Caiguang Liu
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (C.L.); (S.Z.); (N.L.); (T.L.); (D.W.)
| | - Shukai Zhan
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (C.L.); (S.Z.); (N.L.); (T.L.); (D.W.)
| | - Zhenyi Tian
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China;
| | - Na Li
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (C.L.); (S.Z.); (N.L.); (T.L.); (D.W.)
| | - Tong Li
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (C.L.); (S.Z.); (N.L.); (T.L.); (D.W.)
| | - Dongxuan Wu
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (C.L.); (S.Z.); (N.L.); (T.L.); (D.W.)
| | - Zhirong Zeng
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (C.L.); (S.Z.); (N.L.); (T.L.); (D.W.)
- Correspondence: (Z.Z.); (X.Z.)
| | - Xiaojun Zhuang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (C.L.); (S.Z.); (N.L.); (T.L.); (D.W.)
- Correspondence: (Z.Z.); (X.Z.)
| |
Collapse
|
45
|
Abstract
Crohn's disease (CD) is chronic immune-related disease of the gastrointestinal tract hypothesized to be caused by an interplay of genetic predisposition and environmental exposures. With the global incidence increasing, more patients are exploring dietary exposures to explain and treat CD. However, most patients report minimal nutritional education from their provider, and providers report few nutritional resources to help them educate patients. This highlights the previous deficit of literature describing the role and influence of diet in CD. To address this need, this article reviews available literature on the possible roles of diet in the pathogenesis, exacerbation, and treatment of CD.
Collapse
Affiliation(s)
- Phillip Gu
- Division of Digestive and Liver Diseases, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Linda A Feagins
- Department of Medicine, Center for Inflammatory Bowel Diseases, University of Texas at Austin, Dell Medical School, Health Discovery Building, Z0900 1601 Trinity Street, Building B, Austin, TX 78712, USA.
| |
Collapse
|
46
|
Lo CH, Khandpur N, Rossato SL, Lochhead P, Lopes EW, Burke KE, Richter JM, Song M, Korat AVA, Sun Q, Fung TT, Khalili H, Chan AT, Ananthakrishnan AN. Ultra-processed Foods and Risk of Crohn's Disease and Ulcerative Colitis: A Prospective Cohort Study. Clin Gastroenterol Hepatol 2022; 20:e1323-e1337. [PMID: 34461300 PMCID: PMC8882700 DOI: 10.1016/j.cgh.2021.08.031] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The rising incidence of inflammatory bowel disease in regions undergoing Westernization has coincided with the increase in ultra-processed food (UPF) consumption over the past few decades. We aimed to examine the association between consumption of UPFs and the risk of Crohn's disease (CD) and ulcerative colitis (UC). METHODS We performed a prospective cohort study of 3 nationwide cohorts of health professionals in the United States-the Nurses' Health Study (1986-2014), the Nurses' Health Study II (1991-2017), and the Health Professionals Follow-up Study (1986-2012). We employed Cox proportional hazards models with adjustment for confounders to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs) for CD and UC according to self-reported consumption of UPFs. RESULTS The study included 245,112 participants. Over 5,468,444 person-years of follow-up, we documented 369 incident cases of CD and 488 incident cases of UC. The median age at diagnosis was 56 years (range, 29-85 years). Compared with participants in the lowest quartile of simple updated UPF consumption, those in the highest quartile had a significantly increased risk of CD (HR, 1.70; 95% CI, 1.23-2.35; Ptrend = .0008). Among different UPF subgroups, ultra-processed breads and breakfast foods; frozen or shelf-stable ready-to-eat/heat meals; and sauces, cheeses, spreads, and gravies showed the strongest positive associations with CD risk (HR per 1 standard deviation increase in intake, 1.18 [95% CI, 1.07-1.29], 1.11 [95% CI, 1.01-1.22], and 1.14 [95% CI, 1.02-1.27], respectively). There was no consistent association between UPF intake and UC risk. CONCLUSIONS Higher UPF intake was associated with an increased risk of incident CD. Further studies are needed to identify specific contributory dietary components.
Collapse
Affiliation(s)
- Chun-Han Lo
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Neha Khandpur
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA,Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil,Center for Epidemiological Studies in Health and Nutrition, Faculty of Public Health, University of São Paulo, São Paulo, Brazil
| | - Sinara Laurini Rossato
- Graduation course in Public Health, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil
| | - Paul Lochhead
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Emily W. Lopes
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kristin E. Burke
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - James M. Richter
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mingyang Song
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Andres Victor Ardisson Korat
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Qi Sun
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Teresa T. Fung
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA,Department of Nutrition, Simmons University, Boston, Massachusetts, USA
| | - Hamed Khalili
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ashwin N. Ananthakrishnan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
47
|
Excellent Cooperation between Carboxyl-Substituted Porphyrins, k-Carrageenan and AuNPs for Extended Application in CO2 Capture and Manganese Ion Detection. CHEMOSENSORS 2022. [DOI: 10.3390/chemosensors10040133] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Significant tasks of the presented research are the development of multifunctional materials capable both to detect/capture carbon dioxide and to monitor toxic metal ions from waters, thus contributing to maintaining a sustainable and clean environment. The purpose of this work was to synthesize, characterize (NMR, FT-IR, UV-Vis, Fluorescence, AFM) and exploit the optical and emission properties of a carboxyl-substituted A3B porphyrin, 5-(4-carboxy-phenyl)-10,15,20-tris-(4-methyl-phenyl)–porphyrin, and based on it, to develop novel composite material able to adsorb carbon dioxide. This porphyrin-k-carrageenan composite material can capture CO2 in ambient conditions with a performance of 6.97 mmol/1 g adsorbent. Another aim of our research was to extend this porphyrin- k-carrageenan material’s functionality toward Mn2+ detection from polluted waters and from medical samples, relying on its synergistic partnership with gold nanoparticles (AuNPs). The plasmonic porphyrin-k-carrageenan-AuNPs material detected Mn2+ in the range of concentration of 4.56 × 10−5 M to 9.39 × 10−5 M (5–11 mg/L), which can be useful for monitoring health of humans exposed to polluted water sources or those who ingested high dietary manganese.
Collapse
|
48
|
Huang W, Tan H, Nie S. Beneficial effects of seaweed-derived dietary fiber: Highlights of the sulfated polysaccharides. Food Chem 2022; 373:131608. [PMID: 34815114 DOI: 10.1016/j.foodchem.2021.131608] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022]
Abstract
Seaweeds and their derivatives are important bioresources of natural bioactive compounds. Nutritional studies indicate that dietary fibers derived from seaweeds have great beneficial potentials in human health and can be developed as functional food. Moreover, sulfated polysaccharides are more likely to be the main bioactive components which are widely distributed in various species of seaweeds including Phaeophyceae, Rhodophyceae and Chlorophyceae. The catabolism by gut microbiota of the seaweeds-derived dietary fibers (DFs) may be one of the pivotal pathways of their physiological functions. Therefore, in this review, we summarized the latest results of the physiological characteristics of seaweed-derived dietary fiber and highlighted the roles of sulfated polysaccharides in the potential regulatory mechanisms against disorders. Meanwhile, the effects of different types of seaweed-derived dietary fiber on gut microbiota were discussed. The analysis of the structure-function correlations and gut microbiota related mechanisms and will contribute to further better applications in food and biotherapeutics.
Collapse
Affiliation(s)
- Wenqi Huang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Huizi Tan
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| |
Collapse
|
49
|
Pan L, Fu T, Cheng H, Mi J, Shang Q, Yu G. Polysaccharide from edible alga Gloiopeltis furcata attenuates intestinal mucosal damage by therapeutically remodeling the interactions between gut microbiota and mucin O-glycans. Carbohydr Polym 2022; 278:118921. [PMID: 34973740 DOI: 10.1016/j.carbpol.2021.118921] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/11/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022]
Abstract
Gloiopeltis furcata is an edible alga that has long been consumed in China. However, the bioactive polysaccharides from G. furcata have been largely unexplored. Here, we show for the first time that a sulfated polysaccharide from G. furcata (SAO) could improve the integrity of the colonic epithelial layer and protect against dextran sulfate sodium-induced intestinal mucosal damage. Mechanistically, SAO attenuated colonic mucosal damage by therapeutically remodeling the interactions between gut microbiota and mucin O-glycans. Specifically, SAO increased the proportions of complex long-chain mucin O-glycans in the epithelial layer with two terminal N-acetylneuraminic acid residues and promoted the growth of probiotic bacteria including Roseburia spp. and Muribaculaceae. Altogether, our study demonstrates a novel application of SAO for the treatment of inflammatory bowel disease-associated mucosal damage and forms the basis to understand the therapeutic effects of natural polysaccharides from the perspective of symbiotic interactions between host mucin O-glycome and gut microbiome.
Collapse
Affiliation(s)
- Lin Pan
- Key Laboratory of Marine Drugs of Ministry of Education, and Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Tianyu Fu
- Key Laboratory of Marine Drugs of Ministry of Education, and Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Hao Cheng
- Key Laboratory of Marine Drugs of Ministry of Education, and Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jianchen Mi
- Key Laboratory of Marine Drugs of Ministry of Education, and Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Qingsen Shang
- Key Laboratory of Marine Drugs of Ministry of Education, and Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Qingdao Marine Biomedical Research Institute, Qingdao 266071, China.
| | - Guangli Yu
- Key Laboratory of Marine Drugs of Ministry of Education, and Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China.
| |
Collapse
|
50
|
Wu W, Zhou J, Xuan R, Chen J, Han H, Liu J, Niu T, Chen H, Wang F. Dietary κ-carrageenan facilitates gut microbiota-mediated intestinal inflammation. Carbohydr Polym 2022; 277:118830. [PMID: 34893247 DOI: 10.1016/j.carbpol.2021.118830] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/10/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022]
Abstract
The inflammatory effects of carrageenan (CGN), a ubiquitous food additive, remains controversial. Gut microbiota and intestinal homeostasis may be a breakthrough in resolving this controversy. Here we show that, κ-CGN did not cause significant inflammatory symptoms, but it did cause reduced bacteria-derived short-chain fatty acids (SCFAs) and decreased thickness of the mucus layer by altering microbiota composition. Administration of the pathogenic bacterium Citrobacter rodentium, further aggravated the inflammation and mucosal damage in the presence of κ-CGN. Mucus layer degradation and altered SCFA levels could be reproduced by fecal transplantation from κ-CGN-fed mice, but not from germ-free κ-CGN-fed mice. These symptoms could be partially repaired by administering probiotics. Our results suggest that κ-CGN may not be directly inflammatory, but it creates an environment that favors inflammation by perturbation of gut microbiota composition and then facilitates expansion of pathogens, and this effect may be partially reversed by the introduction of probiotics.
Collapse
Affiliation(s)
- Wei Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Jiawei Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Rongrong Xuan
- Department of Gynecology and Obstetrics, the Affiliated Hospital of Medical College of Ningbo University, Ningbo, Zhejiang 315211, China
| | - Juanjuan Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Hui Han
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Jingwangwei Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Tingting Niu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Haimin Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Feng Wang
- Department of Laboratory Medicine, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo 315040, China.
| |
Collapse
|