1
|
Zhao L, Weng W, Ni M, Shen H, Zhang S, Chen Y, Jia R, Fan L, Mao Y, Qin L, Liu S, Wang Y. Rubidium salt can effectively relieve the symptoms of DSS-induced ulcerative colitis. Biomed Pharmacother 2024; 181:117574. [PMID: 39520912 DOI: 10.1016/j.biopha.2024.117574] [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: 07/03/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic condition that afflicts individuals repeatedly and cannot be cured at present, which has seriously affected the quality of life of patients. Minerals Containing Rubidium (MCR) from Guangxi Yuechengling, which Professor Zhao Lichun purified, were explored. Against this backdrop, the present study investigates the efficacy of rubidium salt in ulcerative colitis. Rubidium salt reduced levels of inflammatory markers and improved intestinal barrier function through the Elisa kit, immunohistochemistry, and qPCR. Next, we detected the level of short-chain fatty acid and found that the content of propanoic acid, butyric acid, and n-butyric acid increased after treatment with rubidium salt. We used fecal metagenomics to explore the underlying reasons further and found that rubidium salt significantly adjusted the structure of intestinal flora, increased the abundance of beneficial bacteria such as lactobacillus and bifidobacterium, and inhibited the abundance of harmful bacteria such as Enterobacteriaceae and Escherichia coli. We also learned that rubidium salt directly weakened pathogenic bacteria's infection and survival ability by reducing the expression of virulence factors such as fimH, invA, and hilA and virulence genes such as acrA and ompR. Overall, rubidium salt can reduce harmful bacteria and increase beneficial bacteria. The increased beneficial bacteria help enhance the gut barrier and regulate inflammatory factors by raising the levels of short-chain fatty acids. A strengthened gut barrier further stabilizes microbial homeostasis, ultimately alleviating ulcerative colitis.
Collapse
Affiliation(s)
- Lichun Zhao
- Guang Xi University of Chinese Medicine, Nanning, China; Guangxi Zhuangyao Pharmaceutical Engineering Technology Research Center, Nanning 530200, China
| | - Wenhao Weng
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Mengyue Ni
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Haoyu Shen
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Shuai Zhang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Yaning Chen
- Department of Pharmacology, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Ruining Jia
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Linzi Fan
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Yuanhui Mao
- Guang Xi University of Chinese Medicine, Nanning, China
| | - Linyin Qin
- Guang Xi University of Chinese Medicine, Nanning, China
| | - Shengzhi Liu
- Department of Pharmacology, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China.
| | - Yuji Wang
- Guang Xi University of Chinese Medicine, Nanning, China; Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; Department of Pharmacology, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China; Beijing Laboratory of Oral Health, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
2
|
Van Hul M, Cani PD, Petitfils C, De Vos WM, Tilg H, El-Omar EM. What defines a healthy gut microbiome? Gut 2024; 73:1893-1908. [PMID: 39322314 PMCID: PMC11503168 DOI: 10.1136/gutjnl-2024-333378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/16/2024] [Indexed: 09/27/2024]
Abstract
The understanding that changes in microbiome composition can influence chronic human diseases and the efficiency of therapies has driven efforts to develop microbiota-centred therapies such as first and next generation probiotics, prebiotics and postbiotics, microbiota editing and faecal microbiota transplantation. Central to microbiome research is understanding how disease impacts microbiome composition and vice versa, yet there is a problematic issue with the term 'dysbiosis', which broadly links microbial imbalances to various chronic illnesses without precision or definition. Another significant issue in microbiome discussions is defining 'healthy individuals' to ascertain what characterises a healthy microbiome. This involves questioning who represents the healthiest segment of our population-whether it is those free from illnesses, athletes at peak performance, individuals living healthily through regular exercise and good nutrition or even elderly adults or centenarians who have been tested by time and achieved remarkable healthy longevity.This review advocates for delineating 'what defines a healthy microbiome?' by considering a broader range of factors related to human health and environmental influences on the microbiota. A healthy microbiome is undoubtedly linked to gut health. Nevertheless, it is very difficult to pinpoint a universally accepted definition of 'gut health' due to the complexities of measuring gut functionality besides the microbiota composition. We must take into account individual variabilities, the influence of diet, lifestyle, host and environmental factors. Moreover, the challenge in distinguishing causation from correlation between gut microbiome and overall health is presented.The review also highlights the resource-heavy nature of comprehensive gut health assessments, which hinders their practicality and broad application. Finally, we call for continued research and a nuanced approach to better understand the intricate and evolving concept of gut health, emphasising the need for more precise and inclusive definitions and methodologies in studying the microbiome.
Collapse
Affiliation(s)
- Matthias Van Hul
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Camille Petitfils
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
| | - Willem M De Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medizinische Universitat Innsbruck, Innsbruck, Austria
| | - Emad M El-Omar
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
3
|
Van Hul M, Neyrinck AM, Everard A, Abot A, Bindels LB, Delzenne NM, Knauf C, Cani PD. Role of the intestinal microbiota in contributing to weight disorders and associated comorbidities. Clin Microbiol Rev 2024; 37:e0004523. [PMID: 38940505 PMCID: PMC11391702 DOI: 10.1128/cmr.00045-23] [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] [Indexed: 06/29/2024] Open
Abstract
SUMMARYThe gut microbiota is a major factor contributing to the regulation of energy homeostasis and has been linked to both excessive body weight and accumulation of fat mass (i.e., overweight, obesity) or body weight loss, weakness, muscle atrophy, and fat depletion (i.e., cachexia). These syndromes are characterized by multiple metabolic dysfunctions including abnormal regulation of food reward and intake, energy storage, and low-grade inflammation. Given the increasing worldwide prevalence of obesity, cachexia, and associated metabolic disorders, novel therapeutic strategies are needed. Among the different mechanisms explaining how the gut microbiota is capable of influencing host metabolism and energy balance, numerous studies have investigated the complex interactions existing between nutrition, gut microbes, and their metabolites. In this review, we discuss how gut microbes and different microbiota-derived metabolites regulate host metabolism. We describe the role of the gut barrier function in the onset of inflammation in this context. We explore the importance of the gut-to-brain axis in the regulation of energy homeostasis and glucose metabolism but also the key role played by the liver. Finally, we present specific key examples of how using targeted approaches such as prebiotics and probiotics might affect specific metabolites, their signaling pathways, and their interactions with the host and reflect on the challenges to move from bench to bedside.
Collapse
Affiliation(s)
- Matthias Van Hul
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, France/Belgium
| | - Audrey M Neyrinck
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
| | - Amandine Everard
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | | | - Laure B Bindels
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Nathalie M Delzenne
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
| | - Claude Knauf
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, France/Belgium
- INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), Université Paul Sabatier, Toulouse III, CHU Purpan, Toulouse, France
| | - Patrice D Cani
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, France/Belgium
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), Brussels, Belgium
| |
Collapse
|
4
|
Zhang X, Zhang X, Yang Y. Update of gut gas metabolism in ulcerative colitis. Expert Rev Gastroenterol Hepatol 2024; 18:339-349. [PMID: 39031456 DOI: 10.1080/17474124.2024.2383635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 07/19/2024] [Indexed: 07/22/2024]
Abstract
INTRODUCTION Ulcerative colitis (UC) is a chronic, nonspecific inflammatory disease of the intestine. The intestinal microbiota is essential in the occurrence and development of UC. Gut gases are produced via bacterial fermentation or chemical interactions, which can reveal altered intestinal microbiota, abnormal cellular metabolism, and inflammation responses. Recent studies have demonstrated that UC patients have an altered gut gas metabolism. AREAS COVERED In this review, we integrate gut gas metabolism advances in UC and discuss intestinal gases' clinical values as new biomarkers or therapeutic targets for UC, providing the foundation for further research. Literature regarding gut gas metabolism and its significance in UC from inception to October 2023 was searched on the MEDLINE database and references from relevant articles were investigated. EXPERT OPINION Depending on their type, concentration, and volume, gut gases can induce or alleviate clinical symptoms and regulate intestinal motility, inflammatory responses, immune function, and oxidative stress, significantly impacting UC. Gut gases may function as new biomarkers and provide potential diagnostic or therapeutic targets for UC.
Collapse
Affiliation(s)
- Xiaohan Zhang
- Medical School, Nankai University, Tianjin, China
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiuli Zhang
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yunsheng Yang
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
5
|
Halmos EP, Godny L, Vanderstappen J, Sarbagili-Shabat C, Svolos V. Role of diet in prevention versus treatment of Crohn's disease and ulcerative colitis. Frontline Gastroenterol 2024; 15:247-257. [PMID: 38665795 PMCID: PMC11042448 DOI: 10.1136/flgastro-2023-102417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/10/2023] [Indexed: 04/28/2024] Open
Abstract
Diet is a modifiable risk factor for disease course and data over the past decade have emerged to indicate its role in Crohn's disease (CD) and ulcerative colitis (UC). However, literature is riddled with misinterpretation of data, often leading to unexpected or conflicting results. The key understanding is that causative factors in disease development do not always proceed to an opportunity to change disease course, once established. Here, we discuss the data on dietary influences in three distinct disease states for CD and UC-predisease, active disease and quiescent disease. We appraise the literature for how our dietary recommendations should be shaped to prevent disease development and if or how that differs for CD and UC induction therapy and maintenance therapy. In UC, principles of healthy eating are likely to play a role in all states of disease. Conversely, data linking dietary factors to CD prevention and treatment are paradoxical with the highest quality evidence for CD treatment being exclusive enteral nutrition, a lactose, gluten and fibre-free diet comprising solely of ultraprocessed food-all dietary factors that are not associated or inversely associated with CD prevention. High-quality evidence from dietary trials is much awaited to expand our understanding and ultimately lead our dietary recommendations for targeted patient populations.
Collapse
Affiliation(s)
- Emma P Halmos
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Victoria, Australia
| | - Lihi Godny
- Division of Gastroenterology and Nutrition Unit, Rabin Medical Center, Petah Tikva, Israel
| | - Julie Vanderstappen
- Department of Gastroenterology and Hepatology, University Hospitals of Leuven, Leuven, Belgium
| | - Chen Sarbagili-Shabat
- Pediatric Gastroenterology and Nutrition Unit, The E. Wolfson Medical Center, Holon, Israel
- The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Vaios Svolos
- School of Medicine, Dentistry and Nursing, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Laboratory of Clinical Nutrition and Dietetics, Department of Nutrition and Dietetics, School of Physical Education, Sports Science and Dietetics, University of Thessaly, Trikala, Greece
| |
Collapse
|
6
|
Huangfu W, Cao S, Li S, Zhang S, Liu M, Liu B, Zhu X, Cui Y, Wang Z, Zhao J, Shi Y. In vitro and in vivo fermentation models to study the function of dietary fiber in pig nutrition. Appl Microbiol Biotechnol 2024; 108:314. [PMID: 38683435 PMCID: PMC11058960 DOI: 10.1007/s00253-024-13148-9] [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: 01/16/2024] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
The importance of dietary fiber (DF) in animal diets is increasing with the advancement of nutritional research. DF is fermented by gut microbiota to produce metabolites, which are important in improving intestinal health. This review is a systematic review of DF in pig nutrition using in vitro and in vivo models. The fermentation characteristics of DF and the metabolic mechanisms of its metabolites were summarized in an in vitro model, and it was pointed out that SCFAs and gases are the important metabolites connecting DF, gut microbiota, and intestinal health, and they play a key role in intestinal health. At the same time, some information about host-microbe interactions could have been improved through traditional animal in vivo models, and the most direct feedback on nutrients was generated, confirming the beneficial effects of DF on sow reproductive performance, piglet intestinal health, and growing pork quality. Finally, the advantages and disadvantages of different fermentation models were compared. In future studies, it is necessary to flexibly combine in vivo and in vitro fermentation models to profoundly investigate the mechanism of DF on the organism in order to promote the development of precision nutrition tools and to provide a scientific basis for the in-depth and rational utilization of DF in animal husbandry. KEY POINTS: • The fermentation characteristics of dietary fiber in vitro models were reviewed. • Metabolic pathways of metabolites and their roles in the intestine were reviewed. • The role of dietary fiber in pigs at different stages was reviewed.
Collapse
Affiliation(s)
- Weikang Huangfu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Shixi Cao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Shouren Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Shuhang Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Mengqi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Boshuai Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
- Henan Forage Engineering Technology Research Center, Zhengzhou, 450002, Henan, China
| | - Xiaoyan Zhu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
- Henan Forage Engineering Technology Research Center, Zhengzhou, 450002, Henan, China
| | - Yalei Cui
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
- Henan Forage Engineering Technology Research Center, Zhengzhou, 450002, Henan, China
| | - Zhichang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
- Henan Forage Engineering Technology Research Center, Zhengzhou, 450002, Henan, China
| | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Yinghua Shi
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China.
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China.
- Henan Forage Engineering Technology Research Center, Zhengzhou, 450002, Henan, China.
| |
Collapse
|
7
|
Sasama Y, Yoshimura K, Hoshino M, Sasa K, Akaike T, Morita M, Baba K, Shirota T, Miyamoto Y. Supersulfides support bone growth by promoting chondrocyte proliferation in the growth plates. J Oral Biosci 2024; 66:76-81. [PMID: 37979656 DOI: 10.1016/j.job.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023]
Abstract
OBJECTIVES While chondrocytes have mitochondria, they receive little O2 from the bloodstream. Sulfur respiration, an essential energy production system in mitochondria, uses supersulfides instead of O2. Supersulfides are inorganic and organic sulfides with catenated sulfur atoms and are primarily produced by cysteinyl tRNA synthetase-2 (CARS2). Here, we investigated the role of supersulfides in chondrocyte proliferation and bone growth driven by growth plate chondrocyte proliferation. METHODS We examined the effects of NaHS, an HS-/H2S donor, and cystine, the cellular source of cysteine, on the proliferation of mouse primary chondrocytes and growth of embryonic mouse tibia in vitro. We also examined the effect of RNA interference acting on the Cars2 gene on chondrocyte proliferation in the presence of cystine. RESULTS NaHS (30 μmol/L) enhanced tibia longitudinal growth in vitro with expansion of the proliferating zone of their growth plates. While NaHS (30 μmol/L) also promoted chondrocyte proliferation only under normoxic conditions (20 % O2), cystine (0.5 mmol/L) promoted it under both normoxic and hypoxic (2 % O2) conditions. Cars2 gene knockdown abrogated the ability of cystine (0.5 mmol/L) to promote chondrocyte proliferation under normoxic conditions, indicating that supersulfides produced by CARS2 were responsible for the cystine-dependent promotion of bone growth. CONCLUSIONS The presented results indicate that supersulfides play a vital role in bone growth achieved by chondrocyte proliferation in the growth plates driven by sulfur respiration.
Collapse
Affiliation(s)
- Yuji Sasama
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan; Department of Oral and Maxillofacial Surgery, Showa University School of Dentistry, Tokyo, Japan
| | - Kentaro Yoshimura
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan
| | - Marie Hoshino
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan; Department of Prosthodontics, Showa University School of Dentistry, Tokyo, Japan
| | - Kiyohito Sasa
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masanobu Morita
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuyoshi Baba
- Department of Prosthodontics, Showa University School of Dentistry, Tokyo, Japan
| | - Tatsuo Shirota
- Department of Oral and Maxillofacial Surgery, Showa University School of Dentistry, Tokyo, Japan
| | - Yoichi Miyamoto
- Department of Biochemistry, Showa University School of Dentistry, Tokyo, Japan.
| |
Collapse
|
8
|
Sun T, Ren K, Xu G, Ma R, Wang X, Min T, Xie X, Sun A, Ma Y, Wang H, Zhang Y, Zhu K, Dang C, Zhang G, Zhang H. Plasma-Activated Solutions Mitigates DSS-Induced Colitis via Restoring Redox Homeostasis and Reversing Microbiota Dysbiosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304044. [PMID: 37870220 PMCID: PMC10700679 DOI: 10.1002/advs.202304044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/10/2023] [Indexed: 10/24/2023]
Abstract
Ulcerative colitis is a chronic disease that increases the risk of developing colorectal cancer. Conventional medications are limited by drug delivery and a weak capacity to modulate the inflammatory microenvironment. Further, gut microbiota dysbiosis caused by mucosal damage and dysregulated redox homeostasis leads to frequent recurrence. Therefore, promoting mucosal healing and restoring redox homeostasis is considered the initial step in treating ulcerative colitis. Plasma-activated solutions (PAS) are liquids rich in various reactive nitrogen species (RNS) and reactive oxygen species (ROS) and are used to treat multiple diseases. However, its effect on ulcerative colitis remains to be examined. Therefore, using a DSS-induced mice colitis model, it is found that PAS has the potential to treat colitis and prevent its recurrence by promoting intestinal mucosal repair, reducing inflammation, improving redox homeostasis, and reversing gut microbiota dysbiosis. Further, an equipment is designed for preparing PAS without using nitrogen; however, after treatment with the Nitro-free PAS, the therapeutic effect of PAS is significantly weakened or even lost, indicating that RNS may be the main mediator by which PAS exerts its therapeutic effects. Overall, this study demonstrates the treatment of ulcerative colitis as a novel application of PAS.
Collapse
Affiliation(s)
- Tuanhe Sun
- Department of Surgical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Kaijie Ren
- Department of Surgical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Guimin Xu
- State Key Laboratory of Electrical Insulation and Power EquipmentSchool of Electrical EngineeringXi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Rulan Ma
- Department of Surgical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Xueni Wang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Tianhao Min
- Department of Surgical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Xin Xie
- Department of Nuclear MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Anbang Sun
- State Key Laboratory of Electrical Insulation and Power EquipmentSchool of Electrical EngineeringXi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Yuyi Ma
- Department of Surgical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Haonan Wang
- Department of Surgical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Yong Zhang
- Department of Surgical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Kun Zhu
- Department of Surgical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Chengxue Dang
- Department of Surgical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Guanjun Zhang
- State Key Laboratory of Electrical Insulation and Power EquipmentSchool of Electrical EngineeringXi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Hao Zhang
- Department of Surgical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| |
Collapse
|
9
|
Talebi S, Zeraattalab-Motlagh S, Rahimlou M, Naeini F, Ranjbar M, Talebi A, Mohammadi H. The Association between Total Protein, Animal Protein, and Animal Protein Sources with Risk of Inflammatory Bowel Diseases: A Systematic Review and Meta-Analysis of Cohort Studies. Adv Nutr 2023; 14:752-761. [PMID: 37187455 PMCID: PMC10334156 DOI: 10.1016/j.advnut.2023.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/17/2023] Open
Abstract
We aimed to conduct this dose-dependent meta-analysis to examine the relation between total protein, animal protein, and its sources with inflammatory bowel disease (IBD). We searched databases, comprising PubMed/Medline, Web of Science (ISI), Embase, and Google Scholar, for the published studies up to 28 March 2023. Prospective cohort study designs that investigated associations between dietary intake of various animal protein sources and with risk of IBD in the general population were identified. Eleven prospective cohort studies with 4,302,554 participants and 8067 cases were considered eligible. Findings indicated that higher intake of dairy was significantly associated with a lower risk of IBD (relative risk [RR]: 0.81; 95% confidence interval [CI]: 0.72, 0.90), Crohn disease (RR: 0.69; 95% CI: 0.56, 0.86), and ulcerative colitis (RR: 0.84; 95% CI: 0.75, 0.94). There was no association between different sources of animal protein and the risk of IBD. The dose-response analysis suggested that each 100 g/d increment in dietary total meat consumption was associated with a 38% greater risk of IBD. Moreover, a positive linear association was found between total meat intake and risk of IBD (Pnonlinearity = 0.522, Pdose-response = 0.005). Overall, among the dietary sources of protein, the risk of IBD increased only with increasing total meat intake, and the consumption of protein from dairy products was found to be a protective factor against the IBD risk. This trial was registered at PROSPERO as CRD42023397719.
Collapse
Affiliation(s)
- Sepide Talebi
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran; Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Sheida Zeraattalab-Motlagh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehran Rahimlou
- Department of Nutrition, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Fatemeh Naeini
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran; Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Ranjbar
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Talebi
- Clinical Pharmacy Department, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Mohammadi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Yao CK, Sarbagili-Shabat C. Gaseous metabolites as therapeutic targets in ulcerative colitis. World J Gastroenterol 2023; 29:682-691. [PMID: 36742165 PMCID: PMC9896612 DOI: 10.3748/wjg.v29.i4.682] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/19/2022] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
Diet therapies are currently under-utilised in optimising clinical outcomes for patients with active ulcerative colitis (UC). Furthermore, existing dietary therapies are framed by poorly defined mechanistic targets to warrant its success. There is good evidence to suggest that microbial production of gaseous metabolites, hydrogen sulfide (H2S) and nitric oxide (NO) are implicated in the development of mucosal inflammation in UC. On a cellular level, exposure of the colonic epithelium to excessive concentrations of these gases are shown to promote functional defects described in UC. Hence, targeting bacterial production of these gases could provide an opportunity to formulate new dietary therapies in UC. Despite the paucity of evidence, there is epidemiological and clinical data to support the concept of reducing mucosal inflammation in UC via dietary strategies that reduce H2S. Several dietary components, namely sulphur-containing amino acids and inorganic sulphur have been shown to be influential in enhancing colonic H2S production. More recent data suggests increasing the supply of readily fermentable fibre as an effective strategy for H2S reduction. Conversely, very little is known regarding how diet alters microbial production of NO. Hence, the current evidence suggest that a whole diet approach is needed. Finally, biomarkers for assessing changes in microbial gaseous metabolites in response to dietary interventions are very much required. In conclusion, this review identifies a great need for high quality randomised-controlled trials to demonstrate the efficacy of a sulphide-reducing dietary therapy for patients with active UC.
Collapse
Affiliation(s)
- Chu K Yao
- Department of Gastroenterology, Monash University, Melbourne 3004, Australia
| | - Chen Sarbagili-Shabat
- Pediatric Gastroenterology Unit, PIBD Research Center, Wolfson Medical Center, Holon 5822012, Israel
| |
Collapse
|
11
|
Nüse B, Holland T, Rauh M, Gerlach RG, Mattner J. L-arginine metabolism as pivotal interface of mutual host-microbe interactions in the gut. Gut Microbes 2023; 15:2222961. [PMID: 37358082 PMCID: PMC10294761 DOI: 10.1080/19490976.2023.2222961] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 06/05/2023] [Indexed: 06/27/2023] Open
Abstract
L-arginine (L-arg) is a versatile amino acid and a central intestinal metabolite in mammalian and microbial organisms. Thus, L-arg participates as precursor of multiple metabolic pathways in the regulation of cell division and growth. It also serves as a source of carbon, nitrogen, and energy or as a substrate for protein synthesis. Consequently, L-arg can simultaneously modify mammalian immune functions, intraluminal metabolism, intestinal microbiota, and microbial pathogenesis. While dietary intake, protein turnover or de novo synthesis usually supply L-arg in sufficient amounts, the expression of several key enzymes of L-arg metabolism can change rapidly and dramatically following inflammation, sepsis, or injury. Consequently, the availability of L-arg can be restricted due to increased catabolism, transforming L-arg into an essential amino acid. Here, we review the enzymatic pathways of L-arg metabolism in microbial and mammalian cells and their role in immune function, intraluminal metabolism, colonization resistance, and microbial pathogenesis in the gut.
Collapse
Affiliation(s)
- Björn Nüse
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Tim Holland
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Roman G. Gerlach
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, FAUErlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
12
|
Abdelbary MMH, Hatting M, Bott A, Dahlhausen A, Keller D, Trautwein C, Conrads G. The oral-gut axis: Salivary and fecal microbiome dysbiosis in patients with inflammatory bowel disease. Front Cell Infect Microbiol 2022; 12:1010853. [PMID: 36275026 PMCID: PMC9585322 DOI: 10.3389/fcimb.2022.1010853] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a group of chronic inflammatory disorders that fall into two main categories: Crohn's disease (CD) and ulcerative colitis (UC). The gastrointestinal tract extends from the mouth to the anus and harbors diverse bacterial communities. Several sequencing-based studies have identified an intestinal enrichment of oral-associated bacteria and demonstrated their ability to induce intestinal inflammation in mice, suggesting that intestinal pathobionts originate from the oral cavity, particularly members of the genus Streptococcus. This study aimed to investigate the composition of the salivary and fecal microbiome of IBD patients (n = 14) compared to healthy controls (n = 12) and to determine the abundance of common bacterial taxa in both niches. Metagenomic DNA was extracted from saliva and fecal samples, and the 16S rRNA gene was targeted for sequencing. Our results revealed that the overall microbial composition of saliva was significantly altered in the IBD patients compared to the control subjects (p = 0.038). At the genus level, Veillonella and Prevotella were highly abundant in IBD (median: 25.4% and 22.2%, respectively) compared to the control group (17.9% and 13.4%, respectively). In contrast, Neisseria, Streptococcus, Haemophilus, and Fusobacterium were associated with a healthy gut state. Regarding the fecal microbiome, the IBD group had a significantly higher abundance of Clostridium sensu stricto 1 and Escherichia-Shigella (both comprising pathogenic bacteria) compared with the control group. Members of both bacterial groups have previously been shown to positively correlate with intestinal inflammation and high expression of pro-inflammatory cytokines that disrupt intestinal barrier integrity. In addition, we demonstrate that the increased abundance of Clostridium sensu stricto 1 and Escherichia-Shigella has also been associated with significant upregulation of certain metabolic pathways in the feces of the IBD group, including bacterial invasion of epithelial cells. Streptococcus was the only common genus detected in both the salivary and fecal microbiome and represented the oral-gut axis in our study. Using culture-based methods, we isolated 57 and 91 Streptococcus strains from saliva as well as 40 and 31 strains from fecal samples of the controls and IBD patients, respectively. The phylogenetic tree of streptococci based on sodA sequences revealed several patient-specific clusters comprising salivary and fecal streptococcal isolates from the same patient and belonging to the same species, suggesting that the oral cavity is an endogenous reservoir for intestinal strains.
Collapse
Affiliation(s)
- Mohamed M. H. Abdelbary
- Division of Oral Microbiology and Immunology, Department of Operative Dentistry, Periodontology and Preventive Dentistry, Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital, Aachen, Germany
| | | | - Alexandra Bott
- Division of Oral Microbiology and Immunology, Department of Operative Dentistry, Periodontology and Preventive Dentistry, Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital, Aachen, Germany
| | - Andrea Dahlhausen
- University Medical Center for Occupational Medicine, RWTH University, Aachen, Germany
| | - Doris Keller
- University Medical Center for Occupational Medicine, RWTH University, Aachen, Germany
| | | | - Georg Conrads
- Division of Oral Microbiology and Immunology, Department of Operative Dentistry, Periodontology and Preventive Dentistry, Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital, Aachen, Germany
| |
Collapse
|
13
|
Day AS, Yao CK, Costello SP, Ruszkiewicz A, Andrews JM, Gibson PR, Bryant RV. Therapeutic Potential of the 4 Strategies to SUlfide-REduction (4-SURE) Diet in Adults with Mild to Moderately Active Ulcerative Colitis: An Open-Label Feasibility Study. J Nutr 2022; 152:1690-1701. [PMID: 35451489 DOI: 10.1093/jn/nxac093] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/15/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diet therapy may bridge the therapeutic gap in ulcerative colitis (UC). OBJECTIVES The novel 4-SURE diet (4-strategies-to-SUlfide-REduction), designed to modulate colonic fermentation and influence production of excess hydrogen sulfide, was examined in a feasibility study for tolerability, clinical efficacy, and effects on microbial endpoints. METHODS Adults aged ≥18 y old with mild to moderately active UC were advised to increase intake of fermentable fibers, restrict total and sulfur-containing proteins, and avoid specific food additives for 8 wk. The primary outcome was tolerability of diet [100-mm visual analogue scale (VAS) with 100-mm being intolerable]. Secondary exploratory outcomes were self-reported adherence (always adherent ≥76-100%), clinical and endoscopic response (reduction in partial Mayo ≥2 and Mayo endoscopic subscore ≥1), modulation of fecal characteristics including markers of protein and carbohydrate fermentation, and food-related quality of life (IBD-FRQoL-29). Primary analysis was by intention to treat, performed using paired t and Wilcoxon signed-rank statistical tests. RESULTS Twenty-eight adults with UC [mean (range) age: 42 (22-72) y, 15 females, 3 proctitis, 14 left-sided, and 11 extensive] were studied. Prescribed dietary targets were achieved overall. The diet was well tolerated (VAS: 19 mm; 95% CI: 7, 31 mm) with 95% frequently or always adherent. Clinical response occurred in 13 of 28 (46%) and endoscopic improvement in 10 of 28 participants (36%). Two participants (7%) worsened. Fecal excretion of SCFAs increased by 69% (P < 0.0001), whereas the proportion of branched-chain fatty acids to SCFAs was suppressed by 27% (-1.34%; 95% CI: -2.28%, -0.40%; P = 0.007). The FRQoL improved by 10 points (95% CI: 4, 16; P < 0.001). CONCLUSIONS The 4-SURE dietary strategy is considered tolerable and an acceptable diet by adults with mild to moderately active UC. The dietary teachings achieved the prescribed dietary and fecal targets. Given signals of therapeutic efficacy, further evaluation of this diet is warranted in a placebo-controlled trial. This trial was registered at https://www.anzctr.org.au (Australian New Zealand Clinical Trials Registry) as ACTRN12619000063112.
Collapse
Affiliation(s)
- Alice S Day
- Inflammatory Bowel Disease Services, Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, Woodville, Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
- Basil Hetzel Research Institute, Woodville, Australia
| | - Chu Kion Yao
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Australia
| | - Samuel P Costello
- Inflammatory Bowel Disease Services, Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, Woodville, Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
- Basil Hetzel Research Institute, Woodville, Australia
| | - Andrew Ruszkiewicz
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
- Gastroenterology Research Laboratory, SA Pathology, Adelaide, Australia
| | - Jane M Andrews
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
- Inflammatory Bowel Disease Service, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, Australia
| | - Peter R Gibson
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Australia
| | - Robert V Bryant
- Inflammatory Bowel Disease Services, Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, Woodville, Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
- Basil Hetzel Research Institute, Woodville, Australia
| |
Collapse
|
14
|
A novel fluorescent probe for real-time imaging of thionitrous acid under inflammatory and oxidative conditions. Redox Biol 2022; 54:102372. [PMID: 35728302 PMCID: PMC9214870 DOI: 10.1016/j.redox.2022.102372] [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: 04/30/2022] [Revised: 06/07/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
Thionitrous acid (HSNO), a crosstalk intermediate of two crucial gasotransmitters nitric oxide and hydrogen sulfide, plays a critical role in redox regulation of cellular signaling and functions. However, real-time and facile detection of HSNO with high selectivity and sensitivity remains highly challenging. Herein we report a novel fluorescent probe (SNP-1) for HSNO detection. SNP-1 has a simple molecular structure, but showing strong fluorescence, a low detection limit, a broad linear detection range (from nanomolar to micromolar concentrations), ultrasensitivity, and high selectivity for HSNO in both aqueous media and cells. Benefiting from these unique features, SNP-1 could effectively visualize changes of HSNO levels in mouse models of acute ulcerative colitis and renal ischemia/reperfusion injury. Moreover, the good correlation between colonic HSNO levels and disease activity index demonstrated that HSNO is a promising new diagnostic agent for acute ulcerative colitis. Therefore, SNP-1 can serve as a useful fluorescent probe for precision detection of HSNO in various biological systems, thereby facilitating mechanistic studies, therapeutic assessment, and high-content drug screening for corresponding diseases. HSNO was the preferred intermediate to study crosstalk between H2S and NO. HSNO displayed translational potential for diagnosis and assessment of diseases. SNP-1 displayed excellent fluorescence performance for HSNO detection. SNP-1 could effectively image HSNO in cells and mouse models.
Collapse
|
15
|
González-Soltero R, Bailén M, de Lucas B, Ramírez-Goercke MI, Pareja-Galeano H, Larrosa M. Role of Oral and Gut Microbiota in Dietary Nitrate Metabolism and Its Impact on Sports Performance. Nutrients 2020; 12:E3611. [PMID: 33255362 PMCID: PMC7760746 DOI: 10.3390/nu12123611] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/30/2020] [Accepted: 11/20/2020] [Indexed: 12/20/2022] Open
Abstract
Nitrate supplementation is an effective, evidence-based dietary strategy for enhancing sports performance. The effects of dietary nitrate seem to be mediated by the ability of oral bacteria to reduce nitrate to nitrite, thus increasing the levels of nitrite in circulation that may be further reduced to nitric oxide in the body. The gut microbiota has been recently implicated in sports performance by improving muscle function through the supply of certain metabolites. In this line, skeletal muscle can also serve as a reservoir of nitrate. Here we review the bacteria of the oral cavity involved in the reduction of nitrate to nitrite and the possible changes induced by nitrite and their effect on gastrointestinal balance and gut microbiota homeostasis. The potential role of gut bacteria in the reduction of nitrate to nitrite and as a supplier of the signaling molecule nitric oxide to the blood circulation and muscles has not been explored in any great detail.
Collapse
Affiliation(s)
- Rocío González-Soltero
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (M.B.); (M.I.R.-G.)
| | - María Bailén
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (M.B.); (M.I.R.-G.)
| | - Beatriz de Lucas
- Faculty of Sports Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (B.d.L.); (H.P.-G.); (M.L.)
| | - Maria Isabel Ramírez-Goercke
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (M.B.); (M.I.R.-G.)
| | - Helios Pareja-Galeano
- Faculty of Sports Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (B.d.L.); (H.P.-G.); (M.L.)
| | - Mar Larrosa
- Faculty of Sports Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (B.d.L.); (H.P.-G.); (M.L.)
| |
Collapse
|
16
|
Ardalan ZS, Yao CK, Sparrow MP, Gibson PR. Review article: the impact of diet on ileoanal pouch function and on the pathogenesis of pouchitis. Aliment Pharmacol Ther 2020; 52:1323-1340. [PMID: 32955120 DOI: 10.1111/apt.16085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/30/2020] [Accepted: 08/24/2020] [Indexed: 12/09/2022]
Abstract
BACKGROUND There is expanding interest in the role that diet plays in ileoanal pouch function and in the pathogenesis of pouchitis. AIMS To present a narrative review of published literature regarding the relationship of diet with pouch function and the pathogenesis of pouchitis, and to provide potentially beneficial dietary strategies. METHODS Current relevant literature was summarised and critically examined. RESULTS Dietary components influence pouch function via their effect on upper gastrointestinal transit, small bowel water content and the structure and fermentative activity of the pouch microbiota. FODMAPs in fruits and vegetables appear to affect pouch function the most, with intake positively associated with increased stool frequency and reduced consistency. Dietary factors that influence the pathogenesis of pouchitis appear different and, at times, opposite to those better for optimising function. For example, risk of pouchitis appears to be inversely associated with intake of fruits. The food components mechanistically responsible for this observation are not known, but a rich supply of fermentable fibres and micronutrients in such foods might play a beneficial role via modulation of microbial community structure (such as increasing diversity and/or changing microbial communities to favour 'protective' over 'pathogenic' bacteria) and function and/or anti-inflammatory effects. CONCLUSION Available data are weak but suggest tailoring dietary recommendations according to pouch phenotype/behaviour and pouchitis risk might improve outcomes. More sophisticated dietary strategies that utilise the physiological and pathophysiological effects of dietary components on ileoanal pouches have potential to further improve outcomes. Well designed, adequately powered studies are required.
Collapse
Affiliation(s)
- Zaid S Ardalan
- Department of Gastroenterology, Alfred Health and Monash University, Melbourne, VIC, Australia
| | - Chu K Yao
- Department of Gastroenterology, Alfred Health and Monash University, Melbourne, VIC, Australia
| | - Miles P Sparrow
- Department of Gastroenterology, Alfred Health and Monash University, Melbourne, VIC, Australia
| | - Peter R Gibson
- Department of Gastroenterology, Alfred Health and Monash University, Melbourne, VIC, Australia
| |
Collapse
|
17
|
Costello SP, Day A, Yao CK, Bryant RV. Faecal microbiota transplantation (FMT) with dietary therapy for acute severe ulcerative colitis. BMJ Case Rep 2020; 13:e233135. [PMID: 32843418 PMCID: PMC7449292 DOI: 10.1136/bcr-2019-233135] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2020] [Indexed: 01/25/2023] Open
Abstract
A 19-year-old man presented with acute severe ulcerative colitis. He was taking azathioprine (therapeutic metabolites) and sulphasalazine as well as infliximab with a therapeutic drug level. On day 3 of hydrocortisone therapy, he met day Oxford criteria with >8 bloody stools per day and was given faecal microbiota transplantation and subsequently commenced on dietary therapy combining several strategies-(1) increased intake of fermentable fibres, (2) reduced intake of overall and sulfur-containing protein and (3) restriction of sulfate and sulfite food additives. At week 8 assessment, he was in clinical and endoscopic remission and remained in clinical and endoscopic remission at 12 months.
Collapse
Affiliation(s)
- Samuel Paul Costello
- Department of Gastroenterology, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Department of Medicine, The University of Adelaide Faculty of Health Sciences, Adelaide, South Australia, Australia
| | - Alice Day
- Department of Gastroenterology, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Department of Medicine, The University of Adelaide Faculty of Health Sciences, Adelaide, South Australia, Australia
| | - Chu K Yao
- Department of Medicine, Monash University, Clayton, Victoria, Australia
| | - Robert Venning Bryant
- Department of Gastroenterology, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Department of Medicine, The University of Adelaide Faculty of Health Sciences, Adelaide, South Australia, Australia
| |
Collapse
|
18
|
Gibson PR, Halmos EP, Muir JG. Review article: FODMAPS, prebiotics and gut health-the FODMAP hypothesis revisited. Aliment Pharmacol Ther 2020; 52:233-246. [PMID: 32562590 DOI: 10.1111/apt.15818] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/27/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Restriction of dietary FODMAP intake can alleviate symptoms in patients with irritable bowel syndrome. Because many FODMAPs have prebiotic actions, there is concern that their dietary restriction leads to dysbiosis with health consequences, and their intake is being encouraged by addition to foods and via supplements. AIMS To examine the hazards and benefits of high and low FODMAP intake. METHODS Current literature was reviewed and alternative hypotheses formulated. RESULTS Low FODMAP intake reduces abundance of faecal Bifidobacteria without known adverse outcomes and has no effect on diversity, but the reduction in bacterial density may potentially be beneficial to gut health. Supplementary prebiotics can markedly elevate the intake of FODMAPs over levels consumed in the background diet. While this increases the abundance of Bifidobacteria, it adversely affects gut health in animal studies by inducing colonic mucosal barrier dysfunction, mucosal inflammation and visceral hypersensitivity. Rapid colonic fermentation is central to the identified mechanisms that include injury from high luminal concentrations of short-chain fatty acids and low pH, and inflammatory effects of increased endotoxin load and glycation of macromolecules. Whether these observations translate into humans requires further study. Opposing hypotheses are presented whereby excessive intake of FODMAPs might have health benefits via prebiotic effects, but might also be injurious and contribute to the apparent increase in functional intestinal disorders. CONCLUSIONS Reduced FODMAP intake has few deleterious effects on gut microbiota. Consequences (both positive and negative) of excessive carbohydrate fermentation in the human intestines from elevated FODMAP intake require more attention.
Collapse
Affiliation(s)
- Peter R Gibson
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Vic., Australia
| | - Emma P Halmos
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Vic., Australia
| | - Jane G Muir
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Vic., Australia
| |
Collapse
|
19
|
Intestinal gases: influence on gut disorders and the role of dietary manipulations. Nat Rev Gastroenterol Hepatol 2019; 16:733-747. [PMID: 31520080 DOI: 10.1038/s41575-019-0193-z] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/29/2019] [Indexed: 02/06/2023]
Abstract
The inner workings of the intestines, in which the body and microbiome intersect to influence gut function and systemic health, remain elusive. Carbon dioxide, hydrogen, methane and hydrogen sulfide, as well as a variety of trace gases, are generated by the chemical interactions and microbiota within the gut. Profiling of these intestinal gases and their responses to dietary changes can reveal the products and functions of the gut microbiota and their influence on human health. Indeed, different tools for measuring these intestinal gases have been developed, including newly developed gas-sensing capsule technology. Gases can, according to their type, concentration and volume, induce or relieve abdominal symptoms, and might also have physiological, pathogenic and therapeutic effects. Thus, profiling and modulating intestinal gases could be powerful tools for disease prevention and/or therapy. As the interactions between the microbiota, chemical constituents and fermentative substrates of the gut are principally influenced by dietary intake, altering the diet, which, in turn, changes gas profiles, is the main therapeutic approach for gastrointestinal disorders. An improved understanding of the complex interactions within the intestines that generate gases will enhance our ability to prevent, diagnose, treat and monitor many gastrointestinal disorders.
Collapse
|
20
|
Tsiaoussis J, Antoniou MN, Koliarakis I, Mesnage R, Vardavas CI, Izotov BN, Psaroulaki A, Tsatsakis A. Effects of single and combined toxic exposures on the gut microbiome: Current knowledge and future directions. Toxicol Lett 2019; 312:72-97. [PMID: 31034867 DOI: 10.1016/j.toxlet.2019.04.014] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/12/2022]
Abstract
Human populations are chronically exposed to mixtures of toxic chemicals. Predicting the health effects of these mixtures require a large amount of information on the mode of action of their components. Xenobiotic metabolism by bacteria inhabiting the gastrointestinal tract has a major influence on human health. Our review aims to explore the literature for studies looking to characterize the different modes of action and outcomes of major chemical pollutants, and some components of cosmetics and food additives, on gut microbial communities in order to facilitate an estimation of their potential mixture effects. We identified good evidence that exposure to heavy metals, pesticides, nanoparticles, polycyclic aromatic hydrocarbons, dioxins, furans, polychlorinated biphenyls, and non-caloric artificial sweeteners affect the gut microbiome and which is associated with the development of metabolic, malignant, inflammatory, or immune diseases. Answering the question 'Who is there?' is not sufficient to define the mode of action of a toxicant in predictive modeling of mixture effects. Therefore, we recommend that new studies focus to simulate real-life exposure to diverse chemicals (toxicants, cosmetic/food additives), including as mixtures, and which combine metagenomics, metatranscriptomics and metabolomic analytical methods achieving in that way a comprehensive evaluation of effects on human health.
Collapse
Affiliation(s)
- John Tsiaoussis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, 71110 Heraklion, Greece
| | - Michael N Antoniou
- Gene Expression and Therapy Group, King's College London, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, 8th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Ioannis Koliarakis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, 71110 Heraklion, Greece
| | - Robin Mesnage
- Gene Expression and Therapy Group, King's College London, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, 8th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Constantine I Vardavas
- Laboratory of Toxicology, Medical School, University of Crete, Voutes, 71409 Heraklion, Crete, Greece
| | - Boris N Izotov
- Department of Analytical, Toxicology, Pharmaceutical Chemistry and Pharmacognosy, Sechenov University, 119991 Moscow, Russia
| | - Anna Psaroulaki
- Department of Clinical Microbiology and Microbial Pathogenesis, Medical School, University of Crete, 71110 Heraklion, Greece
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, Voutes, 71409 Heraklion, Crete, Greece; Department of Analytical, Toxicology, Pharmaceutical Chemistry and Pharmacognosy, Sechenov University, 119991 Moscow, Russia.
| |
Collapse
|
21
|
Dietary intake of fish, n-3 polyunsaturated fatty acids, and risk of inflammatory bowel disease: a systematic review and meta-analysis of observational studies. Eur J Nutr 2019; 59:1-17. [PMID: 30680455 DOI: 10.1007/s00394-019-01901-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 01/10/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Fish consumption and dietary intake of n-3 polyunsaturated acids (PUFAs) may be associated with inflammatory bowel disease (IBD). We aimed to conduct a systematic review and summarize published articles on the association between fish consumption and dietary intake of n-3 PUFAs with the risk of IBD. METHODS PubMed, Scopus, and Web of Science databases were used to conduct a comprehensive search and identify eligible literature published prior to January 2019. Fixed-effects model or random-effects models (DerSimonian-Laird method) were applied to pool the effect sizes. Cochrane Q test was used to trace the potential source of heterogeneity across studies. RESULTS 12 studies (5 prospective and 7 case-control) were included in the systematic review, which ten of them were eligible for inclusion in the meta-analysis. Studies were included a total sample size of 282610 participants which 2002 of them were cases of IBD [1061 Crohn's disease (CD) and 937 ulcerative colitis (UC)]. A negative association was found between fish consumption and the incidence of CD (pooled effect size: 0.54, 95%CI: 0.31-0.96, P = 0.03). There was no relationship between total dietary n-3 PUFAs intake and IBD (pooled effect size: 1.17, 95%CI: 0.80-1.72, P = 0.41). A significant inverse association was observed between dietary long-chain n-3 PUFAs and the risk of UC (pooled effect size: 0.75, 95%CI: 0.57-0.98, P = 0.03). Moreover, no association was found between α-Linolenic acid (ALA) and IBD (pooled effect size: 1.17, 95%CI: 0.63-2.17, P = 0.62). CONCLUSIONS Findings showed a negative association between fish consumption and the risk of CD. Moreover, there was a significant inverse association between dietary long-chain n-3 PUFAs and the risk of UC.
Collapse
|
22
|
Abstract
Despite the revolution in inflammatory bowel disease (IBD) treatment over the past two decades with the advent of biological therapies, there remains a substantial proportion of patients with inadequate or unsustained response to existent therapies. The overwhelming focus of IBD therapeutics has been targeting mucosal immunity, however with the developing evidence base pointing to the role of gut microbes in the inflammatory process, renewed focus should be placed on the impact of manipulating the microbiome in IBD management. This review provides an overview of the evidence implicating bacteria in the pathogenesis of gut inflammation in IBD and provides an overview of the evidence of antibiotics in IBD treatment. We also suggest a potential role of antibiotics in clinical practice based on available evidence and clinical experience.
Collapse
Affiliation(s)
- Oren Ledder
- Shaare Zedek Medical Center, Jerusalem, Israel.,The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
23
|
Peng S, Gao J, Liu W, Jiang C, Yang X, Sun Y, Guo W, Xu Q. Andrographolide ameliorates OVA-induced lung injury in mice by suppressing ROS-mediated NF-κB signaling and NLRP3 inflammasome activation. Oncotarget 2018; 7:80262-80274. [PMID: 27793052 PMCID: PMC5348318 DOI: 10.18632/oncotarget.12918] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/14/2016] [Indexed: 12/20/2022] Open
Abstract
In this study, we attempted to explore the effect and possible mechanism of Andrographolide on OVA-induced asthma. OVA challenge induced significant airway inflammatory cell recruitment and lung histological alterations, which were ameliorated by Andrographolide. The protein levels of cytokines in bron-choalveolar fluid (BALF) and serum were reduced by Andrographolide administration as well as the mRNA levels in lung tissue. Mechanically, Andrographolide markedly hampered the activation of nuclear factor-κB (NF-κB) and NLRP3 inflammasome both in vivo and vitro thus decreased levels of TNF-α and IL-1β. Finally, we confirmed that ROS scavenging was responsible for Andrographolide's inactivation of NF-κB and NLRP3 inflammasome signaling. Our study here revealed the effect and possible mechanism of Andrographolide on asthma, which may represent a new therapeutic approach for treating this disease.
Collapse
Affiliation(s)
- Shuang Peng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Jian Gao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Wen Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Chunhong Jiang
- State Key Laboratory of Innovative Nature Medicine and TCM Injections, Jiangxi Qingfeng Pharmaceutical Co., Ltd., Ganzhou, China
| | - Xiaoling Yang
- State Key Laboratory of Innovative Nature Medicine and TCM Injections, Jiangxi Qingfeng Pharmaceutical Co., Ltd., Ganzhou, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
24
|
Monasta L, Pierobon C, Princivalle A, Martelossi S, Marcuzzi A, Pasini F, Perbellini L. Inflammatory bowel disease and patterns of volatile organic compounds in the exhaled breath of children: A case-control study using Ion Molecule Reaction-Mass Spectrometry. PLoS One 2017; 12:e0184118. [PMID: 28859138 PMCID: PMC5578606 DOI: 10.1371/journal.pone.0184118] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 08/20/2017] [Indexed: 12/22/2022] Open
Abstract
Inflammatory bowel diseases (IBD) profoundly affect quality of life and have been gradually increasing in incidence, prevalence and severity in many areas of the world, and in children in particular. Patients with suspected IBD require careful history and clinical examination, while definitive diagnosis relies on endoscopic and histological findings. The aim of the present study was to investigate whether the alveolar air of pediatric patients with IBD presents a specific volatile organic compounds’ (VOCs) pattern when compared to controls. Patients 10–17 years of age, were divided into four groups: Crohn’s disease (CD), ulcerative colitis (UC), controls with gastrointestinal symptomatology, and surgical controls with no evidence of gastrointestinal problems. Alveolar breath was analyzed by ion molecule reaction mass spectrometry. Four models were built starting from 81 molecules plus the age of subjects as independent variables, adopting a penalizing LASSO logistic regression approach: 1) IBDs vs. controls, finally based on 18 VOCs plus age (sensitivity = 95%, specificity = 69%, AUC = 0.925); 2) CD vs. UC, finally based on 13 VOCs plus age (sensitivity = 94%, specificity = 76%, AUC = 0.934); 3) IBDs vs. gastroenterological controls, finally based on 15 VOCs plus age (sensitivity = 94%, specificity = 65%, AUC = 0.918); 4) IBDs vs. controls, built starting from the 21 directly or indirectly calibrated molecules only, and finally based on 12 VOCs plus age (sensitivity = 94%, specificity = 71%, AUC = 0.888). The molecules identified by the models were carefully studied in relation to the concerned outcomes. This study, with the creation of models based on VOCs profiles, precise instrumentation and advanced statistical methods, can contribute to the development of new non–invasive, fast and relatively inexpensive diagnostic tools, with high sensitivity and specificity. It also represents a crucial step towards gaining further insights on the etiology of IBD through the analysis of specific molecules which are the expression of the particular metabolism that characterizes these patients.
Collapse
Affiliation(s)
- Lorenzo Monasta
- Institute for Maternal and Child Health – IRCCS “Burlo Garofolo”, Trieste, Italy
- * E-mail:
| | - Chiara Pierobon
- Institute for Maternal and Child Health – IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Andrea Princivalle
- Occupational Medicine, Department of Public Health and Community Medicine, University of Verona, Verona, Italy
| | - Stefano Martelossi
- Institute for Maternal and Child Health – IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Annalisa Marcuzzi
- Institute for Maternal and Child Health – IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Francesco Pasini
- Occupational Medicine, Department of Public Health and Community Medicine, University of Verona, Verona, Italy
| | - Luigi Perbellini
- Occupational Medicine, Department of Public Health and Community Medicine, University of Verona, Verona, Italy
| |
Collapse
|
25
|
Costello SP, Soo W, Bryant RV, Jairath V, Hart AL, Andrews JM. Systematic review with meta-analysis: faecal microbiota transplantation for the induction of remission for active ulcerative colitis. Aliment Pharmacol Ther 2017; 46:213-224. [PMID: 28612983 DOI: 10.1111/apt.14173] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 04/07/2017] [Accepted: 05/12/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Faecal microbiota transplantation (FMT) is emerging as a novel therapy for ulcerative colitis (UC). Interpretation of efficacy of FMT for UC is complicated by differences among studies in blinding, FMT administration procedures, intensity of therapy and donor stool processing methods. AIM To determine whether FMT is effective and safe for the induction of remission in active UC. METHODS Medline (Ovid), Embase and the Cochrane Library were searched from inception through February 2017. Original studies reporting remission rates following FMT for active UC were included. All study designs were included in the systematic review and a meta-analysis performed including only randomised controlled trials (RCTs). RESULTS There were 14 cohort studies and four RCTs that used markedly different protocols. In the meta-analysis of RCTs, clinical remission was achieved in 39 of 140 (28%) patients in the donor FMT groups compared with 13 of 137 (9%) patients in the placebo groups; odds ratio 3.67 (95% CI: 1.82-7.39, P<.01). Clinical response was achieved in 69 of 140 (49%) donor FMT patients compared to 38 of 137 (28%) placebo patients; odds ratio 2.48 (95% CI: 1.18-5.21, P=.02). In cohort studies, 39 of 168 (24%; 95% CI: 11%-40%) achieved clinical remission. CONCLUSIONS Despite variation in processes, FMT appears to be effective for induction of remission in UC, with no major short-term safety signals. Further studies are needed to better define dose frequency and preparation methods, and to explore its feasibility, efficacy and safety as a maintenance agent.
Collapse
Affiliation(s)
- S P Costello
- Department of Gastroenterology, The Queen Elizabeth Hospital, Woodville, SA, Australia.,School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - W Soo
- Department of Gastroenterology, The Queen Elizabeth Hospital, Woodville, SA, Australia
| | - R V Bryant
- Department of Gastroenterology, The Queen Elizabeth Hospital, Woodville, SA, Australia.,School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - V Jairath
- Departments of Medicine, Epidemiology and Biostatistics, Western University, London, ON, Canada
| | - A L Hart
- St Mark's Hospital, Harrow, UK.,Department of Surgery and Cancer, Imperial College, London, UK
| | - J M Andrews
- School of Medicine, University of Adelaide, Adelaide, SA, Australia.,IBD Service Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
26
|
Yu L, Yan J, Sun Z. D-limonene exhibits anti-inflammatory and antioxidant properties in an ulcerative colitis rat model via regulation of iNOS, COX-2, PGE2 and ERK signaling pathways. Mol Med Rep 2017; 15:2339-2346. [PMID: 28260017 DOI: 10.3892/mmr.2017.6241] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 12/09/2016] [Indexed: 11/06/2022] Open
Abstract
D-limonene has been demonstrated to have important immunomodulatory properties, including antitumor effects, and may alleviate asthma and allergies. In the present study, the anti‑inflammatory effects of D‑limonene were investigated in an ulcerative colitis (UC) rat model. Healthy male Sprague‑Dawley rats were randomly divided into control, untreated UC, and treatment with 50 or 100 mg/kg D‑limonene UC groups. In UC rats, disease activity and colonic mucosa damage were significantly reduced by the anti‑inflammatory effects of D‑limonene, via suppression of matrix metalloproteinase (MMP)‑2 and ‑9 gene expression. In addition, treatment with D‑limonene significantly increased antioxidant, inducible nitric oxide synthase (iNOS) and cyclooxygenase‑2 (COX‑2) protein expression levels in UC rats. A decrease in prostaglandin E2 (PGE2) production, transforming growth factor‑β (TGF‑β) gene expression and an increase phosphorylated‑extracellular signal regulated kinase (ERK) 1/2 expression levelswere observed in UC rats treated with D‑limonene. In conclusion, D‑limonene reduced MMP‑2 and ‑9 mRNA expression levels via regulation of the iNOS, COX‑2, PGE2, TGF‑β and ERK1/2 signaling pathways in a UC rat model, indicating its potential antioxidant and anti‑inflammatory properties.
Collapse
Affiliation(s)
- Lihua Yu
- Department of Gastroenterology, Wuxi Third People's Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu 214041, P.R. China
| | - Jing Yan
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Zhiguang Sun
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
27
|
Rotbart A, Yao C, Ha N, Chrisp MD, Muir JG, Gibson PR, Kalantar-zadeh K, Ou JZ. Designing an in-vitro gas profiling system for human faecal samples. SENSORS AND ACTUATORS B: CHEMICAL 2017; 238:754-764. [DOI: 10.1016/j.snb.2016.07.120] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
28
|
Yao CK, Muir JG, Gibson PR. Review article: insights into colonic protein fermentation, its modulation and potential health implications. Aliment Pharmacol Ther 2016; 43:181-196. [PMID: 26527169 DOI: 10.1111/apt.13456] [Citation(s) in RCA: 285] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 09/11/2015] [Accepted: 10/13/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Beneficial effects of carbohydrate fermentation on gastrointestinal health are well established. Conversely, protein fermentation generates harmful metabolites but their relevance to gastrointestinal health is poorly understood. AIM To review the effects of increased protein fermentation on biomarkers of colonic health, factors influencing fermentative activity and potential for dietary modulation to minimise detrimental effects. METHODS A literature search was performed in PubMed, Medline, EMBASE and Google scholar for clinical and pre-clinical studies using search terms - 'dietary protein', 'fermentation', 'putrefaction', 'phenols', 'sulphide', 'branched-chain fatty acid', 'carbohydrate fermentation', 'gastrointestinal'. RESULTS High protein, reduced carbohydrate diets alter the colonic microbiome, favouring a potentially pathogenic and pro-inflammatory microbiota profile, decreased short-chain fatty acid production and increased ammonia, phenols and hydrogen sulphide concentrations. These metabolites largely compromise the colonic epithelium structure, causing mucosal inflammation but may also directly modulate the enteric nervous system and intestinal motility. Increased protein fermentation as a result of a high-protein intake can be attenuated by addition of oligosaccharides, resistant starch and nonstarch polysaccharides and a reduction in total protein or specifically, aromatic and sulphur-containing amino acids. These factors may have clinical importance as novel therapeutic approaches to problems, in which protein fermentation may be implicated, such as malodorous flatus, irritable bowel syndrome, ulcerative colitis and prevention of colorectal cancer. CONCLUSIONS The direct clinical relevance of excessive protein fermentation in the pathogenesis of irritable bowel syndrome, malodorous flatus and ulcerative colitis are underexplored. Manipulating dietary carbohydrate and protein intake have potential therapeutic applications in such settings and warrant further clinical studies.
Collapse
Affiliation(s)
- C K Yao
- Department of Gastroenterology, Monash University, Alfred Health, Melbourne, Vic., Australia
| | - J G Muir
- Department of Gastroenterology, Monash University, Alfred Health, Melbourne, Vic., Australia
| | - P R Gibson
- Department of Gastroenterology, Monash University, Alfred Health, Melbourne, Vic., Australia
| |
Collapse
|
29
|
Dinc S, Caydere M, Akgul G, Yenidogan E, Hücümenoglu S, Rajesh M. Methylene Blue inhibits the inflammatory process of the acetic acid-induced colitis in the rat colonic mucosa. Int Surg 2015; 100:1364-1374. [PMID: 26062761 DOI: 10.9738/intsurg-d-15-00118.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Inflammatory bowel disease is a serious health problem. Although it has been widely investigated, treatment of inflammatory bowel diseases currently remains as a challenging clinical problem. Over production of nitric oxide has been demonstrated to cause tissue damage and inflammation. In this study, the effect of methylene blue (MB), a well-known inhibitor of nitric oxide synthesis, was investigated in acetic acid (AA)-induced colitis model in Sprague-Dawley rats. Eighty male rats randomized into 4 groups (control, control MB, colitis, colitis + MB). AA was applied to groups 3 and 4. MB was added into group 2 and 4. Three days later, animals were sacrificed and 8 cm distal colonic segment resected and the specimens are examined using macroscopical, histological, and biochemical methods. The results of the macroscopic and microscopic examination showed that in group 4 the mucosal damage and inflammation score significantly lower than group 3. Increased intestinal permeability in acetic acid-administered group was significantly reversed by MB application. Myeloperoxidase activity and malondialdehyde levels increased significantly, while superoxide dismutase and catalase activities were suppressed after AA-administration. These biochemical parameters were reversed in MB-treated group. Administration of acetic acid resulted in increased levels of tumor necrosis factor-α, interleukin-1β, interleukin-6, total nitrite/nitrate levels and nitric oxide synthase activity. These biochemical alterations were significantly reversed by MB application also. In conclusion, our results indicate that MB decreases the level of nitric oxide and decreases inflammation in acetic acid-induced colitis.
Collapse
Affiliation(s)
- Soykan Dinc
- b ankara research and traing hospital, ankara, 06800, Turkey
| | | | | | | | | | | |
Collapse
|
30
|
Magierowski M, Magierowska K, Kwiecien S, Brzozowski T. Gaseous mediators nitric oxide and hydrogen sulfide in the mechanism of gastrointestinal integrity, protection and ulcer healing. Molecules 2015; 20:9099-123. [PMID: 25996214 PMCID: PMC6272495 DOI: 10.3390/molecules20059099] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 04/29/2015] [Accepted: 05/13/2015] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are known as biological messengers; they play an important role in human organism and contribute to many physiological and pathophysiological processes. NO is produced from l-arginine by constitutive NO synthase (NOS) and inducible NOS enzymatic pathways. This gaseous mediator inhibits platelet aggregation, leukocyte adhesion and contributes to the vessel homeostasis. NO is known as a vasodilatory molecule involved in control of the gastric blood flow (GBF) and the maintenance of gastric mucosal barrier integrity in either healthy gastric mucosa or that damaged by strong irritants. Biosynthesis of H2S in mammals depends upon two enzymes cystathionine-β-synthase and cystathionine γ-lyase. This gaseous mediator, similarly to NO and carbon monoxide, is involved in neuromodulation, vascular contractility and anti-inflammatory activities. For decades, H2S has been known to inhibit cytochrome c oxidase and reduce cell energy production. Nowadays it is generally considered to act through vascular smooth muscle ATP-dependent K+ channels, interacting with intracellular transcription factors and promote sulfhydration of protein cysteine moieties within the cell, but the mechanism of potential gastroprotective and ulcer healing properties of H2S has not been fully explained. The aim of this review is to compare current results of the studies concerning the role of H2S and NO in gastric mucosa protection and outline areas that may pose new opportunities for further development of novel therapeutic targets.
Collapse
Affiliation(s)
- Marcin Magierowski
- Department of Physiology, Jagiellonian University Medical College, Cracow 31-531, Poland.
| | - Katarzyna Magierowska
- Department of Physiology, Jagiellonian University Medical College, Cracow 31-531, Poland.
| | - Slawomir Kwiecien
- Department of Physiology, Jagiellonian University Medical College, Cracow 31-531, Poland.
| | - Tomasz Brzozowski
- Department of Physiology, Jagiellonian University Medical College, Cracow 31-531, Poland.
| |
Collapse
|
31
|
Zhang CB, Tang YC, Xu XJ, Guo SX, Wang HZ. Hydrogen gas inhalation protects against liver ischemia/reperfusion injury by activating the NF-κB signaling pathway. Exp Ther Med 2015; 9:2114-2120. [PMID: 26136944 DOI: 10.3892/etm.2015.2385] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 05/30/2014] [Indexed: 11/05/2022] Open
Abstract
Hydrogen has been demonstrated to function as a novel antioxidant and exert therapeutic antioxidant activity in a number of diseases. The present study was designed to investigate the effect of hydrogen inhalation on liver ischemia/reperfusion (I/R) injury in rats. The portal triad to the left lobe and the left middle lobe of the liver were completely occluded for 90 min. This was followed by reperfusion for 180 min. The rats subsequently underwent syngeneic orthotopic liver transplantation. Inhalation of various concentrations (1, 2 and 3%) of hydrogen gas and its administration for different durations (1, 3 and 6 h) immediately prior to the I/R injury allowed the optimal dose and duration of administration to be determined. Liver injury was evaluated through biochemical and histopathological examinations. The expression levels of proinflammatory cytokines, including tumor necrosis factor (TNF)-α and interleukin (IL)-6, were measured by enzyme-linked immunosorbent assay and quantitative polymerase chain reaction (qPCR). Liver nuclear factor κB (NF-κB) was detected by qPCR and western blot analysis. Inhalation of hydrogen gas at 2% concentration for 1 h significantly reduced the serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities, the expression of cytokines, including IL-6, TNF-α, early growth response protein 1 (Egr-1) and IL-1β, and morphological damage. In addition, the mRNA and protein expression levels of NF-κB, heme oxygenase-1 (HO-1), B-cell lymphoma 2 (Bcl-2) and zinc finger protein A20 (A20) in rats where only the donors received hydrogen were significantly increased compared with those in rats where both the donor and recipient, or only the recipient received hydrogen. The results indicate that hydrogen inhalation at 2% concentration for 1 h prior to liver transplantation protected the rats from ischemia/reperfusion injury by activation of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Chao-Bin Zhang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yi-Chen Tang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Xue-Jun Xu
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Shi-Xiang Guo
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Huai-Zhi Wang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
32
|
Abstract
The current general interest in the use of food choice or diet in maintaining good health and in preventing and treating disease also applies to patients with IBD, who often follow poor or nutritionally challenging dietary plans. Unfortunately, dietary advice plays only a minor part in published guidelines for management of IBD, which sends a message that diet is not of great importance. However, a considerable evidence base supports a focused and serious attention to nutrition and diet in patients with IBD. In this Review, a step-wise approach in the evaluation and management of these patients is proposed. First, dietary intake and eating habits as well as current nutritional state should be documented, and corrective measures instituted. Secondly, dietary strategies as primary or adjunctive therapy for the reduction of inflammation and/or prevention of relapse of IBD should be seriously contemplated. Thirdly, use of diet to improve symptoms or lessen the effects of complications should be considered. Finally, dietary advice regarding disease prevention should be discussed when relevant. An increasing need exists for applying improved methodologies into establishing the value of current and new ways of using food choice as a therapeutic and preventive tool in IBD.
Collapse
|
33
|
Sanei MH, Hadizadeh F, Adibi P, Alavi SA. Inflammatory cells' role in acetic acid-induced colitis. Adv Biomed Res 2014; 3:193. [PMID: 25337523 PMCID: PMC4202508 DOI: 10.4103/2277-9175.140666] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Accepted: 07/25/2012] [Indexed: 02/07/2023] Open
Abstract
Background: Free radicals are the known mechanisms responsible for inducing colitis with two origins: Inflammatory cells and tissues. Only the inflammatory cells can be controlled by corticosteroids. Our aim was to assess the importance of neutrophils as one of the inflammatory cells in inducing colitis and to evaluate the efficacy of corticosteroids in the treatment of inflammatory bowel disease (IBD). Materials and Methods: Thirty-six mice were divided into six groups of six mice each. Colitis was induced in three groups by exposing them to acetic acid through enema (group 1), ex vivo (group 3), and enema after immune suppression (group 5). Each group had one control group that was exposed to water injection instead of acetic acid. Tissue samples were evaluated and compared based on macroscopic damages and biochemical and pathological results. Results: Considering neutrophilic infiltration, there were significant differences between groups 1, 3, 5, and the control of group 1. Groups 3, 5, and their controls, and group 1 and the control of group 3 had significant differences in terms of goblet depletion. Based on tissue originated H2O2, we found significant differences between group 1 and its control and group 3, and also between groups 5 and the control of group 3. All the three groups were significantly different from their controls based on Ferric Reducing Ability of Plasma (FRAP) and such differences were also seen between group 1 with two other groups. Conclusion: Neutrophils may not be the only cause of oxidation process in colitis, and also makes the effectiveness of corticosteroids in the treatment of this disease doubtful.
Collapse
Affiliation(s)
- Mohammad H Sanei
- Department of Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Hadizadeh
- Integrated Functional Gastroenterology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran ; East Sage Co, Isfahan Science and Technology Town, Isfahan, Iran
| | - Peyman Adibi
- Integrated Functional Gastroenterology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sayyed Ali Alavi
- East Sage Co, Isfahan Science and Technology Town, Isfahan, Iran
| |
Collapse
|
34
|
Liu W, Guo W, Guo L, Gu Y, Cai P, Xie N, Yang X, Shu Y, Wu X, Sun Y, Xu Q. Andrographolide sulfonate ameliorates experimental colitis in mice by inhibiting Th1/Th17 response. Int Immunopharmacol 2014; 20:337-45. [DOI: 10.1016/j.intimp.2014.03.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 03/11/2014] [Accepted: 03/20/2014] [Indexed: 01/24/2023]
|
35
|
Vermeiren J, Hindryckx P, Van Nieuwenhuyse G, Laukens D, De Vos M, Boon N, Van de Wiele T. Intrarectal nitric oxide administration prevents cellular infiltration but not colonic injury during dextran sodium sulfate colitis. Dig Dis Sci 2012; 57:1832-7. [PMID: 22419056 DOI: 10.1007/s10620-012-2105-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Accepted: 02/21/2012] [Indexed: 02/08/2023]
Abstract
BACKGROUND During inflammation in the gastrointestinal tract, the production of nitric oxide (NO) is mediated by the mucosal conversion of L-arginine. Recently, it was shown that the gut microbiota can also produce NO. AIMS The effect of gut luminal NO on inflammatory processes of an experimental colitis mice model was investigated by administrating NO directly to the colon, mimicking microbial NO production. METHODS Twenty-four mice received daily intrarectal treatment with a NO donor in 2 doses and 8 mice were treated with placebo. Starting 1 day later, 18 of these mice were fed ad libitum with 4% of dextran sodium sulfate (DSS) in their drinking water to induce colitis. At day 6, histopathology (both the inflammation and damage score), myeloperoxidase (MPO)-activity, colon length and colonic permeability were evaluated. RESULTS Co-administration of NO during DSS exposure inhibited the induction of an increasing colonic MPO-activity. This protective effect of NO was confirmed by the histological inflammation score showing a similar trend. The colonic permeability was restored when very low levels of NO were administered to the DSS-mice. On the other hand, the colon length of the NO-treated DSS-mice was negatively correlated with the NO dose and the histological damage score was not improved. CONCLUSIONS Our results indicate that intrarectal administration of NO has clear anti-inflammatory effects in experimental colitis, but does not prevent colonic damage. Therefore, NO-producing microorganisms in the gut lumen should be accounted as a modulating process during colitis.
Collapse
Affiliation(s)
- Joan Vermeiren
- Faculty of Bioscience Engineering, Laboratory of Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | | | | | | | | | | | | |
Collapse
|
36
|
Vermeiren J, Van de Wiele T, Van Nieuwenhuyse G, Boeckx P, Verstraete W, Boon N. Sulfide- and nitrite-dependent nitric oxide production in the intestinal tract. Microb Biotechnol 2011; 5:379-87. [PMID: 22129449 PMCID: PMC3821680 DOI: 10.1111/j.1751-7915.2011.00320.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In the gut ecosystem, nitric oxide (NO) has been described to have damaging effects on the energy metabolism of colonocytes. Described mechanisms of NO production are microbial reduction of nitrate via nitrite to NO and conversion of l‐arginine by NO synthase. The aim of this study was to investigate whether dietary compounds can stimulate the production of NO by representative cultures of the human intestinal microbiota and whether this correlates to other processes in the intestinal tract. We have found that the addition of a reduced sulfur compound, i.e. cysteine, contributed to NO formation. This increase was ascribed to higher sulfide concentrations generated from cysteine that in turn promoted the chemical conversion of nitrite to NO. The NO release from nitrite was of the order of 4‰ at most. Overall, it was shown that two independent biological processes contribute to the chemical formation of NO in the intestinal tract: (i) the production of sulfide by fermentation of sulfur containing amino acids or reduction of sulfate by sulfate reducing bacteria, and (ii) the reduction of nitrate to nitrite. Our results indicate that dietary thiol compounds in combination with nitrate may contribute to colonocytes damaging processes by promoting NO formation.
Collapse
Affiliation(s)
- Joan Vermeiren
- Laboratory of Microbial Ecology and Technology, Ghent University, Coupure Links 653, B-9000 Gent, Belgium
| | | | | | | | | | | |
Collapse
|
37
|
Högberg L, Webb C, Fälth-Magnusson K, Forslund T, Magnusson KE, Danielsson L, Ivarsson A, Sandström O, Sundqvist T. Children with screening-detected coeliac disease show increased levels of nitric oxide products in urine. Acta Paediatr 2011; 100:1023-7. [PMID: 21284717 DOI: 10.1111/j.1651-2227.2011.02186.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
AIM Increased concentration of nitric oxide (NO) metabolites, nitrite and nitrate, in the urine is a strong indication of ongoing small intestinal inflammation, which is a hallmark of the enteropathy of coeliac disease (CD). It has previously been shown that children with symptomatic, untreated CD have increased levels of NO oxidation products in their urine. The aim of this study was to investigate whether screening-detected, asymptomatic coeliac children display the same urinary nitrite/nitrate pattern. METHODS In a multicenter screening study, serum samples were collected from 7208 12-year-old children without previously diagnosed CD. Sera were analysed for anti-human tissue transglutaminase (tTG) of isotype IgA. Small bowel biopsy was performed in antibody-positive children, yielding 153 new cases of CD. In the screening-detected individuals, the sum of nitrite and nitrate concentrations in the urine was analysed and used as an indicator of NO production. For comparison, 73 children with untreated, symptomatic CD were studied. RESULTS The nitrite/nitrate levels in children with screening-detected CD and those with untreated symptomatic CD did not differ significantly. Both groups had significantly increased urinary nitrite/nitrate concentrations compared to the children with normal small bowel biopsy (p < 0.001). CONCLUSION Children with screening-detected CD have increased production of NO just as children with untreated symptomatic CD. High NO metabolite levels in the urine may indicate a pathogenetic feature of CD and be a marker of major clinical importance.
Collapse
Affiliation(s)
- L Högberg
- Pediatric Clinic, Norrköping Hospital, Norrköping, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Jantchou P, Morois S, Clavel-Chapelon F, Boutron-Ruault MC, Carbonnel F. Animal protein intake and risk of inflammatory bowel disease: The E3N prospective study. Am J Gastroenterol 2010; 105:2195-201. [PMID: 20461067 DOI: 10.1038/ajg.2010.192] [Citation(s) in RCA: 297] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Diet composition has long been suspected to contribute to inflammatory bowel disease (IBD), but has not been thoroughly assessed, and has been assessed only in retrospective studies that are prone to recall bias. The aim of the present study was to evaluate the role of dietary macronutrients in the etiology of IBD in a large prospective cohort. METHODS The Etude Épidémiologique des femmes de la Mutuelle Générale de l'Education Nationale cohort consists of women living in France, aged 40-65 years, and free of major diseases at inclusion. A self-administered questionnaire was used to record dietary habits at baseline. Questionnaires on disease occurrence and lifestyle factors were completed every 24 months. IBDs were assessed in each questionnaire until June 2005, and subsequently validated using clinical and pathological criteria. We estimated the association between nutrients or foods and IBD using Cox proportional hazards models adjusted for energy intake. RESULTS Among 67,581 participants (705,445 person-years, mean follow-up since completion of the baseline dietary questionnaire 10.4 years), we validated 77 incident IBD cases. High total protein intake, specifically animal protein, was associated with a significantly increased risk of IBD, (hazards ratio for the third vs. first tertile and 95% confidence interval being 3.31 and 1.41-7.77 (P trend=0.007), and 3.03 and 1.45-6.34 (P trend=0.005) for total and animal protein, respectively). Among sources of animal protein, high consumption of meat or fish but not of eggs or dairy products was associated with IBD risk. CONCLUSIONS High protein intake is associated with an increased risk of incident IBD in French middle-aged women.
Collapse
Affiliation(s)
- Prévost Jantchou
- INSERM, UMRS, Centre for Research in Epidemiology and Population Health, Institut Gustave Roussy, Université Paris Sud, Villejuif, France
| | | | | | | | | |
Collapse
|
39
|
Abstract
Inflammatory diseases of the intestine, including Crohn's disease, ulcerative colitis, and celiac disease are now very common in Australia and remain major challenges for clinicians. Australian (and New Zealand) clinicians and scientists have made considerable contributions to our current understanding of these diseases over the last 50 years, including pathogenesis (such as the 'butyrate hypothesis', 'endoplasmic reticulum (ER) stress', and the identification of the peptide sequences that incite celiac disease), true population epidemiology (albeit in New Zealand), precise clinical observation, new investigative tools, innovative new potential therapies, influential clinical drug trials (such as triple antibiotics for Crohn's disease), and a dietary approach with efficacy for functional gut symptoms (the low FODMAP diet). Underpinning the success has been clinical excellence and adaptation of clinicians to the changing landscape of disease severity and therapeutic options. The future is indeed bright if such trends continue.
Collapse
Affiliation(s)
- Peter R Gibson
- Monash University Department of Medicine, Box Hill Hospital, Box Hill, Victoria, Australia.
| |
Collapse
|
40
|
Packey CD, Sartor RB. Commensal bacteria, traditional and opportunistic pathogens, dysbiosis and bacterial killing in inflammatory bowel diseases. Curr Opin Infect Dis 2009; 22:292-301. [PMID: 19352175 DOI: 10.1097/qco.0b013e32832a8a5d] [Citation(s) in RCA: 249] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The authors present evidence published during the past 2 years of the roles of commensal and pathogenic bacteria in the pathogenesis of the inflammatory bowel diseases. RECENT FINDINGS Rodent models conclusively implicate commensal enteric bacteria in chronic, immune-mediated, experimental colitis, and genetically determined defects in bacterial killing by innate immune cells are found in a subset of patients with Crohn's disease. There is no evidence that a single pathogen, including Mycobacterium avium subspecies paratuberculosis, causes Crohn's disease or ulcerative colitis. However, adherent/invasive Escherichia coli are associated with ileal Crohn's disease, with the mechanisms and genetics of adherent/invasive E. coli virulence being elucidated. Molecular characterization of the microbiota in patients with inflammatory bowel diseases reveals decreased biodiversity of commensal bacteria, most notably the phyla Bacteroidetes and Firmicutes, including the clinically relevant Faecalibacterium prausnitzii, and increased E. coli concentrations. VSL#3 is one probiotic preparation shown to be efficacious in certain clinical situations in small clinical trials. SUMMARY Further characterization of altered microbiota in patients with inflammatory bowel diseases and linking dysbiosis with host genetic alterations in immunoregulation, innate microbial killing and barrier function are critical, so that individualized treatments to increase beneficial commensals and their metabolic products (probiotic and prebiotic administration) and diminish deleterious species such as adherent/invasive E. coli can be tailored for defined patient subsets.
Collapse
Affiliation(s)
- Christopher D Packey
- Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
41
|
Abstract
OBJECTIVE Hepcidin is an endogenous antimicrobial peptide with a key role in iron homoeostasis. Hepcidin is similar to defensin, the deficiency of which is associated with Crohn's disease. To date there has been no validated method to reliably assay serum hepcidin. We studied iron indices in inflammatory bowel disease (IBD) including hepcidin. DESIGN We assessed serum hepcidin concentrations (using a newly developed competitive radioimmunoassay) and ferritin in patients with IBD. Haematinics including serum soluble transferrin receptor, serum iron, serum vitamin B12 and red cell folate levels were also measured. The hepcidin results were compared with a control group of healthy volunteers from the local community. SETTING This study was based in a hospital. PATIENTS Sixty-one patients with IBD (51 patients with ulcerative colitis and 10 with Crohn's disease). Their mean hepcidin results were compared with 25 healthy controls. MAIN OUTCOME MEASURE hepcidin concentration in serum samples in IBD patients compared with normal volunteers. RESULTS We found significantly low serum hepcidin levels in patients with IBD. The hepcidin levels were low in IBD patients without iron deficiency anaemia as evidenced by normal ferritin and serum iron levels (n=41, mean hepcidin 6.81 ng/ml, SEM 1.2) and in IBD patients with iron deficiency anaemia (n=18, mean hepcidin 4.14 ng/ml, SEM 0.72) compared with healthy controls (n=25, mean hepcidin 15.3 ng/ml, SEM 3.14) (P=0.0045 and P=0.0050 on unpaired t-tests, respectively). We also measured IL-6 (enzyme-linked immunosorbent assay method, Abcam plc) in 21 of the 61 patients with IBD and compared the results with samples from 10 healthy volunteers. The IL-6 level was significantly higher (P=0.0222 on unpaired t-tests) in this group of IBD patients (n=21, IL-6 mean 2.94 pg/ml, SEM 0.64) compared with controls (n=10, IL-6 mean 0.663 pg/ml SEM 0.14). A significant positive correlation (Pearson's correlation coefficient r=0.6331) was present between hepcidin and IL-6, but not between hepcidin and serum soluble transferrin receptor (r=-0.235). CONCLUSION The low hepcidin results in IBD patients may reflect a causal or perpetuator effect on intestinal inflammation.
Collapse
|