51
|
Sanekommu H, Taj S, Mah Noor R, Umair Akmal M, Akhtar R, Hossain M, Asif A. Probiotics and Fecal Transplant: An Intervention in Delaying Chronic Kidney Disease Progression? Clin Pract 2023; 13:881-888. [PMID: 37623261 PMCID: PMC10453439 DOI: 10.3390/clinpract13040080] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/01/2023] [Accepted: 07/09/2023] [Indexed: 08/26/2023] Open
Abstract
Chronic kidney disease (CKD) is a global health challenge affecting nearly 700 million people worldwide. In the United States alone, the Medicare costs for CKD management has reached nearly USD 80 billion per year. While reversing CKD may be possible in the future, current strategies aim to slow its progression. For the most part, current management strategies have focused on employing Renin Angiotensin Aldosterone (RAS) inhibitors and optimizing blood pressure and diabetes mellitus control. Emerging data are showing that a disruption of the gut-kidney axis has a significant impact on delaying CKD progression. Recent investigations have documented promising results in using microbiota-based interventions to better manage CKD. This review will summarize the current evidence and explore future possibilities on the use of probiotics, prebiotics, synbiotics, and fecal microbial transplant to reduce CKD progression.
Collapse
Affiliation(s)
- Harshavardhan Sanekommu
- Department of Medicine, Jersey Shore University Medical Center, 1945 NJ-33, Neptune City, NJ 07753, USA; (S.T.)
| | - Sobaan Taj
- Department of Medicine, Jersey Shore University Medical Center, 1945 NJ-33, Neptune City, NJ 07753, USA; (S.T.)
| | - Rida Mah Noor
- School of Medicine, Eastern Campus, International University of Kyrgyzstan-International, Bishkek 720007, Kyrgyzstan;
| | | | - Reza Akhtar
- Department of Gastroenterology, Jersey Shore University Medical Center, 1945 NJ-33, Neptune City, NJ 07753, USA
| | - Mohammad Hossain
- Department of Medicine, Jersey Shore University Medical Center, 1945 NJ-33, Neptune City, NJ 07753, USA; (S.T.)
| | - Arif Asif
- Department of Medicine, Jersey Shore University Medical Center, 1945 NJ-33, Neptune City, NJ 07753, USA; (S.T.)
| |
Collapse
|
52
|
Li W, Jia Y, Gong Z, Dong Z, Yu F, Fu Y, Jiang C, Kong W. Ablation of the gut microbiota alleviates high-methionine diet-induced hyperhomocysteinemia and glucose intolerance in mice. NPJ Sci Food 2023; 7:36. [PMID: 37460578 DOI: 10.1038/s41538-023-00212-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 07/12/2023] [Indexed: 07/20/2023] Open
Abstract
A high-methionine (HM) diet leads to hyperhomocysteinemia (HHcy), while gastrointestinal tissue is an important site of net homocysteine (Hcy) production. However, the role of the gut microbiota in host HHcy remains obscure. This study aimed to determine whether gut microbiota ablation could alleviate host HHcy and glucose intolerance and reveal the underlying mechanism. The results showed that the HM diet-induced HHcy and glucose intolerance in mice, while antibiotic administration decreased the plasma level of Hcy and reversed glucose intolerance. HM diet increased intestinal epithelial homocysteine levels, while antibiotic treatment decreased intestinal epithelial homocysteine levels under the HM diet. Gut microbiota depletion had no effect on the gene expression and enzyme activity of CBS and BHMT in the livers of HM diet-fed mice. The HM diet altered the composition of the gut microbiota with marked increases in the abundances of Faecalibaculum and Dubosiella, which were also positively correlated with plasma Hcy concentrations. An in-depth analysis of the bacterial cysteine and methionine metabolism pathways showed that the abundances of two homocysteine biosynthesis-related KEGG orthologies (KOs) were markedly increased in the gut microbiota in HM diet-fed mice. Hcy was detected from Dubosiella newyorkensis-cultured supernatant by liquid chromatography-tandem mass spectrometry (LC‒MS) analysis. In conclusion, these findings suggested that the HM diet-induced HHcy and glucose intolerance in mice, by reshaping the composition of the gut microbiota, which might produce and secrete Hcy.
Collapse
Affiliation(s)
- Wenqiang Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yiting Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhao Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Third Hospital, Peking University, Beijing, China.
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| |
Collapse
|
53
|
Hajjar R, Gonzalez E, Fragoso G, Oliero M, Alaoui AA, Calvé A, Vennin Rendos H, Djediai S, Cuisiniere T, Laplante P, Gerkins C, Ajayi AS, Diop K, Taleb N, Thérien S, Schampaert F, Alratrout H, Dagbert F, Loungnarath R, Sebajang H, Schwenter F, Wassef R, Ratelle R, Debroux E, Cailhier JF, Routy B, Annabi B, Brereton NJB, Richard C, Santos MM. Gut microbiota influence anastomotic healing in colorectal cancer surgery through modulation of mucosal proinflammatory cytokines. Gut 2023; 72:1143-1154. [PMID: 36585238 DOI: 10.1136/gutjnl-2022-328389] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/08/2022] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Colorectal cancer (CRC) is the third most diagnosed cancer, and requires surgical resection and reconnection, or anastomosis, of the remaining bowel to re-establish intestinal continuity. Anastomotic leak (AL) is a major complication that increases mortality and cancer recurrence. Our objective is to assess the causal role of gut microbiota in anastomotic healing. DESIGN The causal role of gut microbiota was assessed in a murine AL model receiving faecal microbiota transplantation (FMT) from patients with CRC collected before surgery and who later developed or not, AL. Anastomotic healing and gut barrier integrity were assessed after surgery. Bacterial candidates implicated in anastomotic healing were identified using 16S rRNA gene sequencing and were isolated from faecal samples to be tested both in vitro and in vivo. RESULTS Mice receiving FMT from patients that developed AL displayed poor anastomotic healing. Profiling of gut microbiota of patients and mice after FMT revealed correlations between healing parameters and the relative abundance of Alistipes onderdonkii and Parabacteroides goldsteinii. Oral supplementation with A. onderdonkii resulted in a higher rate of leaks in mice, while gavage with P. goldsteinii improved healing by exerting an anti-inflammatory effect. Patients with AL and mice receiving FMT from AL patients presented upregulation of mucosal MIP-1α, MIP-2, MCP-1 and IL-17A/F before surgery. Retrospective analysis revealed that patients with AL present higher circulating neutrophil and monocyte counts before surgery. CONCLUSION Gut microbiota plays an important role in surgical colonic healing in patients with CRC. The impact of these findings may extend to a vast array of invasive gastrointestinal procedures.
Collapse
Affiliation(s)
- Roy Hajjar
- Nutrition and Microbiome Laboratory, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Department of Surgery, Université de Montréal, Montréal, Québec, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Emmanuel Gonzalez
- Canadian Centre for Computational Genomics, McGill Genome Centre, Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montréal, Québec, Canada
| | - Gabriela Fragoso
- Nutrition and Microbiome Laboratory, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Manon Oliero
- Nutrition and Microbiome Laboratory, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Ahmed Amine Alaoui
- Nutrition and Microbiome Laboratory, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Department of Surgery, Université de Montréal, Montréal, Québec, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Annie Calvé
- Nutrition and Microbiome Laboratory, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Hervé Vennin Rendos
- Nutrition and Microbiome Laboratory, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Souad Djediai
- Molecular Oncology Laboratory, Department of Chemistry, Université du Québec à Montréal (UQAM), Montréal, Québec, Canada
| | - Thibault Cuisiniere
- Nutrition and Microbiome Laboratory, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Patrick Laplante
- Institut du cancer de Montréal, Montréal, Québec, Canada
- Axe Cancer, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Claire Gerkins
- Nutrition and Microbiome Laboratory, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Ayodeji Samuel Ajayi
- Nutrition and Microbiome Laboratory, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Khoudia Diop
- Institut du cancer de Montréal, Montréal, Québec, Canada
- Laboratory of Immunotherapy and Oncomicrobiome, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Nassima Taleb
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Sophie Thérien
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Frédéricke Schampaert
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Hefzi Alratrout
- (Current address: Department of General Surgery, King Fahd Hospital of the University, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia). Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - François Dagbert
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Rasmy Loungnarath
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Herawaty Sebajang
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Frank Schwenter
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Ramses Wassef
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Richard Ratelle
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Eric Debroux
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Jean-François Cailhier
- Institut du cancer de Montréal, Montréal, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
- Renal Division, Department of Medicine, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Bertrand Routy
- Institut du cancer de Montréal, Montréal, Québec, Canada
- Laboratory of Immunotherapy and Oncomicrobiome, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Hemato-oncology Division, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Borhane Annabi
- Molecular Oncology Laboratory, Department of Chemistry, Université du Québec à Montréal (UQAM), Montréal, Québec, Canada
| | - Nicholas J B Brereton
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
- Institut de Recherche en Biologie Végétale, Université de Montréal, Montréal, Québec, Canada
| | - Carole Richard
- Digestive Surgery Service, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Manuela M Santos
- Nutrition and Microbiome Laboratory, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du cancer de Montréal, Montréal, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
54
|
Sun J, Fang D, Wang Z, Liu Y. Sleep Deprivation and Gut Microbiota Dysbiosis: Current Understandings and Implications. Int J Mol Sci 2023; 24:ijms24119603. [PMID: 37298553 DOI: 10.3390/ijms24119603] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Gut microbiota comprises the microbial communities inhabiting our gastrointestinal (GI) tracts. Accordingly, these complex communities play a fundamental role in many host processes and are closely implicated in human health and diseases. Sleep deprivation (SD) has become increasingly common in modern society, partly owing to the rising pressure of work and the diversification of entertainment. It is well documented that sleep loss is a significant cause of various adverse outcomes on human health including immune-related and metabolic diseases. Furthermore, accumulating evidence suggests that gut microbiota dysbiosis is associated with these SD-induced human diseases. In this review, we summarize the gut microbiota dysbiosis caused by SD and the succedent diseases ranging from the immune system and metabolic system to various organs and highlight the critical roles of gut microbiota in these diseases. The implications and possible strategies to alleviate SD-related human diseases are also provided.
Collapse
Affiliation(s)
- Jingyi Sun
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Dan Fang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zhiqiang Wang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China
| | - Yuan Liu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
55
|
Wang T, Tian J, Su W, Yang F, Yin J, Jiang Q, Li Y, Yao K, Li T, Yin Y. Effect of Ornithine α-Ketoglutarate on Intestinal Microbiota and Serum Inflammatory Cytokines in Dextran Sulfate Sodium Induced Colitis. Nutrients 2023; 15:nu15112476. [PMID: 37299439 DOI: 10.3390/nu15112476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Ornithine α-ketoglutarate (OKG), a nutritional compound, is an amino acid salt with anti-oxidative and anti-inflammatory effects on humans and animals. Ulcerative colitis (UC), as an inflammatory bowel disease (IBD), leads to chronic intestinal inflammatory dysfunction. This study evaluated the optimal dosage of OKG in healthy mice. Then, a mouse model of acute colitis was established using dextran sodium sulfate (DSS), and the preventive effect of OKG on DSS-induced colitis in mice was explored through analysis of serum inflammatory cytokines and fecal microbiota. Initially, the mice were randomly divided into a control group, a group given a low dose of OKG (LOKG: 0.5%), a group given a medium dose of OKG (MOKG: 1%), and a group given a high dose of OKG (HOKG: 1.5%); they remained in these groups for the entire 14-day experimental period. Our results demonstrated that 1% OKG supplementation increased body weight, serum growth hormone (GH), insulin (INS), alkaline phosphatase (ALP), Tyr, and His and decreased urea nitrogen (BUN), NH3L, and Ile. Then, a 2 × 2 factor design was used for a total of 40 mice, with diet (a standard diet or a 1% OKG diet) and challenge (4% DSS or not) as the main factors. During days 14 to 21, the DSS mice were administered 4% DSS to induce colitis. The results revealed that OKG alleviated weight loss and reversed the increases in colonic histological damage induced by DSS. OKG also increased serum IL-10 secretion. Moreover, OKG enhanced the abundance of Firmicutes and decreased that of Bacteriodetes at the phylum level and particularly enhanced the abundance of Alistipes and reduced that of Parabacterioides at the genus level. Our results indicated that OKG promotes growth performance and hormone secretion and regulates serum biochemical indicators and amino acid concentrations. Furthermore, 1% OKG supplementation prevents DSS-induced colitis in mice via altering microbial compositions and reducing the secretion of inflammatory cytokines in serum.
Collapse
Affiliation(s)
- Tao Wang
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
| | - Junquan Tian
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
| | - Wenxuan Su
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
| | - Fan Yang
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410125, China
| | - Qian Jiang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410125, China
| | - Yuying Li
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, China
| | - Kang Yao
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
| | - Tiejun Li
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410125, China
| |
Collapse
|
56
|
Venneri T, Giorgini G, Leblanc N, Flamand N, Borrelli F, Silvestri C, Di Marzo V. Altered endocannabinoidome bioactive lipid levels accompany reduced DNBS-induced colonic inflammation in germ-free mice. Lipids Health Dis 2023; 22:63. [PMID: 37189092 DOI: 10.1186/s12944-023-01823-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/24/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Gut microbiota are involved in the onset and development of chronic intestinal inflammation. The recently described endocannabinoidome (eCBome), a diverse and complex system of bioactive lipid mediators, has been reported to play a role in various physio-pathological processes such as inflammation, immune responses and energy metabolism. The eCBome and the gut microbiome (miBIome) are closely linked and form the eCBome - miBIome axis, which may be of special relevance to colitis. METHODS Colitis was induced in conventionally raised (CR), antibiotic-treated (ABX) and germ-free (GF) mice with dinitrobenzene sulfonic acid (DNBS). Inflammation was assessed by Disease Activity Index (DAI) score, body weight change, colon weight-length ratio, myeloperoxidase (MPO) activity and cytokine gene expression. Colonic eCBome lipid mediator concentrations were measured by HPLC-MS /MS. RESULTS GF mice showed increased levels of anti-inflammatory eCBome lipids (LEA, OEA, DHEA and 13- HODE-EA) in the healthy state and higher MPO activity. DNBS elicited reduced inflammation in GF mice, having lower colon weight/length ratios and lower expression levels of Il1b, Il6, Tnfa and neutrophil markers compared to one or both of the other DNBS-treated groups. Il10 expression was also lower and the levels of several N-acyl ethanolamines and 13-HODE-EA levels were higher in DNBS-treated GF mice than in CR and ABX mice. The levels of these eCBome lipids negatively correlated with measures of colitis and inflammation. CONCLUSIONS These results suggest that the depletion of the gut microbiota and subsequent differential development of the gut immune system in GF mice is followed by a compensatory effect on eCBome lipid mediators, which may explain, in part, the observed lower susceptibility of GF mice to develop DNBS-induced colitis.
Collapse
Affiliation(s)
- Tommaso Venneri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Giada Giorgini
- Joint International Research Unit (JIRU) for Chemical and Biomolecular Research on the Microbiome and its impact on Metabolic Health and Nutrition (MicroMeNu) between Université Laval and the Consiglio Nazionale delle Ricerche (CNR), Institute of Biomolecular Chemistry, Pozzuoli, NA, Italy
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Département de médecine, Faculté de Médecine, Université Laval, Québec, Canada
| | - Nadine Leblanc
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Département de médecine, Faculté de Médecine, Université Laval, Québec, Canada
| | - Nicolas Flamand
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Département de médecine, Faculté de Médecine, Université Laval, Québec, Canada
| | - Francesca Borrelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Cristoforo Silvestri
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Département de médecine, Faculté de Médecine, Université Laval, Québec, Canada.
- Centre NUTRISS, École de nutrition, Faculté des sciences de l'agriculture et de l'alimentation (FSAA), Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec, Canada.
| | - Vincenzo Di Marzo
- Joint International Research Unit (JIRU) for Chemical and Biomolecular Research on the Microbiome and its impact on Metabolic Health and Nutrition (MicroMeNu) between Université Laval and the Consiglio Nazionale delle Ricerche (CNR), Institute of Biomolecular Chemistry, Pozzuoli, NA, Italy.
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Département de médecine, Faculté de Médecine, Université Laval, Québec, Canada.
- Centre NUTRISS, École de nutrition, Faculté des sciences de l'agriculture et de l'alimentation (FSAA), Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec, Canada.
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, Canada.
| |
Collapse
|
57
|
He Q, Guo K, Wang L, Xie F, Zhao Q, Jiang X, He Z, Wang P, Li S, Huang Y, Zhang C, Huang R, Liu Y, Wang F, Zhou X, Niu R, Zuo T, Wang Y, Li C. Tannins amount determines whether tannase-containing bacteria are probiotic or pathogenic in IBD. Life Sci Alliance 2023; 6:e202201702. [PMID: 36759174 PMCID: PMC9911794 DOI: 10.26508/lsa.202201702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/01/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
The role of dietary tannin in inflammatory bowel disease (IBD) is still not clear. Therefore, we aim to study the effect of TA in the progression of IBD. Dextran sulphate sodium (DSS)-induced model was used to mimic IBD. Metagenomics and metabolomics were performed to study the alteration of intestinal microbiota and metabolites. NCM460 and THP-1 cells were used for in vitro study. The amount of TA was associated with the outcomes of DSS-induced IBD as evidenced by in vivo and in vitro studies. Metabolomic and metagenomic analyses revealed that TA-induced enrichment of microbial metabolite gallic acid (GA) was responsible for the action of TA. Mechanistically, protective dose of GA promoted colonic mucus secretion to suppress bacterial infection and that it ameliorated DSS-induced epithelial damage by inhibiting p53 signaling, whereas toxic dose of GA directly caused epithelial damage by promoting cell cycle arrest. Therapeutic experiment showed protective dose of GA-promoted recovery of DSS-induced colonic inflammation. The role of tannase-containing bacteria can be transformed under different conditions in IBD progression.
Collapse
Affiliation(s)
- Qiuyue He
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Kenan Guo
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Lulu Wang
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Fei Xie
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Qingyuan Zhao
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Xianhong Jiang
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Zhongming He
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Peng Wang
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Shiqiang Li
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Yan Huang
- Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Cong Zhang
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Rongjuan Huang
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Yang Liu
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Fengchao Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Xiaoyang Zhou
- Department of Biological Safety, Army Medical University, Chongqing, China
| | - Rong Niu
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Tao Zuo
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yong Wang
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| | - Chuangen Li
- Department of Laboratory Animal Science, Army Medical University, Chongqing, China
| |
Collapse
|
58
|
Yang XJ, Wang XH, Yang MY, Ren HY, Chen H, Zhang XY, Liu QF, Yang G, Yang Y, Yang XJ. Exploring choices of early nutritional support for patients with sepsis based on changes in intestinal microecology. World J Gastroenterol 2023; 29:2034-2049. [PMID: 37155528 PMCID: PMC10122787 DOI: 10.3748/wjg.v29.i13.2034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/21/2023] [Accepted: 03/20/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Sepsis exacerbates intestinal microecological disorders leading to poor prognosis. Proper modalities of nutritional support can improve nutrition, immunity, and intestinal microecology. AIM To identify the optimal modality of early nutritional support for patients with sepsis from the perspective of intestinal microecology. METHODS Thirty patients with sepsis admitted to the intensive care unit of the General Hospital of Ningxia Medical University, China, between 2019 and 2021 with indications for nutritional support, were randomly assigned to one of three different modalities of nutritional support for a total of 5 d: Total enteral nutrition (TEN group), total parenteral nutrition (TPN group), and supplemental parenteral nutrition (SPN group). Blood and stool specimens were collected before and after nutritional support, and changes in gut microbiota, short-chain fatty acids (SCFAs), and immune and nutritional indicators were detected and compared among the three groups. RESULTS In comparison with before nutritional support, the three groups after nutritional support presented: (1) Differences in the gut bacteria (Enterococcus increased in the TEN group, Campylobacter decreased in the TPN group, and Dialister decreased in the SPN group; all P < 0.05); (2) different trends in SCFAs (the TEN group showed improvement except for Caproic acid, the TPN group showed improvement only for acetic and propionic acid, and the SPN group showed a decreasing trend); (3) significant improvement of the nutritional and immunological indicators in the TEN and SPN groups, while only immunoglobulin G improved in the TPN group (all P < 0.05); and (4) a significant correlation was found between the gut bacteria, SCFAs, and nutritional and immunological indicators (all P < 0.05). CONCLUSION TEN is recommended as the preferred mode of early nutritional support in sepsis based on clinical nutritional and immunological indicators, as well as changes in intestinal microecology.
Collapse
Affiliation(s)
- Xiao-Juan Yang
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Xiao-Hong Wang
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ming-Yue Yang
- Department of Emergency Medicine, Affiliated Hospital of Jining Medical University, Jining 272030, Shandong Province, China
| | - Hong-Yan Ren
- Shanghai Mobio Biomedical Technology Co., Ltd., Shanghai 201318, China
| | - Hui Chen
- Shanghai Mobio Biomedical Technology Co., Ltd., Shanghai 201318, China
| | - Xiao-Ya Zhang
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Qin-Fu Liu
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ge Yang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Yang
- Department of Critical Care Medicine, Southeast University School of Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Xiao-Jun Yang
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
59
|
Lv R, Sun N, Mao C, Zheng Z, Lin S. Prevention and potential repair of colitis: Beneficial effects and regulatory mechanisms of food-derived anti-inflammatory peptides. Crit Rev Food Sci Nutr 2023; 64:8184-8202. [PMID: 37017113 DOI: 10.1080/10408398.2023.2197068] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
Intestinal inflammatory diseases are increasingly prevalent worldwide, and their pathogenesis is still not fully understood. As of late, studies have discovered that food-derived peptides have specific anti-inflammatory activity and can play a positive role in intestinal health. At the same time, it has broad application prospects in the prevention and treatment of colitis because of its wide source, fast absorption, and high safety. This article reviews the structure-activity and quantity-effect relationships of food-derived peptides for their anti-inflammatory effects. It then discusses their mechanism of action in inhibiting colitis from four aspects. Food-derived anti-inflammatory peptides can delay the progression of the disease by stimulating innate immunity, inhibiting inflammation, and promoting wound healing. Further experiments showed that food-derived anti-inflammatory peptides could prevent and treat colitis through four mechanisms: (a) regulation of inflammatory cytokines; (b) regulation of inflammatory pathways; (c) regulation of intestinal epithelial barrier; (d) regulation of intestinal flora balance. However, due to the treatment of colitis having limitations, there is an urgent to develop food-derived anti-inflammatory peptides as a treatment or adjunctive treatment for colitis. This review highlights the positive effects of food-derived peptides on colitis and anticipates the appearance of mitigating peptides for the therapy of colitis.
Collapse
Affiliation(s)
- Renzhi Lv
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Na Sun
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Chuwen Mao
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Zhihong Zheng
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Songyi Lin
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| |
Collapse
|
60
|
Yee SM, Choi H, Seon JE, Ban YJ, Kim MJ, Seo JE, Seo JH, Kim S, Moon SH, Yun CH, Lee HB, Kang HS. Axl alleviates DSS-induced colitis by preventing dysbiosis of gut microbiota. Sci Rep 2023; 13:5371. [PMID: 37005456 PMCID: PMC10067963 DOI: 10.1038/s41598-023-32527-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/29/2023] [Indexed: 04/04/2023] Open
Abstract
Axl is a tyrosine kinase receptor, a negative regulator for innate immune responses and inflammatory bowel disease (IBD). The gut microbiota regulates intestinal immune homeostasis, but the role of Axl in the pathogenesis of IBD through the regulation of gut microbiota composition remains unresolved. In this study, mice with DSS-induced colitis showed increased Axl expression, which was almost entirely suppressed by depleting the gut microbiota with antibiotics. Axl-/- mice without DSS administration exhibited increased bacterial loads, especially the Proteobacteria abundant in patients with IBD, significantly consistent with DSS-induced colitis mice. Axl-/- mice also had an inflammatory intestinal microenvironment with reduced antimicrobial peptides and overexpression of inflammatory cytokines. The onset of DSS-induced colitis occurred faster with an abnormal expansion of Proteobacteria in Axl-/- mice than in WT mice. These findings suggest that a lack of Axl signaling exacerbates colitis by inducing aberrant compositions of the gut microbiota in conjunction with an inflammatory gut microenvironment. In conclusion, the data demonstrated that Axl signaling could ameliorate the pathogenesis of colitis by preventing dysbiosis of gut microbiota. Therefore, Axl may act as a potential novel biomarker for IBD and can be a potential candidate for the prophylactic or therapeutic target of diverse microbiota dysbiosis-related diseases.
Collapse
Affiliation(s)
- Su-Min Yee
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Harim Choi
- Department of Nursing, Nambu University, 23 Chumdan Jungang-Ro, Gwangsan-Gu, Gwangju, 62271, Republic of Korea
| | - Jeong-Eun Seon
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Yu-Jin Ban
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Min-Jae Kim
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Jae-Eun Seo
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Ja Hun Seo
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Sehyeon Kim
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Seo Hee Moon
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Chul-Ho Yun
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Hyang Burm Lee
- Environmental Microbiology Lab, Department of Agricultural Biological Chemistry, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea
| | - Hyung-Sik Kang
- School of Biological Sciences and Technology, Chonnam National University, 77 Yongbong-Ro, Buk-Gu, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
61
|
Zhong P, Wu H, Ma Y, Xu X, Jiang Y, Jin C, Zhu Q, Liu X, Suo Z, Wang J. P2X4 receptor modulates gut inflammation and favours microbial homeostasis in colitis. Clin Transl Med 2023; 13:e1227. [PMID: 37085966 PMCID: PMC10122071 DOI: 10.1002/ctm2.1227] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 03/02/2023] [Accepted: 03/08/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a non-specific chronic inflammatory disease of the intestine. In addition to genetic susceptibility, environmental factors and dysregulated host immunity, the gut microbiota is implicated in the pathogenesis of Crohn's disease (CD) or ulcerative colitis (UC), the two primary types of IBD. The P2X4 receptor has been demonstrated to have a crucial role in preventing infection, inflammation, and organ damage. However, it remains unclear whether the P2X4 receptor affects IBD and the underlying mechanisms. METHODS Colitis was induced in mice administrated with dextran sodium sulphate (DSS). 16S rDNA sequencing was used to analyze the gut microbiota in knockout and wild-type mice. Clinical and histopathological parameters were monitored throughout the disease progression. RESULTS Gene Expression Omnibus analysis showed the downregulation of P2RX4 (P2rx4) expression in colonic tissues from patients or mice with IBD. However, its expression at the protein levels was upregulated on day 4 or 6 and then downregulated on day 7 in C57BL/6 mice treated with DSS. Gene ablation of P2rx4 aggravated DSS-induced colitis accompanying gut microbiota dysbiosis in mice. Moreover, P2X4 receptor-positive modulator ivermectin alleviated colitis and corrected dysregulated microbiota in wild-type C57BL/6 mice. Further antibiotic-treated gut microbiota depletion, cohousing experiment, and fecal microbiota transplantation proved that gut microbiota dysbiosis was associated with the aggravation of colitis in the mouse model initiated by P2rx4. CONCLUSIONS Our findings elaborate on an unrevealed etiopathophysiological mechanism by which microbiota dysbiosis induced by the P2X4 receptor influences the development of colitis, indicating that the P2X4 receptor represents a promising target for treating patients with CD and UC.
Collapse
Affiliation(s)
- Peijie Zhong
- Infection and Immunity Institute and Translational Medical Center, Huaihe HospitalHenan UniversityKaifengChina
| | - Hang Wu
- Infection and Immunity Institute and Translational Medical Center, Huaihe HospitalHenan UniversityKaifengChina
| | - Yuanqiao Ma
- Infection and Immunity Institute and Translational Medical Center, Huaihe HospitalHenan UniversityKaifengChina
| | - Xiaoxiao Xu
- Infection and Immunity Institute and Translational Medical Center, Huaihe HospitalHenan UniversityKaifengChina
| | - Yizhuo Jiang
- Infection and Immunity Institute and Translational Medical Center, Huaihe HospitalHenan UniversityKaifengChina
| | - Chaolei Jin
- Infection and Immunity Institute and Translational Medical Center, Huaihe HospitalHenan UniversityKaifengChina
| | - Qiaozhen Zhu
- Infection and Immunity Institute and Translational Medical Center, Huaihe HospitalHenan UniversityKaifengChina
| | - Xinlei Liu
- Infection and Immunity Institute and Translational Medical Center, Huaihe HospitalHenan UniversityKaifengChina
| | - Zhimin Suo
- Infection and Immunity Institute and Translational Medical Center, Huaihe HospitalHenan UniversityKaifengChina
| | - Junpeng Wang
- Infection and Immunity Institute and Translational Medical Center, Huaihe HospitalHenan UniversityKaifengChina
| |
Collapse
|
62
|
Raftery AL, O’Brien CA, Harris NL, Tsantikos E, Hibbs ML. Development of severe colitis is associated with lung inflammation and pathology. Front Immunol 2023; 14:1125260. [PMID: 37063825 PMCID: PMC10102339 DOI: 10.3389/fimmu.2023.1125260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
Inflammatory bowel diseases (IBD) such as Crohn’s disease and ulcerative colitis are chronic relapsing diseases that affect the gastrointestinal tract, most commonly the colon. A link between the gut and the lung is suggested since patients with IBD have an increased susceptibility for chronic inflammatory lung disease. Furthermore, in the absence of overt lung disease, IBD patients have worsened lung function and more leukocytes in sputum than healthy individuals, highlighting a conduit between the gut and lung in disease. To study the gut-lung axis in the context of IBD, we used TCRδ-/- mice, which are highly susceptible to dextran sulfate sodium (DSS) due to the importance of γδ T cells in maintenance of barrier integrity. After induction of experimental colitis using DSS, the lungs of TCRδ-/- mice exhibited signs of inflammation and mild emphysema, which was not observed in DSS-treated C57BL/6 mice. Damage to the lung tissue was accompanied by a large expansion of neutrophils in the lung parenchyma and an increase in alveolar macrophages in the lung wash. Gene expression analyses showed a significant increase in Csf3, Cxcl2, Tnfa, and Il17a in lung tissue in keeping with neutrophil infiltration. Expression of genes encoding reactive oxygen species enzymes and elastolytic enzymes were enhanced in the lungs of both C57BL/6 and TCRδ-/- mice with colitis. Similarly, surfactant gene expression was also enhanced, which may represent a protective mechanism. These data demonstrate that severe colitis in a susceptible genetic background is sufficient to induce lung inflammation and tissue damage, providing the research community with an important tool for the development of novel therapeutics aimed at reducing co-morbidities in IBD patients.
Collapse
|
63
|
Wang S, Wen Q, Qin Y, Xia Q, Shen C, Song S. Gut microbiota and host cytochrome P450 characteristics in the pseudo germ-free model: co-contributors to a diverse metabolic landscape. Gut Pathog 2023; 15:15. [PMID: 36945019 PMCID: PMC10029254 DOI: 10.1186/s13099-023-00540-5] [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: 11/04/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND The pseudo germ-free (PGF) model has been widely used to research the role of intestinal microbiota in drug metabolism and efficacy, while the modelling methods and the utilization of the PGF model are still not standardized and unified. A comprehensive and systematic research of the PGF model on the composition and function of the intestinal microbiota, changes in host cytochrome P450 (CYP450) enzymes expression and intestinal mucosal permeability in four different modelling cycles of the PGF groups are provided in this paper. RESULTS 16S rRNA gene amplicon sequencing was employed to compare and analyze the alpha and beta diversity, taxonomic composition, taxonomic indicators and predicted function of gut microbiota in the control and PGF groups. Bacterial richness and diversity decreased significantly in the PGF group beginning after the first week of establishment of the PGF model with antibiotic exposure. The PGF group exposed to antibiotics for 4-week-modelling possessed the fewest indicator genera. Moreover, increased intestinal mucosal permeability occurred in the second week of PGF model establishment, indicating that one week of antibiotic exposure is an appropriate time to establish the PGF model. The results of immunoblots revealed that CYP1A2, CYP2C19 and CYP2E1 expression was significantly upregulated in the PGF group compared with the control group, implying that the metabolic clearance of related drugs would change accordingly. The abundance of functional pathways predicted in the gut microbiota changed dramatically between the control and PGF groups. CONCLUSIONS This study provides information concerning the microbial and CYP450 enzyme expression profiles as a reference for evaluating drug metabolism differences co-affected by gut microbiota and host CYP450 enzymes in the PGF model.
Collapse
Affiliation(s)
- Shanshan Wang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Qiuyu Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Yan Qin
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Quan Xia
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Chenlin Shen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China.
| | - Shuai Song
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, People's Republic of China.
| |
Collapse
|
64
|
Volkmann ER. Is there a role for the microbiome in systemic sclerosis? Expert Rev Clin Immunol 2023; 19:237-240. [PMID: 36576305 PMCID: PMC9957839 DOI: 10.1080/1744666x.2023.2161512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Affiliation(s)
- Elizabeth R. Volkmann
- Division of Rheumatology, Department of Medicine, University of California, Los Angeles, David Geffen School of Medicine
| |
Collapse
|
65
|
Hutchinson NT, Wang SS, Dokhanchi J, Johnson RW, Buford TW, Allen JM, Woods JA. Effects of Broad-Spectrum Antibiotic Treatment or Germ-Free Status on Endurance Performance and Exercise Adaptations in Mice. Med Sci Sports Exerc 2023; 55:225-234. [PMID: 36170555 PMCID: PMC9840680 DOI: 10.1249/mss.0000000000003051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Endurance exercise alters the gut microbiome independently of diet. The extent to which gut microbes are responsible for physiologic adaptations to exercise training is unknown. The purpose of these experiments was to determine the role of gut microbes in performance and muscle adaptation to 6 wk of voluntary wheel running (VWR) in mice. METHODS We depleted microbes with broad-spectrum antibiotic (ABX) treatment and used germ-free (GF) mice to determine effects on adaptations to VWR. Male and female C57Bl/6 mice ( n = 56) were assigned to daily VWR or sedentary conditions. After the intervention, treadmill endurance and glucose tolerance were assessed, and gastrocnemius and soleus tissues were harvested and analyzed for citrate synthase (CS) enzyme activity and expression of exercise training-sensitive genes. RESULTS ABX treatment and GF status resulted in VWR volumes ~22% and 26% lower than controls, respectively. Analysis of variance revealed that, although VWR increased treadmill endurance, ABX had no effect. GF status significantly reduced treadmill performance in trained GF mice after training. VWR increased gastrocnemius CS enzyme activity in all groups, and ABX and GF status did not reduce the VWR effect. VWR also increased muscle expression of PGC1a, but this was not affected by ABX treatment. CONCLUSIONS ABX treatment and GF status reduced VWR behavior but did not affect VWR-induced adaptations in endurance capacity, CS activity, or expression of muscle metabolic genes. However, GF status reduced endurance capacity. These data indicated that reducing microbes in adulthood does not inhibit endurance training adaptations in C57Bl/6 mice, but that GF mice possess a reduced responsiveness to endurance exercise training, perhaps because of a developmental defect associated with lack of microbes from birth.
Collapse
Affiliation(s)
- Noah T. Hutchinson
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL
| | - Selena S. Wang
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL
| | - Jack Dokhanchi
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL
| | - Rodney W. Johnson
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL
| | - Thomas W. Buford
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Birmingham/Atlanta Geriatric Research, Education, and Clinical Center, Birmingham VA Medical Center, Birmingham, AL
| | - Jacob M. Allen
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL
| | - Jeffrey A. Woods
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL
| |
Collapse
|
66
|
Zhao LP, Wu J, Quan W, Zhou Y, Hong H, Niu GY, Li T, Huang SB, Qiao CM, Zhao WJ, Cui C, Shen YQ. DSS-induced colitis activates the kynurenine pathway in serum and brain by affecting IDO-1 and gut microbiota. Front Immunol 2023; 13:1089200. [PMID: 36776388 PMCID: PMC9908955 DOI: 10.3389/fimmu.2022.1089200] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/28/2022] [Indexed: 01/27/2023] Open
Abstract
Accumulative studies suggest that inflammatory bowel disease (IBD) may cause multiple central nervous system (CNS) pathologies. Studies have found that indoleamine-2,3-dioxygenase (IDO, rate-limiting enzyme of the kynurenine (Kyn) pathway) deficient mice were protected from endotoxin induced cognitive impairment, and Kyn administration induced cognitive memory deficits in both control and IDO-deficient mice. However, there is no investigation of the brain Kyn pathway in IBD, thus we investigated whether dextran sulfate sodium (DSS)-induced colitis could cause dysregulation of Kyn pathway in brain, and also in serum. C57BL/6J mice were given drinking water with 2% DSS for 10 consecutive days to induce colitis. In serum, we found significant increase in Kyn and kynurenic acid (Kyna) level, which was regulated by IDO-1 and KAT2 (rate-limiting enzymes of Trp-Kyn-Kyna pathway). Similarly, by analyzing GEO datasets, higher IDO-1 levels in peripheral blood monocytes and colon of UC patients was found. Furthermore, the Kyn pathway was significantly upregulated in the cerebral cortex under the action of IDO-1 after DSS treatment, which ultimately induced the neurotoxic phenotype of astrocytes. To investigate whether gut microbiota is involved in IBD-induced Kyn pathway dysregulation, we performed intestinal flora 16S rRNA sequencing and found that DSS-induced colitis significantly altered the composition and diversity of the gut microbiota. Metabolic function analysis also showed that Tryptophan metabolism, NOD-like receptor signaling pathway and MAPK signaling pathway were significantly up-regulated in the 2% DSS group. A significant association between intestinal flora and Trp metabolism (both in serum and brain) was found by correlation analysis. Overall, this study revealed that DSS-induced colitis causes dysregulation of the Kyn pathway in serum and brain by affecting rate-limiting enzymes and intestinal flora.
Collapse
|
67
|
Li L, Qiu N, Meng Y, Wang C, Mine Y, Keast R, Guyonnet V. Preserved egg white alleviates DSS-induced colitis in mice through the reduction of oxidative stress, modulation of infl ammatory cytokines, NF-κB, MAPK and gut microbiota composition. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
68
|
Antibiotic-resistant bacteria originating from the gut may modulate the mucosal immune response during sepsis and septic shock. Drug Target Insights 2022; 16:81-87. [PMID: 36755640 PMCID: PMC9886009 DOI: 10.33393/dti.2022.2520] [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: 11/16/2022] [Accepted: 12/30/2022] [Indexed: 12/31/2022] Open
Abstract
The enrichment and diversity of gut microbiota play an important role in sepsis, but the role of gut microbiota composition and early-life colonization in sepsis and septic shock has not yet been characterized. The impact of gut microbiota diversity on host immunological disorders and future treatments of inflammatory diseases are not yet fully elucidated. Further, the association between the microbiota and immune development in sepsis remains unknown, and the underlying mechanisms are not well understood. The altered composition of gut microbiota during sepsis is profoundly associated with a loss of commensal bacteria and an overgrowth of potentially pathogenic bacteria, especially AMR bacteria. Disruptions of gut microbiota diversity are directly associated with susceptibility to sepsis and a higher risk of adverse outcomes. Several studies have confirmed that a mutual association between gut microbiota and the host is important for the metabolism of essential nutrients for the organism, for gut development, and for the maturation and development of a fully functional immune system. Therefore, understanding the gut microbiota diversity, composition, and function during various inflammatory conditions and sepsis may provide a comprehensive knowledge of the mechanisms behind the pathogenesis of gut-derived infection in diseases and the design of new treatment options (e.g., probiotics or fecal microbiota transplantation). Emerging evidence displays an important role of gut microbiota and their derived metabolites in modulating the host mucosal immune response and determining the susceptibility to, as well as outcomes of sepsis.
Collapse
|
69
|
Vaghari-Tabari M, Moein S, Alipourian A, Qujeq D, Malakoti F, Alemi F, Yousefi B, Khazaie S. Melatonin and inflammatory bowel disease: From basic mechanisms to clinical application. Biochimie 2022; 209:20-36. [PMID: 36535545 DOI: 10.1016/j.biochi.2022.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 10/25/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Inflammatory bowel disease is a chronic inflammatory disease and has periods of recurrence and remission. Improper immune responses to gut flora bacteria, along with genetic susceptibility, appear to be involved in causing this complex disease. It seems dysbiosis and oxidative stress may also be involved in IBD pathogenesis. A significant number of clinical studies have shown an interesting association between sleep disturbances and IBD. Studies in animal models have also shown that sleep deprivation has a significant effect on the pathogenesis of IBD and can aggravate inflammation. These interesting findings have drawn attention to melatonin, a sleep-related hormone. Melatonin is mainly produced by the pineal gland, but many tissues in the body, including the intestines, can produce it. Melatonin can have an interesting effect on the pathogenesis of IBD. Melatonin can enhance the intestinal mucosal barrier, alter the composition of intestinal bacteria in favor of bacteria with anti-inflammatory properties, regulate the immune response, alleviate inflammation and attenuate oxidative stress. It seems that, melatonin supplementation is effective in relieving inflammation and healing intestinal ulcers in IBD animal models. Some clinical studies have also shown that melatonin supplementation as an adjuvant therapy may be helpful in reducing disease activity in IBD patients. In this review article, in addition to reviewing the effects of sleep disturbances and melatonin on key mechanisms involved in the pathogenesis of IBD, we will review the findings of clinical studies regarding the effects of melatonin supplementation on IBD treatment.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Moein
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Alipourian
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Faezeh Malakoti
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Forough Alemi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Sepideh Khazaie
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
70
|
Hu Y, Chen Z, Xu C, Kan S, Chen D. Disturbances of the Gut Microbiota and Microbiota-Derived Metabolites in Inflammatory Bowel Disease. Nutrients 2022; 14:5140. [PMID: 36501169 PMCID: PMC9735443 DOI: 10.3390/nu14235140] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/10/2022] Open
Abstract
Inflammatory bowel disease (IBD), comprising Crohn's disease (CD) and ulcerative colitis (UC), is characterized as a chronic and recurrent inflammatory disease whose pathogenesis is still elusive. The gut microbiota exerts important and diverse effects on host physiology through maintaining immune balance and generating health-benefiting metabolites. Many studies have demonstrated that IBD is associated with disturbances in the composition and function of the gut microbiota. Both the abundance and diversity of gut microbiota are dramatically decreased in IBD patients. Furthermore, some particular classes of microbiota-derived metabolites, principally short-chain fatty acids, tryptophan, and its metabolites, and bile acids have also been implicated in the pathogenesis of IBD. In this review, we aim to define the disturbance of gut microbiota and the key classes of microbiota-derived metabolites in IBD pathogenesis. In addition, we also focus on scientific evidence on probiotics, not only on the molecular mechanisms underlying the beneficial effects of probiotics on IBD but also the challenges it faces in safe and appropriate application.
Collapse
Affiliation(s)
- Yongjia Hu
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Zhouzhou Chen
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Chengchen Xu
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Shidong Kan
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Daijie Chen
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| |
Collapse
|
71
|
Yan C, Kwek E, Ding HF, He Z, Ma KY, Zhu H, Chen ZY. Dietary Oxidized Cholesterol Aggravates Chemically Induced Murine Colon Inflammation and Alters Gut Microbial Ecology. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:13289-13301. [PMID: 36198042 DOI: 10.1021/acs.jafc.2c05001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Western diet with a higher intake of fat and cholesterol has been claimed as an intestinal inflammation trigger. Human diet contains both cholesterol and oxidized cholesterol. Oxidized cholesterol has been claimed to be associated with various inflammation diseases, but its effects on colitis and gut microbiome remain largely unknown. The present study was the first time to investigate the effect of the oxidized cholesterol on gut microbiota and dextran sodium sulfate-induced colitis using mice as a model. The results showed that oxidized cholesterol promoted colitis by exacerbating bleeding, body weight decrease, colon shortening, gut barrier damage, oxidative stress, and gut inflammation, whereas non-oxidized cholesterol had no effect. Meanwhile, oxidized cholesterol could adversely modulate the gut microbiota by increasing the relative abundance of pro-inflammatory bacteria (including Escherichia-Shigella and Bacteroides) and decreasing that of beneficial bacteria (Lachnospiraceae_NK4A136_group and Odoribacter). In addition, oxidized cholesterol significantly reduced the production of fecal short-chain fatty acids in colitis mice. It was concluded that oxidized cholesterol was a potential dietary factor of gut dysbiosis.
Collapse
Affiliation(s)
- Chi Yan
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
| | - Erika Kwek
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
| | - Hua-Fang Ding
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
| | - Zouyan He
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
- School of Public Health, Guanxi Medical University, Nanning 530021, China
| | - Ka Ying Ma
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
| | - Hanyue Zhu
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
- School of Food Science and Engineering/South China Food Safety Research Center, Foshan University, Foshan 528000, Guangdong, China
| | - Zhen-Yu Chen
- Food and Nutritional Sciences Program, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong 999077, China
| |
Collapse
|
72
|
Effect of DSS-Induced Ulcerative Colitis and Butyrate on the Cytochrome P450 2A5: Contribution of the Microbiome. Int J Mol Sci 2022; 23:ijms231911627. [PMID: 36232929 PMCID: PMC9569822 DOI: 10.3390/ijms231911627] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 12/02/2022] Open
Abstract
Several studies have indicated the beneficial anti-inflammatory effect of butyrate in inflammatory bowel disease (IBD) therapy implying attempts to increase butyrate production in the gut through orally administered dietary supplementation. Through the gut-liver axis, however, butyrate may reach directly the liver and influence the drug-metabolizing ability of hepatic enzymes, and, indirectly, also the outcome of applied pharmacotherapy. The focus of our study was on the liver microsomal cytochrome P450 (CYP) 2A5, which is a mouse orthologue of human CYP2A6 responsible for metabolism of metronidazole, an antibiotic used to treat IBD. Our findings revealed that specific pathogen-free (SPF) and germ-free (GF) mice with dextran sulfate sodium (DSS)-induced colitis varied markedly in enzyme activity of CYP2A and responded differently to butyrate pre-treatment. A significant decrease (to 50%) of the CYP2A activity was observed in SPF mice with colitis; however, an administration of butyrate prior to DSS reversed this inhibition effect. This phenomenon was not observed in GF mice. The results highlight an important role of gut microbiota in the regulation of CYP2A under inflammatory conditions. Due to the role of CYP2A in metronidazole metabolism, this phenomenon may have an impact on the IBD therapy. Butyrate administration, hence, brings promising therapeutic potential for improving symptoms of gut inflammation; however, possible interactions with drug metabolism need to be further studied.
Collapse
|
73
|
Zhang J, Liang F, Chen Z, Chen Y, Yuan J, Xiong Q, Hou S, Huang S, Liu C, Liang J. Vitexin Protects against Dextran Sodium Sulfate-Induced Colitis in Mice and Its Potential Mechanisms. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:12041-12054. [PMID: 36124900 DOI: 10.1021/acs.jafc.2c05177] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Vitexin, one of the major active components in hawthorn, has been shown to possess multiple pharmacological activities. Here, we sought to investigate the effect of vitexin on an ameliorating dextran sodium sulfate (DSS)-induced ulcerative colitis (UC) mouse model and further explored its potential mechanism. The results indicated that vitexin administration could significantly alleviate the signs of colitis via suppressing body weight loss, reducing disease activity index (DAI) score, and mitigating colonic damage. Also, vitexin treatment in colitis mice markedly inhibited the production of pro-inflammation cytokines (such as IL-1β, IL-6, and TNF-α). Meanwhile, vitexin also could markedly down-regulate the phosphorylation levels of p65, IκB, and STAT1. Moreover, vitexin also dose-dependently increased the expressions of muc-2, ZO-1, and occludin proteins in colonic tissues of colitis mice. Further studies revealed that vitexin dramatically modulated the disturbed intestinal flora in colitis mice. Vitexin is beneficial for regulating abundances of some certain bacteria, such as Bacteroides, Helicobacter, Alistipes, Lachnospiraceae_NK4A136_group, and Lachnospiraceae_UCG-006. Interestingly, the correlation analysis indicated that key microbes were strongly correlated with colitis features, such as pro-inflammatory cytokines and gut barrier. Collectively, these results demonstrated that vitexin treatment alleviated inflammation, intestinal barrier dysfunction, and intestinal flora dysbiosis in colitis mice. Vitexin is expected to be a promising compound for UC treatment.
Collapse
Affiliation(s)
- Jing Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Feilin Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Zongwen Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
- Gaozhou Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Chinese Medicine, Gaozhou, Guangdong 510006, China
| | - Yonger Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Jun Yuan
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Qingping Xiong
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Shaozhen Hou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Song Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
- Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, Guangdong 510006, China
| | - Changhui Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Jian Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, China
- Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, Guangdong 510006, China
| |
Collapse
|
74
|
Zhang B, Zhang Y, Liu X, Yin J, Li X, Zhang X, Xing X, Wang J, Wang S. Differential Protective Effect of Resveratrol and Its Microbial Metabolites on Intestinal Barrier Dysfunction is Mediated by the AMPK Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:11301-11313. [PMID: 36066018 DOI: 10.1021/acs.jafc.2c04101] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The effectiveness of resveratrol (RES) on intestinal barrier dysfunction and colitis has been extensively studied. However, the specific effects of its microbial metabolites on gut barrier function remain unclear. Hence, we compared the protective effects of RES and its microbial metabolites dihydroresveratrol (DHR) and 3-(4-hydroxyphenyl)-propionic acid (4HPP) against intestinal barrier injury and colitis. Only 4HPP and RES significantly reduced paracellular permeability and the secretion of proinflammatory cytokines in lipopolysaccharides (LPS)-treated intestinal Caco-2 cells, which was consistent with the upregulation in tight junction (TJ) proteins. Furthermore, RES and 4HPP ameliorated intestinal barrier dysfunction and colonic inflammation in colitis mice, while DHR did not. In particular, the expressions of intestinal TJ proteins and Muc2 were restored by RES and 4HPP. The molecular mechanism involved the adenosine monophosphate-activated protein kinase (AMPK)-mediated activation of CDX2 and the regulation of the SIRT1/NF-κB pathway. These findings provide new insights into understanding the protective effects of RES against intestinal barrier damage and colitis.
Collapse
Affiliation(s)
- Bowei Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yunhui Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiaoxia Liu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jia Yin
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiang Li
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Xuejiao Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiaolong Xing
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jin Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
75
|
Seki N, Kimizuka T, Gondo M, Yamaguchi G, Sugiura Y, Akiyama M, Yakabe K, Uchiyama J, Higashi S, Haneda T, Suematsu M, Hase K, Kim YG. D-tryptophan suppresses enteric pathogens and pathobionts and prevents colitis by modulating microbial tryptophan metabolism. iScience 2022; 25:104838. [PMID: 35996581 PMCID: PMC9391578 DOI: 10.1016/j.isci.2022.104838] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/17/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022] Open
Abstract
D-Amino acids (D-AAs) have various functions in mammals and microbes. D-AAs are produced by gut microbiota and can act as potent bactericidal molecules. Thus, D-AAs regulate the ecological niche of the intestine; however, the actual impacts of D-AAs in the gut remain unknown. In this study, we show that D-Tryptophan (D-Trp) inhibits the growth of enteric pathogen and colitogenic pathobionts. The growth of Citrobacter rodentium in vitro is strongly inhibited by D-Trp treatment. Moreover, D-Trp protects mice from lethal C. rodentium infection via reduction of the pathogen. Additionally, D-Trp prevents the development of experimental colitis by the depletion of specific microbes in the intestine. D-Trp increases the intracellular level of indole acrylic acid (IA), a key molecule that determines the susceptibility of enteric microbes to D-Trp. Treatment with IA improves the survival of mice infected with C. rodentium. Hence, D-Trp could act as a gut environmental modulator that regulates intestinal homeostasis. D-Trp inhibits the growth of Citrobacter rodentium in vitro and in vivo D-Trp suppresses experimental colitis by the depletion of specific gut microbes IA is the metabolite that determines the susceptibility of enteric microbes to D-Trp
Collapse
Affiliation(s)
- Natsumi Seki
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
- Department of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
| | - Tatsuki Kimizuka
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
- Department of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
| | - Monica Gondo
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
- Department of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
| | - Genki Yamaguchi
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
- Department of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masahiro Akiyama
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
| | - Kyosuke Yakabe
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
- Department of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
| | - Jun Uchiyama
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
- Department of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
| | - Seiichiro Higashi
- Co-Creation Center, Meiji Holdings Co., Ltd., 1-29-1 Nanakuni, Hachiouji, Tokyo 192-0919, Japan
| | - Takeshi Haneda
- Laboratory of Microbiology, School of Pharmacy, Kitasato University, Tokyo 108-8641, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Koji Hase
- Department of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
| | - Yun-Gi Kim
- Research Center for Drug Discovery, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
- Corresponding author
| |
Collapse
|
76
|
Li L, Wang M, Bao J, Wang N, Huang Y, He S, Chen B, Yan F. Periodontitis may impair the homeostasis of systemic bone through regulation of gut microbiota in
ApoE
‐/‐
mice. J Clin Periodontol 2022; 49:1304-1319. [PMID: 35871602 DOI: 10.1111/jcpe.13708] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/01/2022] [Accepted: 07/21/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Lili Li
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Min Wang
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Jun Bao
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Nannan Wang
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Yuezhen Huang
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Shasha He
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Bin Chen
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Fuhua Yan
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| |
Collapse
|
77
|
Sex-Biased Immune Responses to Antibiotics during Anti-PD-L1 Treatment in Mice with Colon Cancer. J Immunol Res 2022; 2022:9202491. [PMID: 35903754 PMCID: PMC9325566 DOI: 10.1155/2022/9202491] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 11/17/2022] Open
Abstract
Colitis is a frequently occurred side effect of immune checkpoint inhibitors (ICIs), which are increasingly used in cancer treatment, whereas antibiotics are widely used to treat colitis, their effectiveness in ICI-associated colitis remains controversial. In this study, we firstly assessed the effectiveness of several commonly used antibiotics and antibiotic cocktails in alleviating of dextran sulfate sodium- (DSS-) induced colitis. We observed that two narrow-spectrum antibiotics, neomycin and metronidazole, were more effective in alleviating colitis, as evidenced by the remission of loss of the body weight, enlargement of the spleen, shortening of the colon, secretion of proinflammatory cytokines, and histological score of the colon tissue. Moreover, these two antibiotics resulted in better relief of colitis symptoms in the MC38 tumor-bearing male mice receiving the anti-PD-L1 mAb (αPD-L1) treatment, compared to the females. In the meantime, an enhanced response to αPD-L1 efficiency against mice colon cancer was observed in the male mouse group upon the application of these two antibiotics. In contrast, both neomycin and metronidazole showed destructive effects on the antitumor efficiency of αPD-L1 in female mice, despite relief from colitis. We found that antibiotic treatment attenuated the increased infiltration of granulocytes and myeloid cells in colon tissue induced by DSS in female mice, while reducing the proportion of Th17 cells in male mice. These differences were further associated with the sex-biased differences in the gut microbiota. These findings indicated that sex-dependent alterations in the gut microbiota should be considered when applying antibiotics for the treatment of ICI-associated colitis.
Collapse
|
78
|
Li H, Li J, Xiao T, Hu Y, Yang Y, Gu X, Jin G, Cao H, Zhou H, Yang C. Nintedanib Alleviates Experimental Colitis by Inhibiting CEBPB/PCK1 and CEBPB/EFNA1 Pathways. Front Pharmacol 2022; 13:904420. [PMID: 35910380 PMCID: PMC9331303 DOI: 10.3389/fphar.2022.904420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/13/2022] [Indexed: 11/22/2022] Open
Abstract
The super-enhancer, a cluster of enhancers with strong transcriptional activity, has become one of the most interesting topics in recent years. This study aimed to investigate pathogenic super-enhancer–driven genes in IBD and screen therapeutic drugs based on the results. In this study, through the analysis of differentially expressed genes in colitis patients from the GEO database and the analysis of the super-enhancer–associated database, we found that the super-enhancer pathogenic genes PCK1 and EFNA1 were simultaneously regulated by transcription factor CEBPB through two super-enhancers (sc-CHR20-57528535 and sc-CHR1-155093980). Silencing CEBPB could significantly inhibit the expression of PCK1 and EFNA1 and enhance the expression of epithelial barrier proteins claudin-1, occludin, and ZO-1. In LPS-induced Caco-2 cells, drugs commonly used in clinical colitis including tofacitinib, oxalazine, mesalazine, and sulfasalazine inhibited mRNA levels of CEBPB, PCK1, and EFNA1. In the drug screening, we found that nintedanib significantly inhibited the mRNA and protein levels of CEBPB, PCK1, and EFNA1. In vivo experiments, nintedanib significantly alleviated DSS-induced colitis in mice by inhibiting CEBPB/PCK1 and CEBPB/EFNA1 signaling pathways. At the genus level, nintedanib improved the composition of the gut microbiota in mice with DSS-induced experimental colitis. In conclusion, we found that PCK1 and EFNA1 are highly expressed in colitis and they are regulated by CEBPB through two super-enhancers, and we further demonstrate their role in vivo and in vitro. Nintedanib may be a potential treatment for IBD. Super-enhancers may be a new way to explore the pathogenesis of colitis.
Collapse
Affiliation(s)
- Hailong Li
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Jinhe Li
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
- High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Ting Xiao
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Yayue Hu
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
- High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Ying Yang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Xiaoting Gu
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Ge Jin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
- *Correspondence: Hailong Cao, ; Honggang Zhou, ; Cheng Yang,
| | - Honggang Zhou
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
- High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin, China
- *Correspondence: Hailong Cao, ; Honggang Zhou, ; Cheng Yang,
| | - Cheng Yang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
- High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin, China
- *Correspondence: Hailong Cao, ; Honggang Zhou, ; Cheng Yang,
| |
Collapse
|
79
|
Zhang Y, Si X, Yang L, Wang H, Sun Y, Liu N. Association between intestinal microbiota and inflammatory bowel disease. Animal Model Exp Med 2022; 5:311-322. [PMID: 35808814 PMCID: PMC9434590 DOI: 10.1002/ame2.12255] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/21/2022] [Indexed: 12/15/2022] Open
Abstract
Inflammatory bowel disease (IBD), which includes Crohn's disease (CD) and ulcerative colitis (UC), has emerged as a global disease with high incidence, long duration, devastating clinical symptoms, and low curability (relapsing immune response and barrier function defects). Mounting studies have been performed to investigate its pathogenesis to provide an ever‐expanding arsenal of therapeutic options, while the precise etiology of IBD is not completely understood yet. Recent advances in high‐throughput sequencing methods and animal models have provided new insights into the association between intestinal microbiota and IBD. In general, dysbiosis characterized by an imbalanced microbiota has been widely recognized as a pathology of IBD. However, intestinal microbiota alterations represent the cause or result of IBD process remains unclear. Therefore, more evidences are needed to identify the precise role of intestinal microbiota in the pathogenesis of IBD. Herein, this review aims to outline the current knowledge of commonly used, chemically induced, and infectious mouse models, gut microbiota alteration and how it contributes to IBD, and dysregulated metabolite production links to IBD pathogenesis.
Collapse
Affiliation(s)
- Yunchang Zhang
- Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xuemeng Si
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Ling Yang
- Department of Food and Bioengineering, Beijing Vocational College of Agriculture, Beijing, China
| | - Hui Wang
- Department of Food and Bioengineering, Beijing Vocational College of Agriculture, Beijing, China
| | - Ye Sun
- Institute of Medical Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing, China
| | - Ning Liu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
80
|
Chen C, Liang H, Wang J, Ren G, Li R, Cui ZG, Zhang C. Heterophyllin B an Active Cyclopeptide Alleviates Dextran Sulfate Sodium-induced Colitis by Modulating Gut Microbiota and Repairing Intestinal Mucosal Barrier via AMPK Activation. Mol Nutr Food Res 2022; 66:e2101169. [PMID: 35796402 DOI: 10.1002/mnfr.202101169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 05/12/2022] [Indexed: 12/07/2022]
Abstract
SCOPE Advances in pathology broaden our perception of the intimate interaction between gut microbiota dysbiosis and the pathogenesis of ulcerative colitis (UC), but the potential modulating roles remain to be elucidated. METHODS AND RESULTS DSS-induced colitis was used to investigate the effect of Heterophyllin B (HB), a typical active cyclopeptide extracted from Pseudostellaria heterophylla, on colitis and gut microbiota. Administration of HB substantially mitigated the symptoms of UC as evidenced by increasing body weight and colon length, as well as decreased macrophages infiltration in the colon. Meanwhile, HB significantly alleviated intestinal mucosal barrier dysfunction by reducing the production of inflammatory cytokines, while all the mentioned beneficial effects were significantly eliminated by co-treatment with compound C, a selective AMPK inhibitor. In addition, 16S rDNA gene analyses and fecal microbiota transplantation also revealed that HB dramatically prevented against UC by reshaping intestinal dysbiosis, especially elevated the relative abundance of Akkermansia muciniphila. CONCLUSION These findings illustrated that HB prominently improved intestinal epithelial homeostasis via activating AMPK and ameliorated the colonic inflammation in a gut microbiota-dependent manner, which provide evidence for microbial contribution to UC pathogenesis and suggesting a novel approach for colitis prevention. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ce Chen
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China.,Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Han Liang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Jiaoyang Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Guoqing Ren
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Renshi Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China.,Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Zheng-Guo Cui
- Department of Environmental Health, University of Fukui School of Medical Science, 23-3 Matsuoka Shimoaizuki, Eiheiji, Fukui, 910-1193, Japan
| | - Chaofeng Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China.,Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
81
|
Novel Gut Microbiota Modulator, Which Markedly Increases Akkermansia muciniphila Occupancy, Ameliorates Experimental Colitis in Rats. Dig Dis Sci 2022; 67:2899-2911. [PMID: 34259969 DOI: 10.1007/s10620-021-07131-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/21/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Since gut microbiota is involved in the pathogenesis of inflammatory bowel disease (IBD), antibiotics or probiotics may be attractive options for the treatment of IBD. Akkermansia muciniphila is expected as a next-generation probiotic for IBD, and OPS-2071 is a novel quinolone with potent antibacterial activity against Clostridioides difficile. AIMS The aim of this study is to assess the potential of OPS-2071 as a gut microbiota modulator for IBD. METHODS Minimum inhibitory concentrations of several bacteria in the human intestinal microbiota were determined. Microbiota changes in the feces were typed using metagenomic analysis after oral administration of OPS-2071 (100 mg/kg) twice a day to normal rats. The amounts of mucin were determined using the Fecal Mucin Assay Kit. The effects of OPS-2071 (1, 3, 10 mg/kg) twice a day on fecal symptoms and fecal microbiota were evaluated in a colitis rat model induced by free access to drinking water containing 3% dextran sulfate sodium for 10 days. RESULTS OPS-2071 showed notably low antibacterial activity against only A. muciniphila in spite of higher antimicrobial activity against other strains of intestinal bacteria. OPS-2071 rapidly and dramatically increased the occupancy of A. muciniphila as well as the amount of mucin in the feces of normal rats. OPS-2071 (10 mg/kg) significantly suppressed the exacerbation of stool scores, especially the bloody stool score, with the increase in A. muciniphila occupancy. CONCLUSIONS OPS-2071 is expected to be a new therapeutic option for IBD as a gut microbiota modulator by significantly increasing A. muciniphila occupancy.
Collapse
|
82
|
Tao W, An X, Guo Z, Yang N, Wu M, Oliveira H, Zhang R, He J. Structural characterization, acute toxicity assessment and protective effects of selenylated apple pectin on dextran sulfate sodium-induced ulcerative colitis. Food Funct 2022; 13:7320-7332. [PMID: 35726791 DOI: 10.1039/d1fo04189d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study was aimed at investigating the structural characterization, acute toxicity and protective effect of selenylated apple pectin on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in mice. Selenylated apple pectin was characterized by ion chromatography, NMR and SEC-RI-MALLS. The acute toxicity and protective effect of selenylated apple pectin against UC were investigated by gavage administration in mice. The organ state and coefficients, inflammatory cytokine (IL-6, IL-10 and TNF-α) contents in serum, GSH-Px activity and MPO content in colon tissues were also evaluated. The results indicated that selenylated apple pectin was non-toxic and contained 244.28 μgselenium per g. The monosaccharide composition with different molar ratios, different relative molecular weights and a weakened signal peak (CH2-O group) at 3-4 ppm were observed after selenylation. The selenylated apple pectin showed the protective effect against UC by down-regulating IL-6 and TNF-α contents and up-regulating the IL-10 content in serum, as well as increasing the GSH-Px activity and decreasing the MPO content in colon tissues. Moreover, DSS-induced alterations were effectively recovered by a high-dose sample. These findings provide evidence in support of selenylated apple pectin as a novel dietary selenium supplement for UC protection.
Collapse
Affiliation(s)
- Wen Tao
- National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, No. 36 Huanhu Middle Road, Jinyinhu District, Wuhan 430023, P. R. China.
| | - Xiaoyu An
- National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, No. 36 Huanhu Middle Road, Jinyinhu District, Wuhan 430023, P. R. China.
| | - Ziqi Guo
- National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, No. 36 Huanhu Middle Road, Jinyinhu District, Wuhan 430023, P. R. China.
| | - Ning Yang
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, No. 36 Huanhu Middle Road, Jinyinhu District, Wuhan 430023, P. R. China.
| | - Muci Wu
- National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, No. 36 Huanhu Middle Road, Jinyinhu District, Wuhan 430023, P. R. China. .,Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, No. 36 Huanhu Middle Road, Jinyinhu District, Wuhan 430023, P. R. China.
| | - Hélder Oliveira
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Rui Zhang
- National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, No. 36 Huanhu Middle Road, Jinyinhu District, Wuhan 430023, P. R. China. .,Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, No. 36 Huanhu Middle Road, Jinyinhu District, Wuhan 430023, P. R. China.
| | - Jingren He
- National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, No. 36 Huanhu Middle Road, Jinyinhu District, Wuhan 430023, P. R. China. .,Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, No. 36 Huanhu Middle Road, Jinyinhu District, Wuhan 430023, P. R. China.
| |
Collapse
|
83
|
Rashed R, Valcheva R, Dieleman LA. Manipulation of Gut Microbiota as a Key Target for Crohn's Disease. Front Med (Lausanne) 2022; 9:887044. [PMID: 35783604 PMCID: PMC9244564 DOI: 10.3389/fmed.2022.887044] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Crohn's disease (CD) is an inflammatory bowel disease (IBD) sub-type characterized by transmural chronic inflammation of the gastrointestinal tract. Research indicates a complex CD etiology involving genetic predisposition and immune dysregulation in response to environmental triggers. The chronic mucosal inflammation has been associated with a dysregulated state, or dysbiosis, of the gut microbiome (bacteria), mycobiome (fungi), virome (bacteriophages and viruses), and archeaome (archaea) further affecting the interkingdom syntrophic relationships and host metabolism. Microbiota dysbiosis in CD is largely described by an increase in facultative anaerobic pathobionts at the expense of strict anaerobic Firmicutes, such as Faecalibacterium prausnitzii. In the mycobiome, reduced fungal diversity and fungal-bacteria interactions, along with a significantly increased abundance of Candida spp. and a decrease in Saccharomyces cerevisiae are well documented. Virome analysis also indicates a significant decrease in phage diversity, but an overall increase in phages infecting bacterial groups associated with intestinal inflammation. Finally, an increase in methanogenic archaea such as Methanosphaera stadtmanae exhibits high immunogenic potential and is associated with CD etiology. Common anti-inflammatory medications used in CD management (amino-salicylates, immunomodulators, and biologics) could also directly or indirectly affect the gut microbiome in CD. Other medications often used concomitantly in IBD, such as antibiotics, antidepressants, oral contraceptives, opioids, and proton pump inhibitors, have shown to alter the gut microbiota and account for increased susceptibility to disease onset or worsening of disease progression. In contrast, some environmental modifications through alternative therapies including fecal microbiota transplant (FMT), diet and dietary supplements with prebiotics, probiotics, and synbiotics have shown potential protective effects by reversing microbiota dysbiosis or by directly promoting beneficial microbes, together with minimal long-term adverse effects. In this review, we discuss the different approaches to modulating the global consortium of bacteria, fungi, viruses, and archaea in patients with CD through therapies that include antibiotics, probiotics, prebiotics, synbiotics, personalized diets, and FMT. We hope to provide evidence to encourage clinicians and researchers to incorporate these therapies into CD treatment options, along with making them aware of the limitations of these therapies, and indicate where more research is needed.
Collapse
|
84
|
Zhu Y, Xu Y, Wang X, Rao L, Yan X, Gao R, Shen T, Zhou Y, Kong C, Zhou L. Probiotic Cocktail Alleviates Intestinal Inflammation Through Improving Gut Microbiota and Metabolites in Colitis Mice. Front Cell Infect Microbiol 2022; 12:886061. [PMID: 35782138 PMCID: PMC9240319 DOI: 10.3389/fcimb.2022.886061] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/16/2022] [Indexed: 12/17/2022] Open
Abstract
The modulation of the gut microbiome has been widely suggested as a promising therapeutic strategy for inflammatory bowel disease (IBD). Here, we established a novel probiotic cocktail to investigate its therapeutic role in acute colitis mice. During dextran sulfate sodium (DSS)-induced colitis, the mice were treated with the probiotic cocktail, fecal microbiota transplantation (FMT) from a healthy mice donor, or 5-aminosalicylic acid (5-ASA), respectively. The inflammatory responses were assessed by symptoms, serum inflammatory factors, and histological scoring. The intestinal barrier function was assessed by detecting tight junction proteins. Gut microbiota and its metabolites were further identified using 16S rDNA sequencing and a liquid chromatograph mass spectrometer (LC-MS/MS). Compared with FMT and 5-ASA treatment, the probiotic cocktail performed better in alleviating symptoms of colitis and decreasing disease activity score and mucosal inflammation. The probiotic cocktail also significantly decreased serum IL-17 level and increased JAM-1 expression in colon. The gut microbiota analysis confirmed that the beneficial effects of the probiotic cocktail were attributed to increasing anti-inflammatory bacteria Akkermansia, Bifidobacterium, and Blautia, while decreasing pro-inflammatory bacteria Parasutterella. The targeted metabolome analysis further indicated a rise in the production of Bifidobacterium-related short-chain fatty acids (SCFAs) such as propanoic acid and isobutyric acid after probiotics treatment. Taken together, the probiotic cocktail effectively alleviated intestinal inflammation through improving gut microbiota and metabolites in colitis mice, suggesting its great potential to be a novel therapeutic approach for IBD patients.
Collapse
Affiliation(s)
- Yefei Zhu
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
- Department of General Surgery, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Medicine, Nantong University, Nantong, China
| | - Yu Xu
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
- School of Medicine, Nantong University, Nantong, China
| | - Xinyue Wang
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
| | - Leiping Rao
- Department of General Surgery, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xuebing Yan
- Department of Oncology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Renyuan Gao
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
| | - Tongyi Shen
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
| | - Yuan Zhou
- School of Biological Engineering, Hefei Technology College, Hefei, China
| | - Cheng Kong
- Research Institute of Intestinal Diseases, Tongji University, School of Medicine, Shanghai, China
- *Correspondence: Longxiang Zhou, ; Cheng Kong,
| | - Longxiang Zhou
- Department of General Surgery, Jinshan Branch of Shanghai Sixth People’s Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Longxiang Zhou, ; Cheng Kong,
| |
Collapse
|
85
|
Lucarini E, Di Pilato V, Parisio C, Micheli L, Toti A, Pacini A, Bartolucci G, Baldi S, Niccolai E, Amedei A, Rossolini GM, Nicoletti C, Cryan JF, O'Mahony SM, Ghelardini C, Di Cesare Mannelli L. Visceral sensitivity modulation by faecal microbiota transplantation: the active role of gut bacteria in pain persistence. Pain 2022; 163:861-877. [PMID: 34393197 PMCID: PMC9009324 DOI: 10.1097/j.pain.0000000000002438] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/19/2021] [Accepted: 08/02/2021] [Indexed: 11/25/2022]
Abstract
Recent findings linked gastrointestinal disorders characterized by abdominal pain to gut microbiota composition. The present work aimed to evaluate the power of gut microbiota as a visceral pain modulator and, consequently, the relevance of its manipulation as a therapeutic option in reversing postinflammatory visceral pain persistence. Colitis was induced in mice by intrarectally injecting 2,4-dinitrobenzenesulfonic acid (DNBS). The effect of faecal microbiota transplantation from viscerally hypersensitive DNBS-treated and naive donors was evaluated in control rats after an antibiotic-mediated microbiota depletion. Faecal microbiota transplantation from DNBS donors induced a long-lasting visceral hypersensitivity in control rats. Pain threshold trend correlated with major modifications in the composition of gut microbiota and short chain fatty acids. By contrast, no significant alterations of colon histology, permeability, and monoamines levels were detected. Finally, by manipulating the gut microbiota of DNBS-treated animals, a counteraction of persistent visceral pain was achieved. The present results provide novel insights into the relationship between intestinal microbiota and visceral hypersensitivity, highlighting the therapeutic potential of microbiota-targeted interventions.
Collapse
Affiliation(s)
- Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Vincenzo Di Pilato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Clinical Microbiology and Virology Unit, Florence Careggi University Hospital, Florence, Italy
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - Carmen Parisio
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Alessandra Toti
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine, Anatomy and Histology Section, University of Florence, Florence, Italy
| | - Gianluca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences University of Florence, Florence, Italy
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Clinical Microbiology and Virology Unit, Florence Careggi University Hospital, Florence, Italy
| | - Claudio Nicoletti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Siobhain M. O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| |
Collapse
|
86
|
Gupta B, Rai R, Oertel M, Raeman R. Intestinal Barrier Dysfunction in Fatty Liver Disease: Roles of Microbiota, Mucosal Immune System, and Bile Acids. Semin Liver Dis 2022; 42:122-137. [PMID: 35738255 PMCID: PMC9307091 DOI: 10.1055/s-0042-1748037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) describes a spectrum of progressive liver diseases ranging from simple steatosis to steatohepatitis and fibrosis. Globally, NAFLD is the leading cause of morbidity and mortality associated with chronic liver disease, and NAFLD patients are at a higher risk of developing cirrhosis and hepatocellular carcinoma. While there is a consensus that inflammation plays a key role in promoting NAFLD progression, the underlying mechanisms are not well understood. Recent clinical and experimental evidence suggest that increased hepatic translocation of gut microbial antigens, secondary to diet-induced impairment of the intestinal barrier may be important in driving hepatic inflammation in NAFLD. Here, we briefly review various endogenous and exogenous factors influencing the intestinal barrier and present recent advances in our understanding of cellular and molecular mechanisms underlying intestinal barrier dysfunction in NAFLD.
Collapse
Affiliation(s)
- Biki Gupta
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ravi Rai
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael Oertel
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania,Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,McGowan Institute for Regenerative Medicine, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Reben Raeman
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania,Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
87
|
Kuttke M, Hromadová D, Yildirim C, Brunner JS, Vogel A, Paar H, Peters S, Weber M, Hofmann M, Kerndl M, Kieler M, Datler H, Musiejovsky L, Salzmann M, Lang M, Soukup K, Halfmann A, Sharif O, Schabbauer G. PI3K Signaling in Dendritic Cells Aggravates DSS-Induced Colitis. Front Immunol 2022; 13:695576. [PMID: 35514976 PMCID: PMC9063450 DOI: 10.3389/fimmu.2022.695576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Aberrant innate immune responses to the gut microbiota are causally involved in the pathogenesis of inflammatory bowel diseases (IBD). The exact triggers and main signaling pathways activating innate immune cells and how they modulate adaptive immunity in IBD is still not completely understood. Here, we report that the PI3K/PTEN signaling pathway in dendritic cells enhances IL-6 production in a model of DSS-induced colitis. This results in exacerbated Th1 cell responses and increased mortality in DC-specific PTEN knockout (PTENΔDC) animals. Depletion of the gut microbiota using antibiotics as well as blocking IL-6R signaling rescued mortality in PTENΔDC mice, whereas adoptive transfer of Flt3L-derived PTEN-/- DCs into WT recipients exacerbated DSS-induced colitis and increased mortality. Taken together, we show that the PI3K signaling pathway in dendritic cells contributes to disease pathology by promoting IL-6 mediated Th1 responses.
Collapse
Affiliation(s)
- Mario Kuttke
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Dominika Hromadová
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Ceren Yildirim
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Julia S. Brunner
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Andrea Vogel
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Hannah Paar
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Sophie Peters
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Maria Weber
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Melanie Hofmann
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Martina Kerndl
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Markus Kieler
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Hannes Datler
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Laszlo Musiejovsky
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Manuel Salzmann
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michaela Lang
- Department of Gastroenterology and Hepatology, Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Klara Soukup
- St. Anna Children’s Cancer Research Institute, Vienna, Austria
| | - Angela Halfmann
- St. Anna Children’s Cancer Research Institute, Vienna, Austria
| | - Omar Sharif
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Gernot Schabbauer
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
88
|
Black Tea Reduces Diet-Induced Obesity in Mice via Modulation of Gut Microbiota and Gene Expression in Host Tissues. Nutrients 2022; 14:nu14081635. [PMID: 35458198 PMCID: PMC9027533 DOI: 10.3390/nu14081635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 12/17/2022] Open
Abstract
Black tea was reported to alter the microbiome populations and metabolites in diet-induced obese mice and displays properties that prevent obesity, but the underlying mechanism of the preventative effect of black tea on high-fat diet (HFD) induced obesity has not been elucidated. Epigenetic studies are a useful tool for determining the relationship between obesity and environment. Here, we show that the water extract of black tea (Lapsang souchong, LS) reverses HFD-induced gut dysbiosis, alters the tissue gene expression, changes the level of a major epigenetic modification (DNA methylation), and prevents obesity in HFD feeding mice. The anti-obesity properties of black tea are due to alkaloids, which are the principal active components. Our data indicate that the anti-obesity benefits of black tea are transmitted via fecal transplantation, and the change of tissue gene expression and the preventative effects on HFD-induced obesity in mice of black tea are dependent on the gut microbiota. We further show that black tea could regulate the DNA methylation of imprinted genes in the spermatozoa of high-fat diet mice. Our results show a mechanistic link between black tea, changes in the gut microbiota, epigenetic processes, and tissue gene expression in the modulation of diet-induced metabolic dysfunction.
Collapse
|
89
|
Microbial drivers of DSS variability. Nat Microbiol 2022; 7:478-479. [PMID: 35365788 DOI: 10.1038/s41564-022-01097-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
90
|
Sasaki T, Nagashima H, Okuma A, Yamauchi T, Yamasaki K, Aiba S, So T, Ishii N, Owada Y, MaruYama T, Kobayashi S. Functional Analysis of the Transcriptional Regulator IκB-ζ in Intestinal Homeostasis. Dig Dis Sci 2022; 67:1252-1259. [PMID: 33818662 DOI: 10.1007/s10620-021-06958-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 03/12/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND The Toll-like receptor signaling pathway contributes to the regulation of intestinal homeostasis through interactions with commensal bacteria. Although the transcriptional regulator IκB-ζ can be induced by Toll-like receptor signaling, its role in intestinal homeostasis is still unclear. AIMS To investigate the role of IκB-ζ in gut homeostasis. METHODS DSS-administration induced colitis in control and IκB-ζ-deficient mice. The level of immunoglobulins in feces was detected by ELISA. The immunological population in lamina propria (LP) was analyzed by FACS. RESULTS IκB-ζ-deficient mice showed severe inflammatory diseases with DSS administration in the gut. The level of IgM in the feces after DSS administration was less in IκB-ζ-deficient mice compared to control mice. Upon administration of DSS, IκB-ζ-deficient mice showed exaggerated intestinal inflammation (more IFN-g-producing CD4+ T cells in LP), and antibiotic treatment canceled this inflammatory phenotype. CONCLUSION IκB-ζ plays a crucial role in maintaining homeostasis in the gut.
Collapse
Affiliation(s)
- Tomoki Sasaki
- Laboratory of Cell Recognition and Response, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Hiroyuki Nagashima
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Okuma
- Laboratory of Cell Recognition and Response, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Takeshi Yamauchi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kenshi Yamasaki
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takanori So
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuji Owada
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi, Japan
| | - Takashi MaruYama
- Laboratory of Cell Recognition and Response, Graduate School of Life Sciences, Tohoku University, Sendai, Japan.,Mucosal Immunology Unit, NIDCR, NIH, Bethesda, MD, USA
| | - Shuhei Kobayashi
- Laboratory of Cell Recognition and Response, Graduate School of Life Sciences, Tohoku University, Sendai, Japan. .,Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan. .,Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi, Japan.
| |
Collapse
|
91
|
Forster SC, Clare S, Beresford-Jones BS, Harcourt K, Notley G, Stares MD, Kumar N, Soderholm AT, Adoum A, Wong H, Morón B, Brandt C, Dougan G, Adams DJ, Maloy KJ, Pedicord VA, Lawley TD. Identification of gut microbial species linked with disease variability in a widely used mouse model of colitis. Nat Microbiol 2022; 7:590-599. [PMID: 35365791 PMCID: PMC8975739 DOI: 10.1038/s41564-022-01094-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 02/24/2022] [Indexed: 12/12/2022]
Abstract
Experimental mouse models are central to basic biomedical research; however, variability exists across genetically identical mice and mouse facilities making comparisons difficult. Whether specific indigenous gut bacteria drive immunophenotypic variability in mouse models of human disease remains poorly understood. We performed a large-scale experiment using 579 genetically identical laboratory mice from a single animal facility, designed to identify the causes of disease variability in the widely used dextran sulphate sodium mouse model of inflammatory bowel disease. Commonly used treatment endpoint measures-weight loss and intestinal pathology-showed limited correlation and varied across mouse lineages. Analysis of the gut microbiome, coupled with machine learning and targeted anaerobic culturing, identified and isolated two previously undescribed species, Duncaniella muricolitica and Alistipes okayasuensis, and demonstrated that they exert dominant effects in the dextran sulphate sodium model leading to variable treatment endpoint measures. We show that the identified gut microbial species are common, but not ubiquitous, in mouse facilities around the world, and suggest that researchers monitor for these species to provide experimental design opportunities for improved mouse models of human intestinal diseases.
Collapse
Affiliation(s)
- Samuel C Forster
- Experimental Cancer Genetics Lab, Wellcome Sanger Institute, Hinxton, UK
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Simon Clare
- Experimental Cancer Genetics Lab, Wellcome Sanger Institute, Hinxton, UK
| | - Benjamin S Beresford-Jones
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Katherine Harcourt
- Experimental Cancer Genetics Lab, Wellcome Sanger Institute, Hinxton, UK
| | - George Notley
- Experimental Cancer Genetics Lab, Wellcome Sanger Institute, Hinxton, UK
| | - Mark D Stares
- Experimental Cancer Genetics Lab, Wellcome Sanger Institute, Hinxton, UK
| | - Nitin Kumar
- Experimental Cancer Genetics Lab, Wellcome Sanger Institute, Hinxton, UK
| | - Amelia T Soderholm
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Anne Adoum
- Experimental Cancer Genetics Lab, Wellcome Sanger Institute, Hinxton, UK
| | | | - Bélen Morón
- Experimental Medicine Division, University of Oxford, Oxford, UK
| | - Cordelia Brandt
- Experimental Cancer Genetics Lab, Wellcome Sanger Institute, Hinxton, UK
| | - Gordon Dougan
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - David J Adams
- Experimental Cancer Genetics Lab, Wellcome Sanger Institute, Hinxton, UK
| | - Kevin J Maloy
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Virginia A Pedicord
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Trevor D Lawley
- Experimental Cancer Genetics Lab, Wellcome Sanger Institute, Hinxton, UK.
| |
Collapse
|
92
|
Intestinal Microbiota-Mediated Biotransformations Alter the Pharmacokinetics of the Major Metabolites of Azathioprine in Rats after Oral Administration. Drug Metab Pharmacokinet 2022; 45:100458. [DOI: 10.1016/j.dmpk.2022.100458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/24/2022] [Accepted: 03/11/2022] [Indexed: 11/22/2022]
|
93
|
Chen M, Liu C, Dai M, Wang Q, Li C, Hung W. Bifidobacterium lactis BL-99 modulates intestinal inflammation and functions in zebrafish models. PLoS One 2022; 17:e0262942. [PMID: 35171916 PMCID: PMC9126502 DOI: 10.1371/journal.pone.0262942] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/07/2022] [Indexed: 01/10/2023] Open
Abstract
This study was designed to explore the therapeutics and the mechanisms of a patented and marked gastric acid and intestine juice-resistant probiotics Bifidobacterium lactis BL-99 (B. lactis BL-99) on the intestinal inflammation and functions in the zebrafish models. After feeding for 6 hours, B. lactis BL-99 was fully retained in the larval zebrafish intestinal tract and stayed for over 24 hours. B. lactis BL-99 promoted the intestinal motility and effectively alleviated aluminum sulfate-induced larval zebrafish constipation (p < 0.01). Irregular high glucose diet induced adult zebrafish intestinal functional and metabolic disorders. After fed with B. lactis BL-99, IL-1β gene expression was significantly down-regulated, and IL-10 and IL-12 gene levels were markedly up-regulated in this model (p < 0.05). The intestinal lipase activity was elevated in the adult zebrafish intestinal functional disorder model after B. lactis BL-99 treatment (p < 0.05), but tryptase content had no statistical changes (p > 0.05). B. lactis BL-99 improved the histopathology of the adult zebrafish intestinal inflammation, increased the goblet cell numbers, and up-and-down metabolites were markedly recovered after treatment of B. lactis BL-99 (p < 0.05). These results suggest that B. lactis BL-99 could relieve intestinal inflammation and promote intestinal functions, at least in part, through modulating intestinal and microbial metabolism to maintain intestinal health.
Collapse
Affiliation(s)
- Meng Chen
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot,
China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd.,
Hohhot, China
| | - Chinfeng Liu
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot,
China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd.,
Hohhot, China
| | - Mingzhu Dai
- Hunter Biotechnology, Inc., F1A, Hangzhou, China
| | - Qinwen Wang
- Hunter Biotechnology, Inc., F1A, Hangzhou, China
| | - Chunqi Li
- Hunter Biotechnology, Inc., F1A, Hangzhou, China
| | - Weilian Hung
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot,
China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd.,
Hohhot, China
| |
Collapse
|
94
|
Olivier S, Diounou H, Pochard C, Frechin L, Durieu E, Foretz M, Neunlist M, Rolli-Derkinderen M, Viollet B. Intestinal Epithelial AMPK Deficiency Causes Delayed Colonic Epithelial Repair in DSS-Induced Colitis. Cells 2022; 11:cells11040590. [PMID: 35203241 PMCID: PMC8869996 DOI: 10.3390/cells11040590] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 12/17/2022] Open
Abstract
Dysfunctions in the intestinal barrier, associated with an altered paracellular pathway, are commonly observed in inflammatory bowel disease (IBD). The AMP-activated protein kinase (AMPK), principally known as a cellular energy sensor, has also been shown to play a key role in the stabilization and assembly of tight junctions. Here, we aimed to investigate the contribution of intestinal epithelial AMPK to the initiation, progression and resolution of acute colitis. We also tested the hypothesis that protection mediated by metformin administration on intestinal epithelium damage required AMPK activation. A dextran sodium sulfate (DSS)-induced colitis model was used to assess disease progression in WT and intestinal epithelial cell (IEC)-specific AMPK KO mice. Barrier integrity was analyzed by measuring paracellular permeability following dextran-4kDa gavage and pro-inflammatory cytokines and tight junction protein expression. The deletion of intestinal epithelial AMPK delayed intestinal injury repair after DSS exposure and was associated with a slower re-epithelization of the intestinal mucosa coupled with severe ulceration and inflammation, and altered barrier function. Following intestinal injury, IEC AMPK KO mice displayed a lower goblet cell counts with concomitant decreased Muc2 gene expression, unveiling an impaired restitution of goblet cells and contribution to wound healing process. Metformin administration during the recovery phase attenuated the severity of DSS-induced colitis through improvement in intestinal repair capacity in both WT and IEC AMPK KO mice. Taken together, these findings demonstrate a critical role for IEC-expressed AMPK in regulating mucosal repair and epithelial regenerative capacity following acute colonic injury. Our studies further underscore the therapeutic potential of metformin to support repair of the injured intestinal epithelium, but this effect is conferred independently of intestinal epithelial AMPK.
Collapse
Affiliation(s)
- Séverine Olivier
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014 Paris, France; (S.O.); (H.D.); (L.F.); (M.F.)
| | - Hanna Diounou
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014 Paris, France; (S.O.); (H.D.); (L.F.); (M.F.)
| | - Camille Pochard
- Université de Nantes, TENS, The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l’Appareil Digestif, F-44093 Nantes, France; (C.P.); (E.D.); (M.N.); (M.R.-D.)
| | - Lisa Frechin
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014 Paris, France; (S.O.); (H.D.); (L.F.); (M.F.)
| | - Emilie Durieu
- Université de Nantes, TENS, The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l’Appareil Digestif, F-44093 Nantes, France; (C.P.); (E.D.); (M.N.); (M.R.-D.)
| | - Marc Foretz
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014 Paris, France; (S.O.); (H.D.); (L.F.); (M.F.)
| | - Michel Neunlist
- Université de Nantes, TENS, The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l’Appareil Digestif, F-44093 Nantes, France; (C.P.); (E.D.); (M.N.); (M.R.-D.)
| | - Malvyne Rolli-Derkinderen
- Université de Nantes, TENS, The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l’Appareil Digestif, F-44093 Nantes, France; (C.P.); (E.D.); (M.N.); (M.R.-D.)
| | - Benoit Viollet
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014 Paris, France; (S.O.); (H.D.); (L.F.); (M.F.)
- Correspondence: ; Tel.: +33-1-4441-2401
| |
Collapse
|
95
|
Wang R, Moniruzzaman M, Wong KY, Wiid P, Harding A, Giri R, Tong W(H, Creagh J, Begun J, McGuckin MA, Hasnain SZ. Gut microbiota shape the inflammatory response in mice with an epithelial defect. Gut Microbes 2022; 13:1-18. [PMID: 33645438 PMCID: PMC7928202 DOI: 10.1080/19490976.2021.1887720] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Intestinal epithelial cell endoplasmic reticulum (ER) stress has been implicated in intestinal inflammation. It remains unclear whether ER stress is an initiator of or a response to inflammation. Winnie mice, carrying a Muc2 gene mutation resulting in intestinal goblet cell ER stress, develop spontaneous colitis with a depleted mucus barrier and increased bacterial translocation. This study aims to determine whether the microbiota was required for the development of Winnie colitis, and whether protein misfolding itself can initiate inflammation directly in absence of the microbiota. To assess the role of microbiota in driving Winnie colitis, WT and Winnie mice on the same background were rederived into the germ-free facility and housed in the Trexler-type soft-sided isolators. The colitis phenotype of these mice was assessed and compared to WT and Winnie mice housed within a specific pathogen-free facility. We found that Winnie colitis was substantially reduced but not abolished under germ-free conditions. Expression of inflammatory cytokine genes was reduced but several chemokines remained elevated in absence of microbiota. Concomitantly, ER stress was also diminished, although mucin misfolding persisted. RNA-Seq revealed that Winnie differentiated colon organoids have decreased expression of the negative regulators of the inflammatory response compared to WT. This data along with the increase in Mip2a chemokine expression, suggests that the epithelial cells in the Winnie mice are more responsive to stimuli. Moreover, the data demonstrate that intestinal epithelial intrinsic protein misfolding can prime an inflammatory response without initiating the unfolded protein response in the absence of the microbiota. However, the microbiota is necessary for the amplification of colitis in Winnie mice. Genetic predisposition to mucin misfolding in secretory cells initiates mild inflammatory signals. However, the inflammatory signal sets a forward-feeding cycle establishing progressive inflammation in the presence of microbiota.Abbreviations: Endoplasmic Reticulum: ER; Mucin-2: Muc-2; GF: Germ-Free; Inflammatory Bowel Disease: IBD.
Collapse
Affiliation(s)
- Ran Wang
- Immunopathology Group, Mater Research Institute – The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Md Moniruzzaman
- Immunopathology Group, Mater Research Institute – The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Kuan Yau Wong
- Immunopathology Group, Mater Research Institute – The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Percival Wiid
- Immunopathology Group, Mater Research Institute – The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Alexa Harding
- Immunopathology Group, Mater Research Institute – The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Rabina Giri
- Inflammatory Bowel Disease Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Wendy (Hui) Tong
- Immunopathology Group, Mater Research Institute – The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Jackie Creagh
- Immunopathology Group, Mater Research Institute – The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Jakob Begun
- Inflammatory Bowel Disease Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, Australia,Mater Adult Hospital, Mater Health Services, South Brisbane, Australia
| | - Michael A. McGuckin
- Immunopathology Group, Mater Research Institute – The University of Queensland, Translational Research Institute, Brisbane, Australia,Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Australia
| | - Sumaira Z. Hasnain
- Immunopathology Group, Mater Research Institute – The University of Queensland, Translational Research Institute, Brisbane, Australia,Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, Australia,CONTACT Sumaira Z. Hasnain Mater Research Institute – University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, Qld4102, Australia; Ran Wang Mater Research Institute – University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, Qld 4102, Australia
| |
Collapse
|
96
|
Szychowiak P, Villageois-Tran K, Patrier J, Timsit JF, Ruppé É. The role of the microbiota in the management of intensive care patients. Ann Intensive Care 2022; 12:3. [PMID: 34985651 PMCID: PMC8728486 DOI: 10.1186/s13613-021-00976-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022] Open
Abstract
The composition of the gut microbiota is highly dynamic and changes according to various conditions. The gut microbiota mainly includes difficult-to-cultivate anaerobic bacteria, hence knowledge about its composition has significantly arisen from culture-independent methods based on next-generation sequencing (NGS) such as 16S profiling and shotgun metagenomics. The gut microbiota of patients hospitalized in intensive care units (ICU) undergoes many alterations because of critical illness, antibiotics, and other ICU-specific medications. It is then characterized by lower richness and diversity, and dominated by opportunistic pathogens such as Clostridioides difficile and multidrug-resistant bacteria. These alterations are associated with an increased risk of infectious complications or death. Specifically, at the time of writing, it appears possible to identify distinct microbiota patterns associated with severity or infectivity in COVID-19 patients, paving the way for the potential use of dysbiosis markers to predict patient outcomes. Correcting the microbiota disturbances to avoid their consequences is now possible. Fecal microbiota transplantation is recommended in recurrent C. difficile infections and microbiota-protecting treatments such as antibiotic inactivators are currently being developed. The growing interest in the microbiota and microbiota-associated therapies suggests that the control of the dysbiosis could be a key factor in the management of critically ill patients. The present narrative review aims to provide a synthetic overview of microbiota, from healthy individuals to critically ill patients. After an introduction to the different techniques used for studying the microbiota, we review the determinants involved in the alteration of the microbiota in ICU patients and the latter's consequences. Last, we assess the means to prevent or correct microbiota alteration.
Collapse
Affiliation(s)
- Piotr Szychowiak
- Université de Paris, IAME, INSERM, 75018, Paris, France
- Service de Médecine Intensive-Réanimation, Centre Hospitalier Régional Universitaire de Tours, 37000, Tours, France
| | - Khanh Villageois-Tran
- Université de Paris, IAME, INSERM, 75018, Paris, France
- Laboratoire de Bactériologie, AP-HP, Hôpital Beaujon, 92110, Paris, France
| | - Juliette Patrier
- Université de Paris, IAME, INSERM, 75018, Paris, France
- Service de Réanimation Médicale Et Infectieuse, AP-HP, Hôpital Bichat, 75018, Paris, France
| | - Jean-François Timsit
- Université de Paris, IAME, INSERM, 75018, Paris, France
- Service de Réanimation Médicale Et Infectieuse, AP-HP, Hôpital Bichat, 75018, Paris, France
| | - Étienne Ruppé
- Université de Paris, IAME, INSERM, 75018, Paris, France.
- Laboratoire de Bactériologie, AP-HP, Hôpital Bichat-Claude Bernard, 46 rue Henri Huchard, 75018, Paris, France.
| |
Collapse
|
97
|
Xia B, Liu X, Li X, Wang Y, Wang D, Kou R, Zhang L, Shi R, Ye J, Bo X, Liu Q, Zhao B, Liu X. Sesamol Ameliorates Dextran Sulfate Sodium-induced Depression-like and Anxiety-like Behaviors in Colitis Mice:The potential involvement of Gut-Brain Axis. Food Funct 2022; 13:2865-2883. [DOI: 10.1039/d1fo03888e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Inflammatory bowel disease (IBD) is accompanied by some psychiatric disorders, including anxiety and depression. Sesamol has been reported to alleviate colitis symptoms and depression-like behaviors caused by chronic unpredictable mild...
Collapse
|
98
|
El-Sahhar S, Varga-Weisz P. The gut microbiome in health and disease: Inflammatory bowel diseases. ADV ECOL RES 2022. [DOI: 10.1016/bs.aecr.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
99
|
Hu Y, Ye Z, Wu M, She Y, Li L, Xu Y, Qin K, Hu Z, Yang M, Lu F, Ye Q. The Communication Between Intestinal Microbiota and Ulcerative Colitis: An Exploration of Pathogenesis, Animal Models, and Potential Therapeutic Strategies. Front Med (Lausanne) 2021; 8:766126. [PMID: 34966755 PMCID: PMC8710685 DOI: 10.3389/fmed.2021.766126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022] Open
Abstract
Ulcerative Colitis (UC) is a chronic inflammatory bowel disease. The prolonged course of UC and the lack of effective treatment management make it difficult to cure, affecting the health and life safety of patients. Although UC has received more attention, the etiology and pathogenesis of UC are still unclear. Therefore, it is urgent to establish an updated and comprehensive understanding of UC and explore effective treatment strategies. Notably, sufficient evidence shows that the intestinal microbiota plays an important role in the pathogenesis of UC, and the treating method aimed at improving the balance of the intestinal microbiota exhibits a therapeutic potential for UC. This article reviews the relationship between the genetic, immunological and microbial risk factors with UC. At the same time, the UC animal models related to intestinal microbiota dysbiosis induced by chemical drugs were evaluated. Finally, the potential value of the therapeutic strategies for restoring intestinal microbial homeostasis and treating UC were also investigated. Comprehensively, this study may help to carry out preclinical research, treatment theory and methods, and health management strategy of UC, and provide some theoretical basis for TCM in the treatment of UC.
Collapse
Affiliation(s)
- Yu Hu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mingquan Wu
- Department of Pharmacy, Sichuan Provincial Orthopedic Hospital, Chengdu, China
| | - Yingqi She
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Linzhen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yujie Xu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kaihua Qin
- Health Preservation and Rehabilitation College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhipeng Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maoyi Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fating Lu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiaobo Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
100
|
Casado-Bedmar M, Viennois E. MicroRNA and Gut Microbiota: Tiny but Mighty-Novel Insights into Their Cross-talk in Inflammatory Bowel Disease Pathogenesis and Therapeutics. J Crohns Colitis 2021; 16:992-1005. [PMID: 34918052 PMCID: PMC9282881 DOI: 10.1093/ecco-jcc/jjab223] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/06/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022]
Abstract
MicroRNAs [miRNAs], small non-coding RNAs, have recently been described as crucial contributors to intestinal homeostasis. They can interact with the gut microbiota in a reciprocal manner and deeply affect host health status, leading to several disorders when unbalanced. Inflammatory bowel disease [IBD] is a chronic inflammation of the gastrointestinal tract that co-occurs with alterations of the gut microbiota, and whose aetiology remains largely unclear. On one hand, host miRNA could be playing a relevant role in IBD pathophysiology by shaping the gut microbiota. The gut microbiome, on the other hand, may regulate the expression of host miRNAs, resulting in intestinal epithelial dysfunction, altered autophagy, and immune hyperactivation. Interestingly, it has been hypothesised that their reciprocal impact may be used for therapeutic goals. This review describes the latest research and suggests mechanisms through which miRNA and intestinal microbiota, as joint actors, may participate specifically in IBD pathophysiology. Furthermore, we discuss the diagnostic power and therapeutic potential resulting from their bidirectional communication after faecal transplantation, probiotics intake, or anti-miRNAs or miRNA mimics administration. The current literature is summarised in the present work in a comprehensive manner, hoping to provide a better understanding of the miRNA-microbiota cross-talk and to facilitate their application in IBD.
Collapse
Affiliation(s)
- Maite Casado-Bedmar
- INSERM, U1149, Center for Research on Inflammation, Université de Paris, Paris, France
| | - Emilie Viennois
- Corresponding author: Emilie Viennois, INSERM, U1149, Center for Research on Inflammation, Université de Paris, 75018 Paris, France.
| |
Collapse
|