1
|
Sullivan O, Sie C, Ng KM, Cotton S, Rosete C, Hamden JE, Singh AP, Lee K, Choudhary J, Kim J, Yu H, Clayton CA, Carranza Garcia NA, Voznyuk K, Deng BD, Plett N, Arora S, Ghezzi H, Huan T, Soma KK, Yu JPJ, Tropini C, Ciernia AV. Early-life gut inflammation drives sex-dependent shifts in the microbiome-endocrine-brain axis. Brain Behav Immun 2024; 125:117-139. [PMID: 39674560 DOI: 10.1016/j.bbi.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 11/13/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024] Open
Abstract
Despite recent advances in understanding the connection between the gut microbiota and the adult brain, significant knowledge gaps remain regarding how gut inflammation affects brain development. We hypothesized that gut inflammation during early life would negatively affect neurodevelopment by disrupting microbiota communication to the brain. We therefore developed a novel pediatric chemical model of inflammatory bowel disease (IBD), an incurable condition affecting millions of people worldwide. IBD is characterized by chronic intestinal inflammation, and is associated with comorbid symptoms such as anxiety, depression and cognitive impairment. Notably, 25% of patients with IBD are diagnosed during childhood, and the effects of chronic inflammation during this critical developmental period remain poorly understood. This study investigated the effects of early-life gut inflammation induced by DSS (dextran sulfate sodium) on a range of microbiota, endocrine, and behavioral outcomes, focusing on sex-specific impacts. DSS-treated mice exhibited increased intestinal inflammation and altered microbiota membership, which correlated with changes in microbiota-derived circulating metabolites. The majority of behavioral measures were unaffected, with the exception of impaired mate-seeking behaviors in DSS-treated males. DSS-treated males also showed significantly smaller seminal vesicles, lower circulating androgens, and decreased intestinal hormone-activating enzyme activity compared to vehicle controls. In the brain, DSS treatment led to chronic, sex-specific alterations in microglial morphology. These results suggest that early-life gut inflammation causes changes in gut microbiota composition, affecting short-chain fatty acid (SCFA) producers and glucuronidase (GUS) activity, correlating with altered SCFA and androgen levels. The findings highlight the developmental sensitivity to inflammation-induced changes in endocrine signalling and emphasize the long-lasting physiological and microbiome changes associated with juvenile IBD.
Collapse
Affiliation(s)
- Olivia Sullivan
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Claire Sie
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Katharine M Ng
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Sophie Cotton
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Cal Rosete
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Jordan E Hamden
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Ajay Paul Singh
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kristen Lee
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Jatin Choudhary
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Jennifer Kim
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Huaxu Yu
- Department of Chemistry, University of British Columbia, Vancouver, Canada
| | - Charlotte A Clayton
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | | | - Kateryna Voznyuk
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Brian D Deng
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Nadine Plett
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Sana Arora
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Hans Ghezzi
- Department of Bioinformatics, University of British Columbia, Vancouver, Canada
| | - Tao Huan
- Department of Chemistry, University of British Columbia, Vancouver, Canada
| | - Kiran K Soma
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Psychology, University of British Columbia, Vancouver Canada
| | - John-Paul J Yu
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Carolina Tropini
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, Canada; Humans and the Microbiome Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Canada.
| | - Annie Vogel Ciernia
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
2
|
Dong S, Zhang Y, Ye L, Cao Q. Identification of a Novel Activated NK-Associated Gene Score Associated with Diagnosis and Biological Therapy Response in Ulcerative Colitis. Digestion 2024:1-22. [PMID: 39182484 DOI: 10.1159/000540939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION Natural killer (NK) cells are associated with the pathogenesis of ulcerative colitis (UC); however, their precise contributions remain unclear. The present study aimed to investigate the diagnostic value of the activated NK-associated gene (ANAG) score in UC and evaluate its predictive value in response to biological therapy. METHODS Bulk RNA-seq and scRNA-seq datasets were obtained from the Gene Expression Omnibus (GEO) and Single Cell Portal (SCP) databases. In the bulk RNA-seq, differentially expressed genes (DEGs) were screened by the "Batch correction" and "Robust rank aggregation" (RRA) methods. The immune infiltration landscape was estimated using single-sample gene set enrichment analysis (ssGSEA) and CIBERSORT. DEGs that correlated with activated NK cells were identified as activated NK-associated genes (ANAGs). Protein-protein interaction (PPI) analysis and least absolute shrinkage and selection operator (LASSO) regression were used to screen key ANAGs and establish an ANAG score. The expression levels of the four key ANAGs were validated in human samples by real-time quantitative polymerase chain reaction (RT-qPCR) and immunofluorescence. The potential therapeutic drugs for UC were identified using the DSigDB database. Through scRNA-seq data analysis, the cell scores based on the ANAGs were calculated by "AddModuleScore" and "AUCell." RESULTS Immune infiltration analysis revealed a higher abundance of activated NK cells in noninflamed UC tissues (ssGSEA, p < 0.001; CIBERSORT, p < 0.01). Fifty-four DEGs correlated with activated NK cells were identified as ANAGs. The ANAG score was established using four key ANAGs (SELP, TIMP1, MMP7, and ABCG2). The ANAG scores were significantly higher in inflamed tissues (p < 0.001) and in biological therapy nonresponders (NR) tissues before treatment (golimumab, p < 0.05; ustekinumab, p < 0.001). The ANAG score demonstrated an excellent diagnostic value (AUC = 0.979). Patients with higher ANAG scores before treatment were more likely to experience a lack of response to golimumab or ustekinumab (golimumab, p < 0.05; ustekinumab, p < 0.001). CONCLUSION This study established a novel ANAG score with the ability to precisely diagnose UC and distinguish the efficacy of biological treatment.
Collapse
Affiliation(s)
- Siyuan Dong
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Yu Zhang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Lingna Ye
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Qian Cao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| |
Collapse
|
3
|
Madani AMA, Muhlisin M, Kurniawati A, Baskara AP, Anas MA. Dietary jack bean ( Canavalia ensiformis L.) supplementation enhanced intestinal health by modulating intestinal integrity and immune responses of broiler chickens. Heliyon 2024; 10:e34389. [PMID: 39130426 PMCID: PMC11315099 DOI: 10.1016/j.heliyon.2024.e34389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 08/13/2024] Open
Abstract
This study investigated the influence of supplementing with jack beans on jejunal morphology, cecal short-chain fatty acids production, gene expression both of pro- and anti-inflammatory cytokines and tight junctions. Four treatment groups including 288 Indian River chicks that were one day old were randomized at random. While the treatment groups received jack bean supplementation at levels of 5 %, 10 %, and 15 %, the control group (0 %) was given a basal diet. For 11-35 days, each treatment consisted of 8 pens with 9 birds each. Supplementing with jack beans significantly enhanced butyrate production (P < 0.001), while at 10 % supplementation did not differ from control. Villus height (VH) and the ratio (VH:CD) were significantly (P < 0.001) increased by dietary treatments, while villus width (VW) and crypt depth (CD) were significantly (P < 0.05) decreased. TLR-3, TNF-a, and IL-6 were all significantly (P < 0.001) increased by dietary supplementation. However, at 15 %, TLR-3 and IL-6 were same with control. IL-18 was significantly (P < 0.05) decreased at 15 %. IL-10 decreased significantly (P < 0.001), but at 10 % same with control. At 5 and 10 %, IL-13 increased significantly (P < 0.001), whereas dietary treatments decreased at 15 % compared to control. Although ZO1 decreased significantly (P < 0.001) and OLCN increased significantly (P < 0.001), both ZO1 and OCLN were not significantly different from the control at 15 %. Dietary treatments significantly (P < 0.001) increased CLDN1 but did not differ from the control at 10 %. JAM2 decreased significantly (P < 0.001) with dietary treatments. In conclusion, jack bean supplementation may increase broiler chicken performance and intestinal health due to butyrate production. It may affect intestinal morphology and integrity by upregulating a tight junction protein gene. Jack beans also impacted jejunum immune responses and inflammatory cytokine gene expression.
Collapse
Affiliation(s)
- Abd Majid Ahmad Madani
- Animal Nutrition and Feed Science Department, Faculty of Animal Science, Universitas Gadjah Mada, Indonesia
| | - Muhlisin Muhlisin
- Animal Nutrition and Feed Science Department, Faculty of Animal Science, Universitas Gadjah Mada, Indonesia
| | - Asih Kurniawati
- Animal Nutrition and Feed Science Department, Faculty of Animal Science, Universitas Gadjah Mada, Indonesia
| | - Aji Praba Baskara
- Animal Nutrition and Feed Science Department, Faculty of Animal Science, Universitas Gadjah Mada, Indonesia
| | - Muhsin Al Anas
- Animal Nutrition and Feed Science Department, Faculty of Animal Science, Universitas Gadjah Mada, Indonesia
| |
Collapse
|
4
|
Li J, Zhao J, Tian C, Dong L, Kang Z, Wang J, Zhao S, Li M, Tong X. Mechanisms of regulation of glycolipid metabolism by natural compounds in plants: effects on short-chain fatty acids. Nutr Metab (Lond) 2024; 21:49. [PMID: 39026248 PMCID: PMC11256480 DOI: 10.1186/s12986-024-00829-5] [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: 05/15/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Natural compounds can positively impact health, and various studies suggest that they regulate glucose‒lipid metabolism by influencing short-chain fatty acids (SCFAs). This metabolism is key to maintaining energy balance and normal physiological functions in the body. This review explores how SCFAs regulate glucose and lipid metabolism and the natural compounds that can modulate these processes through SCFAs. This provides a healthier approach to treating glucose and lipid metabolism disorders in the future. METHODS This article reviews relevant literature on SCFAs and glycolipid metabolism from PubMed and the Web of Science Core Collection (WoSCC). It also highlights a range of natural compounds, including polysaccharides, anthocyanins, quercetins, resveratrols, carotenoids, and betaines, that can regulate glycolipid metabolism through modulation of the SCFA pathway. RESULTS Natural compounds enrich SCFA-producing bacteria, inhibit harmful bacteria, and regulate operational taxonomic unit (OTU) abundance and the intestinal transport rate in the gut microbiota to affect SCFA content in the intestine. However, most studies have been conducted in animals, lack clinical trials, and involve fewer natural compounds that target SCFAs. More research is needed to support the conclusions and to develop healthier interventions. CONCLUSIONS SCFAs are crucial for human health and are produced mainly by the gut microbiota via dietary fiber fermentation. Eating foods rich in natural compounds, including fruits, vegetables, tea, and coarse fiber foods, can hinder harmful intestinal bacterial growth and promote beneficial bacterial proliferation, thus increasing SCFA levels and regulating glucose and lipid metabolism. By investigating how these compounds impact glycolipid metabolism via the SCFA pathway, novel insights and directions for treating glucolipid metabolism disorders can be provided.
Collapse
Affiliation(s)
- Jiarui Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jinyue Zhao
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Chuanxi Tian
- Beijing University of Chinese Medicine, Beijing, China
| | - Lishuo Dong
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Zezheng Kang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jingshuo Wang
- The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Shuang Zhao
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Min Li
- Research Laboratory of Molecular Biology, Guang'anmen Hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xiaolin Tong
- Guang'anmen Hospital, Academician of Chinese Academy of Sciences, China Academy of Traditional Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
5
|
Facchin S, Bertin L, Bonazzi E, Lorenzon G, De Barba C, Barberio B, Zingone F, Maniero D, Scarpa M, Ruffolo C, Angriman I, Savarino EV. Short-Chain Fatty Acids and Human Health: From Metabolic Pathways to Current Therapeutic Implications. Life (Basel) 2024; 14:559. [PMID: 38792581 PMCID: PMC11122327 DOI: 10.3390/life14050559] [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: 03/25/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
The gastrointestinal tract is home to trillions of diverse microorganisms collectively known as the gut microbiota, which play a pivotal role in breaking down undigested foods, such as dietary fibers. Through the fermentation of these food components, short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate are produced, offering numerous health benefits to the host. The production and absorption of these SCFAs occur through various mechanisms within the human intestine, contingent upon the types of dietary fibers reaching the gut and the specific microorganisms engaged in fermentation. Medical literature extensively documents the supplementation of SCFAs, particularly butyrate, in the treatment of gastrointestinal, metabolic, cardiovascular, and gut-brain-related disorders. This review seeks to provide an overview of the dynamics involved in the production and absorption of acetate, propionate, and butyrate within the human gut. Additionally, it will focus on the pivotal roles these SCFAs play in promoting gastrointestinal and metabolic health, as well as their current therapeutic implications.
Collapse
Affiliation(s)
- Sonia Facchin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Luisa Bertin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Erica Bonazzi
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Greta Lorenzon
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Caterina De Barba
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Brigida Barberio
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Fabiana Zingone
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Daria Maniero
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Marco Scarpa
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Cesare Ruffolo
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Imerio Angriman
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Edoardo Vincenzo Savarino
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| |
Collapse
|
6
|
Yu S, Wang X, Li Z, Jin D, Yu M, Li J, Li Y, Liu X, Zhang Q, Liu Y, Liu R, Wang X, Fang B, Zhang C, Wang R, Ren F. Solobacterium moorei promotes the progression of adenomatous polyps by causing inflammation and disrupting the intestinal barrier. J Transl Med 2024; 22:169. [PMID: 38368407 DOI: 10.1186/s12967-024-04977-3if:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/11/2024] [Indexed: 07/26/2024] Open
Abstract
BACKGROUND Adenomatous polyps (APs) with inflammation are risk factors for colorectal cancer. However, the role of inflammation-related gut microbiota in promoting the progression of APs is unknown. METHODS Sequencing of the 16S rRNA gene was conducted to identify characteristic bacteria in AP tissues and normal mucosa. Then, the roles of inflammation-related bacteria were clarified by Spearman correlation analysis. Furthermore, colorectal HT-29 cells, normal colon NCM460 cells, and azoxymethane-treated mice were used to investigate the effects of the characteristic bacteria on progression of APs. RESULTS The expression levels of inflammation-related markers (diamine oxidase, D-lactate, C-reactive protein, tumor necrosis factor-α, interleukin-6 and interleukin-1β) were increased, whereas the expression levels of anti-inflammatory factors (interleukin-4 and interleukin-10) were significantly decreased in AP patients as compared to healthy controls. Solobacterium moorei (S. moorei) was enriched in AP tissues and fecal samples, and significantly positively correlated with serum inflammation-related markers. In vitro, S. moorei preferentially attached to HT-29 cells and stimulated cell proliferation and production of pro-inflammatory factors. In vivo, the incidence of intestinal dysplasia was significantly increased in the S. moorei group. Gavage of mice with S. moorei upregulated production of pro-inflammatory factors, suppressed proliferation of CD4+ and CD8+cells, and disrupted the integrity of the intestinal barrier, thereby accelerating progression of APs. CONCLUSIONS S. moorei accelerated the progression of AP in mice via activation of the NF-κB signaling pathway, chronic low-grade inflammation, and intestinal barrier disruption. Targeted reduction of S. moorei presents a potential strategy to prevent the progression of APs.
Collapse
Affiliation(s)
- Shoujuan Yu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Xifan Wang
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, 10032, USA
| | - Ziyang Li
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Dekui Jin
- Department of General Practice, The Third Centers of Chinese PLA General Hospital, Beijing, 100039, China
| | - Mengyang Yu
- Department of General Practice, The Third Centers of Chinese PLA General Hospital, Beijing, 100039, China
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Yixuan Li
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Xiaoxue Liu
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Qi Zhang
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Yinghua Liu
- Department of Nutrition, The First Center of Chinese PLA General Hospital, Beijing, 100037, China
| | - Rong Liu
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Xiaoyu Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Bing Fang
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Chengying Zhang
- Department of General Practice, The Third Centers of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Ran Wang
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China.
| | - Fazheng Ren
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
7
|
Yu S, Wang X, Li Z, Jin D, Yu M, Li J, Li Y, Liu X, Zhang Q, Liu Y, Liu R, Wang X, Fang B, Zhang C, Wang R, Ren F. Solobacterium moorei promotes the progression of adenomatous polyps by causing inflammation and disrupting the intestinal barrier. J Transl Med 2024; 22:169. [PMID: 38368407 PMCID: PMC10874563 DOI: 10.1186/s12967-024-04977-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/11/2024] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND Adenomatous polyps (APs) with inflammation are risk factors for colorectal cancer. However, the role of inflammation-related gut microbiota in promoting the progression of APs is unknown. METHODS Sequencing of the 16S rRNA gene was conducted to identify characteristic bacteria in AP tissues and normal mucosa. Then, the roles of inflammation-related bacteria were clarified by Spearman correlation analysis. Furthermore, colorectal HT-29 cells, normal colon NCM460 cells, and azoxymethane-treated mice were used to investigate the effects of the characteristic bacteria on progression of APs. RESULTS The expression levels of inflammation-related markers (diamine oxidase, D-lactate, C-reactive protein, tumor necrosis factor-α, interleukin-6 and interleukin-1β) were increased, whereas the expression levels of anti-inflammatory factors (interleukin-4 and interleukin-10) were significantly decreased in AP patients as compared to healthy controls. Solobacterium moorei (S. moorei) was enriched in AP tissues and fecal samples, and significantly positively correlated with serum inflammation-related markers. In vitro, S. moorei preferentially attached to HT-29 cells and stimulated cell proliferation and production of pro-inflammatory factors. In vivo, the incidence of intestinal dysplasia was significantly increased in the S. moorei group. Gavage of mice with S. moorei upregulated production of pro-inflammatory factors, suppressed proliferation of CD4+ and CD8+cells, and disrupted the integrity of the intestinal barrier, thereby accelerating progression of APs. CONCLUSIONS S. moorei accelerated the progression of AP in mice via activation of the NF-κB signaling pathway, chronic low-grade inflammation, and intestinal barrier disruption. Targeted reduction of S. moorei presents a potential strategy to prevent the progression of APs.
Collapse
Affiliation(s)
- Shoujuan Yu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Xifan Wang
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, 10032, USA
| | - Ziyang Li
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Dekui Jin
- Department of General Practice, The Third Centers of Chinese PLA General Hospital, Beijing, 100039, China
| | - Mengyang Yu
- Department of General Practice, The Third Centers of Chinese PLA General Hospital, Beijing, 100039, China
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Yixuan Li
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Xiaoxue Liu
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Qi Zhang
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Yinghua Liu
- Department of Nutrition, The First Center of Chinese PLA General Hospital, Beijing, 100037, China
| | - Rong Liu
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Xiaoyu Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Bing Fang
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China
| | - Chengying Zhang
- Department of General Practice, The Third Centers of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Ran Wang
- Key Laboratory of Functional Dairy, Co-Constructed By Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, 100190, China.
| | - Fazheng Ren
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
8
|
Chu Z, Hu Z, Luo Y, Zhou Y, Yang F, Luo F. Targeting gut-liver axis by dietary lignans ameliorate obesity: evidences and mechanisms. Crit Rev Food Sci Nutr 2023; 65:243-264. [PMID: 37870876 DOI: 10.1080/10408398.2023.2272269] [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] [Indexed: 10/24/2023]
Abstract
An imbalance between energy consumption and energy expenditure causes obesity. It is characterized by increased adipose accumulation and accompanied by chronic low-grade inflammation. Many studies have suggested that the gut microbiota of the host mediates the relationship between high-fat diet consumption and the development of obesity. Diet and nutrition of the body are heavily influenced by gut microbiota. The alterations in the microbiota in the gut may have effects on the homeostasis of the host's energy levels, systemic inflammation, lipid metabolism, and insulin sensitivity. The liver is an important organ for fat metabolism and gut-liver axis play important role in the fat metabolism. Gut-liver axis is a bidirectional relationship between the gut and its microbiota and the liver. As essential plant components, lignans have been shown to have different biological functions. Accumulating evidences have suggested that lignans may have lipid-lowering properties. Lignans can regulate the level of the gut microbiota and their metabolites in the host, thereby affecting signaling pathways related to fat synthesis and metabolism. These signaling pathways can make a difference in inhibiting fat accumulation, accelerating energy metabolism, affecting appetite, and inhibiting chronic inflammation. It will provide the groundwork for future studies on the lipid-lowering impact of lignans and the creation of functional meals based on those findings.
Collapse
Affiliation(s)
- Zhongxing Chu
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| | - Zuomin Hu
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| | - Yi Luo
- Department of Clinic Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, P.R. China
| | - Yaping Zhou
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| | - Feiyan Yang
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| | - Feijun Luo
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| |
Collapse
|
9
|
Pant K, Venugopal SK, Lorenzo Pisarello MJ, Gradilone SA. The Role of Gut Microbiome-Derived Short-Chain Fatty Acid Butyrate in Hepatobiliary Diseases. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1455-1467. [PMID: 37422149 PMCID: PMC10548274 DOI: 10.1016/j.ajpath.2023.06.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/09/2023] [Accepted: 06/21/2023] [Indexed: 07/10/2023]
Abstract
The short-chain fatty acid butyrate, produced from fermentable carbohydrates by gut microbiota in the colon, has multiple beneficial effects on human health. At the intestinal level, butyrate regulates metabolism, helps in the transepithelial transport of fluids, inhibits inflammation, and induces the epithelial defense barrier. The liver receives a large amount of short-chain fatty acids via the blood flowing from the gut via the portal vein. Butyrate helps prevent nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, inflammation, cancer, and liver injuries. It ameliorates metabolic diseases, including insulin resistance and obesity, and plays a direct role in preventing fatty liver diseases. Butyrate has different mechanisms of action, including strong regulatory effects on the expression of many genes by inhibiting the histone deacetylases and modulating cellular metabolism. The present review highlights the wide range of beneficial therapeutic and unfavorable adverse effects of butyrate, with a high potential for clinically important uses in several liver diseases.
Collapse
Affiliation(s)
- Kishor Pant
- The Hormel Institute, University of Minnesota, Austin, Minnesota.
| | - Senthil K Venugopal
- Laboratory of Molecular Medicine and Hepatology, Faculty of Life Science and Biotechnology, South Asian University, New Delhi, India
| | - Maria J Lorenzo Pisarello
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA), National Council of Scientific and Technological Research, San Miguel de Tucuman, Argentina; Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Sergio A Gradilone
- The Hormel Institute, University of Minnesota, Austin, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
10
|
Yu C, Liu J, Liu Z, Ding Q, Zhu Q, Chen N, Fu J, Valencak TG, Ren D. Lactobacillus plantarum ZJUIDS04 alleviates DSS-induced colitis via modulating gut microbiota. J Funct Foods 2023; 109:105794. [DOI: 10.1016/j.jff.2023.105794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
11
|
Couto M, Andrade N, Magro F, Martel F. Taurocholate uptake by Caco-2 cells is inhibited by pro-inflammatory cytokines and butyrate. Cytokine 2023; 169:156307. [PMID: 37487380 DOI: 10.1016/j.cyto.2023.156307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/26/2023]
Abstract
Inflammatory bowel disease (IBD) is a group of chronic and life-threating inflammatory diseases of the gastrointestinal tract. The active intestinal absorption of bile salts is reduced in IBD, resulting in higher luminal concentrations of these agents that contribute to the pathophysiology of IBD-associated diarrhea. Butyrate (BT) is a short-chain fatty acid produced by colonic bacterial fermentation of dietary fibers. BT utilization is impaired in the intestinal inflamed mucosa of IBD patients. Our aim was to investigate the link between IBD and bile acid absorption, by testing the effect of the pro-inflammatory cytokines TNF-α and IFN-γ and of BT upon 3H-TC uptake by Caco-2 cells. The proinflammatory cytokines TNF-α and IFN-γ inhibit Na+-independent, non-ASBT (sodium-dependent bile acid transporter)-mediated 3H-TC uptake by Caco-2 cells. The inhibitory effect of these cytokines on Na+-independent 3H-TC uptake is PI3K- and JAK/STAT1-mediated. These two compounds upregulate ASBT expression levels, but no corresponding increase in Na+-dependent component of 3H-TC is observed. Moreover, BT was also found to inhibit 3H-TC uptake and showed an additive effect with IFN-γ in reducing 3H-TC uptake. We conclude that an interaction between BT and bile acids appears to exist in IBD, which may participate in the link between diet, microbiota and IBD.
Collapse
Affiliation(s)
- Mafalda Couto
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Nelson Andrade
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Portugal
| | - Fernando Magro
- Unit of Pharmacology and Therapeutics, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Gastroenterology Unit, Department of Medicine, Centro Hospitalar S. João, Porto, Portugal
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal.
| |
Collapse
|
12
|
Zhang X, Wang Y, Fan R, Zhang L, Li Z, Zhang Y, Zheng W, Wang L, Liu B, Quan C. Quantitative Proteomic Analysis of Outer Membrane Vesicles from Fusobacterium nucleatum Cultivated in the Mimic Cancer Environment. Microbiol Spectr 2023; 11:e0039423. [PMID: 37341631 PMCID: PMC10434195 DOI: 10.1128/spectrum.00394-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 05/25/2023] [Indexed: 06/22/2023] Open
Abstract
Fusobacterium nucleatum is a Gram-negative bacterium that has been identified as an important pathogenic gut bacterium associated with colorectal cancer. Compared with the normal intestine, the pH value of the tumor microenvironment is weakly acidic. The metabolic changes of F. nucleatum in the tumor microenvironment, especially the protein composition of its outer membrane vesicles, remain unclear. Here, we systematically analyzed the effect of environmental pH on the proteome of outer membrane vesicles (OMVs) from F. nucleatum by tandem mass tag (TMT) labeling-high-resolution liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis. A total of 991 proteins were identified in acidic OMVs (aOMVs) and neutral OMVs (nOMVs), including known virulence proteins and putative virulence proteins. Finally, 306 upregulated proteins and 360 downregulated proteins were detected in aOMVs, and approximately 70% of the expression of OMV proteins was altered under acidic conditions. A total of 29 autotransporters were identified in F. nucleatum OMVs, and 13 autotransporters were upregulated in aOMVs. Interestingly, three upregulated autotransporters (D5REI9, D5RD69, and D5RBW2) show homology to the known virulence factor Fap2, suggesting that they may be involved in various pathogenic pathways such as the pathway for binding with colorectal cancer cells. Moreover, we found that more than 70% of MORN2 domain-containing proteins may have toxic effects on host cells. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses demonstrated that a number of proteins were significantly enriched in multiple pathways involving fatty acid synthesis and butyrate synthesis. Seven metabolic enzymes involved in fatty acid metabolism pathways were identified in the proteomic data, of which 5 were upregulated and 2 were downregulated in aOMVs, while 14 metabolic enzymes involved in the butyric acid metabolic pathway were downregulated in aOMVs. In conclusion, we found a key difference in virulence proteins and pathways in the outer membrane vesicles of F. nucleatum between the tumor microenvironment pH and normal intestinal pH, which provides new clues for the prevention and treatment of colorectal cancer. IMPORTANCE F. nucleatum is an opportunistic pathogenic bacterium that can be enriched in colorectal cancer tissues, affecting multiple stages of colorectal cancer development. OMVs have been demonstrated to play key roles in pathogenesis by delivering toxins and other virulence factors to host cells. By employing quantitative proteomic analysis, we found that the pH conditions could affect the protein expression of the outer membrane vesicles of F. nucleatum. Under acidic conditions, approximately 70% of the expression of proteins in OMVs was altered. Several virulence factors, such as type 5a secreted autotransporter (T5aSSs) and membrane occupation and recognition nexus (MORN) domain-containing proteins, were upregulated under acidic conditions. A large number of proteins showed significant enrichments in multiple pathways involving fatty acid synthesis and butyrate synthesis. Proteomics analysis of the outer membrane vesicles secreted by pathogenic bacteria in the acidic tumor microenvironment is of great significance for elucidating the pathogenicity mechanism and its application in vaccine and drug delivery vehicles.
Collapse
Affiliation(s)
- Xuqiang Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Yuxin Wang
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Ruochen Fan
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, Liaoning, China
| | - Liying Zhang
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, Liaoning, China
| | - Zhuting Li
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Yanmei Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Wei Zheng
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Lulu Wang
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, Liaoning, China
| | - Baoquan Liu
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| | - Chunshan Quan
- Key Laboratory of Biotechnology and Bioresources Utilization of the Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
| |
Collapse
|
13
|
Couto MR, Andrade N, Magro F, Martel F. Bile salts and proinflammatory cytokines inhibit MCT1-mediated cellular uptake of butyrate and interfere with its antiproliferative properties. Exp Cell Res 2023; 429:113670. [PMID: 37290498 DOI: 10.1016/j.yexcr.2023.113670] [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: 11/25/2022] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023]
Abstract
Butyrate (BT) is important in the prevention and inhibition of colorectal cancer (CRC). Inflammatory bowel disease, a risk factor for CRC, is associated with higher levels of proinflammatory cytokines and bile acids. The aim of this work was to investigate the interaction of these compounds in inhibiting BT uptake by Caco-2 cells, as a mechanism contributing to the link between IBD and CRC. TNF-α, IFN-γ, chenodeoxycholic acid (CDCA) and deoxycholic acid (DCA) markedly reduce 14C-BT uptake. All these compounds appear to inhibit MCT1-mediated BT cellular uptake at a posttranscriptional level, and, because their effect is not additive, they are most probably inhibiting MCT1 by a similar mechanism. Correspondingly, the antiproliferative effect of BT (MCT1-dependent) and of the proinflammatory cytokines and CDCA were not additive. In contrast, the cytotoxic effect of BT (MCT1-independent) and of the proinflammatory cytokines and CDCA were additive. In conclusion, proinflammatory cytokines (TNF-α and IFN-γ) and bile acids (DCA and CDCA) inhibit MCT1-mediated BT cellular uptake. These proinflammatory cytokines and CDCA were found to interfere with the antiproliferative effect of BT, mediated by an inhibitory effect upon MCT1-mediated cellular uptake of BT.
Collapse
Affiliation(s)
- Mafalda R Couto
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal
| | - Nelson Andrade
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal
| | - Fernando Magro
- Unit of Pharmacology and Therapeutics, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Gastroenterology Unit, Department of Medicine, Centro Hospitalar S. João, Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal.
| |
Collapse
|
14
|
Vega-Cárdenas M, Martínez-Gutierrez F, Lara-Ramírez EE, Reynaga-Hernandez E, Yañez-Estrada L, Ratering S, Schnell S, Godínez-Hernández CI, Vargas-Morales JM, Portales-Pérez DP. Agave fructans enhance the effects of fermented milk products on obesity biomarkers: a randomised trial. Benef Microbes 2023; 14:153-164. [PMID: 36856122 DOI: 10.3920/bm2022.0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Dysbiosis has been implicated in childhood obesity. Oral intake of fermented milk containing Lacticaseibacillus casei strain Shirota preserves gut microbiota (GM) diversity in children and adults. This study was a double-blind trial involving 37 overweight or obese children aged 6-10 years. Children were followed over a 6-week intervention period in which they received different fermented milk products containing L. casei Shirota: 10 in the first group received just L. casei Shirota; 13 received L. casei Shirota with 3 g/day of inulin (L. casei+inulin); and 14 received L. casei Shirota with 3 g/day of fructans from Agave salmiana (L. casei+fructans). Principal component analysis showed the relationship between microbial abundance, GM metabolites, and other obesity-related markers. Supplementation with probiotics and synbiotics improved the HDL-cholesterol levels of overweight and obese children, although no changes in body composition were detected. We observed an increase in butyrate or propionate concentrations in the L. casei+fructans group compared to the end of the intervention (P<0.03). A diminished level of ANGPTL4 within the L. casei+fructans group (P=0.04) was also found, but no differences when lipopolysaccharide-binding protein was evaluated. The FFAR2+ cell frequency decreased between baseline and at the end of 6-week intervention in L. casei+inulin (P=0.02) and L. casei+fructans groups (P=0.04). In contrast, the percentage of CD14+FFAR3+ frequency increased in the same groups (P=0.04). The L. casei Shirota with inulin or fructans modulates GM, which improves the lipid profile and changes at a molecular level, such as expression of FFAR3 and FFAR2, ANGPTL4, propionate, and butyrate. It, therefore, could be considered an interesting therapeutic possibility for treating childhood overweight and obesity. The study was registered at ClinicalTrials.gov (ID: NCT05423015).
Collapse
Affiliation(s)
- M Vega-Cárdenas
- Research Centre for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí (UASLP), Av. Sierra Leona 550, Lomas de San Luis 78210, San Luis Potosí, SLP, Mexico
| | - F Martínez-Gutierrez
- Research Centre for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí (UASLP), Av. Sierra Leona 550, Lomas de San Luis 78210, San Luis Potosí, SLP, Mexico.,Faculty of Chemical Sciences, UASLP, Av. Dr. Manuel Nava 6, Zona Universitaria 78210, San Luis Potosí, SLP, Mexico
| | - E E Lara-Ramírez
- Zacatecas Biomedical Research Unit, Mexican Social Security Institute (IMSS), Alameda Trinidad García de La Cadena 438, Zacatecas Centro 98000 Zacatecas, Zac, Mexico
| | - E Reynaga-Hernandez
- Faculty of Chemical Sciences, UASLP, Av. Dr. Manuel Nava 6, Zona Universitaria 78210, San Luis Potosí, SLP, Mexico
| | - L Yañez-Estrada
- Faculty of Medicine, UASLP, Av. Dr. Manuel Nava 6, Zona Universitaria 78210, San Luis Potosí, SLP, Mexico
| | - S Ratering
- Institute of Applied Microbiology, Justus Liebig University, Schubertstr. 81 35392, Giessen, Germany
| | - S Schnell
- Institute of Applied Microbiology, Justus Liebig University, Schubertstr. 81 35392, Giessen, Germany
| | - C I Godínez-Hernández
- Desert Zones Research Institute, UASLP, De Altair 200, Col del Llano, 78377, San Luis Potosí, SLP, Mexico
| | - J M Vargas-Morales
- Faculty of Chemical Sciences, UASLP, Av. Dr. Manuel Nava 6, Zona Universitaria 78210, San Luis Potosí, SLP, Mexico
| | - D P Portales-Pérez
- Research Centre for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí (UASLP), Av. Sierra Leona 550, Lomas de San Luis 78210, San Luis Potosí, SLP, Mexico.,Faculty of Chemical Sciences, UASLP, Av. Dr. Manuel Nava 6, Zona Universitaria 78210, San Luis Potosí, SLP, Mexico
| |
Collapse
|
15
|
Xue Z, Li R, Liu J, Zhou J, Zhang X, Zhang T, Zhang M, Yang Y, Chen H. Preventive and synbiotic effects of the soluble dietary fiber obtained from Lentinula edodes byproducts and Lactobacillus plantarum LP90 against dextran sulfate sodium-induced colitis in mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:616-626. [PMID: 36054505 DOI: 10.1002/jsfa.12173] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/22/2022] [Accepted: 08/11/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Soluble dietary fiber (SDF) obtained from Lentinula edodes byproducts has beneficial effects on human intestinal health. This study aimed to examine the combined preventive and ameliorative effects of a kind of synbiotic (SDF with a molecular weight of 1.58 × 102 kDa and Lactobacillus plantarum LP90 (LP) at 1 × 109 CFU kg-1 ) on dextran sulfate sodium-induced colitis mice. RESULTS The results demonstrated that synbiotic treatment could alleviate weight loss, decrease the disease activity index level and cause histological amelioration. Synbiotic treatment also promoted the production of goblet cells, increased the expression of tight junction proteins, and adjusted the production of myeloperoxidase, malondialdehyde and superoxide dismutase to repair intestinal epithelial injury. Clinical symptoms were alleviated by maintaining Th17/Treg balance, increasing interleukin 10 and immunoglobulin A levels, reducing interleukin 17a and tumor necrosis factor α production, and promoting mRNA to highly express of Foxp3 and vitamin D receptors. Moreover, synbiotic treatment could upregulate butyric acid production (4.71 ± 0.46 mol g-1 feces, P < 0.05) and diversity of intestinal microbial to maintain intestinal homeostasis. CONCLUSION This study suggested that the combination of LP and SDF as a synbiotic has the potential for use as a nutritional supplement to alleviate colitis. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zihan Xue
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Ruilin Li
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Junyu Liu
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Jingna Zhou
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Xiaoyu Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Tingting Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Min Zhang
- College of Food Science and Bioengineering, Tianjin Agricultural University, Tianjin, PR China
- State Key Laboratory of Nutrition and Safety, Tianjin University of Science and Technology, Tianjin, PR China
| | - Yang Yang
- Department of Orthopedics, Tianjin Hospital, Tianjin, PR China
| | - Haixia Chen
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| |
Collapse
|
16
|
Liu S, Yang X. Intestinal flora plays a role in the progression of hepatitis-cirrhosis-liver cancer. Front Cell Infect Microbiol 2023; 13:1140126. [PMID: 36968098 PMCID: PMC10034054 DOI: 10.3389/fcimb.2023.1140126] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/23/2023] [Indexed: 03/29/2023] Open
Abstract
The liver is a vital metabolism and detoxification organ of human body, which is involved in the biotransformation and metabolism of the organism. Hepatitis - cirrhosis - liver cancer are significant and common part of liver diseases. The pathogenesis of liver diseases is generally as followed: inflammation and other pathogenic factors cause persistent damage to the liver, leading to the activation of hepatic stellate cells (HSCs) and excessive deposition of extracellular matrix. Patients with chronic hepatitis have a high risk of developing into liver fibrosis, cirrhosis, and even life-threatening liver cancer, which poses a great threat to public health.As the first organ to come into contact with blood from the gut, the liver is profoundly affected by the intestinal flora and its metabolites, with leaky gut and flora imbalance being the triggers of the liver's pathological response. So far, no one has reviewed the role of intestinal flora in this process from the perspective of the progression of hepatitis-cirrhosis-liver cancer and this article reviews the evidence supporting the effect of intestinal flora in the progression of liver disease.
Collapse
|
17
|
Abstract
The diet and gut microbiota have been extensively interrogated as a fuel for gut inflammation in inflammatory bowel diseases (IBDs) in the last few years. Here, we review how specific nutrients, typically enriched in a Western diet, instigate or deteriorate experimental gut inflammation in a genetically susceptible host and we discuss microbiota-dependent and independent mechanisms. We depict the study landscape of nutritional trials in paediatric and adult IBD and delineate common grounds for dietary advice. Conclusively, the diet reflects a critical rheostat of microbial dysbiosis and gut inflammation in IBD. Dietary restriction by exclusive enteral nutrition, with or without a specific exclusion diet, is effectively treating paediatric Crohn's disease, while adult IBD trials are less conclusive. Insights into molecular mechanisms of nutritional therapy will change the perception of IBD and will allow us to enter the era of precision nutrition. To achieve this, we discuss the need for carefully designed nutritional trials with scientific rigour comparable to medical trials, which also requires action from stake holders. Establishing evidence-based dietary therapy for IBD does not only hold promise to avoid long-term immunosuppression, but to provide a widely accessible therapy at low cost. Identification of dietary culprits disturbing gut health also bears the potential to prevent IBD and allows informed decision making in food politics.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Medicine I, Gastroenterology, Hepatology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Jingwan Zhang
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
18
|
Sori N, Kunnummal SP, Peddha MS, Khan M. Prophylactic effect of pectic oligosaccharides against poly I: C- induced virus-like infection in BALB/c mice. J Food Biochem 2022; 46:e14459. [PMID: 36240117 DOI: 10.1111/jfbc.14459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/08/2022] [Accepted: 09/21/2022] [Indexed: 01/14/2023]
Abstract
Pectin oligosaccharides (POS) are pectin-derived prebiotics that exerts anti-inflammatory effects on the host and stimulates an innate immune response. The role of POS in protective immunity against viral infections is not very obvious. Therefore, the prophylactic effect of POS in the mouse model induced by Poly I: C mimicking viral infection was examined. Mice fed POS showed a significant (p ≤ .05) increase in IgG, sIgA, IgA, IL-12, and a significant (p ≤ .05) decrease in the concentration of pro-inflammatory cytokines IL-5, IL-6, IL-13 and IL-17 in lung and blood serum after Poly I: C stimulation. However, the control group could not inhibit pro-inflammatory cytokines. POS also promoted the growth of the Lactobacillus, Prevotella, Rilenellaceae, and Lachanospiraceae groups. Therefore, this study demonstrate that POS has the potiential to protect against viral inflammation by altering gut microbiota and activating mucosal immunity. PRACTICAL APPLICATIONS: POS is 2-10 mer oligomers of pectin. The human gastrointestinal tract lacks the enzyme to break down POS. They are fermented by gut bacteria in the colon and stimulate the proliferation of specific gut bacteria that are positively correlated with the production of anti-inflammatory cytokines and SCFA. POS also stimulates the secretion of IgA, which inhibits bacterial and viral adhesion and protects the host. Therefore, POS can be used as a functional food ingredient in food to stimulate a specific group of gut bacteria and enhance preventive immunity.
Collapse
Affiliation(s)
- Nidhi Sori
- Department of Microbiology and Fermentation Technology, CSIR - Central Food Technological Research Institute, Mysuru, India
| | - Saarika Pothuvan Kunnummal
- Department of Microbiology and Fermentation Technology, CSIR - Central Food Technological Research Institute, Mysuru, India.,CSIR-Academy of Scientific & Innovative Research, Ghaziabad, India
| | - MuthuKumar Serva Peddha
- CSIR-Academy of Scientific & Innovative Research, Ghaziabad, India.,Department of Biochemistry, CSIR - Central Food Technological Research Institute, Mysuru, India
| | - Mahejibin Khan
- Department of Microbiology and Fermentation Technology, CSIR - Central Food Technological Research Institute, Mysuru, India.,CSIR-Academy of Scientific & Innovative Research, Ghaziabad, India
| |
Collapse
|
19
|
Guo XR, He CW, Gao H, Hua RX, Liang C, Du YX, Shang HW, Lu X, Xu JD. Insight into role of short chain fatty acids in regulating intestinal mucosal barrier and alleviating inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2022; 30:928-940. [DOI: 10.11569/wcjd.v30.i21.928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
In recent years, the importance of intestinal microbiota and its metabolites in maintaining the human intestinal environment has been gradually revealed. Therefore, short chain fatty acids (SCFAs), as the metabolites produced by the intestinal microbiota, play a momentous part in regulating the balance between the function and morphology of the mucosal barrier, regulating the proliferation and differentiation of mucosal cells, protecting the integrity and permeability of the mucosal barrier, and maintainingthe stability of tight junctions. Inflammatory bowel disease (IBD) is a chronic, inflammatory condition of the gastrointestinal tract, associated with a disturbance of intestinal barrier function and dysregulation of the intestinal immune responses, the etiology and pathogenesis of which, however, are not yet fully uncovered. Animal models and human studies have corroborated the contribution of SCFAs in enhancing the barrier function through protective effects. This review will summarize the potential role of SCFAs in IBD with regard to regulating intestinal function, hoping to provide a new target for clinical treatment of IBD.
Collapse
Affiliation(s)
- Xue-Ran Guo
- 2019 Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Cheng-Wei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Han Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Rong-Xuan Hua
- 2020 Clinical Medicine of "5+3" Program, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Chen Liang
- 2019 Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yi-Xuan Du
- 2020 Oral Medicine of "5+3" Program, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Hong-Wei Shang
- Teaching Laboratory of Morphology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xin Lu
- Teaching Laboratory of Morphology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jing-Dong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| |
Collapse
|
20
|
Tan YL, Zhao YE. Advances in understanding of effects of dietary nutrients in inflammatory bowel disease patients. Shijie Huaren Xiaohua Zazhi 2022; 30:921-927. [DOI: 10.11569/wcjd.v30.i21.921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The occurrence of inflammatory bowel disease (IBD) is influenced by environmental factors. Diet, as an important environmental factor, is closely associated with intestinal ecology and plays a critical role in the pathological process of IBD. This review summarizes the role of major dietary nutrients in the pathogenesis of IBD, with an aim to provide clues for the prevention and treatment of IBD.
Collapse
Affiliation(s)
- Yan-Li Tan
- Department of Gastroenterology, TongJi Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Yu-E Zhao
- Department of Gastroenterology, TongJi Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| |
Collapse
|
21
|
Sommer KM, Jespersen JC, Sutkus LT, Lee Y, Donovan SM, Dilger RN. Oral gamma-cyclodextrin-encapsulated tributyrin supplementation in young pigs with experimentally induced colitis. J Anim Sci 2022; 100:skac314. [PMID: 36161319 PMCID: PMC9671115 DOI: 10.1093/jas/skac314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Disruption of intestinal integrity and barrier function due to tissue inflammation has negative implications on overall growth and well-being in young pigs. In this study, we investigated the effects of oral gamma-cyclodextrin-encapsulated tributyrin (TBCD) in young pigs experiencing dextran sodium sulfate (DSS)-induced colitis. Pigs (n = 32 boars) were weaned from the sow at postnatal day (PND) 2, allotted to treatment based on the litter of origin and body weight (BW), and reared artificially over a 26-d feeding period. Treatment groups included: 1) nutritionally adequate (control) milk replacer, no DSS (Control n = 8), 2) control milk replacer plus oral DSS (DSS, n = 7), and 3) control diet supplemented with 8.3 g of TBCD per kg of reconstituted milk replacer plus oral DSS (TBCD + DSS, n = 8). Colitis was induced by administering DSS at 1.25 g of DSS/kg BW daily in a reconstituted milk replacer from PND 14-18. Milk replacer and water were provided ad libitum throughout the 26-d study. All the data were analyzed using a one-way ANOVA using the MIXED procedure of SAS. Control and DSS pigs had similar BW throughout the study, while TBCD + DSS pigs exhibited decreased (P < 0.05) BW starting at approximately PND 15. Additionally, average daily gain (ADG) before and after initiation of DSS dosing, along with over the total study duration, was decreased (P < 0.05) in pigs receiving TBCD + DSS compared with the Control. Milk disappearance was decreased (P < 0.05) in TBCD + DSS pigs when compared with Control and DSS groups. Both the concentration and molar ratio of cecal butyrate concentrations were increased (P < 0.05) in TBCD + DSS pigs compared with the Control group. The DSS and TBCD + DSS treatments also increased (P < 0.05) butyrate concentrations in the luminal contents with the proximal colon compared with Control. TBCD + DSS and DSS pigs had increased (P < 0.05) mucosal width in the distal colon compared with Control, thereby indicating heightened intestinal inflammation. Overall, oral supplementation of encapsulated tributyrin increased the concentration of butyrate in the colon, but was unable to mitigate the negative effects of DSS-induced colitis.
Collapse
Affiliation(s)
- Kaitlyn M Sommer
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | | | - Loretta T Sutkus
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | - Youngsoo Lee
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, USA
| | - Sharon M Donovan
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | - Ryan N Dilger
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| |
Collapse
|
22
|
Hong M, Cheng L, Liu Y, Wu Z, Zhang P, Zhang X. Mechanisms Underlying the Interaction Between Chronic Neurological Disorders and Microbial Metabolites via Tea Polyphenols Therapeutics. Front Microbiol 2022; 13:823902. [PMID: 35401435 PMCID: PMC8991060 DOI: 10.3389/fmicb.2022.823902] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/24/2022] [Indexed: 12/14/2022] Open
Abstract
The number of hydroxyl groups and existence of characteristic structural groups in tea polyphenols (TP) make them have antioxidant activity, which gives TP anti-inflammatory effects, toward protecting the intestinal flora and brain neurons. Host-associated microbial metabolites are emerging as dominant modifiers of the central nervous system. As yet, the investigations on host-microbiota crosstalking remain challenging, studies focusing on metabolites such as serotonin, short-chain fatty acids, and others have pinpointed multiple actionable signaling pathways relevant to host health. However, there are still complexities and apparent limitations inherent in transforming complex human diseases to corresponding animal models. Here, we choose to discuss several intestinal metabolites with research value, as crucial areas for assessing TP-mediated chronic brain diseases interactions with microbial.
Collapse
Affiliation(s)
- Mengyu Hong
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Lu Cheng
- Department of Food Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Yanan Liu
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Zufang Wu
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Peng Zhang
- Department of Student Affairs, Xinyang Normal University, Xinyang, China
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| |
Collapse
|
23
|
Kopper JJ, Iennarella-Servantez C, Jergens AE, Sahoo DK, Guillot E, Bourgois-Mochel A, Martinez MN, Allenspach K, Mochel JP. Harnessing the Biology of Canine Intestinal Organoids to Heighten Understanding of Inflammatory Bowel Disease Pathogenesis and Accelerate Drug Discovery: A One Health Approach. FRONTIERS IN TOXICOLOGY 2022; 3:773953. [PMID: 35295115 PMCID: PMC8915821 DOI: 10.3389/ftox.2021.773953] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/07/2021] [Indexed: 12/13/2022] Open
Abstract
In a recent issue of the Lancet, the prevalence of Inflammatory Bowel Disease (IBD) was estimated at 7 million worldwide. Overall, the burden of IBD is rising globally, with direct and indirect healthcare costs ranging between $14.6 and $31.6 billion in the U.S. alone in 2014. There is currently no cure for IBD, and up to 40% of patients do not respond to medical therapy. Although the exact determinants of the disease pathophysiology remain unknown, the prevailing hypothesis involves complex interplay among host genetics, the intestinal microenvironment (primarily bacteria and dietary constituents), and the mucosal immune system. Importantly, multiple chronic diseases leading to high morbidity and mortality in modern western societies, including type II diabetes, IBD and colorectal cancer, have epidemiologically been linked to the consumption of high-calorie, low-fiber, high monosaccharide, and high-fat diets (HFD). More specifically, data from our laboratory and others have shown that repeated consumption of HFD triggers dysbiotic changes of the gut microbiome concomitant with a state of chronic intestinal inflammation and increased intestinal permeability. However, progress in our understanding of the effect of dietary interventions on IBD pathogenesis has been hampered by a lack of relevant animal models. Additionally, current in vitro cell culture systems are unable to emulate the in vivo interplay between the gut microbiome and the intestinal epithelium in a realistic and translatable way. There remains, therefore, a critical need to develop translatable in vitro and in vivo models that faithfully recapitulate human gut-specific physiological functions to facilitate detailed mechanistic studies on the impact of dietary interventions on gut homeostasis. While the study of murine models has been pivotal in advancing genetic and cellular discoveries, these animal systems often lack key clinical signs and temporal pathological changes representative of IBD. Specifically, some limitations of the mouse model are associated with the use of genetic knockouts to induce immune deficiency and disease. This is vastly different from the natural course of IBD developing in immunologically competent hosts, as is the case in humans and dogs. Noteworthily, abundant literature suggests that canine and human IBD share common clinical and molecular features, such that preclinical studies in dogs with naturally occurring IBD present an opportunity to further our understanding on disease pathogenesis and streamline the development of new therapeutic strategies. Using a stepwise approach, in vitro mechanistic studies investigating the contribution of dietary interventions to chronic intestinal inflammation and "gut leakiness" could be performed in intestinal organoids and organoid derived monolayers. The biologic potential of organoids stems from the method's ability to harness hard-wired cellular programming such that the complexity of the disease background can be reflected more accurately. Likewise, the effect of therapeutic drug candidates could be evaluated in organoids prior to longitudinal studies in dog and human patients with IBD. In this review, we will discuss the value (and limitations) of intestinal organoids derived from a spontaneous animal disease model of IBD (i.e., the dog), and how it can heighten understanding of the interplay between dietary interventions, the gut microbiota and intestinal inflammation. We will also review how intestinal organoids could be used to streamline the preclinical development of therapeutic drug candidates for IBD patients and their best four-legged friends.
Collapse
Affiliation(s)
- Jamie J Kopper
- Veterinary Clinical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States.,SMART Translational Medicine, Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States
| | - Chelsea Iennarella-Servantez
- SMART Pharmacology, Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States.,SMART Translational Medicine, Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States
| | - Albert E Jergens
- Veterinary Clinical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States
| | - Dipak K Sahoo
- Veterinary Clinical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States.,SMART Translational Medicine, Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States
| | - Emilie Guillot
- 3D Health Solutions, Inc., ISU Research Park, Ames, IA, United States
| | - Agnes Bourgois-Mochel
- Veterinary Clinical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States
| | - Marilyn N Martinez
- Office of New Animal Drug Evaluation, Center for Veterinary Medicine, Food and Drug Administration, Rockville, MD, United States
| | - Karin Allenspach
- Veterinary Clinical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States.,SMART Translational Medicine, Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States.,3D Health Solutions, Inc., ISU Research Park, Ames, IA, United States
| | - Jonathan P Mochel
- SMART Pharmacology, Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States.,SMART Translational Medicine, Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States.,3D Health Solutions, Inc., ISU Research Park, Ames, IA, United States
| |
Collapse
|
24
|
Huang YA, Huang ZA, Li JQ, You ZH, Wang L, Yi HC, Yu CQ. GBDR: a Bayesian model for precise prediction of pathogenic microorganisms using 16S rRNA gene sequences. BMC Genomics 2022; 22:916. [PMID: 35296232 PMCID: PMC8925046 DOI: 10.1186/s12864-022-08423-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 02/25/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recent evidences have suggested that human microorganisms participate in important biological activities in the human body. The dysfunction of host-microbiota interactions could lead to complex human disorders. The knowledge on host-microbiota interactions can provide valuable insights into understanding the pathological mechanism of diseases. However, it is time-consuming and costly to identify the disorder-specific microbes from the biological "haystack" merely by routine wet-lab experiments. With the developments in next-generation sequencing and omics-based trials, it is imperative to develop computational prediction models for predicting microbe-disease associations on a large scale. RESULTS Based on the known microbe-disease associations derived from the Human Microbe-Disease Association Database (HMDAD), the proposed model shows reliable performance with high values of the area under ROC curve (AUC) of 0.9456 and 0.8866 in leave-one-out cross validations and five-fold cross validations, respectively. In case studies of colorectal carcinoma, 80% out of the top-20 predicted microbes have been experimentally confirmed via published literatures. CONCLUSION Based on the assumption that functionally similar microbes tend to share the similar interaction patterns with human diseases, we here propose a group based computational model of Bayesian disease-oriented ranking to prioritize the most potential microbes associating with various human diseases. Based on the sequence information of genes, two computational approaches (BLAST+ and MEGA 7) are leveraged to measure the microbe-microbe similarity from different perspectives. The disease-disease similarity is calculated by capturing the hierarchy information from the Medical Subject Headings (MeSH) data. The experimental results illustrate the accuracy and effectiveness of the proposed model. This work is expected to facilitate the characterization and identification of promising microbial biomarkers.
Collapse
Affiliation(s)
- Yu-An Huang
- Department of Information Engineering, Xijing University, Xi'an, 710123, China.
| | - Zhi-An Huang
- Center for Computer Science and Information Technology, City University of Hong Kong Dongguan Research Institute, Dongguan, China
| | - Jian-Qiang Li
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, 518000, China.
| | - Zhu-Hong You
- Department of Information Engineering, Xijing University, Xi'an, 710123, China
| | - Lei Wang
- Guangxi Academy of Science, Nanning, 530000, China
| | - Hai-Cheng Yi
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Ürümqi, 830000, China
| | - Chang-Qing Yu
- Department of Information Engineering, Xijing University, Xi'an, 710123, China
| |
Collapse
|
25
|
Kang S, You HJ, Ju Y, Kim HJ, Jeong YJ, Johnston TV, Ji GE, Ku S, Park MS. Butyl-fructooligosaccharides modulate gut microbiota in healthy mice and ameliorate ulcerative colitis in a DSS-induced model. Food Funct 2022; 13:1834-1845. [DOI: 10.1039/d1fo03337a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Butyl-fructooligosaccharides (B-FOSs) are newly synthesized prebiotics composed of short-chain FOS (GF2, 1-kestose; GF3, nystose; GF4, fructofuranosyl-nystose; GF5, 1-F-(1-b-D-fructofuranosyl)-2-nystose) bound with one or two butyric groups by ester bonds. Previous in...
Collapse
|
26
|
Ahn E, Jeong H, Kim E. Differential effects of various dietary proteins on dextran sulfate sodium-induced colitis in mice. Nutr Res Pract 2022; 16:700-715. [DOI: 10.4162/nrp.2022.16.6.700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/13/2022] [Accepted: 03/25/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Eunyeong Ahn
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan 38430, Korea
| | - Hyejin Jeong
- Gyeongsangbuk-do Institute of Health & Environment, Yeongcheon 38874, Korea
| | - Eunjung Kim
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan 38430, Korea
| |
Collapse
|
27
|
Beaumont M, Mussard E, Barilly C, Lencina C, Gress L, Painteaux L, Gabinaud B, Cauquil L, Aymard P, Canlet C, Paës C, Knudsen C, Combes S. Developmental Stage, Solid Food Introduction, and Suckling Cessation Differentially Influence the Comaturation of the Gut Microbiota and Intestinal Epithelium in Rabbits. J Nutr 2021; 152:723-736. [PMID: 34875085 PMCID: PMC8891179 DOI: 10.1093/jn/nxab411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/16/2021] [Accepted: 12/01/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND In mammals, the establishment around weaning of a symbiotic relationship between the gut microbiota and its host determines long-term health. OBJECTIVES The aim of this study was to identify the factors driving the comaturation of the gut microbiota and intestinal epithelium at the suckling-to-weaning transition. We hypothesized that the developmental stage, solid food ingestion, and suckling cessation contribute to this process. METHODS From birth to day 18, Hyplus rabbits were exclusively suckling. From day 18 to day 25, rabbits were 1) exclusively suckling; 2) suckling and ingesting solid food; or 3) exclusively ingesting solid food. The microbiota (16S amplicon sequencing), metabolome (nuclear magnetic resonance), and epithelial gene expression (high-throughput qPCR) were analyzed in the cecum at days 18 and 25. RESULTS The microbiota structure and metabolic activity were modified with age when rabbits remained exclusively suckling. The epithelial gene expression of nutrient transporters, proliferation markers, and innate immune factors were also regulated with age (e.g., 1.5-fold decrease of TLR5). Solid food ingestion by suckling rabbits had a major effect on the gut microbiota by increasing its α diversity, remodeling its structure (e.g., 6.3-fold increase of Ruminococcaceae), and metabolic activity (e.g., 4.6-fold increase of butyrate). Solid food introduction also regulated the gene expression of nutrient transporters, differentiation markers, and innate immune factors in the epithelium (e.g., 3-fold increase of nitric oxide synthase). Suckling cessation had no effect on the microbiota, while it regulated the expression of genes involved in epithelial differentiation and immunoglobulin transport (e.g., 2.5-increase of the polymeric immunoglobulin receptor). CONCLUSIONS In rabbits, the maturation of the microbiota at the suckling-to-weaning transition is driven by the introduction of solid food and, to a lesser extent, by the developmental stage. In contrast, the maturation of the intestinal epithelium at the suckling-to-weaning transition is under the influence of the developmental stage, solid food introduction, and suckling cessation.
Collapse
Affiliation(s)
| | - Eloïse Mussard
- GenPhySE, Université de Toulouse, INRAE, ENVT, F-31326, Castanet-Tolosan, France
| | - Céline Barilly
- GenPhySE, Université de Toulouse, INRAE, ENVT, F-31326, Castanet-Tolosan, France
| | - Corinne Lencina
- GenPhySE, Université de Toulouse, INRAE, ENVT, F-31326, Castanet-Tolosan, France
| | - Laure Gress
- GenPhySE, Université de Toulouse, INRAE, ENVT, F-31326, Castanet-Tolosan, France
| | - Louise Painteaux
- GenPhySE, Université de Toulouse, INRAE, ENVT, F-31326, Castanet-Tolosan, France
| | - Béatrice Gabinaud
- GenPhySE, Université de Toulouse, INRAE, ENVT, F-31326, Castanet-Tolosan, France
| | - Laurent Cauquil
- GenPhySE, Université de Toulouse, INRAE, ENVT, F-31326, Castanet-Tolosan, France
| | - Patrick Aymard
- GenPhySE, Université de Toulouse, INRAE, ENVT, F-31326, Castanet-Tolosan, France
| | - Cécile Canlet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Charlotte Paës
- GenPhySE, Université de Toulouse, INRAE, ENVT, F-31326, Castanet-Tolosan, France
| | - Christelle Knudsen
- GenPhySE, Université de Toulouse, INRAE, ENVT, F-31326, Castanet-Tolosan, France
| | - Sylvie Combes
- GenPhySE, Université de Toulouse, INRAE, ENVT, F-31326, Castanet-Tolosan, France
| |
Collapse
|
28
|
Sato DT, Campos FG, Kotze PG, Mendonça RLS, Kanno DT, Pereira JA, Martinez CAR. Sucralfate enemas reduce the oxidative tissue damage and preserves the contents of E-cadherin and ?-catenin in colonic mucosa without fecal stream. Acta Cir Bras 2021; 36:e361007. [PMID: 34852133 PMCID: PMC8650803 DOI: 10.1590/acb361007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/15/2021] [Indexed: 11/21/2022] Open
Abstract
PURPOSE To evaluate the effects of sucralfate enemas in tissue contents of E-cadherin and ?-catenin in an experimental diversion colitis. METHODS Thirty-six male Wistar rats were submitted to a proximal colostomy and a distal mucous fistula. They were allocated into three groups: first group received daily saline enemas (2 mL/day) and the two other groups daily enemas with sucralfate at dosage of 1 or 2 g/kg/day, respectively. Six animals of each group were euthanized after two weeks and six animals after four weeks. The inflammation of the excluded mucosa was evaluated by histological analysis. The oxidative damage was quantified by measurement of malondialdehyde tissue levels. The expression of E-cadherin and ?-catenin was identified by immunohistochemistry, and its contents were quantified by computer-assisted image analysis. RESULTS Sucralfate enemas reduced inflammation in animals subjected to treatment with 2 g/kg/day by four weeks, and the levels of oxidative damage in mucosa without fecal stream irrespective of concentration and time of intervention. E-cadherin and ?-catenin content increased in segments without fecal stream in those animals subjected to treatment with sucralfate. CONCLUSIONS Sucralfate reduces the inflammation and oxidative stress and increases the tissue content of E-cadherin and ?-catenin in colonic mucosa devoid to the fecal stream.
Collapse
|
29
|
Zhang M, Wang Y, Zhao X, Liu C, Wang B, Zhou J. Mechanistic basis and preliminary practice of butyric acid and butyrate sodium to mitigate gut inflammatory diseases: a comprehensive review. Nutr Res 2021; 95:1-18. [PMID: 34757305 DOI: 10.1016/j.nutres.2021.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 01/02/2023]
Abstract
A key event featured in the early stage of chronic gut inflammatory diseases is the disordered recruitment and excess accumulation of immune cells in the gut lamina propria. This process is followed by the over-secretion of pro-inflammatory factors and the prolonged overactive inflammatory responses. Growing evidence has suggested that gut inflammatory diseases may be mitigated by butyric acid (BA) or butyrate sodium (NaB). Laboratory studies show that BA and NaB can enhance gut innate immune function through G-protein-mediated signaling pathways while mitigating the overactive inflammatory responses by inhibiting histone deacetylase. The regulatory effects may occur in both epithelial enterocytes and the immune cells in the lamina propria. Prior to further clinical trials, comprehensive literature reviews and rigid examination concerning the underlying mechanism are necessary. To this end, we collected and reviewed 197 published reports regarding the mechanisms, bioactivities, and clinical effects of BA and NaB to modulate gut inflammatory diseases. Our review found insufficient evidence to guarantee the safety of clinical practice of BA and NaB, either by anal enema or oral administration of capsule or tablet. The safety of clinical use of BA and NaB should be further evaluated. Alternatively, dietary patterns rich in "fruits, vegetables and beans" may be an effective and safe approach to prevent gut inflammatory disease, which elevates gut microbiota-dependent production of BA. Our review provides a comprehensive reference to future clinical trials of BA and NaB to treat gut inflammatory diseases.
Collapse
Affiliation(s)
- Mingbao Zhang
- Department of Gastroenterology and Hepatology, Second Hospital of Shandong University, Shandong University, 250012 China
| | - Yanan Wang
- Department of Gastroenterology and Hepatology, Second Hospital of Shandong University, Shandong University, 250012 China
| | - Xianqi Zhao
- School of Public Health, Cheeloo College of Medicine, Shandong University, 250012 China
| | - Chang Liu
- School of Public Health, Cheeloo College of Medicine, Shandong University, 250012 China
| | - Baozhen Wang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 250012 China.
| | - Jun Zhou
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 250012 China.
| |
Collapse
|
30
|
Shute A, Callejas BE, Li S, Wang A, Jayme TS, Ohland C, Lewis IA, Layden BT, Buret AG, McKay DM. Cooperation between host immunity and the gut bacteria is essential for helminth-evoked suppression of colitis. MICROBIOME 2021; 9:186. [PMID: 34517928 PMCID: PMC8438845 DOI: 10.1186/s40168-021-01146-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 07/30/2021] [Indexed: 05/06/2023]
Abstract
BACKGROUND Studies on the inhibition of inflammation by infection with helminth parasites have, until recently, overlooked a key determinant of health: the gut microbiota. Infection with helminths evokes changes in the composition of their host's microbiota: one outcome of which is an altered metabolome (e.g., levels of short-chain fatty acids (SCFAs)) in the gut lumen. The functional implications of helminth-evoked changes in the enteric microbiome (composition and metabolites) are poorly understood and are explored with respect to controlling enteric inflammation. METHODS Antibiotic-treated wild-type, germ-free (GF) and free fatty-acid receptor-2 (ffar2) deficient mice were infected with the tapeworm Hymenolepis diminuta, then challenged with DNBS-colitis and disease severity and gut expression of the il-10 receptor-α and SCFA receptors/transporters assessed 3 days later. Gut bacteria composition was assessed by 16 s rRNA sequencing and SCFAs were measured. Other studies assessed the ability of feces or a bacteria-free fecal filtrate from H. diminuta-infected mice to inhibit colitis. RESULTS Protection against disease by infection with H. diminuta was abrogated by antibiotic treatment and was not observed in GF-mice. Bacterial community profiling revealed an increase in variants belonging to the families Lachnospiraceae and Clostridium cluster XIVa in mice 8 days post-infection with H. diminuta, and the transfer of feces from these mice suppressed DNBS-colitis in GF-mice. Mice treated with a bacteria-free filtrate of feces from H. diminuta-infected mice were protected from DNBS-colitis. Metabolomic analysis revealed increased acetate and butyrate (both or which can reduce colitis) in feces from H. diminuta-infected mice, but not from antibiotic-treated H. diminuta-infected mice. H. diminuta-induced protection against DNBS-colitis was not observed in ffar2-/- mice. Immunologically, anti-il-10 antibodies inhibited the anti-colitic effect of H. diminuta-infection. Analyses of epithelial cell lines, colonoids, and colon segments uncovered reciprocity between butyrate and il-10 in the induction of the il-10-receptor and butyrate transporters. CONCLUSION Having defined a feed-forward signaling loop between il-10 and butyrate following infection with H. diminuta, this study identifies the gut microbiome as a critical component of the anti-colitic effect of this helminth therapy. We suggest that any intention-to-treat with helminth therapy should be based on the characterization of the patient's immunological and microbiological response to the helminth.
Collapse
Affiliation(s)
- Adam Shute
- Gastrointestinal Research Group, Inflammation Research Network and Host-Parasite Interaction Group, Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Blanca E Callejas
- Gastrointestinal Research Group, Inflammation Research Network and Host-Parasite Interaction Group, Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - ShuHua Li
- Gastrointestinal Research Group, Inflammation Research Network and Host-Parasite Interaction Group, Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Arthur Wang
- Gastrointestinal Research Group, Inflammation Research Network and Host-Parasite Interaction Group, Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Timothy S Jayme
- Gastrointestinal Research Group, Inflammation Research Network and Host-Parasite Interaction Group, Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Christina Ohland
- International Microbiome Center, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Ian A Lewis
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Canada
| | - Brian T Layden
- Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - André G Buret
- Gastrointestinal Research Group, Inflammation Research Network and Host-Parasite Interaction Group, Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Canada
| | - Derek M McKay
- Gastrointestinal Research Group, Inflammation Research Network and Host-Parasite Interaction Group, Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
31
|
Koller KR, Wilson A, Normolle DP, Nicholson JK, Li JV, Kinross J, Lee FR, Flanagan CA, Merculieff ZT, Iyer P, Lammers DL, Thomas TK, O'Keefe SJD. Dietary fibre to reduce colon cancer risk in Alaska Native people: the Alaska FIRST randomised clinical trial protocol. BMJ Open 2021; 11:e047162. [PMID: 34452959 PMCID: PMC8404459 DOI: 10.1136/bmjopen-2020-047162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 08/09/2021] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Diet, shown to impact colorectal cancer (CRC) risk, is a modifiable environmental factor. Fibre foods fermented by gut microbiota produce metabolites that not only provide food for the colonic epithelium but also exert regulatory effects on colonic mucosal inflammation and proliferation. We describe methods used in a double-blinded, randomised, controlled trial with Alaska Native (AN) people to determine if dietary fibre supplementation can substantially reduce CRC risk among people with the highest reported CRC incidence worldwide. METHODS AND ANALYSES Eligible patients undergoing routine screening colonoscopy consent to baseline assessments and specimen/data collection (blood, urine, stool, saliva, breath and colon mucosal biopsies) at the time of colonoscopy. Following an 8-week stabilisation period to re-establish normal gut microbiota post colonoscopy, study personnel randomise participants to either a high fibre supplement (resistant starch, n=30) or placebo (digestible starch, n=30) condition, repeating stool sample collection. During the 28-day supplement trial, each participant consumes their usual diet plus their supplement under direct observation. On day 29, participants undergo a flexible sigmoidoscopy to obtain mucosal biopsy samples to measure the effect of the supplement on inflammatory and proliferative biomarkers of cancer risk, with follow-up assessments and data/specimen collection similar to baseline. Secondary outcome measures include the impact of a high fibre supplement on the oral and colonic microbiome and biofluid metabolome. ETHICS AND DISSEMINATION Approvals were obtained from the Alaska Area and University of Pittsburgh Institutional Review Boards and Alaska Native Tribal Health Consortium and Southcentral Foundation research review bodies. A data safety monitoring board, material transfer agreements and weekly study team meetings provide regular oversight throughout the study. Study findings will first be shared with AN tribal leaders, health administrators, providers and community members. Peer-reviewed journal articles and conference presentations will be forthcoming once approved by tribal review bodies. TRIAL REGISTRATION NUMBER NCT03028831.
Collapse
Affiliation(s)
- Kathryn R Koller
- Research Services, Alaska Native Tribal Health Consortium, Anchorage, Alaska, USA
| | - Annette Wilson
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Daniel P Normolle
- Hillman Cancer Center Biostatistics Facility, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jeremy K Nicholson
- Australian National Phenome Center, Murdoch University, Perth, Western Australia, Australia
| | - Jia V Li
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - James Kinross
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Flora R Lee
- Research Services, Alaska Native Tribal Health Consortium, Anchorage, Alaska, USA
| | - Christie A Flanagan
- Alaska Native Epidemiology Center, Alaska Native Tribal Health Consortium, Anchorage, Alaska, USA
| | - Zoe T Merculieff
- Wellness and Prevention, Alaska Native Tribal Health Consortium, Anchorage, Alaska, USA
| | - Priya Iyer
- Hillman Cancer Center Biostatistics Facility, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Daniela L Lammers
- Research Services, Alaska Native Tribal Health Consortium, Anchorage, Alaska, USA
| | - Timothy K Thomas
- Research Services, Alaska Native Tribal Health Consortium, Anchorage, Alaska, USA
| | - Stephen J D O'Keefe
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
32
|
Sauvaitre T, Etienne-Mesmin L, Sivignon A, Mosoni P, Courtin CM, Van de Wiele T, Blanquet-Diot S. Tripartite relationship between gut microbiota, intestinal mucus and dietary fibers: towards preventive strategies against enteric infections. FEMS Microbiol Rev 2021; 45:5918835. [PMID: 33026073 DOI: 10.1093/femsre/fuaa052] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
The human gut is inhabited by a large variety of microorganims involved in many physiological processes and collectively referred as to gut microbiota. Disrupted microbiome has been associated with negative health outcomes and especially could promote the onset of enteric infections. To sustain their growth and persistence within the human digestive tract, gut microbes and enteric pathogens rely on two main polysaccharide compartments, namely dietary fibers and mucus carbohydrates. Several evidences suggest that the three-way relationship between gut microbiota, dietary fibers and mucus layer could unravel the capacity of enteric pathogens to colonise the human digestive tract and ultimately lead to infection. The review starts by shedding light on similarities and differences between dietary fibers and mucus carbohydrates structures and functions. Next, we provide an overview of the interactions of these two components with the third partner, namely, the gut microbiota, under health and disease situations. The review will then provide insights into the relevance of using dietary fibers interventions to prevent enteric infections with a focus on gut microbial imbalance and impaired-mucus integrity. Facing the numerous challenges in studying microbiota-pathogen-dietary fiber-mucus interactions, we lastly describe the characteristics and potentialities of currently available in vitro models of the human gut.
Collapse
Affiliation(s)
- Thomas Sauvaitre
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France.,Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| | - Adeline Sivignon
- Université Clermont Auvergne, UMR 1071 Inserm, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH), Clermont-Ferrand, France
| | - Pascale Mosoni
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| | - Christophe M Courtin
- KU Leuven, Faculty of Bioscience Engineering, Laboratory of Food Chemistry and Biochemistry & Leuven Food Science and Nutrition Research Centre (LFoRCe), Leuven, Belgium
| | - Tom Van de Wiele
- Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| |
Collapse
|
33
|
Butyrate and the Intestinal Epithelium: Modulation of Proliferation and Inflammation in Homeostasis and Disease. Cells 2021; 10:cells10071775. [PMID: 34359944 PMCID: PMC8304699 DOI: 10.3390/cells10071775] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/29/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
The microbial metabolite butyrate serves as a link between the intestinal microbiome and epithelium. The monocarboxylate transporters MCT1 and SMCT1 are the predominant means of butyrate transport from the intestinal lumen to epithelial cytoplasm, where the molecule undergoes rapid β-oxidation to generate cellular fuel. However, not all epithelial cells metabolize butyrate equally. Undifferentiated colonocytes, including neoplastic cells and intestinal stem cells at the epithelial crypt base preferentially utilize glucose over butyrate for cellular fuel. This divergent metabolic conditioning is central to the phenomenon known as “butyrate paradox”, in which butyrate induces contradictory effects on epithelial proliferation in undifferentiated and differentiated colonocytes. There is evidence that accumulation of butyrate in epithelial cells results in histone modification and altered transcriptional activation that halts cell cycle progression. This manifests in the apparent protective effect of butyrate against colonic neoplasia. A corollary to this process is butyrate-induced inhibition of intestinal stem cells. Yet, emerging research has illustrated that the evolution of the crypt, along with butyrate-producing bacteria in the intestine, serve to protect crypt base stem cells from butyrate’s anti-proliferative effects. Butyrate also regulates epithelial inflammation and tolerance to antigens, through production of anti-inflammatory cytokines and induction of tolerogenic dendritic cells. The role of butyrate in the pathogenesis and treatment of intestinal neoplasia, inflammatory bowel disease and malabsorptive states is evolving, and holds promise for the potential translation of butyrate’s cellular function into clinical therapies.
Collapse
|
34
|
Fredericks E, Theunissen R, Roux S. Short chain fatty acids and monocarboxylate transporters in irritable bowel syndrome. TURKISH JOURNAL OF GASTROENTEROLOGY 2021; 31:840-847. [PMID: 33625995 DOI: 10.5152/tjg.2020.19856] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND/AIMS Gut microbiota ferments indigestible food that rests in the colon to produce short-chain fatty acids (SCFAs) acetate, propionate, and butyrate. Colonic SCFA stimulate the synthesis of serotonin which is central in irritable bowel syndrome (IBS) pathophysiology. Reduced SCFA have been linked to specific IBS symptoms like colonic hyperalgesia and hypersensitivity. SCFA enter the colonocyte mainly via 2 energy-dependent monocarboxylate transporters, MCT1 (SLC16A1) and SMCT1 (SLC5A8). We investigated specific gut microbiota, SCFA concentrations, and monocarboxylate transporter mRNA expression in patients with IBS. MATERIAL AND METHODS A total of 30 IBS patients-15 constipation-predominant (C-IBS) and 15 diarrhoea-predominant (D-IBS)-and 15 healthy controls were recruited. Bacteroidetes and Bifidobacterium species were analyzed using quantitative polymerase chain reaction (qPCR) on stool samples. SCFA concentrations were determined by gas chromatography/mass spectroscopy of stool samples. Monocarboxylate transporter mRNA was quantified by qPCR on colon biopsy specimens. RESULTS Bacteroides was significantly increased in the D-IBS group compared with the C-IBS group and healthy controls. Bifidobacterium was significantly reduced in both IBS groups. SCFA ratios were altered in both IBS groups with a reduction of all 3 measured SCFA in C-IBS and acetic acid in D-IBS. MCT1 and SMCT1 were significantly reduced in C-IBS and D-IBS. CONCLUSION In agreement with findings of previous studies, the microbiota assessed were significantly altered inferring dysbiosis in IBS. SCFA and their ratios were significantly altered in both IBS groups. SCFA transporters, MCT1 and SMCT1 were significantly reduced in both IBS groups, suggesting reduced colonocyte SCFA transfer. SCFA availability and transfer into the colonocytes may be important in IBS pathogenesis and should be prospectively studied.
Collapse
Affiliation(s)
- Ernst Fredericks
- Department of Physiology, Nelson Mandela University School of Science, Port Elizabeth, South Africa
| | - Reza Theunissen
- Department of Physiology, Nelson Mandela University School of Science, Port Elizabeth, South Africa
| | - Saartjie Roux
- Department of Physiology, Nelson Mandela University School of Science, Port Elizabeth, South Africa
| |
Collapse
|
35
|
Abstract
Inflammatory Bowel Disease (IBD) is a term used to describe a group of complex disorders of the gastrointestinal (GI) tract. IBDs include two main forms: Crohn’s Disease (CD) and Ulcerative Colitis (UC), which share similar clinical symptoms but differ in the anatomical distribution of the inflammatory lesions. The etiology of IBDs is undetermined. Several hypotheses suggest that Crohn’s Disease and Ulcerative Colitis result from an abnormal immune response against endogenous flora and luminal antigens in genetically susceptible individuals. While there is no cure for IBDs, most common treatments (medication and surgery) aim to reduce inflammation and help patients to achieve remission. There is growing evidence and focus on the prophylactic and therapeutic potential of probiotics in IBDs. Probiotics are live microorganisms that regulate the mucosal immune system, the gut microbiota and the production of active metabolites such as Short-Chain Fatty Acids (SCFAs). This review will focus on the role of intestinal dysbiosis in the immunopathogenesis of IBDs and understanding the health-promoting effects of probiotics and their metabolites.
Collapse
|
36
|
Butyric Acid Added Apically to Intestinal Caco-2 Cells Elevates Hepatic ApoA-I Transcription and Rescues Lower ApoA-I Expression in Inflamed HepG2 Cells Co-Cultured in the Basolateral Compartment. Biomolecules 2021; 11:biom11010071. [PMID: 33430253 PMCID: PMC7825706 DOI: 10.3390/biom11010071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/29/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Apolipoprotein A-I (ApoA-I) concentrations are decreased during inflammation, which may reduce high-density lipoprotein (HDL) functionality. Thus, rescuing ApoA-I concentrations during inflammation might help to prevent atherosclerosis. Recent studies have shown that butyric acid (C4) has anti-inflammatory effects and rescues ApoA-I production. However, whether intestinal short chain fatty acids (SCFAs) are able to influence hepatic processes is unknown. Therefore, we investigated C4 anti-inflammatory effects on ApoA-I transcription in the intestine-liver co-culture model. C4 dose-response experiments in the presence or absence of cytokines were performed in a co-culture system including Caco-2 cells, HepG2 cells, or both. Changes in ApoA-I transcription in Caco-2 cells and HepG2 cells were analyzed using qPCR. C4 increased ApoA-I expression in HepG2 cells that cultured alone. When both cells were cultured together, C4 decreased ApoA-I expression in Caco-2 cells and increased ApoA-I expression in HepG2 cells. However, adding C4 to apical Caco-2 cells resulted in a smaller effect in HepG2 cells compared with adding C4 directly to the hepatocytes. Moreover, C4 rescued ApoA-I expression in inflamed HepG2 cells. These findings suggests that intestinal SCFAs can affect hepatic processes. However, the smaller effect in the co-culture experiment indicates cross-talk between intestine and liver.
Collapse
|
37
|
Lee JS, Wang RX, Alexeev EE, Colgan SP. Intestinal Inflammation as a Dysbiosis of Energy Procurement: New Insights into an Old Topic. Gut Microbes 2021; 13:1-20. [PMID: 33583319 PMCID: PMC7889129 DOI: 10.1080/19490976.2021.1880241] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel disease (IBD) coincides with profound shifts in microbiota and host metabolic energy supply and demand. The gastrointestinal epithelium is anatomically positioned to provide a selective barrier between the anaerobic luminal microbiota and host lamina propria, with the microbiota and epithelium participating in an intricate energy exchange necessary for homeostasis. Maintenance and restoration of the barrier requires high energy flux and places significant demands on available substrates to generate ATP. It is recently appreciated that components of the microbiota contribute significantly to a multitude of biochemical pathways within and outside of the mucosa. Decades-old studies have appreciated that byproducts of the microbiota provide essential sources of energy to the intestinal epithelium, especially the colon. More recent work has unveiled the existence of numerous microbial-derived metabolites that support energy procurement within the mucosa. It is now appreciated that disease-associated shifts in the microbiota, termed dysbiosis, places significant demands on energy acquisition within the mucosa. Here, we review the topic of host- and microbial-derived components that influence tissue energetics in health and during disease.
Collapse
Affiliation(s)
- J. Scott Lee
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, United States
| | - Ruth X. Wang
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, United States
| | - Erica E. Alexeev
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, United States
- Department of Gastroenterology, Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, United States
| | - Sean P. Colgan
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, United States
| |
Collapse
|
38
|
Wang RX, Henen MA, Lee JS, Vögeli B, Colgan SP. Microbiota-derived butyrate is an endogenous HIF prolyl hydroxylase inhibitor. Gut Microbes 2021; 13:1938380. [PMID: 34190032 PMCID: PMC8253137 DOI: 10.1080/19490976.2021.1938380] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 02/04/2023] Open
Abstract
The gut microbiota is essential for human health. Microbial supply of short-chain fatty acids (SCFAs), particularly butyrate, is a well-established contributor to gut homeostasis and disease resistance. Reaching millimolar luminal concentrations, butyrate is sequestered and utilized in the colon as the favored energy source for intestinal epithelia. Given the steep oxygen gradient across the anoxic lumen and the highly oxygenated lamina propria, the colon provides a particularly interesting environment to study oxygen sensing. Previous studies have shown that the transcription factor hypoxia-inducible factor (HIF) is stabilized in healthy colonic epithelia. Here we show that butyrate directly inhibits HIF prolyl hydroxylases (PHDs) to stabilize HIF. We find that butyrate stabilizes HIF in vitro despite eliminating β-oxidation and resultant oxygen consumption. Using recombinant PHD protein in combination with nuclear magnetic resonance and enzymatic biochemical assays, we identify butyrate to bind and function as a unique, noncompetitive inhibitor of PHDs relative to other SCFAs. Butyrate inhibited PHD with a noncompetitive Ki of 5.3 ± 0.5 mM, a physiologically relevant concentration. We also confirm that microbiota-derived butyrate is necessary to stabilize HIF in mice colonic tissue through antibiotic-induced butyrate depletion and reconstitution experiments. Our results suggest that the co-evolution of mammals and mutualistic microbiota has selected for butyrate to impact a critical gene regulation pathway that can be extended beyond the mammalian gut. As PHDs are a major target for drug development in the stabilization of HIF, butyrate holds great potential as a well-tolerated endogenous inhibitor with far-reaching therapeutic impact.
Collapse
Affiliation(s)
- Ruth X. Wang
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- School of Medicine, Medical Scientist Training Program, University of Colorado, Aurora, CO, USA
| | - Morkos A. Henen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmaceutical Organic Chemistry, Mansoura University, Mansoura, Egypt
| | - J. Scott Lee
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Beat Vögeli
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sean P. Colgan
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
39
|
Ma C, Gao Q, Zhang W, Zhu Q, Tang W, Blachier F, Ding H, Kong X. Supplementing Synbiotic in Sows' Diets Modifies Beneficially Blood Parameters and Colonic Microbiota Composition and Metabolic Activity in Suckling Piglets. Front Vet Sci 2020; 7:575685. [PMID: 33330695 PMCID: PMC7734190 DOI: 10.3389/fvets.2020.575685] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/04/2020] [Indexed: 01/12/2023] Open
Abstract
Nutrients in the maternal diet favor the growth and development of suckling piglets and alter their gut microbiota composition and metabolic activity, thus affecting the hosts. The present study analyzed, in suckling piglets from sows receiving antibiotic or synbiotic supplements from pregnancy to lactation, several biochemical parameters, oxidative/anti-oxidative indices, inflammatory cytokines, and ingestion-related factor levels in plasma, as well as colonic microbiota composition and metabolic activity, and mucosal expression of genes related to the intestinal barrier function. Compared with the control group, maternal synbiotic supplementation decreased (P < 0.05) the plasma levels of glucose, AMM, TC, low-density lipoprotein-cholesterol (LDL-C), MDA, H2O2, ghrelin, CCK, PP, IL-1β, IL-2, IL-6, TNF-α, Ala, Cys, Tau, and β-AiBA, the levels of propionate and total short-chain fatty acids (SCFAs) in the colonic luminal content, and colonic abundances of RFN20, Anaerostipes, and Butyricimonas; while increased (P < 0.05) the plasma levels of urea nitrogen (UN), Ile, Leu, α-AAA, α-ABA, and 1-Mehis, as well as colonic abundances of Sphingomonas, Anaerovorax, Sharpea, and Butyricicoccus. Compared with the antibiotic group, maternal synbiotic supplementation decreased (P < 0.05) the plasma levels of glucose, gastrin, and Ala, as well as abundances of Pasteurella and RFN20 and propionate level in the colonic content. Expression of genes coding for E-cadherin, Occludin, ZO-1, ZO-2, IL-10, and interferon-α were down-regulated in the colonic mucosa. The synbiotic supplementation increased (P < 0.05) the plasma levels of UN, Leu, α-ABA, and 1-Mehis, the abundances of Anaerovorax, Sharpea, and Butyricicoccus and expression of genes coding for E-cadherin, Occludin, ZO-1, ZO-2, IL-10, and interferon-α. Spearman correlation analysis showed that there was a positive correlation between colonic Anaerostipes abundance and acetate and SCFAs levels; whereas a negative correlation between Fusobacteria and Fusobacterium abundances and acetate level. These findings suggest that synbiotic supplementation in the maternal diet improved nutrient metabolism and intestinal barrier permeability, reduced oxidative stress, and modified colonic microbiota composition and metabolic activity in suckling piglets.
Collapse
Affiliation(s)
- Cui Ma
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qiankun Gao
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Wanghong Zhang
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qian Zhu
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Wu Tang
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Francois Blachier
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, Paris, France
| | - Hao Ding
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Xiangfeng Kong
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
40
|
Zou Y, Chen T. Engineered Akkermansia muciniphila: A promising agent against diseases (Review). Exp Ther Med 2020; 20:285. [PMID: 33209129 PMCID: PMC7668130 DOI: 10.3892/etm.2020.9415] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 09/15/2020] [Indexed: 12/23/2022] Open
Abstract
Achieving a harmonious gut microbial ecosystem has been hypothesized to be a successful method for alleviating metabolic disorders. The administration of probiotics, such as Lactobacillus and Bifidobacteria, is a known traditional and safe pathway to regulate human commensal microbes. With advancements in genetic sequencing and genetic editing tools, more bacteria are able to function as engineered probiotics with multiple therapeutic properties. As one of the next-generation probiotic candidates, Akkermansia muciniphila (A. muciniphila) has been discovered to enhance the gut barrier function and moderate inflammatory responses, exhibit improved effects with pasteurization and display beneficial probiotic effects in individuals with obesity, type 2 diabetes, atherosclerosis and autism-related gastrointestinal disturbances. In view of this knowledge, the present review aimed to summarize the effects of A. muciniphila in the treatment of metabolic disorders and to discuss several mature recombination systems for the genetic modification of A. muciniphila. From gaining an enhanced understanding of its genetic background, ingested A. muciniphila is expected to be used in various applications, including as a diagnostic tool, and in the site-specific delivery of therapeutic drugs.
Collapse
Affiliation(s)
- Yixuan Zou
- Institute of Translational Medicine, National Engineering Research Center for Bioengineering Drugs and Technologies, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Tingtao Chen
- Institute of Translational Medicine, National Engineering Research Center for Bioengineering Drugs and Technologies, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| |
Collapse
|
41
|
Effect of Dietary Sodium Acetate on Skin Mucus Immune Parameters and Expression of Gene Related to Growth, Immunity and Antioxidant System in Common Carp (Cyprinus carpio) Intestine. ANNALS OF ANIMAL SCIENCE 2020. [DOI: 10.2478/aoas-2020-0050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Abstract
The present study investigated the possible effects of including salt of short chain fatty acid, sodium acetate (SA), on skin mucus immune parameters and immune, antioxidant and growth-related genes expression in common carp. There is little data available about the effective role of SA on immune, antioxidant and growth related genes expression as well as skin mucus immune parameters. The aim of this study was to analyse the effect of SA intake on these factors using common carp (Cyprinus carpio) as model organism. Two hundred and forty healthy common carps (mean weight = 15 ± 0.9 g) were supplied and randomly stocked into 12 fiberglass tanks 200 L (20 fish per tank) assigned to four treatments and triplicates. The study was performed in a completely randomized design. The treatments were feeding carps with experimental diets containing different levels (0.0 [control], 0.5, 1 and 2%) of SA. The skin mucus total immunoglobulin and total protein levels in fish fed 2% SA showed significant increase compared to the control group (P<0.05). Results showed a significant increase in the GH gene expression in 1 and 2% SA treatments (P<0.05). The carps fed the diet with 2% SA showed a significant increase in IGF-1 expression (P<0.05). The expression of GSTa, and GPX (antioxidant genes) revealed a significant increase in the GSTa (fish fed SA at 1% and 2% levels) and GPX gene expression with fish fed 2% SA (P<0.05). Supplementation of fish diet with SA induced a slight elevation in the intestine of all immune-related genes (TNF-α, IL-1β, IL8 and Lyz) compared to the control group (P>0.05). However, Lyz gene was significantly up-regulated in 1 or 2% SA treatments. These results confirmed beneficial effects of SA as a feed additive in common carp culture.
Collapse
|
42
|
Kent-Dennis C, Penner GB. Effects of a proinflammatory response on metabolic function of cultured, primary ruminal epithelial cells. J Dairy Sci 2020; 104:1002-1017. [PMID: 33131809 DOI: 10.3168/jds.2020-19092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022]
Abstract
Inflammation of ruminal epithelium may occur during ruminal acidosis as a result of translocation and interaction of ruminal epithelial cells (REC) with molecules such as lipopolysaccharide (LPS). Such inflammation has been reported to alter cellular processes such as nutrient absorption, metabolic regulation, and energy substrate utilization in other cell types but has not been investigated for REC. The objectives of this study were to investigate the effects of LPS on metabolism of short-chain fatty acids by primary REC, as well as investigating the effects of media containing short-chain fatty acids on the proinflammatory response. Ruminal papillae from 9 yearling Speckle Park beef heifers were used to isolate and culture primary REC. Cells were grown in minimum essential medium (MEM) for 12 d before use and then reseeded in 24-well culture plates. The study was conducted as a 2 × 2 factorial, where cells were grown in unaltered MEM (REG) or medium containing 2 mM butyrate and 5 mM propionate (SCFA) with (50,000 EU/mL; +LPS) or without LPS (-LPS) for 24 h. Supernatant samples were collected for analysis of glucose and SCFA consumption. Cells were collected to determine the expression of mRNA for genes associated with inflammation (TNF, IL1B, CXCL2, CXCL8, PTGS2, and TLR4), purinergic signaling (P2RX7, ADORAB2, and CD73), nutrient transport [SLC16A1 (MCT1), SLC16A3 (MCT4), SLC5A8, and MCU], and cell metabolism [ACAT1, SLC2A1 (GLUT1), IGFBP3, and IGFBP5]. Protein expression of TLR4 and ketogenic enzymes (BDH1 and HMGCS1) were also analyzed using flow cytometry. Statistical analysis was conducted with the MIXED model of SAS version 9.4 (SAS Institute Inc., Cary, NC) with medium, LPS exposure, and medium × LPS interaction as fixed effects and animal within plate as a random effect. Cells tended to consume more glucose when exposed to LPS as opposed to no LPS exposure (31.8 vs. 28.7 ± 2.7), but consumption of propionate and butyrate was not influenced by LPS. Expression of TNF and IL1B was upregulated when exposed to LPS, and expression of CXCL2 and CXCL8 increased following LPS exposure with SCFA (medium × LPS). For cells exposed to LPS, we found a downregulation of ACAT1 and IGFBP5 and an upregulation of SLC2A1, SLC16A3, MCU, and IGFBP3. Medium with SCFA led to greater expression of MCU. SLC16A1 was upregulated in cells incubated with SCFA and without LPS compared with the other groups. Protein expression of ketogenic enzymes was not affected; however, BDH1 mean fluorescence intensity (MFI) expression tended to be less in cells exposed to LPS. These data are interpreted to indicate that when REC are exposed to LPS, they may increase glucose metabolism. Moreover, transport of solutes was affected by SCFA in the medium and by exposure to LPS. Overall, the results suggest that metabolic function of REC in vitro is altered by a proinflammatory response, which may lead to a greater glucose requirement.
Collapse
Affiliation(s)
- C Kent-Dennis
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK, Canada, S7N 5A8
| | - G B Penner
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK, Canada, S7N 5A8.
| |
Collapse
|
43
|
Couto MR, Gonçalves P, Magro F, Martel F. Microbiota-derived butyrate regulates intestinal inflammation: Focus on inflammatory bowel disease. Pharmacol Res 2020; 159:104947. [DOI: 10.1016/j.phrs.2020.104947] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/04/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
|
44
|
Effect of dietary sodium acetate on skin mucus immune parameters and expression of gene related to growth, immunity and antioxidant system in common carp ( Cyprinus carpio) intestine. ANNALS OF ANIMAL SCIENCE 2020. [DOI: 10.2478/aoas-2020-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
The present study investigated the possible effects of including salt of short chain fatty acid, sodium acetate (SA), on skin mucus immune parameters and immune, antioxidant and growth-related genes expression in common carp. There is a little data available about the effective role of SA on immune, antioxidant and growth related genes expression as well as skin mucus immune parameters. The aim of this study was to analysis the effect of SA intake on these factors using common carp (Cyprinus carpio) as model organism. Two hundred and forty healthy common carp (mean weight = 15 ± 0.9 g) supplied and randomly stocked into 12 fiberglass tanks 200 L (20 fish per tank) assigned to four treatments and triplicates. The study was performed in a completely randomized design. The treatments were feeding carps with experimental diets containing different levels (0.0 [control], 0.5, 1 and 2%) of SA. The skin mucus total immunoglobulin and total protein levels in fish fed 2% SA showed significant increase compared to the control group (P < 0.05). Results showed a significant increase in the GH gene expression in 1 and 2% SA treatments (P < 0.05). The carps were fed with diet content 2% SA showed significantly increase in IGF-1 expression (P < 0.05). The expression of GSTa, and GPX (antioxidant genes) revealed a significant increase in the GSTa (fish fed SA at 1% and 2% levels) and GPX gene expression with fish fed 2% SA (P < 0.05). Supplementation of fish diet with SA induce slight elevation in the intestine of all immune-related genes (TNF-α, IL-1β, IL8 and Lyz) compared to the control group (P > 0.05). However, Lyz gene significantly up-regulated in 1 or 2% SA treatments. These results confirmed beneficial effects of SA as a feed additive in common carp culture.
Collapse
|
45
|
Jakubczyk D, Leszczyńska K, Górska S. The Effectiveness of Probiotics in the Treatment of Inflammatory Bowel Disease (IBD)-A Critical Review. Nutrients 2020; 12:nu12071973. [PMID: 32630805 PMCID: PMC7400428 DOI: 10.3390/nu12071973] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/19/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
Inflammatory bowel disease (IBD), which affects millions of people worldwide, includes two separate diseases: Crohn's disease (CD) and ulcerative colitis (UC). Although the background (chronic inflammatory state) and some of the symptoms of CD and UC are similar, both diseases differ from each other. It is becoming clear that a combination of many factors, in particular genetic background, host immune response and microbial reduced diversity status are associated with IBD. One potential strategy to prevent/treat IBD is gut modulation by probiotics. Over the last twenty years, many publications have focused on the role of probiotics in the course of IBD. The review discusses the utility of different strains of probiotics, especially Bifidobacterium spp., in all factors potentially involved in the etiology of IBD. The probiotic modulatory properties among different study models (cell lines, animal models of colitis, clinical study) are discussed and probiotic usefulness is assessed in relation to the treatment, prevention, and remission of diseases.
Collapse
|
46
|
Ion Transport Basis of Diarrhea in a Mouse Model of Adoptive T Cell Transfer Colitis. Dig Dis Sci 2020; 65:1700-1709. [PMID: 31741140 PMCID: PMC7230007 DOI: 10.1007/s10620-019-05945-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Diarrhea, a major pathological hallmark of inflammatory bowel disease, is characterized by a significant reduction in the expression and function of key intestinal ion transporters. The adoptive naïve CD4+ T cell transfer colitis is an immune-based, chronic colitis mouse model which resembles human Crohn's disease. Although mice with T cell transfer colitis demonstrate diarrhea, the ion transporter basis of this phenotype has not been explored. AIMS/METHODS In the current studies, we aimed to determine the mRNA and protein levels of the key NaCl transporters DRA and NHE3 along with the mRNA expression of other transporters in the inflamed intestine. RESULTS Naïve CD4+ T cells, transferred to Rag2 knockout mice, induced severe colonic inflammation characterized by histological damage and increased mRNA levels of cytokines in the colon with no effect in the ileum. Diarrheal phenotype was a key feature of the excised colons of mice where loose stools were evident. Our results demonstrated that the key chloride transporter DRA, mRNA, and protein levels were significantly reduced in the inflamed colon. However, expression of the key sodium hydrogen exchanger NHE3 was unaffected. The mRNA expression of other important transporters was also determined; in this regard, the sodium channel ENACα and the monocarboxylate transporters MCT1 and SMCT1 mRNA levels were also significantly lower compared to control mice. However, CFTR mRNA was not altered in the colon or ileum. CONCLUSIONS The studies conducted herein for the first time demonstrate the downregulation of important intestinal ion transporters in proximal and distal colon in T cell transfer colitis mouse model, providing valuable evidence for the ion transporter basis of diarrhea in this chronic model of inflammation.
Collapse
|
47
|
Wang RX, Lee JS, Campbell EL, Colgan SP. Microbiota-derived butyrate dynamically regulates intestinal homeostasis through regulation of actin-associated protein synaptopodin. Proc Natl Acad Sci U S A 2020; 117:11648-11657. [PMID: 32398370 PMCID: PMC7260972 DOI: 10.1073/pnas.1917597117] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The intestinal mucosa exists in dynamic balance with trillions of luminal microbes. Disruption of the intestinal epithelial barrier, commonly observed in mucosal inflammation and diseases such as inflammatory bowel diseases (IBDs), is often associated with dysbiosis, particularly decreases in species producing short-chain fatty acids (SCFAs), such as butyrate. It remains unclear to what extent microbiota-derived factors contribute to the overall maintenance of intestinal homeostasis. Initial studies revealed that butyrate selectively promotes epithelial barrier function and wound healing. We aimed to define the specific mechanism(s) through which butyrate contributes to these epithelial responses. Guided by an unbiased profiling approach, we identified the dominant regulation of the actin-binding protein synaptopodin (SYNPO). Extensions of this work revealed a role for SYNPO in intestinal epithelial barrier function and wound healing. SYNPO was localized to the intestinal epithelial tight junction and within F-actin stress fibers where it is critical for barrier integrity and cell motility. Butyrate, but not other SCFAs, induced SYNPO in epithelial cell lines and murine colonic enteroids through mechanisms possibly involving histone deacetylase inhibition. Moreover, depletion of the microbiota abrogated expression of SYNPO in the mouse colon, which was rescued with butyrate repletion. Studies in Synpo-deficient mice demonstrated exacerbated disease susceptibility and increased intestinal permeability in a dextran sulfate sodium colitis model. These findings establish a critical role for the microbiota and their products, specifically butyrate, in the regulated expression of SYNPO for intestinal homeostasis and reveal a direct mechanistic link between microbiota-derived butyrate and barrier restoration.
Collapse
Affiliation(s)
- Ruth X Wang
- Mucosal Inflammation Program, Department of Medicine, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Medical Scientist Training Program, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - J Scott Lee
- Mucosal Inflammation Program, Department of Medicine, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Eric L Campbell
- Centre for Experimental Medicine, Queens University Belfast, Belfast BT9 7BL, Northern Ireland
| | - Sean P Colgan
- Mucosal Inflammation Program, Department of Medicine, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045;
| |
Collapse
|
48
|
Nutrition, IBD and Gut Microbiota: A Review. Nutrients 2020; 12:nu12040944. [PMID: 32235316 PMCID: PMC7230231 DOI: 10.3390/nu12040944] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/11/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic relapsing–remitting systemic disease of the gastrointestinal tract, characterized by an inflammatory process that requires lifelong treatment. The underlying causes of IBD are still unclear, as this heterogeneous disorder results from a complex interplay between genetic variability, the host immune system and environmental factors. The current knowledge recognizes diet as a risk factor for the development of IBD and attributes a substantial pathogenic role to the intestinal dysbiosis inducing an aberrant mucosal immune response in genetically predisposed individuals. This review focused on the clinical evidence available that considers the impact of some nutrients on IBD onset and the role of different diets in the management of IBD and their effects on the gut microbiota composition. The effects of the Specific Carbohydrate Diet, low fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAP) diet, gluten free diet, anti-inflammatory diet and Mediterranean diet are investigated with regard to their impact on microbiota and on the evolution of the disease. At present, no clear indications toward a specific diet are available but the assessment of dysbiosis prior to the recommendation of a specific diet should become a standard clinical approach in order to achieve a personalized therapy.
Collapse
|
49
|
Kang S, You HJ, Lee YG, Jeong Y, Johnston TV, Baek NI, Ku S, Ji GE. Production, Structural Characterization, and In Vitro Assessment of the Prebiotic Potential of Butyl-Fructooligosaccharides. Int J Mol Sci 2020; 21:ijms21020445. [PMID: 31936703 PMCID: PMC7013684 DOI: 10.3390/ijms21020445] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/11/2022] Open
Abstract
Short-chain fatty acids (SCFAs), especially butyrate, produced in mammalian intestinal tracts via fermentation of dietary fiber, are known biofunctional compounds in humans. However, the variability of fermentable fiber consumed on a daily basis and the diversity of gut microbiota within individuals often limits the production of short-chain fatty acids in the human gut. In this study, we attempted to enhance the butyrate levels in human fecal samples by utilizing butyl-fructooligosaccharides (B-FOS) as a novel prebiotic substance. Two major types of B-FOS (GF3-1B and GF3-2B), composed of short-chain fructooligosaccharides (FOS) bound to one or two butyric groups by ester bonds, were synthesized. Qualitative analysis of these B-FOS using Fourier transform infrared (FT-IR) spectroscopy, matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS), nuclear magnetic resonance (NMR) and low-resolution fast-atom bombardment mass spectra (LR-FAB-MS), showed that the chemical structure of GF3-1B and GF3-2B were [O-(1-buty-β-D-fru-(2→1)-O-β-D-fru-(2→1)-O-β-D-fru-O-α-D-glu] and [O-(1-buty)-β-D-fru-(2→1)-O-β-D-fru-(2→1)-O-(4-buty)-β-D-fru-O-α-D-glu], respectively. The ratio of these two compounds was approximately 5:3. To verify their biofunctionality as prebiotic oligosaccharides, proliferation and survival patterns of human fecal microbiota were examined in vitro via 16S rRNA metagenomics analysis compared to a positive FOS control and a negative control without a carbon source. B-FOS treatment showed different enrichment patterns on the fecal microbiota community during fermentation, and especially stimulated the growth of major butyrate producing bacterial consortia and modulated specific butyrate producing pathways with significantly enhanced butyrate levels. Furthermore, the relative abundance of Fusobacterium and ammonia production with related metabolic genes were greatly reduced with B-FOS and FOS treatment compared to the control group. These findings indicate that B-FOS differentially promotes butyrate production through the enhancement of butyrate-producing bacteria and their metabolic genes, and can be applied as a novel prebiotic compound in vivo.
Collapse
Affiliation(s)
- Sini Kang
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea; (S.K.); (Y.J.)
| | - Hyun Ju You
- Institute of Health and Environment, Graduate School of Public Health, Seoul National University, Seoul 08826, Korea;
| | - Yeong-Geun Lee
- Graduate School of Biotechnology and Department of Oriental Medicinal Biotechnology, Kyung Hee University, Yongin 17104, Korea; (Y.-G.L.); (N.-I.B.)
| | - Yunju Jeong
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea; (S.K.); (Y.J.)
| | - Tony V. Johnston
- Fermentation Science Program, School of Agriculture, College of Basic and Applied Sciences, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
| | - Nam-In Baek
- Graduate School of Biotechnology and Department of Oriental Medicinal Biotechnology, Kyung Hee University, Yongin 17104, Korea; (Y.-G.L.); (N.-I.B.)
| | - Seockmo Ku
- Fermentation Science Program, School of Agriculture, College of Basic and Applied Sciences, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
- Correspondence: (S.K.); (G.E.J.); Tel.: +1-615-904-8290 (S.K.); +82-2-880-6282 (G.E.J.)
| | - Geun Eog Ji
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea; (S.K.); (Y.J.)
- Research Center, BIFIDO Co., Ltd., Hongcheon 25117, Korea
- Correspondence: (S.K.); (G.E.J.); Tel.: +1-615-904-8290 (S.K.); +82-2-880-6282 (G.E.J.)
| |
Collapse
|
50
|
Ferrer-Picón E, Dotti I, Corraliza AM, Mayorgas A, Esteller M, Perales JC, Ricart E, Masamunt MC, Carrasco A, Tristán E, Esteve M, Salas A. Intestinal Inflammation Modulates the Epithelial Response to Butyrate in Patients With Inflammatory Bowel Disease. Inflamm Bowel Dis 2020; 26:43-55. [PMID: 31211831 PMCID: PMC6905302 DOI: 10.1093/ibd/izz119] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Butyrate-producing gut bacteria are reduced in patients with active inflammatory bowel disease (IBD), supporting the hypothesis that butyrate supplementation may be beneficial in this setting. Nonetheless, earlier studies suggest that the oxidation of butyrate in IBD patients is altered. We propose that inflammation may decrease epithelial butyrate consumption. METHODS Non-IBD controls and IBD patients were recruited for the study. Stool samples were used for short-chain fatty acid and bacterial butyryl CoA:acetate CoA-transferase quantification. Colonic biopsies and ex vivo differentiated epithelial organoids (d-EpOCs) treated with butyrate and/or tumor necrosis factor alpha (TNFα) were used for analyzing the expression of transporters MCT1 and ABCG2, metabolic enzyme ACADS, and butyrate receptor GPR43, and for butyrate metabolism and consumption assays. RESULTS We observed that lower stool content of butyrate-producing bacteria in active IBD patients did not correlate with decreased butyrate concentrations. Indeed, the intestinal epithelial expression of MCT1, ABCG2, ACADS, and GPR43 was altered in active IBD patients. Nonetheless, d-EpOCs derived from IBD patients showed SLC16A1 (gene encoding for MCT1 protein), ABCG2, ACADS, and GPR43 expression levels comparable to controls. Moreover, IBD- and non-IBD-derived d-EpOCs responded similarly to butyrate, as assessed by transcriptional regulation. TNFα significantly altered SLC16A1, ABCG2, and GPR43 transcription in d-EpOCs, mimicking the expression profile observed in biopsies from active IBD patients and resulting in reduced butyrate consumption. CONCLUSIONS We provide evidence that the response to butyrate is not intrinsically altered in IBD patients. However, TNFα renders the epithelium less responsive to this metabolite, defeating the purpose of butyrate supplementation during active inflammation.
Collapse
Affiliation(s)
- Elena Ferrer-Picón
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - Isabella Dotti
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - Ana M Corraliza
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - Aida Mayorgas
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - Miriam Esteller
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - José Carlos Perales
- Department of Physiological Sciences, Faculty of Medicine, University of Barcelona, L’Hospitalet del Llobregat, Barcelona, Spain
| | - Elena Ricart
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - Maria C Masamunt
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain
| | - Anna Carrasco
- Department of Gastroenterology, Hospital Universitari Mútua Terrassa, CIBERehd, Barcelona, Spain
| | - Eva Tristán
- Department of Gastroenterology, Hospital Universitari Mútua Terrassa, CIBERehd, Barcelona, Spain
| | - Maria Esteve
- Department of Gastroenterology, Hospital Universitari Mútua Terrassa, CIBERehd, Barcelona, Spain
| | - Azucena Salas
- Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Barcelona, Spain,Address correspondence to: Azucena Salas, Department of Gastroenterology, IDIBAPS, Hospital Clínic, CIBERehd, Rosselló 149-153, 08036, Barcelona, Spain ()
| |
Collapse
|