1
|
Mukhatayev Z, Kovenskiy A, Ren Z, Rangel SM, Katkenov N, Khuanbai Y, Shivde R, Daniel M, Dellacecca ER, Cedercreutz K, Ostapchuk Y, Nurgozhina A, Chulenbayeva L, Nurgaziyev M, Jarmukhanov Z, Nurlankyzy M, Kozhdan K, Seidulla S, Mukhanbetzhanova Z, Sergazy S, Kozhakhmetov S, Ali Y, Daftary KM, Green SJ, Kundu RV, Kushugulova A, Le Poole IC. Escherichia abundance and metabolism align with vitiligo disease activity. J Invest Dermatol 2025:S0022-202X(25)00119-8. [PMID: 39983982 DOI: 10.1016/j.jid.2025.01.032] [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: 11/12/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 02/23/2025]
Abstract
Vitiligo is a cutaneous autoimmune disorder characterized by progressive depigmentation due to melanocyte destruction by cytotoxic T cells. Genetic factors predispose patients to the disease, supported by environmental factors often initiating new disease episodes. We questioned whether disease outcomes are partially defined by pathogenic microbes driving nutrient deficiencies and inflammation. Our study presents results from research on the diet and gut microbiome composition of vitiligo patients and healthy controls from Kazakhstan and the USA. Dietary nutrient intake was assessed using NIH-generated Diet History Questionnaires. Vitiligo patients with active disease exhibited limited intake of specific fatty acids, amino acids, and zinc. Disease activity was further characterized by an abundance of Odoribacter and Escherichia genera in the gut. Metabolic pathway analysis supported a role for the Escherichia genus in disease activity by limiting energy metabolism and amino acid biosynthetic pathways. Disease activity also aligned with elevated circulating pro-inflammatory cytokines. These findings suggest that nutritional limitations are not compensated by metabolites from the gut microbiome in active disease, potentially leaving room for inflammation and exacerbating vitiligo. The intricate relationship between diet, gut microbiome composition, and disease progression in vitiligo highlights potential avenues for targeted interventions to reduce autoimmune activity and improve patient outcomes.
Collapse
Affiliation(s)
| | - Artur Kovenskiy
- National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - Ziyou Ren
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Stephanie M Rangel
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Nurlubek Katkenov
- National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | | | - Rohan Shivde
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Moriel Daniel
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Emilia R Dellacecca
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | | | | | | | | | | | | - Kamilya Kozhdan
- National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - Symbat Seidulla
- National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | | | - Shynggyss Sergazy
- National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | | | - Yasmeen Ali
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Karishma M Daftary
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Stefan J Green
- Genomics and Microbiome Core Facility, Rush University, Chicago, Illinois, USA
| | - Roopal V Kundu
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - I Caroline Le Poole
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
2
|
Zhou W, Yuan S, Kang W, Deng X, Zhou H, Ruan J, Feng X, Qi M, Chen B. Replication Study and Meta-Analysis of the Contribution of Seven Genetic Polymorphisms in Immune-Related Genes to the Risk of Gastric and Colorectal Cancers. Int J Immunogenet 2025; 52:39-55. [PMID: 39800859 DOI: 10.1111/iji.12705] [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: 09/27/2024] [Revised: 12/27/2024] [Accepted: 01/05/2025] [Indexed: 01/30/2025]
Abstract
Recently, it has been realized that immune processes participate in the pathogenesis of human cancers. A large number of genetic polymorphisms in immune-related genes have been extensively examined for their roles in the susceptibility of gastric cancer (GC) and colorectal cancer (CRC), including IL4 gene rs2070874, IL4RA gene rs1801275, IL18 gene rs187238, IL18RAP gene rs917997, IL17A gene rs8193036, IL23R gene rs1884444 and IL23R gene rs10889677. However, there is no consistent conclusion, which calls for further research. In this case-control study, these 7 genetic polymorphisms were genotyped by Sanger sequencing in a total of 1247 patients with cancer (GC/CRC: 460/787) and 800 healthy individuals. A total of 31 previous studies and our present study were included in this meta-analysis. The case-control study revealed that in Hubei Chinese population, rs2070874, rs187238 and rs10889677 were significantly associated with CRC risk, whereas only rs917997 was significantly associated with GC risk. The meta-analysis showed that rs2070874, rs917997, rs8193036 and rs1884444 were significantly associated with GC risk in Chinese population, whereas rs2070874 in total population, rs1801275 in Asian population and rs187238 in Chinese population were significantly associated with CRC risk. IL4 gene rs2070874, IL18RAP gene rs917997, IL17A gene rs8193036 and IL23R gene rs1884444 may serve as the susceptible factors for GC carcinogenesis in Chinese population. IL4 gene rs2070874 in total population, IL4RA gene rs1801275 in Asian population and IL18 gene rs187238 in Chinese population may be genetic biomarkers for CRC susceptibility.
Collapse
Affiliation(s)
- Weiguang Zhou
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Siqi Yuan
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Wenqiang Kang
- Institute of WUT-AMU, Wuhan University of Technology, Wuhan, Hubei, China
| | - Xiangyuan Deng
- Institute of WUT-AMU, Wuhan University of Technology, Wuhan, Hubei, China
| | - Hang Zhou
- Institute of WUT-AMU, Wuhan University of Technology, Wuhan, Hubei, China
| | - Jiangyi Ruan
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Xianhong Feng
- Department of Clinical Laboratory, Wuhan Xinzhou District People's Hospital, Wuhan, Hubei, China
| | - Meifang Qi
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Bifeng Chen
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
- Institute of WUT-AMU, Wuhan University of Technology, Wuhan, Hubei, China
| |
Collapse
|
3
|
Pietrasanta C, Carlosama C, Lizier M, Fornasa G, Jost TR, Carloni S, Giugliano S, Silvestri A, Brescia P, De Ponte Conti B, Braga D, Mihula M, Morosi L, Bernardinello A, Ronchi A, Martano G, Mosca F, Penna G, Grassi F, Pugni L, Rescigno M. Prenatal antibiotics reduce breast milk IgA and induce dysbiosis in mouse offspring, increasing neonatal susceptibility to bacterial sepsis. Cell Host Microbe 2024; 32:2178-2194.e6. [PMID: 39603245 DOI: 10.1016/j.chom.2024.11.001] [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: 01/25/2023] [Revised: 07/25/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Antibiotics (Abx) are administered to 20%-30% of pregnant women, but their effects on neonatal immune development are poorly understood. We show that newborn mice born to Abx-treated dams are more susceptible to late-onset sepsis. This susceptibility is linked to lower maternal breast milk immunoglobulin A (IgA), neonatal fecal IgA, and IgA coating of intestinal bacteria, thus causing the translocation of intestinal pathobionts. Weaned young adults born to Abx-treated mothers had reduced IgA+ plasma cells in the ileum and colon, fecal secretory IgA (SIgA), colonic CD4+ T regulatory lymphocytes and T helper 17-like lymphocytes, and a less diverse fecal microbiome. However, treatment with apyrase, which restores SIgA secretion, prompted IgA production in breast milk and protected pups from sepsis. Additionally, breast milk from untreated mothers rescued the phenotypes of pups born to Abx-treated mothers. Our data highlight the impact of prenatal Abx on breast milk IgA and their long-term influence on intestinal mucosal immune function mediated by breastfeeding.
Collapse
Affiliation(s)
- Carlo Pietrasanta
- Department of Clinical Sciences and Community Health, Dipartimento di Eccellenza 2023-2027, University of Milan, Via della Commenda 19, Milan, Italy; NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via della Commenda 12, Milan, Italy.
| | - Carolina Carlosama
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Michela Lizier
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Giulia Fornasa
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Tanja Rezzonico Jost
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Sara Carloni
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Milan 20072, Italy
| | - Silvia Giugliano
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Milan 20072, Italy
| | | | - Paola Brescia
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Milan 20072, Italy
| | - Benedetta De Ponte Conti
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Daniele Braga
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Martin Mihula
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Department of Medical Biotechnology, Università di Siena, Via Banchi di Sotto 55, 53100 Siena, Italy
| | - Lavinia Morosi
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Alessandro Bernardinello
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Milan 20072, Italy
| | - Andrea Ronchi
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via della Commenda 12, Milan, Italy
| | - Giuseppe Martano
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Institute of Neuroscience, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Via Manzoni 56, Rozzano, Milan, Italy
| | - Fabio Mosca
- Department of Clinical Sciences and Community Health, Dipartimento di Eccellenza 2023-2027, University of Milan, Via della Commenda 19, Milan, Italy; NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via della Commenda 12, Milan, Italy
| | - Giuseppe Penna
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Fabio Grassi
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland; Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Lorenza Pugni
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via della Commenda 12, Milan, Italy
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Milan 20072, Italy.
| |
Collapse
|
4
|
Rizzo SM, Alessandri G, Tarracchini C, Bianchi MG, Viappiani A, Mancabelli L, Lugli GA, Milani C, Bussolati O, van Sinderen D, Ventura M, Turroni F. Molecular cross-talk among human intestinal bifidobacteria as explored by a human gut model. Front Microbiol 2024; 15:1435960. [PMID: 39314876 PMCID: PMC11418510 DOI: 10.3389/fmicb.2024.1435960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Bifidobacteria are well known as common and abundant colonizers of the human gut and are able to exert multiple beneficial effects on their host, although the cooperative and competitive relationships that may occur among bifidobacterial strains are still poorly investigated. Therefore, to dissect possible molecular interactions among bifidobacterial species that typically colonize the human gut, three previously identified bifidobacterial prototypes, i.e., B. bifidum PRL2010, B. breve PRL2012, and B. longum PRL2022 were cultivated individually as well as in bi- and tri-association in a human gut-simulating medium. Transcriptomic analyses of these co-associations revealed up-regulation of genes predicted to be involved in the production of extracellular structures including pili (i.e., flp pilus assembly TadE protein gene), exopolysaccharides (i.e., GtrA family protein gene) and teichoic acids (i.e., ABC transporter permease), along with carbohydrate, amino acid and vitamin metabolism-related genes (i.e., exo-alpha-sialidase; beta-galactosidase and pyridoxamine kinase), suggesting that co-cultivation of bifidobacteria induces a response, in individual bifidobacterial strains, aimed at enhancing their proliferation and survival, as well as their ability to cooperate with their host to promote their persistence. Furthermore, exposure of the selected prototypes to human cell line monolayers unveiled the ability of the bifidobacterial tri-association to communicate with their host by increasing the expression of genes involved in adherence to/interaction with intestinal human cells. Lastly, bifidobacterial tri-association promoted the transcriptional upregulation of genes responsible for maintaining the integrity and homeostasis of the intestinal epithelial barrier.
Collapse
Affiliation(s)
- Sonia Mirjam Rizzo
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Chiara Tarracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Massimiliano G. Bianchi
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | | | - Leonardo Mancabelli
- Microbiome Research Hub, University of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Ovidio Bussolati
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
5
|
Li H, Cui X, Lin Y, Huang F, Tian A, Zhang R. Gut microbiota changes in patients with Alzheimer's disease spectrum based on 16S rRNA sequencing: a systematic review and meta-analysis. Front Aging Neurosci 2024; 16:1422350. [PMID: 39175809 PMCID: PMC11338931 DOI: 10.3389/fnagi.2024.1422350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024] Open
Abstract
Background The gut microbiota (GM) is hypothesized to play roles in Alzheimer's disease (AD) pathogenesis. In recent years, many GM composition and abundance investigations in AD patients have been conducted; however, despite this work, some results remain controversial. Therefore, we conducted a systematic review and meta-analysis using 16S ribosomal RNA (16S rRNA) sequencing to explore GM alterations between patients with AD spectrum and healthy controls (HCs). Methods A systematic and comprehensive literature search of PubMed, Web of Science, Embase, the Cochrane Library, China National Knowledge Infrastructure, China Biology Medicine disc database, WanFang database and Social Sciences Citation Index databases was conducted from inception to January 2023. Inclusion and exclusion criteria were strictly defined, and two researchers independently screened and extracted information from selected studies. Data quality were evaluated according to the "Cochrane system evaluator manual" and pooled data were comprehensively analyzed using Stata 14 software with standardized mean differences (SMDs) and 95% confidence intervals (95% CIs) used to measure effect sizes. Also, geographical heterogeneity effects (related to cohorts) on GM abundance were examined based on subgroup meta-analyses if sufficient studies reported outcomes. Finally, publication bias was assessed using funnel plots. Results Out of 1566 articles, 13 studies involving 581 patients with AD spectrum and 445 HCs were deemed eligible and included in our analysis. In summary, a decreased microbiota alpha diversity and a significantly distinct pattern of clustering with regard to beta diversity were observed in AD spectrum patients when compared with HCs. Comparative analyses revealed a decreased Ruminococcus, Faecalibacterium, Lachnospira, Dialister, Lachnoclostridium, and Roseburia abundance in AD spectrum patients while Phascolarctobacterium, Lactobacillus, and Akkermansia muciniphila were more enriched in patients when compared to HCs. Furthermore, regional variations may have been in play for intestinal microbes such as Bacteroides, Bifidobacterium, and Alistipes. Conclusion Our meta-analysis identified alterations in GM abundance in patients with AD spectrum, with 12 genera from four major phyla significantly associated with AD. Moreover, we provided evidence for region-specific alterations in Bacteroides, Bifidobacterium, and Alistipes abundance. These findings may have profound implications for the development of innovative GM-based strategies to prevent and treat AD. Systematic review registration https://doi.org/10.37766/inplasy2024.6.0067, identifier INPLASY202460067.
Collapse
Affiliation(s)
- Hui Li
- Department of Gerontology and Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaopan Cui
- Department of Gerontology and Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuxiu Lin
- Department of Gerontology and Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fengqiong Huang
- Department of Gerontology and Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ayong Tian
- Department of Anesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Rongwei Zhang
- Department of Gerontology and Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
6
|
Little RB, Carter SJ, Motl RW, Hunter G, Cook A, Liu N, Krontiras H, Lefkowitz EJ, Turan B, Schleicher E, Rogers LQ. Role of Gut Microbe Composition in Psychosocial Symptom Response to Exercise Training in Breast Cancer Survivors (ROME) study: protocol for a randomised controlled trial. BMJ Open 2024; 14:e081660. [PMID: 38702085 PMCID: PMC11086582 DOI: 10.1136/bmjopen-2023-081660] [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: 11/02/2023] [Accepted: 04/12/2024] [Indexed: 05/06/2024] Open
Abstract
INTRODUCTION Breast cancer survivors have an increased risk for chronic fatigue and altered gut microbiota composition, both with negative health and quality of life affects. Exercise modestly improves fatigue and is linked to gut microbial diversity and production of beneficial metabolites. Studies suggest that gut microbiota composition is a potential mechanism underlying fatigue response to exercise. Randomised controlled trials testing the effects of exercise on the gut microbiome are limited and there is a scarcity of findings specific to breast cancer survivors. The objective of this study is to determine if fitness-related modifications to gut microbiota occur and, if so, mediate the effects of aerobic exercise on fatigue response. METHODS AND ANALYSIS The research is a randomised controlled trial among breast cancer survivors aged 18-74 with fatigue. The primary aim is to determine the effects of aerobic exercise training compared with an attention control on gut microbiota composition. The secondary study aims are to test if exercise training (1) affects the gut microbiota composition directly and/or indirectly through inflammation (serum cytokines), autonomic nervous system (heart rate variability) or hypothalamic-pituitary-adrenal axis mediators (hair cortisol assays), and (2) effects on fatigue are direct and/or indirect through changes in the gut microbiota composition. All participants receive a standardised controlled diet. Assessments occur at baseline, 5 weeks, 10 weeks and 15 weeks (5 weeks post intervention completion). Faecal samples collect the gut microbiome and 16S gene sequencing will identify the microbiome. Fatigue is measured by a 13-item multidimensional fatigue scale. ETHICS AND DISSEMINATION The University of Alabama at Birmingham Institutional Review Board (IRB) approved this study on 15 May 2019, UAB IRB#30000320. A Data and Safety Monitoring Board convenes annually or more often if indicated. Findings will be disseminated in peer-reviewed journals and conference presentations. TRIAL REGISTRATION NUMBER ClinicalTrials.gov, NCT04088708.
Collapse
Affiliation(s)
- Rebecca B Little
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Stephen J Carter
- Department of Kinesiology, Indiana University Bloomington, Bloomington, Indiana, USA
| | - Robert W Motl
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Gary Hunter
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Abby Cook
- Baylor Scott & White Medical Center Temple, Temple, Texas, USA
| | - Nianjun Liu
- Department of Epidemiology and Biostatistics, Indiana University Bloomington, Bloomington, Indiana, USA
| | - Helen Krontiras
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Elliot J Lefkowitz
- Department of Computer Science, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Bulent Turan
- Department of Psychology, Koc University, Istanbul, Turkey
| | - Erica Schleicher
- Department of Health Behavior, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Laura Q Rogers
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
7
|
Huang X, Hu X, Li S, Li T. Vitexin-rhamnoside encapsulated with zein-pectin nanoparticles relieved high-fat diet induced lipid metabolism disorders in mice by altering the gut microbiota. Int J Biol Macromol 2024; 264:130704. [PMID: 38460630 DOI: 10.1016/j.ijbiomac.2024.130704] [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: 12/04/2023] [Revised: 01/26/2024] [Accepted: 03/05/2024] [Indexed: 03/11/2024]
Abstract
This study aimed to investigate the modulatory effects of Vitexin-rhamnoside (VR) and Zein-VR-pectin nanoparticles (VRN) on lipid metabolism disorders induced by high-fat diet (HFD). The ingestion of VR or VRN attenuated dyslipidemia and fat accumulation in HFD mice, and improved intestinal dysbiosis by regulating the relative abundance of dominant bacteria, alleviating chronic inflammation and hepatic injury in HFD mice. The intervention effect of VRN was significantly higher than that of VR. After fecal microbiota transplantation (FMT) treatment, the fecal microbiota of VRN-treated donor mice significantly attenuated the symptoms associated with hyperlipidemia, confirming that VRN ameliorates HFD-induced disorders of lipid metabolism by modulating the gut microbiota, especially increasing the abundance of Rombousia and Faecalibaculum. Overall, VRN can regulate the gut microbiota and thus improve lipid metabolism. The present study provided new evidence that nanoparticles enhance the bioavailability of food bioactive ingredients.
Collapse
Affiliation(s)
- Xin Huang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Xiaopei Hu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China.
| | - Suhong Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China.
| | - Tuoping Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China.
| |
Collapse
|
8
|
Guo J, Shi W, Li X, Yang B, Qin C, Su L. Comparative Analysis of Gut Microbiomes in Laboratory Chinchillas, Ferrets, and Marmots: Implications for Pathogen Infection Research. Microorganisms 2024; 12:646. [PMID: 38674591 PMCID: PMC11051751 DOI: 10.3390/microorganisms12040646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Gut microbes play a vital role in the health and disease of animals, especially in relation to pathogen infections. Chinchillas, ferrets, and marmots are commonly used as important laboratory animals for infectious disease research. Here, we studied the bacterial and fungal microbiota and discovered that chinchillas had higher alpha diversity and a higher abundance of bacteria compared to marmots and ferrets by using the metabarcoding of 16S rRNA genes and ITS2, coupled with co-occurrence network analysis. The dominant microbes varied significantly among the three animal species, particularly in the gut mycobiota. In the ferrets, the feces were dominated by yeast such as Rhodotorula and Kurtzmaniella, while in the chinchillas, we found Teunomyces and Penicillium dominating, and Acaulium, Piromyces, and Kernia in the marmots. Nevertheless, the dominant bacterial genera shared some similarities, such as Clostridium and Pseudomonas across the three animal species. However, there were significant differences observed, such as Vagococcus and Ignatzschineria in the ferrets, Acinetobacter and Bacteroides in the chinchillas, and Bacteroides and Cellvibrio in the marmots. Additionally, our differential analysis revealed significant differences in classification levels among the three different animal species, as well as variations in feeding habitats that resulted in distinct contributions from the host microbiome. Therefore, our data are valuable for monitoring and evaluating the impacts of the microbiome, as well as considering potential applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Lei Su
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing 100021, China; (J.G.); (W.S.); (X.L.); (B.Y.); (C.Q.)
| |
Collapse
|
9
|
Salvadori M, Rosso G. Update on the gut microbiome in health and diseases. World J Methodol 2024; 14:89196. [PMID: 38577200 PMCID: PMC10989414 DOI: 10.5662/wjm.v14.i1.89196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/18/2023] [Accepted: 01/27/2024] [Indexed: 03/07/2024] Open
Abstract
The Human Microbiome Project, Earth Microbiome Project, and next-generation sequencing have advanced novel genome association, host genetic linkages, and pathogen identification. The microbiome is the sum of the microbes, their genetic information, and their ecological niche. This study will describe how millions of bacteria in the gut affect the human body in health and disease. The gut microbiome changes in relation with age, with an increase in Bacteroidetes and Firmicutes. Host and environmental factors affecting the gut microbiome are diet, drugs, age, smoking, exercise, and host genetics. In addition, changes in the gut microbiome may affect the local gut immune system and systemic immune system. In this study, we discuss how the microbiome may affect the metabolism of healthy subjects or may affect the pathogenesis of metabolism-generating metabolic diseases. Due to the high number of publications on the argument, from a methodologically point of view, we decided to select the best papers published in referred journals in the last 3 years. Then we selected the previously published papers. The major goals of our study were to elucidate which microbiome and by which pathways are related to healthy and disease conditions.
Collapse
Affiliation(s)
- Maurizio Salvadori
- Department of Renal Transplantation, Careggi University Hospital, Florence 50139, Tuscany, Italy
| | - Giuseppina Rosso
- Division of Nephrology, San Giovanni di Dio Hospital, Florence 50143, Toscana, Italy
| |
Collapse
|
10
|
Qiao X, Li X, Wang Z, Feng Y, Wei X, Li L, Pan Y, Zhang K, Zhou R, Yan L, Li P, Xu C, Lv Z, Tian Z. Gut microbial community and fecal metabolomic signatures in different types of osteoporosis animal models. Aging (Albany NY) 2024; 16:1192-1217. [PMID: 38284894 PMCID: PMC10866450 DOI: 10.18632/aging.205396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/13/2023] [Indexed: 01/30/2024]
Abstract
BACKGROUND The gut microbiota (GM) constitutes a critical factor in the maintenance of physiological homeostasis. Numerous studies have empirically demonstrated that the GM is closely associated with the onset and progression of osteoporosis (OP). Nevertheless, the characteristics of the GM and its metabolites related to different forms of OP are poorly understood. In the present study, we examined the changes in the GM and its metabolites associated with various types of OP as well as the correlations among them. METHODS We simultaneously established rat postmenopausal, disuse-induced, and glucocorticoid-induced OP models. We used micro-CT and histological analyses to observe bone microstructure, three-point bending tests to measure bone strength, and enzyme-linked immunosorbent assay (ELISA) to evaluate the biochemical markers of bone turnover in the three rat OP models and the control. We applied 16s rDNA to analyze GM abundance and employed untargeted metabolomics to identify fecal metabolites in all four treatment groups. We implemented multi-omics methods to explore the relationships among OP, the GM, and its metabolites. RESULTS The 16S rDNA sequencing revealed that both the abundance and alterations of the GM significantly differed among the OP groups. In the postmenopausal OP model, the bacterial genera g__Bacteroidetes_unclassified, g__Firmicutes_unclassified, and g__Eggerthella had changed. In the disuse-induced and glucocorticoid-induced OP models, g__Akkermansia and g__Rothia changed, respectively. Untargeted metabolomics disclosed that the GM-derived metabolites significantly differed among the OP types. However, a Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that it was mainly metabolites implicated in lipid and amino acid metabolism that were altered in all cases. An association analysis indicated that the histidine metabolism intermediate 4-(β-acetylaminoethyl) imidazole was common to all OP forms and was strongly correlated with all bone metabolism-related bacterial genera. Hence, 4-(β-acetylaminoethyl) imidazole might play a vital role in OP onset and progression. CONCLUSIONS The present work revealed the alterations in the GM and its metabolites that are associated with OP. It also disclosed the changes in the GM that are characteristic of each type of OP. Future research should endeavor to determine the causal and regulatory effects of the GM and the metabolites typical of each form of OP.
Collapse
Affiliation(s)
- Xiaochen Qiao
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, Shanxi, P.R. China
- Department of Orthopedics, Jinzhong Hospital Affiliated to Shanxi Medical University, Jinzhong 030600, Shanxi, P.R. China
| | - Xiaoyan Li
- Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, Shanxi, P.R. China
| | - Zhichao Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, Shanxi, China
| | - Yi Feng
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, Shanxi, P.R. China
| | - Xiaochun Wei
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, Shanxi, P.R. China
| | - Lu Li
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, Shanxi, P.R. China
| | - Yongchun Pan
- Department of Orthopedics, Third People’s Hospital of Datong City, Datong 037006, Shanxi, P.R. China
| | - Kun Zhang
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, Shanxi, P.R. China
| | - Ruhao Zhou
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, Shanxi, P.R. China
| | - Lei Yan
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, Shanxi, P.R. China
| | - Pengcui Li
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, Shanxi, P.R. China
| | - Chaojian Xu
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, Shanxi, P.R. China
| | - Zhi Lv
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, Shanxi, P.R. China
| | - Zhi Tian
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, Shanxi, P.R. China
| |
Collapse
|
11
|
Gan Z, Zhao M, Xia Y, Yan Y, Ren W. Carbon metabolism in the regulation of macrophage functions. Trends Endocrinol Metab 2024; 35:62-73. [PMID: 37778898 DOI: 10.1016/j.tem.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 10/03/2023]
Abstract
Carbon metabolism, including one-carbon (1C) metabolism and central carbon metabolism (CCM), provides energy for the cell and generates metabolites with signaling activities. The regulation of macrophage polarization involves complex signals and includes an epigenetic level. Epigenetic modifications through changes in carbon metabolism allow macrophages to respond in a timely manner to their environment and adapt to metabolic demands during macrophage polarization. Here we summarize the current understanding of the crosstalk between carbon metabolism and epigenetic modifications in macrophages under physiological conditions and in the tumor microenvironment (TME) and provide targets and further directions for macrophage-associated diseases.
Collapse
Affiliation(s)
- Zhending Gan
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510000, Guangdong, China
| | - Muyang Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510000, Guangdong, China
| | - Yaoyao Xia
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Yuqi Yan
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Wenkai Ren
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510000, Guangdong, China.
| |
Collapse
|
12
|
Liu L, Zhang S, Zhi F, Song Y, Li B, Gao P, Zhang Y, Ma K, Xu J, Jiang B, Chu Y, Li Y, Qin J. RNA helicase DExD/H-box 5 modulates intestinal microbiota in mice. Microb Pathog 2023; 182:106265. [PMID: 37482112 DOI: 10.1016/j.micpath.2023.106265] [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: 06/19/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 07/25/2023]
Abstract
The RNA helicase DExD/H-box (DDX) family of proteins plays a central role in host cellular RNA metabolism, including mRNA regulation, microRNA biogenesis, and ribosomal processing. DDX5, also known as p68, promotes viral replication and tumorigenesis. However, there have been no studies on the regulation of the intestinal microbiota by DDX family proteins. We constructed DDX5 knockout mice (Ddx5+/-) using CRISPR/CAS9 technology. Subsequently, DDX5 knockout mice were analyzed for PCR products, mRNA levels, protein expression, immunohistochemistry, and histopathological lesions. Fecal (n = 12) and ileum (n = 12) samples were collected from the Ddx5+/- and wild-type (Ddx5+/+) mice. The diversity, richness, and structural separation of the intestinal microbiota of the Ddx5+/- and Ddx5+/+ mice were determined by 16S rRNA sequencing and analysis. Ddx5+/- mice were successfully established, and the ileum had normal morphology, a clear layer of tissue structures, and neatly arranged cupped cells. DDX5 knockout mice did not exhibit adverse effects on the ileal tissue. Microbial diversity and abundance were not significantly different, but the microbial structure of the intestinal microbiota was clustered separately between Ddx5+/+ and Ddx5+/- mice. Furthermore, we found that the relative abundance of Akkermansia and Clostridium_sensu_stricto_1 in the Ddx5+/- mice was significantly lower than in the Ddx5+/+ mice. These analyses indicated specific interactions between the intestinal microbiota and DDX5 protein. Our results indicate that DDX5 has a significant effect on the composition of the intestinal microbiota in mice, suggesting its potential as a promising novel target for the treatment of inflammation and tumorigenesis in the intestine.
Collapse
Affiliation(s)
- Liyuan Liu
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei Province, 071001, China; State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China
| | - Silan Zhang
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang Province, 830091, China
| | - Feijie Zhi
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China
| | - Yinjuan Song
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China
| | - Bin Li
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang Province, 830091, China
| | - Pengchen Gao
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China
| | - Ying Zhang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China
| | - Ke Ma
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China
| | - Jian Xu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang Province, 830091, China
| | - Bo Jiang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing, 100097, China
| | - Yuefeng Chu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang Province, 830091, China
| | - Yongqing Li
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing, 100097, China
| | - Jianhua Qin
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei Province, 071001, China.
| |
Collapse
|
13
|
Huang P, Cao L, Du J, Gao J, Zhang Y, Sun Y, Li Q, Nie Z, Xu G. Effects of Prometryn Exposure on Hepatopancreas Oxidative Stress and Intestinal Flora in Eriocheir sinensis (Crustacea: Decapoda). Antioxidants (Basel) 2023; 12:1548. [PMID: 37627543 PMCID: PMC10451815 DOI: 10.3390/antiox12081548] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
There is growing evidence that long-term exposure to prometryn (a widely used herbicide) can induce toxicity in bony fish and shrimp. Our previous study demonstrated its 96 h acute toxicity on the crab Eriocheir sinensis. However, studies on whether longer exposure to prometryn with a lower dose induces toxicity in E. sinensis are scarce. Therefore, we conducted a 20 d exposure experiment to investigate its effects on the hepatopancreas and intestine of E. sinensi. Prometryn reduce the activities of antioxidant enzymes, increase the level of lipid peroxidation and cause oxidative stress. Moreover, long-term exposure resulted in immune and detoxification fatigue, while short-term exposure to prometryn could upregulate the expression of genes related to immunity, inflammation and detoxification. Prometryn altered the morphological structure of the hepatopancreas (swollen lumen) and intestine (shorter intestinal villi, thinner muscle layer and thicker peritrophic membrane). In addition, prometryn changed the species composition of the intestinal flora. In particular, Bacteroidota and Proteobacteria showed a dose-dependent decrease accompanied by a dose-dependent increase in Firmicutes at the phylum level. At the genus level, all exposure groups significantly increased the abundance of Zoogloea and a Firmicutes bacterium ZOR0006, but decreased Shewanella abundance. Interestingly, Pearson correlation analysis indicated a potential association between differential flora and hepatopancreatic disorder. Phenotypic abundance analysis indicated that changes in the gut flora decreased the intestinal organ's resistance to stress and increased the potential for opportunistic infection. In summary, our research provides new insights into the prevention and defense strategies in response to external adverse environments and contributes to the sustainable development of E. sinensis culture.
Collapse
Affiliation(s)
- Peng Huang
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (P.H.); (L.C.); (J.D.); (Y.Z.)
| | - Liping Cao
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (P.H.); (L.C.); (J.D.); (Y.Z.)
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (J.G.)
| | - Jinliang Du
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (P.H.); (L.C.); (J.D.); (Y.Z.)
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (J.G.)
| | - Jiancao Gao
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (J.G.)
| | - Yuning Zhang
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (P.H.); (L.C.); (J.D.); (Y.Z.)
| | - Yi Sun
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (J.G.)
| | - Quanjie Li
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (J.G.)
| | - Zhijuan Nie
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (P.H.); (L.C.); (J.D.); (Y.Z.)
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (J.G.)
| | - Gangchun Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (P.H.); (L.C.); (J.D.); (Y.Z.)
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (J.G.)
| |
Collapse
|
14
|
Wang YC, Cao Y, Pan C, Zhou Z, Yang L, Lusis AJ. Intestinal cell type-specific communication networks underlie homeostasis and response to Western diet. J Exp Med 2023; 220:213924. [PMID: 36880999 PMCID: PMC10038833 DOI: 10.1084/jem.20221437] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/14/2022] [Accepted: 02/10/2023] [Indexed: 03/08/2023] Open
Abstract
The small intestine plays a key role in immunity and mediates inflammatory responses to high fat diets. We have used single-cell RNA-sequencing (scRNA-seq) and statistical modeling to examine gaps in our understanding of the dynamic properties of intestinal cells and underlying cellular mechanisms. Our scRNA-seq and flow cytometry studies of different layers of intestinal cells revealed new cell subsets and modeled developmental trajectories of intestinal intraepithelial lymphocytes, lamina propria lymphocytes, conventional dendritic cells, and enterocytes. As compared to chow-fed mice, a high-fat high-sucrose (HFHS) "Western" diet resulted in the accumulation of specific immune cell populations and marked changes to enterocytes nutrient absorption function. Utilizing ligand-receptor analysis, we profiled high-resolution intestine interaction networks across all immune cell and epithelial structural cell types in mice fed chow or HFHS diets. These results revealed novel interactions and communication hubs among intestinal cells, and their potential roles in local as well as systemic inflammation.
Collapse
Affiliation(s)
- Yu-Chen Wang
- Department of Medicine, Division of Cardiology, University of California, Los Angeles , Los Angeles, CA, USA
| | - Yang Cao
- Department of Medicine, Division of Cardiology, University of California, Los Angeles , Los Angeles, CA, USA
| | - Calvin Pan
- Department of Medicine, Division of Cardiology, University of California, Los Angeles , Los Angeles, CA, USA
| | - Zhiqiang Zhou
- Department of Medicine, Division of Cardiology, University of California, Los Angeles , Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles , Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, Los Angeles , Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, the David Geffen School of Medicine, University of California, Los Angeles , Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles , Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, University of California, Los Angeles , Los Angeles, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles , Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine at UCLA , Los Angeles, CA, USA
| |
Collapse
|
15
|
Grüner N, Ortlepp AL, Mattner J. Pivotal Role of Intestinal Microbiota and Intraluminal Metabolites for the Maintenance of Gut-Bone Physiology. Int J Mol Sci 2023; 24:ijms24065161. [PMID: 36982235 PMCID: PMC10048911 DOI: 10.3390/ijms24065161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Intestinal microbiota, and their mutual interactions with host tissues, are pivotal for the maintenance of organ physiology. Indeed, intraluminal signals influence adjacent and even distal tissues. Consequently, disruptions in the composition or functions of microbiota and subsequent altered host-microbiota interactions disturb the homeostasis of multiple organ systems, including the bone. Thus, gut microbiota can influence bone mass and physiology, as well as postnatal skeletal evolution. Alterations in nutrient or electrolyte absorption, metabolism, or immune functions, due to the translocation of microbial antigens or metabolites across intestinal barriers, affect bone tissues, as well. Intestinal microbiota can directly and indirectly alter bone density and bone remodeling. Intestinal dysbiosis and a subsequently disturbed gut-bone axis are characteristic for patients with inflammatory bowel disease (IBD) who suffer from various intestinal symptoms and multiple bone-related complications, such as arthritis or osteoporosis. Immune cells affecting the joints are presumably even primed in the gut. Furthermore, intestinal dysbiosis impairs hormone metabolism and electrolyte balance. On the other hand, less is known about the impact of bone metabolism on gut physiology. In this review, we summarized current knowledge of gut microbiota, metabolites and microbiota-primed immune cells in IBD and bone-related complications.
Collapse
Affiliation(s)
- Niklas Grüner
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anna Lisa Ortlepp
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
16
|
Lin J, Wen J, Xiao N, Cai YT, Xiao J, Dai W, Chen JP, Zeng KW, Liu F, Du B, Li P. Anti-diabetic and gut microbiota modulation effects of sacha inchi (Plukenetia volubilis L.) leaf extract in streptozotocin-induced type 1 diabetic mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:4304-4312. [PMID: 35043419 DOI: 10.1002/jsfa.11782] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 09/17/2021] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Sacha inchi (Plukenetia volubilis L.) tea has been used as an adjuvant treatment for diabetes in Pu'er, in the Yunnan province of China. The effects of sacha inchi tea on diabetes and the underlying mechanisms remain unknown. This study was conducted to investigate the influence of a water extract of sacha inchi (P. volubilis L.) leaves (PWE) on hypoglycemic activity and gut microbiota composition in mice with streptozotocin (STZ)-induced type 1 diabetes mellitus (T1DM). During the 6 weeks of the study, T1DM mice were administered PWE intragastrically at 400 mg kg-1 body weight (BW) per day. RESULTS Treatment with PWE reduced excessive loss of BW and excessive intake of food. It significantly decreased blood glucose levels and improved oral glucose tolerance. The treatment caused protective histopathological transformations in sections of the pancreas, leading to decreased insulin resistance and improved insulin sensitivity. Treatment with PWE also significantly ameliorated disorders of the gut microbiota structure and increased the richness and diversity of intestinal microbial species in T1DM mice. At the genus level, the populations of several crucial bacteria, such as Akkermansia, Parabacteroides, and Muribaculum increased in the PWE treatment group but the abundance of Ruminiclostridium and Oscillibacter decreased. CONCLUSIONS Treatment with PWE can ameliorate hyperglycemic symptoms in STZ-induced T1DM mice, and the anti-diabetic effect of PWE was related to the amelioration of gut microbial structural disorder and the enrichment of functional bacteria. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jinming Lin
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jiamin Wen
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Nan Xiao
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yu-Tong Cai
- School of Data Science, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen, China
| | - Jie Xiao
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wenhao Dai
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jian-Ping Chen
- School of Chinese Medicine, LKS faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Fengsong Liu
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Bing Du
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Pan Li
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
Thomas MS, Blesso CN, Calle MC, Chun OK, Puglisi M, Fernandez ML. Dietary Influences on Gut Microbiota with a Focus on Metabolic Syndrome. Metab Syndr Relat Disord 2022; 20:429-439. [PMID: 35704900 DOI: 10.1089/met.2021.0131] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
There is a clear correlation between gut microbiota, diet, and metabolic outcomes. A diet high in fiber has been shown to decrease inflammation, increase insulin sensitivity, and reduce dyslipidemias whereas a diet high in fat and sugar leads to dyslipidemia, insulin resistance, and low-grade inflammation. There is recent evidence suggesting that the human gut microbiota has a significant role in the development or the resolution of metabolic syndrome (MetS) and associated conditions. Leading a stressful, sedentary lifestyle with limited or no physical activity and consuming an unhealthy diet high in saturated fat, simple carbohydrates, and sodium and low in dietary fiber and in high-quality protein are some of the contributing factors. Unhealthy diets have been shown to induce alterations in the gut microbiota and contribute to the pathogenesis of MetS by altering microbiota composition and disrupting the intestinal barrier, which leads to low-grade systemic inflammation. In contrast, healthy diets can lead to changes in microbiota that increase gut barrier function and increase the production of anti-inflammatory biomarkers. This review aims at providing a more in-depth discussion of diet-induced dysbiosis of the gut microbiota and its effect on MetS. Here, we discuss the possible mechanisms involved in the development of the metabolic biomarkers that define MetS, with an emphasis on the role of sugar and dietary fiber in microbiome-mediated changes in low-grade systemic inflammation and metabolic dysfunction.
Collapse
Affiliation(s)
- Minu S Thomas
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Christopher N Blesso
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Mariana C Calle
- Health Sciences Department ST 110-M, Worcester University, Worcester, Massachusetts, USA
| | - Ock K Chun
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Michael Puglisi
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Maria Luz Fernandez
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
18
|
Han Y, Zhang Y, Ouyang K, Chen L, Zhao M, Wang W. Sulfated Cyclocarya paliurus polysaccharides improve immune function of immunosuppressed mice by modulating intestinal microbiota. Int J Biol Macromol 2022; 212:31-42. [PMID: 35597376 DOI: 10.1016/j.ijbiomac.2022.05.110] [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: 02/27/2022] [Revised: 05/07/2022] [Accepted: 05/14/2022] [Indexed: 01/03/2023]
Abstract
The study was aimed to investigate the effect of Cyclocarya paliurus polysaccharides (CPP) and the sulfation derivative (S-CPP) on modulate intestinal mucosal immunity and intestinal microbiota in cyclophosphamide-induced mice. The results showed that CPP and S-CPP effectively alleviated intestinal villi injury, enhanced the contents of interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) in small intestinal tissue and serum, and upregulated IL-1β at gene levels, zonula occludens-1 (ZO-1), Occludin and Claudin-1 at gene and protein levels, thereby promoting the repair of intestinal mechanical barrier and enhancing intestinal mucosal immunity. Moreover, the beneficial modulation of CPP and S-CPP on the overall structure of intestinal microbiota was revealed by performing 16S ribosomal RNA (16S rRNA) sequencing. Sulfated modification could improve the protection of CPP on the intestinal barrier and the regulation of systemic immunity. S-CPP had a stronger potential to reduce the damage of cyclophosphamide (Cy) on immunity and intestinal microbiota.
Collapse
Affiliation(s)
- Yi Han
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China; School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yang Zhang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Kehui Ouyang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Lingli Chen
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Meng Zhao
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Wenjun Wang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
19
|
Liu L, Lu Y, Xu C, Chen H, Wang X, Wang Y, Cai B, Li B, Verstrepen L, Ghyselinck J, Marzorati M, Yao Q. The Modulation of Chaihu Shugan Formula on Microbiota Composition in the Simulator of the Human Intestinal Microbial Ecosystem Technology Platform and its Influence on Gut Barrier and Intestinal Immunity in Caco-2/THP1-Blue™ Cell Co-Culture Model. Front Pharmacol 2022; 13:820543. [PMID: 35370677 PMCID: PMC8964513 DOI: 10.3389/fphar.2022.820543] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
The traditional Chinese medicine (TCM)–Chaihu Shugan Formula (CSF), consisting of several Chinese botanical drugs like Bupleurum, is derived from the ancient Chinese pharmacopeia. It has been used for more than thousands of years in various suboptimal health statuses and diseases induced by chronic stress based on empirical therapy. Recent studies confirm the role of CSF in the development of many diseases, including depression, stress-induced hepatic injury and tumors. However, little has been known about the mechanisms behind the health effects of CSF. Here, we investigate the influence of CSF on the modulation of the simulated colonic microbiota of five healthy donors, gut barrier integrity, and intestinal immunity by combining the simulator of the human intestinal microbial ecosystem (SHIME®) technology platform with co-culture of intestinal and immune cells. This approach revealed that CSF stimulated the production of SCFA (acetate, propionate and butyrate) across donors while significantly lowering the production of branched SCFA (bSCFA). In terms of community composition, CSF stimulated a broad spectrum of health-related Bifidobacterium species, which are potent acetate and lactate producers. At the same time, it lowered the abundance of opportunistic pathogenic Escherichia coli. Later, we explore the effect of colonic fermentation of CSF on the gut barrier and intestinal immunity in the Caco-2/THP1-blue™ cell co-culture model. Based on the study using SHIME technology platform, CSF showed protective effects on inflammation-induced intestinal epithelial barrier disruption in all donors. Also, the treatment of CSF showed pronounced anti-inflammatory properties by strongly inducing anti-inflammatory cytokines IL-6 and IL-10 and reducing pro-inflammatory cytokine TNF-α. These findings demonstrate a significant modulatory effect of CSF on intestinal gut microbiota. CSF-microbial fermentation products improved the gut barrier and controlled intestinal inflammation.
Collapse
Affiliation(s)
- Ling Liu
- Department of Integrated Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yi Lu
- Department of Clinical Nutrition, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Chao Xu
- Department of Integrated Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Haitao Chen
- Department of Integrated Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xuanying Wang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yijie Wang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Biyu Cai
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bing Li
- Leuven Health Technology Centre China Centre, Hangzhou, China
| | | | | | - Massimo Marzorati
- Center of Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium.,ProDigest BV, Technologiepark, Zwijnaarde, Belgium
| | - Qinghua Yao
- Department of Integrated Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China.,Key Laboratory of Traditional Chinese Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China.,Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
20
|
Miranda-Ribera A, Serena G, Liu J, Fasano A, Kingsbury MA, Fiorentino MR. The Zonulin-transgenic mouse displays behavioral alterations ameliorated via depletion of the gut microbiota. Tissue Barriers 2021; 10:2000299. [PMID: 34775911 DOI: 10.1080/21688370.2021.2000299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The gut-brain axis hypothesis suggests that interactions in the intestinal milieu are critically involved in regulating brain function. Several studies point to a gut-microbiota-brain connection linking an impaired intestinal barrier and altered gut microbiota composition to neurological disorders involving neuroinflammation. Increased gut permeability allows luminal antigens to cross the gut epithelium, and via the blood stream and an impaired blood-brain barrier (BBB) enters the brain impacting its function. Pre-haptoglobin 2 (pHP2), the precursor protein to mature HP2, is the first characterized member of the zonulin family of structurally related proteins. pHP 2 has been identified in humans as the thus far only endogenous regulator of epithelial and endothelial tight junctions (TJs). We have leveraged the Zonulin-transgenic mouse (Ztm) that expresses a murine pHP2 (zonulin) to determine the role of increased gut permeability and its synergy with a dysbiotic intestinal microbiota on brain function and behavior. Here we show that Ztm mice display sex-dependent behavioral abnormalities accompanied by altered gene expression of BBB TJs and increased expression of brain inflammatory genes. Antibiotic depletion of the gut microbiota in Ztm mice downregulated brain inflammatory markers ameliorating some anxiety-like behavior. Overall, we show that zonulin-dependent alterations in gut permeability and dysbiosis of the gut microbiota are associated with an altered BBB integrity, neuroinflammation, and behavioral changes that are partially ameliorated by microbiota depletion. Our results suggest the Ztm model as a tool for the study of the cross-talk between the microbiome/gut and the brain in the context of neurobehavioral/neuroinflammatory disorders.
Collapse
Affiliation(s)
- Alba Miranda-Ribera
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Gloria Serena
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Jundi Liu
- Department of Poultry Science, University of Georgia, Athens, GA, USA
| | - Alessio Fasano
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Marcy A Kingsbury
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA, USA.,Lurie Center for Autism, Boston, MA, USA
| | - Maria R Fiorentino
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA, USA
| |
Collapse
|
21
|
Chen J, Liu Y, Huang Y, Tong A, Liu B, Zeng F. Schizochytrium
oil and its Mixture with Fish Oil and
Sacha inchi
Oil Ameliorate Gut Microbiota Composition and Lipid Metabolism via the FAS/HMGCR/SREBP Signaling Pathway. EUR J LIPID SCI TECH 2021. [DOI: 10.1002/ejlt.202100108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jie Chen
- College of Food Science Fujian Agriculture and Forestry University Fuzhou 350002 China
| | - Yilin Liu
- College of Food Science Fujian Agriculture and Forestry University Fuzhou 350002 China
| | - Ying Huang
- College of Food Science Fujian Agriculture and Forestry University Fuzhou 350002 China
| | - Aijun Tong
- College of Food Science Fujian Agriculture and Forestry University Fuzhou 350002 China
| | - Bin Liu
- College of Food Science Fujian Agriculture and Forestry University Fuzhou 350002 China
- National Engineering Research Center of JUNCAO Technology Fujian Agriculture and Forestry University Fuzhou 350002 China
| | - Feng Zeng
- College of Food Science Fujian Agriculture and Forestry University Fuzhou 350002 China
- National Engineering Research Center of JUNCAO Technology Fujian Agriculture and Forestry University Fuzhou 350002 China
| |
Collapse
|
22
|
Administration of Lactobacillus reuteri Combined with Clostridium butyricum Attenuates Cisplatin-Induced Renal Damage by Gut Microbiota Reconstitution, Increasing Butyric Acid Production, and Suppressing Renal Inflammation. Nutrients 2021; 13:nu13082792. [PMID: 34444952 PMCID: PMC8402234 DOI: 10.3390/nu13082792] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/28/2021] [Accepted: 08/09/2021] [Indexed: 01/02/2023] Open
Abstract
Cisplatin-induced nephrotoxicity is associated with gut microbiota disturbance. The present study aimed to investigate whether supplementation of Lactobacillus reuteri and Clostridium butyricum (LCs) had a protective effect on cisplatin-induced nephrotoxicity through reconstruction of gut microbiota. Wistar rats were given different treatments: control, cisplatin (Cis), cisplatin + C. butyricum and L. reuteri (Cis+LCs), and C. butyricum and L. reuteri (LCs). We observed that cisplatin-treated rats supplemented with LCs exhibited significantly decreased renal inflammation (KIM-1, F4/80, and MPO), oxidative stress, fibrosis (collagen IV, fibronectin, and a-SMA), apoptosis, concentration of blood endotoxin and indoxyl sulfate, and increased fecal butyric acid production compared with those without supplementation. In addition, LCs improved the cisplatin-induced microbiome dysbiosis by maintaining a healthy gut microbiota structure and diversity; depleting Escherichia-Shigella and the Enterobacteriaceae family; and enriching probiotic Bifidobacterium, Ruminococcaceae, Ruminiclostridium_9, and Oscillibacter. Moreover, the LCs intervention alleviated the cisplatin-induced intestinal epithelial barrier impairment. This study indicated LCs probiotic serves as a mediator of the gut–kidney axis in cisplatin-induced nephrotoxicity to restore the intestinal microbiota composition, thereby suppressing uremic toxin production and enhancing butyrate production. Furthermore, the renoprotective effect of LCs is partially mediated by increasing the anti-inflammatory effects and maintaining the integrity of the intestinal barrier.
Collapse
|
23
|
Morita Y, Jounai K, Tomita Y, Maruyama M. Long-term intake of Lactobacillus paracasei KW3110 prevents age-related circadian locomotor activity and changes in gut metabolism in physiologically aged mice. Exp Gerontol 2021; 153:111477. [PMID: 34271136 DOI: 10.1016/j.exger.2021.111477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 11/20/2022]
Abstract
Aging involves age-progressive loss of physiological functions in organs and tissues. We previously showed that Lactobacillus paracasei KW3110 suppressed age-related inflammation and prevented age-related retinal ganglion cell (RGC) loss. As RGCs mediate biological behaviors associated with responses to ambient light, we assessed whether L. paracasei KW3110 affects circadian locomotor activities in physiologically aged mice. The ratio of locomotor activity during the nighttime (active phase) to daytime (inactive phase) significantly decreased in physiologically aged mice compared with young mice: intake of L. paracasei KW3110 prevented this decrease. We also performed metabolomics analysis of cecal contents using both capillary electrophoresis and liquid chromatography time-of-flight mass spectrometry to better understand the benefical effects for aging of L. paracasei KW3110 through a gut retina axis, since our previous study showed that L. paracasei KW3110 mitigated not only age-related expansions of intestinal inflammatory immune cells but age-related alternation of gut microbiome composition. Principal component analysis showed clear changes in metabolites between physiologically aged mice fed a diet containing L. paracasei KW3110 and age-matched control mice. Furthermore, we found that intake of L. paracasei KW3110 mitigated age-related changes in some fatty acids compared with age-matched control mice. Taken together, L. paracasei KW3110 might regulate age-related alternation of metabolites in cecal contents, potentially leading to suppression of age-related decline in physiological functions, including impairment of circadian locomotor activities.
Collapse
Affiliation(s)
- Yuji Morita
- KIRIN Central Research Institute, Kirin Holdings Co., Ltd., Kanagawa, Japan.
| | - Kenta Jounai
- Technical Development Center, Koiwai Dairy Products Co., Ltd., Sayama, Saitama 350-1321, Japan
| | - Yasuyuki Tomita
- KIRIN Central Research Institute, Kirin Holdings Co., Ltd., Kanagawa, Japan
| | - Mitsuo Maruyama
- Department of Inflammation and Immunosenescence, Geroscience Research Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan.; Department of Aging Research, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
| |
Collapse
|
24
|
Miller MO, Kashyap PC, Becker SL, Thomas RM, Hodin RA, Miller G, Hundeyin M, Pushalkar S, Cohen D, Saxena D, Shogan BD, Morris-Stiff GJ. SSAT State-of-the-Art Conference: Advancements in the Microbiome. J Gastrointest Surg 2021; 25:1885-1895. [PMID: 32989690 DOI: 10.1007/s11605-020-04551-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The microbiome plays a major role in human physiology by influencing obesity, inducing inflammation, and impacting cancer therapies. During the 60th Annual Meeting of the Society of the Alimentary Tract (SSAT) at the State-of-the-Art Conference, experts in the field discussed the influence of the microbiome. This paper is a summary of the influence of the microbiome on obesity, inflammatory bowel disease, pancreatic cancer, cancer therapies, and gastrointestinal optimization. This review shows how the microbiome plays an important role in the development of diseases and surgical complications. Future studies are needed in targeting the gut microbiome to develop individualized therapies.
Collapse
Affiliation(s)
- Miquell O Miller
- Department of General Surgery, Stanford University, 300 Pasteur Dr, Stanford, CA, 94305, USA.
| | - Purna C Kashyap
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Sarah L Becker
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ryan M Thomas
- Departments of Surgery, Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, 32610, USA
| | - Richard A Hodin
- Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - George Miller
- Departments of Surgery and Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Mautin Hundeyin
- Departments of Surgery and Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Smruti Pushalkar
- Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Deirdre Cohen
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Deepak Saxena
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Benjamin D Shogan
- Department of Surgery, University of Chicago, Chicago, IL, 60637, USA
| | | |
Collapse
|
25
|
Alam MS, Gangiredla J, Hasan NA, Barnaba T, Tartera C. Aging-Induced Dysbiosis of Gut Microbiota as a Risk Factor for Increased Listeria monocytogenes Infection. Front Immunol 2021; 12:672353. [PMID: 33995413 PMCID: PMC8115019 DOI: 10.3389/fimmu.2021.672353] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/12/2021] [Indexed: 12/15/2022] Open
Abstract
Invasive foodborne Listeria monocytogenes infection causes gastroenteritis, septicemia, meningitis, and chorioamnionitis, and is associated with high case-fatality rates in the elderly. It is unclear how aging alters gut microbiota, increases risk of listeriosis, and causes dysbiosis post-infection. We used a geriatric murine model of listeriosis as human surrogate of listeriosis for aging individuals to study the effect of aging and L. monocytogenes infection. Aging and listeriosis-induced perturbation of gut microbiota and disease severity were compared between young-adult and old mice. Young-adult and old mice were dosed intragastrically with L. monocytogenes. Fecal pellets were collected pre- and post-infection for microbiome analysis. Infected old mice had higher Listeria colonization in liver, spleen, and feces. Metagenomics analyses of fecal DNA-sequences showed increase in α-diversity as mice aged, and infection reduced its diversity. The relative abundance of major bacterial phylum like, Bacteroidetes and Firmicutes remained stable over aging or infection, while the Verrucomicrobia phylum was significantly reduced only in infected old mice. Old mice showed a marked reduction in Clostridaiceae and Lactobacillaceae bacteria even before infection when compared to uninfected young-adult mice. L. monocytogenes infection increased the abundance of Porphyromonadaceae and Prevotellaceae in young-adult mice, while members of the Ruminococcaceae and Lachnospiraceae family were significantly increased in old mice. The abundance of the genera Blautia and Alistipes were significantly reduced post-infection in young-adult and in old mice as compared to their uninfected counterparts. Butyrate producing, immune-modulating bacterial species, like Pseudoflavonifractor and Faecalibacterium were significantly increased only in old infected mice, correlating with increased intestinal inflammatory mRNA up-regulation from old mice tissue. Histologic analyses of gastric tissues showed extensive lesions in the Listeria-infected old mice, more so in the non-glandular region and fundus than in the pylorus. Commensal species like Lactobacillus, Clostridiales, and Akkermansia were only abundant in infected young-adult mice but their abundance diminished in the infected old mice. Listeriosis in old mice enhances the abundance of butyrate-producing inflammatory members of the Ruminococcaceae/Lachnospiraceae bacteria while reducing/eliminating beneficial commensals in the gut. Results of this study indicate that, aging may affect the composition of gut microbiota and increase the risk of invasive L. monocytogenes infection.
Collapse
Affiliation(s)
- Mohammad S Alam
- Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, United States
| | - Jayanthi Gangiredla
- Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, United States
| | | | - Tammy Barnaba
- Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, United States
| | - Carmen Tartera
- Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, United States
| |
Collapse
|
26
|
Tu Y, Yang R, Xu X, Zhou X. The microbiota-gut-bone axis and bone health. J Leukoc Biol 2021; 110:525-537. [PMID: 33884666 DOI: 10.1002/jlb.3mr0321-755r] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/16/2021] [Accepted: 04/05/2021] [Indexed: 02/05/2023] Open
Abstract
The gastrointestinal tract is colonized by trillions of microorganisms, consisting of bacteria, fungi, and viruses, known as the "second gene pool" of the human body. In recent years, the microbiota-gut-bone axis has attracted increasing attention in the field of skeletal health/disorders. The involvement of gut microbial dysbiosis in multiple bone disorders has been recognized. The gut microbiota regulates skeletal homeostasis through its effects on host metabolism, immune function, and hormonal secretion. Owing to the essential role of the gut microbiota in skeletal homeostasis, novel gut microbiota-targeting therapeutics, such as probiotics and prebiotics, have been proven effective in preventing bone loss. However, more well-controlled clinical trials are still needed to evaluate the long-term efficacy and safety of these ecologic modulators in the treatment of bone disorders.
Collapse
Affiliation(s)
- Ye Tu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Ran Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Xin Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
27
|
Dietary Fucose Affects Macrophage Polarization and Reproductive Performance in Mice. Nutrients 2021; 13:nu13030855. [PMID: 33807914 PMCID: PMC8001062 DOI: 10.3390/nu13030855] [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: 02/22/2021] [Accepted: 03/02/2021] [Indexed: 12/30/2022] Open
Abstract
Intestinal mucus protects epithelial and immune cells from the gut resident microorganisms, and provides growth-promoting factors as mucus-derived O-glycans for beneficial bacteria. A lack of intestinal protective mucus results in changes in the commensal microflora composition, mucosal immune system reprogramming, and inflammation. Previous work has shown that fucose, the terminal glycan chain component of the intestinal glycoprotein Mucin2, and fucoidan polysaccharides have an anti-inflammatory effect in some mouse models of colitis. This study evaluates the effect of fucose on reproductive performance in heterozygous mutant Muc2 female mice. We found that even though Muc2+/− females are physiologically indistinguishable from C57Bl/6 mice, they have a significantly reduced reproductive performance upon dietary fucose supplementation. Metagenomic analysis reveals that the otherwise healthy wild-type siblings of Muc2−/− animals have reduced numbers of some of the intestinal commensal bacterial species, compared to C57BL/6 mice. We propose that the changes in beneficial microflora affect the immune status in Muc2+/− mice, which causes implantation impairment. In accordance with this hypothesis, we find that macrophage polarization during pregnancy is impaired in Muc2+/− females upon addition of fucose. Metabolic profiling of peritoneal macrophages from Muc2+/− females reveals their predisposition towards anaerobic glycolysis in favor of oxidative phosphorylation, compared to C57BL/6-derived cells. In vitro experiments on phagocytosis activity and mitochondrial respiration suggest that fucose affects oxidative phosphorylation in a genotype-specific manner, which might interfere with implantation depending on the initial status of macrophages. This hypothesis is further confirmed in BALB/c female mice, where fucose caused pregnancy loss and opposed implantation-associated M2 macrophage polarization. Taken together, these data suggest that intestinal microflora affects host immunity and pregnancy outcome. At the same time, dietary fucose might act as a differential regulator of macrophage polarization during implantation, depending on the immune status of the host.
Collapse
|
28
|
Characterization and proteomic analysis of outer membrane vesicles from a commensal microbe, Enterobacter cloacae. J Proteomics 2021; 231:103994. [PMID: 33007464 DOI: 10.1016/j.jprot.2020.103994] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 12/16/2022]
Abstract
Outer membrane vesicles (OMVs) are membrane-enclosed spherical entities released by gram-negative bacteria and are important for bacterial survival under stress conditions. There have been numerous studies on OMVs released by gram-negative pathogenic bacteria, but an understanding of the functions and characteristics of the OMVs produced by commensal microbes is still lacking. Enterobacter cloacae is a gram-negative commensal bacterium present in the human gut microbiome, but this organism can also function as an opportunistic pathogen. Understanding the OMV-mediated communication route between bacteria-bacteria or bacteria-host is essential for the determination of the biological functions of the commensal bacterium in the gut and delineating between benign and virulent characteristics. In this study, we have described a proteome of E. cloacae OMVs, which are membrane vesicles in a size range of 20-300 nm. Proteomic analysis showed the presence of membrane-bound proteins, including transporters, receptors, signaling molecules, and protein channels. The physical and proteomic analyses also indicate this bacterium uses two mechanisms for OMV production. This study is one of the few existing descriptions of the proteomic profile of OMVs generated by a commensal Proteobacteria, and the first report of OMVs produced by E. cloacae. SIGNIFICANCE: This study prioritizes the importance of understanding the vesicular proteome of the human commensal bacterium, Enterobacter cloacae. We demonstrate for the first time that the gram-negative bacterium E. cloacae ATCC 13047 produces outer membrane vesicles (OMVs). The proteomic analysis showed enrichment of membrane-bound proteins in these vesicles. Understanding the cargo proteins of OMVs will help in exploring the physiological and functional role of these vesicles in the human microbiome and how they assist in the conversion of a bacterium from commensal to pathogen under certain conditions. While EM images reveal vesicles budding from the bacterial surface, the presence of cytoplasmic proteins and genomic DNA within the OMVs indicate that explosive cell lysis is an additional mechanism of biogenesis for these OMVs along with outer membrane blebbing. This research encourages future work on characterizing membrane vesicles produced by commensal bacterial and investigating their role in cell to cell communication.
Collapse
|
29
|
Yago T, Asano T, Fujita Y, Migita K. Familial Mediterranean fever phenotype progression into anti-cyclic citrullinated peptide antibody-positive rheumatoid arthritis:a case report. Fukushima J Med Sci 2020; 66:160-166. [PMID: 33162488 PMCID: PMC7790467 DOI: 10.5387/fms.2020-07] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Familial Mediterranean fever (FMF) is caused by dysfunction of the MEFV gene product, pyrin. Here we report a case of FMF phenotype which developed into rheumatoid arthritis (RA), based on a positive result for anti-cyclic citrullinated peptide (CCP) antibody (Ab). A 42-year-old woman presented to our clinic with more than 6 months of intermittent arthralgia in the wrists, feet, and fingers associated with menstruation. No fever was reported and there was no family history of FMF or other autoimmune diseases. Laboratory tests revealed elevated C-reactive protein (CRP) and rheumatoid factor (RF). Tests for autoantibodies including anti-CCP Ab, antinuclear Ab, and anti-DNA Ab were all negative. Genetic analysis identified an R304R homozygous mutation in MEFV; however, the pathological significance is unclear because this mutation does not cause amino acid substitution. We diagnosed incomplete FMF phenotype despite the lack of fever due to periodic arthritis, lack of autoantibodies, and complete resolution of arthritis following colchicine treatment within a day. Several months later, increased stiffness and arthralgia persistently occurred in finger joints on both sides. Ultrasonography revealed synovitis at the metacarpophalangeal and metatarsophalangeal joints. Laboratory analysis revealed the patient to be positive for anti-CCP Ab. Therefore, we finally diagnosed RA. Her arthritis diminished following administration of methotrexate and salazosulfapyridine. We consider the possibility that pyrin dysfunction may have affected the acquired immunity, contributing to the onset of RA as an autoimmune disease. This is an interesting case of equivalent FMF progressing into RA and will be valuable to raise awareness of a continuum from autoinflammatory to autoimmune disease.
Collapse
Affiliation(s)
- Toru Yago
- Department of Rheumatology, Fukushima Medical University School of Medicine
| | - Tomoyuki Asano
- Department of Rheumatology, Fukushima Medical University School of Medicine
| | - Yuya Fujita
- Department of Rheumatology, Fukushima Medical University School of Medicine
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University School of Medicine
| |
Collapse
|
30
|
Yao Y, Cai X, Fei W, Ye Y, Zhao M, Zheng C. The role of short-chain fatty acids in immunity, inflammation and metabolism. Crit Rev Food Sci Nutr 2020; 62:1-12. [PMID: 33261516 DOI: 10.1080/10408398.2020.1854675] [Citation(s) in RCA: 321] [Impact Index Per Article: 64.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Short-chain fatty acids (SCFAs) are carboxylic acids with carbon atom numbers less than 6, which are important metabolites of gut microbiome. Existing research shows that SCFAs play a vital role in the health and disease of the host. First, SCFAs are the key energy source for colon and ileum cells, and affect the intestinal epithelial barrier and defense functions by regulating related gene expression. Second, SCFAs regulate the function of innate immune cells to participate in the immune system, such as macrophages, neutrophils and dendritic cells. Third, SCFAs can also regulate the differentiation of T cells and B cells and the antigen-specific adaptive immunity mediated by them. Besides, SCFAs are raw materials for sugar and lipid synthesis, which provides a theoretical basis for studying the potential role of SCFAs in regulating energy homeostasis and metabolism. There are also studies showing that SCFAs inhibit tumor cell proliferation and promote apoptosis. In this article, we summarized in detail the role of SCFAs in immunity, inflammation and metabolism, and briefly introduced the role of SCFAs in tumor cell survival. It provides a systematic theoretical basis for the study of SCFAs as potential drugs to promote human health.
Collapse
Affiliation(s)
- Yao Yao
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Xiaoyu Cai
- Department of Pharmacy, Hangzhou First People's Hospital, Hangzhou, China
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Yiqing Ye
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Mengdan Zhao
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| |
Collapse
|
31
|
Carpita B, Marazziti D, Palego L, Giannaccini G, Betti L, Dell'Osso L. Microbiota, Immune System and Autism Spectrum Disorders: An Integrative Model towards Novel Treatment Options. Curr Med Chem 2020; 27:5119-5136. [PMID: 31448708 DOI: 10.2174/0929867326666190328151539] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Autism Spectrum Disorder (ASD) is a condition strongly associated with genetic predisposition and familial aggregation. Among ASD patients, different levels of symptoms severity are detectable, while the presence of intermediate autism phenotypes in close relatives of ASD probands is also known in literature. Recently, increasing attention has been paid to environmental factors that might play a role in modulating the relationship between genomic risk and development and severity of ASD. Within this framework, an increasing body of evidence has stressed a possible role of both gut microbiota and inflammation in the pathophysiology of neurodevelopment. The aim of this paper is to review findings about the link between microbiota dysbiosis, inflammation and ASD. METHODS Articles ranging from 1990 to 2018 were identified on PUBMED and Google Scholar databases, with keyword combinations as: microbiota, immune system, inflammation, ASD, autism, broad autism phenotype, adult. RESULTS Recent evidence suggests that microbiota alterations, immune system and neurodevelopment may be deeply intertwined, shaping each other during early life. However, results from both animal models and human samples are still heterogeneous, while few studies focused on adult patients and ASD intermediate phenotypes. CONCLUSION A better understanding of these pathways, within an integrative framework between central and peripheral systems, might not only shed more light on neural basis of ASD symptoms, clarifying brain pathophysiology, but it may also allow to develop new therapeutic strategies for these disorders, still poorly responsive to available treatments.
Collapse
Affiliation(s)
- Barbara Carpita
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Donatella Marazziti
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Lionella Palego
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Gino Giannaccini
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Laura Betti
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Liliana Dell'Osso
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| |
Collapse
|
32
|
Gut Microbiota Profile in Patients with Type 1 Diabetes Based on 16S rRNA Gene Sequencing: A Systematic Review. DISEASE MARKERS 2020; 2020:3936247. [PMID: 32908614 PMCID: PMC7474751 DOI: 10.1155/2020/3936247] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 08/07/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023]
Abstract
The gut microbiota has been presumed to have a role in the pathogenesis of type 1 diabetes (T1D). Significant changes in the microbial composition of T1D patients have been reported in several case-control studies. This study is aimed at systematically reviewing the existing literature, which has investigated the alterations of the intestinal microbiome in T1D patients compared with healthy controls (HCs) using 16S ribosomal RNA-targeted sequencing. The databases of MEDLINE, EMBASE, Web of Science, and the Cochrane Library were searched until April 2019 for case-control studies comparing the composition of the intestinal microbiome in T1D patients and HCs based on 16S rRNA gene sequencing techniques. The Newcastle-Ottawa Scale was used to assess the methodological quality. Ten articles involving 260 patients with T1D and 276 HCs were included in this systematic review. The quality scores of all included studies were 6–8 points. In summary, a decreased microbiota diversity and a significantly distinct pattern of clustering with regard to β-diversity were observed in T1D patients when compared with HCs. At the phylum level, T1D was characterised by a reduced ratio of Firmicutes/Bacteroidetes in the structure of the gut community, although no consistent conclusion was reached. At the genus or species level, T1D patients had a reduced abundance of Clostridium and Prevotella compared with HCs, whereas Bacteroides and Ruminococcus were found to be more enriched in T1D patients. This systematic review identified that there is a close association between the gut microbiota and development of T1D. Moreover, gut dysbiosis might be involved in the pathogenesis of T1D, although the causative role of gut microbiota remains to be established. Further well-controlled prospective studies are needed to better understand the role of the intestinal microbiome in the pathogenesis of T1D, which may help explore novel microbiota-based strategies to prevent and treat T1D.
Collapse
|
33
|
Li MM, Zhou Y, Zuo L, Nie D, Li XA. Dietary fiber regulates intestinal flora and suppresses liver and systemic inflammation to alleviate liver fibrosis in mice. Nutrition 2020; 81:110959. [PMID: 33059126 DOI: 10.1016/j.nut.2020.110959] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Liver fibrosis is a common pathologic process related to chronic liver disease. However, there are currently no effective methods to reverse liver fibrosis. Chronic liver disease is typically associated with a major imbalance in the intestinal flora, and targeting the regulation of the intestinal flora structure may facilitate the prevention and treatment of chronic liver disease. Therefore, in this study, we explored the effects of dietary fiber on the prevention of liver fibrosis in mice. METHODS C57BL/6J mice were randomly divided into 4 groups: olive oil group (control), fibrosis (CCl4) group, resistant maltodextrin (RM) + CCl4 group, and wheat fiber (WF) + CCl4 group. In the latter 3 groups, liver fibrosis was established by treatment with CCl4. In the RM + CCl4 and WF + CCl4 groups, the mice were treated with soluble dietary fiber (RM) or insoluble dietary fiber (WF) for 3 wk before receiving CCl4. The effects of dietary fiber on various indexes of liver fibrosis in mice induced by CCl4 were observed. RESULTS The results showed that increasing dietary fiber intake prevented liver fibrosis in mice, reduced serum levels of proinflammatory factors (e.g., tumor necrosis factor-alpha, interleukin [IL] 1-beta and IL-6) and increased IL-10 and interferon-gamma levels. Moreover, increased dietary fiber intake also reduced the infiltration of cluster of differentiation (CD) 3+, 4+, and 8+ T lymphocytes in the liver, regulated the structure of the intestinal flora, and increased the Bacteroidetes/Firmicutes ratio. CONCLUSIONS Our findings revealed the complex relationships between dietary fiber, intestinal flora, and immunity, and suggested that dietary therapy could alleviate liver fibrosis.
Collapse
Affiliation(s)
- Ming-Mei Li
- The Gastroenterology Tumor and Microenvironment Laboratory, Department Of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Yan Zhou
- The Gastroenterology Tumor and Microenvironment Laboratory, Department Of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Luo Zuo
- The Gastroenterology Tumor and Microenvironment Laboratory, Department Of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Dan Nie
- The Gastroenterology Tumor and Microenvironment Laboratory, Department Of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China
| | - Xiao-An Li
- The Gastroenterology Tumor and Microenvironment Laboratory, Department Of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, Sichuan, People's Republic of China; Department of Gastroenterology, Mianyang Central Hospital, Mianyang, Sichuan, People's Republic of China.
| |
Collapse
|
34
|
Zhai S, Qin S, Li L, Zhu L, Zou Z, Wang L. Dietary butyrate suppresses inflammation through modulating gut microbiota in high-fat diet-fed mice. FEMS Microbiol Lett 2020; 366:5531309. [PMID: 31295342 DOI: 10.1093/femsle/fnz153] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 07/10/2019] [Indexed: 11/14/2022] Open
Abstract
Butyrate, a key metabolite fermented by gut microbiota mainly from undigested carbohydrates such as dietary fibers is widely used as feed additive. However, mechanisms of its contributions in maintaining host health are relatively poorly revealed. The aim of this study was to investigate how butyrate impacts gut microbiota and immunity response in high-fat diet-fed mice. Gut microbial analysis exhibited that butyrate intervention increased short-chain fatty acids (SCFAs)-producing bacteria and decreased pathogenic bacteria, such as endotoxin-secreting bacteria. Our result also demonstrated that butyrate intervention enhanced fecal SCFAs concentrations, and inhibited endotoxin levels in feces and serum. Correlation analysis indicated positive relation between endotoxin level and Desulfovibrionaceae abundance. Furthermore, butyrate intervention inhibited expressions of IL-1β, IL-6 and MCP1/CCL2 in liver, as well as TLR4 in adipose tissue. Apart from inhibiting expressions of proinflammatory cytokines, butyrate exerted anti-inflammation effect through selectively modulating gut microbiota, such as increasing SCFAs-producing bacteria and decreasing endotoxin-secreting bacteria, as well as via regulating levels of microbiota-dependent metabolites and components, such as SCFAs and endotoxin.
Collapse
Affiliation(s)
- Shixiang Zhai
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China.,College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Song Qin
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China
| | - Lili Li
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China
| | - Limeng Zhu
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China.,Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100080, China
| | - Zhiqiang Zou
- Department of Hepatology, Infectious Disease Hospital of Yantai, Yantai 264001, China
| | - Li Wang
- Department of Hepatology, Infectious Disease Hospital of Yantai, Yantai 264001, China
| |
Collapse
|
35
|
Association of gut microbiota composition and function with an aged rat model of senile osteoporosis using 16S rRNA and metagenomic sequencing analysis. Aging (Albany NY) 2020; 12:10795-10808. [PMID: 32487781 PMCID: PMC7346068 DOI: 10.18632/aging.103293] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022]
Abstract
Recently, more interest has been paid to the association between bone mass and gut microecological dysbiosis. The results of clinical studies comparing gut microbiota (GM) in osteoporosis patients have been inconsistent due to different inclusion and exclusion criteria. To date, the association between the GM and senile osteoporosis remains poorly understood. Here, we utilized an aged rat model (22 months old) of senile osteoporosis to study the association of the composition and function of the GM with osteoporosis by 16S rRNA and metagenomic sequencing. The results showed that there was a significant reduction in alpha diversity and the F/B (Firmicutes/Bacteroidetes) ratio in aged rats. At the genus level, the enrichment of Helicobacter was potentially related to osteoporosis as a risk factor. Metagenomics results based on two databases indicated that shifts in the GM contribute to senile osteoporosis through metabolic pathways and subsequent immune disorders. In conclusion, our study reveals the association of gut microbiota composition and function with senile osteoporosis in an aged rat model in a brand new way, and variations in the GM might contribute to senile osteoporosis through metabolic pathways.
Collapse
|
36
|
Chen D, Ding Y, Ye H, Sun Y, Zeng X. Effect of long-term consumption of tea (Camellia sinensis L.) flower polysaccharides on maintaining intestinal health in BALB/c mice. J Food Sci 2020; 85:1948-1955. [PMID: 32424941 DOI: 10.1111/1750-3841.15155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 03/23/2020] [Accepted: 04/05/2020] [Indexed: 12/19/2022]
Abstract
Polysaccharides have various health-promoting functions. However, dietary polysaccharides cannot be digested by the human alimentary tract, thus the gut is the most important location where polysaccharides play their role. The effect of polysaccharides from tea (Camellia sinensis L.) flower (TFPS) on intestinal health was investigated in the present study. TFPS with the molecular weight of 1,316.29 kDa was prepared, and twenty 6-week-old BALB/c male mice were randomly allotted to a chow diet (normal control group, NC group) or with 200 mg/kg (body weight)/day of TFPS for 13 weeks (n = 10 each). Histomorphology observation of jejunum and colons showed that TFPS maintained the adequate gut barrier. qPCR analysis revealed that the expression of colonic tight junction proteins of claudin1 (1.29 ± 0.15 compared with 1.00 ± 0.13, P < 0.05) and claudin5 (2.91 ± 0.44 compared with 1.00 ± 0.27, P < 0.01) at mRNA level with a significant difference between TFPS supplement or not, while the expression of TLR4 and TNF-α mRNA was not changed statistically. 16S rDNA amplicons sequencing was applied to measure the compositions of gut microbiota from feces of mice. TFPS treatment exhibited similar relative abundances in Bacteroidetes and Firmicutes; however, it decreased the relative abundance of Akkermansia and increased that of Lactobacillus compared with the NC group. The contents of short-chain fatty acids after TFPS supplementation, both in cecal contents and feces, were significantly higher than those of the NC group. Besides, TFPS significantly increased IgA production. These results suggest that TFPS is beneficial to intestinal health and can improve intestinal adaptive immune tolerance. PRACTICAL APPLICATION: Dietary polysaccharides improve human intestinal health. Understanding the effect of TFPS, safe and healthy food components, on gut health increases the likelihood that TFPS will be developed as a functional food.
Collapse
Affiliation(s)
- Dan Chen
- College of Food Science and Technology, Nanjing Agricultural Univ., Nanjing, 210095, People's Republic of China
| | - Yu Ding
- College of Food Science and Technology, Nanjing Agricultural Univ., Nanjing, 210095, People's Republic of China
| | - Hong Ye
- College of Food Science and Technology, Nanjing Agricultural Univ., Nanjing, 210095, People's Republic of China
| | - Yi Sun
- College of Food Science and Technology, Nanjing Agricultural Univ., Nanjing, 210095, People's Republic of China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural Univ., Nanjing, 210095, People's Republic of China
| |
Collapse
|
37
|
Zhang F, Xiang X, Dong Y, Yan S, Song Y, Zhou L. Significant Differences in the Gut Bacterial Communities of Hooded Crane ( Grus monacha) in Different Seasons at a Stopover Site on the Flyway. Animals (Basel) 2020; 10:E701. [PMID: 32316467 PMCID: PMC7222709 DOI: 10.3390/ani10040701] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/10/2020] [Accepted: 04/15/2020] [Indexed: 02/08/2023] Open
Abstract
Intestinal bacterial communities form an integral component of the organism. Many factors influence gut bacterial community composition and diversity, including diet, environment and seasonality. During seasonal migration, birds use many habitats and food resources, which may influence their intestinal bacterial community structure. Hooded crane (Grus monacha) is a migrant waterbird that traverses long distances and occupies varied habitats. In this study, we investigated the diversity and differences in intestinal bacterial communities of hooded cranes over the migratory seasons. Fecal samples from hooded cranes were collected at a stopover site in two seasons (spring and fall) in Lindian, China, and at a wintering ground in Shengjin Lake, China. We analyzed bacterial communities from the fecal samples using high throughput sequencing (Illumina Mi-seq). Firmicutes, Proteobacteria, Tenericutes, Cyanobacteria, and Actinobacteria were the dominant phyla across all samples. The intestinal bacterial alpha-diversity of hooded cranes in winter was significantly higher than in fall and spring. The bacterial community composition significantly differed across the three seasons (ANOSIM, P = 0.001), suggesting that seasonal fluctuations may regulate the gut bacterial community composition of migratory birds. This study provides baseline information on the seasonal dynamics of intestinal bacterial community structure in migratory hooded cranes.
Collapse
Affiliation(s)
- Fengling Zhang
- School of Resources and Environmental Engineering, Anhui University, Hefei 230601, China
- Anhui Province Key Laboratory of Wetland Ecosystem Protection and Restoration (Anhui University), Hefei 230601, China
| | - Xingjia Xiang
- School of Resources and Environmental Engineering, Anhui University, Hefei 230601, China
- Anhui Province Key Laboratory of Wetland Ecosystem Protection and Restoration (Anhui University), Hefei 230601, China
| | - Yuanqiu Dong
- School of Resources and Environmental Engineering, Anhui University, Hefei 230601, China
- Anhui Province Key Laboratory of Wetland Ecosystem Protection and Restoration (Anhui University), Hefei 230601, China
| | - Shaofei Yan
- School of Resources and Environmental Engineering, Anhui University, Hefei 230601, China
- Anhui Province Key Laboratory of Wetland Ecosystem Protection and Restoration (Anhui University), Hefei 230601, China
| | - Yunwei Song
- Shengjin Lake National Nature Reserve of Anhui Province, Dongzhi 247200, China
| | - Lizhi Zhou
- School of Resources and Environmental Engineering, Anhui University, Hefei 230601, China
- Anhui Province Key Laboratory of Wetland Ecosystem Protection and Restoration (Anhui University), Hefei 230601, China
| |
Collapse
|
38
|
Buzhylov M, Kaprelyants L, Pozhitkova L, Kishenya A, Soboleva A. ENZYMATIC MODIFICATION OF WHEAT RICE. FOOD SCIENCE AND TECHNOLOGY 2020. [DOI: 10.15673/fst.v14i1.1643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The article presents conceptual approaches to solving technological and technical problems in the creation of functional foods. General approaches are proposed to change existing technologies to improve the efficiency of integrated raw material processing and to increase the production of high-quality foods and food ingredients with antioxidant properties. Cereal crops are the richest source of functional ingredients and a major component of human nutrition. It is proved that most of the nutrients are in the products of its processing. For the first time, polyphenols from cereal raw materials were obtained by biotechnological means. The feasibility of pretreatment of raw materials with amylolytic and proteolytic enzymes for purification and cleavage of polysaccharide matrix has been established. Based on the regularities of enzymatic hydrolysis of polysaccharides, we used the processing of wheat bran with multifunctional drug Viscozyme L with hemicellulase, cellulase, pectinesterase and feruloesterase activities, which resulted in a high effect of degradation of certain covalent cells, ferulic acid from 40.99 to 2507.9 mcg / g. It is determined that this method of obtaining the target components allows to preserve their native structure, especially the supramolecular structure, which determines their physiological effect. The influence of plant polyphenols on the cultivation of probiotic microorganisms is characterized. the comparative characterization of the prebiotic properties of the polyphenols obtained from wheat bran and the concentrate of the polyphenols from the grape buds "ENOANT" are substantiated. The possibility of increasing the proportion of free polyphenols by fermentation of wheat bran is shown. It is established that the extract of polyphenols from wheat bran can be used for its purpose as an effective antioxidant, which does not have a negative effect on the state of the basic physiological systems of the body.
Collapse
|
39
|
De Spiegeleer A, Elewaut D, Van Den Noortgate N, Janssens Y, Debunne N, Van Langenhove S, Govindarajan S, De Spiegeleer B, Wynendaele E. Quorum sensing molecules as a novel microbial factor impacting muscle cells. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165646. [DOI: 10.1016/j.bbadis.2019.165646] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 12/15/2019] [Indexed: 02/07/2023]
|
40
|
Takai A, Kikuchi K, Ichimura M, Tsuneyama K, Moritoki Y, Matsumoto K, Tsunashima H, Onda T, Kuniyoshi N, Nariyama T, Ohyatsu S, Kubota J, Nagumo K, Sato S, Hara M, Miyakawa H. Fructo-oligosaccharides ameliorate steatohepatitis, visceral adiposity, and associated chronic inflammation via increased production of short-chain fatty acids in a mouse model of non-alcoholic steatohepatitis. BMC Gastroenterol 2020; 20:46. [PMID: 32103741 PMCID: PMC7045471 DOI: 10.1186/s12876-020-01194-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 02/17/2020] [Indexed: 02/08/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a hepatic manifestation of metabolic syndrome. Within the spectrum of NAFLD, non-alcoholic steatohepatitis (NASH) in combination with hepatic inflammation and fibrosis can lead to liver cirrhosis and hepatocellular carcinoma. Dysbiosis was reported to contribute to NASH pathogenesis. This study aimed to determine the effects of fructo-oligosaccharides (FOS) on steatohepatitis and visceral adiposity in an obese mouse model of NASH. Methods Twelve newborn C57BL/6 J male mice were subcutaneously injected with monosodium glutamate (MSG) to induce obesity on a conventional diet. Six mice were also administered 5% FOS via drinking water from 10 weeks of age. At 18 weeks, histological characteristics of the liver and epididymal fat were compared between the groups. Hepatic mRNA expression of lipid metabolism enzymes and SCFA in feces and sera were measured. Results Hepatic steatosis, inflammatory cell infiltration, and hepatocyte ballooning in the liver and increased hepatic mRNA expression of fatty acid synthase and glycerol-3-phosphate acyltransferase were observed in the MSG-treated mice. FOS treatment improved the liver pathology and blunted the increases in the mRNA expression levels of lipid metabolism enzymes. In addition, FOS inhibited adipocyte enlargement and formation of crown-like structures and reduced the M1 macrophage frequency in the epididymal fat of the MSG mice (39.4% ± 3.0% vs. 22.8% ± 0.7%; P = 0.001). FOS increased not only the fecal concentrations of n-butyric acid (0.04 ± 0.01 vs. 0.38 ± 0.14 mg/g, P = 0.02), propionic acid (0.09 ± 0.03 vs. 0.42 ± 0.16 mg/g, P = 0.02), and acetic acid (0.65 ± 0.16 vs. 1.48 ± 0.29 mg/g, P = 0.03) but also the serum concentration of propionic acid (3.9 ± 0.5 vs. 8.2 ± 0.5 μmol/L, P = 0.001). Conclusions FOS ameliorates steatohepatitis, visceral adiposity, and chronic inflammation by increasing SCFA production.
Collapse
Affiliation(s)
- Atsuko Takai
- Fourth Department of Internal Medicine, Teikyo University Mizonokuchi Hospital, 5-1-1 Futako, Takatsu-ku, Kawasaki-shi, Kanagawa, 213-8507, Japan
| | - Kentaro Kikuchi
- Fourth Department of Internal Medicine, Teikyo University Mizonokuchi Hospital, 5-1-1 Futako, Takatsu-ku, Kawasaki-shi, Kanagawa, 213-8507, Japan.
| | - Mayuko Ichimura
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima-shi, Tokushima, Japan
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima-shi, Tokushima, Japan
| | - Yuki Moritoki
- Department of General Internal Medicine and Clinical Laboratory Medicine, Akita University Graduate School of Medicine, Akita-shi, Akita, Japan
| | - Kotaro Matsumoto
- Department of Gastroenterology, Teikyo University Mizonokuchi Hospital, Kawasaki-shi, Kanagawa, Japan
| | - Hiromichi Tsunashima
- Department of Gastroenterology, Teikyo University Mizonokuchi Hospital, Kawasaki-shi, Kanagawa, Japan
| | - Takeshi Onda
- Department of Gastroenterology, Teikyo University Mizonokuchi Hospital, Kawasaki-shi, Kanagawa, Japan.,Department of Gastroenterology, Nippon Medical School Chiba Hokusoh Hospital, Inzai-shi, Chiba, Japan
| | - Noriyuki Kuniyoshi
- Department of Gastroenterology, Teikyo University Mizonokuchi Hospital, Kawasaki-shi, Kanagawa, Japan.,Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Tomoyuki Nariyama
- Fourth Department of Internal Medicine, Teikyo University Mizonokuchi Hospital, 5-1-1 Futako, Takatsu-ku, Kawasaki-shi, Kanagawa, 213-8507, Japan
| | - Sho Ohyatsu
- Fourth Department of Internal Medicine, Teikyo University Mizonokuchi Hospital, 5-1-1 Futako, Takatsu-ku, Kawasaki-shi, Kanagawa, 213-8507, Japan
| | - Juri Kubota
- Fourth Department of Internal Medicine, Teikyo University Mizonokuchi Hospital, 5-1-1 Futako, Takatsu-ku, Kawasaki-shi, Kanagawa, 213-8507, Japan
| | - Kozue Nagumo
- Fourth Department of Internal Medicine, Teikyo University Mizonokuchi Hospital, 5-1-1 Futako, Takatsu-ku, Kawasaki-shi, Kanagawa, 213-8507, Japan
| | - Shinpei Sato
- Fourth Department of Internal Medicine, Teikyo University Mizonokuchi Hospital, 5-1-1 Futako, Takatsu-ku, Kawasaki-shi, Kanagawa, 213-8507, Japan
| | - Masumi Hara
- Fourth Department of Internal Medicine, Teikyo University Mizonokuchi Hospital, 5-1-1 Futako, Takatsu-ku, Kawasaki-shi, Kanagawa, 213-8507, Japan
| | - Hiroshi Miyakawa
- Fourth Department of Internal Medicine, Teikyo University Mizonokuchi Hospital, 5-1-1 Futako, Takatsu-ku, Kawasaki-shi, Kanagawa, 213-8507, Japan
| |
Collapse
|
41
|
The Interplay between Immune System and Microbiota in Osteoporosis. Mediators Inflamm 2020; 2020:3686749. [PMID: 32184701 PMCID: PMC7061131 DOI: 10.1155/2020/3686749] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/27/2020] [Accepted: 02/04/2020] [Indexed: 01/04/2023] Open
Abstract
Osteoporosis is a disease characterized by low bone mass and alterations of bone microarchitecture, with an increased risk of fractures. It is a multifactorial disorder that is more frequent in postmenopausal women but can be associated to other diseases (inflammatory and metabolic diseases). At present, several options are available to treat osteoporosis trying to block bone reabsorption and reduce the risk of fracture. Anyway, these drugs have safety and tolerance problems in long-term treatment. Recently, gut microbiota has been highlighted to have strong influence on bone metabolism, becoming a potential new target to modify bone mineral density. Such evidences are mainly based on mouse models, showing an involvement in modulating the interaction between the immune system and bone cells. Germ-free mice represent a basic model to understand the interaction between microbiota, immune system, and bone cells, even though data are controversial. Anyway, such models have unequivocally demonstrated a connection between such systems, even if the mechanism is unclear. Gut microbiota is a complex system that influences calcium and vitamin D absorption and modulates gut permeability, hormonal secretion, and immune response. A key role is played by the T helper 17 lymphocytes, TNF, interleukin 17, and RANK ligand system. Other important pathways include NOD1, NOD2, and Toll-like receptor 5. Prebiotics and probiotics are a wide range of substances and germs that can influence and modify microbiota. Several studies demonstrated actions by different prebiotics and probiotics in different animals, differing according to sex, age, and hormonal status. Data on the effects on humans are poor and controversial. Gut microbiota manipulation appears a possible strategy to prevent and treat osteopenia and/or osteoporosis as well as other possible bone alterations, even though further clinical studies are necessary to identify correct procedures in humans.
Collapse
|
42
|
Aguilar-López BA, Moreno-Altamirano MMB, Dockrell HM, Duchen MR, Sánchez-García FJ. Mitochondria: An Integrative Hub Coordinating Circadian Rhythms, Metabolism, the Microbiome, and Immunity. Front Cell Dev Biol 2020; 8:51. [PMID: 32117978 PMCID: PMC7025554 DOI: 10.3389/fcell.2020.00051] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/20/2020] [Indexed: 12/25/2022] Open
Abstract
There is currently some understanding of the mechanisms that underpin the interactions between circadian rhythmicity and immunity, metabolism and immune response, and circadian rhythmicity and metabolism. In addition, a wealth of studies have led to the conclusion that the commensal microbiota (mainly bacteria) within the intestine contributes to host homeostasis by regulating circadian rhythmicity, metabolism, and the immune system. Experimental studies on how these four biological domains interact with each other have mainly focused on any two of those domains at a time and only occasionally on three. However, a systematic analysis of how these four domains concurrently interact with each other seems to be missing. We have analyzed current evidence that signposts a role for mitochondria as a key hub that supports and integrates activity across all four domains, circadian clocks, metabolic pathways, the intestinal microbiota, and the immune system, coordinating their integration and crosstalk. This work will hopefully provide a new perspective for both hypothesis-building and more systematic experimental approaches.
Collapse
Affiliation(s)
- Bruno A Aguilar-López
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Hazel M Dockrell
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Michael R Duchen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Francisco Javier Sánchez-García
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
43
|
Zhou H, Sun L, Zhang S, Zhao X, Gang X, Wang G. Evaluating the Causal Role of Gut Microbiota in Type 1 Diabetes and Its Possible Pathogenic Mechanisms. Front Endocrinol (Lausanne) 2020; 11:125. [PMID: 32265832 PMCID: PMC7105744 DOI: 10.3389/fendo.2020.00125] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 02/25/2020] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) is a multifactorial autoimmune disease mediated by genetic, epigenetic, and environmental factors. In recent years, the emergence of high-throughput sequencing has allowed us to investigate the role of gut microbiota in the development of T1D. Significant changes in the composition of gut microbiome, also termed dysbiosis, have been found in subjects with clinical or preclinical T1D. However, whether the dysbiosis is a cause or an effect of the disease remains unclear. Currently, increasing evidence has supported a causal link between intestine microflora and T1D development. The current review will focus on recent research regarding the associations between intestine microbiome and T1D progression with an intention to evaluate the causality. We will also discuss the possible mechanisms by which imbalanced gut microbiota leads to the development of T1D.
Collapse
|
44
|
Abstract
Chronic heart failure, diabetes, depression, and other chronic diseases are associated with high mortality rate and low cure rate. Exercise induces muscle contraction and secretes multiple myokines, which affects the signaling pathways in skeletal muscle tissues and regulate remote organ functions. Exercise is known to be effective in treating a variety of chronic diseases. Here we summarize how exercise influences skeletal muscle, heart, brain, gut, and liver, and prevents heart failure, cognitive dysfunction, obesity, fatty liver, and other diseases. Exercise training may achieve additional benefits as compared to the present medication for these chronic diseases through cross talk among skeletal muscle and other organs.
Collapse
Affiliation(s)
- Zhiqing Fan
- Department of Cardiology, Daqing Qilfield General Hospital, Daqing, China
| | - Minjun Xu
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| |
Collapse
|
45
|
The Potential Influence of the Bacterial Microbiome on the Development and Progression of ADHD. Nutrients 2019; 11:nu11112805. [PMID: 31744191 PMCID: PMC6893446 DOI: 10.3390/nu11112805] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
The latest research cumulates staggering information about the correlation between the microbiota-gut-brain axis and neurodevelopmental disorders. This review aims to shed light on the potential influence of the microbiome on the development of the most prevalent neurodevelopmental disease, attention-deficit-hyperactive disorder (ADHD). As the etiology and pathophysiology of ADHD are still unclear, finding viable biomarkers and effective treatment still represent a challenge. Therefore, we focused on factors that have been associated with a higher risk of developing ADHD, while simultaneously influencing the microbial composition. We reviewed the effect of a differing microbial makeup on neurotransmitter concentrations important in the pathophysiology of ADHD. Additionally, we deduced factors that correlate with a high prevalence of ADHD, while simultaneously affecting the gut microbiome, such as emergency c-sections, and premature birth as the former leads to a decrease of the gut microbial diversity and the latter causes neuroprotective Lactobacillus levels to be reduced. Also, we assessed nutritional influences, such as breastfeeding, ingestion of short-chain fatty acids (SCFAs) and polyunsaturated fatty acids (PUFAs) on the host′s microbiome and development of ADHD. Finally, we discussed the potential significance of Bifidobacterium as a biomarker for ADHD, the importance of preventing premature birth as prophylaxis and nutrition as a prospective therapeutic measurement against ADHD.
Collapse
|
46
|
De Spiegeleer A, Elewaut D, Van Den Noortgate N, Janssens Y, Debunne N, Van Langenhove S, Govindarajan S, De Spiegeleer B, Wynendaele E. WITHDRAWN: This article has been withdrawn. Biochim Biophys Acta Mol Basis Dis 2019:165585. [PMID: 31678164 DOI: 10.1016/j.bbadis.2019.165585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/11/2019] [Accepted: 09/22/2019] [Indexed: 11/21/2022]
Abstract
This article has been withdrawn at the request of the author for administrative reasons. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Anton De Spiegeleer
- Department of Geriatrics, Faculty of Medicine and Health Sciences, Ghent University Hospital, C. Heymanslaan 10, 9000 Ghent, Belgium; Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Unit for Molecular Immunology and Inflammation, VIB-Center for Inflammation Research, Technologiepark 71, 9052, Zwijnaarde, Ghent, Belgium
| | - Dirk Elewaut
- Unit for Molecular Immunology and Inflammation, VIB-Center for Inflammation Research, Technologiepark 71, 9052, Zwijnaarde, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University Hospital, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - Nele Van Den Noortgate
- Department of Geriatrics, Faculty of Medicine and Health Sciences, Ghent University Hospital, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - Yorick Janssens
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Nathan Debunne
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Selien Van Langenhove
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Srinath Govindarajan
- Unit for Molecular Immunology and Inflammation, VIB-Center for Inflammation Research, Technologiepark 71, 9052, Zwijnaarde, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University Hospital, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - Bart De Spiegeleer
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Evelien Wynendaele
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
47
|
Nicoletti A, Ponziani FR, Biolato M, Valenza V, Marrone G, Sganga G, Gasbarrini A, Miele L, Grieco A. Intestinal permeability in the pathogenesis of liver damage: From non-alcoholic fatty liver disease to liver transplantation. World J Gastroenterol 2019; 25:4814-4834. [PMID: 31543676 PMCID: PMC6737313 DOI: 10.3748/wjg.v25.i33.4814] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/04/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
The intimate connection and the strict mutual cooperation between the gut and the liver realizes a functional entity called gut-liver axis. The integrity of intestinal barrier is crucial for the maintenance of liver homeostasis. In this mutual relationship, the liver acts as a second firewall towards potentially harmful substances translocated from the gut, and is, in turn, is implicated in the regulation of the barrier. Increasing evidence has highlighted the relevance of increased intestinal permeability and consequent bacterial translocation in the development of liver damage. In particular, in patients with non-alcoholic fatty liver disease recent hypotheses are considering intestinal permeability impairment, diet and gut dysbiosis as the primary pathogenic trigger. In advanced liver disease, intestinal permeability is enhanced by portal hypertension. The clinical consequence is an increased bacterial translocation that further worsens liver damage. Furthermore, this pathogenic mechanism is implicated in most of liver cirrhosis complications, such as spontaneous bacterial peritonitis, hepatorenal syndrome, portal vein thrombosis, hepatic encephalopathy, and hepatocellular carcinoma. After liver transplantation, the decrease in portal pressure should determine beneficial effects on the gut-liver axis, although are incompletely understood data on the modifications of the intestinal permeability and gut microbiota composition are still lacking. How the modulation of the intestinal permeability could prevent the initiation and progression of liver disease is still an uncovered area, which deserves further attention.
Collapse
Affiliation(s)
- Alberto Nicoletti
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Francesca Romana Ponziani
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Marco Biolato
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Venanzio Valenza
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Giuseppe Marrone
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Gabriele Sganga
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Antonio Gasbarrini
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Luca Miele
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Antonio Grieco
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| |
Collapse
|
48
|
Gupta S, Fernandes J, Kiron V. Antibiotic-Induced Perturbations Are Manifested in the Dominant Intestinal Bacterial Phyla of Atlantic Salmon. Microorganisms 2019; 7:E233. [PMID: 31382431 PMCID: PMC6723382 DOI: 10.3390/microorganisms7080233] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 12/25/2022] Open
Abstract
The intestinal microbiota of certain farmed fish are often exposed to antimicrobial substances, such as antibiotics, that are used to prevent and treat bacterial diseases. Antibiotics that kill or inhibit the growth of harmful microbes can rapidly alter intestinal microbial diversity and composition, with potential effects on the host health. In this study, we have elucidated the impact of two antibiotics, florfenicol and oxolinic acid, by employing a high-throughput 16S rRNA gene amplicon sequencing technique on the distal and mid intestinal microbial communities of Atlantic salmon (Salmo salar). For this, Atlantic salmon were offered diets with or without antibiotics. We then investigated the bacterial communities in the intestinal mucus of the fish. Our results showed that antibiotic exposure shifts the intestinal microbial profile differentially. In addition, the bacterial compositions of the control and antibiotic-fed groups were significantly different. Antibiotic feeding altered the composition and abundance of the dominant bacterial phyla, namely Proteobacteria, Actinobacteria, Firmicutes, Spirochaetes, Bacteroidetes, Tenericutes, and Thermotogae. The bacterial association network analysis also indicated the differential pattern of co-occurrence of bacteria in the three study groups. The results regarding the differences in the structure and association of the intestinal microbiota of Atlantic salmon after florfenicol and oxolinic acid feeding can be employed to attenuate the adverse effects of antibiotic feeding on fish.
Collapse
Affiliation(s)
- Shruti Gupta
- Faculty of Biosciences and Aquaculture, Nord University, 8049 Bodø, Norway
| | - Jorge Fernandes
- Faculty of Biosciences and Aquaculture, Nord University, 8049 Bodø, Norway
| | - Viswanath Kiron
- Faculty of Biosciences and Aquaculture, Nord University, 8049 Bodø, Norway.
| |
Collapse
|
49
|
Basmaciyan L, Bon F, Paradis T, Lapaquette P, Dalle F. " Candida Albicans Interactions With The Host: Crossing The Intestinal Epithelial Barrier". Tissue Barriers 2019; 7:1612661. [PMID: 31189436 PMCID: PMC6619947 DOI: 10.1080/21688370.2019.1612661] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 02/08/2023] Open
Abstract
Formerly a commensal organism of the mucosal surfaces of most healthy individuals, Candida albicans is an opportunistic pathogen that causes infections ranging from superficial to the more life-threatening disseminated infections, especially in the ever-growing population of vulnerable patients in the hospital setting. In these situations, the fungus takes advantage of its host following a disturbance in the host defense system and/or the mucosal microbiota. Overwhelming evidence suggests that the gastrointestinal tract is the main source of disseminated C. albicans infections. Major risk factors for disseminated candidiasis include damage to the mucosal intestinal barrier, immune dysfunction, and dysbiosis of the resident microbiota. A better understanding of C. albicans' interaction with the intestinal epithelial barrier will be useful for designing future therapies to avoid systemic candidiasis. In this review, we provide an overview of the current knowledge regarding the mechanisms of pathogenicity that allow the fungus to reach and translocate the gut barrier.
Collapse
Affiliation(s)
- Louise Basmaciyan
- Laboratoire de Parasitologie-Mycologie, Plateforme de Biologie Hospitalo-Universitaire Gérard Mack, Dijon France
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Fabienne Bon
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Tracy Paradis
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Pierre Lapaquette
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Frédéric Dalle
- Laboratoire de Parasitologie-Mycologie, Plateforme de Biologie Hospitalo-Universitaire Gérard Mack, Dijon France
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| |
Collapse
|
50
|
Impact of endurance exercise and probiotic supplementation on the intestinal microbiota: a cross-over pilot study. Pilot Feasibility Stud 2019; 5:76. [PMID: 31198580 PMCID: PMC6556026 DOI: 10.1186/s40814-019-0459-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 05/13/2019] [Indexed: 02/08/2023] Open
Abstract
Background The human microbiota has a broad range of functions contributing to metabolic processes and the activities of our immune system. Its influence on health, well-being and chronic diseases are discussed in various studies. The intestinal microbiota and the mucosal integrity are influenced by diet, environment and other lifestyle factors, including physical activity. There are correlations between cardiorespiratory fitness and important markers of intestinal health. However, data linking endurance exercise to microbiota composition are sparse. Many endurance athletes take probiotics to reduce gastrointestinal symptoms linked to exercise or immunosuppression, but the longitudinal data is insufficient. This randomised, controlled cross-over pilot study will examine the impact of specific endurance training and probiotic supplementation on the intestinal microbiota and mucosa in healthy, athletic students. Objective The aim of this pilot study is to elucidate the impact of physical activity on the intestinal microbiota and mucosa with regard to the effects of a probiotic supplementation. Methods In this pilot study, thirty non-specifically trained student athletes will participate in an intervention consisting of a two-week rest (baseline) period, a four-week exercise programme and a four-week probiotic intervention using SymbioLactComp®. The exercise programme consists of three 60-min running workouts per week at 70–85% of the peak heart rate (HRpeak). Primary endpoint of this pilot study is the feasibility and practicality of the intervention as well as a sample size estimation. Furthermore, anthropometric measurements and information on nutrition and lifestyle will be obtained. The peak oxygen uptake (VO2peak) and peak heart rate (HRpeak) (determined during a shuttle run test) as well as selected blood and saliva parameters (haemogram, cytokines) will be evaluated. Changes to the intestinal microbiota will be analysed by stool diagnostics (KyberKompaktPRO®, KyberPlus®). The potential changes may include microbiota composition, bacterial metabolites and mucosa- and immune markers. Conclusion Results will be used for the design of a main randomised controlled trial with a larger collective based on feasibility, validity and sample size estimation as well as the potential effects of endurance exercise on intestinal microbiota and mucosa. Evidence-based information of an exercise-altered microbiota could be of importance for the prevention and therapy of intestinal or immune disorders. Trial registration German Clinical Trials Register: DRKS00011108. Retrospectively registered on 28 November 2016. Electronic supplementary material The online version of this article (10.1186/s40814-019-0459-9) contains supplementary material, which is available to authorized users.
Collapse
|