1
|
Slager J, Simpson HL, Gacesa R, Chen L, Tan IL, Gelderloos J, Maatman A, Wijmenga C, Zhernakova A, Fu J, Weersma RK, Gonera G, Jonkers IH, Withoff S. High-resolution analysis of the treated coeliac disease microbiome reveals strain-level variation. Gut Microbes 2025; 17:2489071. [PMID: 40289251 PMCID: PMC12036492 DOI: 10.1080/19490976.2025.2489071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/05/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Coeliac disease (CeD) is an immune-mediated disorder primarily affecting the small intestine, characterized by an inflammatory immune reaction to dietary gluten. CeD onset results from a multifaceted interplay of genetic and environmental factors. While recent data show that alterations in gut microbiome composition could play an important role, many current studies are constrained by small sample sizes and limited resolution. METHODS To address these limitations, we analyzed fecal gut microbiota from two Dutch cohorts, CeDNN (128 treated CeD patients (tCeD), 106 controls) and the Lifelines Dutch Microbiome Project (24 self-reported tCeD, 654 controls), using shotgun metagenomic sequencing. Self-reported IBS (570 cases, 1710 controls) and IBD (93 cases, 465 controls) were used as comparative conditions of the gastrointestinal tract. Interindividual variation within the case and control groups was calculated at whole microbiome and strain level. Finally, species-specific gene repertoires were analyzed in tCeD patients and controls. RESULTS Within-individual microbiome diversity was decreased in patients with self-reported IBS and IBD but not in tCeD patients. Each condition displayed a unique microbial pattern and, in addition to confirming previously reported microbiome associations, we identify an increase in the levels of Clostridium sp. CAG:253, Roseburia hominis, and Eggerthella lenta, amongst others. We further show that the observed changes can partially be explained by gluten-free diet adherence. We also observe increased interindividual variation of gut microbiome composition among tCeD patients and a higher bacterial mutation frequency in tCeD that contributes to higher interindividual variation at strain level. In addition, the immotile European subspecies of Eubacterium rectale, which has a distinct carbohydrate metabolism potential, was nearly absent in tCeD patients. CONCLUSION Our study sheds light on the complex interplay between the gut microbiome and CeD, revealing increased interindividual variation and strain-level variation in tCeD patients. These findings expand our understanding of the microbiome's role in intestinal health and disease.
Collapse
Affiliation(s)
- Jelle Slager
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hanna L. Simpson
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ranko Gacesa
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Lianmin Chen
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Cardiology, Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Ineke L. Tan
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jody Gelderloos
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Astrid Maatman
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rinse K. Weersma
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gieneke Gonera
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pediatrics, Wilhelmina Hospital Assen, Assen, The Netherlands
| | - Iris H. Jonkers
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sebo Withoff
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
2
|
Mugo CW, Church E, Horniblow RD, Mollan SP, Botfield H, Hill LJ, Sinclair AJ, Grech O. Unravelling the gut-brain connection: a systematic review of migraine and the gut microbiome. J Headache Pain 2025; 26:125. [PMID: 40399789 PMCID: PMC12096802 DOI: 10.1186/s10194-025-02039-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/16/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND There is substantial evidence linking migraines to gastrointestinal (GI) issues. Conditions such as irritable bowel syndrome and colitis often co-occur with migraines and GI symptoms are common among migraine patients. However, the evidence supporting the efficacy of gut microbiome-targeted therapies for managing migraines is limited. This systematic review aimed to describe the existing evidence of the gut microbiome in patients with migraine compared to healthy individuals. Additionally, it sought to examine how therapies targeting the gut microbiome including prebiotics, probiotics and synbiotics, might influence clinical outcomes. METHODS We performed searches on Embase, PubMed, and the Cochrane Library to identify studies in migraines and the gut microbiome, focusing on those which investigated the gut microbiome composition and gut microbiome-targeted therapies. Key data was extracted and analysed including study details, patient demographics, migraine type, comorbidities, and clinical outcomes. For gut microbiome composition studies, bacterial diversity and abundance was noted. For gut microbiome-targeted therapies studies, treatment types, dosages, and patient outcomes was recorded. RESULTS A significant difference between various genera of microbes was reported between migraine patients and controls in several studies. Bacteroidetes (also named Bacteroidota), proteobacteria, and firmicutes (also named Bacillota) phyla groups were found significantly abundant in migraine, while studies were conflicted in the abundance of Actinobacteria and Clostridia with regards to increased migraine risk in migraine patients. Patients with migraine had a gut microbiome with reduced species number and relative abundance, as well as a distinct bacterial composition compared to controls. Synbiotic and synbiotic/probiotic combination treatments have been shown in five randomised controlled trials and one open label pilot study to significantly decrease migraine severity, frequency, duration and painkiller consumption. CONCLUSIONS The significant alterations in microbial phyla observed in migraine patients suggest a potential microbial signature that may be associated with migraine risk or chronic progression. However, the mechanistic underpinnings of these associations remain unclear. This systemic review found that probiotic and synbiotic/probiotic combination therapies may be promising interventions for migraine management, offering significant reductions in migraine frequency and painkiller use. Future randomised controlled studies are needed to evaluate the optimal length of treatment and impact on patient related quality of life.
Collapse
Affiliation(s)
- Caroline W Mugo
- Biomedical Sciences, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ella Church
- Biomedical Sciences, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
| | - Richard D Horniblow
- Biomedical Sciences, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
| | - Susan P Mollan
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- Neuro-Ophthalmology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2GW, UK
| | - Hannah Botfield
- Inflammation and Ageing, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
| | - Lisa J Hill
- Biomedical Sciences, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2GW, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, B15 2TH, UK
| | - Alexandra J Sinclair
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2GW, UK.
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, B15 2TH, UK.
| | - Olivia Grech
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
3
|
Ye H, Meehan D, Timmons S, O'Toole PW. Effects of Prebiotics and a Synthetic Microbiome Consortium on the Composition and Metabolites of the Elderly Gut Microbiota In Vitro. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11720-11729. [PMID: 40324006 DOI: 10.1021/acs.jafc.5c00364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
A 24 h artificial colon fermentation was performed to assess the effects of a prebiotic MIX and synthetic consortium (S7) on gut microbiota composition and microbial metabolite production in clinically stratified older adults (healthy and frail). Treatments included supplementation of the donor microbiota with a synthetic microbial consortium (S7), a prebiotic mix (MIX), and a combination of MIX and S7 (MIX+S7). The S7 treatment decreased the alpha diversity of long-stay-dwelling donor (LS, "frail") microbiota and increased the relative abundance of S7 taxa at 16 h. MIX alone caused the enrichment of reportedly beneficial genera such as Coprobacillus and Eubacterium in community-dwelling donor (CM, "healthy") microbiota and Citrobacter and Faecalibacterium in LS microbiota. The MIX+S7 treatment sustained higher overall S7 species richness and consortium taxon abundance at 24 h. Both MIX and MIX+S7 treatments enhanced short-chain acid production compared to control. These findings highlight the differential responses of microbiota from distinct elderly health strata to prebiotic and microbial consortia supplementation.
Collapse
Affiliation(s)
- Huimin Ye
- School of Microbiology, and APC Microbiome Ireland, University College Cork, Western Road, T12 Y337 Cork, Ireland
| | - Dara Meehan
- School of Microbiology, and APC Microbiome Ireland, University College Cork, Western Road, T12 Y337 Cork, Ireland
| | - Suzanne Timmons
- Centre for Gerontology and Rehabilitation, School of Medicine, University College Cork, Western Road, T12 Y337 Cork, Ireland
| | - Paul W O'Toole
- School of Microbiology, and APC Microbiome Ireland, University College Cork, Western Road, T12 Y337 Cork, Ireland
| |
Collapse
|
4
|
Hu Y, Ni X, Chen Q, Qu Y, Chen K, Zhu G, Zhang M, Xu N, Bai X, Wang J, Ma Y, Luo Q, Cai K. Predicting diabetic kidney disease with serum metabolomics and gut microbiota. Sci Rep 2025; 15:12179. [PMID: 40204798 PMCID: PMC11982385 DOI: 10.1038/s41598-025-91281-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025] Open
Abstract
This study aims to identify biomarkers for reliably predicting diabetic kidney disease (DKD), systematically characterize serum metabolites and gut microbiota in DKD patients, and investigate the correlation between differential serum metabolites and gut microbiota. From September 2021 to January 2023, 90 subjects were recruited: 30 with DKD, 30 with type 2 diabetes mellitus (T2DM), and 30 normal controls (NCs). Serum metabolites, including 180 different metabolites, were analyzed using untargeted metabolomics UPLC-MS/MS, and gut microbiota were assessed via 16S rRNA sequencing. Differential metabolites were identified through univariate (t-test or Mann-Whitney U-test, P < 0.05) and multivariate analyses (OPLS-DA model, VIP > 1, P < 0.05), followed by selection using the least absolute shrinkage and selection operator (LASSO). The selected overlapping serum metabolites, along with DKD-associated differential gut microbiota, were used to develop a logistic regression prediction model for DKD based on six markers. In the DKD group compared to the DM and NC groups, 39 and 60 differential serum metabolites were identified, respectively (VIP > 1, P < 0.01). Among these, 36 serum metabolites, including alpha-Hydroxyisobutyric acid, were significantly elevated in DKD patients compared to those with DM. Of these, 28 metabolites showed a negative correlation with estimated glomerular filtration rate (eGFR), while 29 showed a positive correlation with urine albumin creatinine ratio (UACR). Patients with DKD were further categorized into subgroups (DKD middle and DKD early) based on eGFR (eGFR < 90 ml/min/1.73m2, eGFR ≥ 90 ml/min/1.73m2), revealing 23 differential metabolites. Dysbiosis of the gut microbiota was evident in DKD patients, with lower relative abundances of g_Prevotella and g_Faecalibacterium compared to the DM and NC groups. Subgroup analysis indicated decreased relative abundances of g_Prevotella and g_Faecalibacterium in the DKD middle group, along with a decrease in g_Klebsiella compared to the DKD early group, which correlated positively with DKD patients' eGFR. There were 11 common metabolites among the three groups of differential metabolites. Among these, three serum metabolites-imidazolepropionic acid, adipoylcarnitine, and 1-methylhistidine-were identified as predictive serum metabolic markers. Disease prediction models (logistic regression models) were constructed based on these three metabolites combined with three genera of bacteria. These models demonstrated strong discriminatory power for diagnosing patients with DKD compared to patients with DM (area under the receiver operating characteristic curve (AUROC) = 0.939 and precision-recall curve (AUPR) = 0.940). The models also effectively discriminated between patients with DKD and NCs (0.976, 0.973). This study revealed distinctive serum metabolites and gut microbiota in patients with DKD. It demonstrated the potential utility of three specific serum metabolites and three genera of bacteria in diagnosing patients with DKD and assessing their renal dysfunction.
Collapse
Affiliation(s)
- Yuyun Hu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Xue Ni
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Qinghuo Chen
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Yihui Qu
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Kanan Chen
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Gaohui Zhu
- Department of Endocrinology, Ningbo Zhenhai Hospital of Traditional Chinese Medicine, Ningbo, Zhejiang, China
| | - Minqiao Zhang
- Department of Nephrology, The First People's Hospital of Xiangshan, Ningbo, 315700, China
| | - Ningjie Xu
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Xu Bai
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Jing Wang
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Yanhong Ma
- Department of Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qun Luo
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Kedan Cai
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China.
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, 315010, China.
| |
Collapse
|
5
|
Guo Y, Pan Y, Feng X, Guo H, Liu L, Zhang K, Xie H, Zhu B, Gong S, Chu Q, Fang H, Chen P. Reshaped local microbiology metabolism by raw tea according to pile fermentation in the dark tea. J Adv Res 2025:S2090-1232(25)00137-7. [PMID: 40064439 DOI: 10.1016/j.jare.2025.02.039] [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: 09/16/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025] Open
Abstract
INTRODUCTION Traditionally, the mechanism of dark tea quality formation has centered on microorganisms, with quality regulated by manipulating microorganisms and their fermentation environment. Nevertheless, raw teas, the natural selective medium of microbial community, was completely ignored in the formation of dark tea unique flavors. OBJECTIVES This study aims to uncover the previously unappreciated interactions between raw tea and microorganisms, demonstrating the significant role of raw tea in the formation of dark tea quality. METHODS Sun-dried raw tea (SDT), baked raw tea (BT), and pan-fried raw tea (PFT) were pile fermented. Chemical profiles, microbial communities, and sensory qualities were assessed by metabolomics, high-throughput sequencing, and sensory evaluation, with correlation and multiple factor analyses used to explore their relationships. RESULTS Compared to PFT and BT, SDT had 18 % lower flavonoid content and 26 % lower catechin content, which favored dominant Agathobacter and Wickerhamomyces. Wickerhamomyces contributed to flower aroma by producing alcohols, esters and terpenes, while Agathobacter amplified acid production. The distinctive dominant bacterium Acidovorax in BT was positively correlated with alcohols and hydrocarbons, with Pearson's r > 0.6, resulting in a 47 % increase in volatile alcohol level, enhancing the fresh and refreshing attributes. A 70-80 % increase in iron concentration in PFT compared to SDT and BT resulted in the predominance of Geobacter, which exhibited a negative correlation with aldehydes. The presence of distinctive bacteria, Streptococcus and Ligilactobacillus, in PFT led to a significant rise in volatile acid content, increasing from 5 % to 25 %. CONCLUSION The chemical profiles of raw tea could reshape local microbiota, which then drives unique qualities of dark tea. This indicates dark tea quality is not passively shaped by the environmental microorganisms, but actively screened by raw tea chemistry. This study paves the way for targeted manipulation of raw tea chemical profiles to achieve desired dark tea flavor characteristics.
Collapse
Affiliation(s)
- Yating Guo
- Tea Research Institute, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Yani Pan
- Tea Research Institute, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Xinyu Feng
- Tea Research Institute, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Haowei Guo
- Tea Research Institute, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Liping Liu
- Huzhou Key Laboratory of Innovation and Application of Agricultural Germplasm Resources, Huzhou Academy of Agricultural Sciences, Huzhou 313001, China
| | - Kexin Zhang
- Huzhou Key Laboratory of Innovation and Application of Agricultural Germplasm Resources, Huzhou Academy of Agricultural Sciences, Huzhou 313001, China
| | - Haojun Xie
- Yuyao Agricultural Mechanization Technology Promotion Center, Yuyao 315400, China
| | - Binkai Zhu
- Zhejiang Wuyi Camelkowloon Brick Tea Co., LTD, Wuyi 321200, China
| | - Shuying Gong
- Tea Research Institute, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Qiang Chu
- Tea Research Institute, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou 310058, China.
| | - Hua Fang
- Institute of Pesticide and Environmental Toxicology, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou 310058, China.
| | - Ping Chen
- Tea Research Institute, College of Agriculture & Biotechnology, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
6
|
Tursi A, Turroni S, De Bastiani R, Procaccianti G, D'Amico F, Allegretta L, Antonino N, Baldi E, Casamassima C, Casella G, Ciuffi M, De Bastiani M, Lazzarotto L, Licci C, Mancuso M, Penna A, Pranzo G, Sanna G, Tosetti C, Zamparella M, Picchio M. Gut microbiota in symptomatic uncomplicated diverticular disease stratifies by severity of abdominal pain. Eur J Gastroenterol Hepatol 2025; 37:147-153. [PMID: 39514266 DOI: 10.1097/meg.0000000000002884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
OBJECTIVE Patients with symptomatic uncomplicated diverticular disease (SUDD) may have a disrupted gut microbiota. However, current data are from small sample studies, and reported associations vary widely across studies. We aimed to profile the fecal microbiota in SUDD patients enrolled in primary care. METHODS A retrospective study was conducted in SUDD ( N = 72) and asymptomatic diverticulosis (AD) ( N = 30), the latter serving as a control group. RESULTS No significant differences in alpha and beta diversity were found between SUDD and AD, but SUDD was discriminated by a higher relative abundance of the family Streptococcaceae and the genera Alistipes , Agathobacter , and Butyricimonas . Interestingly, the gut microbiota of SUDD patients stratified by the severity of abdominal pain [according to the visual analog scale (VAS)]. In particular, higher diversity and health-associated taxa (such as Bifidobacterium , Eubacterium coprostanoligenes group, and Dorea ) characterized mild (VAS score 1-3) SUDD, Proteobacteria , Veillonellaceae and Blautia moderate (VAS score 4-7) SUDD, and Prevotellaceae and Megasphaera severe (VAS score 8-10) SUDD. CONCLUSION Our analysis suggests that specific taxa may be related to SUDD, but the associations vary depending on the severity of abdominal pain. In addition to advancing our ecological understanding of this complex disease, our findings may pave the way for the incorporation of gut microbiota profiling into clinical practice to aid patient management, including stratification and treatment.
Collapse
Affiliation(s)
- Antonio Tursi
- Territorial Gastroenterology Service, Barletta-Andria-Trani Local Health Agency, Andria
- Department of Medical and Surgical Sciences, School of Medicine, Catholic University, Rome
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna
| | - Rudi De Bastiani
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | - Giorgia Procaccianti
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna
| | - Federica D'Amico
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna
| | | | - Natale Antonino
- General Pratictioner, Private Practice Gastroenterologist, Bisceglie
- Barletta-Andria-Trani Local Health Agency, Andria
| | - Elisabetta Baldi
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | - Carlo Casamassima
- Barletta-Andria-Trani Local Health Agency, Andria
- General Pratictioner, Private Practice Gastroenterologist, San Ferdinando di Puglia
| | - Giovanni Casella
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | - Mario Ciuffi
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | - Marco De Bastiani
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | | | | | - Maurizio Mancuso
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | | | - Giuseppe Pranzo
- Ambulatory for IBD Treatment, 'Valle D'Itria' Hospital, Martina Franca
| | - Guido Sanna
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | - Cesare Tosetti
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | - Maria Zamparella
- GIGA-CP Italian Association for Primary Care Gastroenterology, Feltre
| | | |
Collapse
|
7
|
Trezzi S, Scaccabarozzi G, Nossa R, Piazza C, Bianchi AR, Rosi E, Tizzoni F, Mauri M, Camillo L, Baragetti A, Molteni M, Campanella V, Mauro L, Cremonesi P, Severgnini M, Monroy MM, Castiglioni B, Sparvoli F, Pisano S, Pozzi M, Crippa A, Nobile M. Behavioural, cognitive, and neurophysiological effects of a synbiotic supplementation enriched with pigmented corn extract or cornstarch in drug-naïve children with attention-deficit hyperactivity disorder: A randomised, double-blind, comparison-controlled clinical trial. Clin Nutr ESPEN 2025; 65:408-417. [PMID: 39710171 DOI: 10.1016/j.clnesp.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/21/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND & AIMS Considerable interest has been recently given to the potential role of the gut-brain axis (GBA) -a two-way communication network between the gut microbiota and the central nervous system- in the pathogenesis of attention-deficit hyperactivity disorder (ADHD), suggesting the potential usefulness of probiotic and synbiotic supplementations. In light of the limited available evidence, synbiotic efficacy in ADHD children not taking medications should be clarified. This study aimed to investigate the efficacy of a synbiotic dietary supplementation on fatty acids levels as well as on microbiota composition, behaviour, cognition, and brain function in children with ADHD. METHODS A total of 41 drug-naïve school-aged children diagnosed with ADHD were enrolled in a 3-month randomised, double-blind, comparison-controlled clinical trial, receiving either a synbiotic mix (COMP group) or the same synbiotic mix enriched with an additional extract from pigmented corn (EXP group). Changes in levels of some specific short-chain and branched-chain fatty acids were considered as primary outcomes. Secondary outcome measures included gut microbiota profiling, Child Behaviour Checklist, Conners Parent Rating Scale-revised, computerised cognitive tasks, and haemodynamic response to a Go-NoGo task measured by fNIRS. RESULTS No superiority of the EXP synbiotic mix was observed. Analysis of fatty acids did not reveal any significant difference between groups. Children in the COMP group reported a slightly greater improvement than those in the EXP group in focused attention and in the haemodynamic response to a cognitive task. CONCLUSIONS This study shows that pigmented corn extract does not enhance the effects of the synbiotic supplementation in ADHD children in terms of fatty acid production, microbiota composition, clinical, cognitive and neurophysiological measures. However, a synbiotic mix of probiotics plus prebiotic acacia fibre and cornstarch could have some promising effects on ADHD symptoms, which warrants further research. Future studies should also continue to explore the potential of fNIRS for monitoring the effects of interventions that target the GBA. TRIAL REGISTRATION ClinicalTrials.gov: NCT06005506.
Collapse
Affiliation(s)
- Sara Trezzi
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Gaia Scaccabarozzi
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Roberta Nossa
- Bioengineering Lab, Scientific Institute, IRCSS Eugenio Medea, Bosisio Parini, LC, Italy
| | - Caterina Piazza
- Bioengineering Lab, Scientific Institute, IRCSS Eugenio Medea, Bosisio Parini, LC, Italy
| | - Alessandro Rodolfo Bianchi
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Eleonora Rosi
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Federica Tizzoni
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Maddalena Mauri
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Laura Camillo
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy
| | - Massimo Molteni
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | | | - Luca Mauro
- NEMO Clinical Centre, Serena Onlus Foundation, Milan, MI, Italy
| | - Paola Cremonesi
- Institute of Agricultural Biology and Biotechnology, National Research Council, Lodi, LO, Italy
| | - Marco Severgnini
- Institute of Biomedical Technologies, National Research Council, Segrate, MI, Italy
| | - Mariela Mejia Monroy
- Institute of Agricultural Biology and Biotechnology, National Research Council, Pisa, PI, Italy
| | - Bianca Castiglioni
- Institute of Agricultural Biology and Biotechnology, National Research Council, Lodi, LO, Italy
| | - Francesca Sparvoli
- Institute of Agricultural Biology and Biotechnology, National Research Council, Milan, MI, Italy
| | - Simone Pisano
- Department of Neuroscience, AORN Santobono-Pausilipon, Via Mario Fiore 6, Naples, Italy; Department of Translational Medical Sciences, Federico II University, Via Pansini 5, Naples, Italy
| | - Marco Pozzi
- Child and Adolescent Psychiatry Unit, Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, LC, Italy
| | - Alessandro Crippa
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy.
| | - Maria Nobile
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| |
Collapse
|
8
|
McClellan KA, Fowler EC, Perez-Palencia JY, St-Pierre B, Weaver EM, Levesque CL, Koch K, Mueller S, Hong J. Supplemental effects of acidifier and encapsulated butyrate solely and combined in high canola meal diets for nursery pigs. J Anim Sci 2025; 103:skaf111. [PMID: 40233213 PMCID: PMC12065414 DOI: 10.1093/jas/skaf111] [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: 12/02/2024] [Accepted: 03/31/2025] [Indexed: 04/17/2025] Open
Abstract
Incorporating canola meal (CM) into nursery pig diets has been limited due to the adverse effects of glucosinolates, sulfur-containing glycosides, presented in Brassica family oilseeds. This study investigated the effects of acidifier supplementation in high CM diets on the performance, organ weights, blood profiles, and hindgut microbial composition of nursery pigs. A total of 315 nursery pigs, average 6.0 ± 1.2 kg body weight (BW), were assigned to 1 of 5 dietary treatments with 9 replicates (7 pigs/pen) in a randomized complete block design. Treatments included; a corn-soybean meal (SBM) based diet with 20% CM (positive control, PC) and corn-SBM-based diet with 40% CM without (negative control, NC) or with acidifier (NCA), or encapsulated butyrate (NCB), or both (NCAB). Diets met nutrient requirements of NRC (2012) in 3 phases; phase 1 (common diet; 0 to 1 wk), phase 2 (experimental diets; 1 to 3 wk), and phase 3 (experimental diets; 3 to 6 wk). Pig BW and pen feed consumption were measured by phase to investigate the growth performance of pigs. At the end of each phase, 1 pig per pen was bled and then euthanized for measurement of organ weight. Cecal digesta was collected only at the end of phase 3 for microbial composition analysis. Pigs fed the NCA diets had greater (P < 0.05) final BW and average daily gain during phase 3 and had a tendency of greater (P = 0.084) overall gain-to-feed ratio than pigs fed the NC diet. Pigs fed the NCA diet had a greater (χ2 < 0.05) percentage of non-diarrhea feces than pigs fed the PC diet for phase 2 and the overall period. Pigs fed PC diet had higher serum thyroxine (T4) concentration at the end of phase 3 (P < 0.05) than pigs fed NC diets. The addition of acidifier in NC diet decreased (P < 0.05) spleen weight and tended to decrease (P = 0.064) liver weight. Pigs fed the NCAB diet had a lower (P < 0.05) large intestine weight compared with those fed NC diet. Pigs fed the PC or NCA diets tended to have less (P = 0.06) the relative abundance of OTU Ssd-00188 (Agathobacter rectalis 99.23%) and NCAB diet increased (P < 0.05) the relative abundance of OTU Ssd-00001 (Lactobacillus amylovorus 99.64%) in the cecal microbiota compared with those fed NC diet. Overall, these findings support the incorporation of high CM levels (up to 40%) with acidifier supplementation in nursery pig diets, mitigating the negative implications of dietary glucosinolates and providing benefits for both canola growers and pig farmers in regions with substantial canola production.
Collapse
Affiliation(s)
- Katlyn A McClellan
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA
| | - Emily C Fowler
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA
| | | | - Benoit St-Pierre
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA
| | - Eric M Weaver
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA
| | - Crystal L Levesque
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA
| | - Kim Koch
- Northern Crops Institute, Fargo, ND 58105, USA
| | | | - Jinsu Hong
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA
- Department of Animal Science, University of Minnesota, Saint-Paul, MN 55108, USA
| |
Collapse
|
9
|
te Pas MFW, Kluivers-Poodt M, van Riel JW, Schokker D, Rebel JMJ. Restricted vs. ad libitum feeding during sow gestation affects piglet performance, behavior, and fecal microbiota composition. J Anim Sci 2025; 103:skaf118. [PMID: 40244229 PMCID: PMC12124256 DOI: 10.1093/jas/skaf118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/09/2025] [Indexed: 04/18/2025] Open
Abstract
This study investigated how the nutrition of gestating sows affects piglet performance, behavior, and fecal microbiota. Twenty-four sows were divided into 2 feeding groups: those on a restricted diet (once a day) and those fed ad libitum (as much as they wanted), with all receiving the same diet during lactation. The piglets were categorized based on their feeding groups; RG-RL: Piglets born and nursed by restricted-fed sows with restricted feeding, AG-AL: Piglets born and nursed by ad lib-fed sows, RG-AL: Piglets born by restricted-fed sows and nursed by ad lib-fed sows, and AG-RL: Piglets born by ad lib-fed sows and nursed by restricted-fed sows. Performance traits were analyzed using the model including treatment, switching piglets, and room effects. Piglet behavior was evaluated with a generalized linear mixed model (GLMM) using binomial distribution, testing interactions of treatment, switching, gender, and other factors while accounting for random effects related to room, pen, and sow. Results showed that ad lib-fed sows had higher feed intake (P < 0.001), weight gain (P = 0.04), and backfat gain (P = 0.01) compared to restricted-fed sows. They also had lower cortisol levels during gestation (P = 0.02) and lactation (P = 0.04). Restricted-fed sows displayed more air-chewing behavior (P = 0.002), while ad lib-fed sows were more active (P = 0.03) and engaged in eating (P < 0.001). Birth weights and the number of piglets were similar across groups. During lactation, backfat loss varied among piglet groups, with the highest loss in AG-RL (P < 0.01). Piglets from ad lib-fed sows performed better overall, regardless of nursing source. At weaning, piglet weights (P = 0.01) were highest in AG-AL, followed by RG-AL, RG-RL, and AG-RL (interaction effect P = 0.006). Switching piglets initially reduced their playtime but later increased it (P = 0.04). The novel object test indicated that gilts became more active, resembling boars. Additionally, gut microbiota composition varied among sow groups during gestation (P = 0.04) and lactation (P = 0.02), suggesting that maternal diet influences piglet gut health. Overall, these findings highlight the potential role of epigenetic mechanisms in shaping these traits.
Collapse
Affiliation(s)
- Marinus F W te Pas
- Animal Breeding and Genetics, Wageningen Livestock Research, Wageningen, The Netherlands
| | | | - Johan W van Riel
- Animal Breeding and Genetics, Wageningen Livestock Research, Wageningen, The Netherlands
- Department of Animal Health and Welfare, Wageningen Livestock Research, Wageningen, The Netherlands
| | - Dirkjan Schokker
- Epidemiology, Bioinformatic, Animal models, and Vaccine Development, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Johanna M J Rebel
- Epidemiology, Bioinformatic, Animal models, and Vaccine Development, Wageningen Bioveterinary Research, Lelystad, The Netherlands
- Adaptation Physiology Group, Department of Animal Science, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
10
|
Hertz S, Anderson JM, Nielsen HL, Schachtschneider C, McCauley KE, Özçam M, Larsen L, Lynch SV, Nielsen H. Fecal microbiota is associated with extraintestinal manifestations in inflammatory bowel disease. Ann Med 2024; 56:2338244. [PMID: 38648495 PMCID: PMC11036898 DOI: 10.1080/07853890.2024.2338244] [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: 09/29/2023] [Revised: 02/15/2024] [Accepted: 03/17/2024] [Indexed: 04/25/2024] Open
Abstract
INTRODUCTION A large proportion of patients with inflammatory bowel disease (IBD) experience IBD-related inflammatory conditions outside of the gastrointestinal tract, termed extraintestinal manifestations (EIMs) which further decreases quality of life and, in extreme cases, can be life threatening. The pathogenesis of EIMs remains unknown, and although gut microbiota alterations are a well-known characteristic of patients with IBD, its relationship with EIMs remains sparsely investigated. This study aimed to compare the gut microbiota of patients with IBD with and without EIMs. METHODS A total of 131 Danish patients with IBD were included in the study, of whom 86 had a history of EIMs (IBD-EIM) and 45 did not (IBD-C). Stool samples underwent 16S rRNA sequencing. Amplicon sequence variants (ASVs) were mapped to the Silva database. Diversity indices and distance matrices were compared between IBD-EIM and IBD-C. Differentially abundant ASVs were identified using a custom multiple model statistical analysis approach, and modules of co-associated bacteria were identified using sparse correlations for compositional data (SparCC) and related to patient EIM status. RESULTS Patients with IBD and EIMs exhibited increased disease activity, body mass index, increased fecal calprotectin levels and circulating monocytes and neutrophils. Microbiologically, IBD-EIM exhibited lower fecal microbial diversity than IBD-C (Mann-Whitney's test, p = .01) and distinct fecal microbiota composition (permutational multivariate analysis of variance; weighted UniFrac, R2 = 0.018, p = .01). A total of 26 ASVs exhibited differential relative abundances between IBD-EIM and IBD-C, including decreased Agathobacter and Blautia and increased Eggerthella lenta in the IBD-EIM group. SparCC analysis identified 27 bacterial co-association modules, three of which were negatively related to EIM (logistic regression, p < .05) and included important health-associated bacteria, such as Agathobacter and Faecalibacterium. CONCLUSIONS The fecal microbiota in IBD patients with EIMs is distinct from that in IBD patients without EIM and could be important for EIM pathogenesis.
Collapse
Affiliation(s)
- Sandra Hertz
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Jacqueline Moltzau Anderson
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Hans Linde Nielsen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Clinical Microbiology, Aalborg University Hospital, Aalborg, Denmark
| | - Claire Schachtschneider
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Kathryn E. McCauley
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Mustafa Özçam
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Lone Larsen
- Department of Gastroenterology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Center for Molecular Prediction of Inflammatory Bowel Disease, PREDICT, Aalborg University, Aalborg, Denmark
| | - Susan V. Lynch
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Henrik Nielsen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
11
|
Taye B, Mekonnen Z, Belanger KD, Davenport ER. Gut-microbiome profiles among Soil-transmitted helminths (STHs) infected Ethiopian children enrolled in the school-based mass deworming program. PLoS Negl Trop Dis 2024; 18:e0012485. [PMID: 39405336 PMCID: PMC11478818 DOI: 10.1371/journal.pntd.0012485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/26/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Soil-transmitted helminths (STHs) and mutualistic gut microbes coexist in the gastrointestinal tract. However, limited data exist regarding how STH infections are associated with gut microbiome profiles. METHOD We conducted a cross-sectional analysis of baseline data collected in a longitudinal study to identify and explain differences in microbial communities between STH-infected and non-infected Ethiopian school children. We collected 138 stool samples and analyzed them for STH infection using standard direct wet mount and Kato Katz methods. The gut microbiome profiles were characterized using targeted amplicon sequencing of the 16S rRNA gene from the total DNA extracted from the stools. RESULTS Children infected with Trichuris trichiura showed significantly lower microbial diversity than those who were non-infected (p<0.05). We also observed significant difference in microbiome composition based on Trichuris trichiura infection status (PERMANOVA p< 0.01). A comparison of microbial taxa at the genus level among participants infected with different helminth species showed a significant increase in Agathobacter relative abundance among children infected with Trichuris trichiura compared to non-infected subjects (adjusted p = 0.001). CONCLUSIONS Our results indicate that changes in the gut microbiome composition may vary depending on the species of helminth present. Further studies should investigate how Trichuris trichiura selectively alters microbiome composition compared to other STH species.
Collapse
Affiliation(s)
- Bineyam Taye
- Department of Biology, Colgate University, Hamilton, New York, United States of America
| | - Zeleke Mekonnen
- Institute of Health, School of Medical Laboratory Sciences, Jimma University, Jimma, Ethiopia
| | - Kenneth D. Belanger
- Department of Biology, Colgate University, Hamilton, New York, United States of America
| | - Emily R. Davenport
- Department of Biology, Huck Institutes of the Life Sciences, Institute for Computational and Data Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
12
|
Kim YJ, Jung DH, Park CS. Important roles of Ruminococcaceae in the human intestine for resistant starch utilization. Food Sci Biotechnol 2024; 33:2009-2019. [PMID: 39130658 PMCID: PMC11315831 DOI: 10.1007/s10068-024-01621-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/19/2024] [Accepted: 05/27/2024] [Indexed: 08/13/2024] Open
Abstract
Intricate ecosystem of the human gut microbiome is affected by various environmental factors, genetic makeup of the individual, and diet. Specifically, resistant starch (RS) is indigestible in the small intestine but nourishes the gut microbiota in the colon. Degradation of RS in the gut begins with primary degraders, such as Bifidobacterium adolescentis and Ruminococcus bromii. Recently, new RS degraders, such as Ruminococcoides bili, have been reported. These microorganisms play crucial roles in the transformation of RS into short-chain fatty acids (SCFAs), such as acetate, propionate, and butyrate. SCFAs are necessary to maintain optimal intestinal health, regulate inflammation, and protect against various illnesses. This review discusses the effects of RS on gut and highlights its complex interactions with gut flora, especially the Ruminococcaceae family.
Collapse
Affiliation(s)
- Ye-Jin Kim
- Department of Food Science and Biostechnology, Graduate School of Biotechnology and Institute of Life Science and Resources, Kyung Hee University, Yongin, 17104 Republic of Korea
| | - Dong-Hyun Jung
- Division of Food and Nutrition, Chonnam National University, Gwangju, 61186 Republic of Korea
| | - Cheon-Seok Park
- Department of Food Science and Biostechnology, Graduate School of Biotechnology and Institute of Life Science and Resources, Kyung Hee University, Yongin, 17104 Republic of Korea
| |
Collapse
|
13
|
Zhao Y, Li C, Wu K, Chen H, Wang Q, Xiao Y, Yao S, Hong A, Zhang M, Lei S, Yang W, Zhong S, Umar A, Huang J, Yu Z. Exploring the Impact of Short Term Travel on Gut Microbiota and Probiotic Bacteria Mediated Stability. Biomedicines 2024; 12:1378. [PMID: 39061954 PMCID: PMC11274169 DOI: 10.3390/biomedicines12071378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
Although travelers are frequently accompanied by abdominal discomfort and even diarrhea, not every trip can cause this issue. Many studies have reported that intestinal microbes play an important role in it. However, little is known about the reason for the dynamics of these intestinal microbes. Here, we delved into the effects of short-term travel on the gut microbiota of 12 healthy individuals. A total of 72 fecal samples collected before and after one-week travel, alongside non-traveling controls, underwent amplicon sequencing and a series of bioinformatic analyses. We found that travel significantly increased intra-individual gut microbiota fluctuations without diarrhea symptoms. In addition, the initial composition of the gut microbiota before travel emerged as a crucial factor in understanding these fluctuations. Travelers with stable microbiota exhibited an enrichment of specific probiotic bacteria (Agathobaculum, Faecalibacterium, Bifidobacterium, Roseburia, Lactobacillus) before travel. Another batch of data validated their predictive role in distinguishing travelers with and without the gut microbial disorder. This work provided valuable insights into understanding the relationship between gut microbiota and travel. It offered a microbiota-centric perspective and a potential avenue for interventions to preserve gut health during travel.
Collapse
Affiliation(s)
- Yiming Zhao
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Chunyan Li
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Kaijuan Wu
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410013, China; (K.W.); (A.H.); (W.Y.); (S.Z.); (J.H.)
| | - Hao Chen
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410013, China; (K.W.); (A.H.); (W.Y.); (S.Z.); (J.H.)
| | - Qingqun Wang
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Ying Xiao
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Siqi Yao
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Ao Hong
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410013, China; (K.W.); (A.H.); (W.Y.); (S.Z.); (J.H.)
| | - Man Zhang
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Shibo Lei
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Wenyu Yang
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410013, China; (K.W.); (A.H.); (W.Y.); (S.Z.); (J.H.)
| | - Shukun Zhong
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410013, China; (K.W.); (A.H.); (W.Y.); (S.Z.); (J.H.)
| | - Abdulrahim Umar
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| | - Jing Huang
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha 410013, China; (K.W.); (A.H.); (W.Y.); (S.Z.); (J.H.)
| | - Zheng Yu
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha 410013, China; (Y.Z.); (C.L.); (H.C.); (Q.W.); (Y.X.); (S.Y.); (M.Z.); (S.L.); (A.U.)
| |
Collapse
|
14
|
Boston TE, Wang F, Lin X, Kim SW, Fellner V, Scott MF, Ziegler AL, Van Landeghem L, Blikslager AT, Odle J. Prebiotic galactooligosaccharide improves piglet growth performance and intestinal health associated with alterations of the hindgut microbiota during the peri-weaning period. J Anim Sci Biotechnol 2024; 15:88. [PMID: 38867260 PMCID: PMC11170840 DOI: 10.1186/s40104-024-01047-y] [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: 02/23/2024] [Accepted: 05/07/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Weaning stress reduces growth performance and health of young pigs due in part to an abrupt change in diets from highly digestible milk to fibrous plant-based feedstuffs. This study investigated whether dietary galactooligosaccharide (GOS), supplemented both pre- and post-weaning, could improve growth performance and intestinal health via alterations in the hindgut microbial community. METHODS Using a 3 × 2 factorial design, during farrowing 288 piglets from 24 litters received either no creep feed (FC), creep without GOS (FG-) or creep with 5% GOS (FG+) followed by a phase 1 nursery diet without (NG-) or with 3.8% GOS (NG+). Pigs were sampled pre- (D22) and post-weaning (D31) to assess intestinal measures. RESULTS Creep fed pigs grew 19% faster than controls (P < 0.01) prior to weaning, and by the end of the nursery phase (D58), pigs fed GOS pre-farrowing (FG+) were 1.85 kg heavier than controls (P < 0.05). Furthermore, pigs fed GOS in phase 1 of the nursery grew 34% faster (P < 0.04), with greater feed intake and efficiency. Cecal microbial communities clustered distinctly in pre- vs. post-weaned pigs, based on principal coordinate analysis (P < 0.01). No effects of GOS were detected pre-weaning, but gruel creep feeding increased Chao1 α-diversity and altered several genera in the cecal microbiota (P < 0.05). Post-weaning, GOS supplementation increased some genera such as Fusicatenibacter and Collinsella, whereas others decreased such as Campylobacter and Frisingicoccus (P < 0.05). Changes were accompanied by higher molar proportions of butyrate in the cecum of GOS-fed pigs (P < 0.05). CONCLUSIONS Gruel creep feeding effectively improves suckling pig growth regardless of GOS treatment. When supplemented post-weaning, prebiotic GOS improves piglet growth performance associated with changes in hindgut microbial composition.
Collapse
Affiliation(s)
- Timothy E Boston
- Department of Animal Science, College of Ag and Life Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Feng Wang
- Department of Animal Science, College of Ag and Life Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Xi Lin
- Department of Animal Science, College of Ag and Life Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Sung Woo Kim
- Department of Animal Science, College of Ag and Life Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Vivek Fellner
- Department of Animal Science, College of Ag and Life Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Mark F Scott
- Milk Specialties Global, Eden Prairie, MN, 55344, USA
| | - Amanda L Ziegler
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Laurianne Van Landeghem
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27695, USA
| | - Anthony T Blikslager
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Jack Odle
- Department of Animal Science, College of Ag and Life Sciences, North Carolina State University, Raleigh, NC, 27695, USA.
| |
Collapse
|
15
|
Baraille M, Buttet M, Grimm P, Milojevic V, Julliand S, Julliand V. Changes of faecal bacterial communities and microbial fibrolytic activity in horses aged from 6 to 30 years old. PLoS One 2024; 19:e0303029. [PMID: 38829841 PMCID: PMC11146703 DOI: 10.1371/journal.pone.0303029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/17/2024] [Indexed: 06/05/2024] Open
Abstract
Horse owners and veterinarians report that from the age of 15, their horses can lose body condition and be more susceptible to diseases. Large intestinal microbiome changes may be involved. Indeed, microbiota is crucial for maintaining the condition and health of herbivores by converting fibres into nutrients. This study aimed to compare the faecal microbiome in horses aged from 6 to 30 years old (yo), living in the same environment and consuming the same diet, in order to assess whether the parameters changed linearly with age and whether there was a pivotal age category. Fifty horses were selected from the same environment and distributed across four age categories: 6-10 (n = 12), 11-15 (n = 11), 16-20 (n = 13), and 21-30 (n = 14) yo. All horses had no digestive problems, had teeth suitable for consuming their feed, and were up to date with their vaccination and deworming programmes. After three weeks of constant diet (ad libitum hay and 860 g of concentrate per day), one faecal sample per horse was collected on the same day. The bacterial communities' richness and intra-sample diversity were negatively correlated with age. There was a new distribution of non-beneficial and beneficial taxa, particularly in the 21-30 yo category. Although the faecal concentration of short-chain fatty acids remained stable, the acetate proportion was negatively correlated with age while it was the opposite for the proportions of butyrate, valerate, and iso-valerate. Additionally, the faecal pH was negatively correlated with age. Differences were more pronounced when comparing the 6-10 yo and 21-30 yo categories. The values of the parameters studied became more dispersed from the 16-20 yo category onwards, which appeared as a transitional moment, as it did not differ significantly from the younger and older categories for most of these parameters. Our data suggest that the microbiome changes with age. By highlighting the pivotal age of 16-20, this gives the opportunity to intervene before individuals reach extremes that could lead to pathological conditions.
Collapse
Affiliation(s)
- Marylou Baraille
- Institut Agro Dijon, Université de Bourgogne Franche–Comté, PAM UMR A 02.102, Dijon, France
- Lab To Field, Dijon, France
| | | | | | | | | | - Véronique Julliand
- Institut Agro Dijon, Université de Bourgogne Franche–Comté, PAM UMR A 02.102, Dijon, France
| |
Collapse
|
16
|
Rosas-Campos R, Sandoval-Rodríguez AS, Rodríguez-Sanabria JS, Vazquéz-Esqueda ÁO, Alfaro-Martinez CR, Escutia-Gutiérrez R, Vega-Magaña N, Peña-Rodríguez M, Zepeda-Nuño JS, Andrade-Marcial M, Campos-Uscanga Y, Jave-Suárez LF, Santos A, Cerda-Reyes E, Almeida-López M, Martínez-López E, Herrera LA, Armendariz-Borunda J. A Novel Foodstuff Mixture Improves the Gut-Liver Axis in MASLD Mice and the Gut Microbiota in Overweight/Obese Patients. Antioxidants (Basel) 2024; 13:664. [PMID: 38929103 PMCID: PMC11200377 DOI: 10.3390/antiox13060664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Microbial community control is crucial for maintaining homeostasis of the gut-liver axis in metabolic dysfunction-associated steatotic liver disease (MASLD). Here, we show that supplementation with a mixture of Mexican foodstuffs (MexMix)-Opuntia ficus indica (nopal), Theobroma cacao (cocoa) and Acheta domesticus (crickets)-enriches several beneficial taxa in MASLD mice and overweight/obese humans. Thus, MexMix induces an important prebiotic effect. In mice, a restoration of intestinal health was observed due to the increased short-chain fatty acids (SCFAs) and intestinal crypt depth, Ocln and Cldn1 expression, and decreased Il6 and Tnfa expression. MexMix significantly reduced steatosis in the mice's liver and modified the expression of 1668 genes. By PCR, we corroborated a Tnfa and Pparg decrease, and a Cat and Sod increase. In addition, MexMix increased the hepatic NRF2 nuclear translocation and miRNA-34a, miRNA-103, and miRNA-33 decline. In overweight/obese humans, MexMix improved the body image satisfaction and reduced the fat intake. These findings indicate that this new food formulation has potential as a therapeutic approach to treat conditions associated with excessive consumption of fats and sugars.
Collapse
Affiliation(s)
- Rebeca Rosas-Campos
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico; (R.R.-C.); (A.S.S.-R.); (J.S.R.-S.); (Á.O.V.-E.); (C.R.A.-M.); (R.E.-G.)
| | - Ana Soledad Sandoval-Rodríguez
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico; (R.R.-C.); (A.S.S.-R.); (J.S.R.-S.); (Á.O.V.-E.); (C.R.A.-M.); (R.E.-G.)
| | - Jonathan Samael Rodríguez-Sanabria
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico; (R.R.-C.); (A.S.S.-R.); (J.S.R.-S.); (Á.O.V.-E.); (C.R.A.-M.); (R.E.-G.)
- Departamento Académico de Ciencias Básicas, Universidad Autónoma de Guadalajara, Zapopan 45129, Mexico
| | - Ángel Omar Vazquéz-Esqueda
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico; (R.R.-C.); (A.S.S.-R.); (J.S.R.-S.); (Á.O.V.-E.); (C.R.A.-M.); (R.E.-G.)
| | - Carlos Roberto Alfaro-Martinez
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico; (R.R.-C.); (A.S.S.-R.); (J.S.R.-S.); (Á.O.V.-E.); (C.R.A.-M.); (R.E.-G.)
| | - Rebeca Escutia-Gutiérrez
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico; (R.R.-C.); (A.S.S.-R.); (J.S.R.-S.); (Á.O.V.-E.); (C.R.A.-M.); (R.E.-G.)
| | - Natali Vega-Magaña
- Instituto de Investigación en Ciencias Biomédicas (IICB), Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico;
| | - Marcela Peña-Rodríguez
- Laboratorio de Diagnóstico de Enfermedades Emergentes y Reemergentes (LaDEER), Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico;
| | - José Sergio Zepeda-Nuño
- Centro de Investigación y Diagnóstico de Patología, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico;
| | | | | | - Luis Felipe Jave-Suárez
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico;
| | - Arturo Santos
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Zapopan 45138, Mexico; (A.S.); (L.A.H.)
| | | | - Mónica Almeida-López
- Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico;
| | - Erika Martínez-López
- Institute of Translational Nutrigenetics and Nutrigenomics, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico;
| | - Luis Alonso Herrera
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Zapopan 45138, Mexico; (A.S.); (L.A.H.)
- Cancer Research Unit, National Institute of Cancerology-Institute of Biomedical Research, National Autonomous University of Mexico (UNAM), Mexico City 70228, Mexico
| | - Juan Armendariz-Borunda
- Institute for Molecular Biology in Medicine and Gene Therapy, Department of Molecular Biology and Genomics, Health Sciences University Center, University of Guadalajara, Guadalajara 44340, Mexico; (R.R.-C.); (A.S.S.-R.); (J.S.R.-S.); (Á.O.V.-E.); (C.R.A.-M.); (R.E.-G.)
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Zapopan 45138, Mexico; (A.S.); (L.A.H.)
| |
Collapse
|
17
|
Okuma K, Hatayama K, Tokuno H, Ebara A, Odachi A, Masuyama H, Hoshiko N, Tanaka N. A risk estimation method for depression based on the dysbiosis of intestinal microbiota in Japanese patients. Front Psychiatry 2024; 15:1382175. [PMID: 38863614 PMCID: PMC11165696 DOI: 10.3389/fpsyt.2024.1382175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/06/2024] [Indexed: 06/13/2024] Open
Abstract
Introduction Early detection of depression is important for preventing depression-related suicides and reducing the risk of recurrence. This study explored the association between depression and intestinal microbiota and developed a depression risk-estimation method based on this. Methods The intestinal microbiota of Japanese patients with depression (33 males and 35 females) and disease-free controls (246 males and 384 females) in their 20's to 60's were compared by sex using 16S rRNA gene amplicon sequencing. A depression-risk estimation method was developed using structural equation modeling. Results Intestinal bacteria taxa that differed between depression and control groups were identified based on effect size (absolute value greater than 0.2). Neglecta was more abundant, while Coprobacter, Butyricimonas, Clostridium_XlVb, and Romboutsia were less abundant in the male depression group compared to the male control group. In the female depression group, Massilimicrobiota, Merdimonas, and Sellimonas were more abundant, whereas Dorea and Agathobacter were less abundant compared to the female control group. Several of the intestinal bacterial taxa that were less abundant in depression were associated with butyrate or hydrogen production. Using these depression-associated intestinal bacteria as indicators, risk-estimation models using structural equation modeling for depression were developed. In the risk-estimation models for males and females, the areas under the receiver operating characteristic curve were 0.72 and 0.70, respectively, indicating that these models can distinguish between individuals with and without depression. Conclusions This study provides insights into depression etiology and aids in its early detection and treatment.
Collapse
Affiliation(s)
| | | | - Hidetaka Tokuno
- Symbiosis Solutions Inc., Tokyo, Japan
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Aya Ebara
- Symbiosis Solutions Inc., Tokyo, Japan
| | | | | | | | | |
Collapse
|
18
|
Liang L, Su X, Guan Y, Wu B, Zhang X, Nian X. Correlation between intestinal flora and GLP-1 receptor agonist dulaglutide in type 2 diabetes mellitus treatment-A preliminary longitudinal study. iScience 2024; 27:109784. [PMID: 38711446 PMCID: PMC11070333 DOI: 10.1016/j.isci.2024.109784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/30/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
GLP-1 receptor agonists (GLP-1 RA) are presently used as the first-line drugs for the clinical treatment of type 2 diabetes mellitus (T2DM). It can regulate blood glucose by stimulating insulin secretion and lowering glucagon levels. We used 16S rRNA amplicon sequencing to detect structural changes in the composition of the intestinal flora of newly diagnosed T2DM after 1 and 48 weeks of dulaglutide administration. Our research found no significant changes in the intestinal flora after the administration of dulaglutide for 1 week to subjects with newly diagnosed T2DM. Nevertheless, after 48 weeks of dulaglutide administration, the composition of the intestinal flora changed significantly, with a significant reduction in the abundance of intestinal flora. Furthermore, we found that fasting glucose levels, fasting c-peptide levels, HbA1c levels, and BMI are also closely associated with intestinal flora. This reveals that intestinal flora may be one of the mechanisms by which dulaglutide treats T2DM.
Collapse
Affiliation(s)
- Lei Liang
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Department of Endocrinology, Anhui Provincial Hospital, the First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - XiaoYun Su
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yaxin Guan
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bin Wu
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuxiang Zhang
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xin Nian
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
19
|
Zhao S, Lau R, Chen MH. Influence of chain length on the colonic fermentation of xylooligosaccharides. Carbohydr Polym 2024; 331:121869. [PMID: 38388037 DOI: 10.1016/j.carbpol.2024.121869] [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: 11/03/2023] [Revised: 01/12/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024]
Abstract
Xylooligosaccharides (XOS) have been employed as prebiotics containing oligomers of varying sizes or molecular ratios. XOS with a low degree of polymerization (DP) has been demonstrated to have high prebiotic potential. However, there is limited information regarding the specific chain length of XOS required to elicit distinct responses in the gut microbiota. In this study, we aimed to explore whether variations in XOS DP could alter the fate of colonic fermentation. Five XOS fractions (BWXFs) with DP ranges of >40, 20-40, 10-20, 5-10, and 2-4 were prepared by beechwood xylan autohydrolysis and tested on human gut microbiota. Extracellular XOS degradation was observed for molecules with a DP exceeding 5. BWXF treatments altered the microbial community structures, and substrate size-dependent effects on the microbial composition and metabolic outputs were observed. Bacteroidaceae were specifically enriched by xylan. Lachnospiraceae were particularly stimulated by XOS with a DP of 20-40 and 2-4. Bifidobacteriaceae were notably enriched by XOS with a DP of 5-20. High butyrate yields were obtained from cultures containing long-chain BWXFs. Microbiota responses differed with XOS DP composition changes, and microbial competition with XOS with a DP of 2-4 requires further exploration.
Collapse
Affiliation(s)
- Sainan Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Block N1.2, 62 Nanyang Drive, 637459, Singapore.
| | - Raymond Lau
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Block N1.2, 62 Nanyang Drive, 637459, Singapore.
| | - Ming-Hsu Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Block N1.2, 62 Nanyang Drive, 637459, Singapore; Institute of Food Science and Technology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 10617, Taiwan.
| |
Collapse
|
20
|
Conde‐Pérez K, Aja‐Macaya P, Buetas E, Trigo‐Tasende N, Nasser‐Ali M, Rumbo‐Feal S, Nión P, Arribas EM, Estévez LS, Otero‐Alén B, Noguera JF, Concha Á, Pardiñas‐López S, Carda‐Diéguez M, Gómez‐Randulfe I, Martínez‐Lago N, Ladra S, Aparicio LMA, Bou G, Mira Á, Vallejo JA, Poza M. The multispecies microbial cluster of Fusobacterium, Parvimonas, Bacteroides and Faecalibacterium as a precision biomarker for colorectal cancer diagnosis. Mol Oncol 2024; 18:1093-1122. [PMID: 38366793 PMCID: PMC11076999 DOI: 10.1002/1878-0261.13604] [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: 08/24/2023] [Revised: 12/27/2023] [Accepted: 01/26/2024] [Indexed: 02/18/2024] Open
Abstract
The incidence of colorectal cancer (CRC) has increased worldwide, and early diagnosis is crucial to reduce mortality rates. Therefore, new noninvasive biomarkers for CRC are required. Recent studies have revealed an imbalance in the oral and gut microbiomes of patients with CRC, as well as impaired gut vascular barrier function. In the present study, the microbiomes of saliva, crevicular fluid, feces, and non-neoplastic and tumor intestinal tissue samples of 93 CRC patients and 30 healthy individuals without digestive disorders (non-CRC) were analyzed by 16S rRNA metabarcoding procedures. The data revealed that Parvimonas, Fusobacterium, and Bacteroides fragilis were significantly over-represented in stool samples of CRC patients, whereas Faecalibacterium and Blautia were significantly over-abundant in the non-CRC group. Moreover, the tumor samples were enriched in well-known periodontal anaerobes, including Fusobacterium, Parvimonas, Peptostreptococcus, Porphyromonas, and Prevotella. Co-occurrence patterns of these oral microorganisms were observed in the subgingival pocket and in the tumor tissues of CRC patients, where they also correlated with other gut microbes, such as Hungatella. This study provides new evidence that oral pathobionts, normally located in subgingival pockets, can migrate to the colon and probably aggregate with aerobic bacteria, forming synergistic consortia. Furthermore, we suggest that the group composed of Fusobacterium, Parvimonas, Bacteroides, and Faecalibacterium could be used to design an excellent noninvasive fecal test for the early diagnosis of CRC. The combination of these four genera would significantly improve the reliability of a discriminatory test with respect to others that use a single species as a unique CRC biomarker.
Collapse
Affiliation(s)
- Kelly Conde‐Pérez
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Pablo Aja‐Macaya
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Elena Buetas
- Genomic and Health Department, FISABIO FoundationCenter for Advanced Research in Public HealthValenciaSpain
| | - Noelia Trigo‐Tasende
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Mohammed Nasser‐Ali
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Soraya Rumbo‐Feal
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Paula Nión
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Elsa Martín‐De Arribas
- Database Laboratory, Research Center for Information and Communication Technologies (CITIC)University of A Coruña (UDC)A CoruñaSpain
| | - Lara S. Estévez
- Pathology Service and BiobankUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Begoña Otero‐Alén
- Pathology Service and BiobankUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - José F. Noguera
- Surgery ServiceUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Ángel Concha
- Pathology Service and BiobankUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Simón Pardiñas‐López
- Periodontology and Oral Surgery, Pardiñas Medical Dental Clinic – Cell Therapy and Regenerative Medicine GroupInstitute of Biomedical Research (INIBIC)A CoruñaSpain
| | - Miguel Carda‐Diéguez
- Genomic and Health Department, FISABIO FoundationCenter for Advanced Research in Public HealthValenciaSpain
| | - Igor Gómez‐Randulfe
- Medical Oncology DepartmentUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | | | - Susana Ladra
- Database Laboratory, Research Center for Information and Communication Technologies (CITIC)University of A Coruña (UDC)A CoruñaSpain
| | - Luis M. A. Aparicio
- Medical Oncology DepartmentUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Germán Bou
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Álex Mira
- Genomic and Health Department, FISABIO FoundationCenter for Advanced Research in Public HealthValenciaSpain
| | - Juan A. Vallejo
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
| | - Margarita Poza
- Microbiome and Health Group (meiGAbiome), Microbiology Research Group, Institute of Biomedical Research (INIBIC) – Interdisciplinary Center for Chemistry and Biology (CICA) – University of A Coruña (UDC) – CIBER de Enfermedades Infecciosas (CIBERINFEC‐ISCIII), Servicio de MicrobiologíaUniversity Hospital of A Coruña (CHUAC)A CoruñaSpain
- Microbiome and Health Group, Faculty of SciencesUniversity of A Coruña (UDC)A CoruñaSpain
| |
Collapse
|
21
|
Xiao Y, Wang Y, Tong B, Gu Y, Zhou X, Zhu N, Xu X, Yin X, Kou Y, Tan Y, Wang J, Li W. Eubacterium rectale is a potential marker of altered gut microbiota in psoriasis and psoriatic arthritis. Microbiol Spectr 2024; 12:e0115423. [PMID: 38441468 PMCID: PMC10986482 DOI: 10.1128/spectrum.01154-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 01/27/2024] [Indexed: 04/06/2024] Open
Abstract
Previous studies have profiled the gut microbiota among psoriatic patients compared to that among healthy individuals. However, a comprehensive understanding of the magnitude, direction, and detailed compositional and functional profiles remains limited. Additionally, research exploring the gut microbiota in the context of both plaque psoriasis (PsO) and psoriatic arthritis (PsA) is lacking. To assess the taxonomic and functional characteristics of the gut microbiota in PsO and PsA patients and investigate potential links between the gut microbiota and disease pathogenesis. We collected fecal samples from 70 psoriatic patients (44 PsO and 26 PsA) and 25 age- and gender-matched healthy controls (HC) and employed deep metagenomic sequencing to characterize their gut microbiota. We noted significant alternations in the gut microbiota compositions of both PsO and PsA patients compared to those of HC. Despite limited effect sizes in alpha diversity (12.3% reduction of microbial richness but unchanged evenness in psoriatic patients) and beta diversity (disease accounts for 3.5% of total variations), we consistently observed substantial reductions of Eubacterium rectale in both PsO and PsA patients, with PsA patients exhibiting even lower levels of E. rectale than PsO patients. Additionally, two Alistipes species were also depleted in psoriatic patients. These microorganisms are known to play crucial roles in carbohydrate metabolism pathways, mainly producing short-chain fatty acids with anti-inflammatory effects. Overall, our observations supplemented the profiling of altered gut microbiota in patients with PsO and PsA at the species level and described a link between the dominant short-chain fatty acid-producing bacterial species and systemic immunity in psoriatic patients. IMPORTANCE In this observational clinical study with sufficient sample size and metagenomic sequencing to profile the gut microbiota, we identified consistent signals of the depleted abundance of Eubacterium rectale and related functional genes among psoriatic patients, including those with psoriatic arthritis. E. rectale may serve as an ecologically important functional unit in the gut microbiota, holding potential as a diagnostic marker and target for therapeutic interventions to achieve lasting effects. Our findings provide comprehensive gut microbiota profiling in psoriasis, resolving previous contradictions and generating new hypotheses for further investigation. These insights may significantly impact psoriasis management and related conditions.
Collapse
Affiliation(s)
- Yue Xiao
- Department of Dermatology and Venereology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yiyi Wang
- Department of Dermatology and Venereology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | - Yuanxia Gu
- Department of Dermatology and Venereology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xingli Zhou
- Department of Dermatology and Venereology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | | | | | | | | | | | - Wei Li
- Department of Dermatology and Venereology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
22
|
Bläckberg A, Holm K, Liderot K, Nilson B, Sunnerhagen T. Eubacterium bacteremia - a retrospective observational study of a seldom found anaerobic pathogen. Diagn Microbiol Infect Dis 2024; 108:116185. [PMID: 38232640 DOI: 10.1016/j.diagmicrobio.2024.116185] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/28/2023] [Accepted: 01/11/2024] [Indexed: 01/19/2024]
Abstract
BACKGROUND Human infections due to Eubacterium are rare and knowledge of the condition is limited. This study aimed to describe clinical characteristics and outcome in patients with Eubacterium bacteremia. METHODS Episodes of Eubacterium bacteremia were identified through the clinical microbiology laboratory in Lund, Sweden. Medical records were retrospectively reviewed. Blood isolates of Eubacterium were collected and antibiotic susceptibility testing was performed with agar dilution. RESULTS Seventeen patients with Eubacterium bacteremia were identified of whom six had monomicrobial bacteremia. The incidence was 1.7 cases of Eubacterium bacteremia per million inhabitants and year. The median age was 67 years (interquartile range 63-79 years), and six patients had some form of malignancy. Most of the patients an abdominal focus of infection and the 30-day mortality was low (n = 1). CONCLUSIONS Invasive infections with Eubacterium have a low incidence. The condition has a low mortality and an abdominal focus of infection, and malignancy, is common.
Collapse
Affiliation(s)
- Anna Bläckberg
- Department of Infectious Diseases, Skåne University Hospital Lund, Sweden; Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University Sweden
| | - Karin Holm
- Department of Infectious Diseases, Skåne University Hospital Lund, Sweden; Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University Sweden
| | - Karin Liderot
- Division of Clinical Microbiology, Karolinska Institutet and Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Bo Nilson
- Clinical Microbiology, Infection Prevention and Control, Office for Medical Services, Lund, Sweden; Division of Medical Microbiology, Department of Laboratory Medicine Lund, Lund University, Lund, Sweden
| | - Torgny Sunnerhagen
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University Sweden; Clinical Microbiology, Infection Prevention and Control, Office for Medical Services, Lund, Sweden.
| |
Collapse
|
23
|
Sato Y, Kanayama M, Nakajima S, Hishida Y, Watanabe Y. Sialyllactose Enhances the Short-Chain Fatty Acid Production and Barrier Function of Gut Epithelial Cells via Nonbifidogenic Modification of the Fecal Microbiome in Human Adults. Microorganisms 2024; 12:252. [PMID: 38399656 PMCID: PMC10892346 DOI: 10.3390/microorganisms12020252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/12/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Although various benefits of human milk oligosaccharides (HMOs) have been reported, such as promoting Bifidobacterium growth in the infant gut, their effects on adults have not been fully studied. This study investigated the effects of two types of sialyllactose, 3'-sialyllactose (3'-SL) and 6'-sialyllactose (6'-SL), on the adult intestinal microbiome using the simulator of human intestinal microbial ecosystem (SHIME®), which can simulate human gastrointestinal conditions. HPLC metabolite analysis showed that sialyllactose (SL) supplementation increased the short-chain fatty acid content of SHIME culture broth. Moreover, 16S rRNA gene sequencing analysis revealed that SL promoted the growth of Phascolarctobacterium and Lachnospiraceae, short-chain fatty acid-producing bacteria, but not the growth of Bifidobacterium. Altogether, both types of SL stimulated an increase in short-chain fatty acids, including propionate and butyrate. Additionally, SHIME culture supernatant supplemented with SL improved the intestinal barrier function in Caco-2 cell monolayers. These results suggest that SL could act as a unique prebiotic among other HMOs with a nonbifidogenic effect, resulting in intestinal barrier protection.
Collapse
Affiliation(s)
- Yohei Sato
- Institute of Health Science, Kirin Holdings Co., Ltd., 2-26-1 Muraoka-Higashi, Fujisawa 251-8555, Japan; (Y.S.); (M.K.); (Y.H.)
| | - Masaya Kanayama
- Institute of Health Science, Kirin Holdings Co., Ltd., 2-26-1 Muraoka-Higashi, Fujisawa 251-8555, Japan; (Y.S.); (M.K.); (Y.H.)
| | - Shiori Nakajima
- Health Science Business Department, Kirin Holdings Co., Ltd., 4-10-2 Nakano, Tokyo 164-0001, Japan;
| | - Yukihiro Hishida
- Institute of Health Science, Kirin Holdings Co., Ltd., 2-26-1 Muraoka-Higashi, Fujisawa 251-8555, Japan; (Y.S.); (M.K.); (Y.H.)
| | - Yuta Watanabe
- Institute of Health Science, Kirin Holdings Co., Ltd., 2-26-1 Muraoka-Higashi, Fujisawa 251-8555, Japan; (Y.S.); (M.K.); (Y.H.)
| |
Collapse
|
24
|
Román-Ochoa Y, M Cantu-Jungles T, Choque Delgado GT, Bulut N, Tejada TR, Yucra HR, Duran AE, Hamaker BR. Specific dietary fibers prevent heavy metal disruption of the human gut microbiota in vitro. Food Res Int 2024; 176:113858. [PMID: 38163737 DOI: 10.1016/j.foodres.2023.113858] [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: 11/02/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024]
Abstract
Heavy metal exposure is a growing concern due to its adverse effects on human health, including the disruption of gut microbiota composition and function. Dietary fibers have been shown to positively impact the gut microbiota and could mitigate some of the heavy metal negative effects. This study aimed to investigate the effects of different heavy metals (As, Cd and Hg in different concentrations) on gut microbiota in the presence and absence of different dietary fibers that included fructooligosaccharides, pectin, resistant starch, and wheat bran. We observed that whereas heavy metals impaired fiber fermentation outcomes for some fiber types, the presence of fibers generally protected gut microbial communities from heavy metal-induced changes, especially for As and Cd. Notably, the protective effects varied depending on fiber types, and heavy metal type and concentration and were overall stronger for wheat bran and pectin than other fiber types. Our findings suggest that dietary fibers play a role in mitigating the adverse effects of heavy metal exposure on gut microbiota health and may have implications for the development of dietary interventions to reduce dysbiosis associated with heavy metal exposure. Moreover, fiber-type specific outcomes highlight the importance of evidence-based selection of prebiotic dietary fibers to mitigate heavy metal toxicity to the gut microbiota.
Collapse
Affiliation(s)
- Yony Román-Ochoa
- Whistler Center for Carbohydrate Research and Department of Food Science, Purdue University, West Lafayette, IN, USA; Carbohydrate Biochemistry Research Group (BIOCAB), Department of Chemistry, Universidad de Los Andes, Carrera 1 No. 18A-10, Bogotá 111711, Colombia
| | - Thaisa M Cantu-Jungles
- Whistler Center for Carbohydrate Research and Department of Food Science, Purdue University, West Lafayette, IN, USA.
| | | | - Nuseybe Bulut
- Whistler Center for Carbohydrate Research and Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - Teresa R Tejada
- Academic Department of Food Industries Engineering, National University of San Agustin, Arequipa, Peru
| | - Harry R Yucra
- Academic Department of Food Industries Engineering, National University of San Agustin, Arequipa, Peru
| | - Antonio E Duran
- Academic Department of Food Industries Engineering, National University of San Agustin, Arequipa, Peru
| | - Bruce R Hamaker
- Whistler Center for Carbohydrate Research and Department of Food Science, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
25
|
Jing Y, Yang D, Bai F, Wang Q, Zhang C, Yan Y, Li Z, Li Y, Chen Z, Li J, Yu Y. Spinal cord injury-induced gut dysbiosis influences neurological recovery partly through short-chain fatty acids. NPJ Biofilms Microbiomes 2023; 9:99. [PMID: 38092763 PMCID: PMC10719379 DOI: 10.1038/s41522-023-00466-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023] Open
Abstract
Spinal cord injury (SCI) can reshape gut microbial composition, significantly affecting clinical outcomes in SCI patients. However, mechanisms regarding gut-brain interactions and their clinical implications have not been elucidated. We hypothesized that short-chain fatty acids (SCFAs), intestinal microbial bioactive metabolites, may significantly affect the gut-brain axis and enhance functional recovery in a mouse model of SCI. We enrolled 59 SCI patients and 27 healthy control subjects and collected samples. Thereafter, gut microbiota and SCFAs were analyzed using 16 S rDNA sequencing and gas chromatography-mass spectrometry, respectively. We observed an increase in Actinobacteriota abundance and a decrease in Firmicutes abundance. Particularly, the SCFA-producing genera, such as Faecalibacterium, Megamonas, and Agathobacter were significantly downregulated among SCI patients compared to healthy controls. Moreover, SCI induced downregulation of acetic acid (AA), propionic acid (PA), and butyric acid (BA) in the SCI group. Fecal SCFA contents were altered in SCI patients with different injury course and injury segments. Main SCFAs (AA, BA, and PA) were administered in combination to treat SCI mice. SCFA supplementation significantly improved locomotor recovery in SCI mice, enhanced neuronal survival, promoted axonal formation, reduced astrogliosis, and suppressed microglial activation. Furthermore, SCFA supplementation downregulated NF-κB signaling while upregulating neurotrophin-3 expression following SCI. Microbial sequencing and metabolomics analysis showed that SCI patients exhibited a lower level of certain SCFAs and related bacterial strains than healthy controls. SCFA supplementation can reduce inflammation and enhance nourishing elements, facilitating the restoration of neurological tissues and the improvement of functional recuperation. Trial registration: This study was registered in the China Clinical Trial Registry ( www.chictr.org.cn ) on February 13, 2017 (ChiCTR-RPC-17010621).
Collapse
Affiliation(s)
- Yingli Jing
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
| | - Degang Yang
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Department of Spinal and Neural Function Reconstruction, Beijing Bo'ai Hospital, Beijing, 100068, China
| | - Fan Bai
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
| | - Qiuying Wang
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
| | - Chao Zhang
- Department of Neurosurgery, Linyi People's Hospital, Shangdong, 276034, China
| | - Yitong Yan
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
| | - Zihan Li
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
| | - Yan Li
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
| | - Zhiguo Chen
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China.
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China.
| | - Jianjun Li
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China.
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China.
| | - Yan Yu
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, and School of Rehabilitation Medicine, Capital Medical University, Beijing, 100068, China.
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China.
| |
Collapse
|
26
|
Bai L, Paek J, Shin Y, Kim H, Kim SH, Shin JH, Kook JK, Chang YH. Description of Fusibacillus kribbianus gen. nov., sp. nov., a fusiform anaerobe isolated from pig feces. Anaerobe 2023; 84:102792. [PMID: 37925064 DOI: 10.1016/j.anaerobe.2023.102792] [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: 08/30/2023] [Revised: 09/12/2023] [Accepted: 10/25/2023] [Indexed: 11/06/2023]
Abstract
OBJECTIVE The family Lachnospiraceae is affiliated with the order Clostridiales and was originally contained within Clostridial cluster XIVa. The members of Lachnospiraceae inhabiting the gut comprise the chemoorganotrophic genera, generating sundry short-chain fatty acids to supply energy to the host, and are considered to be related to obesity and gut health. METHODS The polyphasic taxonomic approach was used to characterize the isolate YH-rum2234T. A detailed metabolic analysis was conducted to compare the novel isolate with related strains within the family Lachnospiraceae. RESULTS A fusiform, obligately anaerobic, Gram-stain-negative bacterium, YH-rum2234T, was isolated from pig feces. Analysis of the 16S rRNA gene sequence revealed that the similarities between the isolate and the familiarly interrelated strain Lientehia hominis KCTC 25345T was 94.3%. The average nucleotide identities and genome-to-genome distances of YH-rum2234T and its closely related strains were below 85.5% and 32.5%, respectively. The G + C content of the genomic DNA was 49.2 mol%. The main fatty acids were C16:0, C14:0, and C14:0 DMA. The major polar lipids were aminophospholipids. The cell wall did not contain the peptidoglycan meso-diaminopimelic acid. CONCLUSION Given the chemotaxonomic, phenotypic, and phylogenetic properties, YH-rum2234T (=KCTC 25710T = DSMZ 116041T) represents a new genus and species in the family Lachnospiraceae. Fusibacillus kribbianus gen. nov., sp. nov. is the proposed name.
Collapse
Affiliation(s)
- Lu Bai
- ABS Research Support Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jayoung Paek
- ABS Research Support Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Yeseul Shin
- ABS Research Support Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hongik Kim
- Vitabio, Inc., Daejeon, 305-500, Republic of Korea
| | - Si Hyun Kim
- Department of Clinical Laboratory Science, Semyung University, Jecheon, Republic of Korea
| | - Jeong Hwan Shin
- Department of Laboratory Medicine, Inje University College of Medicine, Busan, 614-735, Republic of Korea
| | - Joong-Ki Kook
- Korean Collection for Oral Microbiology and Department of Oral Biochemistry, School of Dentistry, Chosun University, Gwangju, 501-759, Republic of Korea.
| | - Young-Hyo Chang
- ABS Research Support Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
27
|
Nguyen TQ, Martínez-Álvaro M, Lima J, Auffret MD, Rutherford KMD, Simm G, Dewhurst RJ, Baima ET, Roehe R. Identification of intestinal and fecal microbial biomarkers using a porcine social stress model. Front Microbiol 2023; 14:1197371. [PMID: 38029169 PMCID: PMC10670831 DOI: 10.3389/fmicb.2023.1197371] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Understanding the relationships between social stress and the gastrointestinal microbiota, and how they influence host health and performance is expected to have many scientific and commercial implementations in different species, including identification and improvement of challenges to animal welfare and health. In particular, the study of the stress impact on the gastrointestinal microbiota of pigs may be of interest as a model for human health. A porcine stress model based on repeated regrouping and reduced space allowance during the last 4 weeks of the finishing period was developed to identify stress-induced changes in the gut microbiome composition. The application of the porcine stress model resulted in a significant increase in salivary cortisol concentration over the course of the trial and decreased growth performance and appetite. The applied social stress resulted in 32 bacteria being either enriched (13) or depleted (19) in the intestine and feces. Fecal samples showed a greater number of microbial genera influenced by stress than caecum or colon samples. Our trial revealed that the opportunistic pathogens Treponema and Clostridium were enriched in colonic and fecal samples from stressed pigs. Additionally, genera such as Streptococcus, Parabacteroides, Desulfovibrio, Terrisporobacter, Marvinbryantia, and Romboutsia were found to be enriched in response to social stress. In contrast, the genera Prevotella, Faecalibacterium, Butyricicoccus, Dialister, Alloprevotella, Megasphaera, and Mitsuokella were depleted. These depleted bacteria are of great interest because they synthesize metabolites [e.g., short-chain fatty acids (SCFA), in particular, butyrate] showing beneficial health benefits due to inhibitory effects on pathogenic bacteria in different animal species. Of particular interest are Dialister and Faecalibacterium, as their depletion was identified in a human study to be associated with inferior quality of life and depression. We also revealed that some pigs were more susceptible to pathogens as indicated by large enrichments of opportunistic pathogens of Clostridium, Treponema, Streptococcus and Campylobacter. Generally, our results provide further evidence for the microbiota-gut-brain axis as indicated by an increase in cortisol concentration due to social stress regulated by the hypothalamic-pituitary-adrenal axis, and a change in microbiota composition, particularly of bacteria known to be associated with pathogenicity and mental health diseases.
Collapse
Affiliation(s)
- Tuan Q. Nguyen
- Scotland’s Rural College, Edinburgh, United Kingdom
- Department of Animal Breeding, Faculty of Animal Science and Veterinary Medicine, Nong Lam University – Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | | | - Joana Lima
- Scotland’s Rural College, Edinburgh, United Kingdom
| | | | | | - Geoff Simm
- Global Academy of Agriculture and Food Security, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Eric T. Baima
- Zoetis Inc., Parsippany-Troy Hills, NJ, United States
| | - Rainer Roehe
- Scotland’s Rural College, Edinburgh, United Kingdom
| |
Collapse
|
28
|
Martinez E, Crevecoeur S, Thirion C, Grandjean J, Fall PA, Hayette MP, Michel M, Taminiau B, Louis E, Daube G. Gut Microbiota Associated with Clostridioides difficile Carriage in Three Clinical Groups (Inflammatory Bowel Disease, C. difficile Infection and Healthcare Workers) in Hospital Field. Microorganisms 2023; 11:2527. [PMID: 37894185 PMCID: PMC10609531 DOI: 10.3390/microorganisms11102527] [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: 09/05/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Clostridioides difficile is an anaerobic spore-forming Gram-positive bacterium. C. difficile carriage and 16S rDNA profiling were studied in three clinical groups at three different sampling times: inflammatory bowel disease (IBD) patients, C. difficile infection (CDI) patients and healthcare workers (HCWs). Diversity analysis was realized in the three clinical groups, the positive and negative C. difficile carriage groups and the three analysis periods. Concerning the three clinical groups, β-diversity tests showed significant differences between them, especially between the HCW group and IBD group and between IBD patients and CDI patients. The Simpson index (evenness) showed a significant difference between two clinical groups (HCWs and IBD). Several genera were significantly different in the IBD patient group (Sutterella, Agathobacter) and in the CDI patient group (Enterococcus, Clostridioides). Concerning the positive and negative C. difficile carriage groups, β-diversity tests showed significant differences. Shannon, Simpson and InvSimpson indexes showed significant differences between the two groups. Several genera had significantly different relative prevalences in the negative group (Agathobacter, Sutterella, Anaerostipes, Oscillospira) and the positive group (Enterococcus, Enterobacteriaceae_ge and Enterobacterales_ge). A microbiota footprint was detected in C. difficile-positive carriers. More experiments are needed to test this microbiota footprint to see its impact on C. difficile infection.
Collapse
Affiliation(s)
- Elisa Martinez
- Food Microbiology Lab, Fundamental and Applied Research for Animals and Health (FARAH), Department of Food Sciences, Faculty of Veterinary Medicine, University of Liege, 4000 Liège, Belgium
| | - Sebastien Crevecoeur
- Food Microbiology Lab, Fundamental and Applied Research for Animals and Health (FARAH), Department of Food Sciences, Faculty of Veterinary Medicine, University of Liege, 4000 Liège, Belgium
| | - Carine Thirion
- Department of Clinical Sciences, Immunopathology—Infectious Diseases and General Internal Medicine, University Hospital CHU of Liege, 4000 Liège, Belgium
| | - Jessica Grandjean
- Department of Gastroenterology, University Hospital CHU of Liege, 4000 Liège, Belgium
| | | | - Marie-Pierre Hayette
- Department of Biomedical and Preclinical Sciences, Faculty of Medicine, University of Liege, 4000 Liège, Belgium
| | - Moutschen Michel
- Department of Clinical Sciences, Immunopathology—Infectious Diseases and General Internal Medicine, University Hospital CHU of Liege, 4000 Liège, Belgium
| | - Bernard Taminiau
- Food Microbiology Lab, Fundamental and Applied Research for Animals and Health (FARAH), Department of Food Sciences, Faculty of Veterinary Medicine, University of Liege, 4000 Liège, Belgium
| | - Edouard Louis
- Department of Gastroenterology, University Hospital CHU of Liege, 4000 Liège, Belgium
| | - Georges Daube
- Food Microbiology Lab, Fundamental and Applied Research for Animals and Health (FARAH), Department of Food Sciences, Faculty of Veterinary Medicine, University of Liege, 4000 Liège, Belgium
| |
Collapse
|
29
|
Hackmann TJ, Zhang B. The phenotype and genotype of fermentative prokaryotes. SCIENCE ADVANCES 2023; 9:eadg8687. [PMID: 37756392 PMCID: PMC10530074 DOI: 10.1126/sciadv.adg8687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
Fermentation is a type of metabolism pervasive in oxygen-deprived environments. Despite its importance, we know little about the range and traits of organisms that carry out this metabolism. Our study addresses this gap with a comprehensive analysis of the phenotype and genotype of fermentative prokaryotes. We assembled a dataset with phenotypic records of 8350 organisms plus 4355 genomes and 13.6 million genes. Our analysis reveals fermentation is both widespread (in ~30% of prokaryotes) and complex (forming ~300 combinations of metabolites). Furthermore, it points to previously uncharacterized proteins involved in this metabolism. Previous studies suggest that metabolic pathways for fermentation are well understood, but metabolic models built in our study show gaps in our knowledge. This study demonstrates the complexity of fermentation while showing that there is still much to learn about this metabolism. All resources in our study can be explored by the scientific community with an online, interactive tool.
Collapse
Affiliation(s)
| | - Bo Zhang
- Department of Chemical Engineering, University of California, Santa Barbara, CA, USA
| |
Collapse
|
30
|
Hatayama K, Ebara A, Okuma K, Tokuno H, Hasuko K, Masuyama H, Ashikari I, Shirasawa T. Characteristics of Intestinal Microbiota in Japanese Patients with Mild Cognitive Impairment and a Risk-Estimating Method for the Disorder. Biomedicines 2023; 11:1789. [PMID: 37509429 PMCID: PMC10376419 DOI: 10.3390/biomedicines11071789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Intestinal microbiota may play a significant role in the development and progression of mild cognitive impairment (MCI). In addition, sex differences in the prevalence of MCI and intestinal microbiota are likely to exist. Therefore, this study investigated the association between MCI and intestinal microbiota by comparing Japanese patients in their 70s with MCI (11 males and 18 females) and disease-free controls (17 males and 23 females), taking sex into account. In both sexes, Clostridium_XVIII, Eggerthella, Erysipelatoclostridium, Flavonifractor, and Ruminococcus 2 were the more abundant taxa in the MCI group, whereas Megasphaera, Oscillibacter, Prevotella, Roseburia, and Victivallis were less abundant. Based on these characteristics, it was hypothesized that the composition of the intestinal microbiota in the MCI group leads to dysregulation of the intestinal microbiota, increased intestinal and blood-brain barrier permeability, and increased chronic neuroinflammation, with the long-term persistence of these abnormalities ultimately leading to cognitive decline. Furthermore, risk estimation models for MCI based on intestinal microbiota data were developed using structural equation modeling. These tests discriminated between the MCI and control groups. Incorporating these factors into intestinal microbiota testing using stool samples may be an efficient method to screen individuals with MCI.
Collapse
Affiliation(s)
| | - Aya Ebara
- Symbiosis Solutions Inc., Tokyo 101-0064, Japan
| | - Kana Okuma
- Symbiosis Solutions Inc., Tokyo 101-0064, Japan
| | | | | | | | | | | |
Collapse
|
31
|
Walters KA, Mohan G, Myers KS, Ingle AT, Donohue TJ, Noguera DR. A metagenome-level analysis of a microbial community fermenting ultra-filtered milk permeate. Front Bioeng Biotechnol 2023; 11:1173656. [PMID: 37324413 PMCID: PMC10263058 DOI: 10.3389/fbioe.2023.1173656] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/03/2023] [Indexed: 06/17/2023] Open
Abstract
Fermentative microbial communities have the potential to serve as biocatalysts for the conversion of low-value dairy coproducts into renewable chemicals, contributing to a more sustainable global economy. To develop predictive tools for the design and operation of industrially relevant strategies that utilize fermentative microbial communities, there is a need to determine the genomic features of community members that are characteristic to the accumulation of different products. To address this knowledge gap, we performed a 282-day bioreactor experiment with a microbial community that was fed ultra-filtered milk permeate, a low-value coproduct from the dairy industry. The bioreactor was inoculated with a microbial community from an acid-phase digester. A metagenomic analysis was used to assess microbial community dynamics, construct metagenome-assembled genomes (MAGs), and evaluate the potential for lactose utilization and fermentation product synthesis of community members represented by the assembled MAGs. This analysis led us to propose that, in this reactor, members of the Actinobacteriota phylum are important in the degradation of lactose, via the Leloir pathway and the bifid shunt, and the production of acetic, lactic, and succinic acids. In addition, members of the Firmicutes phylum contribute to the chain-elongation-mediated production of butyric, hexanoic, and octanoic acids, with different microbes using either lactose, ethanol, or lactic acid as the growth substrate. We conclude that genes encoding carbohydrate utilization pathways, and genes encoding lactic acid transport into the cell, electron confurcating lactate dehydrogenase, and its associated electron transfer flavoproteins, are genomic features whose presence in Firmicutes needs to be established to infer the growth substrate used for chain elongation.
Collapse
Affiliation(s)
- Kevin A. Walters
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, WI, United States
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Geethaanjali Mohan
- Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, WI, United States
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Kevin S. Myers
- Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, WI, United States
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Abel T. Ingle
- Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, WI, United States
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI, United States
- Department of Civil and Environmental Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Timothy J. Donohue
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, WI, United States
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Daniel R. Noguera
- Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, WI, United States
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI, United States
- Department of Civil and Environmental Engineering, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
32
|
Baltazar-Díaz TA, Amador-Lara F, Andrade-Villanueva JF, González-Hernández LA, Cabrera-Silva RI, Sánchez-Reyes K, Álvarez-Zavala M, Valenzuela-Ramírez A, Del Toro-Arreola S, Bueno-Topete MR. Gut Bacterial Communities in HIV-Infected Individuals with Metabolic Syndrome: Effects of the Therapy with Integrase Strand Transfer Inhibitor-Based and Protease Inhibitor-Based Regimens. Microorganisms 2023; 11:microorganisms11040951. [PMID: 37110374 PMCID: PMC10146710 DOI: 10.3390/microorganisms11040951] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/27/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Antiretroviral therapies (ART) are strongly associated with weight gain and metabolic syndrome (MetS) development in HIV-infected patients. Few studies have evaluated the association between gut microbiota and integrase strand transfer inhibitor (INSTI)-based and protease inhibitor (PI)-based regimens in HIV-infected patients with MetS. To assess this, fecal samples were obtained from HIV-infected patients treated with different regimens (16 PI + MetS or 30 INSTI + MetS) and 18 healthy controls (HCs). The microbial composition was characterized using 16S rRNA amplicon sequencing. The INSTI-based and PI-based regimens were associated with a significant decrease in α-diversity compared to HCs. The INSTI + MetS group showed the lowest α-diversity between both regimens. A significant increase in the abundance of short-chain fatty acid (SCFA)-producing genera (Roseburia, Dorea, Ruminococcus torques, and Coprococcus) was observed in the PI + MetS group, while Prevotella, Fusobacterium, and Succinivibrio were significantly increased in the INSTI + MetS group. Moreover, the Proteobacteria/Firmicutes ratio was overrepresented, and functional pathways related to the biosynthesis of LPS components were increased in the INSTI + MetS group. The gut microbiota of patients receiving INSTIs showed a more pronounced dysbiosis orchestrated by decreased bacterial richness and diversity, with an almost complete absence of SCFA-producing bacteria and alterations in gut microbiota functional pathways. These findings have not been previously observed.
Collapse
Affiliation(s)
- Tonatiuh Abimael Baltazar-Díaz
- Departamento de Biología Molecular y Genómica, Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44350, Mexico
| | - Fernando Amador-Lara
- Unidad de VIH, Hospital Civil de Guadalajara "Fray Antonio Alcalde", Guadalajara 44350, Mexico
- Departamento de Clínicas Médicas, Instituto de Investigación en Inmunodeficiencias y VIH, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44350, Mexico
| | - Jaime F Andrade-Villanueva
- Unidad de VIH, Hospital Civil de Guadalajara "Fray Antonio Alcalde", Guadalajara 44350, Mexico
- Departamento de Clínicas Médicas, Instituto de Investigación en Inmunodeficiencias y VIH, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44350, Mexico
| | - Luz Alicia González-Hernández
- Unidad de VIH, Hospital Civil de Guadalajara "Fray Antonio Alcalde", Guadalajara 44350, Mexico
- Departamento de Clínicas Médicas, Instituto de Investigación en Inmunodeficiencias y VIH, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44350, Mexico
| | - Rodolfo Ismael Cabrera-Silva
- Departamento de Clínicas Médicas, Instituto de Investigación en Inmunodeficiencias y VIH, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44350, Mexico
| | - Karina Sánchez-Reyes
- Departamento de Clínicas Médicas, Instituto de Investigación en Inmunodeficiencias y VIH, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44350, Mexico
| | - Monserrat Álvarez-Zavala
- Departamento de Clínicas Médicas, Instituto de Investigación en Inmunodeficiencias y VIH, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44350, Mexico
| | - Aldo Valenzuela-Ramírez
- Unidad de VIH, Hospital Civil de Guadalajara "Fray Antonio Alcalde", Guadalajara 44350, Mexico
| | - Susana Del Toro-Arreola
- Departamento de Biología Molecular y Genómica, Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44350, Mexico
| | - Miriam Ruth Bueno-Topete
- Departamento de Biología Molecular y Genómica, Instituto de Investigación en Enfermedades Crónico-Degenerativas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44350, Mexico
| |
Collapse
|
33
|
Gaffney J, Embree J, Gilmore S, Embree M. Chordicoccus furentiruminis, gen. nov., sp. nov., a novel succinic acid producing bacterium isolated from a steer on a high grain diet. Int J Syst Evol Microbiol 2023; 73. [PMID: 36861667 DOI: 10.1099/ijsem.0.005751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
This study presents MP1D12T (=NRRL B-67553T=NCTC 14480T), an isolate from the ruminal content of an Angus steer fed a high grain diet. Phenotypic and genotypic traits of the isolate were explored. MP1D12T was found to be a strictly anaerobic, catalase-negative, oxidase-negative, coccoid bacterium that frequently grows in chains. Analysis of metabolic products as a result of carbohydrate fermentation showed succinic acid as the major organic acid produced with lactic acid and acetic acid as minor products. Phylogenetic analysis of MP1D12T based on 16S rRNA nucleotide sequence and amino acid sequences from the whole genome presents a divergent lineage from other members in the family Lachnospiraceae. 16S rRNA sequence comparison, whole genome average nucleotide identity digital DNA-DNA hybridization and average amino acid identity results suggest that MP1D12T represents a novel species in a novel genus within the family Lachnospiraceae. We propose the creation of the genus Chordicoccus in which MP1D12T represents the type strain for the novel species Chordicoccus furentiruminis.
Collapse
Affiliation(s)
- James Gaffney
- Native Microbials, 10255 Science Center Drive Suite C2, San Diego, CA 92121, USA
| | - Jordan Embree
- Native Microbials, 10255 Science Center Drive Suite C2, San Diego, CA 92121, USA
| | - Sean Gilmore
- Native Microbials, 10255 Science Center Drive Suite C2, San Diego, CA 92121, USA
| | - Mallory Embree
- Native Microbials, 10255 Science Center Drive Suite C2, San Diego, CA 92121, USA
| |
Collapse
|
34
|
Lin D, Sun Q, Liu Z, Pan J, Zhu J, Wang S, Jia S, Zheng M, Li X, Gong F. Gut microbiota and bile acids partially mediate the improvement of fibroblast growth factor 21 on methionine-choline-deficient diet-induced non-alcoholic fatty liver disease mice. Free Radic Biol Med 2023; 195:199-218. [PMID: 36586452 DOI: 10.1016/j.freeradbiomed.2022.12.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by hepatic steatosis, inflammation, and fibrosis, as well as gut dysbiosis. Fibroblast growth factor 21 (FGF21), which regulates glucose and lipid metabolism, has been proven to have a good effect on NAFLD. However, the modulating process between FGF21 and gut microbiota remains unclear in treating NAFLD. Here, the fecal microbiota composition of 30 patients with NAFLD who had undergone liver biopsy and 29 matched healthy participants were studied, together with the fecal bile acid (BA) profile. Treatment with FGF21 was given in methionine-choline-deficient (MCD) diet-induced NAFLD model C57BL/6 mice. An antibiotic cocktail and fecal microbiota transplantation were used to further confirm the benefits of FGF21 that were partially attributable to the change in gut microbiota. Patients with NAFLD had higher serum FGF21 levels and dysregulated fecal microbiota compositions and fecal BA profiles. In NAFLD mice, FGF21 significantly reduced steatohepatitis and collagen deposition in vivo and restored intestinal structure. FGF21 treatment also changed gut microbiota composition and regulated dysbiosis in BA metabolism. After treatment with an antibiotic cocktail, FGF21 partially alleviated hepatic and intestinal damage in NAFLD mice. Furthermore, fecal microbiota transplantation from FGF21-treated mice showed benefits similar to FGF21 therapy. The improvement using FGF21 in MCD diet-induced NAFLD mice is partially mediated via gut microbiota and BA. Gut microbiota-regulated BA metabolism may be a potential target of FGF21 in improving NAFLD.
Collapse
Affiliation(s)
- Danfeng Lin
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qiyan Sun
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhaoyang Liu
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jiaxuan Pan
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jing Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Shangwen Wang
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China
| | - Sining Jia
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China
| | - Minghua Zheng
- Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Xiaokun Li
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Fanghua Gong
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
35
|
Tokuno H, Itoga T, Kasuga J, Okuma K, Hasuko K, Masuyama H, Benno Y. Method for estimating disease risk from microbiome data using structural equation modeling. Front Microbiol 2023; 14:1035002. [PMID: 36778866 PMCID: PMC9909428 DOI: 10.3389/fmicb.2023.1035002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/09/2023] [Indexed: 01/28/2023] Open
Abstract
The relationship between the human gut microbiota and disease is of increasing scientific interest. Previous investigations have focused on the differences in intestinal bacterial abundance between control and affected groups to identify disease biomarkers. However, different types of intestinal bacteria may have interacting effects and thus be considered biomarker complexes for disease. To investigate this, we aimed to identify a new kind of biomarker for atopic dermatitis using structural equation modeling (SEM). The biomarkers identified were latent variables, which are complex and derived from the abundance data for bacterial marker candidates. Groups of females and males classified as healthy participants [normal control (NC) (female: 321 participants, male: 99 participants)], and patients afflicted with atopic dermatitis only [AS (female: 45 participants, male: 13 participants)], with atopic dermatitis and other diseases [AM (female: 75 participants, male: 34 participants)], and with other diseases but without atopic dermatitis [OD (female: 1,669 participants, male: 866 participants)] were used in this investigation. The candidate bacterial markers were identified by comparing the intestinal microbial community compositions between the NC and AS groups. In females, two latent variables (lv) were identified; for lv1, the associated components (bacterial genera) were Alistipes, Butyricimonas, and Coprobacter, while for lv2, the associated components were Agathobacter, Fusicatenibacter, and Streptococcus. There was a significant difference in the lv2 scores between the groups with atopic dermatitis (AS, AM) and those without (NC, OD), and the genera identified for lv2 are associated with the suppression of inflammatory responses in the body. A logistic regression model to estimate the probability of atopic dermatitis morbidity with lv2 as an explanatory variable had an area under the curve (AUC) score of 0.66 when assessed using receiver operating characteristic (ROC) analysis, and this was higher than that using other logistic regression models. The results indicate that the latent variables, especially lv2, could represent the effects of atopic dermatitis on the intestinal microbiome in females. The latent variables in the SEM could thus be utilized as a new type of biomarker. The advantages identified for the SEM are as follows: (1) it enables the extraction of more sophisticated information when compared with models focused on individual bacteria and (2) it can improve the accuracy of the latent variables used as biomarkers, as the SEM can be expanded.
Collapse
Affiliation(s)
- Hidetaka Tokuno
- Symbiosis Solutions Inc., Tokyo, Japan,*Correspondence: Hidetaka Tokuno,
| | | | | | | | | | | | | |
Collapse
|
36
|
Kuo YJ, Chen CJ, Hussain B, Tsai HC, Hsu GJ, Chen JS, Asif A, Fan CW, Hsu BM. Inferring Bacterial Community Interactions and Functionalities Associated with Osteopenia and Osteoporosis in Taiwanese Postmenopausal Women. Microorganisms 2023; 11:234. [PMID: 36838199 PMCID: PMC9959971 DOI: 10.3390/microorganisms11020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/30/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Growing evidence suggests that the gut microbiota and their metabolites are associated with bone homeostasis and fragility. However, this association is limited to microbial taxonomic differences. This study aimed to explore whether gut bacterial community associations, composition, and functions are associated with osteopenia and osteoporosis. We compared the gut bacterial community composition and interactions of healthy postmenopausal women with normal bone density (n = 8) with those of postmenopausal women with osteopenia (n = 18) and osteoporosis (n = 21) through 16S rRNA sequencing coupled with network biology and statistical analyses. The results of this study showed reduced alpha diversity in patients with osteoporosis, followed by that in patients with osteopenia, then in healthy controls. Taxonomic analysis revealed that significantly enriched bacterial genera with higher abundance was observed in patients with osteoporosis and osteopenia than in healthy subjects. Additionally, a co-occurrence network revealed that, compared to healthy controls, bacterial interactions were higher in patients with osteoporosis, followed by those with osteopenia. Further, NetShift analysis showed that a higher number of bacteria drove changes in the microbial community structure of patients with osteoporosis than osteopenia. Correlation analysis revealed that most of these driver bacteria had a significant positive relationship with several significant metabolic pathways. Further, ordination analysis revealed that height and T-score were the primary variables influencing the gut microbial community structure. Taken together, this study evaluated that microbial community interaction is more important than the taxonomic differences in knowing the critical role of gut microbiota in postmenopausal women associated with osteopenia and osteoporosis. Additionally, the significantly enriched bacteria and functional pathways might be potential biomarkers for the prognosis and treatment of postmenopausal women with osteopenia and osteoporosis.
Collapse
Affiliation(s)
- Yi-Jie Kuo
- Department of Orthopedic Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| | - Chia-Jung Chen
- Department of Chinese Medicine, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Chiayi 622, Taiwan
| | - Bashir Hussain
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 621, Taiwan
| | - Hsin-Chi Tsai
- Department of Psychiatry, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
- Department of Psychiatry, Tzu-Chi General Hospital, Hualien 970, Taiwan
| | - Gwo-Jong Hsu
- Division of Infectious Disease, Department of Internal Medicine, Chia-Yi Christian Hospital, Chiayi 621, Taiwan
| | - Jung-Sheng Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824, Taiwan
| | - Aslia Asif
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
- Doctoral Program in Science, Technology, Environment and Mathematics, National Chung Cheng University, Chiayi 621, Taiwan
| | - Cheng-Wei Fan
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
| | - Bing-Mu Hsu
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
| |
Collapse
|
37
|
Sheridan PO, Odat MA, Scott KP. Establishing genetic manipulation for novel strains of human gut bacteria. MICROBIOME RESEARCH REPORTS 2023; 2:1. [PMID: 38059211 PMCID: PMC10696588 DOI: 10.20517/mrr.2022.13] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/29/2022] [Accepted: 12/12/2022] [Indexed: 12/08/2023]
Abstract
Recent years have seen the development of high-accuracy and high-throughput genetic manipulation techniques, which have greatly improved our understanding of genetically tractable microbes. However, challenges remain in establishing genetic manipulation techniques in novel organisms, owing largely to exogenous DNA defence mechanisms, lack of selectable markers, lack of efficient methods to introduce exogenous DNA and an inability of genetic vectors to replicate in their new host. In this review, we describe some of the techniques that are available for genetic manipulation of novel microorganisms. While many reviews exist that focus on the final step in genetic manipulation, the editing of recipient DNA, we particularly focus on the first step in this process, the transfer of exogenous DNA into a strain of interest. Examples illustrating the use of these techniques are provided for a selection of human gut bacteria in which genetic tractability has been established, such as Bifidobacterium, Bacteroides and Roseburia. Ultimately, this review aims to provide an information source for researchers interested in developing genetic manipulation techniques for novel bacterial strains, particularly those of the human gut microbiota.
Collapse
Affiliation(s)
- Paul O. Sheridan
- School of Biological and Chemical Sciences, University of Galway, Galway H91 TK33, Ireland
| | - Ma’en Al Odat
- Gut Health Group, Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, Scotland AB25 2ZD, UK
| | - Karen P. Scott
- Gut Health Group, Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, Scotland AB25 2ZD, UK
| |
Collapse
|
38
|
Wang Z, Li Q, Lan X, Shen W, Wan F, He J, Tang S, Tan Z. Evaluation of stirring time through a rumen simulation technique: Influences on rumen fermentation and bacterial community. Front Microbiol 2023; 14:1103222. [PMID: 36950158 PMCID: PMC10026382 DOI: 10.3389/fmicb.2023.1103222] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Rumen motility is a key element that influences ruminant nutrition, whereas little is known about the effects of rumen contraction duration on rumen fermentation and ruminal microbiome. We previously reported that proper rotation speed of a rumen simulation technique (RUSITEC) system enhanced rumen fermentation and microbial protein (MCP) production. In the present study, different contraction durations and intervals were simulated by setting different stirring times and intervals of the stirrers in a RUSITEC system. The objective of this trial was to evaluate the influences of stirring time on rumen fermentation characteristics, nutrient degradation, and ruminal bacterial microbiota in vitro. Methods This experiment was performed in a 3 × 3 Latin square design, with each experimental period comprising 4 d for adjustment and 3 d for sample collection. Three stirring time treatments were set: the constant stir (CS), the intermittent stir 1 (each stir for 5 min with an interval of 2 min, IS1), and the intermittent stir 2 (each stir for 4 min with an interval of 3 min, IS2). Results The total volatile fatty acid (TVFA) concentration, valerate molar proportion, ammonia nitrogen level, MCP density, protozoa count, disappearance rates of dry matter, organic matter, crude protein, neutral detergent fiber, and acid detergent fiber, emissions of total gas and methane, and the richness index Chao 1 for the bacterial community were higher (p < 0.05) in the IS1 when compared to those in the CS. The greatest TVFA, MCP, protozoa count, nutrient disappearance rates, gas productions, and bacterial richness indices of Ace and Chao 1 amongst all treatments were observed in the IS2. The relative abundance of the genus Treponema was enriched (p < 0.05) in CS, while the enrichment (p < 0.05) of Agathobacter ruminis and another two less known bacterial genera were identified in IS2. Discussion It could be concluded that the proper reduction in the stirring time might help to enhance the feed fermentation, MCP synthesis, gas production, and the relative abundances of specific bacterial taxa.
Collapse
Affiliation(s)
- Zuo Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Quan Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Xinyi Lan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Weijun Shen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
- *Correspondence: Weijun Shen,
| | - Fachun Wan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Jianhua He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Shaoxun Tang
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Zhiliang Tan
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| |
Collapse
|
39
|
Muller E, Algavi YM, Borenstein E. The gut microbiome-metabolome dataset collection: a curated resource for integrative meta-analysis. NPJ Biofilms Microbiomes 2022; 8:79. [PMID: 36243731 PMCID: PMC9569371 DOI: 10.1038/s41522-022-00345-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/04/2022] [Indexed: 12/03/2022] Open
Abstract
Integrative analysis of microbiome and metabolome data obtained from human fecal samples is a promising avenue for better understanding the interplay between bacteria and metabolites in the human gut, in both health and disease. However, acquiring, processing, and unifying such datasets from multiple sources is a daunting and challenging task. Here we present a publicly available, simple-to-use, curated dataset collection of paired fecal microbiome-metabolome data from multiple cohorts. This data resource allows researchers to easily obtain multiple fully processed and integrated microbiome-metabolome datasets, facilitating the discovery of universal microbe-metabolite links, benchmark various microbiome-metabolome integration tools, and compare newly identified microbe-metabolite findings to other published datasets.
Collapse
Affiliation(s)
- Efrat Muller
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Yadid M Algavi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elhanan Borenstein
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel.
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Santa Fe Institute, Santa Fe, NM, USA.
| |
Collapse
|
40
|
Lee MJ, Lai HC, Kuo YL, Chen VCH. Association between Gut Microbiota and Emotional-Behavioral Symptoms in Children with Attention-Deficit/Hyperactivity Disorder. J Pers Med 2022; 12:jpm12101634. [PMID: 36294773 PMCID: PMC9605220 DOI: 10.3390/jpm12101634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
Previous studies have explored the role of the microbiome in attention-deficit/hyperactivity disorder (ADHD). However, whether the microbiome is correlated with emotional-behavioral disturbances, the most common comorbid symptom of ADHD, remains unclear. We established a cross-sectional study in which 6- to 18-year-old children with ADHD who were receiving no medication and a healthy control group of children without ADHD were recruited to analyze their microbiome composition. Microbiota of fecal samples were collected and analyzed using a 16s rRNA gene sequencing approach. In comparison with the healthy control group, the gut microbiota in children with ADHD exhibited significantly lower beta diversity. The abundance of the phylum Proteobacteria and the genera Agathobacter, Phascolarctobacterium, Prevotella_2, Acidaminococcus, Roseburia, and Ruminococcus gnavus group was increased in the ADHD group compared with the healthy group. Linear discriminant effect size (LEfSe) analysis was used to highlight specific bacteria phylotypes that were differentially altered between the ADHD and control groups. A regression analysis was performed to investigate the association between microbiota and emotional-behavioral symptoms in children with ADHD. A significant association was noted between withdrawal and depression symptoms and Agathobacter (p = 0.044), and between rule-breaking behavior and the Ruminococcus gnavus group (p = 0.046) after adjusting for sex, age, and the ADHD core symptoms score. This study advances the knowledge of how gut microbiota composition may contribute to emotional-behavioral symptoms in children with ADHD. The detailed mechanisms underlying the role of the gut microbiota in ADHD pathophysiology still require further investigation.
Collapse
Affiliation(s)
- Min-Jing Lee
- Department of Psychiatry, Chang Gung Memorial Hospital, Chiayi Branch, Chiayi 613, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Hsin-Chih Lai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Microbiota Research Center and Emerging Viral Infections Research Center, Chang Gung University, Taoyuan 333, Taiwan
- Central Research Laboratory, Xiamen Chang Gung Hospital, Xiamen 361, China
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
| | - Yu-Lun Kuo
- Biotools Co., Ltd., New Taipei City 221, Taiwan
| | - Vincent Chin-Hung Chen
- Department of Psychiatry, Chang Gung Memorial Hospital, Chiayi Branch, Chiayi 613, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Correspondence: ; Tel.: +886-5-3621000 (ext. 2315); Fax: +886-5-3623002
| |
Collapse
|
41
|
Shang J, Zhang Y, Guo R, Liu W, Zhang J, Yan G, Wu F, Cui W, Wang P, Zheng X, Wang T, Dong Y, Zhao J, Wang L, Xiao J, Zhao Z. Gut Microbiome Analysis Can Be Used as a Noninvasive Diagnostic Tool and Plays an Essential Role in the Onset of Membranous Nephropathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201581. [PMID: 35975460 PMCID: PMC9534961 DOI: 10.1002/advs.202201581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/06/2022] [Indexed: 05/15/2023]
Abstract
Membranous nephropathy (MN) is a common cause of nephrotic syndrome. The aim is to establish a non-invasive diagnostic model of MN using differential gut microbiome analysis, and to explore the relationship between the gut microbiome and MN pathogenesis in vivo. 825 fecal samples from MN patients and healthy participants are collected from multiple medical centers across China. Key operational taxonomic units (OTUs) obtained through 16S rRNA sequencing are used to establish a diagnostic model. A rat model of MN is developed to explore the relationship between the gut microbiome and the pathogenesis of MN. The diversity and richness of the gut microbiome are significantly lower in patients with MN than in healthy individuals. The diagnostic model based on seven OTUs achieves an excellent efficiency of 98.36% in the training group and also achieves high efficiency in cross-regional cohorts. In MN rat model, gut microbiome elimination prevents model establishment, but fecal microbiome transplantation restores the phenotype of protein urine. Gut microbiome analysis can be used as a non-invasive tool for MN diagnosis. The onset of MN depends on the presence of naturally colonized microbiome. Early intervention in the gut microbiome may help reduce urinary protein level in MN.
Collapse
Affiliation(s)
- Jin Shang
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
- Laboratory Animal Platform of Academy of Medical SciencesZhengzhou UniversityZhengzhouHenan450000P. R. China
- Laboratory of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052P. R. China
| | - Yiding Zhang
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Ruixue Guo
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Wenli Liu
- Department of Clinical LaboratoryPeking Union Medical College HospitalPeking Union Medical College & China Academy of Medical ScienceBeijing100730China
| | - Jun Zhang
- Department of Nephrologythe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdong510630China
| | - Ge Yan
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Feng Wu
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Wen Cui
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Peipei Wang
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Xuejun Zheng
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Ting Wang
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Yijun Dong
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Jing Zhao
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Li Wang
- Biobank of The First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052P. R. China
| | - Jing Xiao
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
- Laboratory of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052P. R. China
| | - Zhanzheng Zhao
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
- Laboratory Animal Platform of Academy of Medical SciencesZhengzhou UniversityZhengzhouHenan450000P. R. China
- Laboratory of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052P. R. China
| |
Collapse
|
42
|
Qiu L, Gong F, Wu J, You D, Zhao Y, Xu L, Cao X, Bao F. Exercise Interventions Improved Sleep Quality through Regulating Intestinal Microbiota Composition. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph191912385. [PMID: 36231686 PMCID: PMC9564517 DOI: 10.3390/ijerph191912385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/18/2022] [Accepted: 09/26/2022] [Indexed: 05/08/2023]
Abstract
(1) Background: Sleep quality is closely related to the physical and mental health of college students. The objectives of this study were to obtain data on the sleep quality of university students and to investigate the relationship between intestinal flora and the improvement in sleep quality through exercise intervention. (2) Methods: Here, 11 university students with a body mass index (BMI) ≤ 18 and Pittsburgh Sleep Quality Index (PSQI) ≥ 7 were selected as experimental subjects, and another 11 healthy people were recruited as control subjects. The experimental group and control group were each intervened with exercise for 8 weeks. We used 16SrDNA sequencing technology to analyze the variations of the intestinal flora and the relation of the variations and sleep quality improvement between the experimental group and the control group before and after the exercise intervention. (3) Results: The differences in gut flora composition between people with sleep disorders and healthy people were statistically significant (p < 0.05). Before and after the exercise intervention, the differences were also statistically significant (p < 0.05) in people with sleep disorders. The sleep-disordered population had a larger proportion compared with the healthy population (p < 0.05). Blautia and Eubacterium hallii were microbe markers in the sleep-disordered population before and after the exercise intervention, while there was no microbe marker found in the healthy population. (4) Conclusions: The increase in Blautia and Eubacterium hallii, and the decrease in Agathobacter are associated with healthy sleep. Gut flora may be related to sleep disorders. Exercise intervention can improve sleep quality while changing the diversity of the gut flora, and exercise intervention targeting the gut flora is a new concept for preventing and treating sleep disorders.
Collapse
Affiliation(s)
- Liangwu Qiu
- Department of Physical Education, Kunming Medical University, Kunming 650500, China
| | - Fuhong Gong
- Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, China
| | - Jiang Wu
- Department of Physical Education, Kunming Medical University, Kunming 650500, China
| | - Dingyun You
- Biomedical Engineering Research Center, Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Kunming Medical University, Kunming 650500, China
| | - Yinzhou Zhao
- Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, China
| | - Lianwu Xu
- Department of Physical Education, Kunming Medical University, Kunming 650500, China
- Correspondence: (L.X.); (X.C.); (F.B.)
| | - Xue Cao
- Department of Laboratory Animal Science, Kunming Medical University, Kunming 650500, China
- Correspondence: (L.X.); (X.C.); (F.B.)
| | - Fukai Bao
- Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, China
- Correspondence: (L.X.); (X.C.); (F.B.)
| |
Collapse
|
43
|
Li K, Methé BA, Fitch A, Gentry H, Kessinger C, Patel A, Petraglia V, Swamy P, Morris A. Gut and oral microbiota associations with viral mitigation behaviors during the COVID-19 pandemic. Front Cell Infect Microbiol 2022; 12:966361. [PMID: 36159641 PMCID: PMC9500509 DOI: 10.3389/fcimb.2022.966361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Imposition of social and health behavior mitigations are important control measures in response to the coronavirus disease 2019 (COVID-19) pandemic caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Although postulated that these measures may impact the human microbiota including losses in diversity from heightened hygiene and social distancing measures, this hypothesis remains to be tested. Other impacts on the microbiota and host mental and physical health status associations from these measures are also not well-studied. Here we examine changes in stool and oral microbiota by analyzing 16S rRNA gene sequence taxonomic profiles from the same individuals during pre-pandemic (before March 2020) and early pandemic (May-November 2020) phases. During the early pandemic phase, individuals were also surveyed using questionnaires to report health histories, anxiety, depression, sleep and other lifestyle behaviors in a cohort of predominantly Caucasian adults (mean age = 61.5 years) with the majority reporting at least one underlying co-morbidity. We identified changes in microbiota (stool n = 288; oral n = 89) between pre-pandemic and early pandemic time points from the same subject and associated these differences with questionnaire responses using linear statistical models and hierarchical clustering of microbiota composition coupled to logistic regression. While a trend in loss of diversity was identified between pre-pandemic and early pandemic time points it was not statistically significant. Paired difference analyses between individuals identified fewer significant changes between pre-pandemic and early pandemic microbiota in those who reported fewer comorbidities. Cluster transition analyses of stool and saliva microbiota determined most individuals remained in the same cluster assignments from the pre-pandemic to early pandemic period. Individuals with microbiota that shifted in composition, causing them to depart a pre-pandemic cluster, reported more health issues and pandemic-associated worries. Collectively, our study identified that stool and saliva microbiota from the pre-pandemic to early pandemic periods largely exhibited ecological stability (especially stool microbiota) with most associations in loss of diversity or changes in composition related to more reported health issues and pandemic-associated worries. Longitudinal observational cohorts are necessary to monitor the microbiome in response to pandemics and changes in public health measures.
Collapse
Affiliation(s)
- Kelvin Li
- Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Barbara A. Methé
- Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Adam Fitch
- Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Heather Gentry
- Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Cathy Kessinger
- Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Asha Patel
- Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Vickie Petraglia
- Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Pruthvi Swamy
- Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Alison Morris
- Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| |
Collapse
|
44
|
Lu D, Zou X, Zhang H. The Relationship Between Atrial Fibrillation and Intestinal Flora With Its Metabolites. Front Cardiovasc Med 2022; 9:948755. [PMID: 35845042 PMCID: PMC9283774 DOI: 10.3389/fcvm.2022.948755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/02/2022] [Indexed: 01/01/2023] Open
Abstract
Atrial fibrillation (AF) is characterized by high morbidity and disability rate. The incidence of AF has rapidly increased due to increased aging population, causing a serious burden on society and patients. Therefore, it is necessary to determine the prevention and treatment of AF. Several studies have assessed the occurrence, development mechanism, and intervention measures of AF. The human gut has several non-pathogenic microorganisms forming the gut flora. The human gut microbiota plays a crucial role in the construction and operation of the metabolic system and immune system. Emerging clinical studies and basic experiments have confirmed that intestinal flora and its metabolites have a role in some metabolic disorders and chronic inflammatory diseases. Moreover, the gut microbiota has a role in cardiovascular diseases, such as hypertension and heart failure. However, the relationship between AF and gut microbiota is unclear. This review summarizes the relevant literature on the relationship between AF and intestinal flora with its metabolites, including Trimethylamine N-Oxide, short-chain fatty acids, lipopolysaccharide and bile acids. Therefore, this review may enhance further development of related research.
Collapse
Affiliation(s)
- Dasheng Lu
- Department of Cardiology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
- Vascular Diseases Research Center of Wannan Medical College, Wuhu, China
- *Correspondence: Dasheng Lu
| | - Xinyue Zou
- Department of Cardiology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Hongxiang Zhang
- Department of Cardiology, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
- Vascular Diseases Research Center of Wannan Medical College, Wuhu, China
- Hongxiang Zhang
| |
Collapse
|
45
|
Escherichia/ Shigella, SCFAs, and Metabolic Pathways-The Triad That Orchestrates Intestinal Dysbiosis in Patients with Decompensated Alcoholic Cirrhosis from Western Mexico. Microorganisms 2022; 10:microorganisms10061231. [PMID: 35744749 PMCID: PMC9229093 DOI: 10.3390/microorganisms10061231] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/07/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022] Open
Abstract
Gut microbiota undergoes profound alterations in alcohol cirrhosis. Microbiota-derived products, e.g., short chain fatty acids (SCFA), regulate the homeostasis of the gut-liver axis. The objective was to evaluate the composition and functions of the intestinal microbiota in patients with alcohol-decompensated cirrhosis. Fecal samples of 18 patients and 18 healthy controls (HC) were obtained. Microbial composition was characterized by 16S rRNA amplicon sequencing, SCFA quantification was performed by gas chromatography (GC), and metagenomic predictive profiles were analyzed by PICRUSt2. Gut microbiota in the cirrhosis group revealed a significant increase in the pathogenic/pathobionts genera Escherichia/Shigella and Prevotella, a decrease in beneficial bacteria, such as Blautia, Faecalibacterium, and a decreased α-diversity (p < 0.001) compared to HC. Fecal SCFA concentrations were significantly reduced in the cirrhosis group (p < 0.001). PICRUSt2 analysis indicated a decrease in acetyl-CoA fermentation to butyrate, as well as an increase in pathways related to antibiotics resistance, and aromatic amino acid biosynthesis. These metabolic pathways have been poorly described in the progression of alcohol-related decompensated cirrhosis. The gut microbiota of these patients possesses a pathogenic/inflammatory environment; therefore, future strategies to balance intestinal dysbiosis should be implemented. These findings are described for the first time in the population of western Mexico.
Collapse
|
46
|
Oh HS, Min U, Jang H, Kim N, Lim J, Chalita M, Chun J. Proposal of a health gut microbiome index based on a meta-analysis of Korean and global population datasets. JOURNAL OF MICROBIOLOGY (SEOUL, KOREA) 2022; 60:533-549. [PMID: 35362897 DOI: 10.1007/s12275-022-1526-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/03/2022] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
Abstract
The disruption of the human gut microbiota has been linked to host health conditions, including various diseases. However, no reliable index for measuring and predicting a healthy microbiome is currently available. Here, the sequencing data of 1,663 Koreans were obtained from three independent studies. Furthermore, we pooled 3,490 samples from public databases and analyzed a total of 5,153 fecal samples. First, we analyzed Korean gut microbiome covariates to determine the influence of lifestyle on variation in the gut microbiota. Next, patterns of microbiota variations across geographical locations and disease statuses were confirmed using a global cohort and di-sease data. Based on comprehensive comparative analysis, we were able to define three enterotypes among Korean cohorts, namely, Prevotella type, Bacteroides type, and outlier type. By a thorough categorization of dysbiosis and the evaluation of microbial characteristics using multiple datasets, we identified a wide spectrum of accuracy levels in classifying health and disease states. Using the observed microbiome patterns, we devised an index named the gut microbiome index (GMI) that could consistently predict health conditions from human gut microbiome data. Compared to ecological metrics, the microbial marker index, and machine learning approaches, GMI distinguished between healthy and non-healthy individuals with a higher accuracy across various datasets. Thus, this study proposes a potential index to measure health status of gut microbiome that is verified from multiethnic data of various diseases, and we expect this model to facilitate further clinical application of gut microbiota data in future.
Collapse
Affiliation(s)
- Hyun-Seok Oh
- ChunLab Inc., Seoul, 06194, Republic of Korea.,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Uigi Min
- ChunLab Inc., Seoul, 06194, Republic of Korea
| | - Hyejin Jang
- ChunLab Inc., Seoul, 06194, Republic of Korea
| | - Namil Kim
- ChunLab Inc., Seoul, 06194, Republic of Korea
| | | | | | - Jongsik Chun
- ChunLab Inc., Seoul, 06194, Republic of Korea. .,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea. .,School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
47
|
The Effects of High Fiber Rye, Compared to Refined Wheat, on Gut Microbiota Composition, Plasma Short Chain Fatty Acids, and Implications for Weight Loss and Metabolic Risk Factors (the RyeWeight Study). Nutrients 2022; 14:nu14081669. [PMID: 35458231 PMCID: PMC9032876 DOI: 10.3390/nu14081669] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 02/04/2023] Open
Abstract
Consumption of whole grain and cereal fiber have been inversely associated with body weight and obesity measures in observational studies but data from large, long-term randomized interventions are scarce. Among the cereals, rye has the highest fiber content and high rye consumption has been linked to increased production of gut fermentation products, as well as reduced risks of obesity and metabolic disease. The effects on body weight and metabolic risk factors may partly be mediated through gut microbiota and/or their fermentation products. We used data from a randomized controlled weight loss trial where participants were randomized to a hypocaloric diet rich in either high fiber rye foods or refined wheat foods for 12 weeks to investigate the effects of the intervention on gut microbiota composition and plasma short chain fatty acids, as well as the potential association with weight loss and metabolic risk markers. Rye, compared to wheat, induced some changes in gut microbiota composition, including increased abundance of the butyrate producing Agathobacter and reduced abundance of [Ruminococcus] torques group, which may be related to reductions in low grade inflammation caused by the intervention. Plasma butyrate increased in the rye group. In conclusion, intervention with high fiber rye foods induced some changes in gut microbiota composition and plasma short chain fatty acid concentration, which were associated with improvements in metabolic risk markers as a result of the intervention.
Collapse
|
48
|
Gut Microbiota and Metabolites in Atrial Fibrillation Patients and Their Changes after Catheter Ablation. Microbiol Spectr 2022; 10:e0107721. [PMID: 35384710 PMCID: PMC9045169 DOI: 10.1128/spectrum.01077-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The gut microbiota has been shown to be associated with multiple cardiovascular diseases, but there is little research on the gut microbiota and atrial fibrillation (AF); thus, how the gut microbiota and metabolites change in AF patients after catheter ablation is unclear. In this study, we used 16S rRNA high-throughput sequencing and nontargeted metabolomic detection to conduct horizontal and longitudinal analyses of the gut microbiota and metabolites of AF patients. Compared with a control group, species richness and diversity increased significantly in AF patients. Among them, opportunistic pathogenic bacteria, such as Klebsiella, Haemophilus, Streptococcus, and Enterococcus, were significantly increased, and symbiotic bacteria, such as Agathobacter and Butyrivibrio, were significantly reduced. After catheter ablation, intestinal symbiotic bacteria (Lactobacillus, Agathobacter, Lachnospira, etc.) were increased in most AF patients, while pathogenic bacteria (Ruminococcus, etc.) were reduced. Moreover, in AF patients, caffeine, which was negatively correlated with Klebsiella, was downregulated, and estradiol and ascorbic acid, which were positively correlated with Agathobacter, were also downregulated. After catheter ablation, citrulline, which was positively correlated with Ralstonia and Lactobacillus, was increased. Oleanolic acid, which was negatively correlated with Ralstonia was downregulated. In conclusion, our results not only show overall changes in the gut microbiota and metabolites in AF patients but also indicate their changes in the short term after catheter ablation. These data will provide novel possibilities for the future clinical diagnosis and treatment of AF. IMPORTANCE Gut microbiota and metabolites play a very important role in human health and can not only assess human health but also treat and prevent diseases. We analyzed the characteristics of the microbiota and metabolites in the human gut and found the effect of disease on gut microbiota and metabolites, which may be of important value in the pathogenesis of atrial fibrillation. At the same time, we also observed dynamic changes in gut microbiota and metabolites with the intervention of catheter ablation, which was not available in previous studies.
Collapse
|
49
|
Andriulli A, Bevilacqua A, Palmieri O, Latiano A, Fontana R, Gioffreda D, Castellana S, Mazza T, Panza A, Menzaghi C, Grandone E, di Mauro L, Decina I, Tricarico M, Musaico D, Mäki M, Isola J, Popp A, Taavela J, Petruzzi L, Sinigaglia M, Rosaria Corbo M, Lamacchia C. Healthy and pro-inflammatory gut ecology plays a crucial role in the digestion and tolerance of a novel Gluten Friendly™ bread in celiac subjects: a randomized, double blind, placebo control in vivo study. Food Funct 2022; 13:1299-1315. [PMID: 35029612 DOI: 10.1039/d1fo00490e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gluten Friendly™ (GF) is a new gluten achieved through a physicochemical process applied to wheat kernels. The goal of this research was to assess the in vivo effects of Gluten Friendly™ bread on celiac gut mucosa and microbiota. In a double-blind placebo-controlled intervention study, 48 celiac disease (CD) patients were randomized into 3 groups to eat 100 g of bread daily, containing different doses (0; 3 g; 6 g) of GF for 12 weeks. The small-bowel morphology (VH/CrD), intraepithelial densities of CD3+, celiac serology, MUC2, CB1, gut permeability, proinflammatory cytokines, gluten in stools, symptoms, and gut microbial composition were assessed. All 48 CD subjects experienced no symptoms. K-means analysis evidenced celiac subjects clustering around unknown parameters independent of GF dosage: K1 35%; K2 30%; K3 35%. VH/CrD significantly decreased in K1 and K2. VH/CrD did not correlate with IEL increase in K2. 33-mer was not detected in 47% and 73% of patients in both K1 and K2, respectively. VH/CrD and IEL did not change significantly and strongly correlated with the absence of 33-mer in K3. Inflammation and VH/CrD decrease are strongly related with the presence of proinflammatory species at the baseline. A boost in probiotic, butyrate-producing genera, is strongly related with GF tolerance at the end of the trial. Our research suggests that a healthy and proinflammatory ecology could play a crucial role in the digestion and tolerance of the new gluten molecule in celiac subjects. However, GF can be completely digested by gut microbiota of CD subjects and shapes it toward gut homeostasis by boosting healthy butyrate-producing populations. The clinical trial registry number is NCT03137862 (https://clinicaltrials.gov).
Collapse
Affiliation(s)
- Angelo Andriulli
- Division of Gastroenterology, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Antonio Bevilacqua
- Università degli Studi di Foggia, Dipartimento di Scienze dell'Agricoltura, degli Alimenti e dell'Ambiente, Foggia, Italy.
| | - Orazio Palmieri
- Division of Gastroenterology, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Anna Latiano
- Division of Gastroenterology, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Rosanna Fontana
- Division of Gastroenterology, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Domenica Gioffreda
- Division of Gastroenterology, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Stefano Castellana
- Bioinformatics Unit, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (Foggia), Italy
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (Foggia), Italy
| | - Anna Panza
- Division of Gastroenterology, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Claudia Menzaghi
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Elvira Grandone
- Atherosclerosis and Thrombosis Unit, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Lazzaro di Mauro
- Immunohaematology and Transfusion Medicine Service, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | | | | | - Daniela Musaico
- Università degli Studi di Foggia, Dipartimento di Scienze dell'Agricoltura, degli Alimenti e dell'Ambiente, Foggia, Italy.
| | - Markku Mäki
- Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital Tampere, Finland
| | - Jorma Isola
- Faculty of Medicine and Health Technology, Tampere University and Jilab Inc., Tampere, Finland
| | - Alina Popp
- University of Medicine and Pharmacy "Carol Davila" and National Institute for Mother and Child Health "Alessandrescu-Rusescu", Bucharest, Romania
| | - Juha Taavela
- Department of Internal Medicine, Central Finland Central Hospital, Jyväskylä, Finland
| | - Leonardo Petruzzi
- Università degli Studi di Foggia, Dipartimento di Scienze dell'Agricoltura, degli Alimenti e dell'Ambiente, Foggia, Italy.
| | - Milena Sinigaglia
- Università degli Studi di Foggia, Dipartimento di Scienze dell'Agricoltura, degli Alimenti e dell'Ambiente, Foggia, Italy.
| | - Maria Rosaria Corbo
- Università degli Studi di Foggia, Dipartimento di Scienze dell'Agricoltura, degli Alimenti e dell'Ambiente, Foggia, Italy.
| | - Carmela Lamacchia
- Università degli Studi di Foggia, Dipartimento di Scienze dell'Agricoltura, degli Alimenti e dell'Ambiente, Foggia, Italy. .,New Gluten World s.r.l., Foggia, Italy
| |
Collapse
|
50
|
Chen L, Zheng T, Yang Y, Chaudhary PP, Teh JPY, Cheon BK, Moses D, Schuster SC, Schlundt J, Li J, Conway PL. Integrative multiomics analysis reveals host-microbe-metabolite interplays associated with the aging process in Singaporeans. Gut Microbes 2022; 14:2070392. [PMID: 35549618 PMCID: PMC9116421 DOI: 10.1080/19490976.2022.2070392] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/18/2022] [Indexed: 02/04/2023] Open
Abstract
The age-associated alterations in microbiomes vary across populations due to the influence of genetics and lifestyles. To the best of our knowledge, the microbial changes associated with aging have not yet been investigated in Singapore adults. We conducted shotgun metagenomic sequencing of fecal and saliva samples, as well as fecal metabolomics to characterize the gut and oral microbial communities of 62 healthy adult male Singaporeans, including 32 young subjects (age, 23.1 ± 1.4 years) and 30 elderly subjects (age, 69.0 ± 3.5 years). We identified 8 gut and 13 oral species that were differentially abundant in elderly compared to young subjects. By combining the gut and oral microbiomes, 25 age-associated oral-gut species connections were identified. Moreover, oral bacteria Acidaminococcus intestine and Flavonifractor plautii were less prevalent/abundant in elderly gut samples than in young gut samples, whereas Collinsella aerofaciens and Roseburia hominis showed the opposite trends. These results indicate the varied gut-oral communications with aging. Subsequently, we expanded the association studies on microbiome, metabolome and host phenotypic parameters. In particular, Eubacterium eligens increased in elderly compared to young subjects, and was positively correlated with triglycerides, which implies that the potential role of E. eligens in lipid metabolism is altered during the aging process. Our results demonstrated aging-associated changes in the gut and oral microbiomes, as well as the connections between metabolites and host-microbe interactions, thereby deepening the understanding of alterations in the human microbiome during the aging process in a Singapore population.
Collapse
Affiliation(s)
- Liwei Chen
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore
- Nanyang Technological University Food Technology Centre (NAFTEC), College of Engineering, Nanyang Technological University, Singapore
| | - Tingting Zheng
- Department of Infectious Diseases and Public Health, The Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, China
| | - Yifan Yang
- Office of Education Research, and Physical Education and Sports Science, National Institute of Education, Nanyang Technological University, Singapore
| | - Prem Prashant Chaudhary
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore
- Nanyang Technological University Food Technology Centre (NAFTEC), College of Engineering, Nanyang Technological University, Singapore
- Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - Jean Pui Yi Teh
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore
- Nanyang Technological University Food Technology Centre (NAFTEC), College of Engineering, Nanyang Technological University, Singapore
| | - Bobby K. Cheon
- School of Social Sciences, Nanyang Technological University, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research (A*STAR), Singapore
- Eunice Kenndy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniela Moses
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore
| | - Stephan C. Schuster
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore
| | - Joergen Schlundt
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore
- Nanyang Technological University Food Technology Centre (NAFTEC), College of Engineering, Nanyang Technological University, Singapore
| | - Jun Li
- Department of Infectious Diseases and Public Health, The Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, China
- School of Data Science, City University of Hong Kong, Hong Kong, China
| | - Patricia L. Conway
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore
- Nanyang Technological University Food Technology Centre (NAFTEC), College of Engineering, Nanyang Technological University, Singapore
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences,The University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|