1
|
Choi H, Cho SH, Park JH, Seok YJ. Fructose-responsive regulation by FruR in Faecalibacterium prausnitzii for its intestinal colonization. Commun Biol 2025; 8:426. [PMID: 40082586 PMCID: PMC11906611 DOI: 10.1038/s42003-025-07878-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
Faecalibacterium prausnitzii, a dominant member of healthy human gut microbiota, exhibits a strong positive correlation with fecal fructose levels, suggesting fructose as a key energy source for its colonization and persistence. This study explores the regulatory mechanisms governing the fru operon in F. prausnitzii, responsible for fructose uptake and metabolism. Here, we demonstrate that FruR, a DeoR family transcriptional regulator, orchestrates fru operon expression through interactions with fructose-1-phosphate (F1P) and HPr2, the histidine-containing phosphocarrier protein. The F1P-HPr2(Ser-P)-FruR complex enhances RNA polymerase binding to the fru promoter, with stronger affinity for specific operator motifs compared to apo-FruR. F1P induces structural modifications in FruR that strengthen its interaction with HPr2 and alter its DNA recognition pattern, facilitating RNA polymerase access to the promoter. In vivo experiments in mice demonstrate increased F. prausnitzii abundance alongside upregulated fru operon expression in fructose-rich environments. This study provides new insights into how fructose availability modulates fru operon regulation and promotes F. prausnitzii colonization in the host intestine.
Collapse
Affiliation(s)
- HyeLim Choi
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang-Hyun Cho
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Joo-Hong Park
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yeong-Jae Seok
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Wang L, Li A, Zhang X, Iqbal M, Aabdin ZU, Xu M, Mo Q, Li J. Effect of Bacillus subtilis isolated from yaks on D-galactose-induced oxidative stress and hepatic damage in mice. Front Microbiol 2025; 16:1550556. [PMID: 40109966 PMCID: PMC11920168 DOI: 10.3389/fmicb.2025.1550556] [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: 12/23/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Acute hepatic injury is a severe condition that is always accompanied by oxidative stress and inflammation, seriously threatening the health of the host. Probiotics have been shown to be involved in the regulation of antioxidant system and gut microbiota activity, but studies on the effects of yak derived Bacillus subtilis (B. subtilis) on acute liver injury and oxidative stress remain scarce. Here, we aim to explore the ameliorative effects of B. subtilis isolated from yaks on oxidative stress and hepatic injury caused by D-galactose, as well as the underlying processes. Results indicated that B. subtilis administration, particularly the BS3, significantly mitigated hepatic damage induced by D-galactose in mice as evidenced by ameliorating liver tissue damage as well as decreasing ALT (p < 0.05) and AST (p < 0.05) levels. Additionally, the B. subtilis intervention was demonstrated to enhance the antioxidant system in D-galactose-exposed mice, as manifested by increased T-AOC and SOD, alongside a decrease in MDA levels (p < 0.05). Meanwhile, B. subtilis intervention could effectively mitigate oxidative damage via modulating the Keap1/Nrf2 signaling pathway. Importantly, B. subtilis exhibited a pronounced protective effect against D-galactose-induced intestinal barrier dysfunction through improving tight junction proteins. The gut microbiota results suggest that BS3 alters the abundance of some gut flora such as Firmicutes phylum and Oscillibacter and Lachnospiraceae_NK4A136 genera, which affects the composition of the gut microbiota and reverses the decrease in the microbial richness index in mice. In summary, these findings demonstrated that B. subtilis isolated from yaks serve as a promising candidate to ameliorate oxidative damage and hepatic injury. Meanwhile, the positive regulation effect of B. subtilis on gut microbiota and intestinal mucosal barrier may be one of its underlying mechanisms to alleviate oxidative stress and hepatic injury.
Collapse
Affiliation(s)
- Lei Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Aoyun Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xiaohu Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Mudassar Iqbal
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zain Ul Aabdin
- Department of Preventive Veterinary Medicine and Public Health, Faculty of Veterinary and Animal Sciences, Ziauddin University, Karachi, Pakistan
| | - Mengen Xu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Quan Mo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Animal Science, Xizang Agricultural and Animal Husbandry University, Nyingchi, China
| |
Collapse
|
3
|
González A, Fullaondo A, Rodríguez J, Tirnauca C, Odriozola I, Odriozola A. Conjugated linoleic acid metabolite impact in colorectal cancer: a potential microbiome-based precision nutrition approach. Nutr Rev 2025; 83:e602-e614. [PMID: 38728013 PMCID: PMC11723137 DOI: 10.1093/nutrit/nuae046] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
Colorectal cancer (CRC) is the second most deadly and the third most diagnosed cancer in both sexes worldwide. CRC pathogenesis is associated with risk factors such as genetics, alcohol, smoking, sedentariness, obesity, unbalanced diets, and gut microbiota dysbiosis. The gut microbiota is the microbial community living in symbiosis in the intestine, in a dynamic balance vital for health. Increasing evidence underscores the influence of specific gut microbiota bacterial species on CRC incidence and pathogenesis. In this regard, conjugated linoleic acid (CLA) metabolites produced by certain gut microbiota have demonstrated an anticarcinogenic effect in CRC, influencing pathways for inflammation, proliferation, and apoptosis. CLA production occurs naturally in the rumen, and human bioavailability is through the consumption of food derived from ruminants. In recent years, biotechnological attempts to increase CLA bioavailability in humans have been unfruitful. Therefore, the conversion of essential dietary linoleic acid to CLA metabolite by specific intestinal bacteria has become a promising process. This article reviews the evidence regarding CLA and CLA-producing bacteria as therapeutic agents against CRC and investigates the best strategy for increasing the yield and bioavailability of CLA. Given the potential and limitations of the present strategies, a new microbiome-based precision nutrition approach based on endogenous CLA production by human gut bacteria is proposed. A literature search in the PubMed and PubMed Central databases identified 794 papers on human gut bacteria associated with CLA production. Of these, 51 studies exploring association consistency were selected. After excluding 19 papers, due to health concerns or discrepancies between studies, 32 papers were selected for analysis, encompassing data for 38 CLA-producing bacteria, such as Bifidobacterium and Lactobacillus species. The information was analyzed by a bioinformatics food recommendation system patented by our research group, Phymofood (EP22382095). This paper presents a new microbiome-based precision nutrition approach targeting CLA-producing gut bacterial species to maximize the anticarcinogenic effect of CLA in CRC.
Collapse
Affiliation(s)
- Adriana González
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country UPV/EHU, Bilbao, Spain
| | - Asier Fullaondo
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country UPV/EHU, Bilbao, Spain
| | - Javier Rodríguez
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Cristina Tirnauca
- Departamento de Matemáticas, Estadística y Computación, Universidad de Cantabria, Santander, Spain
| | - Iñaki Odriozola
- Health Department of Basque Government, Gipuzkoa, Donostia-San Sebastián, Spain
| | - Adrian Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country UPV/EHU, Bilbao, Spain
| |
Collapse
|
4
|
Chen Q, Shi J, Yu G, Xie H, Yu S, Xu J, Liu J, Sun J. Gut microbiota dysbiosis in patients with Alzheimer's disease and correlation with multiple cognitive domains. Front Aging Neurosci 2024; 16:1478557. [PMID: 39665039 PMCID: PMC11632125 DOI: 10.3389/fnagi.2024.1478557] [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: 08/10/2024] [Accepted: 10/31/2024] [Indexed: 12/13/2024] Open
Abstract
Background Accumulating evidence suggested that Alzheimer's disease (AD) was associated with altered gut microbiota. However, the relationships between gut microbiota and specific cognitive domains of AD patients have yet been fully elucidated. The aim of this study was to explore microbial signatures associated with global cognition and specific cognitive domains in AD patients and to determine their predictive value as biomarkers. Methods A total of 64 subjects (18 mild AD, 23 severe AD and 23 healthy control) were recruited in the study. 16 s rDNA sequencing was performed for the gut bacteria composition, followed by liquid chromatography electrospray ionization tandem mass spectrometry (LC/MS/MS) analysis of short-chain fatty acids (SCFAs). The global cognition, specific cognitive domains (abstraction, orientation, attention, language, etc.) and severity of cognitive impairment, were evaluated by Montreal Cognitive Assessment (MoCA) scores. We further identified characteristic bacteria and SCFAs, and receiver operating characteristic (ROC) curve was used to determine the predictive value. Results Our results showed that the microbiota dysbiosis index was significantly higher in the severe and mild AD patients compared to the healthy control (HC). Linear discriminant analysis (LDA) showed that 12 families and 17 genera were identified as key microbiota among three groups. The abundance of Butyricicoccus was positively associated with abstraction, and the abundance of Lachnospiraceae_UCG-004 was positively associated with attention, language, orientation in AD patients. Moreover, the levels of isobutyric acid and isovaleric acid were both significantly negatively correlated with abstraction, and level of propanoic acid was significantly positively associated with the attention. In addition, ROC models based on the characteristic bacteria Lactobacillus, Butyricicoccus and Lachnospiraceae_UCG-004 could effectively distinguished between low and high orientation in AD patients (area under curve is 0.891), and Butyricicoccus and Agathobacter or the combination of SCFAs could distinguish abstraction in AD patients (area under curve is 0.797 and 0.839 respectively). Conclusion These findings revealed the signatures gut bacteria and metabolite SCFAs of AD patients and demonstrated the correlations between theses characteristic bacteria and SCFAs and specific cognitive domains, highlighting their potential value in early detection, monitoring, and intervention strategies for AD patients.
Collapse
Affiliation(s)
- Qionglei Chen
- Department of Geriatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiayu Shi
- Department of Geriatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gaojie Yu
- Department of Geriatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huijia Xie
- Department of Geriatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shicheng Yu
- Department of Geriatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jin Xu
- Department of Geriatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiaming Liu
- Department of Preventive Medicine, School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Jing Sun
- Department of Geriatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
5
|
Ferri I, Canala B, Rossi L. Unravelling the Role of Chitin and Chitosan in Prebiotic Activity and Correlation With Cancer: A Narrative Review. Nutr Rev 2024:nuae168. [PMID: 39530850 DOI: 10.1093/nutrit/nuae168] [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] [Indexed: 11/16/2024] Open
Abstract
This review describes the state of the art regarding the prebiotic role of chitin and the interactions of chitin and chitosan with cancer cells. Chitin is the second most abundant polysaccharide in nature and a constitutive component of crustacean shells and the exoskeleton of insects. Chitosan is the deacetylated form of chitin, which is obtained by chemical processing or the enzymatic activity of deacetylases found in microorganisms and insects. Edible insects have recently been introduced in Western countries, thus raising concerns regarding food safety and due to their chitin content and the release of chitosan during the digestive process. The roles of insect chitin and chitosan in the gastrointestinal tract, microbiome modulation, and cancer have been widely investigated. Several in vitro and in vivo studies have shown the possible microbiota modulation of chitin and its relevant communication with the immune system, thus confirming its prebiotic activity. No evidence has been provided on the cancerogenic activity of chitin; however, studies have suggested that chitin has a cytotoxic effect on cancer cell lines. Chitosan has been confirmed to exhibit apoptotic and cytotoxic activities on cancer cells in several in vitro studies on cancer cell lines and in vivo models. In conclusion, the literature does not show a direct connection between the presence of chitin or chitosan and the onset of cancer. However, cytotoxic and apoptotic activities in relation to cancerous lines have been demonstrated.
Collapse
Affiliation(s)
- Irene Ferri
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Lodi 26900, Italy
| | - Benedetta Canala
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Lodi 26900, Italy
| | - Luciana Rossi
- Department of Veterinary Medicine and Animal Sciences (DIVAS), University of Milan, Lodi 26900, Italy
| |
Collapse
|
6
|
Seethaler B, Basrai M, Neyrinck AM, Vetter W, Delzenne NM, Kiechle M, Bischoff SC. Effect of the Mediterranean diet on the faecal long-chain fatty acid composition and intestinal barrier integrity: an exploratory analysis of the randomised controlled LIBRE trial. Br J Nutr 2024; 132:1-9. [PMID: 39431570 PMCID: PMC11617107 DOI: 10.1017/s0007114524001788] [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: 03/13/2024] [Revised: 07/23/2024] [Accepted: 08/16/2024] [Indexed: 10/22/2024]
Abstract
We recently showed that adherence to the Mediterranean diet increased the proportion of plasma n-3 PUFA, which was associated with an improved intestinal barrier integrity. In the present exploratory analysis, we assessed faecal fatty acids in the same cohort, aiming to investigate possible associations with intestinal barrier integrity. Women from the Lifestyle Intervention Study in Women with Hereditary Breast and Ovarian Cancer (LIBRE) randomised controlled trial, characterised by an impaired intestinal barrier integrity, followed either a Mediterranean diet (intervention group, n 33) or a standard diet (control group, n 35). At baseline (BL), month 3 (V1) and month 12 (V2), plasma lipopolysaccharide-binding protein, faecal zonulin and faecal fatty acids were measured. In the intervention group, faecal proportions of palmitoleic acid (16:1, n-7) and arachidonic acid (20:4, n-6) decreased, while the proportion of linoleic acid (18:2, n-6) and α linoleic acid (18:3, n-3) increased (BL-V1 and BL-V2, all P < 0·08). In the control group, faecal proportions of palmitic acid and arachidic acid increased, while the proportion of linoleic acid decreased (BL-V1, all P < 0·05). The decrease in the proportion of palmitoleic acid correlated with the decrease in plasma lipopolysaccharide-binding protein (ΔV1-BL r = 0·72, P < 0·001; ΔV2-BL r = 0·39, P < 0·05) and correlated inversely with adherence to the Mediterranean diet (Mediterranean diet score; ΔV1-BL r = -0·42, P = 0·03; ΔV2-BL r = -0·53, P = 0·005) in the intervention group. Our data show that adherence to the Mediterranean diet induces distinct changes in the faecal fatty acid composition. Furthermore, our data indicate that the faecal proportion of palmitoleic acid, but not faecal n-3 PUFA, is associated with intestinal barrier integrity in the intervention group.
Collapse
Affiliation(s)
- Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Maryam Basrai
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Audrey M. Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Walter Vetter
- Institute of Food Chemistry, University of Hohenheim, Stuttgart, Germany
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Marion Kiechle
- Department of Gynecology, Center for Hereditary Breast and Ovarian Cancer, Klinikum Rechts der Isar, Technical University Munich and Comprehensive Cancer Center Munich, Munich, Germany
| | - Stephan C. Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
7
|
Capirchio L, Rousseaux C, Dubuquoy C, Ouwehand AC, Maquet V, Modica S, Louis E, Desreumaux P, Tack J. A Synbiotic Combining Chitin-Glucan and Lactobacillus acidophilus NCFM Induces a Colonic Molecular Signature Soothing Intestinal Pain and Inflammation in an Animal Model of IBS. Int J Mol Sci 2024; 25:10732. [PMID: 39409061 PMCID: PMC11476380 DOI: 10.3390/ijms251910732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Chitin-glucan (CG) is a new generation of prebiotic. Lactobacillus acidophilus NCFM® (NCFM) is a probiotic with the ability to decrease abdominal pain. We evaluate the functional and molecular gastrointestinal responses to a synbiotic administration combining CG and NCFM in a rat model of long-lasting colon hypersensitivity. The intracolonic pressure was assessed during the 9-week experiment in animals receiving CG in association or not with NCFM and compared to that in Lacticaseibacillus paracasei Lpc-37®-treated animals and control rats receiving tap water. The effects of the synbiotic were evaluated using the Wallace score, the quantification of colon myeloperoxidase (MPO) and the master genes driving analgesia and inflammation. CG 1.5 alone and NCFM 109 colony forming units (CFU) alone similarly decreased the visceral pain sensitivity. Lpc-37 had no significant effect. The best profile of pain perception inhibition was obtained with the combination of CG 1.5 g and NCFM 109 CFU, confirming a synbiotic property. This synbiotic treatment significantly reduced macroscopic colonic lesions and MPO concentrations, and induced master genes involved in analgesia (CB1, CB2, MOR, PPARα), with a downregulation of inflammatory cytokines (IL-1β, TNFα) and an induction of IL-10 and PPARγ. In conclusion, CG 1.5 g + NCFM 109 CFU significantly decreased visceral pain perception and intestinal inflammation through the regulation of master genes.
Collapse
Affiliation(s)
- Lena Capirchio
- Hepato-Gastroenterology Department, Centre Hospitalier Wallonie-Picarde, 7500 Tournai, Belgium;
| | | | | | | | - Véronique Maquet
- KitoZyme SA, Parc Industriel des Hauts Sarts Zone 2, Rue de Milmort 680, 4040 Herstal, Belgium; (V.M.); (S.M.)
| | - Salvatore Modica
- KitoZyme SA, Parc Industriel des Hauts Sarts Zone 2, Rue de Milmort 680, 4040 Herstal, Belgium; (V.M.); (S.M.)
| | - Edouard Louis
- Department of Gastroenterology, Centre Hospitalier Universitaire de Liège, 4000 Liège, Belgium;
| | - Pierre Desreumaux
- U1286—INFINITE—Institute for Translational Research in Inflammation, University Lille, Inserm, CHU Lille, 59000 Lille, France
- Hepato-Gastroenterology Department, Lille University Hospital, 59000 Lille, France
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium;
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
- Rome Foundation, Raleigh, NC 27614, USA
| |
Collapse
|
8
|
Vieujean S, Peyrin-Biroulet L. Pharmacokinetics of S1P receptor modulators in the treatment of ulcerative colitis. Expert Opin Drug Metab Toxicol 2024; 20:881-892. [PMID: 39252206 DOI: 10.1080/17425255.2024.2402931] [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: 04/10/2024] [Revised: 07/17/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Ulcerative colitis is a chronic inflammatory bowel disease, affecting the colorectal mucosae, with a relapsing-remitting course, characterized by the trafficking and gathering of lymphocytes in the inflammatory intestinal mucosa. Sphingosine-1-phosphate (S1P) receptor modulators preventing lymphocytes egress from lymphoid tissues to the active inflammation site is an alternative therapeutic option in this condition. AREA COVERED We carried out a comprehensive review of the literature available on Medline, Scopus and Embase regarding the pharmacokinetics of S1P receptor modulators. For each compound, we reviewed the mechanism of action, pharmacokinetic data and efficacy and safety data from phase 3 studies and real-life studies when available. EXPERT OPINION S1P receptor modulators, including ozanimod and etrasimod (both currently on the market) as well as VTX002 (under development), are a new class of drugs for the treatment of moderate to severe ulcerative colitis, inducing and maintaining the remission. Due to its pharmacokinetic features, this class of drugs has certain advantages such as an oral administration, a short half-life, a high volume of distribution, and no immunogenicity. On the other hand, there are risks of cardiological and ophthalmological side-effects, as well as drug-drug interactions risk, that require special attention from the healthcare providers.
Collapse
Affiliation(s)
- Sophie Vieujean
- Hepato-Gastroenterology and Digestive Oncology, University Hospital CHU of Liège, Liège, Belgium
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, INFINY Institute, INSERM NGERE, CHRU Nancy, Vandœuvre-lès-Nancy, France
- Groupe Hospitalier privé Ambroise Paré - Hartmann, Paris IBD center, Neuilly sur Seine, France
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Wang L, Jin S, Wang P, Li X, Liu C, Sun S, Zhang G, Chang J, Yin Q, Zhang H, Zhu Q. Fermented total mixed ration enhances nutrient digestibility and modulates the milk components and fecal microbial community in lactating Holstein dairy cows. Front Vet Sci 2024; 11:1408348. [PMID: 39205803 PMCID: PMC11350115 DOI: 10.3389/fvets.2024.1408348] [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: 03/28/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
Fermented total mixed ration (FTMR) is an effective method of preserving high-moisture byproducts with higher aerobic stability after fermentation. FTMR has the potential to fulfill the daily nutritional requirements of cattle and enhance their production performance. The objective of this research was to examine the influence of FTMR on lactation performance, total tract apparent digestibility, fecal microbiota communities, and fermentation profiles in lactating dairy cows. A total of 12 cows were randomly assigned into two groups: the TMR group and the FTMR group. The TMR group was fed a total mixed ration (TMR) diet, and the FTMR group was fed an FTMR diet. The FTMR did not impact milk yield in dairy cows despite a decrease in dry matter intake, which increased the efficiency of the feed. In contrast to that in the TMR group, the milk fat content in the FTMR group was greater. The FTMR group showed greater digestibility of neutral detergent fiber (NDF), organic matter (OM), dry matter (DM), crude protein (CP), and acid detergent fiber (ADF) in the total digestive tract than did the TMR group. The FTMR increased the concentration of butyrate in the fecal matter and reduced the pH of the feces. The Chao1, ACE, and Shannon indices of the archaeal community in dairy cow feces were significantly higher in cow fed the FTMR compared to those fed the TMR. LefSe analysis revealed higher levels of Oscillospira, Lactobacillus, Prevotella, and Dehalobacterium in the feces of dairy cows fed the FTMR than in those fed the TMR. However, the abundances of Roseburia, rc4-4, Bulleidia and Sharpea exhibited the opposite trend. The abundances of Halobacteria, Halobacteriales, and Halobacteriaceae, which are biomarkers for distinguishing fecal archaea in the TMR from the FTMR, were substantially greater in the feces of dairy cows that consumed the TMR than in those that consumed the FTMR. Therefore, FTMR can improve the milk fat content, total tract apparent feed digestibility efficiency, and diversity of archaea in the feces. Additionally, this work provides a theoretical basis for the feasibility of FTMR feeding for dairy cows.
Collapse
Affiliation(s)
- Lijun Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Sanjun Jin
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Ping Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Xinxin Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Chaoqi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Siying Sun
- Institute of International Education, Henan Agricultural University, Zhengzhou, China
| | - Guangning Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Juan Chang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Qingqiang Yin
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Haiyang Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Qun Zhu
- Henan Delin Biological Products Co., Ltd., Xinxiang, China
| |
Collapse
|
10
|
Talbodec N, Le Roy P, Fournier P, Lesage B, Lepoutre E, Castex F, Godchaux JM, Vandeville L, Bismuth B, Lesage X, Bayart P, Genin M, Rousseaux C, Maquet V, Modica S, Desreumaux P, Valibouze C. Efficacy and tolerability of chitin-glucan combined with simethicone (GASTRAP ® DIRECT) in irritable bowel syndrome: A prospective, open-label, multicenter study. World J Gastrointest Pharmacol Ther 2024; 15:90757. [PMID: 38846967 PMCID: PMC11151880 DOI: 10.4292/wjgpt.v15.i3.90757] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/19/2024] [Accepted: 05/08/2024] [Indexed: 05/27/2024] Open
Abstract
BACKGROUND Irritable bowel syndrome (IBS), defined according to the Rome IV diagnostic criteria, is a chronic functional gastrointestinal disorder characterized by recurrent abdominal pain related to altered bowel habits. First-line recommended treatments are limited to combining drugs targeting predominant symptoms, particularly pain (antispasmodics), constipation (laxatives), and diarrhea (loperamide), yielding only a limited therapeutic gain. GASTRAP® DIRECT is a class IIa medical formulation composed of a combination of chitin-glucan and simethicone indicated for the symptomatic treatment of gas-related gastrointestinal disorders by combining different mechanisms of action. AIM To evaluate the efficacy, tolerability, and safety of 4-week GASTRAP® DIRECT treatment in patients with IBS. METHODS In this prospective, multicenter, open-label trial, 120 patients with IBS received three sticks of GASTRAP® DIRECT (1.5 g/d of chitin-glucan and 0.75 mg/d of simethicone) per day for 4 weeks. The primary endpoint was the responder rate, defined as the number of patients whose abdominal pain score decreased by ≥ 30% from baseline to week (W) 4. The analysis was performed using the per-protocol set. Cardinal symptoms, impact of global symptoms on daily life, change in stool consistency, and improvement in defecatory disorders were evaluated. RESULTS Overall, 100 patients were evaluated. At W4, 67% (95%CI: 57-75) showed improvement in abdominal pain (score: 5.8 ± 2.4 vs 2.9 ± 2.0, P < 0.0001). Similar improvements were observed for bloating [8.0 ± 1.7 vs 4.7 ± 2.9, P < 0.0001; 60% (95%CI: 50-70) responders], abdominal distension [7.2 ± 2.1 vs 4.4 ± 3.1, P < 0.0001; 53% (95%CI: 43-63) responders], and impact of global symptoms on daily life [7.1 ± 2.0 vs 4.6 ± 2.9, P < 0.0001; 54% (95%CI: 44-64) responders]. Stool consistency improved in most patients (90% and 57% for patients with liquid and hard stools, respectively). Overall, 42% of patients with defecatory disorders reported very much/considerable improvements by W2. No severe adverse event occurred, and tolerability was rated "good" or "very good" by 93% of patients. CONCLUSION GASTRAP® DIRECT is safe and well tolerated, alleviating IBS symptoms rapidly in 2 weeks. This open-label study suggests that the combination of chitin-glucan and simethicone could be beneficial in patients with IBS.
Collapse
Affiliation(s)
- Nathalie Talbodec
- Department of Gastroenterology, Hôpital privé Le Bois, Lille 59000, France
| | - Pauline Le Roy
- Department of Gastroenterology, Hôpital privé de Villeneuve d’Ascq, Villeneuve d’Ascq 59650, France
| | - Peggy Fournier
- Department of Gastroenterology, Hôpital privé Le Bois, Lille 59000, France
| | - Benoit Lesage
- Department of Gastroenterology, Hôpital privé Le Bois, Lille 59000, France
| | - Elodie Lepoutre
- Department of Gastroenterology, Hôpital privé Le Bois, Lille 59000, France
| | - François Castex
- Department of Gastroenterology, Hôpital privé de Villeneuve d’Ascq, Villeneuve d’Ascq 59650, France
| | - Jean Michel Godchaux
- Department of Gastroenterology, Hôpital privé de Villeneuve d’Ascq, Villeneuve d’Ascq 59650, France
| | - Lionel Vandeville
- Department of Gastroenterology, Hôpital privé Le Bois, Lille 59000, France
| | - Benjamin Bismuth
- Department of Gastroenterology, Hôpital privé Le Bois, Lille 59000, France
| | - Xavier Lesage
- Department of Gastroenterology, Hôpital privé Le Bois, Lille 59000, France
| | - Pauline Bayart
- Department of Gastroenterology, Hôpital privé Le Bois, Lille 59000, France
| | - Michael Genin
- Univ. Lille, CHU Lille, ULR 2694–METRICS, Évaluation des Technologies de Santé et des Pratiques Médicales, Lille 59000, France
| | - Christel Rousseaux
- Development of Intestinal Biotech, 1 Avenue Oscar Lambret, Lille 59045, France
| | - Veronique Maquet
- KitoZyme SA, Parc Industriel des hauts Sarts Zone 2, Rue de Milmort, Herstal 4040, Belgium
| | - Salvatore Modica
- BiOkuris A, Parc Industriel des hauts Sarts Zone 2, Rue de Milmort, Herstal 4040, Belgium
| | - Pierre Desreumaux
- Department of Hepato-Gastroenterology, Lille University Hospital, Lille 59000, France
- U1286-INFINITE, Institute for Translational Research in Inflammation, Univ. Lille, Inserm, CHU Lille, Lille 59000, France
| | - Caroline Valibouze
- Department of Digestive Surgery and Transplantation, Lille University Hospital, Lille 59037, France
- U1286-INFINITE, Institute for Translational Research in Inflammation, Univ. Lille, Inserm, CHU Lille, Lille 59000, France
| |
Collapse
|
11
|
Valibouze C, Dubuquoy C, Chavatte P, Genin M, Maquet V, Modica S, Desreumaux P, Rousseaux C. Chitin-glucan improves important pathophysiological features of irritable bowel syndrome. World J Gastroenterol 2024; 30:2258-2271. [PMID: 38690023 PMCID: PMC11056916 DOI: 10.3748/wjg.v30.i16.2258] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/21/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is one of the most frequent and debilitating conditions leading to gastroenterological referrals. However, recommended treatments remain limited, yielding only limited therapeutic gains. Chitin-glucan (CG) is a novel dietary prebiotic classically used in humans at a dosage of 1.5-3.0 g/d and is considered a safe food ingredient by the European Food Safety Authority. To provide an alternative approach to managing patients with IBS, we performed preclinical molecular, cellular, and animal studies to evaluate the role of chitin-glucan in the main pathophysiological mechanisms involved in IBS. AIM To evaluate the roles of CG in visceral analgesia, intestinal inflammation, barrier function, and to develop computational molecular models. METHODS Visceral pain was recorded through colorectal distension (CRD) in a model of long-lasting colon hypersensitivity induced by an intra-rectal administration of TNBS [15 milligrams (mg)/kilogram (kg)] in 33 Sprague-Dawley rats. Intracolonic pressure was regularly assessed during the 9 wk-experiment (weeks 0, 3, 5, and 7) in animals receiving CG (n = 14) at a human equivalent dose (HED) of 1.5 g/d or 3.0 g/d and compared to negative control (tap water, n = 11) and positive control (phloroglucinol at 1.5 g/d HED, n = 8) groups. The anti-inflammatory effect of CG was evaluated using clinical and histological scores in 30 C57bl6 male mice with colitis induced by dextran sodium sulfate (DSS) administered in their drinking water during 14 d. HT-29 cells under basal conditions and after stimulation with lipopolysaccharide (LPS) were treated with CG to evaluate changes in pathways related to analgesia (µ-opioid receptor (MOR), cannabinoid receptor 2 (CB2), peroxisome proliferator-activated receptor alpha, inflammation [interleukin (IL)-10, IL-1b, and IL-8] and barrier function [mucin 2-5AC, claudin-2, zonula occludens (ZO)-1, ZO-2] using the real-time PCR method. Molecular modelling of CG, LPS, lipoteichoic acid (LTA), and phospholipomannan (PLM) was developed, and the ability of CG to chelate microbial pathogenic lipids was evaluated by docking and molecular dynamics simulations. Data were expressed as the mean ± SEM. RESULTS Daily CG orally-administered to rats or mice was well tolerated without including diarrhea, visceral hypersensitivity, or inflammation, as evaluated at histological and molecular levels. In a model of CRD, CG at a dosage of 3 g/d HED significantly decreased visceral pain perception by 14% after 2 wk of administration (P < 0.01) and reduced inflammation intensity by 50%, resulting in complete regeneration of the colonic mucosa in mice with DSS-induced colitis. To better reproduce the characteristics of visceral pain in patients with IBS, we then measured the therapeutic impact of CG in rats with TNBS-induced inflammation to long-lasting visceral hypersensitivity. CG at a dosage of 1.5 g/d HED decreased visceral pain perception by 20% five weeks after colitis induction (P < 0.01). When the CG dosage was increased to 3.0 g/d HED, this analgesic effect surpassed that of the spasmolytic agent phloroglucinol, manifesting more rapidly within 3 wk and leading to a 50% inhibition of pain perception (P < 0.0001). The underlying molecular mechanisms contributing to these analgesic and anti-inflammatory effects of CG involved, at least in part, a significant induction of MOR, CB2 receptor, and IL-10, as well as a significant decrease in pro-inflammatory cytokines IL-1b and IL-8. CG also significantly upregulated barrier-related genes including muc5AC, claudin-2, and ZO-2. Molecular modelling of CG revealed a new property of the molecule as a chelator of microbial pathogenic lipids, sequestering gram-negative LPS and gram-positive LTA bacterial toxins, as well as PLM in fungi at the lowesr energy conformations. CONCLUSION CG decreased visceral perception and intestinal inflammation through master gene regulation and direct binding of microbial products, suggesting that CG may constitute a new therapeutic strategy for patients with IBS or IBS-like symptoms.
Collapse
Affiliation(s)
- Caroline Valibouze
- Department of Digestive Surgery and Transplantation, Lille University, Lille 59037, France
| | - Caroline Dubuquoy
- Intestinal Biotech Development, Faculté de Médicine, Lille 59045, France
| | - Philippe Chavatte
- U1286-INFINITE-Institute for Translational Research in Inflammation, Université de Lille, Lille 59000, France
| | - Michaël Genin
- ULR 2694-METRICS, Évaluation des Technologies de santé et des Pratiques Médicales, University of Lille, Lille 59000, France
| | - Veronique Maquet
- KitoZyme SA, Institution Société Anonyme, Zone 2, Parc des Hauts Sarts, Rue de Milmort, Herstal 4040, Belgium
| | - Salvatore Modica
- KitoZyme SA, Institution Société Anonyme, Zone 2, Parc des Hauts Sarts, Rue de Milmort, Herstal 4040, Belgium
| | - Pierre Desreumaux
- Hepato-Gastroenterology Department, Lille University Hospital, Lille 59037, France
| | - Christel Rousseaux
- Intestinal Biotech Development, Faculté de Médicine, Lille 59045, France
| |
Collapse
|
12
|
Fang M, Liu W, Wang Z, Li J, Hu S, Li Z, Chen W, Zhang N. Causal associations between gut microbiota with intervertebral disk degeneration, low back pain, and sciatica: a Mendelian randomization study. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2024; 33:1424-1439. [PMID: 38285276 DOI: 10.1007/s00586-024-08131-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 11/30/2023] [Accepted: 01/03/2024] [Indexed: 01/30/2024]
Abstract
PURPOSE Although studies have suggested that gut microbiota may be associated with intervertebral disk disease, their causal relationship is unclear. This study aimed to investigate the causal relationship between the gut microbiota and its metabolic pathways with the risk of intervertebral disk degeneration (IVDD), low back pain (LBP), and sciatica. METHODS Genetic variation data for 211 gut microbiota taxa at the phylum to genus level were obtained from the MiBioGen consortium. Genetic variation data for 105 taxa at the species level and 205 metabolic pathways were obtained from the Dutch Microbiome Project. Genetic variation data for disease outcomes were obtained from the FinnGen consortium. The causal relationships between the gut microbiota and its metabolic pathways and the risk of IVDD, LBP, and sciatica were evaluated via Mendelian randomization (MR). The robustness of the results was assessed through sensitivity analysis. RESULTS Inverse variance weighting identified 46 taxa and 33 metabolic pathways that were causally related to IVDD, LBP, and sciatica. After correction by weighted median and MR-PRESSO, 15 taxa and nine pathways remained stable. After FDR correction, only the effect of the genus_Eubacterium coprostanoligenes group on IVDD remained stable. Sensitivity analyses showed no evidence of horizontal pleiotropy, heterogeneity, or reverse causation. CONCLUSION Some microbial taxa and their metabolic pathways are causally related to IVDD, LBP, and sciatica and may serve as potential intervention targets. This study provides new insights into the mechanisms of gut microbiota-mediated development of intervertebral disk disease.
Collapse
Affiliation(s)
- Miaojie Fang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, 310009, Hangzhou, People's Republic of China
- Department of Orthopedics Surgery, International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, N1 Shangcheng Road, Yiwu, Zhejiang, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Wei Liu
- Department of Orthopedics Surgery, International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, N1 Shangcheng Road, Yiwu, Zhejiang, People's Republic of China
| | - Zhan Wang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, 310009, Hangzhou, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Jun Li
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, 310009, Hangzhou, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Shaojun Hu
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, 310009, Hangzhou, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Zilong Li
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, 310009, Hangzhou, People's Republic of China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Weishan Chen
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, 310009, Hangzhou, People's Republic of China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China.
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China.
| | - Ning Zhang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, 310009, Hangzhou, People's Republic of China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China.
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
13
|
Yin P, Yi S, Du T, Zhang C, Yu L, Tian F, Zhao J, Chen W, Zhai Q. Dynamic response of different types of gut microbiota to fructooligosaccharides and inulin. Food Funct 2024; 15:1402-1416. [PMID: 38214586 DOI: 10.1039/d3fo04855a] [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: 01/13/2024]
Abstract
Fructooligosaccharides (FOS) and inulin are beneficial for human health. However, their benefits differ in individuals who consume prebiotics. Several factors contribute to this variation, including host genetics and differences in the gut microbiota. Bifidobacterium and Bacteroides are strong carbohydrate-utilizing bacteria in the gut, and the level of the Bacteroides/Bifidobacterium (Ba/Bi) ratio in the gut is closely related to the body's ability to utilize prebiotics. However, how to select the type of prebiotics more beneficial for populations with specific Ba/Bi backgrounds and the underlying regulatory mechanisms remain unclear. Here, we explored the dynamics of the gut microbiota and metabolic functions during the in vitro fermentation of FOS and inulin in two different groups: Bacteroides/Bifidobacterium high (H) and Bacteroides/Bifidobacterium low (L). This study revealed that the baseline Ba/Bi ratio had a greater impact on the gut microbiota compared to prebiotic species. Noticeable differences were observed between the two groups after prebiotic intervention, with the H group being more likely to benefit from the prebiotic intervention. Compared to the L group, the H group exhibited significantly higher microbial α-diversity; the co-abundance response group 1 (CARG1) members Ruminococcus gnavus and Blautia involved in the synthesis of propionic and butyric acids increased significantly, the abundance of pathogenic bacteria such as Escherichia Shigella decreased significantly, and the ability to degrade carbohydrates and synthesize fatty acids was greater. Regression modeling showed that the key microbiota could predict the short-chain fatty acid (SCFA) levels, with FOS associated with the ecological roles of CARG2 and CARG7 and inulin associated with CARG4, which provides the basis for the use of prebiotics in nutritional applications and the stratification of populations based on pertinent microbiota profiles to explain the incongruent health effects in human intervention studies.
Collapse
Affiliation(s)
- Pingping Yin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Shanrong Yi
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Ting Du
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
14
|
Kawasaki K, Zhao J, Takao N, Sato M, Ban T, Tamamaki K, Kagami M, Yano K. Sustenance Trial to Analyze the Effects of Black Soldier Fly Larvae Meal on the Reproductive Efficiency of Sows and the Hematological Properties of Suckling and Weaning Piglets. Animals (Basel) 2023; 13:3410. [PMID: 37958165 PMCID: PMC10647472 DOI: 10.3390/ani13213410] [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: 10/04/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
The escalating demand for meat, driven by global population growth, necessitates sustainable solutions for animal feed production. This study investigated the effects of substituting conventional protein resources in sow and piglet dietary regimens with black soldier fly (BSF; Hermetia illucens) meal on reproductive efficiency, blood profile, piglet growth, and intestinal tissue morphology. The results indicate that substituting animal-derived and soy proteins with BSF meal does not compromise sow reproductive performance. Although no notable disparities were observed in piglet growth, the feed conversion ratio from the 28- to 35-day age marks were lower in the BSF-fed groups. This suggests that the animal protein-BSF substitution rate may require optimization, potentially involving chitin removal from BSF meal to enhance digestibility. Minor variations in the hematological composition and properties in piglets, with elevated high-density lipoprotein cholesterol levels in the high BSF group at the 28-day mark, were potentially attributable to the unique fatty acid composition of BSF meal. Moreover, this study potentiates future exploration into the efficacy of complete animal protein substitution with BSF meals on piglet nutrition and physiology, particularly in fattening pigs. The practical implementation of BSF meals in animal feed production holds promise for enhancing the sustainability of the swine industry.
Collapse
Affiliation(s)
- Kiyonori Kawasaki
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Ikenobe 2393, Miki-cho, Kita-gun, Kagawa 761-0795, Japan; (J.Z.)
| | - Junliang Zhao
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Ikenobe 2393, Miki-cho, Kita-gun, Kagawa 761-0795, Japan; (J.Z.)
| | - Natsu Takao
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Ikenobe 2393, Miki-cho, Kita-gun, Kagawa 761-0795, Japan; (J.Z.)
| | - Masaki Sato
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Ikenobe 2393, Miki-cho, Kita-gun, Kagawa 761-0795, Japan; (J.Z.)
| | - Takuma Ban
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Ikenobe 2393, Miki-cho, Kita-gun, Kagawa 761-0795, Japan; (J.Z.)
| | - Kaoru Tamamaki
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Ikenobe 2393, Miki-cho, Kita-gun, Kagawa 761-0795, Japan; (J.Z.)
| | - Masanori Kagami
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Ikenobe 2393, Miki-cho, Kita-gun, Kagawa 761-0795, Japan; (J.Z.)
| | - Kiminobu Yano
- University Farm, Kagawa University, Showa 300-2, Sanuki, Kagawa 769-2304, Japan
| |
Collapse
|
15
|
Hu J, Yang X, Ren J, Zhong S, Fan Q, Li W. Identification and verification of characteristic differentially expressed ferroptosis-related genes in osteosarcoma using bioinformatics analysis. Toxicol Mech Methods 2023; 33:781-795. [PMID: 37488941 DOI: 10.1080/15376516.2023.2240879] [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: 05/17/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND This study identified and verified the characteristic differentially expressed ferroptosis-related genes (CDEFRGs) in osteosarcoma (OS). METHODS We extracted ferroptosis-related genes (FRGs), identified differentially expressed FRGs (DEFRGs) in OS, and conducted correlation analysis between DEFRGs. Next, we conducted GO and KEGG analyses to explore the biological functions and pathways of DEFRGs. Furthermore, we used LASSO and SVM-RFE algorithms to screen CDEFRGs, and evaluated its accuracy in diagnosing OS through ROC curves. Then, we demonstrated the molecular function and pathway enrichment of CDEFRGs through GSEA analysis. In addition, we evaluated the differences in immune cell infiltration between OS and NC groups, as well as the correlation between CDEFRGs expressions and immune cell infiltrations. Finally, the expression of CDEFRGs was verified through qRT-PCR, western blotting, and immunohistochemistry experiments. RESULTS We identified 51 DEFRGs and the expression relationship between them. GO and KEGG analysis revealed their key functions and important pathways. Based on four CDEFRGs (PEX3, CPEB1, NOX1, and ALOX5), we built the OS diagnostic model, and verified its accuracy. GSEA analysis further revealed the important functions and pathways of CDEFRGs. In addition, there were differences in immune cell infiltration between OS group and NC group, and CDEFRGs showed significant correlation with certain infiltrating immune cells. Finally, we validated the differential expression levels of four CDEFRGs through external experiments. CONCLUSIONS This study has shed light on the molecular pathological mechanism of OS and has offered novel perspectives for the early diagnosis and immune-targeted therapy of OS patients.
Collapse
Affiliation(s)
- Jianhua Hu
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, P. R. China
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, P. R. China
| | - Xi Yang
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, P. R. China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, P. R. China
| | - Jing Ren
- Department of Spinal Surgery, Qujing No. 1 Hospital, Affiliated Qujing Hospital of Kunming Medical University, Qujing, P. R. China
| | - Shixiao Zhong
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, P. R. China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, P. R. China
| | - Qianbo Fan
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, P. R. China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, P. R. China
| | - Weichao Li
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, P. R. China
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, P. R. China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, P. R. China
| |
Collapse
|
16
|
Cantu-Jungles TM, Hamaker BR. Tuning Expectations to Reality: Don't Expect Increased Gut Microbiota Diversity with Dietary Fiber. J Nutr 2023; 153:3156-3163. [PMID: 37690780 DOI: 10.1016/j.tjnut.2023.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023] Open
Abstract
Dietary approaches, particularly those including fiber supplementation, can be used to promote health benefits by shaping gut microbial communities. Whereas community diversity measures, such as richness and evenness, are often used in microbial ecology to make sense of these complex and vast microbial ecosystems, it is less clear how these concepts apply when dietary fiber supplementation is given. In this perspective, we summarize and demonstrate how factors including experimental approach, number of bacteria sharing a dietary fiber, and initial relative abundances of bacteria that use a fiber can significantly affect diversity outcomes in fiber fermentation studies. We also show that a reduction in alpha diversity is possible, and perhaps expected, for most approaches that use fermentable fibers to beneficially shape the gut microbial community while still achieving health-related improvements.
Collapse
Affiliation(s)
- Thaisa M Cantu-Jungles
- Department of Food Science, Whistler Center for Carbohydrate Research, Purdue University, West Lafayette, IN, United States.
| | - Bruce R Hamaker
- Department of Food Science, Whistler Center for Carbohydrate Research, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
17
|
Seethaler B, Lehnert K, Yahiaoui-Doktor M, Basrai M, Vetter W, Kiechle M, Bischoff SC. Omega-3 polyunsaturated fatty acids improve intestinal barrier integrity-albeit to a lesser degree than short-chain fatty acids: an exploratory analysis of the randomized controlled LIBRE trial. Eur J Nutr 2023; 62:2779-2791. [PMID: 37318580 PMCID: PMC10468946 DOI: 10.1007/s00394-023-03172-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 05/04/2023] [Indexed: 06/16/2023]
Abstract
PURPOSE Adherence to the Mediterranean diet is associated with beneficial health effects, including gastrointestinal disorders. Preclinical studies suggest that omega-3 polyunsaturated fatty acids (n-3 PUFAs), found in Mediterranean foods like nuts and fish, improve intestinal barrier integrity. Here, we assessed possible effects of n-3 PUFAs on barrier integrity in a randomized controlled trial. METHODS We studied 68 women from the open-label LIBRE trial (clinicaltrials.gov: NCT02087592) who followed either a Mediterranean diet (intervention group, IG) or a standard diet (control group, CG). Study visits comprised baseline, month 3, and month 12. Barrier integrity was assessed by plasma lipopolysaccharide binding protein (LBP) and fecal zonulin; fatty acids by gas chromatography with mass spectrometry. Median and interquartile ranges are shown. RESULTS Adherence to the Mediterranean diet increased the proportion of the n-3 docosahexaenoic acid (DHA) (IG + 1.5% [0.9;2.5, p < 0.001]/ + 0.3% [- 0.1;0.9, p < 0.050] after 3/12 months; CG + 0.9% [0.5;1.6, p < 0.001]/ ± 0%) and decreased plasma LBP (IG - 0.3 µg/ml [- 0.6;0.1, p < 0.010]/ - 0.3 µg/ml [- 1.1; - 0.1, p < 0.001]; CG - 0.2 µg/ml [- 0.8; - 0.1, p < 0.001]/ ± 0 µg/ml) and fecal zonulin levels (IG - 76 ng/mg [- 164; - 12, p < 0.010]/ - 74 ng/mg [- 197;15, p < 0.001]; CG - 59 ng/mg [- 186;15, p < 0.050]/ + 10 ng/mg [- 117;24, p > 0.050]). Plasma DHA and LBP (R2: 0.14-0.42; all p < 0.070), as well as plasma DHA and fecal zonulin (R2: 0.18-0.48; all p < 0.050) were found to be inversely associated in bi- and multivariate analyses. Further multivariate analyses showed that the effect of DHA on barrier integrity was less pronounced than the effect of fecal short-chain fatty acids on barrier integrity. CONCLUSIONS Our data show that n-3 PUFAs can improve intestinal barrier integrity. TRIAL REGISTRATION NUMBER The trial was registered prospectively at ClinicalTrials.gov (reference: NCT02087592).
Collapse
Affiliation(s)
- Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Fruwirthstr. 12, 70593, Stuttgart, Germany
| | - Katja Lehnert
- Institute of Food Chemistry, University of Hohenheim, Stuttgart, Germany
| | - Maryam Yahiaoui-Doktor
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
| | - Maryam Basrai
- Institute of Nutritional Medicine, University of Hohenheim, Fruwirthstr. 12, 70593, Stuttgart, Germany
| | - Walter Vetter
- Institute of Food Chemistry, University of Hohenheim, Stuttgart, Germany
| | - Marion Kiechle
- Department of Gynecology, Center for Hereditary Breast and Ovarian Cancer, Klinikum Rechts der Isar, Technical University Munich and Comprehensive Cancer Center Munich, Munich, Germany
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Fruwirthstr. 12, 70593, Stuttgart, Germany.
| |
Collapse
|
18
|
Meiller L, Sauvinet V, Breyton AE, Ranaivo H, Machon C, Mialon A, Meynier A, Bischoff SC, Walter J, Neyrinck AM, Laville M, Delzenne NM, Vinoy S, Nazare JA. Metabolic signature of 13C-labeled wheat bran consumption related to gut fermentation in humans: a pilot study. Eur J Nutr 2023; 62:2633-2648. [PMID: 37222787 DOI: 10.1007/s00394-023-03161-5] [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: 11/07/2022] [Accepted: 04/18/2023] [Indexed: 05/25/2023]
Abstract
PURPOSE The aim of this pilot study was to analyze concomitantly the kinetics of production of 13C-labeled gut-derived metabolites from 13C-labeled wheat bran in three biological matrices (breath, plasma, stools), in order to assess differential fermentation profiles among subjects. METHODS Six healthy women consumed a controlled breakfast containing 13C-labeled wheat bran biscuits. H2, CH4 and 13CO2, 13CH4 24 h-concentrations in breath were measured, respectively, by gas chromatography (GC) and GC-isotope ratio mass spectrometry (GC-IRMS). Plasma and fecal concentrations of 13C-short-chain fatty acids (linear SCFAs: acetate, propionate, butyrate, valerate; branched SCFAs: isobutyrate, isovalerate) were quantified using GC-combustion-IRMS. Gut microbiota composition was assessed by16S rRNA gene sequencing analysis. RESULTS H2 and CH4 24 h-kinetics distinguished two groups in terms of fermentation-related gas excretion: high-CH4 producers vs low-CH4 producers (fasting concentrations: 45.3 ± 13.6 ppm vs 6.5 ± 3.6 ppm). Expired 13CH4 was enhanced and prolonged in high-CH4 producers compared to low-CH4 producers. The proportion of plasma and stool 13C-butyrate tended to be higher in low-CH4 producers, and inversely for 13C-acetate. Plasma branched SCFAs revealed different kinetics of apparition compared to linear SCFAs. CONCLUSION This pilot study allowed to consider novel procedures for the development of biomarkers revealing dietary fiber-gut microbiota interactions. The non-invasive assessment of exhaled gas following 13C-labeled fibers ingestion enabled to decipher distinct fermentation profiles: high-CH4 producers vs low-CH4 producers. The isotope labeling permits a specific in vivo characterisation of the dietary fiber impact consumption on microbiota metabolite production. CLINICAL TRIAL REGISTRATION The study has been registered under the number NCT03717311 at ClinicalTrials.gov on October 24, 2018.
Collapse
Affiliation(s)
- Laure Meiller
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Valérie Sauvinet
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Anne-Esther Breyton
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
- CarMeN Laboratory, INSERM, INRAe, Claude Bernard Lyon1 University, Pierre Bénite, France
| | - Harimalala Ranaivo
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
- CarMeN Laboratory, INSERM, INRAe, Claude Bernard Lyon1 University, Pierre Bénite, France
| | - Christelle Machon
- Service de Biochimie et Biologie Moléculaire, Hospices Civils de Lyon, Pierre Bénite, France
| | - Anne Mialon
- Service de Biochimie et Biologie Moléculaire, Hospices Civils de Lyon, Pierre Bénite, France
| | | | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Jens Walter
- Department of Medicine, School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, UCLouvain, Brussels, Belgium
| | - Martine Laville
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
- CarMeN Laboratory, INSERM, INRAe, Claude Bernard Lyon1 University, Pierre Bénite, France
- Service d'Endocrinologie Diabète Nutrition, Hospices Civils de Lyon, Pierre Bénite, France
| | | | - Sophie Vinoy
- Nutrition Research, Mondelez International, Saclay, France
| | - Julie-Anne Nazare
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, INSERM, INRAe, Claude Bernard Lyon1 University, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France.
- CarMeN Laboratory, INSERM, INRAe, Claude Bernard Lyon1 University, Pierre Bénite, France.
| |
Collapse
|
19
|
Yu L, Gao Y, Ye Z, Duan H, Zhao J, Zhang H, Narbad A, Tian F, Zhai Q, Chen W. Interaction of beta-glucans with gut microbiota: Dietary origins, structures, degradation, metabolism, and beneficial function. Crit Rev Food Sci Nutr 2023; 64:9884-9909. [PMID: 37272431 DOI: 10.1080/10408398.2023.2217727] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Beta-glucan (BG), a polysaccharide comprised of interfacing glucose monomers joined via beta-glycosidic linkages, can be defined as a type of dietary fiber with high specificity based on its interaction with the gut microbiota. It can induce similar interindividual microbiota responses, thereby having beneficial effects on the human body. In this paper, we review the four main sources of BG (cereals, fungi, algae, and bacteria) and their differences in structure and content. The interaction of BG with gut microbiota and the resulting health effects have been highlighted, including immune enhancement, regulation of serum cholesterol and insulin levels, alleviation of obesity and improvement of cognitive disorders. Finally, the application of BG in food products and its beneficial effects on the gut microbiota of consumers were discussed. Although some of the mechanisms of action remain unclear, revealing the beneficial functions of BG from the perspective of gut microbiota can help provide theoretical support for the development of diets that target the regulation of microbiota.
Collapse
Affiliation(s)
- Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Yuhang Gao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Zi Ye
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Hui Duan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Arjan Narbad
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- Gut Health and Microbiome Institute Strategic Programme, Quadram Institute Bioscience, Norwich, UK
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
20
|
Stull VJ, Weir TL. Chitin and omega-3 fatty acids in edible insects have underexplored benefits for the gut microbiome and human health. NATURE FOOD 2023; 4:283-287. [PMID: 37117549 DOI: 10.1038/s43016-023-00728-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 03/02/2023] [Indexed: 04/30/2023]
Abstract
A healthy gut microbiome is critical for nutrient metabolism, pathogen inhibition and immune regulation, and is highly influenced by diet. Edible insects are good sources of protein and micronutrients, but unlike other animal-derived foods, they also contain both dietary fibre and omega-3 fatty acids that can modulate gut microbiota. Here we explore the potential impacts of insect consumption on the microbiome. Laboratory, animal and human studies indicate that insect fibre in the form of chitin and its derivatives can modify gut microbiota with beneficial outcomes. Some insects also contain favourable omega-3/omega-6 ratios. We identify gaps in the literature-especially a dearth of human studies-that must be addressed to better understand health impacts of entomophagy. Insects, already eaten across the globe, can be farmed using fewer resources than conventional livestock. Widening the research scope offers an opportunity to advance use of edible insects to address interconnected environmental and health challenges.
Collapse
Affiliation(s)
- Valerie J Stull
- Center for Sustainability and the Global Environment, University of Wisconsin-Madison, Madison, WI, USA.
| | - Tiffany L Weir
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
21
|
Van Hul M, Cani PD. The gut microbiota in obesity and weight management: microbes as friends or foe? Nat Rev Endocrinol 2023; 19:258-271. [PMID: 36650295 DOI: 10.1038/s41574-022-00794-0] [Citation(s) in RCA: 119] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/18/2023]
Abstract
Obesity is caused by a long-term difference between energy intake and expenditure - an imbalance that is seemingly easily restored by increasing exercise and reducing caloric consumption. However, as simple as this solution appears, for many people, losing excess weight is difficult to achieve and even more difficult to maintain. The reason for this difficulty is that energy intake and expenditure, and by extension body weight, are regulated through complex hormonal, neural and metabolic mechanisms that are under the influence of many environmental factors and internal responses. Adding to this complexity, the microorganisms (microbes) that comprise the gut microbiota exert direct effects on the digestion, absorption and metabolism of food. Furthermore, the gut microbiota exerts a miscellany of protective, structural and metabolic effects both on the intestinal milieu and peripheral tissues, thus affecting body weight by modulating metabolism, appetite, bile acid metabolism, and the hormonal and immune systems. In this Review, we outline historical and recent advances in understanding how the gut microbiota is involved in regulating body weight homeostasis. We also discuss the opportunities, limitations and challenges of using gut microbiota-related approaches as a means to achieve and maintain a healthy body weight.
Collapse
Affiliation(s)
- Matthias Van Hul
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO Department, WEL Research Institute, Wavre, Belgium.
| |
Collapse
|
22
|
Chitin and chitin-based biomaterials: A review of advances in processing and food applications. Carbohydr Polym 2023; 299:120142. [PMID: 36876773 DOI: 10.1016/j.carbpol.2022.120142] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022]
Abstract
Chitin is the most abundant natural amino polysaccharide, showing various practical applications owing to its functional properties. However, there are barriers in the development due to the difficulty of chitin extraction and purification, regarding its high crystallinity and low solubility. In recent years, some novel technologies such as microbial fermentation, ionic liquid, electrochemical extraction have emerged for the green extraction of chitin from new sources. Furthermore, nanotechnology, dissolution systems and chemical modification were applied to develop a variety of chitin-based biomaterials. Remarkably, chitin was used in delivering active ingredients and developing functional foods for weight loss, lipid reduction, gastrointestinal health, and anti-aging. Moreover, the application of chitin-based materials was expanded into medicine, energy and the environment. This review outlined the emerging extraction methods and processing routes of different chitin sources and advances in applying chitin-based materials. We aimed to provide some direction for the multi-disciplinary production and application of chitin.
Collapse
|
23
|
Dietary Fiber Intake and Gut Microbiota in Human Health. Microorganisms 2022; 10:microorganisms10122507. [PMID: 36557760 PMCID: PMC9787832 DOI: 10.3390/microorganisms10122507] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/11/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Dietary fiber is fermented by the human gut microbiota, producing beneficial microbial metabolites, such as short-chain fatty acids. Over the last few centuries, dietary fiber intake has decreased tremendously, leading to detrimental alternations in the gut microbiota. Such changes in dietary fiber consumption have contributed to the global epidemic of obesity, type 2 diabetes, and other metabolic disorders. The responses of the gut microbiota to the dietary changes are specific to the type, amount, and duration of dietary fiber intake. The intricate interplay between dietary fiber and the gut microbiota may provide clues for optimal intervention strategies for patients with type 2 diabetes and other noncommunicable diseases. In this review, we summarize current evidence regarding dietary fiber intake, gut microbiota modulation, and modification in human health, highlighting the type-specific cutoff thresholds of dietary fiber for gut microbiota and metabolic outcomes.
Collapse
|
24
|
Seethaler B, Nguyen NK, Basrai M, Kiechle M, Walter J, Delzenne NM, Bischoff SC. Short-chain fatty acids are key mediators of the favorable effects of the Mediterranean diet on intestinal barrier integrity: data from the randomized controlled LIBRE trial. Am J Clin Nutr 2022; 116:928-942. [PMID: 36055959 DOI: 10.1093/ajcn/nqac175] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/28/2022] [Accepted: 06/25/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The Mediterranean diet is associated with the prevention of diabetes, cardiovascular disease, and cancer, all of which are linked to intestinal barrier impairment. OBJECTIVES Here, we hypothesize that the Mediterranean diet, possibly via the induction of short-chain fatty acids (SCFAs), improves intestinal barrier integrity. Furthermore, we aim to establish novel personalized nutrition advice based on machine learning algorithms. METHODS We studied 260 women with intestinal barrier impairment. The women were allocated to follow either a Mediterranean diet or a control diet for 3 mo. Intestinal permeability was assessed by measuring lipopolysaccharide binding protein (LBP) in plasma and zonulin in feces. SCFA concentrations were analyzed in feces. Bi- and multivariate analyses and machine learning algorithms (random forest classification) were conducted. RESULTS Particularly in the intervention group, adherence to the Mediterranean diet increased, whereas plasma LBP and fecal zonulin concentrations decreased (all q < 0.001 for the intervention group, all q < 0.1 for control group). In the intervention group, fecal SCFA concentrations increased (propionate + 19%; butyrate + 44%; both q < 0.001). Multivariate analyses showed that adherence to the Mediterranean diet was associated with SCFA concentrations (all q < 0.001) and inversely associated with LBP and zonulin concentrations (all q < 0.02). Mediation analyses identified propionate and butyrate as the key mechanistic link between diet and intestinal permeability integrity. Accordingly, using baseline SCFA data, we could predict the effect of the Mediterranean diet on intestinal permeability using a machine learning algorithm (receiver operating characteristic AUC: 0.78-0.96). CONCLUSIONS Our data suggest that SCFAs are key mediators for the relation between diet and gut health. Assessment of SCFAs may form a basis for personalized nutrition in future clinical care. These results need to be verified in larger studies powered for this purpose, comprising different study populations. The trial was registered at clinicaltrials.gov as NCT02087592 and NCT02516540.
Collapse
Affiliation(s)
- Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | | | - Maryam Basrai
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Marion Kiechle
- Department of Gynecology, Center for Hereditary Breast and Ovarian Cancer, Klinikum Rechts der Isar, Technical University Munich and Comprehensive Cancer Center Munich, Munich, Germany
| | - Jens Walter
- APC Microbiome Ireland, Department of Medicine, and School of Microbiology, University College Cork, Cork, Ireland
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
25
|
Rhoades NS, Cinco IR, Hendrickson SM, Slifka MK, Messaoudi I. Taxonomic and Functional Shifts in the Perinatal Gut Microbiome of Rhesus Macaques. Microbiol Spectr 2022; 10:e0081422. [PMID: 35863030 PMCID: PMC9431225 DOI: 10.1128/spectrum.00814-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/17/2022] [Indexed: 11/20/2022] Open
Abstract
Pregnancy and the postpartum period result in some of the most dramatic metabolic, hormonal, and physiological changes that can be experienced by an otherwise healthy adult. The timing and magnitude of these changes is key for both maternal and fetal health. One of the factors believed to critically modulate these physiological changes is the maternal gut microbiome. However, the dynamic changes in this community during the perinatal period remain understudied. Clinical studies can be complicated by confounding variables like diet and other drivers of heterogeneity in the human microbiome. Therefore, in this study, we conducted a longitudinal analysis of the fecal microbiome obtained during the pregnancy and postpartum periods in 26 captive rhesus macaques using 16S rRNA gene amplicon sequencing and shotgun metagenomics. Shifts at both the taxonomic and functional potential level were detected when comparing pregnancy to postpartum samples. Taxonomically, Alloprevotella, Actinobacillus, and Anaerovibrio were enriched in the gut microbiome during pregnancy, while Treponema, Lachnospiraceae, and Methanosphaera were more abundant postpartum. Functionally, the gut microbiome during pregnancy was associated with increased abundance in pathways involving the production of the short-chain fatty acid (SCFA) butyrate, while pathways associated with starch degradation and folate transformation were more abundant during the postpartum period. These data demonstrate dramatic changes in the maternal gut microbiome even in the absence of dietary changes and suggest that rhesus macaques could provide a valuable model to determine how changes in the microbiome correlate to other physiological changes in pregnancy. IMPORTANCE Pregnancy and the postpartum period are characterized by a myriad of metabolic and physiological adaptations needed to support fetal growth and maternal health. The maternal gut microbiome is believed to play a key role during this period but remains underexplored. Here, we report significant shifts in the taxonomic landscape and functional potential of the gut microbiome in 26 pregnant rhesus macaques during the transition from pregnancy to the postpartum period, despite shared dietary and environmental exposures. Increased abundance of pathways involved in the production of the short-chain fatty acid butyrate could play a critical role in modulating the maternal immune system and regulating fetal tolerance. On the other hand, increased abundance of pathways associated with starch degradation and folate transformation during the postpartum period could be important for meeting the metabolic demands of breastfeeding and neonatal growth.
Collapse
Affiliation(s)
- Nicholas S. Rhoades
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, Kentucky, USA
| | - Isaac R. Cinco
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, Kentucky, USA
| | - Sara M. Hendrickson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Sciences University, Beaverton, Oregon, USA
| | - Mark K. Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Sciences University, Beaverton, Oregon, USA
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
26
|
Recent Research Progress of Ionic Liquid Dissolving Silks for Biomedicine and Tissue Engineering Applications. Int J Mol Sci 2022; 23:ijms23158706. [PMID: 35955840 PMCID: PMC9369158 DOI: 10.3390/ijms23158706] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022] Open
Abstract
Ionic liquids (ILs) show a bright application prospect in the field of biomedicine and energy materials due to their unique recyclable, modifiability, structure of cation and anion adjustability, as well as excellent physical and chemical properties. Dissolving silk fibroin (SF), from different species silkworm cocoons, with ILs is considered an effective new way to obtain biomaterials with highly enhanced/tailored properties, which can significantly overcome the shortcomings of traditional preparation methods, such as the cumbersome, time-consuming and the organic toxicity caused by manufacture. In this paper, the basic structure and properties of SF and the preparation methods of traditional regenerated SF solution are first introduced. Then, the dissolving mechanism and main influencing factors of ILs for SF are expounded, and the fabrication methods, material structure and properties of SF blending with natural biological protein, inorganic matter, synthetic polymer, carbon nanotube and graphene oxide in the ILs solution system are introduced. Additionally, our work summarizes the biomedicine and tissue engineering applications of silk-based materials dissolved through various ILs. Finally, according to the deficiency of ILs for dissolving SF at a high melting point and expensive cost, their further study and future development trend are prospected.
Collapse
|
27
|
Xia Y, Meng P, Liu S, Tan Z, Yang X, Liang L, Xie F, Zhang H, Wang G, Xiong Z, Lo J, Ai L. Structural and Potential Functional Properties of Alkali-Extracted Dietary Fiber From Antrodia camphorata. Front Microbiol 2022; 13:921164. [PMID: 35875549 PMCID: PMC9301256 DOI: 10.3389/fmicb.2022.921164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/30/2022] [Indexed: 11/27/2022] Open
Abstract
Antrodia camphorata is rich in a variety of bioactive ingredients; however, the utilization efficiency of the residue of A. camphorata is low, resulting in serious waste. It is necessary to deeply study the functional components of A. camphorata residues to achieve high-value utilization. In this study, the components, structural characteristics, and functional properties of alkali-extracted dietary fiber extracted from residues of A. camphorata (basswood and dish cultured fruiting body, respectively) were investigated. There were similar components and structural characteristics of ACA-DK (extract from basswood cultured) and ACA-DF (extract from dish cultured). The two alkali-extracted dietary fiber were composed of mainly cellulose and xylan. However, ACA-DK has better adsorption capacities than ACA-DF on lipophilic substances such as oil (12.09 g/g), cholesterol (20.99 mg/g), and bile salts (69.68 mg/g). In vitro immunomodulatory assays stated that ACA-DK had a good effect on promoting the proliferation of RAW 264.7 cells and can activate cell phagocytosis, NO synthesis, and other immune capabilities. The edible fungus A. camphorata is a good source of functional dietary fiber. The alkali-extracted dietary fiber of A. camphorata might be used as a functional ingredient in the medicine and food industry.
Collapse
Affiliation(s)
- Yongjun Xia
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Peng Meng
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Shaodong Liu
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Zhuoming Tan
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Xi Yang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Lihong Liang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Fan Xie
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Hui Zhang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Guangqiang Wang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Zhiqiang Xiong
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Jenyu Lo
- Honest and Humble Biotechnology Co., Ltd., New Taipei City, China
| | - Lianzhong Ai
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
- *Correspondence: Lianzhong Ai
| |
Collapse
|
28
|
Neyrinck AM, Rodriguez J, Zhang Z, Nazare JA, Bindels LB, Cani PD, Maquet V, Laville M, Bischoff SC, Walter J, Delzenne NM. Breath volatile metabolome reveals the impact of dietary fibres on the gut microbiota: Proof of concept in healthy volunteers. EBioMedicine 2022; 80:104051. [PMID: 35561452 PMCID: PMC9108873 DOI: 10.1016/j.ebiom.2022.104051] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 01/06/2023] Open
Abstract
Background Current data suggest that dietary fibre (DF) interaction with the gut microbiota largely contributes to their physiological effects. The bacterial fermentation of DF leads to the production of metabolites, most of them are volatile. This study analyzed the breath volatile metabolites (BVM) profile in healthy individuals (n=15) prior and after a 3-week intervention with chitin-glucan (CG, 4.5 g/day), an insoluble fermentable DF. Methods The present exploratory study presents the original data related to the secondary outcomes, notably the analysis of BVM. BVM were analyzed throughout the test days -in fasting state and after standardized meals - using selected ion flow tube mass spectrometry (SIFT-MS). BVM production was correlated to the gut microbiota composition (Illumina sequencing, primary outcome), analyzed before and after the intervention. Findings The data reveal that the post-prandial state versus fasting state is a key determinant of BVM fingerprint. Correlation analyses with fecal microbiota spotlighted butyrate-producing bacteria, notably Faecalibacterium, as dominant bacteria involved in butyrate and other BVM expiration. CG intervention promotes interindividual variations of fasting BVM, and decreases or delays the expiration of most exhaled BVM in favor of H2 expiration, without any consequence on gastrointestinal tolerance. Interpretation Assessing BVM is a non-invasive methodology allowing to analyze the influence of DF intervention on the gut microbiota. Funding FiberTAG project was initiated from a European Joint Programming Initiative “A Healthy Diet for a Healthy Life” (JPI HDHL) and was supported by the Service Public de Wallonie (SPW-EER, convention 1610365, Belgium).
Collapse
Affiliation(s)
- Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), avenue E. Mounier box B1.73.11, Brussels B-1200, Belgium
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), avenue E. Mounier box B1.73.11, Brussels B-1200, Belgium
| | - Zhengxiao Zhang
- Department of Medicine, University of Alberta, Edmonton, Canada; College of Food and Biological Engineering, Jimei University, Xiamen, China
| | - Julie-Anne Nazare
- Rhône-Alpes Research Center for Human Nutrition, CarMeN Laboratory, Hospices Civils de Lyon, Université-Lyon, France
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), avenue E. Mounier box B1.73.11, Brussels B-1200, Belgium
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), avenue E. Mounier box B1.73.11, Brussels B-1200, Belgium; WELBIO- Walloon Excellence in Life Sciences and Biotechnology, UCLouvain (Université catholique de Louvain), Brussels, Belgium
| | - Véronique Maquet
- KitoZyme, Parc Industriel des Hauts-Sart, Zone 2, Rue de Milmort 680, Herstal 4040, Belgium
| | - Martine Laville
- Rhône-Alpes Research Center for Human Nutrition, CarMeN Laboratory, Hospices Civils de Lyon, Université-Lyon, France
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Jens Walter
- Department of Medicine, APC Microbiome Ireland, School of Microbiology, University College Cork, Cork, Ireland
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain (Université catholique de Louvain), avenue E. Mounier box B1.73.11, Brussels B-1200, Belgium.
| |
Collapse
|
29
|
Ranaivo H, Zhang Z, Alligier M, Van Den Berghe L, Sothier M, Lambert-Porcheron S, Feugier N, Cuerq C, Machon C, Neyrinck AM, Seethaler B, Rodriguez J, Roumain M, Muccioli GG, Maquet V, Laville M, Bischoff SC, Walter J, Delzenne NM, Nazare JA. Chitin-glucan supplementation improved postprandial metabolism and altered gut microbiota in subjects at cardiometabolic risk in a randomized trial. Sci Rep 2022; 12:8830. [PMID: 35614185 PMCID: PMC9132890 DOI: 10.1038/s41598-022-12920-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/05/2022] [Indexed: 01/04/2023] Open
Abstract
Chitin-glucan (CG), an insoluble dietary fiber, has been shown to improve cardiometabolic disorders associated with obesity in mice. Its effects in healthy subjects has recently been studied, revealing its interaction with the gut microbiota. In this double-blind, randomized, cross-over, twice 3-week exploratory study, we investigated the impacts of CG on the cardiometabolic profile and gut microbiota composition and functions in 15 subjects at cardiometabolic risk. They consumed as a supplement 4.5 g of CG daily or maltodextrin as control. Before and after interventions, fasting and postprandial metabolic parameters and exhaled gases (hydrogen [H2] and methane [CH4]) were evaluated. Gut microbiota composition (16S rRNA gene sequencing analysis), fecal concentrations of bile acids, long- and short-chain fatty acids (LCFA, SCFA), zonulin, calprotectin and lipopolysaccharide binding protein (LBP) were analyzed. Compared to control, CG supplementation increased exhaled H2 following an enriched-fiber breakfast ingestion and decreased postprandial glycemia and triglyceridemia response to a standardized test meal challenge served at lunch. Of note, the decrease in postprandial glycemia was only observed in subjects with higher exhaled H2, assessed upon lactulose breath test performed at inclusion. CG decreased a family belonging to Actinobacteria phylum and increased 3 bacterial taxa: Erysipelotrichaceae UCG.003, Ruminococcaceae UCG.005 and Eubacterium ventriosum group. Fecal metabolites, inflammatory and intestinal permeability markers did not differ between groups. In conclusion, we showed that CG supplementation modified the gut microbiota composition and improved postprandial glycemic response, an early determinant of cardiometabolic risk. Our results also suggest breath H2 production as a non-invasive parameter of interest for predicting the effectiveness of dietary fiber intervention.
Collapse
Affiliation(s)
- Harimalala Ranaivo
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Zhengxiao Zhang
- College of Food and Biological Engineering, Jimei University, Xiamen, Fujian, China
| | - Maud Alligier
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Laurie Van Den Berghe
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Monique Sothier
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Stéphanie Lambert-Porcheron
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Nathalie Feugier
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Charlotte Cuerq
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
- Service de Biochimie et Biologie Moléculaire, Unité Médicale Dyslipidémies et Dysfonctions Nutritionnelles et Digestives, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Christelle Machon
- Hospices Civils de Lyon, Service de Biochimie, Centre de Biologie Sud, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Martin Roumain
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Véronique Maquet
- KitoZyme, Parc Industriel des Hauts-Sart, Zone 2, Rue de Milmort 680, 4040, Herstal, Belgium
| | - Martine Laville
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Jens Walter
- Department of Medicine, and School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Julie-Anne Nazare
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France.
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France.
| |
Collapse
|
30
|
Yao W, Gong Y, Li L, Hu X, You L. The effects of dietary fibers from rice bran and wheat bran on gut microbiota: An overview. Food Chem X 2022; 13:100252. [PMID: 35498986 PMCID: PMC9040006 DOI: 10.1016/j.fochx.2022.100252] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 12/19/2022] Open
Abstract
The physicochemical properties of DFs are related to their digestive behaviors. DFs are degraded in the intestines due to the fermentation of gut microbiota. DFs and their metabolites exert beneficial effects on gut microbiota. The fermentation of DFs improve gut barrier function and immune function.
Whole grain is the primary food providing abundant dietary fibers (DFs) in the human diet. DFs from rice bran and wheat bran have been well documented in modulating gut microbiota. This review aims to summarize the physicochemical properties and digestive behaviors of DFs from rice bran and wheat bran and their effects on host gut microbiota. The physicochemical properties of DFs are closely related to their fermentability and digestive behaviors. DFs from rice bran and wheat bran modulate specific bacteria and promote SAFCs-producing bacteria to maintain host health. Moreover, their metabolites stimulate the production of mucus-associated bacteria to enhance the intestinal barrier and regulate the immune system. They also reduce the level of related inflammatory cytokines and regulate Tregs activation. Therefore, DFs from rice bran and wheat bran will serve as prebiotics, and diets rich in whole grain will be a biotherapeutic strategy for human health.
Collapse
Affiliation(s)
- Wanzi Yao
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510640, China
| | - Yufeng Gong
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510640, China
| | - Laihao Li
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Xiao Hu
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China
| | - Lijun You
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510640, China
| |
Collapse
|
31
|
Physicochemical characterization, adsorption function and prebiotic effect of chitin-glucan complex from mushroom Coprinus comatus. Int J Biol Macromol 2022; 206:255-263. [PMID: 35240205 DOI: 10.1016/j.ijbiomac.2022.02.152] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/13/2022] [Accepted: 02/25/2022] [Indexed: 11/22/2022]
Abstract
Chitin-glucan complex (CGC) is a novel insoluble dietary fiber with multiple physiological activities. In this work, CGC was extracted from the fruiting body of Coprinus comatus and its physicochemical properties and prebiotic effects were investigated. The results indicated that CGC consisted of glucosamine and glucose in a molar ratio of 67: 33 with degree of acetylation of 61.91% and crystallinity index of 25.40%. The maximum degradation temperature was determined to be 307.52 °C, and a woven fibrous structure was observed by scanning electron microscopy. CGC exhibited higher oil-holding capacity, water-holding capacity and nitrite ion adsorption capacity than commercial chitin, and showed potential prebiotic effects. Compared with control and commercial chitin, CGC significantly (P < 0.05) increased the concentration of propionic and butyric acids. These results suggested that CGC from C. comatus was promising to be an alternative source of CGC products and used as a bioactive ingredient in functional foods.
Collapse
|
32
|
Abstract
The prevalence of overweight and obesity has reached epidemic proportions globally over the past few decades. The search for new management approaches continues and among them, targeting the gut microbiota can be envisioned. To date, numerous data showed the involvement of the gut microbes in the regulation and control of host metabolism. There are also increasing evidences highlighting the interactions between environmental factors, intrinsic factors, gut microbiota, and metabolic diseases. Diet emerges as the most relevant factor influencing the gut microbiome. Eating habits, as well as short-term consumption of specific diets, alter the gut microbiota composition. Moreover, nutritional disorders are associated with changes of the gut microbiota composition and/or function, as shown in obesity or type 2 diabetic patients versus healthy lean subjects. Targeting the gut microbiota for improving metabolic health appears as a new approach to manage obesity and cardio-metabolic risk. In this review, we have detailed the results of human interventions targeting the gut microbiome by prebiotic supplementation, prebiotics being defined as "substrates that are selectively utilized by the host microorganisms conferring a health benefit." If the potential benefit of this approach is obvious in preclinical models, the efficacy of prebiotics in humans is less reproducible. The inter-individual variability of response to dietary intervention can be dependent on the gut microbiota and we summarized the basal gut microbiota characteristics driving the metabolic response to dieting, prebiotic and dietary fiber intervention in the context of obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
33
|
Kurahashi A, Enomoto T, Oguro Y, Kojima-Nakamura A, Kodaira K, Watanabe K, Ozaki N, Goto H, Hirayama M. Intake of Koji Amazake Improves Defecation Frequency in Healthy Adults. J Fungi (Basel) 2021; 7:jof7090782. [PMID: 34575820 PMCID: PMC8470246 DOI: 10.3390/jof7090782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/19/2022] Open
Abstract
Reportedly, the intake of koji amazake, a beverage made from steamed rice fermented by Aspergillus oryzae, improves defecation frequency. However, its functional ingredients and mechanism of action remain unclear. To compare the effects of koji amazake and a placebo beverage on defecation frequency and to identify the functional ingredients and mechanism of action, a randomized, placebo-controlled, double-blind parallel-group comparative trial was performed on two groups. The koji amazake had 302 ± 15.5 mg/118 g of A. oryzae cells, which was not in the placebo. Compared with the placebo group, the koji amazake group showed a significant increase in weekly defecation frequency at 2 weeks (5.09 days vs. 4.14 days), 3 weeks (5.41 days vs. 4.18 days), and 4 weeks (5.09 days vs. 3.95 days), along with an increase in the weekly fecal weight at 4 weeks (724 g vs. 501 g). The intake of koji amazake did not induce significant intergroup differences in the fecal SCFA concentration, whereas it significantly decreased the relative abundance of Blautia and significantly increased that of Bacteroides at 3 weeks. Therefore, koji amazake intake improved defecation frequency, and A. oryzae cells played potentially important roles as functional ingredients.
Collapse
Affiliation(s)
- Atsushi Kurahashi
- Hakkaisan Brewery Co., Ltd., 1051 Nagamori, Minamiuonuma City, Niigata 949-7112, Japan; (T.E.); (Y.O.); (A.K.-N.); (K.K.)
- Correspondence: ; Tel.: +81-25-788-0910
| | - Toshihiko Enomoto
- Hakkaisan Brewery Co., Ltd., 1051 Nagamori, Minamiuonuma City, Niigata 949-7112, Japan; (T.E.); (Y.O.); (A.K.-N.); (K.K.)
| | - Yoshifumi Oguro
- Hakkaisan Brewery Co., Ltd., 1051 Nagamori, Minamiuonuma City, Niigata 949-7112, Japan; (T.E.); (Y.O.); (A.K.-N.); (K.K.)
| | - Ayana Kojima-Nakamura
- Hakkaisan Brewery Co., Ltd., 1051 Nagamori, Minamiuonuma City, Niigata 949-7112, Japan; (T.E.); (Y.O.); (A.K.-N.); (K.K.)
| | - Kazuya Kodaira
- Hakkaisan Brewery Co., Ltd., 1051 Nagamori, Minamiuonuma City, Niigata 949-7112, Japan; (T.E.); (Y.O.); (A.K.-N.); (K.K.)
| | - Kenichi Watanabe
- Department of Endocrinology and Metabolism, Niigata University Graduate School of Medical and Dental Sciences, 754 Ichibancho, Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan;
| | - Nobuhiro Ozaki
- Niigata Medical Association of Occupational Health Inc., 1185-3 Kitaba, Nishi-ku, Niigata City, Niigata 950-1187, Japan;
| | - Hiroshi Goto
- Niigata Bio-Research Park Inc., 316-2 Higashijima, Akiha-ku, Niigata City, Niigata 956-0841, Japan; (H.G.); (M.H.)
| | - Masao Hirayama
- Niigata Bio-Research Park Inc., 316-2 Higashijima, Akiha-ku, Niigata City, Niigata 956-0841, Japan; (H.G.); (M.H.)
| |
Collapse
|
34
|
Calatayud M, Verstrepen L, Ghyselinck J, Van den Abbeele P, Marzorati M, Modica S, Ranjanoro T, Maquet V. Chitin Glucan Shifts Luminal and Mucosal Microbial Communities, Improve Epithelial Barrier and Modulates Cytokine Production In Vitro. Nutrients 2021; 13:nu13093249. [PMID: 34579126 PMCID: PMC8467507 DOI: 10.3390/nu13093249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022] Open
Abstract
The human gut microbiota has been linked to the health status of the host. Modulation of human gut microbiota through pro- and prebiotic interventions has yielded promising results; however, the effect of novel prebiotics, such as chitin-glucan, on gut microbiota-host interplay is still not fully characterized. We assessed the effect of chitin-glucan (CG) and chitin-glucan plus Bifidobacterium breve (CGB) on human gut microbiota from the luminal and mucosal environments in vitro. Further, we tested the effect of filter-sterilized fecal supernatants from CG and CGB fermentation for protective effects on inflammation-induced barrier disruption and cytokine production using a co-culture of enterocytes and macrophage-like cells. Overall, CG and CGB promote health-beneficial short-chain fatty acid production and shift human gut microbiota composition, with a consistent effect increasing Roseburia spp. and butyrate producing-bacteria. In two of three donors, CG and CGB also stimulated Faecalibacterium prausniitzi. Specific colonization of B. breve was observed in the lumen and mucosal compartment; however, no synergy was detected for different endpoints when comparing CGB and CG. Both treatments included a significant improvement of inflammation-disrupted epithelial barrier and shifts on cytokine production, especially by consistent increase in the immunomodulatory cytokines IL10 and IL6.
Collapse
Affiliation(s)
- Marta Calatayud
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium; (L.V.); (J.G.); (P.V.d.A.)
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9052 Ghent, Belgium
- Correspondence: (M.C.); or (M.M.)
| | - Lynn Verstrepen
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium; (L.V.); (J.G.); (P.V.d.A.)
| | - Jonas Ghyselinck
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium; (L.V.); (J.G.); (P.V.d.A.)
| | | | - Massimo Marzorati
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium; (L.V.); (J.G.); (P.V.d.A.)
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9052 Ghent, Belgium
- Correspondence: (M.C.); or (M.M.)
| | - Salvatore Modica
- KitoZyme SA., Parc Industriel des Hauts Sarts, 4040 Herstal, Belgium; (S.M.); (T.R.); (V.M.)
| | - Thibaut Ranjanoro
- KitoZyme SA., Parc Industriel des Hauts Sarts, 4040 Herstal, Belgium; (S.M.); (T.R.); (V.M.)
| | - Véronique Maquet
- KitoZyme SA., Parc Industriel des Hauts Sarts, 4040 Herstal, Belgium; (S.M.); (T.R.); (V.M.)
| |
Collapse
|
35
|
Gao J, Guo X, Wei W, Li R, Hu K, Liu X, Jiang W, Liu S, Wang W, Sun H, Wu H, Zhang Y, Gu W, Li Y, Sun C, Han T. The Association of Fried Meat Consumption With the Gut Microbiota and Fecal Metabolites and Its Impact on Glucose Homoeostasis, Intestinal Endotoxin Levels, and Systemic Inflammation: A Randomized Controlled-Feeding Trial. Diabetes Care 2021; 44:1970-1979. [PMID: 34253560 DOI: 10.2337/dc21-0099] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/03/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE This randomized controlled-feeding trial aimed to determine the impact of fried meat intake on the gut microbiota and fecal cometabolites and whether such impacts influenced host glucose homoeostasis, intestinal endotoxin levels, and systemic inflammation. RESEARCH DESIGN AND METHODS A total of 117 overweight adults were randomized into two groups. Fifty-nine participants were provided fried meat four times per week, and 58 participants were restricted from fried meat intake, while holding food group and nutrient compositions constant, for 4 weeks. The gut microbiota was analyzed by 16S rRNA sequencing. Glucose and insulin concentrations at 0, 30, 60, and 120 min of an oral glucose tolerance test, fecal microbiota-host cometabolite levels, and intestinal endotoxin and inflammation serum biomarker levels were measured. The area under the curve (AUC) for insulin, insulinogenic index (IGI), and muscle insulin resistance index (MIRI) were calculated. RESULTS The participants who consumed fried meat had lower IGI values than the control subjects, but they had higher MIRI and AUC values of insulin and lipopolysaccharide (LPS), TNF-α, IL-10, and IL-1β levels (P < 0.05). Fried meat intake lowered microbial community richness and decreased Lachnospiraceae and Flavonifractor abundances while increasing Dialister, Dorea, and Veillonella abundances (P FDR <0.05), provoking a significant shift in the fecal cometabolite profile, with lower 3-indolepropionic acid, valeric acid, and butyric acid concentrations and higher carnitine and methylglutaric acid concentrations (P FDR <0.05). Changes in these cometabolite levels were significantly associated with changes in IGI and MIRI values and LPS, FGF21, TNF-α, IL-1β, and IL-10 levels (P < 0.05). CONCLUSIONS Fried meat intake impaired glucose homoeostasis and increased intestinal endotoxin and systemic inflammation levels by influencing the gut microbiota and microbial-host cometabolites.
Collapse
Affiliation(s)
- Jian Gao
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Xiaoyu Guo
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Wei Wei
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Ran Li
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China.,Harbin Center for Disease Control and Prevention, Harbin, China
| | - Ke Hu
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Xin Liu
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Wenbo Jiang
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Siyao Liu
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Memphis, TN.,Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| | - Weiqi Wang
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Hu Sun
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Huanyu Wu
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Yuntao Zhang
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Wenbo Gu
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Ying Li
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Harbin Medical University, Ministry of Education, Harbin, China.,NHC Key Laboratory of Cell Translation, Harbin Medical University, Harbin, China
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China
| | - Tianshu Han
- Department of Nutrition and Food Hygiene, National Key Discipline, School of Public Health, Harbin Medical University, Harbin, China .,Department of Endocrinology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
36
|
Neyrinck AM, Rodriguez J, Zhang Z, Seethaler B, Sánchez CR, Roumain M, Hiel S, Bindels LB, Cani PD, Paquot N, Cnop M, Nazare JA, Laville M, Muccioli GG, Bischoff SC, Walter J, Thissen JP, Delzenne NM. Prebiotic dietary fibre intervention improves fecal markers related to inflammation in obese patients: results from the Food4Gut randomized placebo-controlled trial. Eur J Nutr 2021; 60:3159-3170. [PMID: 33544206 PMCID: PMC8354918 DOI: 10.1007/s00394-021-02484-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023]
Abstract
Purpose Inulin-type fructans (ITF) are prebiotic dietary fibre (DF) that may confer beneficial health effects, by interacting with the gut microbiota. We have tested the hypothesis that a dietary intervention promoting inulin intake versus placebo influences fecal microbial-derived metabolites and markers related to gut integrity and inflammation in obese patients. Methods Microbiota (16S rRNA sequencing), long- and short-chain fatty acids (LCFA, SCFA), bile acids, zonulin, and calprotectin were analyzed in fecal samples obtained from obese patients included in a randomized, placebo-controlled trial. Participants received either 16 g/d native inulin (prebiotic n = 12) versus maltodextrin (placebo n = 12), coupled to dietary advice to consume inulin-rich versus inulin-poor vegetables for 3 months, in addition to dietary caloric restriction. Results Both placebo and prebiotic interventions lowered energy and protein intake. A substantial increase in Bifidobacterium was detected after ITF treatment (q = 0.049) supporting our recent data obtained in a larger cohort. Interestingly, fecal calprotectin, a marker of gut inflammation, was reduced upon ITF treatment. Both prebiotic and placebo interventions increased the ratio of tauro-conjugated/free bile acids in feces. Prebiotic treatment did not significantly modify fecal SCFA content but it increased fecal rumenic acid, a conjugated linoleic acid (cis-9, trans-11 CLA) with immunomodulatory properties, that correlated notably to the expansion of Bifidobacterium (p = 0.031; r = 0.052). Conclusions Our study demonstrates that ITF-prebiotic intake during 3 months decreases a fecal marker of intestinal inflammation in obese patients. Our data point to a potential contribution of microbial lipid-derived metabolites in gastro-intestinal dysfunction related to obesity. ClinicalTrials.gov Identifier NCT03852069 (February 22, 2019 retrospectively, registered). Supplementary Information The online version contains supplementary material available at 10.1007/s00394-021-02484-5.
Collapse
Affiliation(s)
- Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium
| | - Zhengxiao Zhang
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Hohenheim, Germany
| | - Cándido Robles Sánchez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium
| | - Martin Roumain
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Sophie Hiel
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium.,WELBIO- Walloon Excellence in Life Sciences and Biotechnology, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Nicolas Paquot
- Laboratory of Diabetology, Nutrition and Metabolic Disease, Université de Liège, Liège, Belgium
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium.,Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Julie-Anne Nazare
- Rhône-Alpes Research Center for Human Nutrition, Université-Lyon, CarMeN Laboratory, Hospices Civils de Lyon, Lyon, France
| | - Martine Laville
- Rhône-Alpes Research Center for Human Nutrition, Université-Lyon, CarMeN Laboratory, Hospices Civils de Lyon, Lyon, France
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Hohenheim, Germany
| | - Jens Walter
- Department of Medicine, University of Alberta, Edmonton, Canada.,Department of Medicine, and School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jean-Paul Thissen
- Pole of Endocrinology, Diabetes and Nutrition, Institut de Recherche Expérimentale et Clinique, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium.
| |
Collapse
|
37
|
Neyrinck AM, Rodriguez J, Zhang Z, Seethaler B, Mailleux F, Vercammen J, Bindels LB, Cani PD, Nazare JA, Maquet V, Laville M, Bischoff SC, Walter J, Delzenne NM. Noninvasive monitoring of fibre fermentation in healthy volunteers by analyzing breath volatile metabolites: lessons from the FiberTAG intervention study. Gut Microbes 2021; 13:1-16. [PMID: 33461385 PMCID: PMC7833774 DOI: 10.1080/19490976.2020.1862028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/09/2020] [Accepted: 11/29/2020] [Indexed: 02/04/2023] Open
Abstract
The fermentation of dietary fibre (DF) leads to the production of bioactive metabolites, the most volatile ones being excreted in the breath. The aim of this study was to analyze the profile of exhaled breath volatile metabolites (BVM) and gastrointestinal symptoms in healthy volunteers after a single ingestion of maltodextrin (placebo) versus chitin-glucan (CG), an insoluble DF previously shown to be fermented into short-chain fatty acids (SCFA) by the human microbiota in vitro. Maltodextrin (4.5 g at day 0) or CG (4.5 g at day 2) were added to a standardized breakfast in fasting healthy volunteers (n = 15). BVM were measured using selected ion flow tube mass spectrometry (SIFT-MS) throughout the day. A single ingestion of 4.5 g CG did not induce significant gastrointestinal discomfort. Untargeted metabolomics analysis of breath highlighted that 13 MS-fragments (among 408 obtained from ionizations of breath) discriminated CG versus maltodextrin acute intake in the posprandial state. The targeted analysis revealed that CG increased exhaled butyrate and 5 other BVM - including the microbial metabolites 2,3-butanedione and 3-hydroxybutanone - with a peak observed 6 h after CG intake. Correlation analyses with fecal microbiota (Illumina 16S rRNA sequencing) spotlighted Mitsuokella as a potential genus responsible for the presence of butyric acid, triethylamine and 3-hydroxybutanone in the breath. In conclusion, measuring BMV in the breath reveals the microbial signature of the fermentation of DF after a single ingestion. This protocol allows to analyze the time-course of released bioactive metabolites that could be proposed as new biomarkers of DF fermentation, potentially linked to their biological properties. Trial registration: Clinical Trials NCT03494491. Registered 11 April 2018 - Retrospectively registered, https://clinicaltrials.gov/ct2/show/NCT03494491.
Collapse
Affiliation(s)
- Audrey M. Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Zhengxiao Zhang
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Florence Mailleux
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Joeri Vercammen
- Interscience, Louvain-la-Neuve, Belgium
- Engineering, Industrial Catalysis and Adsorption Technology (INCAT), Ghent University, Ghent, Belgium
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Patrice D. Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Julie-Anne Nazare
- Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Lyon, France
| | | | - Martine Laville
- Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Lyon, France
| | - Stephan C. Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Jens Walter
- Department of Agricultural, Food & Nutritional Science and Department of Biological Sciences, University of Alberta, Edmonton, Canada
- APC Microbiome Ireland, School of Microbiology, and Department of Medicine, University College Cork – National University of Ireland, Cork, Ireland
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
38
|
Satitsri S, Muanprasat C. Chitin and Chitosan Derivatives as Biomaterial Resources for Biological and Biomedical Applications. Molecules 2020; 25:molecules25245961. [PMID: 33339290 PMCID: PMC7766609 DOI: 10.3390/molecules25245961] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 01/30/2023] Open
Abstract
Chitin is a long-chain polymer of N-acetyl-glucosamine, which is regularly found in the exoskeleton of arthropods including insects, shellfish and the cell wall of fungi. It has been known that chitin can be used for biological and biomedical applications, especially as a biomaterial for tissue repairing, encapsulating drug for drug delivery. However, chitin has been postulated as an inducer of proinflammatory cytokines and certain diseases including asthma. Likewise, chitosan, a long-chain polymer of N-acetyl-glucosamine and d-glucosamine derived from chitin deacetylation, and chitosan oligosaccharide, a short chain polymer, have been known for their potential therapeutic effects, including anti-inflammatory, antioxidant, antidiarrheal, and anti-Alzheimer effects. This review summarizes potential utilization and limitation of chitin, chitosan and chitosan oligosaccharide in a variety of diseases. Furthermore, future direction of research and development of chitin, chitosan, and chitosan oligosaccharide for biomedical applications is discussed.
Collapse
|