1
|
Benech N, Cassir N, Alric L, Barbut F, Batista R, Bleibtreu A, Briot T, Davido B, Galperine T, Joly A, Kapel N, Melchior C, Mosca A, Nebbad B, Pigneur B, Schneider SM, Wasiak M, Scanzi J, Sokol H. Impact of Clinical and Pharmacological Parameters on Faecal Microbiota Transplantation Outcome in Clostridioides difficile Infections: Results of a 5-Year French National Survey. Aliment Pharmacol Ther 2025; 61:159-167. [PMID: 39387234 PMCID: PMC11636172 DOI: 10.1111/apt.18330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/13/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Detailed comparative assessment of procedure-related factors associated with faecal microbiota transplantation (FMT) efficacy in Clostridioides difficile infection (CDI) is limited. AIMS We took advantage of the differences in procedures at the various French FMT centres to determine clinical and procedure-related factors associated with FMT success in CDI. METHODS We performed a nationwide retrospective multicentre cohort study. All FMTs performed within The French Faecal Transplant Group for CDI from 2018 to 2022 were included. Clinical data were collected retrospectively from recipient medical files, characteristics of stool transplant preparations were prospectively collected by each Pharmacy involved. Univariate and multivariate analyses were performed using Fisher's test and multiple logistic regression. RESULTS Six hundred fifty-eight FMTs were performed for 617 patients in 17 centres. The overall efficacy of FMT was 84.3% (520/617), with 0.5% of severe adverse events possibly related to FMT (3/658). Forty-seven patients were treated at the first recurrence of CDI with a similar success rate (85.1%). Severe chronic kidney disease (CKD; OR: 2.18, 95%CI [1.20-3.88]), non-severe refractory CDI (OR: 15.35, [1.94-318.2]), the use of ≥ 80% glycerol (OR: 2.52, [1.11-5.67]), insufficient bowel cleansing (OR: 5.47, [1.57-20.03]) and partial FMT retention (OR: 9.97, [2.62-48.49]) were associated with CDI recurrence within 8 weeks. CONCLUSIONS Conditions of transplant manufacturing, bowel cleansing, and a route of delivery tailored to the patient's characteristics are key factors in optimising FMT efficacy. FMT at first recurrence showed high success in real-life practice, whereas it had lower efficacy in severe CDI and non-severe refractory CDI.
Collapse
Affiliation(s)
- Nicolas Benech
- French Faecal Transplant Group (GFTF)France
- Hepato‐Gastroenterology DepartmentHôpital de la Croix‐Rousse, Hospices Civils de LyonLyonFrance
- Lyon GEM Microbiota Study GroupLyonFrance
- ESGHAMI (ESCMID Study Group for Host and Microbiota Interactions)BaselSwitzerland
- Claude Bernard Lyon 1 University, CRCLLyonFrance
- ESGCD (ESCMID Study Group for Clostridioides difficile)BaselSwitzerland
| | - Nadim Cassir
- French Faecal Transplant Group (GFTF)France
- Pôle Maladies Infectieuses et Tropicales. Assistance Publique‐Hôpitaux de MarseilleMarseilleFrance
- Aix‐Marseille Université, IRD, MEPHIMarseilleFrance
| | - Laurent Alric
- French Faecal Transplant Group (GFTF)France
- Internal Medecine‐Digestive Department, Rangueil HospitalToulouse 3 UniversityFrance
| | - Frédéric Barbut
- French Faecal Transplant Group (GFTF)France
- ESGCD (ESCMID Study Group for Clostridioides difficile)BaselSwitzerland
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR S‐1139Université de Paris CitéParisFrance
- National Reference Laboratory for Clostridioides Difficile, Assistance Publique‐Hôpitaux de Paris (AP‐HP)Saint‐Antoine HospitalParisFrance
| | - Rui Batista
- French Faecal Transplant Group (GFTF)France
- Pharmacy DepartmentCochin Hospital, APHPParisFrance
| | - Alexandre Bleibtreu
- French Faecal Transplant Group (GFTF)France
- Department of Infectious Diseases, Pitié‐Salpêtrière HospitalAPHP and Sorbonne UniversitéParisFrance
- INSERM U 1135, Cimi‐ParisParisFrance
- Assistance Publique‐Hôpitaux de Paris (AP‐HP) FMT CenterParisFrance
| | - Thomas Briot
- French Faecal Transplant Group (GFTF)France
- Pharmacy DepartmentHospices Civils de Lyon, Groupement Hospitalier NordLyonFrance
| | - Benjamin Davido
- French Faecal Transplant Group (GFTF)France
- Maladies Infectieuses, Hôpital Raymond PoincaréUniversité Paris SaclayGarchesFrance
| | - Tatiana Galperine
- French Faecal Transplant Group (GFTF)France
- Service Des Maladies InfectieusesCentre Hospitalier Universitaire VaudoisLausanneSwitzerland
| | - Anne‐Christine Joly
- French Faecal Transplant Group (GFTF)France
- Assistance Publique‐Hôpitaux de Paris (AP‐HP) FMT CenterParisFrance
- Saint‐Antoine HospitalUnité de Préparation Des Transplants de Microbiote (UPTM)ParisFrance
- Paris Center for Microbiome Medicine (PaCeMM) FHUParisFrance
| | - Nathalie Kapel
- French Faecal Transplant Group (GFTF)France
- Laboratoire de Coprologie, APHP, GH Pitié‐SalpêtrièreParisFrance
- INSERM S1139, Faculté de PharmacieUniversité Paris CitéParisFrance
| | - Chloé Melchior
- French Faecal Transplant Group (GFTF)France
- Department of GastroenterologyUniv Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073, Nutrition, Inflammation and Microbiota‐Gut‐Brain Axis, CHU Rouen, CIC‐CRB 1404RouenFrance
| | - Alexis Mosca
- French Faecal Transplant Group (GFTF)France
- Service de Gastroentérologie et Nutrition PédiatriquesHôpital Robert‐Debré, APHPParisFrance
| | - Biba Nebbad
- French Faecal Transplant Group (GFTF)France
- Hôpital Henri Mondor, AP‐HPUnité de Transplantation de Microbiote FaecalCréteilFrance
| | - Bénédicte Pigneur
- French Faecal Transplant Group (GFTF)France
- Service de Gastro‐Entérologie et Nutrition Pédiatrique, Centre de Référence des Maladies Rares Digestives (MARDI), Assistance Publique—Hôpitaux de Paris, Hôpital Necker Enfants MaladesUniversité Paris CitéParisFrance
- INSERM UMR S 1139, Faculté de Pharmacie de ParisUniversité Paris CitéParisFrance
| | - Stéphane M. Schneider
- French Faecal Transplant Group (GFTF)France
- Gastroentérologie et Nutrition, CHU ArchetUniversité Côte d'AzurNiceFrance
| | - Mathieu Wasiak
- French Faecal Transplant Group (GFTF)France
- Pôle Pharmacie, Centre Hospitalier Universitaire de Clermont‐FerrandClermont‐FerrandFrance
| | - Julien Scanzi
- French Faecal Transplant Group (GFTF)France
- Centre Hospitalier de ThiersThiersFrance
- University Hospital of Clermont FerrandClermont FerrandFrance
| | - Harry Sokol
- French Faecal Transplant Group (GFTF)France
- Assistance Publique‐Hôpitaux de Paris (AP‐HP) FMT CenterParisFrance
- Paris Center for Microbiome Medicine (PaCeMM) FHUParisFrance
- Gastroenterology DepartmentINSERM, Centre de Recherche Saint‐Antoine, CRSA, AP‐HP, Saint‐Antoine Hospital, Sorbonne UniversitéParisFrance
- INRAE, AgroParisTech, Micalis InstituteUniversité Paris‐SaclayJouy‐en‐JosasFrance
| |
Collapse
|
2
|
Groenewegen B, van Lingen E, Kovynev A, van den Berg AJ, Berssenbrugge EKL, Sanders IMJG, van Prehn J, van Nood E, Goorhuis A, Kuijper EJ, Smits WK, Wiese M, Keller JJ, Ducarmon QR, Terveer EM. The presence of Clostridioides difficile in faeces before and after faecal microbiota transplantation and its relation with recurrent C. difficile infection and the gut microbiota in a Dutch cohort. Clin Microbiol Infect 2024:S1198-743X(24)00589-5. [PMID: 39662821 DOI: 10.1016/j.cmi.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
OBJECTIVES To study the presence of Clostridioides difficile in faeces of patients with recurrent C. difficile infection (rCDI) before and after faecal microbiota transplantation (FMT) and to identify risk factors for faecal C. difficile and CDI recurrence. METHODS N=83 faecal sample triads (pre-FMT (∼one day), post-FMT (∼three weeks), and a corresponding FMT donor sample), and n=22 long-term (∼1-3 years) follow-up faecal samples (LTFU) were collected from FMT-treated patients. The presence of C. difficile in faeces was assessed by enrichment broth culture and PCR (tcdB gene) and associated with patient characteristics, FMT outcome, duration of pre-FMT vancomycin, FMT donor, post-FMT antibiotic use, and faecal microbiota composition (shotgun metagenomics). RESULTS The FMT cure rate for rCDI was 92.8% (77/83), with six early CDI recurrences ( CONCLUSIONS While eradication of C. difficile is not required for clinical cure of rCDI by FMT, it is associated with reduced prevalence of early CDI recurrence, as are the full completion of pre-FMT vancomycin (at least 10 days) and avoiding post-FMT antibiotics.
Collapse
Affiliation(s)
- Bas Groenewegen
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases (LUCID) Medical Microbiology and Infection Prevention, Leiden University Medical Center (LUMC), Leiden, the Netherlands; Department of Gastroenterology and Hepatology, LUMC, Leiden, the Netherlands
| | - Emilie van Lingen
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases (LUCID) Medical Microbiology and Infection Prevention, Leiden University Medical Center (LUMC), Leiden, the Netherlands; Department of Gastroenterology and Hepatology, LUMC, Leiden, the Netherlands
| | - Artemiy Kovynev
- Center for Microbiome Analyses and Therapeutics, LUCID Research, LUMC, Leiden, the Netherlands
| | - Alexander J van den Berg
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases (LUCID) Medical Microbiology and Infection Prevention, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Eric K L Berssenbrugge
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases (LUCID) Medical Microbiology and Infection Prevention, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | | | - Joffrey van Prehn
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases (LUCID) Medical Microbiology and Infection Prevention, Leiden University Medical Center (LUMC), Leiden, the Netherlands; Center for Microbiome Analyses and Therapeutics, LUCID Research, LUMC, Leiden, the Netherlands
| | - Els van Nood
- Department of Medical Microbiology and Infectious Diseases and department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Abraham Goorhuis
- Department of Internal Medicine, Amsterdam University Medical Centers, location Academic Medical Center, Amsterdam, the Netherlands
| | - Ed J Kuijper
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases (LUCID) Medical Microbiology and Infection Prevention, Leiden University Medical Center (LUMC), Leiden, the Netherlands; Center for Microbiome Analyses and Therapeutics, LUCID Research, LUMC, Leiden, the Netherlands
| | - Wiep Klaas Smits
- Center for Microbiome Analyses and Therapeutics, LUCID Research, LUMC, Leiden, the Netherlands
| | - Maria Wiese
- Center for Microbiome Analyses and Therapeutics, LUCID Research, LUMC, Leiden, the Netherlands
| | - Josbert J Keller
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases (LUCID) Medical Microbiology and Infection Prevention, Leiden University Medical Center (LUMC), Leiden, the Netherlands; Department of Gastroenterology and Hepatology, LUMC, Leiden, the Netherlands; Department of Gastroenterology and Hepatology, Haaglanden Medical Center, Den Haag, The Netherlands
| | - Quinten R Ducarmon
- Center for Microbiome Analyses and Therapeutics, LUCID Research, LUMC, Leiden, the Netherlands
| | - Elisabeth M Terveer
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases (LUCID) Medical Microbiology and Infection Prevention, Leiden University Medical Center (LUMC), Leiden, the Netherlands; Center for Microbiome Analyses and Therapeutics, LUCID Research, LUMC, Leiden, the Netherlands.
| |
Collapse
|
3
|
Zheng H, Xu Y, Wu Y, Huangfu X, Chen W, He K, Yang Y. Effects of Three Modification Methods on the In Vitro Gastrointestinal Digestion and Colonic Fermentation of Dietary Fiber from Lotus Leaves. Foods 2024; 13:3768. [PMID: 39682840 DOI: 10.3390/foods13233768] [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: 10/10/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
Shear emulsifying (SE), ball milling (BM), and autoclave treatment (AT) were utilized for the modification of lotus leaves, and the effects on in vitro gastrointestinal digestion and colonic fermentation of insoluble dietary fiber (IDF) from lotus leaves were compared. Compared with SEIDF and ATIDF, BMIDF released more polyphenols and exhibited better antioxidant capacity during in vitro gastrointestinal digestion. The IDF of lotus leaves changed the gut microbiota composition during in vitro colonic fermentation. SEIDF was beneficial to the diversity of gut microbiota compared with BMIDF and ATIDF. Among the three IDF groups of lotus leaves, six significant differences of OTUs were all in ATIDF; however there was the highest relative abundance of Escherichia-Shigella in ATIDF. In addition, the concentrations of butyric acid and valeric acid produced by SEIDF were significantly higher than that of BMIDF and ATIDF. Overall, SE modification improved the colonic fermentation characteristics of IDFs in lotus leaves more effectively; while BM modification helped to promote the release of polyphenols from IDFs in lotus leaves during in vitro gastrointestinal digestion. The research lays the foundation for the application of the dietary fiber of lotus leaves as a premium fiber additive in functional food.
Collapse
Affiliation(s)
- Hui Zheng
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yao Xu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yuhang Wu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xuantong Huangfu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Wenxiu Chen
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Kai He
- School of Pharmaceutical Science, Hunan University of Medicine, Huaihua 418000, China
| | - Yong Yang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
4
|
Liu J, Zhang S, Weng H. Effects of Clostridium butyricum and inulin supplementation on intestinal microbial composition in high-fat diet fed mice. Food Funct 2024; 15:10870-10884. [PMID: 39415545 DOI: 10.1039/d4fo02963a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Obesity has become a serious epidemic problem in the world, and probiotics and prebiotics have been used to treat obesity. The effectiveness of diet therapy such as Clostridium butyricum (CB) and inulin supplementation in obesity and whether they can cooperate to produce better effects are still unclear. And during this process, intestinal flora play an important role, while the bacteria involved and the metabolic mechanism need to be explored. In this study, we successfully established a mouse obesity model with a high-fat diet (HFD) and divided it into three experimental groups: 7% CB (CB7), 7% CB + 1% inulin (C7G1), and 10% CB + 1% inulin (C10G). Dietary supplementation with CB and inulin could improve the glucose tolerance and intestinal microbial composition of obese mice, among which the simultaneous supplementation with 7% CB and 1% inulin (C7G1) has the most significant effect on obese mice fed with a HFD. It could significantly reduce the amount of total cholesterol, triglyceride, and low-density lipoprotein, improve abnormal glucose tolerance, and reduce abnormal blood glucose in obese mice. The intestinal flora of obese mice changed significantly, among which Lachnospiraceae_unclassified, Porphyromonaceae_unclassified, Olsenella, Bacteria_unclassified and Clostridiales_unclassified decreased due to the HFD, while Megamonas and Clostridium XIVa increased. After the supplementation with CB and inulin, the enrichment of three kinds of beneficial bacteria, Parabacteroides, Bacteroides, and Ruminococcaceae unclassified increased. The high-fat diet could upregulate the expression of FGF21, and the Clostridium butyricum and inulin supplemented diet could decrease the upregulation.
Collapse
Affiliation(s)
- Jing Liu
- Department of Research, Shanghai University of Medicine and Health Sciences Affliated Zhoupu Hospital, The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China.
| | - Suhua Zhang
- Department of Research, Shanghai University of Medicine and Health Sciences Affliated Zhoupu Hospital, The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China.
| | - Huachun Weng
- Department of Research, Shanghai University of Medicine and Health Sciences Affliated Zhoupu Hospital, The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China.
| |
Collapse
|
5
|
Zhao Z, Chen J, Zhao D, Chen B, Wang Q, Li Y, Chen J, Bai C, Guo X, Hu N, Zhang B, Zhao R, Yuan J. Microbial biomarker discovery in Parkinson's disease through a network-based approach. NPJ Parkinsons Dis 2024; 10:203. [PMID: 39461950 PMCID: PMC11513973 DOI: 10.1038/s41531-024-00802-2] [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: 12/13/2023] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Associations between the gut microbiota and Parkinson's disease (PD) have been widely investigated. However, the replicable biomarkers for PD diagnosis across multiple populations remain elusive. Herein, we performed a meta-analysis to investigate the pivotal role of the gut microbiome in PD and its potential diagnostic implications. Six 16S rRNA gene amplicon sequence datasets from five independent studies were integrated, encompassing 550 PD and 456 healthy control samples. The analysis revealed significant alterations in microbial composition and alpha and beta diversity, emphasizing altered gut microbiota in PD. Specific microbial taxa, including Faecalibacterium, Roseburia, and Coprococcus_2, known as butyrate producers, were notably diminished in PD, potentially contributing to intestinal inflammation. Conversely, genera such as Akkermansia and Bilophila exhibited increased relative abundances. A network-based algorithm called NetMoss was utilized to identify potential biomarkers of PD. Afterwards, a classification model incorporating 11 optimized genera demonstrated high performance. Further functional analyses indicated enrichment in pathways related to neurodegeneration and metabolic pathways. These findings illuminate the intricate relationship between the gut microbiota and PD, offering insights into potential therapeutic interventions and personalized diagnostic strategies.
Collapse
Affiliation(s)
- Zhe Zhao
- Department of Pharmacy, Peking University Third Hospital, 100191, Beijing, China
- Institute for Drug Evaluation, Peking University Health Science Center, 100191, Beijing, China
| | - Jing Chen
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Danhua Zhao
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Baoyu Chen
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Qi Wang
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Yuan Li
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Junyi Chen
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Chaobo Bai
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Xintong Guo
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Nan Hu
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
- First Affiliated Hospital of Dalian Medical University, No. 222, Zhongshan Road, Dalian, 116011, Liaoning, China
| | - Bingwei Zhang
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
- First Affiliated Hospital of Dalian Medical University, No. 222, Zhongshan Road, Dalian, 116011, Liaoning, China
| | - Rongsheng Zhao
- Department of Pharmacy, Peking University Third Hospital, 100191, Beijing, China.
- Institute for Drug Evaluation, Peking University Health Science Center, 100191, Beijing, China.
| | - Junliang Yuan
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China.
| |
Collapse
|
6
|
Spigaglia P. Clostridioides difficile and Gut Microbiota: From Colonization to Infection and Treatment. Pathogens 2024; 13:646. [PMID: 39204246 PMCID: PMC11357127 DOI: 10.3390/pathogens13080646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
Clostridioides difficile is the main causative agent of antibiotic-associated diarrhea (AAD) in hospitals in the developed world. Both infected patients and asymptomatic colonized individuals represent important transmission sources of C. difficile. C. difficile infection (CDI) shows a large range of symptoms, from mild diarrhea to severe manifestations such as pseudomembranous colitis. Epidemiological changes in CDIs have been observed in the last two decades, with the emergence of highly virulent types and more numerous and severe CDI cases in the community. C. difficile interacts with the gut microbiota throughout its entire life cycle, and the C. difficile's role as colonizer or invader largely depends on alterations in the gut microbiota, which C. difficile itself can promote and maintain. The restoration of the gut microbiota to a healthy state is considered potentially effective for the prevention and treatment of CDI. Besides a fecal microbiota transplantation (FMT), many other approaches to re-establishing intestinal eubiosis are currently under investigation. This review aims to explore current data on C. difficile and gut microbiota changes in colonized individuals and infected patients with a consideration of the recent emergence of highly virulent C. difficile types, with an overview of the microbial interventions used to restore the human gut microbiota.
Collapse
Affiliation(s)
- Patrizia Spigaglia
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Roma, Italy
| |
Collapse
|
7
|
Zhang Q, Bi Y, Zhang B, Jiang Q, Mou CK, Lei L, Deng Y, Li Y, Yu J, Liu W, Zhao J. Current landscape of fecal microbiota transplantation in treating depression. Front Immunol 2024; 15:1416961. [PMID: 38983862 PMCID: PMC11231080 DOI: 10.3389/fimmu.2024.1416961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
Depression, projected to be the predominant contributor to the global disease burden, is a complex condition with diverse symptoms including mood disturbances and cognitive impairments. Traditional treatments such as medication and psychotherapy often fall short, prompting the pursuit of alternative interventions. Recent research has highlighted the significant role of gut microbiota in mental health, influencing emotional and neural regulation. Fecal microbiota transplantation (FMT), the infusion of fecal matter from a healthy donor into the gut of a patient, emerges as a promising strategy to ameliorate depressive symptoms by restoring gut microbial balance. The microbial-gut-brain (MGB) axis represents a critical pathway through which to potentially rectify dysbiosis and modulate neuropsychiatric outcomes. Preclinical studies reveal that FMT can enhance neurochemicals and reduce inflammatory markers, thereby alleviating depressive behaviors. Moreover, FMT has shown promise in clinical settings, improving gastrointestinal symptoms and overall quality of life in patients with depression. The review highlights the role of the gut-brain axis in depression and the need for further research to validate the long-term safety and efficacy of FMT, identify specific therapeutic microbial strains, and develop targeted microbial modulation strategies. Advancing our understanding of FMT could revolutionize depression treatment, shifting the paradigm toward microbiome-targeting therapies.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Xianning Medical College, Hubei University of Science & Technology, Xianning, Hubei, China
| | - Yajun Bi
- Department of Pediatrics, Dalian Municipal Women and Children’s Medical Center (Group), Dalian Medical University, Dalian, Liaoning, China
| | - Boyu Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiong Jiang
- Xianning Medical College, Hubei University of Science & Technology, Xianning, Hubei, China
| | - Chao Kam Mou
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lelin Lei
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yibo Deng
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yutong Li
- Wuhan Britain-China School, Wuhan, Hubei, China
| | - Jing Yu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Liu
- Department of Public Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinzhu Zhao
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
8
|
Yan L, Wang X, Yu T, Qi Z, Li H, Nan H, Wang K, Luo D, Hua F, Wang W. Characteristics of the gut microbiota and serum metabolites in postmenopausal women with reduced bone mineral density. Front Cell Infect Microbiol 2024; 14:1367325. [PMID: 38912210 PMCID: PMC11190063 DOI: 10.3389/fcimb.2024.1367325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/29/2024] [Indexed: 06/25/2024] Open
Abstract
Introduction Emerging evidence suggests that the gut microbiota is closely associated with bone homeostasis. However, little is known about the relationships among the bone mineral density (BMD) index, bone turnover markers, and the gut microbiota and its metabolites in postmenopausal women. Methods In this study, to understand gut microbiota signatures and serum metabolite changes in postmenopausal women with reduced BMD, postmenopausal individuals with normal or reduced BMD were recruited and divided into normal and OS groups. Feces and serum samples were collected for 16S rRNA gene sequencing, liquid chromatography coupled with mass spectrometry (LC-MS)-based metabolomics and integrated analysis. Results The results demonstrated that bacterial richness and diversity were greater in the OS group than in the normal group. Additionally, distinguishing bacteria were found among the two groups and were closely associated with the BMD index and bone turnover markers. Metabolomic analysis revealed that the expression of serum metabolites, such as etiocholanolone, testosterone sulfate, and indole-3-pyruvic acid, and the corresponding signaling pathways, especially those involved in tryptophan metabolism, fatty acid degradation and steroid hormone biosynthesis, also changed significantly. Correlation analysis revealed positive associations between normal group-enriched Bacteroides abundance and normal group-enriched etiocholanolone and testosterone sulfate abundances; in particular, Bacteroides correlated positively with BMD. Importantly, the tryptophan-indole metabolism pathway was uniquely metabolized by the gut bacteria-derived tnaA gene, the predicted abundance of which was significantly greater in the normal group than in the control group, and the abundance of Bacteroides was strongly correlated with the tnaA gene. Discussion Our results indicated a clear difference in the gut microbiota and serum metabolites of postmenopausal women. Specifically altered bacteria and derived metabolites were closely associated with the BMD index and bone turnover markers, indicating the potential of the gut microbiota and serum metabolites as modifiable factors and therapeutic targets for preventing osteoporosis.
Collapse
Affiliation(s)
- Litao Yan
- Department of Articular Orthopaedics, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xianfeng Wang
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, China
| | - Tiantian Yu
- Department of Gynaecology and Obstetrics, Dalian Municipal Woman and Children’s Medical Center, Dalian, China
| | - Zhiming Qi
- Department of Articular Orthopaedics, The Second People’s Hospital of Dalian, Dalian, China
| | - Huan Li
- Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Hao Nan
- Department of Articular Orthopaedics, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Kun Wang
- Department of Articular Orthopaedics, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Di Luo
- Department of Clinical Laboratory, The Second People’s Hospital of Dalian, Dalian, China
| | - Fei Hua
- Department of Endocrinology and Metabolism, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wendong Wang
- Department of Articular Orthopaedics, The Second People’s Hospital of Dalian, Dalian, China
| |
Collapse
|
9
|
Zhang R, Bai D, Zhen W, Hu X, Zhang H, Zhong J, Zhang Y, Ito K, Zhang B, Yang Y, Li J, Ma Y. Aspirin eugenol ester affects ileal barrier function, inflammatory response and microbiota in broilers under lipopolysaccharide-induced immune stress conditions. Front Vet Sci 2024; 11:1401909. [PMID: 38872795 PMCID: PMC11169880 DOI: 10.3389/fvets.2024.1401909] [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: 03/16/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024] Open
Abstract
Aims The aim of this study was to investigate the effects of aspirin eugenol ester (AEE) on ileal immune function in broilers under lipopolysaccharide (LPS)-induced immune stress. Methods Two hundred and forty one-day-old male Arbor Acres chicks were randomly divided into four groups (saline, LPS, saline + AEE and LPS + AEE) with six replicates of ten broilers each. The saline group and LPS group were fed the normal diet, while the other two groups received normal diet plus 0.1 g/kg AEE. Broilers in the LPS and LPS + AEE groups were injected intraperitoneally with 0.5 mg/kg B.W LPS in saline for seven consecutive days beginning at 14 days of age, while broilers in the saline and saline + AEE groups were injected with saline only. Results The results showed that AEE improved the ileal morphology and increased the ratio of villus height to crypt depth of immune-stressed broilers. LPS-induced immune stress significantly reduced the expression of the genes for the tight junction proteins occludin, zonula occludens-1 (ZO-1), claudin-1 and claudin-2, in the ileum, while AEE significantly up-regulated the expression of these genes. Compared with the saline group, the LPS-treated chickens showed significantly increased mRNA expression of the inflammatory factors tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-10 (IL-10), cyclooxygenase-2 (COX-2), and microsomal Prostaglandin E Synthesase-1 (mPGES-1) in the ileum, while they were significantly decreased by AEE supplementation. In addition, analysis of the ileal bacterial composition showed that compared with saline and LPS + AEE groups, the proportion of Firmicutes and Lactobacillus in the LPS group was lower, while the proportion of Proteobacteria and Escherichia-Shigella was higher. Similarly, Line Discriminant Analysis Effect Size (LEfSe) analysis showed that compared with the LPS group, Brevibacillus was dominant in the saline group, while the LPS + AEE group was rich in Rhizobium, Lachnoclostridium, Ruminococcaceae, Faecalibacterium, Negativibacillus, Oscillospiraceae, and Flavonifractor. Conclusion These results indicate that dietary supplementation with 0.1 g/kg AEE could protect the intestinal health by improving the intestinal villus morphology, enhancing the expression of tight junction genes and alleviating inflammation to resist the immune stress caused by LPS stimulation in broilers, and the mechanism may involve COX-2-related signal transduction and improved intestinal microbiota composition.
Collapse
Affiliation(s)
- Ruilin Zhang
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Dongying Bai
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Wenrui Zhen
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Xiaodi Hu
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Haojie Zhang
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Jiale Zhong
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Yi Zhang
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Koichi Ito
- Department of Food and Physiological Models, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Ibaraki, Japan
| | - Bingkun Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yajun Yang
- Key Lab of New Animal Drug of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jianyong Li
- Key Lab of New Animal Drug of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yanbo Ma
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Longmen Laboratory, Science and Technology Innovation Center for Completed Set Equipment, Luoyang, China
| |
Collapse
|
10
|
DuPont HL, DuPont AW, Tillotson GS. Microbiota restoration therapies for recurrent Clostridioides difficile infection reach an important new milestone. Therap Adv Gastroenterol 2024; 17:17562848241253089. [PMID: 38800353 PMCID: PMC11119484 DOI: 10.1177/17562848241253089] [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: 03/03/2024] [Accepted: 04/18/2024] [Indexed: 05/29/2024] Open
Abstract
Microbiota restoration therapy has become a standard treatment for recurrent Clostridioides difficile infection (rCDI). In this article, we review the studies supporting the licensure of two live biotherapeutic products (LBPs) designed to prevent rCDI and to provide clinicians with a perspective on their differences. PubMed was reviewed on 1 October 2023, for all papers published concerning the current Food and Drug Administration allowance of the use of fecal microbiota transplantation (FMT) and the studies that led to the licensure of RBX2660 (REBYOTA™), generic name, fecal microbiota, live-jslm, and SER-109 (VOWST™), generic name, fecal microbiota spores, live-brpk. OpenBiome continues to produce fecal products for patients with rCDI at their treatment sites, and the American Gastroenterology Association has a National Registry focused on long-term safety of administering fecal microbiota products. The science behind the licensing of fecal microbiota, live-jslm, a consortium of fecal anaerobes found in stool augmented with strains of Bacteroidetes and fecal microbiota spores, live-brpk, a mixture of 50 species of purified Firmicutes spores is reviewed. Both products appear to be safe in clinical trials and effective in reducing rCDI episodes by mechanisms established for FMT, including normalization of α- and β-diversity of the microbiome and by increasing fecal secondary bile acids. The different makeup of the two LBPs suggests that rCDI responds to a variety of engrafting microbiota which explains why nearly all donors in FMT of rCDI are generally effective. Fecal microbiota, live-jslm has also been shown to successfully treat rCDI in elderly patients with advanced comorbidities. With the licensure of two novel LBPs, we are entering a new phase of microbiota replacement therapy. Having standardized manufacturing and proper monitoring of products, harnessing the microbiome to control and prevent disease has a new beginning.
Collapse
Affiliation(s)
- Herbert L. DuPont
- Infectious Diseases and Epidemiology, Department of Epidemiology, University of Texas School of Public Health, 1200 Pressler Street, Houston, TX 77030, USA
- Department of Internal Medicine, University of Texas McGovern Medical School, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Kelsey Research Foundation, Houston, TX, USA
| | - Andrew W. DuPont
- Division of Gastroenterology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | | |
Collapse
|
11
|
Zhang R, Li X, Zhang S. The Role of Bacteria in Central Nervous System Tumors: Opportunities and Challenges. Microorganisms 2024; 12:1053. [PMID: 38930435 PMCID: PMC11205425 DOI: 10.3390/microorganisms12061053] [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: 05/05/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Tumors of the central nervous system (CNS) are severe and refractory diseases with poor prognosis, especially for patients with malignant glioblastoma and brain metastases. Currently, numerous studies have explored the potential role of bacteria and intestinal flora in tumor development and treatment. Bacteria can penetrate the blood-brain barrier (BBB), targeting the hypoxic microenvironment at the core of tumors, thereby eliminating tumors and activating both the innate and adaptive immune responses, rendering them promising therapeutic agents for CNS tumors. In addition, engineered bacteria and derivatives, such as bacterial membrane proteins and bacterial spores, can also be used as good candidate carriers for targeted drug delivery. Moreover, the intestinal flora can regulate CNS tumor metabolism and influence the immune microenvironment through the "gut-brain axis". Therefore, bacterial anti-tumor therapy, engineered bacterial targeted drug delivery, and intervention of the intestinal flora provide therapeutic modalities for the treatment of CNS tumors. In this paper, we performed a comprehensive review of the mechanisms and therapeutic practices of bacterial therapy for CNS tumors and discussed potential future research directions in this field.
Collapse
Affiliation(s)
| | | | - Si Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China; (R.Z.); (X.L.)
| |
Collapse
|
12
|
Reygner J, Delannoy J, Barba-Goudiaby MT, Gasc C, Levast B, Gaschet E, Ferraris L, Paul S, Kapel N, Waligora-Dupriet AJ, Barbut F, Thomas M, Schwintner C, Laperrousaz B, Corvaïa N. Reduction of product composition variability using pooled microbiome ecosystem therapy and consequence in two infectious murine models. Appl Environ Microbiol 2024; 90:e0001624. [PMID: 38651930 PMCID: PMC11107171 DOI: 10.1128/aem.00016-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024] Open
Abstract
Growing evidence demonstrates the key role of the gut microbiota in human health and disease. The recent success of microbiotherapy products to treat recurrent Clostridioides difficile infection has shed light on its potential in conditions associated with gut dysbiosis, such as acute graft-versus-host disease, intestinal bowel diseases, neurodegenerative diseases, or even cancer. However, the difficulty in defining a "good" donor as well as the intrinsic variability of donor-derived products' taxonomic composition limits the translatability and reproducibility of these studies. Thus, the pooling of donors' feces has been proposed to homogenize product composition and achieve higher taxonomic richness and diversity. In this study, we compared the metagenomic profile of pooled products to corresponding single donor-derived products. We demonstrated that pooled products are more homogeneous, diverse, and enriched in beneficial bacteria known to produce anti-inflammatory short chain fatty acids compared to single donor-derived products. We then evaluated pooled products' efficacy compared to corresponding single donor-derived products in Salmonella and C. difficile infectious mouse models. We were able to demonstrate that pooled products decreased pathogenicity by inducing a structural change in the intestinal microbiota composition. Single donor-derived product efficacy was variable, with some products failing to control disease progression. We further performed in vitro growth inhibition assays of two extremely drug-resistant bacteria, Enterococcus faecium vanA and Klebsiella pneumoniae oxa48, supporting the use of pooled microbiotherapies. Altogether, these results demonstrate that the heterogeneity of donor-derived products is corrected by pooled fecal microbiotherapies in several infectious preclinical models.IMPORTANCEGrowing evidence demonstrates the key role of the gut microbiota in human health and disease. Recent Food and Drug Administration approval of fecal microbiotherapy products to treat recurrent Clostridioides difficile infection has shed light on their potential to treat pathological conditions associated with gut dysbiosis. In this study, we combined metagenomic analysis with in vitro and in vivo studies to compare the efficacy of pooled microbiotherapy products to corresponding single donor-derived products. We demonstrate that pooled products are more homogeneous, diverse, and enriched in beneficial bacteria compared to single donor-derived products. We further reveal that pooled products decreased Salmonella and Clostridioides difficile pathogenicity in mice, while single donor-derived product efficacy was variable, with some products failing to control disease progression. Altogether, these findings support the development of pooled microbiotherapies to overcome donor-dependent treatment efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Stéphane Paul
- Team GIMAP, Centre International de Recherche en Infectiologie, Université Jean Monnet, Saint-Etienne, France
- Inserm, Université Claude Bernard Lyon, Lyon, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
- Immunology Department, iBiothera Reference Center, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Nathalie Kapel
- UMR-S 1139, INSERM, Université Paris Cite, Paris, France
- Service de Coprologie fonctionnelle, Hôpital de la Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France
| | | | - Frederic Barbut
- UMR-S 1139, INSERM, Université Paris Cite, Paris, France
- National Reference Laboratory for Clostridioides difficile, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- The European Society of Clinical Microbiology and Infectious Diseases Study Group for Clostridioides difficile, Basel, Switzerland
| | - Muriel Thomas
- UMR1319, Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | | | | | | |
Collapse
|
13
|
Li J, Yan K, Wang S, Wang P, Jiao J, Dong Y. Alteration of the intestinal microbiota and serum metabolites in a mouse model of Pon1 gene ablation. FASEB J 2024; 38:e23611. [PMID: 38597925 DOI: 10.1096/fj.202302344r] [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: 11/13/2023] [Revised: 03/15/2024] [Accepted: 04/01/2024] [Indexed: 04/11/2024]
Abstract
Mutations in the Paraoxonase 1 (Pon1) gene underlie aging, cardiovascular disease, and impairments of the nervous and gastrointestinal systems and are linked to the intestinal microbiome. The potential role of Pon1 in modulating the intestinal microbiota and serum metabolites is poorly understood. The present study demonstrated that mice with genomic excision of Pon1 by a multiplexed guide RNA CRISPR/Cas9 approach exhibited disrupted gut microbiota, such as significantly depressed alpha-diversity and distinctly separated beta diversity, accompanied by varied profiles of circulating metabolites. Furthermore, genomic knock in of Pon1 exerted a distinct effect on the intestinal microbiome and serum metabolome, including dramatically enriched Aerococcus, linoleic acid and depleted Bacillus, indolelactic acid. Specifically, a strong correlation was established between bacterial alterations and metabolites in Pon1 knockout mice. In addition, we identified metabolites related to gut bacteria in response to Pon1 knock in. Thus, the deletion of Pon1 affects the gut microbiome and functionally modifies serum metabolism, which can lead to dysbiosis, metabolic dysfunction, and infection risk. Together, these findings put forth a role for Pon1 in microbial alterations that contribute to metabolism variations. The function of Pon1 in diseases might at least partially depend on the microbiome.
Collapse
Affiliation(s)
- Jing Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Kaixin Yan
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Siyuan Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Pan Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jie Jiao
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ying Dong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Ortiz-Alvarez de la Campa M, Curtis-Joseph N, Beekman C, Belenky P. Gut Biogeography Accentuates Sex-Related Differences in the Murine Microbiome. Microorganisms 2024; 12:221. [PMID: 38276206 PMCID: PMC10821414 DOI: 10.3390/microorganisms12010221] [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: 12/13/2023] [Revised: 01/05/2024] [Accepted: 01/15/2024] [Indexed: 01/27/2024] Open
Abstract
Recent studies have highlighted the influence of factors such as sex and sex-linked hormones on microbiome composition, raising concerns about the generalizability of findings. Here, we explore whether gut geography, specifically the upper and lower gastrointestinal tract (GI), contributes to sex-linked microbiome differences in mice. We collected microbial samples throughout the length of the GI from male and female C57B6/J mice at 6- and 8-weeks old, and conducted 16S rRNA sequencing. Our findings revealed significant sex-related differences, with Clostridium_sensu_stricto_1 more abundant in the male colon, while females exhibited higher levels of Dubosiella newyorkensis across all organs at 6 weeks. We also observed decreased Shannon alpha diversity in the small intestine compared to the lower GI, and this diversity decreased further at 8 weeks. Interestingly, our results suggest that age mitigates sex-related, but not gut geography-related differences in beta diversity, with implications for experimental outcomes and treatment strategies. This study underscores the dynamic nature of microbial diversity, influenced by sex, age, and GI localization, emphasizing the need for a more comprehensive understanding of microbiome dynamics in experimental research and clinical interventions.
Collapse
Affiliation(s)
| | - Noelle Curtis-Joseph
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Chapman Beekman
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| |
Collapse
|
15
|
Sayol-Altarriba A, Aira A, Villasante A, Albarracín R, Faneca J, Casals G, Villanueva-Cañas JL, Casals-Pascual C. Normalization of short-chain fatty acid concentration by bacterial count of stool samples improves discrimination between eubiotic and dysbiotic gut microbiota caused by Clostridioides difficile infection. Gut Microbes 2024; 16:2415488. [PMID: 39395000 PMCID: PMC11485779 DOI: 10.1080/19490976.2024.2415488] [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: 08/01/2024] [Revised: 09/23/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024] Open
Abstract
Short-chain fatty acids (SCFAs) represent a cornerstone of gut health, serving as critical mediators of immune modulation and overall host homeostasis. Patients with dysbiosis caused by Clostridioides difficile infection (CDI) typically exhibit lower SCFAs levels compared to healthy stool donors and, thus, the concentration of SCFAs has been proposed as a proxy marker of a healthy microbiota. However, there is no consistency in the methods used to quantify SCFAs in stool samples and usually, the results are normalized by the weight of the stool samples, which does not address differences in water and fiber content and ignores bacterial counts in the sample (the main component of stool that contributes to the composition of these metabolites in the sample). Here, we show that normalized SCFAs concentrations by the bacterial count improve discrimination between healthy and dysbiotic samples (patients with CDI), particularly when using acetate and propionate levels. After normalization, butyrate is the metabolite that best discriminates eubiotic and dysbiotic samples according to the area under the receiver operating characteristic (ROC) curve (AUC-ROC = 0.860, [95% CI: 0.786-0.934], p < .0001).
Collapse
Affiliation(s)
- Anna Sayol-Altarriba
- Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain
- ISGlobal, Barcelona, Spain
- Department of Clinical Microbiology, Centre for Biomedical Diagnosis, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Andrea Aira
- ISGlobal, Barcelona, Spain
- Department of Clinical Microbiology, Centre for Biomedical Diagnosis, Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBERINFEC), Barcelona, Spain
| | - Anna Villasante
- Department of Clinical Microbiology, Centre for Biomedical Diagnosis, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Rosa Albarracín
- Department of Clinical Microbiology, Centre for Biomedical Diagnosis, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Joana Faneca
- Department of Biochemistry and Molecular Genetics, Centre for Biomedical Diagnosis, Hospital Clínic de Barcelona, IDIBAPS, Barcelona, Spain
| | - Gregori Casals
- Department of Biochemistry and Molecular Genetics, Centre for Biomedical Diagnosis, Hospital Clínic de Barcelona, IDIBAPS, Barcelona, Spain
| | | | - Climent Casals-Pascual
- Faculty of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain
- ISGlobal, Barcelona, Spain
- Department of Clinical Microbiology, Centre for Biomedical Diagnosis, Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
16
|
Cui L, Hua Y, Zou S, Gu C, Li H. Combination of fenchone and sodium hyaluronate ameliorated constipation-predominant irritable bowel syndrome and underlying mechanisms. Chem Biol Drug Des 2024; 103:e14397. [PMID: 38030381 DOI: 10.1111/cbdd.14397] [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: 08/23/2023] [Revised: 10/15/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023]
Abstract
We sought to explore the protective effect of the combination of fenchone (FE) and sodium hyaluronate (SH) on ice water-induced IBS-C rats and the potential mechanism. The neurotransmitter levels, including substance P (SP), motilin (MTL), 5-hydroxytryptamine (5-HT), and vasoactive intestinal peptide (VIP), were determined by ELISA methods. The stem cell factors (SCF)/c-Kit signaling pathway-related protein and mRNA levels were determined by western blot and reverse transcription quantitative polymerase chain reaction (RT-qPCR) analyses, respectively. The expressions of tight ZO-1, Occludin, and Claudin-1 were also measured by western blot assay and immunofluorescence staining. The 16S rRNA gene sequence was used to measure the composition of gut microbiota. The co-administration of FE and SH improved the body weight, number of fecal pellets, fecal moisture, abdominal with drawal reflex score, and gastrointestinal transit rate in IBS-C rats. The unique efficacy of combination depended on the regulation of balance between excitatory and inhibitory neurotransmitters, enhancement of intestinal barrier function, and activation of SCF/c-Kit pathway. The gut microbiota structure was also restored. The ability of FE combined with SH to regulate SCF/c-Kit signaling pathway, enhance intestinal barrier function, and modulate gut microbiota contributes to their efficacy in managing IBS-C in rats.
Collapse
Affiliation(s)
- Li Cui
- Department of Gastroenterology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yongzhi Hua
- Department of Gastroenterology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Digestive Department, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Shuting Zou
- Department of Gastroenterology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chao Gu
- Department of Gastroenterology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hui Li
- Department of Gastroenterology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
17
|
Sun XW, Huang HJ, Wang XM, Wei RQ, Niu HY, Chen HY, Luo M, Abdugheni R, Wang YL, Liu FL, Jiang H, Liu C, Liu SJ. Christensenella strain resources, genomic/metabolomic profiling, and association with host at species level. Gut Microbes 2024; 16:2347725. [PMID: 38722028 PMCID: PMC11085954 DOI: 10.1080/19490976.2024.2347725] [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: 01/03/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
The gut commensal bacteria Christensenellaceae species are negatively associated with many metabolic diseases, and have been seen as promising next-generation probiotics. However, the cultured Christensenellaceae strain resources were limited, and their beneficial mechanisms for improving metabolic diseases have yet to be explored. In this study, we developed a method that enabled the enrichment and cultivation of Christensenellaceae strains from fecal samples. Using this method, a collection of Christensenellaceae Gut Microbial Biobank (ChrisGMB) was established, composed of 87 strains and genomes that represent 14 species of 8 genera. Seven species were first described and the cultured Christensenellaceae resources have been significantly expanded at species and strain levels. Christensenella strains exerted different abilities in utilization of various complex polysaccharides and other carbon sources, exhibited host-adaptation capabilities such as acid tolerance and bile tolerance, produced a wide range of volatile probiotic metabolites and secondary bile acids. Cohort analyses demonstrated that Christensenellaceae and Christensenella were prevalent in various cohorts and the abundances were significantly reduced in T2D and OB cohorts. At species level, Christensenellaceae showed different changes among healthy and disease cohorts. C. faecalis, F. tenuis, L. tenuis, and Guo. tenuis significantly reduced in all the metabolic disease cohorts. The relative abundances of C. minuta, C. hongkongensis and C. massiliensis showed no significant change in NAFLD and ACVD. and C. tenuis and C. acetigenes showed no significant change in ACVD, and Q. tenuis and Geh. tenuis showed no significant change in NAFLD, when compared with the HC cohort. So far as we know, this is the largest collection of cultured resource and first exploration of Christensenellaceae prevalences and abundances at species level.
Collapse
Affiliation(s)
- Xin-Wei Sun
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Hao-Jie Huang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Xiao-Meng Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Rui-Qi Wei
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Han-Yu Niu
- College of Veterinary Medicine, Shanxi Agr icultural University, Taigu, China
| | - Hao-Yu Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Man Luo
- College of Veterinary Medicine, Shanxi Agr icultural University, Taigu, China
| | - Rashidin Abdugheni
- State Key Laboratory of Desert and Oasis Ecology, Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Ürűmqi, P. R. China
| | - Yu-Lin Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Feng-Lan Liu
- College of Life Sciences, Hebei University, Baoding, P. R. China
| | - He Jiang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Chang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
| |
Collapse
|