1
|
Mei Z, Du P, Han Y, Shao Z, Zheng D. Probiotics interventions modulating gut microbiota composition in individuals with intestinal constipation: Protocol of a systemic review and meta-analysis of randomized controlled trials. PLoS One 2025; 20:e0311799. [PMID: 39854346 PMCID: PMC11759984 DOI: 10.1371/journal.pone.0311799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 09/23/2024] [Indexed: 01/26/2025] Open
Abstract
INTRODUCTION Intestinal constipation is a substantive global health concern, significantly impairing patient quality of life. An emerging view is that the gut microbiota plays a critical role in intestinal function, and probiotics could offer therapeutic benefits. This study aims to consolidate evidence from randomized controlled trials (RCTs) that assess the effectiveness of probiotics in modulating microbiota and ameliorating symptoms of constipation. METHODS We will execute a systematic evidence search across Medline (via PubMed), Embase, Cochrane CENTRAL, Web of Science, Scopus, and CINAHL, employing explicit search terms and further reference exploration. Two independent reviewers will ensure study selection and data integrity while assessing methodological quality via the Cochrane Collaboration's Risk of Bias-2 tool. Our primary goal is to outline changes in microbiota composition, with secondary outcomes addressing symptom relief and stool characteristics. Meta-analyses will adopt a random-effects model to quantify the effects of interventions, supplemented by subgroup analyses and publication bias assessments to fortify the rigor of our findings. DISCUSSION This study endeavors to provide a rigorous, synthesized overview of the probiotics interventions evidence for modulating gut microbiota in individuals with intestinal constipation. The insights derived could inform clinical guidelines, nurture the creation of novel constipation management strategies, and direct future research in this field. ETHICS AND DISSEMINATION As this study aggregates and analyzes existing data without direct human subject involvement, no ethical approval is required. We will disseminate the study's findings through scientific forums and seek publication in well-regarded, peer-reviewed journals. TRIAL REGISTRATION OSF registration number: 10.17605/OSF.IO/MEAHT.
Collapse
Affiliation(s)
- Zubing Mei
- Department of Anorectal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Anorectal Disease Institute of Shuguang Hospital, Shanghai, China
| | - Peixin Du
- Department of Anorectal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ye Han
- Department of Anorectal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhuo Shao
- Department of General Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, Shanghai, China
| | - De Zheng
- Department of Anorectal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Anorectal Disease Institute of Shuguang Hospital, Shanghai, China
| |
Collapse
|
2
|
Mirhosseini SM, Mahdavi A, Yarmohammadi H, Razavi A, Rezaei M, Soltanipur M, Karimi Nemch M, Jafari Naeini S, Siadat SD. What is the link between the dietary inflammatory index and the gut microbiome? A systematic review. Eur J Nutr 2024; 63:2407-2419. [PMID: 39069586 DOI: 10.1007/s00394-024-03470-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 07/14/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE One highlighted pathogenesis mechanism of diseases is the negative impact of pro-inflammatory diets (PD) on the gut microbiome. This systematic review aimed to study the link between dietary inflammatory index (DII), as an indicator of PD, and gut microbiome. METHODS A systematic search was done in PubMed and Scopus, adhering to the guidelines of Preferred Reporting Items for Systematic Reviews and Meta-Analysis. The assessment of the included studies' quality was performed using the critical appraisal checklist from the Joanna Briggs Institute. RESULTS Ten articles were included eight cross-sectional, one case-control, and, one cohort study. Seven and three included articles reported a weak and moderate relationship between gut microbiome and DII scores, respectively. DII scores were linked to variety in microbiome composition and diversity/richness. More importantly, anti-inflammatory diets as measured by lower DII scores were linked to a more desirable gut microbiome profile. Prevotella stercorea, Veillonella rogosae, Morganella morganii, Ruminococcus torques, Eubacterium nodatum, Alistipes intestine, Clostridium leptum, Morganellaceae family, Enterobacteriaceae family, and, Bacteroides thetaiotaomicron were related to higher DII scores. While, Butyrate-producing bacteria such as Ruminococcaceae and Lachnospiraceae families, Faecalibacterium prausnitzii, and Akkermansia muciniphila were related to lower DII scores. CONCLUSION An anti-inflammatory diet, as measured by a lower DII score, might be linked to variations in the composition and variety of the microbiome. Therefore, the DII score could be useful in microbiota research, however, this possibility needs to be investigated more precisely in future studies.
Collapse
Affiliation(s)
| | - Azamalsadat Mahdavi
- Avicenna Fertility Center, Avicenna Research Institute (ARI), ACECR, Tehran, Iran
| | - Hossein Yarmohammadi
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Quality of Life Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Razavi
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahdi Rezaei
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Masood Soltanipur
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Quality of Life Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammadreza Karimi Nemch
- Student Research Committee, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
- Oral and Dental Diseases Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Sepideh Jafari Naeini
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
3
|
Feng G, Zhang H, Liu H, Zhang X, Jiang H, Liao S, Luo X, Yao H, Xiang B, Liu S, Zhang J, Zhang J, Fang J. Natural Flavonoid-Derived Enzyme Mimics DHKNase Balance the Two-Edged Reactive Oxygen Species Function for Wound Healing and Inflammatory Bowel Disease Therapy. RESEARCH (WASHINGTON, D.C.) 2024; 7:0464. [PMID: 39253100 PMCID: PMC11381673 DOI: 10.34133/research.0464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024]
Abstract
Rational regulation of reactive oxygen species (ROS) plays a vital importance in maintaining homeostasis of living biological systems. For ROS-related pathologies, chemotherapy technology derived from metal nanomaterials currently occupies a pivotal position. However, they suffer from inherent issues such as complicated synthesis, batch-to-batch variability, high cost, and potential biological toxicity caused by metal elements. Here, we reported for the first time that dual-action 3,5-dihydroxy-1-ketonaphthalene-structured small-molecule enzyme imitator (DHKNase) exhibited 2-edged ROS regulation, catering to the execution of physiology-beneficial ROS destiny among diverse pathologies in living systems. Based on this, DHKNase is validated to enable remarkable therapeutic effects in 2 classic disease models, including the pathogen-infected wound-healing model and the dextran sulfate sodium (DSS)-caused inflammatory bowel disease (IBD). This work provides a guiding landmark for developing novel natural small-molecule enzyme imitator and significantly expands their application potential in the biomedical field.
Collapse
Affiliation(s)
- Guangfu Feng
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Huaizu Zhang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Huipeng Liu
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Xiaoyan Zhang
- College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, P.R. China
| | - Hongmei Jiang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Sijie Liao
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Xingyu Luo
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, P.R. China
| | - Hao Yao
- Changsha IMADEK Intelligent Technology Co. Ltd., Changsha, Hunan 410081, P.R. China
| | - Bo Xiang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Shiyu Liu
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Jiali Zhang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Jiaheng Zhang
- College of Chemistry, Food Laboratory of Zhongyuan, Flavour Science Research Center of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Jun Fang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| |
Collapse
|
4
|
Brandaleone L, Dal Buono A, Gabbiadini R, Marcozzi G, Polverini D, Carvello M, Spinelli A, Hassan C, Repici A, Bezzio C, Armuzzi A. Hereditary Colorectal Cancer Syndromes and Inflammatory Bowel Diseases: Risk Management and Surveillance Strategies. Cancers (Basel) 2024; 16:2967. [PMID: 39272825 PMCID: PMC11394661 DOI: 10.3390/cancers16172967] [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: 07/10/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Background and aims: Hereditary colorectal cancer syndromes (HCCS), including familial adenomatous polyposis (FAP) and Lynch syndrome (LS), are the two most important high-risk conditions for colorectal cancer (CRC). Inflammatory bowel disease (IBD) increases the risk by two to six times compared with that in the general population. The intersection of these two conditions has rarely been documented in literature. We aimed to summarize the prevalence, pathogenesis, and current evidence-based management of IBD and HCCS and the underlying molecular mechanisms of accelerated carcinogenesis due to combined inflammation and genetic predisposition. Methods: PubMed and Scopus were searched until June 2024 to identify relevant studies investigating the epidemiology, pathogenesis, and management of IBD and coexisting hereditary CRC syndromes. Results: Co-occurrence of IBD and hereditary CRC syndromes is exceptionally uncommon. Individuals with LS and IBD tend to develop CRC at a younger age than those without IBD, with patients with ulcerative colitis facing particularly elevated risks. The interaction between mismatch deficiency and chronic inflammation requires further investigation.
Collapse
Affiliation(s)
- Luca Brandaleone
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Arianna Dal Buono
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Roberto Gabbiadini
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Giacomo Marcozzi
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Davide Polverini
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Michele Carvello
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
- Colon and Rectal Surgery Division, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Antonino Spinelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
- Colon and Rectal Surgery Division, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Cesare Hassan
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
- Endoscopy Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Alessandro Repici
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
- Endoscopy Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Cristina Bezzio
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Alessandro Armuzzi
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| |
Collapse
|
5
|
Chen J, Zhu Y, Yuan Q. Predicting potential microbe-disease associations based on dual branch graph convolutional network. J Cell Mol Med 2024; 28:e18571. [PMID: 39086148 PMCID: PMC11291560 DOI: 10.1111/jcmm.18571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/15/2024] [Accepted: 06/27/2024] [Indexed: 08/02/2024] Open
Abstract
Studying the association between microbes and diseases not only aids in the prevention and diagnosis of diseases, but also provides crucial theoretical support for new drug development and personalized treatment. Due to the time-consuming and costly nature of laboratory-based biological tests to confirm the relationship between microbes and diseases, there is an urgent need for innovative computational frameworks to anticipate new associations between microbes and diseases. Here, we propose a novel computational approach based on a dual branch graph convolutional network (GCN) module, abbreviated as DBGCNMDA, for identifying microbe-disease associations. First, DBGCNMDA calculates the similarity matrix of diseases and microbes by integrating functional similarity and Gaussian association spectrum kernel (GAPK) similarity. Then, semantic information from different biological networks is extracted by two GCN modules from different perspectives. Finally, the scores of microbe-disease associations are predicted based on the extracted features. The main innovation of this method lies in the use of two types of information for microbe/disease similarity assessment. Additionally, we extend the disease nodes to address the issue of insufficient features due to low data dimensionality. We optimize the connectivity between the homogeneous entities using random walk with restart (RWR), and then use the optimized similarity matrix as the initial feature matrix. In terms of network understanding, we design a dual branch GCN module, namely GlobalGCN and LocalGCN, to fine-tune node representations by introducing side information, including homologous neighbour nodes. We evaluate the accuracy of the DBGCNMDA model using five-fold cross-validation (5-fold-CV) technique. The results show that the area under the receiver operating characteristic curve (AUC) and area under the precision versus recall curve (AUPR) of the DBGCNMDA model in the 5-fold-CV are 0.9559 and 0.9630, respectively. The results from the case studies using published experimental data confirm a significant number of predicted associations, indicating that DBGCNMDA is an effective tool for predicting potential microbe-disease associations.
Collapse
Affiliation(s)
- Jing Chen
- School of Electronic and Information EngineeringSuzhou University of Science and TechnologySuzhouChina
| | - Yongjun Zhu
- School of Electronic and Information EngineeringSuzhou University of Science and TechnologySuzhouChina
| | - Qun Yuan
- Department of Respiratory Medicine, The Affiliated Suzhou Hospital of NanjingUniversity Medical SchoolSuzhouChina
| |
Collapse
|
6
|
Haque M, Kaminsky L, Abdulqadir R, Engers J, Kovtunov E, Rawat M, Al-Sadi R, Ma TY. Lactobacillus acidophilus inhibits the TNF-α-induced increase in intestinal epithelial tight junction permeability via a TLR-2 and PI3K-dependent inhibition of NF-κB activation. Front Immunol 2024; 15:1348010. [PMID: 39081324 PMCID: PMC11286488 DOI: 10.3389/fimmu.2024.1348010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
Background Defective intestinal epithelial tight junction (TJ), characterized by an increase in intestinal TJ permeability, has been shown to play a critical role in the pathogenesis of inflammatory bowel disease (IBD). Tumor necrosis factor-α (TNF-α) is a key pro-inflammatory cytokine involved in the immunopathology of IBD and has been shown to cause an increase in intestinal epithelial TJ permeability. Although TNF-α antibodies and other biologics have been advanced for use in IBD treatment, these therapies are associated with severe side effects and have limited efficacy, and there is an urgent need for therapies with benign profiles and high therapeutic efficacy. Probiotic bacteria have beneficial effects and are generally safe and represent an important class of potential therapeutic agents in IBD. Lactobacillus acidophilus (LA) is one of the most used probiotics for wide-ranging health benefits, including in gastrointestinal, metabolic, and inflammatory disorders. A specific strain of LA, LA1, was recently demonstrated to have protective and therapeutic effects on the intestinal epithelial TJ barrier. However, the mechanisms of actions of LA1 remain largely unknown. Methods The primary aim of this study was to investigate microbial-epithelial interactions and novel signaling pathways that regulate the effect of LA1 on TNF-α-induced increase in intestinal epithelial TJ permeability, using cell culture and animal model systems. Results and Conclusion Pre-treatment of filter-grown Caco-2 monolayers with LA1 prevented the TNF-α-induced increase in intestinal epithelial TJ permeability by inhibiting TNF-α-induced activation of NF-κB p50/p65 and myosin light chain kinase (MLCK) gene and kinase activity in a TLR-2-dependent manner. LA1 produced a TLR-2- and MyD88-dependent activation of NF-κB p50/p65 in immune cells; however, LA1, in intestinal cells, inhibited the NF-κB p50/p65 activation in a TLR-2-dependent but MyD88-independent manner. In addition, LA1 inhibition of NF-κB p50/p65 and MLCK gene was mediated by TLR-2 pathway activation of phosphatidylinositol 3-kinase (PI3K) and IKK-α phosphorylation. Our results demonstrated novel intracellular signaling pathways by which LA1/TLR-2 suppresses the TNF-α pathway activation of NF-κB p50/p65 in intestinal epithelial cells and protects against the TNF-α-induced increase in intestinal epithelial TJ permeability.
Collapse
Affiliation(s)
- Mohammad Haque
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Lauren Kaminsky
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Raz Abdulqadir
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Jessica Engers
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Evgeny Kovtunov
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Manmeet Rawat
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Rana Al-Sadi
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Thomas Y. Ma
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
7
|
Huang H, Liu X, Lang Y, Cui J, Zhong D, Zhou M. Breaking barriers: bacterial-microalgae symbiotic systems as a probiotic delivery system. J Nanobiotechnology 2024; 22:371. [PMID: 38918805 PMCID: PMC11197275 DOI: 10.1186/s12951-024-02647-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/16/2024] [Indexed: 06/27/2024] Open
Abstract
The gut microbiota is one of the essential contributors of the pathogenesis and progress of inflammatory bowel disease (IBD). Compared with first-line drug therapy, probiotic supplementation has emerged as a viable and secure therapeutic approach for managing IBD through the regulation of both the immune system and gut microbiota. Nevertheless, the efficacy of oral probiotic supplements is hindered by their susceptibility to the gastrointestinal barrier, leading to diminished bioavailability and restricted intestinal colonization. Here, we developed a bacteria-microalgae symbiosis system (EcN-SP) for targeted intestinal delivery of probiotics and highly effective treatment of colitis. The utilization of mircroalge Spirulina platensis (SP) as a natural carrier for the probiotic Escherichia coli Nissle 1917 (EcN) demonstrated potential benefits in promoting EcN proliferation, facilitating effective intestinal delivery and colonization. The alterations in the binding affinity of EcN-SP within the gastrointestinal environment, coupled with the distinctive structural properties of the SP carrier, served to overcome gastrointestinal barriers, minimizing transgastric EcN loss and enabling sustained intestinal retention and colonization. The oral administration of EcN-SP could effectively treat IBD by reducing the expression of intestinal inflammatory factors, maintaining the intestinal barrier and regulating the balance of gut microbiota. This probiotic delivery approach is inspired by symbiotic interactions found in nature and offers advantages in terms of feasibility, safety, and efficacy, thus holding significant promise for the management of gastrointestinal disorders.
Collapse
Affiliation(s)
- Hui Huang
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Xiaoyang Liu
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yutong Lang
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Zhejiang University-University of Edinburgh Institute (ZJE), Zhejiang University, Haining, 314400, China
| | - Jiarong Cui
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Danni Zhong
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China.
| | - Min Zhou
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China.
- Zhejiang University-University of Edinburgh Institute (ZJE), Zhejiang University, Haining, 314400, China.
- State Key Laboratory (SKL) of Biobased Transportation Fuel Technology, Zhejiang University, Hangzhou, 310027, China.
| |
Collapse
|
8
|
Kuo YR, Lin CH, Lin WS, Pan MH. L-Glutamine Substantially Improves 5-Fluorouracil-Induced Intestinal Mucositis by Modulating Gut Microbiota and Maintaining the Integrity of the Gut Barrier in Mice. Mol Nutr Food Res 2024; 68:e2300704. [PMID: 38656560 DOI: 10.1002/mnfr.202300704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/26/2024] [Indexed: 04/26/2024]
Abstract
SCOPE This study investigates the potential of glutamine to mitigate intestinal mucositis and dysbiosis caused by the chemotherapeutic agent 5-fluorouracil (5-FU). METHODS AND RESULTS Over twelve days, Institute of Cancer Research (ICR) mice are given low (0.5 mg kg-1) or high (2 mg kg-1) doses of L-Glutamine daily, with 5-FU (50 mg kg-1) administered between days six and nine. Mice receiving only 5-FU exhibited weight loss, diarrhea, abnormal cell growth, and colonic inflammation, correlated with decreased mucin proteins, increased endotoxins, reduced fecal short-chain fatty acids, and altered gut microbiota. Glutamine supplementation counteracted these effects by inhibiting the Toll-like receptor 4/nuclear factor kappa B (TLR4/NF-κB) pathway, modulating nuclear factor erythroid 2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) oxidative stress proteins, and increasing mammalian target of rapamycin (mTOR) levels, thereby enhancing microbial diversity and protecting intestinal mucosa. CONCLUSIONS These findings underscore glutamine's potential in preventing 5-FU-induced mucositis by modulating gut microbiota and inflammation pathways.
Collapse
Affiliation(s)
- Ya-Ru Kuo
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
| | - Cheng-Hung Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
| | - Wei-Sheng Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
- Department of Food Science, National Quemoy University, Quemoy County, 89250, Taiwan
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung City, 40402, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung City, 41354, Taiwan
| |
Collapse
|
9
|
Cornejo-Pareja I, Amiar MR, Ocaña-Wilhelmi L, Soler-Humanes R, Arranz-Salas I, Garrido-Sánchez L, Gutiérrez-Repiso C, Tinahones FJ. Non-alcoholic fatty liver disease in patients with morbid obesity: the gut microbiota axis as a potential pathophysiology mechanism. J Gastroenterol 2024; 59:329-341. [PMID: 38265508 PMCID: PMC10959783 DOI: 10.1007/s00535-023-02075-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/27/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND/AIM Alterations in gut microbiota are associated with the pathogenesis of metabolic diseases, including metabolic-associated fatty liver disease (MAFLD). The aim of this study was to evaluate gut microbiota composition and functionality in patients with morbid obesity with different degrees of MAFLD, as assessed by biopsy. SUBJECTS/METHODS 110 patients with morbid obesity were evaluated by biopsy obtained during bariatric surgery for MAFLD. Stool samples were collected prior to surgery for microbiota analysis. RESULTS Gut microbiota from patients with steatosis and non-alcoholic steatohepatitis (NASH) were characterized by an enrichment in Enterobacteriaceae (an ethanol-producing bacteria), Acidaminococcus and Megasphaera and the depletion of Eggerthellaceae and Ruminococcaceae (SCFA-producing bacteria). MAFLD was also associated with enrichment of pathways related to proteinogenic amino acid degradation, succinate production, menaquinol-7 (K2-vitamin) biosynthesis, and saccharolytic and proteolytic fermentation. Basic histological hepatic alterations (steatosis, necroinflammatory activity, or fibrosis) were associated with specific changes in microbiota patterns. Overall, the core microbiome related to basic histological alterations in MAFLD showed an increase in Enterobacteriaceae and a decrease in Ruminococcaceae. Specifically, Escherichia coli was associated with steatosis and necroinflammatory activity, whilst Escherichia-shigella was associated with fibrosis and necroinflammatory activity. CONCLUSIONS We established a link between gut microbiota alterations and histological injury in liver diagnosis using biopsy. Harmful products such as ethanol or succinate may be involved in the pathogenesis and progression of MAFLD. Thus, these alterations in gut microbiota patterns and their possible metabolic pathways could add information to the classical predictors of MAFLD severity and suggest novel metabolic targets.
Collapse
Affiliation(s)
- Isabel Cornejo-Pareja
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Malaga University, Campus Teatinos S/N, 29010, Málaga, Spain.
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA), Virgen de la Victoria University Hospital, Malaga University, 2ª Planta, Campus Teatinos S/N, 29010, Málaga, Spain.
- Centro de Investigacion Biomedica en Red de la Fisiopatología de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29010, Málaga, Spain.
- Department of Medicine and Dermatology, Faculty of Medicine, University of Málaga, 29010, Málaga, Spain.
| | - Mohamed Reda Amiar
- Department of Medicine and Dermatology, Faculty of Medicine, University of Málaga, 29010, Málaga, Spain
- Department of Clinical Analysis Laboratory, Virgen de la Victoria Hospital, 29010, Málaga, Spain
| | - Luís Ocaña-Wilhelmi
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA), Virgen de la Victoria University Hospital, Malaga University, 2ª Planta, Campus Teatinos S/N, 29010, Málaga, Spain
- Department of General and Digestive Surgery, Virgen de la Victoria University Hospital, 29010, Málaga, Spain
- Department of Surgical Specialities, Biochemistry and Immunology, Faculty of Medicine, University of Málaga, 29010, Málaga, Spain
| | - Rocío Soler-Humanes
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA), Virgen de la Victoria University Hospital, Malaga University, 2ª Planta, Campus Teatinos S/N, 29010, Málaga, Spain
- Department of General and Digestive Surgery, Virgen de la Victoria University Hospital, 29010, Málaga, Spain
| | - Isabel Arranz-Salas
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA), Virgen de la Victoria University Hospital, Malaga University, 2ª Planta, Campus Teatinos S/N, 29010, Málaga, Spain
- Department of Human Physiology, Human Histology, Anatomical Pathology and Physical Education, Malaga University, 29010, Málaga, Spain
- Department of Anatomical Pathology, Virgen de la Victoria Hospital, Málaga, Spain
| | - Lourdes Garrido-Sánchez
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Malaga University, Campus Teatinos S/N, 29010, Málaga, Spain.
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA), Virgen de la Victoria University Hospital, Malaga University, 2ª Planta, Campus Teatinos S/N, 29010, Málaga, Spain.
- Centro de Investigacion Biomedica en Red de la Fisiopatología de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29010, Málaga, Spain.
| | - Carolina Gutiérrez-Repiso
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Malaga University, Campus Teatinos S/N, 29010, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA), Virgen de la Victoria University Hospital, Malaga University, 2ª Planta, Campus Teatinos S/N, 29010, Málaga, Spain
- Centro de Investigacion Biomedica en Red de la Fisiopatología de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29010, Málaga, Spain
| | - Francisco Jose Tinahones
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Malaga University, Campus Teatinos S/N, 29010, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA), Virgen de la Victoria University Hospital, Malaga University, 2ª Planta, Campus Teatinos S/N, 29010, Málaga, Spain
- Centro de Investigacion Biomedica en Red de la Fisiopatología de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29010, Málaga, Spain
- Department of Medicine and Dermatology, Faculty of Medicine, University of Málaga, 29010, Málaga, Spain
| |
Collapse
|
10
|
Lin Y, Manalili D, Khodabakhsh A, Cristescu SM. Real-Time Measurement of CH 4 in Human Breath Using a Compact CH 4/CO 2 Sensor. SENSORS (BASEL, SWITZERLAND) 2024; 24:1077. [PMID: 38400235 PMCID: PMC10893524 DOI: 10.3390/s24041077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024]
Abstract
The presence of an elevated amount of methane (CH4) in exhaled breath can be used as a non-invasive tool to monitor certain health conditions. A compact, inexpensive and transportable CH4 sensor is thus very interesting for this purpose. In addition, if the sensor is also able to simultaneously measure carbon dioxide (CO2), one can extract the end-tidal concentration of exhaled CH4. Here, we report on such a sensor based on a commercial detection module using tunable diode laser absorption spectroscopy. It was found that the measured CH4/CO2 values exhibit a strong interference with water vapor. Therefore, correction functions were experimentally identified and validated for both CO2 and CH4. A custom-built breath sampler was developed and tested with the sensor for real-time measurements of CH4 and CO2 in exhaled breath. As a result, the breath sensor demonstrated the capability of accurately measuring the exhaled CH4 and CO2 profiles in real-time. We obtained minimum detection limits of ~80 ppbv for CH4 and ~700 ppmv for CO2 in 1.5 s measurement time.
Collapse
Affiliation(s)
| | | | | | - Simona M. Cristescu
- Life Science Trace Detection Laboratory, Department of Analytical Chemistry and Chemometrics, Institute for Molecules and Materials, Radboud University, 6525 AJ Nijmegen, The Netherlands; (Y.L.); (D.M.); (A.K.)
| |
Collapse
|
11
|
Zhu SJ, Ding Z. Association between gut microbiota and seven gastrointestinal diseases: A Mendelian randomized study. J Gene Med 2024; 26:e3623. [PMID: 37957025 DOI: 10.1002/jgm.3623] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/25/2023] [Accepted: 10/16/2023] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Observational research has shed light on the ability of gut microbes to influence the onset and progression of gastrointestinal diseases. The causal relationships between specific gut microbiomes and various gastrointestinal conditions, however, remain unknown. METHODS We investigated the relationship between gut microbiota and seven specific gastrointestinal disorders using a robust two-sample Mendelian randomization (MR) approach. The inverse variance-weighted (IVW) method was used as the primary analysis tool in our study. Furthermore, we conducted multiple sensitivity analyses to strengthen the robustness of our findings and ensure the reliability of the IVW method. RESULTS Our research has discovered significant links between the composition of gut microbiota and a variety of gastrointestinal ailments. We found compelling links between 13 gut microbiota and fatty liver, four gut microbiota and cirrhosis, eight gut microbiota and hepatocellular carcinoma, four gut microbiota and cholelithiasis, 12 gut microbiota and acute pancreatitis, eight gut microbiota and chronic pancreatitis, and 11 gut microbiota and pancreatic cancer. These findings shed light on the intricate relationship between gut microbes and the emergence of these specific gastrointestinal conditions. CONCLUSIONS The findings of this extensive study not only validate the potential role of specific gut microbiota in gastrointestinal diseases, but also fill a critical gap in previous research. The discovery of these specific gut microbiota is a significant step forward because they may serve as novel and promising biomarkers for both the prevention and treatment of gastrointestinal conditions.
Collapse
Affiliation(s)
- Shuang Jing Zhu
- Hepatobiliary Surgery, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Zhen Ding
- Hepatobiliary Surgery, Chaohu Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
12
|
Zhai Q, Wu H, Zheng S, Zhong T, Du C, Yuan J, Peng J, Cai C, Li J. Association between gut microbiota and NAFLD/NASH: a bidirectional two-sample Mendelian randomization study. Front Cell Infect Microbiol 2023; 13:1294826. [PMID: 38106475 PMCID: PMC10722258 DOI: 10.3389/fcimb.2023.1294826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/09/2023] [Indexed: 12/19/2023] Open
Abstract
Background Recent studies have suggested a relationship between gut microbiota and non-alcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH). However, the nature and direction of this potential causal relationship are still unclear. This study used two-sample Mendelian randomization (MR) to clarify the potential causal links. Methods Summary-level Genome-Wide Association Studies (GWAS) statistical data for gut microbiota and NAFLD/NASH were obtained from MiBioGen and FinnGen respectively. The MR analyses were performed mainly using the inverse-variance weighted (IVW) method, with sensitivity analyses conducted to verify the robustness. Additionally, reverse MR analyses were performed to examine any potential reverse causal associations. Results Our analysis, primarily based on the IVW method, strongly supports the existence of causal relationships between four microbial taxa and NAFLD, and four taxa with NASH. Specifically, associations were observed between Enterobacteriales (P =0.04), Enterobacteriaceae (P =0.04), Lachnospiraceae UCG-004 (P =0.02), and Prevotella9 (P =0.04) and increased risk of NAFLD. Dorea (P =0.03) and Veillonella (P =0.04) could increase the risks of NASH while Oscillospira (P =0.04) and Ruminococcaceae UCG-013 (P=0.005) could decrease them. We also identified that NAFLD was found to potentially cause an increased abundance in Holdemania (P =0.007) and Ruminococcus2 (P =0.002). However, we found no evidence of reverse causation in the microbial taxa associations with NASH. Conclusion This study identified several specific gut microbiota that are causally related to NAFLD and NASH. Observations herein may provide promising theoretical groundwork for potential prevention and treatment strategies for NAFLD and its progression to NASH in future.
Collapse
Affiliation(s)
- Qilong Zhai
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongyu Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Siyuan Zheng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Zhong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Changjie Du
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiajun Yuan
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jialun Peng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Can Cai
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinzheng Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Mehramiz M, Porter T, O’Brien EK, Rainey-Smith SR, Laws SM. A Potential Role for Sirtuin-1 in Alzheimer's Disease: Reviewing the Biological and Environmental Evidence. J Alzheimers Dis Rep 2023; 7:823-843. [PMID: 37662612 PMCID: PMC10473168 DOI: 10.3233/adr-220088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 07/08/2023] [Indexed: 09/05/2023] Open
Abstract
Sirtuin-1 (Sirt1), encoded by the SIRT1 gene, is a conserved Nicotinamide adenine dinucleotide (NAD+) dependent deacetylase enzyme, considered as the master regulator of metabolism in humans. Sirt1 contributes to a wide range of biological pathways via several mechanisms influenced by lifestyle, such as diet and exercise. The importance of a healthy lifestyle is of relevance to highly prevalent modern chronic diseases, such as Alzheimer's disease (AD). There is growing evidence at multiple levels for a role of Sirt1/SIRT1 in AD pathological mechanisms. As such, this review will explore the relevance of Sirt1 to AD pathological mechanisms, by describing the involvement of Sirt1/SIRT1 in the development of AD pathological hallmarks, through its impact on the metabolism of amyloid-β and degradation of phosphorylated tau. We then explore the involvement of Sirt1/SIRT1 across different AD-relevant biological processes, including cholesterol metabolism, inflammation, circadian rhythm, and gut microbiome, before discussing the interplay between Sirt1 and AD-related lifestyle factors, such as diet, physical activity, and smoking, as well as depression, a common comorbidity. Genome-wide association studies have explored potential associations between SIRT1 and AD, as well as AD risk factors and co-morbidities. We summarize this evidence at the genetic level to highlight links between SIRT1 and AD, particularly associations with AD-related risk factors, such as heart disease. Finally, we review the current literature of potential interactions between SIRT1 genetic variants and lifestyle factors and how this evidence supports the need for further research to determine the relevance of these interactions with respect to AD and dementia.
Collapse
Affiliation(s)
- Mehrane Mehramiz
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Tenielle Porter
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| | - Eleanor K. O’Brien
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Stephanie R. Rainey-Smith
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia
- School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia
| | - Simon M. Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| |
Collapse
|
14
|
Vedel G, Triadó-Margarit X, Linares O, Moreno-Rojas JM, la Peña ED, García-Bocanegra I, Jiménez-Martín D, Carranza J, Casamayor EO. Exploring the potential links between gut microbiota composition and natural populations management in wild boar (Sus scrofa). Microbiol Res 2023; 274:127444. [PMID: 37421802 DOI: 10.1016/j.micres.2023.127444] [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: 12/03/2022] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/10/2023]
Abstract
We surveyed wild boar (Sus scrofa) populations using 16S rRNA gene analysis of the gut microbiota in fresh faeces taken from 88 animals hunted in 16 hunting estates. The wild boar is a very convenient model system to explore how environmental factors including game management, food availability, disease prevalence, and behaviour may affect different biological components of wild individuals with potential implications in management and conservation. We tested the hypotheses that diet (according to stable carbon isotopes analyses), gender (i.e., animal behaviour studying males and females), and both health (analyses of serum samples to detect exposure to several diseases) and form statutes (i.e., thoracic circumference in adults) are reflected in changes in the intestinal microbiota. We focused on a gut functional biomarker index combining Oscillospiraceae and Ruminococcaceae vs. Enterobacteriaceae. We found that gender and the estate (population) were explanatory variables (c.a. 28% of the variance), albeit a high degree of overlapping among individuals was observed. The individuals with higher abundance of Enterobacteriaceae showed a gut microbiota with low diversity, mostly in males. Significant statistical differences for thoracic circumference were not found between males and females. Interestingly, the thoracic circumference was significantly and inversely related to the relative abundance of Enterobacteriaceae in males. Overall, we found that diet, gender, and form status were major factors that could be related to the composition and diversity of the gut microbiota. A high variability was observed in the biomarker index for populations with natural diet (rich in C3 plants). Although, we noticed a marginally significant negative trend between the index (higher abundance of Enterobacteriaceae) and the continuous feeding of C4 plants (i.e., supplementary maize) in the diet of males. This result suggests that continuous artificial feeding in hunting estates could be one of the factors negatively influencing the gut microbiota and the form status of wild boars that deserves further investigations.
Collapse
Affiliation(s)
- Giovanni Vedel
- Wildlife Research Unit, University of Cordoba (UIRCP-UCO), 14071 Córdoba, Spain
| | - Xavier Triadó-Margarit
- Ecology of the Global Microbiome, Centre of Advanced Studies of Blanes-Spanish Council for Research (CEAB-CSIC), Accés Cala St Francesc, 14, E-17300 Blanes, Spain
| | - Olmo Linares
- Wildlife Research Unit, University of Cordoba (UIRCP-UCO), 14071 Córdoba, Spain
| | - José Manuel Moreno-Rojas
- Department of Food Science and Health, Andalusian Institute of Agricultural and Fisheries Research and Training (IFAPA), Alameda del Obispo, Avda. Menéndez Pidal, s/n, 14071 Córdoba, Spain
| | - Eva de la Peña
- Wildlife Research Unit, University of Cordoba (UIRCP-UCO), 14071 Córdoba, Spain; IREC National Wildlife Research Institute (CSIC-UCLM-JCCM), Ciudad Real, Spain
| | - Ignacio García-Bocanegra
- Department of Animal Health, Animal Health and Zoonosis Research Group (GISAZ), UIC Zoonoses and Emreging Diseases ENZOEM, University of Cordoba, Cordoba, Spain; CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Spain
| | - Débora Jiménez-Martín
- Department of Animal Health, Animal Health and Zoonosis Research Group (GISAZ), UIC Zoonoses and Emreging Diseases ENZOEM, University of Cordoba, Cordoba, Spain
| | - Juan Carranza
- Wildlife Research Unit, University of Cordoba (UIRCP-UCO), 14071 Córdoba, Spain
| | - Emilio O Casamayor
- Ecology of the Global Microbiome, Centre of Advanced Studies of Blanes-Spanish Council for Research (CEAB-CSIC), Accés Cala St Francesc, 14, E-17300 Blanes, Spain.
| |
Collapse
|
15
|
Zaccaria E, Klaassen T, Alleleyn AME, Boekhorst J, Smokvina T, Kleerebezem M, Troost FJ. Endogenous small intestinal microbiome determinants of transient colonisation efficiency by bacteria from fermented dairy products: a randomised controlled trial. MICROBIOME 2023; 11:43. [PMID: 36879297 PMCID: PMC9990280 DOI: 10.1186/s40168-023-01491-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND The effects of fermented food consumption on the small intestine microbiome and its role on host homeostasis are largely uncharacterised as our knowledge on intestinal microbiota relies mainly on faecal samples analysis. We investigated changes in small intestinal microbial composition and functionality, short chain fatty acid (SCFA) profiles, and on gastro-intestinal (GI) permeability in ileostomy subjects upon the consumption of fermented milk products. RESULTS We report the results from a randomised, cross-over, explorative study where 16 ileostomy subjects underwent 3, 2-week intervention periods. In each period, they consumed either milk fermented by Lacticaseibacillus rhamnosus CNCM I-3690, or milk fermented by Streptococcus thermophilus CNCM I-1630 and Lactobacillus delbrueckii subsp. bulgaricus CNCM I-1519, or a chemically acidified milk (placebo) daily. We performed metataxonomic, metatranscriptomic analysis, and SCFA profiling of ileostomy effluents as well as a sugar permeability test to investigate the microbiome impact of these interventions and their potential effect on mucosal barrier function. Consumption of the intervention products impacted the overall small intestinal microbiome composition and functionality, mainly due to the introduction of the product-derived bacteria that reach in several samples 50% of the total microbial community. The interventions did not affect the SCFA levels in ileostoma effluent, or gastro-intestinal permeability and the effects on the endogenous microbial community were negligible. The impact on microbiome composition was highly personalised, and we identified the poorly characterised bacterial family, Peptostreptococcaceae, to be positively associated with a low abundance of the ingested bacteria. Activity profiling of the microbiota revealed that carbon- versus amino acid-derived energy metabolism of the endogenous microbiome could be responsible for the individual-specific intervention effects on the small intestine microbiome composition and function, reflected also on urine microbial metabolites generated through proteolytic fermentation. CONCLUSIONS The ingested bacteria are the main drivers of the intervention effect on the small intestinal microbiota composition. Their transient abundance level is highly personalised and influenced by the energy metabolism of the ecosystem that is reflected by its microbial composition ( http://www. CLINICALTRIALS gov , ID NCT NCT02920294). Video Abstract.
Collapse
Affiliation(s)
- Edoardo Zaccaria
- Host Microbe Interactomics Group, Wageningen University & Research, De Elst 1, 6708WD, Wageningen, The Netherlands
- Food Innovation and Health, Center for Healthy Eating and Food Innovation, Maastricht University, Venlo, 5911AA, The Netherlands
| | - Tim Klaassen
- Food Innovation and Health, Center for Healthy Eating and Food Innovation, Maastricht University, Venlo, 5911AA, The Netherlands
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 5800, 6202AZ, Maastricht, The Netherlands
| | - Annick M E Alleleyn
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 5800, 6202AZ, Maastricht, The Netherlands
| | - Jos Boekhorst
- Host Microbe Interactomics Group, Wageningen University & Research, De Elst 1, 6708WD, Wageningen, The Netherlands
| | - Tamara Smokvina
- Danone Nutricia Research, Av. De la Vauve, 91767, Palaiseau, France
| | - Michiel Kleerebezem
- Host Microbe Interactomics Group, Wageningen University & Research, De Elst 1, 6708WD, Wageningen, The Netherlands.
| | - Freddy J Troost
- Food Innovation and Health, Center for Healthy Eating and Food Innovation, Maastricht University, Venlo, 5911AA, The Netherlands
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 5800, 6202AZ, Maastricht, The Netherlands
| |
Collapse
|
16
|
Effect of a Multistrain Probiotic on Feline Gut Health through the Fecal Microbiota and Its Metabolite SCFAs. Metabolites 2023; 13:metabo13020228. [PMID: 36837847 PMCID: PMC9962843 DOI: 10.3390/metabo13020228] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
With the increasing awareness of raising pets following scientific methods, people are becoming increasingly more interested in the nutrition and health of pets, especially their intestinal health, which has become a research hotspot. Both Saccharomyces boulardii and Pediococcus acidilactici are probiotics with strong probiotic properties that can maintain the balance of intestinal flora. However, the role of Saccharomyces boulardii and Pediococcus acidilactici in felines has not been comprehensively studied to date. The aim of this study is to investigate the effect of multistrain probiotics consisting of Saccharomyces boulardii and Pediococcus acidilactici on the gut health of felines by modulating gut microbes and the production of metabolite SCFAs. The results show that the multistrain probiotic did not alter the intestinal microbial diversity and structure of short-haired domestic cats, promoted the colonization of beneficial bacteria, increased the levels of microbiota-derived SCFAs and fecal antioxidants, and reduced the levels of fecal inflammatory markers. In conclusion, the use of a multistrain probiotic in healthy, short-haired domestic cats can promote gut health by modulating gut microbes, improving microbiota-derived SCFA production, reducing inflammatory conditions, and improving antioxidant status. These results provide new insights for further exploration of the role of probiotics in the gut microbiome of cats.
Collapse
|
17
|
Xu J, Xu J, Shi T, Zhang Y, Chen F, Yang C, Guo X, Liu G, Shao D, Leong KW, Nie G. Probiotic-Inspired Nanomedicine Restores Intestinal Homeostasis in Colitis by Regulating Redox Balance, Immune Responses, and the Gut Microbiome. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207890. [PMID: 36341495 DOI: 10.1002/adma.202207890] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/26/2022] [Indexed: 06/16/2023]
Abstract
Microbiota-based therapeutics offer innovative strategies to treat inflammatory bowel diseases (IBDs). However, the poor clinical outcome so far and the limited flexibility of the bacterial approach call for improvement. Inspired by the health benefits of probiotics in alleviating symptoms of bowel diseases, bioartificial probiotics are designed to restore the intestinal microenvironment in colitis by regulating redox balance, immune responses, and the gut microbiome. The bioartificial probiotic comprises two components: an E. coli Nissle 1917-derived membrane (EM) as the surface and the biodegradable diselenide-bridged mesoporous silica nanoparticles (SeM) as the core. When orally administered, the probiotic-inspired nanomedicine (SeM@EM) adheres strongly to the mucus layer and restored intestinal redox balance and immune regulation homeostasis in a murine model of acute colitis induced by dextran sodium sulfate. In addition, the respective properties of the EM and SeM synergistically alter the gut microbiome to a favorable state by increasing the bacterial diversity and shifting the microbiome profile to an anti-inflammatory phenotype. This work suggests a safe and effective nanomedicine that can restore intestinal homeostasis for IBDs therapy.
Collapse
Affiliation(s)
- Jiaqi Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tongfei Shi
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Yinlong Zhang
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fangman Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 510006, China
| | - Chao Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Xinjing Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dan Shao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
18
|
Salarieh N, Emami Meibodi A, Alipour S, Azimirad M, Looha MA, Asadzadeh Aghdaei H, Yadegar A, Shahrokh S, Zali MR. Characterization of the mucosal microbiota in patients with nodular lymphoid hyperplasia with concurrent irritable bowel syndrome compared to healthy controls. Mol Biol Rep 2023; 50:145-155. [PMID: 36315327 DOI: 10.1007/s11033-022-07974-1] [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: 07/03/2022] [Accepted: 09/21/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND Nodular lymphoid hyperplasia (NLH) is known as a lymphoproliferative lesion in which multiple small nodules appear on the intestinal wall. It has been documented that patients who struggle with irritable bowel syndrome (IBS) are at greater risk of developing NLH. Here, we aimed to investigate the previously reported pathogens and the abundance of a selection of mucosal microbiota in IBS + NLH patients compared to IBS, and healthy controls. METHODS AND RESULTS Terminal ileum biopsies were collected from 37 IBS + NLH, 37 IBS, and 29 healthy controls. Bacterial culture and PCR was performed to detect the presence of pathogens in biopsies. A qPCR assay was applied to assess the abundance of a selection of bacterial taxa. Totally, five bacterial isolates including two enteropathogenic and one enteroaggregative Escherichia coli (EPEC, EAEC), one enterotoxigenic Staphylococcus aureus (SEA), and one Yersinia enterocolitica strains were detected among the IBS + NLH cases. The relative abundance of Bacteroidetes and Streptococcus spp. in IBS + NLH patients was significantly less than IBS and healthy controls. Firmicutes, Pseudomonas spp., Haemophilus spp., and Campylobacter spp. were notably more abundant in IBS + NLH than in IBS patients. The abundance of Verrucomicrobia was higher in NLH + IBS than in healthy controls. Actinobacteria was also significantly more abundant among NLH + IBS patients than the controls. CONCLUSION Our results demonstrated that mucosal microbiota composition in NLH + IBS patients slightly differs from that of IBS patients and healthy controls. Further research using large-scale cohorts are needed to enhance current understanding of the contribution of the mucosal microbiota to NLH pathogenesis with concurrent IBS.
Collapse
Affiliation(s)
- Naghmeh Salarieh
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Armitasadat Emami Meibodi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Alipour
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Azimirad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Azizmohammad Looha
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Shabnam Shahrokh
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Zaccaria E, Klaassen T, Alleleyn AM, Boekhorst J, Chervaux C, Smokvina T, Troost FJ, Kleerebezem M. L. rhamnosus CNCM I-3690 survival, adaptation, and small bowel microbiome impact in human. Gut Microbes 2023; 15:2244720. [PMID: 37589280 PMCID: PMC10438856 DOI: 10.1080/19490976.2023.2244720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/05/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023] Open
Abstract
Fermented foods and beverages are a significant source of dietary bacteria that enter the gastrointestinal (GI) tract. However, little is known about how these microbes survive and adapt to the small intestinal environment. Colony-forming units (CFU) enumeration and viability qPCR of Lacticaseibacillus rhamnosus CNCM I-3690 in the ileal effluent of 10 ileostomy subjects during 12-h post consumption of a dairy product fermented with this strain demonstrated the high level of survival of this strain during human small intestine passage. Metatranscriptome analyses revealed the in situ transcriptome of L. rhamnosus in the small intestine, which was contrasted with transcriptome data obtained from in vitro cultivation. These comparative analyses revealed substantial metabolic adaptations of L. rhamnosus during small intestine transit, including adjustments of carbohydrate metabolism, surface-protein expression, and translation machinery. The prominent presence of L. rhamnosus in the effluent samples did not elicit an appreciable effect on the composition of the endogenous small intestine microbiome, but significantly altered the ecosystem's overall activity profile, particularly of pathways associated with carbohydrate metabolism. Strikingly, two of the previously recognized gut-brain metabolic modules expressed in situ by L. rhamnosus (inositol degradation and glutamate synthesis II) are among the most dominantly enriched activities in the ecosystem's activity profile. This study establishes the survival capacity of L. rhamnosus in the human small intestine and highlights its functional adjustment in situ, which we postulate to play a role in the probiotic effects associated with this strain.
Collapse
Affiliation(s)
- Edoardo Zaccaria
- Host Microbe Interactomics Group, Wageningen University & Research, Wageningen, The Netherlands
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Tim Klaassen
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Venlo, The Netherlands
| | - Annick M.E. Alleleyn
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Venlo, The Netherlands
| | - Jos Boekhorst
- Host Microbe Interactomics Group, Wageningen University & Research, Wageningen, The Netherlands
| | | | - Tamara Smokvina
- Danone Nutricia Research, Centre Daniel Carasso, Palaiseau, France
| | - Freddy J. Troost
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Michiel Kleerebezem
- Host Microbe Interactomics Group, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
20
|
Peckmezian T, Garcia-Larsen V, Wilkins K, Mosli RH, BinDhim NF, John GK, Yasir M, Azhar EI, Mullin GE, Alqahtani SA. Microbiome-Targeted Therapies as an Adjunct to Traditional Weight Loss Interventions: A Systematic Review and Meta-Analysis. Diabetes Metab Syndr Obes 2022; 15:3777-3798. [PMID: 36530587 PMCID: PMC9753565 DOI: 10.2147/dmso.s378396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022] Open
Abstract
Objective This study evaluated the effect of microbiome-targeted therapies (pre-, pro-, and synbiotics) on weight loss and other anthropometric outcomes when delivered as an adjunct to traditional weight loss interventions in overweight and obese adults. Methods A systematic review of three databases (Medline [PubMed], Embase, and the Cochrane Central Register of Controlled Trials) was performed to identify randomized controlled trials published between January 1, 2010 and December 31, 2020, that evaluated anthropometric outcomes following microbiome-targeted supplements in combination with dietary or dietary and exercise interventions. The pooled mean difference (MD) between treatment and control groups was calculated using a random effects model. Results Twenty-one trials with 1233 adult participants (76.4% female) with overweight or obesity were included. Separate meta-analyses were conducted for probiotics (n=11 trials) and synbiotics (n=10 trials) on each anthropometric outcome; prebiotics were excluded as only a single study was found. Patient characteristics and methodologies varied widely between studies. All studies incorporated some degree of caloric restriction, while only six studies included recommendations for adjunct exercise. Compared with dietary or dietary and exercise interventions only, probiotics resulted in reductions in body weight (MD: -0.73 kg; 95% confidence interval [CI]: -1.02 to -0.44, p < 0.001), fat mass (MD: -0.61 kg; 95% CI: -0.77 to -0.45; p<0.001) and waist circumference (MD: -0.53 cm; 95% CI: -0.99 to -0.07, p=0.024) while synbiotics resulted in reductions in fat mass (MD: -1.53 kg; 95% CI: -2.95 to -0.12, p=0.034) and waist circumference (MD: -1.31 cm; 95% CI: -2.05 to -0.57, p<0.001). Conclusion This analysis indicates that microbiome-targeted supplements may enhance weight loss and other obesity outcomes in adults when delivered as an adjunct to dietary or dietary and exercise interventions. Personalized therapy to include microbiome-targeted supplements may help to optimize weight loss in overweight and obese individuals.
Collapse
Affiliation(s)
| | - Vanessa Garcia-Larsen
- Program in Human Nutrition, Department of International Health, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kayla Wilkins
- Environmental GeoScience Research Group, Trent University, Peterborough, ON, Canada
| | - Rana H Mosli
- Clinical Nutrition Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nasser F BinDhim
- Sharik Association for Health Research, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Saudi Food and Drug Authority, Riyadh, Saudi Arabia
| | - George Kunnackal John
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Easton, MD, USA
| | - Muhammad Yasir
- Special Infectious Agents Unit – BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Esam Ibraheem Azhar
- Special Infectious Agents Unit – BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Gerard E Mullin
- Liver Transplant Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Saleh A Alqahtani
- Liver Transplant Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
- Division of Gastroenterology & Hepatology, John Hopkins University, Baltimore, MD, USA
| |
Collapse
|
21
|
Lao L, Yang G, Zhang A, Liu L, Guo Y, Lian L, Pan D, Wu Z. Anti-inflammation and gut microbiota regulation properties of fatty acids derived from fermented milk in mice with dextran sulfate sodium-induced colitis. J Dairy Sci 2022; 105:7865-7877. [PMID: 36055856 DOI: 10.3168/jds.2022-21877] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/01/2022] [Indexed: 01/03/2024]
Abstract
The by-products of milk fermentation by lactic acid bacteria provide potential health benefits to the balance of host intestinal microflora. In this study, the anti-inflammatory properties of fatty acids from monoculture-strain (Lactiplantibacillusplantarum A3) and multiple-strain (Streptococcus thermophilus, Lactobacillus bulgaricus, and L. plantarum A3 1:1:2) fermented milk were evaluated in a mouse model of dextran sulfate sodium-induced colitis, and the gut microbiota regulation properties of the fatty acids were also investigated. Results showed that fatty acids can attenuate the inflammatory response by inhibiting the expression of inflammatory factors IL-6 and tumor necrosis factor-α, and blocking the phosphorylation of the JNK in MAPK signal pathway. In addition, the relative abundance of the taxa Akkermansia and Lactobacillus were both enriched after the fatty acid intervention. This finding suggests that fatty acids from the milk fermentation with mixed lactic acid bacteria starters can reduce the severity of dextran sulfate sodium-induced colitis and enhance the abundance of the probiotics in the mice intestinal tract.
Collapse
Affiliation(s)
- Lifeng Lao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315211, Zhejiang, P. R. China
| | - Guo Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315211, Zhejiang, P. R. China
| | - Ao Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315211, Zhejiang, P. R. China
| | - Lianliang Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315211, Zhejiang, P. R. China
| | - Yuxing Guo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Liwei Lian
- Ningbo Dairy Group, Ningbo, 315211, Zhejiang, China
| | - Daodong Pan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315211, Zhejiang, P. R. China
| | - Zhen Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315211, Zhejiang, P. R. China.
| |
Collapse
|
22
|
Wang Y, Li L, Zhao X, Sui S, Wang Q, Shi G, Xu H, Zhang X, He Y, Gu J. Intestinal Microflora Changes in Patients with Mild Alzheimer's Disease in a Chinese Cohort. J Alzheimers Dis 2022; 88:563-575. [PMID: 35662119 DOI: 10.3233/jad-220076] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Understanding the relationship between Alzheimer's disease (AD) and intestinal flora is still a major scientific topic that continues to advance. OBJECTIVE To determine characterized changes in the intestinal microbe community of patients with mild AD. METHODS Comparison of the 16S ribosomal RNA (rRNA) high-throughput sequencing data was obtained from the Illumina MiSeq platform of fecal microorganisms of the patients and healthy controls (HC) which were selected from cohabiting caregivers of AD patients to exclude environmental and dietary factors. RESULTS We found that the abundance of several bacteria taxa in AD patients was different from that in HC at the genus level, such as Anaerostipes, Mitsuokella, Prevotella, Bosea, Fusobacterium, Anaerotruncus, Clostridium, and Coprobacillus. Interestingly, the abundance of Akkermansia, an emerging probiotic, increased significantly in the AD group compared with that in the HC group. Meanwhile, the quantity of traditional probiotic Bifidobacteria of the AD group also rose. CONCLUSION These alterations in fecal microbiome of the AD group indicate that patients with mild AD have unique gut microbial characteristics. These specific AD-associated intestinal microbes could serve as novel potential targets for early intervention of AD.
Collapse
Affiliation(s)
- Yilin Wang
- College of Biological Science and Technology, University of Jinan, Jinan, China
| | - Lei Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, China
| | - Xiaodong Zhao
- College of Biological Science and Technology, University of Jinan, Jinan, China
| | - Shaomei Sui
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, China
| | - Qi Wang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, China
| | - Guizhi Shi
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Huilian Xu
- College of Biological Science and Technology, University of Jinan, Jinan, China
| | - Xiujun Zhang
- College of Biological Science and Technology, University of Jinan, Jinan, China
| | - Yan He
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, China
| | - Jinsong Gu
- College of Biological Science and Technology, University of Jinan, Jinan, China
| |
Collapse
|
23
|
Manzoor R, Ahmed W, Afify N, Memon M, Yasin M, Memon H, Rustom M, Al Akeel M, Alhajri N. Trust Your Gut: The Association of Gut Microbiota and Liver Disease. Microorganisms 2022; 10:1045. [PMID: 35630487 PMCID: PMC9146349 DOI: 10.3390/microorganisms10051045] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/08/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota composition is important for nutrient metabolism, mucosal barrier function, immunomodulation, and defense against pathogens. Alterations in the gut microbiome can disturb the gut ecosystem. These changes may lead to the loss of beneficial bacteria or an increase in potentially pathogenic bacteria. Furthermore, these have been shown to contribute to the pathophysiology of gastrointestinal and extra-intestinal diseases. Pathologies of the liver, such as non-alcoholic liver disease, alcoholic liver disease, cirrhosis, hepatocellular carcinoma, autoimmune hepatitis, viral hepatitis, and primary sclerosing cholangitis have all been linked to changes in the gut microbiome composition. There is substantial evidence that links gut dysbiosis to the progression and complications of these pathologies. This review article aimed to describe the changes seen in the gut microbiome in liver diseases and the association between gut dysbiosis and liver disease, and finally, explore treatment options that may improve gut dysbiosis in patients with liver disease.
Collapse
Affiliation(s)
- Ridda Manzoor
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (R.M.); (W.A.); (N.A.); (M.M.); (M.Y.); (H.M.); (M.R.)
| | - Weshah Ahmed
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (R.M.); (W.A.); (N.A.); (M.M.); (M.Y.); (H.M.); (M.R.)
| | - Nariman Afify
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (R.M.); (W.A.); (N.A.); (M.M.); (M.Y.); (H.M.); (M.R.)
| | - Mashal Memon
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (R.M.); (W.A.); (N.A.); (M.M.); (M.Y.); (H.M.); (M.R.)
| | - Maryam Yasin
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (R.M.); (W.A.); (N.A.); (M.M.); (M.Y.); (H.M.); (M.R.)
| | - Hamda Memon
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (R.M.); (W.A.); (N.A.); (M.M.); (M.Y.); (H.M.); (M.R.)
| | - Mohammad Rustom
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates; (R.M.); (W.A.); (N.A.); (M.M.); (M.Y.); (H.M.); (M.R.)
| | - Mohannad Al Akeel
- Division of Family Medicine, Department of Health, Abu Dhabi P.O. Box 5674, United Arab Emirates;
| | - Noora Alhajri
- Department of Medicine, Sheikh Shakhbout Medical City (SSMC), Abu Dhabi P.O. Box 11001, United Arab Emirates
| |
Collapse
|
24
|
A Promising Insight: The Potential Influence and Therapeutic Value of the Gut Microbiota in GI GVHD. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2124627. [PMID: 35571252 PMCID: PMC9098338 DOI: 10.1155/2022/2124627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HSCT) is a reconstruction process of hematopoietic and immune functions that can be curative in patients with hematologic malignancies, but it carries risks of graft-versus-host disease (GVHD), thrombotic microangiopathy (TMA), Epstein–Barr virus (EBV) infection, cytomegalovirus infection, secondary hemophagocytic lymphohistiocytosis (sHLH), macrophage activation syndrome (MAS), bronchiolitis obliterans, and posterior reversible encephalopathy syndrome (PRES). Gastrointestinal graft-versus-host disease (GI GVHD), a common complication of allo-HSCT, is one of the leading causes of transplant-related death because of its high treatment difficulty, which is affected by preimplantation, antibiotic use, dietary changes, and intestinal inflammation. At present, human trials and animal studies have proven that a decrease in intestinal bacterial diversity is associated with the occurrence of GI GVHD. Metabolites produced by intestinal bacteria, such as lipopolysaccharides, short-chain fatty acids, and secondary bile acids, can affect the development of GVHD through direct or indirect interactions with immune cells. The targeted damage of GVHD on intestinal stem cells (ISCs) and Paneth cells results in intestinal dysbiosis or dysbacteriosis. Based on the effect of microbiota metabolites on the gastrointestinal tract, the clinical treatment of GI GVHD can be further optimized. In this review, we describe the mechanisms of GI GVHD and the damage it causes to intestinal cells and we summarize recent studies on the relationship between intestinal microbiota and GVHD in the gastrointestinal tract, highlighting the role of intestinal microbiota metabolites in GI GVHD. We hope to elucidate strategies for immunomodulatory combined microbiota targeting in the clinical treatment of GI GVHD.
Collapse
|
25
|
Khan I, Wei J, Li A, Liu Z, Yang P, Jing Y, Chen X, Zhao T, Bai Y, Zha L, Li C, Ullah N, Che T, Zhang C. Lactobacillus plantarum strains attenuated DSS-induced colitis in mice by modulating the gut microbiota and immune response. Int Microbiol 2022; 25:587-603. [PMID: 35414032 DOI: 10.1007/s10123-022-00243-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 03/19/2022] [Accepted: 03/23/2022] [Indexed: 02/07/2023]
Abstract
Gut microbiota has become a new therapeutic target in the treatment of inflammatory Bowel Disease (IBD). Probiotics are known for their beneficial effects and have shown good efficacy in the clinical treatment of IBD and animal models of colitis. However, how these probiotics contribute to the amelioration of IBD is largely unknown. In the current study, the DSS-induced mouse colitis model was treated with oral administration of Lactobacillus plantarum strains to investigate their effects on colitis. The results indicated that the L. plantarum strains improved dysbiosis and enhanced the abundance of beneficial bacteria related to short-chain fatty acids (SCFAs) production. Moreover, L. plantarum strains decreased the level of pro-inflammatory cytokines, i.e., IL-17A, IL-17F, IL-6, IL-22, and TNF-α and increased the level of anti-inflammatory cytokines, i.e., TGF-β, IL-10. Our result suggests that L. plantarum strains possess probiotic effects and can ameliorate DSS colitis in mice by modulating the resident gut microbiota and immune response.
Collapse
Affiliation(s)
- Israr Khan
- School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.,Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou, 730000, China
| | - Junshu Wei
- School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.,Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou, 730000, China
| | - Anping Li
- Gansu Institute of Drug Control, Lanzhou, 730030, China
| | - Zhirong Liu
- Gansu Institute of Drug Control, Lanzhou, 730030, China
| | - Pingrong Yang
- Gansu Institute of Drug Control, Lanzhou, 730030, China
| | - Yaping Jing
- School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.,Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou, 730000, China
| | - Xinjun Chen
- School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.,Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou, 730000, China
| | - Tang Zhao
- School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.,Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou, 730000, China
| | - Yanrui Bai
- School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.,Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou, 730000, China
| | - Lajia Zha
- School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.,Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou, 730000, China
| | - Chenhui Li
- School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.,Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou, 730000, China
| | - Naeem Ullah
- School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.,Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China.,Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou, 730000, China
| | - Tuanjie Che
- Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou, 730000, China
| | - Chunjiang Zhang
- School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China. .,Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, 730000, China. .,Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China. .,Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou, 730000, China.
| |
Collapse
|
26
|
Abuqwider J, Altamimi M, Mauriello G. Limosilactobacillus reuteri in Health and Disease. Microorganisms 2022; 10:microorganisms10030522. [PMID: 35336098 PMCID: PMC8953724 DOI: 10.3390/microorganisms10030522] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 02/01/2023] Open
Abstract
Limosilactobacillus reuteri is a microorganism with valuable probiotic qualities that has been widely employed in humans to promote health. It is a well-studied probiotic bacterium that exerts beneficial health effects due to several metabolic mechanisms that enhance the production of anti-inflammatory cytochines and modulate the gut microbiota by the production of antimicrobial molecules, including reuterin. This review provides an overview of the data that support the role of probiotic properties, and the antimicrobial and immunomodulatory effects of some L. reuteri strains in relation to their metabolite production profile on the amelioration of many diseases and disorders. Although the results discussed in this paper are strain dependent, they show that L. reuteri, by different mechanisms and various metabolites, may control body weight and obesity, improve insulin sensitivity and glucose homeostasis, increase gut integrity and immunomodulation, and attenuate hepatic disorders. Gut microbiota modulation by ingesting probiotic L. reuteri strains could be a promising preventative and therapeutic approach against many diseases and disorders.
Collapse
Affiliation(s)
- Jumana Abuqwider
- Department of Agricultural Science, University of Naples Federico II, 80049 Naples, Italy;
| | - Mohammad Altamimi
- Department of Nutrition and Food Technology, Faculty of Agriculture and Veterinary Medicine, An-Najah National University, Nablus P.O. Box 7, Palestine;
| | - Gianluigi Mauriello
- Department of Agricultural Science, University of Naples Federico II, 80049 Naples, Italy;
- Correspondence: ; Tel.: +39-081-2539452
| |
Collapse
|
27
|
Wen H, Tian H, Liu C, Zhang X, Peng Y, Yang X, Chen F, Li J. Metformin and cyanidin 3- O-galactoside from Aronia melanocarpa synergistically alleviate cognitive impairment in SAMP8 mice. Food Funct 2021; 12:10994-11008. [PMID: 34657937 DOI: 10.1039/d1fo02122b] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cyanidin 3-O-galactoside (Cy3Gal) from Aronia melanocarpa has been reported to alleviate cognitive impairment. Metformin for preventing the neurodegenerative disease is attracting increasing attention. However, the neuroprotective and metabolic health promoting both of their effects are not clear. We chose the senescence accelerated mouse prone 8 (SAMP8) as a model of spontaneous learning and memory impairment. This study aimed to investigate the synergistic neuroprotective effect of metformin and Cy3Gal by behavioral and histopathological assays and metabolite analysis in SAMP8 mice. The SAMR1 mice were the normal group, and the SAMP8 mice were divided into five groups, including the SAMP8 model group, the donepezil (1 mg kg-1, ig) group, the metformin (100 mg kg-1, ig) group, the Cy3Gal (25 mg kg-1, ig) group, and the combination of metformin plus Cy3Gal (Met + Cy3Gal, 100 mg kg-1, 25 mg kg-1, ig) group. The behavior experiments showed that the SAMP8 mice treated with metformin and Cy3Gal showed improved spatial learning and memory compared to the SAMP8 model group. The number of neurons in the Met + Cy3Gal group was significantly higher than that in the SAMP8 group and the Met + Cy3Gal group showed significantly reduced Aβ aggregation in the brain, which was elevated in SAMP8 mice. Compared with SAMP8 mice, the Met + Cy3Gal group showed decreased indole, methyl esters and ketones and increased short-chain fatty acids and alcohols in feces and urine by regulating the fatty acid biosynthesis and degradation. This study confirmed the neuroprotective effects of coadministration of metformin and cyanidin 3-O-galactoside in the SAMP8 mice, and suggested its positive effect on postponing the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Haichao Wen
- College of Food Science & Nutritional Engineering, China Agricultural University, No. 17 Tsinghua Dong Road, Beijing 100083, China. .,Institute of Nutrition and Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Hehe Tian
- College of Food Science & Nutritional Engineering, China Agricultural University, No. 17 Tsinghua Dong Road, Beijing 100083, China.
| | - Chang Liu
- College of Food Science & Nutritional Engineering, China Agricultural University, No. 17 Tsinghua Dong Road, Beijing 100083, China.
| | - Xiaoxu Zhang
- College of Food Science & Nutritional Engineering, China Agricultural University, No. 17 Tsinghua Dong Road, Beijing 100083, China.
| | - Yao Peng
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Xinquan Yang
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Feng Chen
- Department of Food, Nutrition and Packaging Science, Clemson University, Clemson, SC 29634, USA.
| | - Jingming Li
- College of Food Science & Nutritional Engineering, China Agricultural University, No. 17 Tsinghua Dong Road, Beijing 100083, China.
| |
Collapse
|
28
|
Jurek L, Sevil M, Jay A, Schröder C, Baghdadli A, Héry-Arnaud G, Geoffray MM. Is there a dysbiosis in individuals with a neurodevelopmental disorder compared to controls over the course of development? A systematic review. Eur Child Adolesc Psychiatry 2021; 30:1671-1694. [PMID: 32385698 DOI: 10.1007/s00787-020-01544-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/24/2020] [Indexed: 12/16/2022]
Abstract
Many scientific papers reported that an unbalanced gut microbiota could lead to or worsen neurodevelopmental disorders (NDD). A dysbiosis may then be observed in the course of development and mark a dysfunction within what is called the gut-brain axis. The aim of this systematic review is to investigate potential evidence of dysbiosis in children and young adults with NDD compared to controls. Using the PRISMA guidelines we systematically reviewed studies that compared the gut microbiota in NDD participants (with an age inferior to thirty) to the gut microbiota of controls, regardless of the data analysis methods used. The MEDLINE, Scopus and PsycINFO databases were searched up to September 2018. 31 studies with a total sample size of 3002 ASD (Autism Spectrum Disorder) and 84 ADHD (Attention Deficit Hyperactivity Disorder) participants were included in this systematic review. Independent data extraction and quality assessment were conducted. The quality of the studies was rated from low to high. Population characterization and experimental methods were highly heterogeneous in terms of the data available, selection of criteria, and dysbiosis measurement. A dysbiosis was reported in 28 studies in terms of either diversity, bacterial composition or metabolome dysfunction. Due to heterogeneity, a quantitative synthesis was not applicable. In this paper, we discuss the different biases to understand the complexity of microbiota and neurodevelopmental disorders to provide leads for future cohort studies looking to answer the questions raised by the trillions of microorganisms that inhabit key body niches.
Collapse
Affiliation(s)
- Lucie Jurek
- Child and Adolescent Psychiatry Departement, Center for Assessment and Diagnostic of Autism, Le Vinatier Hospital, Bron, France.
- Health Services and Performance Research EA7425, Claude Bernard Lyon 1 University (CBL1), Lyon, France.
| | - Marine Sevil
- Child and Adolescent Psychiatry Departement, Center for Assessment and Diagnostic of Autism, Le Vinatier Hospital, Bron, France
| | - Agathe Jay
- Child and Adolescent Psychiatry Departement, Center for Assessment and Diagnostic of Autism, Le Vinatier Hospital, Bron, France
| | - Carmen Schröder
- Department of Child and Adolescent Psychiatry, Strasbourg University Hospital, Strasbourg, France
- CNRS UPR 3212, Team 9 Institute of Cellular and Integrative Neurosciences (INCI), Strasbourg, France
| | - Amaria Baghdadli
- Child and Adolescent Psychiatry Departement, Montpellier University Hospital, Montpellier, France
- INSERM U1178 Centre de Recherche en Epidémiologie Et Santé Des Populations (CESP), Paris Sud University (UPS) Et Versailles Saint Quentin University (UVSQ), Versailles, France
| | - Geneviève Héry-Arnaud
- UMR1078, Génétique, Génomique Fonctionnelle Et Biotechnologies, INSERM, Université de Brest, EFS, IBSAM, Brest, France
- Unité de Bactériologie, Pôle de Biologie-Pathologie, Hôpital La Cavale Blanche, CHRU de Brest, Brest, France
| | - Marie-Maude Geoffray
- Child and Adolescent Psychiatry Departement, Center for Assessment and Diagnostic of Autism, Le Vinatier Hospital, Bron, France
- Health Services and Performance Research EA7425, Claude Bernard Lyon 1 University (CBL1), Lyon, France
| |
Collapse
|
29
|
Kittana M, Ahmadani A, Al Marzooq F, Attlee A. Dietary Fat Effect on the Gut Microbiome, and Its Role in the Modulation of Gastrointestinal Disorders in Children with Autism Spectrum Disorder. Nutrients 2021; 13:3818. [PMID: 34836074 PMCID: PMC8618510 DOI: 10.3390/nu13113818] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 12/19/2022] Open
Abstract
Children with autism spectrum disorder (ASD) report a higher frequency and severity of gastrointestinal disorders (GID) than typically developing (TD) children. GID-associated discomfort increases feelings of anxiety and frustration, contributing to the severity of ASD. Emerging evidence supports the biological intersection of neurodevelopment and microbiome, indicating the integral contribution of GM in the development and function of the nervous system, and mental health, and disease balance. Dysbiotic GM could be a contributing factor in the pathogenesis of GID in children with ASD. High-fat diets may modulate GM through accelerated growth of bile-tolerant bacteria, altered bacterial ratios, and reduced bacterial diversity, which may increase the risk of GID. Notably, saturated fatty acids are considered to have a pronounced effect on the increase of bile-tolerant bacteria and reduction in microbial diversity. Additionally, omega-3 exerts a favorable impact on GM and gut health due to its anti-inflammatory properties. Despite inconsistencies in the data elaborated in the review, the dietary fat composition, as part of an overall dietary intervention, plays a role in modulating GID, specifically in ASD, due to the altered microbiome profile. This review emphasizes the need to conduct future experimental studies investigating the effect of diets with varying fatty acid compositions on GID-specific microbiome profiles in children with ASD.
Collapse
Affiliation(s)
- Monia Kittana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (M.K.); (A.A.)
| | - Asma Ahmadani
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (M.K.); (A.A.)
| | - Farah Al Marzooq
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Amita Attlee
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (M.K.); (A.A.)
| |
Collapse
|
30
|
Kuo CJ, Lin CY, Chen CW, Hsu CY, Hsieh SY, Chiu CT, Lin WR. Risk of Enteric Infection in Patients with Gastric Acid Supressive Drugs: A Population-Based Case-Control Study. J Pers Med 2021; 11:jpm11111063. [PMID: 34834415 PMCID: PMC8621954 DOI: 10.3390/jpm11111063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/20/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
Long-term use of gastric-acid-suppressive drugs is known to be associated with several adverse effects. However, the association between enteric infection and acid suppression therapy is still uncertain. This study aimed to evaluate the association between gastric acid suppression and the risk of enteric infection. Materials and Methods: We conducted a population-based case-control study using the data from Chang Gung Research Database (CGRD) in Taiwan. Between January 2008 and December 2017, a total of 154,590 adult inpatients (age > 18) were identified. A pool of potential eligible controls according to four propensity scores matching by sex, age, and index year were extracted (n = 89,925). Subjects with missing data or who received less than 7 days of proton pump inhibitors (PPIs) and/or H2-receptor antagonists (H2RAs) were excluded. Finally, 17,186 cases and 69,708 corresponding controls were selected for analysis. The use of PPIs and H2RAs, the result of microbiological samples, and co-morbidity conditions have been analyzed. Confounders were controlled by conditional logistic regression. Results: 32.84% of patients in the case group used PPIs, compared with 7.48% in the control group. Of patients in the case group, 9.9% used H2RAs, compared with 6.9% in the control group. Of patients in the case group, 8.3% used a combination of PPIs and H2RAs, compared with 2.7% in the control group. The most common etiological pathogens were Enterococcus (44.8%), Clostridioides difficile (34.5%), and Salmonella spp. (10.2%). The adjusted odds ratio (OR) for PPI use with enteric infection was 5.526 (95% confidence interval [CI], 5.274–5.791). For H2RAs, the adjusted odds ratio was 1.339 (95% confidence interval [CI], 1.261–1.424). Compared to the control group, persons with enteric infection had more frequent acid-suppressive agent usage. Conclusions: This study demonstrates that gastric-acid-suppressive drug use is associated with an increased risk of enteric infection after adjusting for potential biases and confounders.
Collapse
Affiliation(s)
- Chia-Jung Kuo
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-J.K.); (C.-Y.L.); (C.-W.C.); (S.-Y.H.); (C.-T.C.)
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Chang Gung Microbiota Therapy Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Cheng-Yu Lin
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-J.K.); (C.-Y.L.); (C.-W.C.); (S.-Y.H.); (C.-T.C.)
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chun-Wei Chen
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-J.K.); (C.-Y.L.); (C.-W.C.); (S.-Y.H.); (C.-T.C.)
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chiu-Yi Hsu
- Center for Big Data Analytics and Statistics, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
| | - Sen-Yung Hsieh
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-J.K.); (C.-Y.L.); (C.-W.C.); (S.-Y.H.); (C.-T.C.)
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Cheng-Tang Chiu
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-J.K.); (C.-Y.L.); (C.-W.C.); (S.-Y.H.); (C.-T.C.)
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Chang Gung Microbiota Therapy Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Wey-Ran Lin
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-J.K.); (C.-Y.L.); (C.-W.C.); (S.-Y.H.); (C.-T.C.)
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, 5, Fushin Street, Kweishan, Taoyuan 333, Taiwan
- Correspondence: ; Tel.: +886-3-3281200 (ext. 8102); Fax: +886-3-3272236
| |
Collapse
|
31
|
Hu W, Lu W, Li L, Zhang H, Lee YK, Chen W, Zhao J. Both living and dead Faecalibacterium prausnitzii alleviate house dust mite-induced allergic asthma through the modulation of gut microbiota and short-chain fatty acid production. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:5563-5573. [PMID: 33709404 DOI: 10.1002/jsfa.11207] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 03/03/2021] [Accepted: 03/11/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Asthma is increasingly prevalent worldwide, and novel strategies to prevent or treat this disease are needed. Probiotic intervention has recently been reported to be effective for asthma prevention. Here, we explored the effects of Faecalibacterium prausnitzii on the development of allergic airway inflammation in a murine model of house dust mite (HDM)-induced allergic asthma. RESULTS Supplementation with living and dead F. prausnitzii blocked eosinophil, neutrophil, lymphocyte and macrophage influx and alleviated the pathological changes. Moreover, both living and dead F. prausnitzii administration decreased the levels of interleukin (IL)-4, IL-5, IL-13 and immunoglobulin G1, elevated regulatory T cell (Tregs) ratio, improved microbial dysbiosis and enhanced short-chain fatty acid (SCFA) production. Network correlation analysis revealed that the immune indicators were strongly associated with SCFA production. Based on the linear discriminant analysis effect size, Turicibacter was found to be the core genus related to HDM-induced asthma. Living F. prausnitzii treatment enriched Faecalibaculum, Dubosiella and Streptococcus, while dead F. prausnitzii treatment increased Muribaculaceae and Parabacteroides. Interestingly, both living and dead F. prausnitzii administration enriched Lachnoclostridium and normalized the pathways involving carbohydrate and lipid metabolism, which might be related to SCFA production. CONCLUSION Faecalibacterium prausnitzii exerts an anti-asthmatic effect partly by gut microbiota modulation and SCFA production, suggesting its potential as a probiotic agent for allergic asthma prevention. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Wenbing Hu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, China
| | - Lingzhi Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Yuan-Kun Lee
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
32
|
Ding W, Shangguan Y, Zhu Y, Sultan Y, Feng Y, Zhang B, Liu Y, Ma J, Li X. Negative impacts of microcystin-LR and glyphosate on zebrafish intestine: Linked with gut microbiota and microRNAs? ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 286:117685. [PMID: 34438504 DOI: 10.1016/j.envpol.2021.117685] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 06/13/2023]
Abstract
Microcystin-LR (MC-LR) and glyphosate (GLY) have been classified as a Group 2B and Group 2A carcinogens for humans, respectively, and frequently found in aquatic ecosystems. However, data on the potential hazard of MC-LR and GLY exposure to the fish gut are relatively scarce. In the current study, a subacute toxicity test of zebrafish exposed to MC-LR (35 μg L-1) and GLY (3.5 mg L-1), either alone or in combination was performed for 21 d. The results showed that MC-LR or/and GLY treatment reduced the mRNA levels of tight junction genes (claudin-5, occludin, and zonula occludens-1) and altered the levels of diamine oxidase and D-lactic, indicating increased intestinal permeability in zebrafish. Furthermore, MC-LR and/or GLY treatment remarkably increased the levels of intestinal IL-1β and IL-8 but decreased the levels of IL-10 and TGF-β, indicating that MC-LR and/or GLY exposure induced an inflammatory response in the fish gut. MC-LR and/or GLY exposure also activated superoxide dismutase and catalase, generally upregulated the levels of p53, bax, bcl-2, caspase-3, and caspase-9, downregulated the levels of caspase-8 and caused notable histological injury in the fish gut. Moreover, MC-LR and/or GLY exposure also significantly altered the microbial community in the zebrafish gut and the expression of miRNAs (miR-146a, miR-155, miR-16, miR-21, and miR-223). Chronic exposure to MC-LR and/or GLY can induce intestinal damage in zebrafish, and this study is the first to demonstrate an altered gut microbiome and miRNAs in the zebrafish gut after MC-LR and GLY exposure.
Collapse
Affiliation(s)
- Weikai Ding
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Yingying Shangguan
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Yuqing Zhu
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Yousef Sultan
- Department of Food Toxicology and Contaminants, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Yiyi Feng
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Bangjun Zhang
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Yang Liu
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Junguo Ma
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China.
| | - Xiaoyu Li
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| |
Collapse
|
33
|
Response of Fecal Bacterial Flora to the Exposure of Fumonisin B1 in BALB/c Mice. Toxins (Basel) 2021; 13:toxins13090612. [PMID: 34564616 PMCID: PMC8472543 DOI: 10.3390/toxins13090612] [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: 07/05/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/19/2022] Open
Abstract
Fumonisins are a kind of mycotoxin that has harmful influence on the health of humans and animals. Although some research studies associated with fumonisins have been reported, the regulatory limits of fumonisins are imperfect, and the effects of fumonisins on fecal bacterial flora of mice have not been suggested. In this study, in order to investigate the effects of fumonisin B1 (FB1) on fecal bacterial flora, BALB/c mice were randomly divided into seven groups, which were fed intragastrically with 0 mg/kg, 0.018 mg/kg, 0.054 mg/kg, 0.162 mg/kg, 0.486 mg/kg, 1.458 mg/kg and 4.374 mg/kg of FB1 solutions, once a day for 8 weeks. Subsequently, feces were collected for analysis of microflora. The V3-V4 16S rRNA of fecal bacterial flora was sequenced using the Illumina MiSeq platform. The results revealed that fecal bacterial flora of mice treated with FB1 presented high diversity. Additionally, the composition of fecal bacterial flora of FB1 exposure groups showed marked differences from that of the control group, especially for the genus types including Alloprevotella, Prevotellaceae_NK3B31_group, Rikenellaceae_RC9_gut_group, Parabacteroides and phylum types including Cyanobacteria. In conclusion, our data indicate that FB1 alters the diversity and composition of fecal microbiota in mice. Moreover, the minimum dose of FB1 exposure also causes changes in fecal microbiota to some extent. This study is the first to focus on the dose-related effect of FB1 exposure on fecal microbiota in rodent animals and gives references to the regulatory doses of fumonisins for better protection of human and animal health.
Collapse
|
34
|
Kalenik A, Kardaś K, Rahnama A, Sirojć K, Wolańczyk T. Gut microbiota and probiotic therapy in ADHD: A review of current knowledge. Prog Neuropsychopharmacol Biol Psychiatry 2021; 110:110277. [PMID: 33561522 DOI: 10.1016/j.pnpbp.2021.110277] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 01/18/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023]
Abstract
Attention deficit hyperactivity disorder (ADHD) is a common neurodevelopmental disorder characterized by inattention, impulsivity and hyperactivity. The etiology of the disorder is multi-factorial, with a main focus on genetic factors. However, emerging research shows the involvement of changes and imbalances in the intestinal microbiota. Evidence for the influence of gut microbiota on brain development and neurogenesis is clear. We present a review of emerging research on the microbiota in the ADHD population. The aim of this study was to summarize the current state of knowledge on ADHD, to identify gaps in knowledge, as well as to indicate the directions of new research. Thanks to the researchers that would be possible to better understand the complexity of ADHD etiology, especially the role of the intestinal microbiota in the pathogenesis of the disorder. Pubmed, Scopus and Google Scholar databases were used while writing the review. Numerous studies show that probiotic supplementation can have a positive effect on the course of neurodevelopmental disorders, including ADHD. Unfortunately, clinical studies that were identified are mostly inconclusive, and more high-quality research is needed to produce robust evidence for therapy based on interventions targeting microbiota.
Collapse
Affiliation(s)
- Anna Kalenik
- Department of Child Psychiatry, Medical University of Warsaw, Poland.
| | - Karolina Kardaś
- Department of Child Psychiatry, Medical University of Warsaw, Poland
| | - Anna Rahnama
- Department of Child Psychiatry, Medical University of Warsaw, Poland
| | - Katarzyna Sirojć
- Department of Child Psychiatry, Medical University of Warsaw, Poland
| | - Tomasz Wolańczyk
- Department of Child Psychiatry, Medical University of Warsaw, Poland
| |
Collapse
|
35
|
Abstract
Mice transplanted with human microbiota are essential tools for studying the role of microbiota in health and disease, striving for the development of microbiota-modulating therapeutics. Traditionally, germ-free mice have been the principal option for establishing human microbiota-associated (HMA) mouse models, leading to significant insights into the composition and function of the human microbiota. However, there are limitations in using germ-free mice as recipients of human microbiota, including considerable resource allocation to establish and maintain the model and incomplete development of their immune system and physiological functions. Thus, antibiotic-treated, non-germ-free mice have been developed as an alternative to satisfy the growing demand for an accessible HMA mouse model. Several methods have been described for creating "humanized" mice. These protocols vary in their key components, mainly antibiotic conditioning and frequency of oral gavage. To address this practical challenge and formulate a simple and repeatable protocol, we established a HMA mouse model with antibiotic-treated conventional and specific-pathogen free (SPF) C57BL/6J mice, revealing that a single oral gavage allows stable engraftment of the human microbiota. In this chapter, we present our simple protocol for antibiotic conditioning to prepare mice for stable engraftment of human gut microbiota.
Collapse
|
36
|
Zeng Z, Liew SS, Wei X, Pu K. Hemicyanine‐Based Near‐Infrared Activatable Probes for Imaging and Diagnosis of Diseases. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202107877] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ziling Zeng
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Si Si Liew
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Xin Wei
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
- School of Physical and Mathematical Sciences Nanyang Technological University 21 Nanyang Link Singapore 637371 Singapore
| |
Collapse
|
37
|
|
38
|
Zeng Z, Liew SS, Wei X, Pu K. Hemicyanine-Based Near-Infrared Activatable Probes for Imaging and Diagnosis of Diseases. Angew Chem Int Ed Engl 2021; 60:26454-26475. [PMID: 34263981 DOI: 10.1002/anie.202107877] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Indexed: 12/18/2022]
Abstract
Molecular activatable probes with near-infrared (NIR) fluorescence play a critical role in in vivo imaging of biomarkers for drug screening and disease diagnosis. With structural diversity and high fluorescence quantum yields, hemicyanine dyes have emerged as a versatile scaffold for the construction of activatable optical probes. This Review presents a survey of hemicyanine-based NIR activatable probes (HNAPs) for in vivo imaging and early diagnosis of diseases. The molecular design principles of HNAPs towards activatable optical signaling against various biomarkers are discussed with a focus on their broad applications in the detection of diseases including inflammation, acute organ failure, skin diseases, intestinal diseases, and cancer. This progress not only proves the unique value of HNAPs in preclinical research but also highlights their high translational potential in clinical diagnosis.
Collapse
Affiliation(s)
- Ziling Zeng
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Si Si Liew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Xin Wei
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.,School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| |
Collapse
|
39
|
Wang MC, Zaydi AI, Lin WH, Lin JS, Liong MT, Wu JJ. Putative Probiotic Strains Isolated from Kefir Improve Gastrointestinal Health Parameters in Adults: a Randomized, Single-Blind, Placebo-Controlled Study. Probiotics Antimicrob Proteins 2021; 12:840-850. [PMID: 31749128 DOI: 10.1007/s12602-019-09615-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The dairy products remain as the largest reservoir for isolation of probiotic microorganisms. While probiotics have been immensely reported to exert various health benefits, it is also a common notion that these health potentials are strain and host dependent, leading to the need of more human evidence based on specific strains, health targets, and populations. This randomized, single-blind, and placebo-controlled human study aimed to evaluate the potential benefits of putative probiotic strains isolated from kefir on gastrointestinal parameters in fifty-six healthy adults. The consumption of AB-kefir (Bifidobacterium longum, Lactobacillus acidophilus, L. fermentum, L. helveticus, L. paracasei, L. rhamnosus, and Streptococcus thermophiles; total 10 log CFU/sachet) daily for 3 week reduced symptoms of abdominal pain, bloating (P = 0.014), and appetite (P = 0.041) in male subjects as compared to the control. Gut microbiota distribution profiles were shifted upon consumption of AB-kefir compared to baseline, where the abundance of bifidobacteria was increased in male subjects and maintained upon cessation of AB-kefir consumption. The consumption of AB-kefir also increased gastrointestinal abundance of total anaerobes (P = 0.038) and total bacterial (P = 0.049) in female subjects compared to the control after 3 weeks. Our results indicated that AB-kefir could potentially be developed as a natural strategy to improve gastrointestinal functions in adults.
Collapse
Affiliation(s)
- Ming-Cheng Wang
- Division of Nephrology, Department of Internal Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ahmad Imran Zaydi
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Wei-Hung Lin
- Department of Internal Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jin-Seng Lin
- Culture Collection and Research Institute, SYNBIO TECH INC., Kaohsiung, Taiwan
| | - Min-Tze Liong
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Penang, Malaysia.
| | - Jiunn-Jong Wu
- Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang Ming University, Taipei, Taiwan.
| |
Collapse
|
40
|
Long L, Liu X, Jin L, Simon T, Ma W, Kim MN, Yang W, Meyerhardt JA, Chan AT, Giovannucci E, Zhang X. Association of bowel movement frequency and laxative use with risk of hepatocellular carcinoma in US women and men. Int J Cancer 2021; 149:1529-1535. [PMID: 34028016 DOI: 10.1002/ijc.33699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/16/2021] [Accepted: 04/29/2021] [Indexed: 12/24/2022]
Abstract
Abnormal bowel movements have been related to a variety of hepatocellular carcinoma (HCC) risk factors such as dyslipidemia, diabetes and altered metabolism of bile acids and gut microbiota. However, little is known about whether bowel movement frequency affects the risk of developing HCC. We followed 88 123 women in the Nurses' Health Study (NHS) and 28 824 men in the Health Professionals Follow-up Study (HPFS) for up to 24 years. The Cox proportional hazards regression model was used to calculate multivariable hazard ratios (HRs) and confidence intervals (95%CI). We documented 101 incident HCC cases. Compared to those with daily bowel movements, participants with bowel movement more than once per day had a multivariable HR of 1.93 (95%CI: 1.18 to 3.16) in the pooled cohorts. For the same comparison, the positive association appeared stronger for men (2.72, 95% CI: 1.14 to 6.44) than for women (1.63, 95% CI: 0.87 to 3.06) but there was no statistically significant heterogeneity by sex (P-value = .31). We found null associations between bowel movement every 2 days or less and the risk of HCC (HR = 1.05, 95%CI: 0.62 to 1.79). The HR (95%CI) for participants who used laxatives regularly relative to those who never used laxatives was 1.00 (0.64 to 1.55). Our results suggest participants with bowel movement more than once daily is associated with a higher risk of developing HCC compared to those with daily bowel movements. These findings need to be confirmed and potential mechanisms underlying this association need to be elucidated.
Collapse
Affiliation(s)
- Lu Long
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xing Liu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Lina Jin
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Tracey Simon
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Clinical and Translational Epidemiology Unit (CTEU), Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Wenjie Ma
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Clinical and Translational Epidemiology Unit (CTEU), Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mi Na Kim
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Clinical and Translational Epidemiology Unit (CTEU), Massachusetts General Hospital, Boston, Massachusetts, USA.,Division of Gastroenterology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, South Korea
| | - Wanshui Yang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew T Chan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Edward Giovannucci
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
41
|
Meng M, Zhong K, Jiang T, Liu Z, Kwan HY, Su T. The current understanding on the impact of KRAS on colorectal cancer. Biomed Pharmacother 2021; 140:111717. [PMID: 34044280 DOI: 10.1016/j.biopha.2021.111717] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
KRAS (kirsten rat sarcoma viral oncogene) is a member of the RAS family. KRAS mutations are one of most dominant mutations in colorectal cancer (CRC). The impact of KRAS mutations on the prognosis and survival of CRC patients drives many research studies to explore potential therapeutics or target therapy for the KRAS mutant CRC. This review summarizes the current understanding of the pathological consequences of the KRAS mutations in the development of CRC; and the impact of the mutations on the response and the sensitivity to the current front-line chemotherapy. The current therapeutic strategies for treating KRAS mutant CRC, the difficulties and challenges will also be discussed.
Collapse
Affiliation(s)
- Mingjing Meng
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Keying Zhong
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ting Jiang
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhongqiu Liu
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Hiu Yee Kwan
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Tao Su
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
42
|
Fantini MC, Guadagni I. From inflammation to colitis-associated colorectal cancer in inflammatory bowel disease: Pathogenesis and impact of current therapies. Dig Liver Dis 2021; 53:558-565. [PMID: 33541800 DOI: 10.1016/j.dld.2021.01.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/23/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
The risk of colorectal cancer (CRC) is higher in patients with inflammatory bowel disease (IBD). Population-based data from patients with ulcerative colitis (UC) estimate that the risk of CRC is approximately 2- to 3-fold that of the general population; patients with Crohn's disease appear to have a similar increased risk. However, the true extent of colitis-associated cancer (CAC) in undertreated IBD is unclear. Data suggest that the size (i.e., severity and extent) and persistence of the inflammatory process is largely responsible for the development of CRC in IBD. As patients with IBD and CRC have a worse prognosis than those without a history of IBD, the impact of current therapies for IBD on CAC is of importance. Chronic inflammation of the gut has been shown to increase the risk of developing CAC in both UC and CD. Therefore, control of inflammation is pivotal to the prevention of CAC. This review presents an overview of the current knowledge of CAC in IBD patients, focusing on the role of inflammation in the pathogenesis of CAC and the potential for IBD drugs to interfere with the process of carcinogenesis by reducing the inflammatory process or by modulating pathways directly involved in carcinogenesis.
Collapse
Affiliation(s)
- Massimo Claudio Fantini
- Department of Medical Science and Public Health, Gastroenterology Unit, University of Cagliari, Cittadella Universitaria di Monserrato - Asse Didattico I, SS 554 bivio Sestu, 09042 Monserrato, Cagliari, Italy.
| | | |
Collapse
|
43
|
Niu H, Zhou X, Zhang X, Liu T, Wu Y, Lyu L, Liang C, Chen S, Gong P, Zhang J, Han X, Jiang S, Zhang L. Breast milk contains probiotics with anti-infantile diarrhoea effects that may protect infants as they change to solid foods. Environ Microbiol 2021; 23:1750-1764. [PMID: 33684236 DOI: 10.1111/1462-2920.15390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 01/04/2021] [Indexed: 12/20/2022]
Abstract
Infants often experience complementary food-induced diarrhoea (CFID), which occurs when infants switch from breast milk to solid foods. The relative abundances of Prevotella and Rothia were higher in stools of infants with CFID, while the relative abundances of Enterococcus and Escherichia were higher in healthy infants. The abundance of Lactobacillus spp. normally found in breast milk fed to infants with CFID was significantly reduced, and Enterococcus spp. were less abundant when diarrhoea occurred. Furthermore, Lactobacillus and Enterococcus were present as shared bacteria in both mother and infant, and they were considered potential anti-CFID probiotics as their relative abundances in breast milk were negatively correlated to infant CFID. Kyoto encyclopedia of genes and genomes (KEGG) functional analysis showed that the function of amino acid metabolism differed between infants with CFID and healthy infants. Therefore, CFID might be related to the decomposition of proteins in food supplements. The screening revealed seven hydrolytic casein and five hydrolytic casein and rice protein isolates from 320 suspected Lactobacillus and Enterococcus isolates. The animal experiments demonstrated that a mixture of five isolates effectively hydrolysed the casein and rice protein and prevented diarrhoea in young rats. Thus, the occurrence of CFID was found to be closely related to the intestinal and breast milk microbiota, and bacteria that could assist in the digestion of cereal proteins were involved in CFID.
Collapse
Affiliation(s)
- Haiyue Niu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | | | | | - Tongjie Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| | - Yifan Wu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Linzheng Lyu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Cong Liang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Shiwei Chen
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Pimin Gong
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| | - Jiliang Zhang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Xue Han
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Shilong Jiang
- Heilongjiang Feihe Dairy Co. Ltd., Beijing, 100015, China
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| |
Collapse
|
44
|
Li F, Ye J, Shao C, Zhong B. Compositional alterations of gut microbiota in nonalcoholic fatty liver disease patients: a systematic review and Meta-analysis. Lipids Health Dis 2021; 20:22. [PMID: 33637088 PMCID: PMC7908766 DOI: 10.1186/s12944-021-01440-w] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/01/2021] [Indexed: 02/08/2023] Open
Abstract
Background Although imbalanced intestinal flora contributes to the pathogenesis of nonalcoholic fatty liver disease (NAFLD), conflicting results have been obtained for patient-derived microbiome composition analyses. A meta-analysis was performed to summarize the characteristics of intestinal microbiota at the species level in NAFLD patients. Methods Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) Statement, a completed search (last update: December 30, 2020) of databases was performed to identify eligible case-control studies detecting gut microbiota in NAFLD patients. The meta-analysis results are presented as the standard mean difference (SMD) and 95% confidence interval (CI). Bias controls were evaluated with the Newcastle-Ottawa Scale (NOS), funnel plot analysis, and Egger’s and Begg’s tests. Results Fifteen studies (NOS score range: 6–8) that detected the gut microbiota in the stools of 1265 individuals (577 NAFLD patients and 688 controls) were included. It was found that Escherichia, Prevotella and Streptococcus (SMD = 1.55 [95% CI: 0.57, 2.54], 1.89 [95% CI: 0.02, 3.76] and 1.33 [95% CI: 0.62, 2.05], respectively) exhibited increased abundance while Coprococcus, Faecalibacterium and Ruminococcus (SMD = − 1.75 [95% CI: − 3.13, − 0.37], − 9.84 [95% CI: − 13.21, − 6.47] and − 1.84 [95% CI, − 2.41, − 1.27], respectively) exhibited decreased abundance in the NAFLD patients compared with healthy controls. No differences in the abundance of Bacteroides, Bifidobacterium, Blautia, Clostridium, Dorea, Lactobacillus, Parabacteroides or Roseburia were confirmed between the NAFLD patients and healthy controls. Conclusions This meta-analysis revealed that changes in the abundance of Escherichia, Prevotella, Streptococcus, Coprococcus, Faecalibacterium and Ruminococcus were the universal intestinal bacterial signature of NAFLD. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-021-01440-w.
Collapse
Affiliation(s)
- Fuxi Li
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, NO 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080, P. R. China
| | - Junzhao Ye
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, NO 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080, P. R. China
| | - Congxiang Shao
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, NO 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080, P. R. China
| | - Bihui Zhong
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, NO 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080, P. R. China.
| |
Collapse
|
45
|
Xu F, Li N, Wang C, Xing H, Chen D, Wei Y. Clinical efficacy of fecal microbiota transplantation for patients with small intestinal bacterial overgrowth: a randomized, placebo-controlled clinic study. BMC Gastroenterol 2021; 21:54. [PMID: 33549047 PMCID: PMC7866462 DOI: 10.1186/s12876-021-01630-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 01/26/2021] [Indexed: 12/16/2022] Open
Abstract
Background Small intestinal bacterial overgrowth (SIBO) is characterized by the condition that bacteria overgrowth in the small intestine. Fecal microbiota transplantation (FMT) has been applied as an effective tool for reestablishing the structure of gut microbiota. However, whether FMT could be applied as a routine SIBO treatment has not been investigated. Methods In this trial, 55 SIBO patients were enrolled. All participants were randomized in two groups, and were given FMT capsule or placebo capsules once a week for 4 consecutive weeks. Measurements including the lactulose hydrogen breath test gastrointestinal symptoms, as well as fecal microbiota diversity were assessed before and after FMT therapy. Results Gastrointestinal symptoms significantly improved in SIBO patients after treatment with FMT compared to participants in placebo group. The gut microbiota diversity of FMT group had a significant increase, while placebo group showed none. Conclusions This study suggests that applying FMT for patients with SIBO can alleviate gastrointestinal symptoms, indicating that FMT may be a promising and novel therapeutic regimen for SIBO. Trial registry This study was retrospectively registered with the Chinese Clinical Trial registry on 2019.7.10 (ID: ChiCTR1900024409, http://www.chictr.org.cn).
Collapse
Affiliation(s)
- Fenghua Xu
- Department of Gastroenterology, Army Medical Center of PLA affiliated with Army Medical University, No.10 Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Ning Li
- Department of Gastroenterology, Army Medical Center of PLA affiliated with Army Medical University, No.10 Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Chun Wang
- Department of Gastroenterology, Army Medical Center of PLA affiliated with Army Medical University, No.10 Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Hanyang Xing
- Department of Gastroenterology, Army Medical Center of PLA affiliated with Army Medical University, No.10 Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Dongfeng Chen
- Department of Gastroenterology, Army Medical Center of PLA affiliated with Army Medical University, No.10 Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Yanling Wei
- Department of Gastroenterology, Army Medical Center of PLA affiliated with Army Medical University, No.10 Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China.
| |
Collapse
|
46
|
Funosas G, Triadó-Margarit X, Castro F, Villafuerte R, Delibes-Mateos M, Rouco C, Casamayor EO. Individual fate and gut microbiome composition in the European wild rabbit (Oryctolagus cuniculus). Sci Rep 2021; 11:766. [PMID: 33436896 PMCID: PMC7804928 DOI: 10.1038/s41598-020-80782-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 11/30/2020] [Indexed: 01/12/2023] Open
Abstract
Studies connecting microbiome composition and functional performance in wildlife have received little attention and understanding their connections with wildlife physical condition are sorely needed. We studied the variation in gut microbiota (hard fecal pellets) between allopatric subspecies of the European wild rabbit in wild populations and in captured individuals studied under captivity. We evaluated the influence of environmental and host-specific factors. The microbiome of wild rabbit populations reduced its heterogeneity under controlled conditions. None of the host-specific factors tested correlated with the microbiota composition. We only observed significant intra-group dispersion for the age factor. The most diverse microbiomes were rich in Ruminococcaceae potentially holding an enriched functional profile with dominance of cellulases and xylanases, and suggesting higher efficiency in the digestion of fiber-rich food. Conversely, low diversity gut microbiomes showed dominance of Enterobacteriaceae potentially rich in amylases. We preliminary noticed geographical variations in field populations with higher dominance of Ruminococcaceae in south-western than in north-eastern Spain. Spatial differences appeared not to be subspecies driven, since they were lost in captivity, but environmentally driven, although differences in social structure and behavior may also play a role that deserve further investigations. A marginally significant relationship between the Ruminococcaceae/Enterobacteriaceae ratio and potential life expectancy was observed in captive rabbits. We hypothesize that the gut microbiome may determine the efficiency of feeding resource exploitation, and can also be a potential proxy for life expectancy, with potential applications for the management of declining wild herbivorous populations. Such hypotheses remain to be explored in the future.
Collapse
Affiliation(s)
- Gerard Funosas
- Microbial Community Ecology, Centre for Advanced Studies of Blanes-Spanish Council for Research CEAB-CSIC, Accés Cala St Francesc, 14, 17300, Blanes, Spain
| | - Xavier Triadó-Margarit
- Microbial Community Ecology, Centre for Advanced Studies of Blanes-Spanish Council for Research CEAB-CSIC, Accés Cala St Francesc, 14, 17300, Blanes, Spain
| | - Francisca Castro
- Departamento de Didácticas Específicas, Universidad de Córdoba, Sociedad, Ecología y Gestión del Medio Ambiente, UCO-IESA, Unidad Asociada al CSIC, 14004, Córdoba, Spain
| | - Rafael Villafuerte
- Institute of Advanced Social Studies-Spanish Council for Research (IESA-CSIC), 14004, Córdoba, Spain
| | - Miguel Delibes-Mateos
- Institute of Advanced Social Studies-Spanish Council for Research (IESA-CSIC), 14004, Córdoba, Spain
| | - Carlos Rouco
- Ecology Area, Faculty of Science, University of Cordoba, Sociedad, Ecología y Gestión del Medio Ambiente, UCO-IESA, Unidad Asociada al CSIC, 14071, Córdoba, Spain
| | - Emilio O Casamayor
- Microbial Community Ecology, Centre for Advanced Studies of Blanes-Spanish Council for Research CEAB-CSIC, Accés Cala St Francesc, 14, 17300, Blanes, Spain.
| |
Collapse
|
47
|
Jovanovic MM, Jurisevic MM, Gajovic NM, Arsenijevic NN, Jocic MV, Jovanovic IP, Zdravkovic ND, Djukic AL, Maric VJ, Jovanovic MM. Increased Severity of Ulcerative Colitis in the Terminal Phase of the Metabolic Syndrome. TOHOKU J EXP MED 2021; 254:171-182. [PMID: 34248084 DOI: 10.1620/tjem.254.171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Ulcerative colitis is chronic immune-mediated disorder that affects primarily colonic mucosa. The metabolic syndrome has increasing global prevalence with a significant impact on biology of chronic diseases, such as ulcerative colitis. Today it is known that the metabolic syndrome attenuates severity of ulcerative colitis. Still, there is no evidence that different stages of metabolic syndrome alter the course of the ulcerative colitis. The aim of this study was to dissect out how progression of the metabolic syndrome impacted the biology of ulcerative colitis and severity of clinical presentation. Seventy-two patients (41 men and 31 women, 22-81 years old) were enrolled in this observational cross-sectional study. Concentrations of pro- and anti-inflammatory cytokines in serum and feces samples were measured and phenotype of colon infiltrating cells was analyzed. Patients in the terminal phase of the metabolic syndrome have clinically and pathohistologically more severe form of ulcerative colitis, which is followed by decreased concentrations of systemic galectin-1, increased values of systemic pro-inflammatory mediators and increased influx of lymphocytes in affected colon tissue. Our data suggest that reduced concentrations of galectin-1 and predomination of the pro-inflammatory mediators in patients with terminal stage of the metabolic syndrome enhance local chronic inflammatory response and subsequent tissue damage, and together point on important role of galectin-1 in immune response in ulcerative colitis patients with the metabolic syndrome.
Collapse
Affiliation(s)
| | | | - Nevena Miroslav Gajovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac
| | - Nebojsa Nikola Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac
| | | | - Ivan Petar Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac
| | | | | | - Veljko Jovo Maric
- Department of Surgery, Faculty of Medicine Foca, University of East Sarajevo
| | | |
Collapse
|
48
|
Qu MY, Pan YF, Xie M. Research progress of intestinal microecology in the occurrence and development of precancerous lesions of liver. E3S WEB OF CONFERENCES 2021; 251:02046. [DOI: 10.1051/e3sconf/202125102046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Intestinal microecology refers to the interaction between the host and the microorganisms in the human intestinal tract, which is composed mainly of intestinal flora. Intestinal microflora affects the physiological and pathological changes of the host through metabolic activity and host interaction. Precancerous lesion of liver is a potential benign liver disease, which may lead to malignant transformation of liver. It is the intermediate stage from benign lesion to malignant transformation. Recent studies have shown that intestinal microecology is closely related to the occurrence of precancerous lesions of the liver. This study expounds the interaction of the bridge between intestine and liver, the gutliver axis, the intestinal microecology and the precancerous lesions of liver, hoping to provide a new idea for clinical prevention and treatment of precancerous lesions of liver.
Collapse
|
49
|
Lim HJ, Shin HS. Antimicrobial and Immunomodulatory Effects of Bifidobacterium Strains: A Review. J Microbiol Biotechnol 2020; 30:1793-1800. [PMID: 33144551 PMCID: PMC9728261 DOI: 10.4014/jmb.2007.07046] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/29/2020] [Accepted: 10/18/2020] [Indexed: 12/15/2022]
Abstract
Bifidobacterium strains can provide several health benefits, such as antimicrobial and immunomodulatory effects. Some strains inhibit growth or cell adhesion of pathogenic bacteria, including multidrug-resistant bacteria, and their antibacterial activity can be intensified when combined with certain antibiotics. In addition, some strains of bifidobacteria reduce viral infectivity, leading to less epithelial damage of intestinal tissue, lowering the virus shedding titer, and controlling the release of antiviral substances. Furthermore, bifidobacteria can modulate the immune system by increasing immunoglobulins, and inducing or reducing pro- or antiinflammatory cytokines, respectively. In particular, these anti-inflammatory effects are helpful in the treatment of patients who are already suffering from infection or inflammatory diseases. This review summarizes the antimicrobial effects and mechanisms, and immunomodulatory effects of Bifidobacterium strains, suggesting the potential of bifidobacteria as an alternative or complementary treatment option.
Collapse
Affiliation(s)
- Hyun Jung Lim
- College of Pharmacy, Duksung Women’s University, Seoul 01369, Republic of Korea
| | - Hea Soon Shin
- College of Pharmacy, Duksung Women’s University, Seoul 01369, Republic of Korea,Corresponding author Phone: +82-2-901-8398 Fax: +82-2-901-8386 E-mail:
| |
Collapse
|
50
|
Kim WK, Jang YJ, Han DH, Jeon K, Lee C, Han HS, Ko G. Lactobacillus paracasei KBL382 administration attenuates atopic dermatitis by modulating immune response and gut microbiota. Gut Microbes 2020; 12:1-14. [PMID: 33016202 PMCID: PMC7553742 DOI: 10.1080/19490976.2020.1819156] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 08/10/2020] [Accepted: 08/21/2020] [Indexed: 02/03/2023] Open
Abstract
Administration of probiotics has been linked to immune regulation and changes in gut microbiota composition, with effects on atopic dermatitis (AD). In this study, we investigated amelioration of the symptoms of AD using Lactobacillus paracasei KBL382 isolated from the feces of healthy Koreans. Mice with Dermatophagoides farinae extract (DFE)-induced AD were fed 1 × 109 CFU d-1 of L. paracasei KBL382 for 4 weeks. Oral administration of L. paracasei KBL382 significantly reduced AD-associated skin lesions, epidermal thickening, serum levels of immunoglobulin E, and immune cell infiltration. L. paracasei KBL382-treated mice showed decreased production of T helper (Th)1-, Th2-, and Th17-type cytokines, including thymic stromal lymphopoietin, thymus, and activation-regulated chemokine, and macrophage-derived chemokine, and increased production of the anti-inflammatory cytokine IL-10 and transforming growth factor-β in skin tissue. Intake of L. paracasei KBL382 also increased the proportion of CD4+ CD25+ Foxp3+ regulatory T cells in mesenteric lymph nodes. In addition, administration of L. paracasei KBL382 dramatically changed the composition of gut microbiota in AD mice. Administration of KBL382 significantly ameliorates AD-like symptoms by regulating the immune response and altering the composition of gut microbiota.
Collapse
Affiliation(s)
- Woon-Ki Kim
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - You Jin Jang
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Dae Hee Han
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Kyungchan Jeon
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Cheonghoon Lee
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - Hyuk Seung Han
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - GwangPyo Ko
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
- N-Bio, Seoul National University, Seoul, Republic of Korea
- KoBioLabs, Inc., Seoul, Republic of Korea
- Center for Human and Environmental Microbiome, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|