1
|
Cruz Mosquera FE, Perlaza CL, Naranjo Rojas A, Murillo Rios S, Carrero Gallego A, Fischersworring SI, Rodríguez JS, Liscano Y. Effectiveness of Probiotics, Prebiotics, and Symbiotic Supplementation in Cystic Fibrosis Patients: A Systematic Review and Meta-Analysis of Clinical Trials. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:489. [PMID: 40142300 PMCID: PMC11944062 DOI: 10.3390/medicina61030489] [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] [Received: 02/11/2025] [Revised: 03/02/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025]
Abstract
Background and Objectives: Cystic fibrosis (CF), caused by CFTR gene mutations, primarily affects the respiratory and gastrointestinal systems. Microbiota modulation through probiotics, prebiotics, or synbiotics may help restore microbial diversity and reduce inflammation. This study aimed to evaluate their efficacy in CF. Materials and Methods: A systematic review and meta-analysis of randomized controlled trials (RCTs) published between 2000 and 2024 was conducted in Cochrane, ScienceDirect, Web of Science, LILAC, BMC, PubMed, and SCOPUS following PRISMA guidelines. Methodological quality was assessed using the Jadad scale, and RevMan 5.4® estimated effects on pulmonary function (FEV1), exacerbations, hospitalizations, quality of life, and inflammatory markers. Results: Thirteen RCTs (n = 552), mostly in pediatric populations, were included. Most examined probiotics (e.g., Lactobacillus rhamnosus GG, L. reuteri), while four used synbiotics. Several studies reported reduced fecal calprotectin and proinflammatory interleukins (e.g., IL-6, IL-8), suggesting an anti-inflammatory effect. However, no significant differences were observed regarding hospitalizations or quality of life. Additionally, none of the studies documented serious adverse events associated with the intervention. The meta-analysis showed no significant decrease in exacerbations (RR = 0.81; 95% CI = 0.48-1.37; p = 0.43) or improvements in FEV1 (MD = 4.7; 95% CI = -5.4 to 14.8; p = 0.37), even in subgroup analyses. Sensitivity analyses did not modify the effect of the intervention on pulmonary function or exacerbation frequency, supporting the robustness of the findings. Conclusions: Current evidence suggests that probiotics or synbiotics yield inconsistent clinical benefits in CF, although some reduction in inflammatory markers may occur. Larger, multicenter RCTs with longer follow-up are needed for clearer conclusions. Until more definitive evidence is available, these supplements should be considered experimental adjuncts rather than standard interventions for CF management.
Collapse
Affiliation(s)
- Freiser Eceomo Cruz Mosquera
- Grupo de Investigación en Salud Integral (GISI), Department of Health Sciences Faculty, Universidad Santiago de Cali, Cali 760035, Colombia; (C.L.P.); (A.N.R.); (S.M.R.); (A.C.G.); (S.I.F.); (J.S.R.); (Y.L.)
| | | | | | | | | | | | | | | |
Collapse
|
2
|
Hayden HS, Nelson MT, Ross SE, Verster AJ, Bouzek DC, Eng A, Waalkes A, Penewit K, Kopp BT, Siracusa C, Rock MJ, Salipante SJ, Hoffman LR, Sanders DB. Effects of Therapeutic Antibiotic Exposure on the Oropharyngeal and Fecal Microbiota in Infants With Cystic Fibrosis. Pediatr Pulmonol 2025; 60:e71024. [PMID: 40042126 PMCID: PMC11881217 DOI: 10.1002/ppul.71024] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 01/11/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND Systemic antibiotics can impact all microbes inhabiting patients, regardless of the intended target organism(s). We studied the simultaneous effects on respiratory and fecal microbiomes of β-lactam antibiotics administered for respiratory symptoms in infants with cystic fibrosis (IWCF). OBJECTIVE To compare the magnitude and duration of intended (respiratory) and unintended (fecal) antimicrobial action by analyzing oropharyngeal (OP) and fecal microbiota in IWCF. DESIGN Shotgun metagenomic sequencing and qPCR were performed on OP and fecal samples collected longitudinally from 14 IWCF (ages 1-17 months) during ("On Antibiotics") and after ("Off Antibiotics") β-lactam therapy, and from 5 IWCF (3-16 months) never treated with antibiotics. RESULTS Total bacterial loads (TBL) for On Antibiotics samples were lower than for both Never (OP and fecal) and Off Antibiotics samples (fecal only). α-diversities (within-sample) for OP On Antibiotics samples were lower than for Never and Off Antibiotics samples but did not differ between fecal sample groups. β-diversity (between-sample) differed between all OP sample groups and between fecal On and Never Antibiotics and Off and Never antibiotics samples; however, fecal On and Off Antibiotics sample β-diversities did not differ. Patterns of change in antibiotic resistance gene abundances reflected shifts in microbial community composition. CONCLUSIONS β-lactam antibiotic exposure was followed by marked alterations in both OP and fecal microbiota. While microbiota appeared to rebound after treatment in both sample types, our results suggest that fecal microbiota recovered less than OP. The clinical consequences of these findings should be studied in IWCF and other populations frequently treated with antibiotics.
Collapse
Affiliation(s)
- Hillary S. Hayden
- Department of PediatricsUniversity of WashingtonSeattleWashingtonUSA
| | - Maria T. Nelson
- Department of PediatricsUniversity of WashingtonSeattleWashingtonUSA
| | - Sydney E. Ross
- Department of PediatricsIndiana University School of MedicineIndianapolisIndianaUSA
| | | | - Drake C. Bouzek
- Department of PediatricsUniversity of WashingtonSeattleWashingtonUSA
| | - Alex Eng
- Department of MicrobiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Adam Waalkes
- Department of MicrobiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Kelsi Penewit
- Department of Laboratory MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Benjamin T. Kopp
- Department of PediatricsEmory University School of MedicineAtlantaGeorgiaUSA
| | | | - Michael J. Rock
- Department of PediatricsUniversity of WisconsinMadisonWisconsinUSA
| | | | - Lucas R. Hoffman
- Department of PediatricsUniversity of WashingtonSeattleWashingtonUSA
| | - Don B. Sanders
- Department of PediatricsIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
3
|
Drzymała-Czyż S, Walkowiak J, Colombo C, Alicandro G, Storrösten OT, Kolsgaard M, Bakkeheim E, Strandvik B. Fatty acid abnormalities in cystic fibrosis-the missing link for a cure? iScience 2024; 27:111153. [PMID: 39620135 PMCID: PMC11607544 DOI: 10.1016/j.isci.2024.111153] [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: 01/05/2025] Open
Abstract
The care for cystic fibrosis (CF) has dramatically changed with the development of modulators, correctors, and potentiators of the CFTR molecule, which lead to improved clinical status of most people with CF (pwCF). The modulators influence phospholipids and ceramides, but not linoleic acid (LA) deficiency, associated with more severe phenotypes of CF. The LA deficiency is associated with upregulation of its transfer to arachidonic acid (AA). The AA release from membranes is increased and associated with increase of pro-inflammatory prostanoids and the characteristic inflammation is present before birth and bacterial infections. Docosahexaenoic acid is often decreased, especially in associated liver disease Some endogenously synthesized fatty acids are increased. Cholesterol and ceramide metabolisms are disturbed. The lipid abnormalities are present at birth, and before feeding in transgenic pigs and ferrets. This review focus on the lipid abnormalities and their associations to clinical symptoms in CF, based on clinical studies and experimental research.
Collapse
Affiliation(s)
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Institute of Pediatrics, Poznan University of Medical Sciences, Poznan, Poland
| | - Carla Colombo
- Cystic Fibrosis Centre, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, and Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Gianfranco Alicandro
- Cystic Fibrosis Centre, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, and Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Olav Trond Storrösten
- National Resource Centre for Cystic Fibrosis, Oslo University Hospital, Oslo, Norway
| | - Magnhild Kolsgaard
- National Resource Centre for Cystic Fibrosis, Oslo University Hospital, Oslo, Norway
| | - Egil Bakkeheim
- National Resource Centre for Cystic Fibrosis, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
4
|
Marsh R, Santos CD, Yule A, Dellschaft NS, Hoad CL, Ng C, Major G, Smyth AR, Rivett D, van der Gast C. Impact of extended Elexacaftor/Tezacaftor/Ivacaftor therapy on the gut microbiome in cystic fibrosis. J Cyst Fibros 2024; 23:967-976. [PMID: 38749891 DOI: 10.1016/j.jcf.2024.05.002] [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: 01/22/2024] [Revised: 04/29/2024] [Accepted: 05/04/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND There is a paucity of knowledge on the longer-term effects of CF transmembrane conductance regulator (CFTR) modulator therapies upon the gut microbiome and associated outcomes. In a pilot study, we investigated longitudinal Elexacaftor/Tezacaftor/Ivacaftor (ETI) therapy on the gut microbiota, metabolomic functioning, and clinical outcomes in people with CF (pwCF). STUDY DESIGN Faecal samples from 20 pwCF were acquired before and then following 3, 6, and 17+ months of ETI therapy. Samples were subjected to microbiota sequencing and targeted metabolomics to profile and quantify short-chain fatty acid composition. Ten healthy matched controls were included for comparison. Clinical data, including markers of intestinal function were integrated to investigate relationships. RESULTS Extended ETI therapy increased core microbiota diversity and composition, which translated to gradual shifts in whole microbiota composition towards that observed in healthy controls. Despite becoming more similar over time, CF microbiota and functional metabolite compositions remained significantly different to healthy controls. Antibiotic treatment for pulmonary infection significantly explained a relatively large degree of variation within the whole microbiota and rarer satellite taxa. Clinical outcomes were not significantly different following ETI. CONCLUSIONS Whilst differences persisted, a positive trajectory towards the microbiota observed in healthy controls was found. We posit that progression was predominately impeded by pulmonary antibiotics administration. We recommend future studies use integrated omics approaches within a combination of long-term longitudinal patient studies and model experimental systems. This will deepen our understanding of the impacts of CFTR modulator therapy and respiratory antibiotic interventions upon the gut microbiome and gastrointestinal pathophysiology in CF.
Collapse
Affiliation(s)
- Ryan Marsh
- Department of Applied Sciences, Northumbria University, Newcastle, UK
| | | | - Alexander Yule
- School of Medicine, University of Nottingham, UK; NIHR Nottingham Biomedical Research Centre, UK
| | | | - Caroline L Hoad
- Sir Peter Mansfield Imaging Centre, University of Nottingham, UK
| | - Christabella Ng
- School of Medicine, University of Nottingham, UK; NIHR Nottingham Biomedical Research Centre, UK
| | - Giles Major
- School of Medicine, University of Nottingham, UK; Nestlé Institute of Health Sciences, Société des Produits Nestlé, Lausanne, Switzerland
| | - Alan R Smyth
- School of Medicine, University of Nottingham, UK; NIHR Nottingham Biomedical Research Centre, UK; School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, UK
| | - Damian Rivett
- Department of Natural Sciences, Manchester Metropolitan University, UK
| | - Christopher van der Gast
- Department of Applied Sciences, Northumbria University, Newcastle, UK; Department of Respiratory Medicine, Northern Care Alliance NHS Foundation Trust, Salford, UK.
| |
Collapse
|
5
|
Suppakitjanusant P, Wang Y, Sivapiromrat AK, Hu C, Binongo J, Hunt WR, Weinstein S, Jathal I, Alvarez JA, Chassaing B, Ziegler TR, Gewirtz AT, Tangpricha V. Impact of high-dose cholecalciferol (vitamin D3) and inulin prebiotic on intestinal and airway microbiota in adults with cystic fibrosis: A 2 × 2 randomized, placebo-controlled, double-blind pilot study. J Clin Transl Endocrinol 2024; 37:100362. [PMID: 39188269 PMCID: PMC11345930 DOI: 10.1016/j.jcte.2024.100362] [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: 04/30/2024] [Revised: 06/26/2024] [Accepted: 07/17/2024] [Indexed: 08/28/2024] Open
Abstract
Background Cystic fibrosis (CF) is a multi-organ disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). Individuals with CF often have gastrointestinal (GI) dysbiosis due to chronic inflammation and antibiotic use. Previous studies suggested a role for vitamin D in reversing the GI dysbiosis found in CF. Objective To explore the potential role of a combination of high-dose oral cholecalciferol (vitamin D3) and fermentable dietary fiber, inulin, to impact bacterial composition, richness, and diversity of intestinal and airway microbiota in adults with CF. Methods This was a 2 × 2 factorial, double-blinded, placebo-controlled, randomized, pilot clinical trial in which adults with CF received oral cholecalciferol (vitamin D3) (50,000 IU/week) and/or inulin (12 g/day) for 12 weeks. Thus, there were 4 study groups (n = 10 subjects per group); 1) placebo 2) vitamin D3 3) inulin 4) vitamin D3 plus inulin. Stool and sputum samples were collected at baseline (just before) and after the intervention and were analysed using 16S ribosomal RNA gene sequencing for gut and airway microbiota composition. Statistical analyses assessed alpha and beta diversity to evaluate microbial community changes. Results Of a total of 254 screened participants, 40 eligible participants were randomized to one of the 4 treatment arms. Participants receiving vitamin D3 plus inulin exhibited greater changes in microbiome indexes in both intestinal and airway relative to those in the other study groups. Specific taxonomic changes supported the potential beneficial influence of this combination to mitigate both intestinal and airway dysbiosis in adults with CF. Conclusion This pilot study established that the combination of oral vitamin D3 and the prebiotic inulin was well tolerated over 12 weeks in adults with CF and altered gut and airway bacterial communities. Future research appear warranted to define clinical outcomes and the role of microbiota changes therein with this approach.
Collapse
Affiliation(s)
- Pichatorn Suppakitjanusant
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Yanling Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | | | - Chengcheng Hu
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jose Binongo
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - William R. Hunt
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | - Jessica A. Alvarez
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Benoit Chassaing
- INSERM U1016, Team “Mucosal Microbiota in Chronic Inflammatory Diseases”, CNRS UMR 8104, Université Paris Cité, Paris, France
| | - Thomas R. Ziegler
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrew T. Gewirtz
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Vin Tangpricha
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| |
Collapse
|
6
|
Roca M, Masip E, Colombo C, Boon M, Hulst JM, Garriga M, de Koning BAE, Bulfamante A, de Boeck K, Ribes-Koninckx C, Calvo-Lerma J. Long-term evaluation of faecal calprotectin levels in a European cohort of children with cystic fibrosis. Arch Dis Child 2024; 109:552-556. [PMID: 38123920 DOI: 10.1136/archdischild-2023-326221] [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: 08/20/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE Intestinal inflammation with contradictory data on faecal calprotectin (fCP) levels is documented in patients with cystic fibrosis (CF). The aim of this study was to longitudinally evaluate fCP in a cohort of children with CF and their relationship with clinical variables. DESIGN Prospective observational study to assess evolution of fCP levels, primary aimed at improving fat absorption. Along 1.5 years of follow-up (November 2016-May 2018) with four study visits pertaining to a pilot study (two of four) and to a clinical trial (two of four), the study outcomes were measured. SETTING Six European CF centres in the context of MyCyFAPP Project. SUBJECTS Children with CF and pancreatic insufficiency (2-18 years old). MAIN OUTCOME MEASUREMENTS fCP levels, pulmonary function (percentage of forced expiratory volume in 1 s (FEV1%)) and coefficient of fat absorption (CFA). Additionally, in the last two visits, gastrointestinal (GI) symptoms were evaluated through the PedsQL-GI Questionnaire. Linear mixed regression models were applied to assess association between fCP and FEV1, CFA and GI symptoms. RESULTS Twenty-nine children with CF and pancreatic insufficiency were included. fCP levels were inversely associated with total modified specific PedsQL-GI score (p=0.04) and positively associated with diarrhoea (p=0.03), but not with CFA. Along the four study visits, fCP significantly increased (from 62 to 256 µg/g) and pulmonary function decreased (from 97% to 87%), with a significant inverse association between the two study outcomes (p<0.001). CONCLUSIONS In children with CF, fCP levels are inversely associated with pulmonary function and thus the specificity of fCP as a marker of intestinal inflammation in paediatric patients with CF warrants further investigation.
Collapse
Affiliation(s)
- Maria Roca
- Health Research Institute La Fe, Valencia, Spain
| | - Etna Masip
- Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Carla Colombo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mieke Boon
- Department of Pediatrics, KU Leuven Hospital, Leuven, Belgium
| | - Jessie M Hulst
- Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, Toronto, Ontario, Canada
- Pediatrics, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | - Anna Bulfamante
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Kris de Boeck
- Department of Pediatrics, KU Leuven Hospital, Leuven, Belgium
| | | | - Joaquim Calvo-Lerma
- Health Research Institute La Fe, Valencia, Spain
- Universitat de València, Valencia, Spain
| |
Collapse
|
7
|
Mainz JG, Lester K, Elnazir B, Williamson M, McKone E, Cox D, Linnane B, Zagoya C, Duckstein F, Barucha A, Davies JC, McNally P. Reduction in abdominal symptoms (CFAbd-Score), faecal M2-pyruvate-kinase and Calprotectin over one year of treatment with Elexacaftor-Tezacaftor-Ivacaftor in people with CF aged ≥12 years - The RECOVER study. J Cyst Fibros 2024; 23:474-480. [PMID: 37806792 DOI: 10.1016/j.jcf.2023.10.001] [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: 07/05/2023] [Revised: 08/23/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND RECOVER is a multicentre post-approval study of Elexacaftor/Tezacaftor/Ivacaftor (ETI) in pwCF in Ireland and the UK. The CFAbd-Score is the first validated CF-specific patient reported outcome measure (PROM) focusing on gastrointestinal symptoms; it comprises 28 items in 5 domains. In a preliminary study, we previously reported reductions in abdominal symptoms (AS) in pwCF after 26 weeks of ETI-therapy using the CFAbd-Score. AIM to assess changes in AS in a second, large cohort and explore novel GI-biomarkers of gut inflammation and cell-proliferation in pwCF over one year of ETI-therapy. METHODS Participants were recruited as part of the RECOVER study at 8 sites (Ireland&UK). The CFAbd-Score was administered prior to ETI-initiation, and subsequently at 1,2,6 and 12 months on treatment. Faecal M2-pyruvate kinase (M2-PK) and calprotectin (FC) were quantified in samples collected at baseline, 1 and 6 months. RESULTS 108 CFAbd-Scores and 73 stool samples were collected at baseline. After 12 months of ETI-therapy, total CFAbd-Scores had significantly declined (15.0±1.4→9.8±1.2pts/p<0.001), and so had all its five domains of "pain" (16.9±2.0pts→9.9±1.8pts/p<0.01), "GERD" (14.4±1.8→9.9±1.6/p<0.05), "disorders of bowel movements" (19.2±1.4→14.1±1.5/p<0.01), "appetite" (7.0±1.1→4.6±1.2/p<0.01) and "impaired-QoL" (13.3±1.9→7.5±1.5/p<0.001). Levels of M2-PK and FC significantly decreased during ETI-therapy. DISCUSSION In-depth analysis of AS with the CFAbd-Score reveals a statistically significant, clinically relevant and sustained improvement with ETI. We attribute this to high sensitivity of the implemented CF-specific PROM, developed and validated following FDA-guidelines. Furthermore, for the first time during ETI-therapy a significant decline in faecal M2-PK, a marker of inflammation and cell-proliferation, was found, in parallel to FC.
Collapse
Affiliation(s)
- Jochen G Mainz
- Brandenburg Medical School (MHB) University. Klinikum Westbrandenburg, Brandenburg an der Havel, Germany.
| | - Karen Lester
- RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | | | | | - Ed McKone
- St Vincent's University Hospital, Dublin, Ireland
| | - Des Cox
- Children's Health Ireland, Dublin, Ireland
| | - Barry Linnane
- University of Limerick School of Medicine, Limerick, Ireland
| | - Carlos Zagoya
- Brandenburg Medical School (MHB) University. Klinikum Westbrandenburg, Brandenburg an der Havel, Germany
| | - Franziska Duckstein
- Brandenburg Medical School (MHB) University. Klinikum Westbrandenburg, Brandenburg an der Havel, Germany
| | - Anton Barucha
- Brandenburg Medical School (MHB) University. Klinikum Westbrandenburg, Brandenburg an der Havel, Germany; Department of Gastroenterology, Brandenburg Medical School (MHB) University, Brandenburg an der Havel, Germany
| | - Jane C Davies
- National Heart & Lung Institute, Imperial College London, UK; Royal Brompton Hospital, Guy's & St Thomas' NHS Trust, London, UK
| | - Paul McNally
- RCSI University of Medicine and Health Sciences, Dublin, Ireland; Children's Health Ireland, Dublin, Ireland
| |
Collapse
|
8
|
Sivapiromrat AK, Suppakitjanusant P, Wang Y, Hu C, Binongo J, Hunt WR, Weinstein S, Jathal I, Alvarez JA, Chassaing B, Ziegler TR, Gewirtz AT, Tangpricha V. Vitamin D and prebiotics for intestinal health in cystic fibrosis: Rationale and design for a randomized, placebo-controlled, double-blind, 2 x 2 trial of administration of prebiotics and cholecalciferol (vitamin D 3) (Pre-D trial) in adults with cystic fibrosis. Contemp Clin Trials Commun 2024; 38:101278. [PMID: 38435430 PMCID: PMC10904905 DOI: 10.1016/j.conctc.2024.101278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 01/18/2024] [Accepted: 02/17/2024] [Indexed: 03/05/2024] Open
Abstract
Individuals with cystic fibrosis (CF) have dysfunctional intestinal microbiota and increased gastrointestinal (GI) inflammation also known as GI dysbiosis. It is hypothesized that administration of high-dose cholecalciferol (vitamin D3) together with a prebiotic (inulin) will be effective, and possibly additive or synergistic, in reducing CF-related GI and airway dysbiosis. Thus, a 2 x 2 factorial design, placebo-controlled, double-blinded, pilot and feasibility, clinical trial was proposed to test this hypothesis. Forty adult participants with CF were block-randomized into one of four groups: 1) high-dose oral vitamin D3 (50,000 IU weekly) plus oral prebiotic placebo daily; 2) oral prebiotic (12 g inulin daily) plus oral placebo vitamin D3 weekly; 3) combined oral vitamin D3 weekly and oral prebiotic inulin daily; and 4) oral vitamin D3 placebo weekly and oral prebiotic placebo. The primary endpoints included 12-week changes in the microbial bacterial communities, gut and airway microbiota richness and diversity before and after the intervention. This pilot study examined whether vitamin D3 with or without prebiotics supplementation was feasible, changed airway and gut microbiota, and reduced dysbiosis, which in turn, may improve health outcomes and quality of life of patients with CF.
Collapse
Affiliation(s)
- Alisa K. Sivapiromrat
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Emory College, Emory University, Atlanta, GA, USA
| | - Pichatorn Suppakitjanusant
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Yanling Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Chengcheng Hu
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jose Binongo
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - William R. Hunt
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | - Jessica A. Alvarez
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Benoit Chassaing
- INSERM U1016, Team “Mucosal Microbiota in Chronic Inflammatory Diseases”, CNRS, UMR, 8104, Université Paris Cité, Paris, France
| | - Thomas R. Ziegler
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - Andrew T. Gewirtz
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Vin Tangpricha
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| |
Collapse
|
9
|
Price CE, Valls RA, Ramsey AR, Loeven NA, Jones JT, Barrack KE, Schwartzman JD, Royce DB, Cramer RA, Madan JC, Ross BD, Bliska J, O'Toole GA. Intestinal Bacteroides modulates inflammation, systemic cytokines, and microbial ecology via propionate in a mouse model of cystic fibrosis. mBio 2024; 15:e0314423. [PMID: 38179971 PMCID: PMC10865972 DOI: 10.1128/mbio.03144-23] [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: 11/21/2023] [Accepted: 12/01/2023] [Indexed: 01/06/2024] Open
Abstract
Persons with cystic fibrosis (CF), starting in early life, show intestinal microbiome dysbiosis characterized in part by a decreased relative abundance of the genus Bacteroides. Bacteroides is a major producer of the intestinal short chain fatty acid propionate. We demonstrate here that cystic fibrosis transmembrane conductance regulator-defective (CFTR-/-) Caco-2 intestinal epithelial cells are responsive to the anti-inflammatory effects of propionate. Furthermore, Bacteroides isolates inhibit the IL-1β-induced inflammatory response of CFTR-/- Caco-2 intestinal epithelial cells and do so in a propionate-dependent manner. The introduction of Bacteroides-supplemented stool from infants with cystic fibrosis into the gut of CftrF508del mice results in higher propionate in the stool as well as the reduction in several systemic pro-inflammatory cytokines. Bacteroides supplementation also reduced the fecal relative abundance of Escherichia coli, indicating a potential interaction between these two microbes, consistent with previous clinical studies. For a Bacteroides propionate mutant in the mouse model, pro-inflammatory cytokine KC is higher in the airway and serum compared with the wild-type (WT) strain, with no significant difference in the absolute abundance of these two strains. Taken together, our data indicate the potential multiple roles of Bacteroides-derived propionate in the modulation of systemic and airway inflammation and mediating the intestinal ecology of infants and children with CF. The roles of Bacteroides and the propionate it produces may help explain the observed gut-lung axis in CF and could guide the development of probiotics to mitigate systemic and airway inflammation for persons with CF.IMPORTANCEThe composition of the gut microbiome in persons with CF is correlated with lung health outcomes, a phenomenon referred to as the gut-lung axis. Here, we demonstrate that the intestinal microbe Bacteroides decreases inflammation through the production of the short-chain fatty acid propionate. Supplementing the levels of Bacteroides in an animal model of CF is associated with reduced systemic inflammation and reduction in the relative abundance of the opportunistically pathogenic group Escherichia/Shigella in the gut. Taken together, these data demonstrate a key role for Bacteroides and microbially produced propionate in modulating inflammation, gut microbial ecology, and the gut-lung axis in cystic fibrosis. These data support the role of Bacteroides as a potential probiotic in CF.
Collapse
Affiliation(s)
- Courtney E. Price
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, USA
| | - Rebecca A. Valls
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, USA
| | - Alexis R. Ramsey
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, USA
| | - Nicole A. Loeven
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, USA
| | - Jane T. Jones
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, USA
| | - Kaitlyn E. Barrack
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, USA
| | | | - Darlene B. Royce
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, USA
| | - Robert A. Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, USA
| | - Juliette C. Madan
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Hanove, USA
| | - Benjamin D. Ross
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, USA
| | - James Bliska
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, USA
| | - George A. O'Toole
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, USA
| |
Collapse
|
10
|
Reasoner SA, Bernard R, Waalkes A, Penewit K, Lewis J, Sokolow AG, Brown RF, Edwards KM, Salipante SJ, Hadjifrangiskou M, Nicholson MR. Longitudinal profiling of the intestinal microbiome in children with cystic fibrosis treated with elexacaftor-tezacaftor-ivacaftor. mBio 2024; 15:e0193523. [PMID: 38275294 PMCID: PMC10865789 DOI: 10.1128/mbio.01935-23] [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: 08/14/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
The intestinal microbiome influences growth and disease progression in children with cystic fibrosis (CF). Elexacaftor-tezacaftor-ivacaftor (ELX/TEZ/IVA), the newest pharmaceutical modulator for CF, restores the function of the pathogenic mutated CF transmembrane conductance regulator (CFTR) channel. We performed a single-center longitudinal analysis of the effect of ELX/TEZ/IVA on the intestinal microbiome, intestinal inflammation, and clinical parameters in children with CF. Following ELX/TEZ/IVA, children with CF had significant improvements in body mass index and percent predicted forced expiratory volume in one second, and required fewer antibiotics for respiratory infections. Intestinal microbiome diversity increased following ELX/TEZ/IVA coupled with a decrease in the intestinal carriage of Staphylococcus aureus, the predominant respiratory pathogen in children with CF. There was a reduced abundance of microbiome-encoded antibiotic resistance genes. Microbial pathways for aerobic respiration were reduced after ELX/TEZ/IVA. The abundance of microbial acid tolerance genes was reduced, indicating microbial adaptation to increased CFTR function. In all, this study represents the first comprehensive analysis of the intestinal microbiome in children with CF receiving ELX/TEZ/IVA.IMPORTANCECystic fibrosis (CF) is an autosomal recessive disease with significant gastrointestinal symptoms in addition to pulmonary complications. Recently approved treatments for CF, CF transmembrane conductance regulator (CFTR) modulators, are anticipated to substantially improve the care of people with CF and extend their lifespans. Prior work has shown that the intestinal microbiome correlates with health outcomes in CF, particularly in children. Here, we study the intestinal microbiome of children with CF before and after the CFTR modulator, ELX/TEZ/IVA. We identify promising improvements in microbiome diversity, reduced measures of intestinal inflammation, and reduced antibiotic resistance genes. We present specific bacterial taxa and protein groups which change following ELX/TEZ/IVA. These results will inform future mechanistic studies to understand the microbial improvements associated with CFTR modulator treatment. This study demonstrates how the microbiome can change in response to a targeted medication that corrects a genetic disease.
Collapse
Affiliation(s)
- Seth A. Reasoner
- Department of Pathology, Microbiology, and Immunology, Division of Molecular Pathogenesis, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Rachel Bernard
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Monroe Carrell Junior Children’s Hospital at Vanderbilt, Nashville, Tennessee, USA
| | - Adam Waalkes
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Kelsi Penewit
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Janessa Lewis
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Andrew G. Sokolow
- Department of Pediatrics, Division of Allergy, Immunology, and Pulmonary Medicine, Monroe Carrell Junior Children’s Hospital at Vanderbilt, Nashville, Tennessee, USA
| | - Rebekah F. Brown
- Department of Pediatrics, Division of Allergy, Immunology, and Pulmonary Medicine, Monroe Carrell Junior Children’s Hospital at Vanderbilt, Nashville, Tennessee, USA
| | - Kathryn M. Edwards
- Department of Pediatrics, Division of Infectious Diseases, Monroe Carrell Junior Children’s Hospital at Vanderbilt, Nashville, Tennessee, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Maria Hadjifrangiskou
- Department of Pathology, Microbiology, and Immunology, Division of Molecular Pathogenesis, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Center for Personalized Microbiology (CPMi), Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Maribeth R. Nicholson
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Monroe Carrell Junior Children’s Hospital at Vanderbilt, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
11
|
Duckworth LA, Sutton KA, Shaikh N, Wang J, Hall-Moore C, Holtz LR, Tarr PI, Rubenstein RC. Quantification of Enteric Dysfunction in Cystic Fibrosis: Inter- and Intraindividual Variability. J Pediatr 2024; 265:113800. [PMID: 37866678 PMCID: PMC10869934 DOI: 10.1016/j.jpeds.2023.113800] [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: 06/29/2023] [Revised: 09/18/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
OBJECTIVES To test the utility of various biomarkers as indicators of gut dysfunction in cystic fibrosis (CF) and determine whether intraindividual variations in these measures are repeatable over short intervals and whether interindividual variations correlate with clinical outcomes. STUDY DESIGN We performed a cross-sectional, limited longitudinal study of children with CF aged 1-21 years who provided blood and stool samples at 2 or 3 visits, 2 weeks and 3 months apart, which were assayed for markers of intestinal inflammation (fecal calprotectin [fCal], lipocalin-2 [fLcn2], neopterin), and permeability (plasma lipopolysaccharide [LPS] antibodies, LPS-binding protein) by enzyme immunoassays. Control specimens were obtained from children without CF who had undergone esophagogastroduodenoscopy and had no evidence of gut inflammation. RESULTS Twenty-six of 29 participants with CF completed the study. Sixty-nine stools (57 case/12 control) and 76 plasmas (60 case/16 control) were analyzed. LPS antibody had reliable intraindividual stability. fCal, fLcn2, and neopterin were significantly greater in CF than in control samples. fCal was negatively correlated with 3-month interval change (Δ) in weight-for-age z-score, body mass index/weight-for-length z-score, and forced expiratory volume in 1 second. fLcn2 was negatively correlated with FEV1 but not with anthropometrics. No marker correlated with Δbody mass index/weight-for-length z-score or ΔFEV1. CONCLUSIONS fLcn2 is elevated in people with CF and might predict worse interval pulmonary function. Expanded studies are warranted to test if fLcn2 correlates with changes in additional outcomes.
Collapse
Affiliation(s)
- Laura A Duckworth
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO.
| | - Kimberly A Sutton
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| | - Nurmohammad Shaikh
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| | - Jinli Wang
- Center for Biostatistics and Data Science, Washington University in St Louis, St Louis, MO
| | - Carla Hall-Moore
- Center for Biostatistics and Data Science, Washington University in St Louis, St Louis, MO
| | - Lori R Holtz
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| | - Phillip I Tarr
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| | - Ronald C Rubenstein
- Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University in St Louis, St Louis, MO
| |
Collapse
|
12
|
Sivapiromrat AK, Suppakitjanusant P, Wang Y, Binongo J, Hunt WR, Gewirtz A, Alvarez JA, Hu C, Weinstein S, Jathal I, Ziegler TR, Tangpricha V. Vitamin D and Prebiotics for Intestinal Health in Cystic Fibrosis: Rationale and design for a randomized, placebo-controlled, double-blind, 2 × 2 trial of administration of prebiotics and cholecalciferol (vitamin D 3) (Pre-D Trial) in adults with cystic fibrosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.04.24300860. [PMID: 38343811 PMCID: PMC10854319 DOI: 10.1101/2024.01.04.24300860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Individuals with cystic fibrosis (CF) have dysfunctional intestinal microbiota and increased gastrointestinal (GI) inflammation also known as GI dysbiosis. It is hypothesized that administration of high-dose cholecalciferol (vitamin D3) together with a prebiotic (inulin) will be effective, and possibly additive or synergistic, in reducing CF-related GI dysbiosis and improving intestinal functions. Thus, a 2 × 2 factorial design, placebo-controlled, double-blind, clinical trial was proposed to test this hypothesis. Forty adult participants with CF will be block-randomized into one of four groups: 1) high-dose oral vitamin D3 (50,000 IU weekly) plus oral prebiotic placebo daily; 2) oral prebiotic (12 g inulin daily) plus oral placebo vitamin D3 weekly; 3) combined oral vitamin D3 weekly and oral prebiotic inulin daily; and 4) oral vitamin D3 placebo weekly and oral prebiotic placebo. The primary endpoints will include 12-week changes in the reduced relative abundance of gammaproteobacteria, and gut microbiota richness and diversity before and after the intervention. This clinical study will examine whether vitamin D3 with or without prebiotics will improve intestinal health and reduce GI dysbiosis, which in turn, should improve health outcomes and quality of life of patients with CF.
Collapse
Affiliation(s)
| | - Pichatorn Suppakitjanusant
- Emory University, Atlanta, GA, USA
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Yanling Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Jose Binongo
- Emory University, Atlanta, GA, USA
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - William R Hunt
- Emory University, Atlanta, GA, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrew Gewirtz
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Jessica A Alvarez
- Emory University, Atlanta, GA, USA
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Chengcheng Hu
- Emory University, Atlanta, GA, USA
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | | | | | - Thomas R Ziegler
- Emory University, Atlanta, GA, USA
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Vin Tangpricha
- Emory University, Atlanta, GA, USA
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
13
|
Williams NC, Jayaratnasingam J, Prayle AP, Nevitt SJ, Smyth AR. Prebiotics for people with cystic fibrosis. Cochrane Database Syst Rev 2023; 9:CD015236. [PMID: 37753791 PMCID: PMC10523429 DOI: 10.1002/14651858.cd015236.pub2] [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] [Indexed: 09/28/2023]
Abstract
BACKGROUND Cystic fibrosis (CF) is a multisystem disease; the importance of growth and nutritional status is well established given their implications for lung function and overall survivability. Furthermore, it has been established that intestinal microbial imbalance and inflammation are present in people with CF. Oral prebiotics are commercially available substrates that are selectively utilised by host intestinal micro-organisms and may improve both intestinal and overall health. OBJECTIVES To evaluate the benefits and harms of prebiotics for improving health outcomes in children and adults with CF. SEARCH METHODS We searched the Cochrane Cystic Fibrosis Trials Register compiled from electronic database searches and handsearching of journals and conference abstract books. We also searched the reference lists of relevant articles and reviews. Date of last search: 19 October 2022. We also searched PubMed and online trials registries. Date of last search: 13 January 2023. SELECTION CRITERIA Randomised controlled trials (RCTs) and quasi-RCTs assessing the efficacy of prebiotics in children and adults with CF. We planned to only include the first treatment period from cross-over RCTs, regardless of washout period. DATA COLLECTION AND ANALYSIS We did not identify any relevant trials. MAIN RESULTS We did not identify any relevant trials for inclusion in this review. AUTHORS' CONCLUSIONS This review did not find any evidence for the use of prebiotics in people with CF. Until such evidence is available, it is reasonable for clinicians to follow any local guidelines and to discuss the use of dietary prebiotics with their patients. Large and robust RCTs assessing the dietary prebiotics of inulin or galacto-oligosaccharides or fructo-oligosaccharides, or any combination of these, are needed. Such studies should be of at least 12 months in duration and assess outcomes such as growth and nutrition, gastrointestinal symptoms, pulmonary exacerbations, lung function, inflammatory biomarkers, hospitalisations, intestinal microbial profiling, and faecal short-chain fatty acids. Trials should include both children and adults and aim to be adequately powered to allow for subgroup analysis by age.
Collapse
Affiliation(s)
- Neil C Williams
- Exercise and Health Research Group, Sport Health and Performance Enhancement (SHAPE) Research Centre, School of Science and Technology., Nottingham Trent University, Nottingham, UK
| | - Jacob Jayaratnasingam
- Exercise and Health Research Group, Sport Health and Performance Enhancement (SHAPE) Research Centre, School of Science and Technology., Nottingham Trent University, Nottingham, UK
| | - Andrew P Prayle
- Nottingham Cystic Fibrosis and Chidlren's Respiratory Research Centre, University of Nottingham, Nottingham, UK
| | - Sarah J Nevitt
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Alan R Smyth
- Division of Child Health, Obstetrics & Gynaecology (COG), School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
14
|
Lussac-Sorton F, Charpentier É, Imbert S, Lefranc M, Bui S, Fayon M, Berger P, Enaud R, Delhaes L. The gut-lung axis in the CFTR modulator era. Front Cell Infect Microbiol 2023; 13:1271117. [PMID: 37780857 PMCID: PMC10540301 DOI: 10.3389/fcimb.2023.1271117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
The advent of CFTR modulators represents a turning point in the history of cystic fibrosis (CF) management, changing profoundly the disease's clinical course by improving mucosal hydration. Assessing changes in airway and digestive tract microbiomes is of great interest to better understand the mechanisms and to predict disease evolution. Bacterial and fungal dysbiosis have been well documented in patients with CF; yet the impact of CFTR modulators on microbial communities has only been partially deciphered to date. In this review, we aim to summarize the current state of knowledge regarding the impact of CFTR modulators on both pulmonary and digestive microbiomes. Our analysis also covers the inter-organ connections between lung and gut communities, in order to highlight the gut-lung axis involvement in CF pathophysiology and its evolution in the era of novel modulators therapies.
Collapse
Affiliation(s)
- Florian Lussac-Sorton
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
| | - Éléna Charpentier
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
| | - Sébastien Imbert
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| | - Maxime Lefranc
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| | - Stéphanie Bui
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| | - Michael Fayon
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| | - Patrick Berger
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| | - Raphaël Enaud
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| | - Laurence Delhaes
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- CHU Bordeaux, Service de Parasitologie et Mycologie, Centre de Ressources et de Compétences de la Mucoviscidose (CRCM), Service de Pédiatrie, Service d’Exploration Fonctionnelle Respiratoire, CIC, Bordeaux, France
| |
Collapse
|
15
|
Asensio-Grau A, Calvo-Lerma J, Ferriz-Jordán M, García-Hernández J, Heredia A, Andrés A. Effect of Lactobacillaceae Probiotics on Colonic Microbiota and Metabolite Production in Cystic Fibrosis: A Comparative In Vitro Study. Nutrients 2023; 15:3846. [PMID: 37686878 PMCID: PMC10490339 DOI: 10.3390/nu15173846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Cystic Fibrosis-related gut dysbiosis (CFRGD) has become a recognised complication in children with this condition, and current evidence remains insufficient to guide the selection of probiotic strains for supplementation treatments. The aim of this study was to characterise the effect of three probiotic strains on CFRGD by means of a dynamic in vitro simulation of the colonic fermentation (SHIME®). The configuration of the system included three bioreactors colonised with the faecal inoculum of a child with cystic fibrosis. For 20 days, each bioreactor was supplied daily with either Lacticaseibacillus rhamnosus GG (ATCC 53103 TM), Limosilactobacillus reuteri (DSM 17938) or Lactiplantibacillus plantarum (DSM 22266). The baseline microbiota was characterised by a high abundance of Prevotella, Faecalibacterium and Acidaminococcus genera. After 20 days of supplementation, L. rhamnosus and L. plantarum reduced Prevotella significantly, and the three strains led to increased Faecalibacterium and Bifidobacterium and decreased Acidaminococcus, with some of these changes being maintained 10 days after ceasing supplementation. The metabolic activity remained unaltered in terms of short-chain fatty acids, but branched-chain fatty acids showed a significant decrease, especially with L. plantarum. Additionally, ammonia decreased at 20 days of supplementation, and lactate continuously increased with the three strains. The effects on colonic microbiota of L. rhamnosus, L. reuteri or L. plantarum were established, including increased beneficial bacteria, such as Faecalibacterium, and beneficial metabolites such as lactate; and on the other hand, a reduction in pathogenic genera, including Prevotella or Acidaminococcus and branched-chain fatty acids, overall supported their use as probiotics in the context of CFRGD.
Collapse
Affiliation(s)
- Andrea Asensio-Grau
- Institute of Food Engineering (IIA-FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain; (A.A.-G.); (M.F.-J.); (A.A.)
- Joint Research Unit NutriCura, Avda. Fernando Abril Martorell 106, 46026 València, Spain
| | - Joaquim Calvo-Lerma
- Institute of Food Engineering (IIA-FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain; (A.A.-G.); (M.F.-J.); (A.A.)
- Joint Research Unit NutriCura, Avda. Fernando Abril Martorell 106, 46026 València, Spain
| | - Miguel Ferriz-Jordán
- Institute of Food Engineering (IIA-FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain; (A.A.-G.); (M.F.-J.); (A.A.)
| | - Jorge García-Hernández
- Advanced Food Microbiology Centre (CAMA), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain
| | - Ana Heredia
- Institute of Food Engineering (IIA-FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain; (A.A.-G.); (M.F.-J.); (A.A.)
- Joint Research Unit NutriCura, Avda. Fernando Abril Martorell 106, 46026 València, Spain
| | - Ana Andrés
- Institute of Food Engineering (IIA-FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain; (A.A.-G.); (M.F.-J.); (A.A.)
- Joint Research Unit NutriCura, Avda. Fernando Abril Martorell 106, 46026 València, Spain
| |
Collapse
|
16
|
Price CE, Hampton TH, Valls RA, Barrack KE, O’Toole GA, Madan JC, Coker MO. Development of the intestinal microbiome in cystic fibrosis in early life. mSphere 2023; 8:e0004623. [PMID: 37404016 PMCID: PMC10449510 DOI: 10.1128/msphere.00046-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/12/2023] [Indexed: 07/06/2023] Open
Abstract
Cystic fibrosis (CF) is a heritable disease that causes altered physiology at mucosal sites; these changes result in chronic infections in the lung, significant gastrointestinal complications as well as dysbiosis of the gut microbiome, although the latter has been less well explored. Here, we describe the longitudinal development of the gut microbiome in a cohort of children with CF (cwCF) from birth through early childhood (0-4 years of age) using 16S rRNA gene amplicon sequencing of stool samples as a surrogate for the gut microbiota. Similar to healthy populations, alpha diversity of the gut microbiome increases significantly with age, but diversity plateaus at ~2 years of age for this CF cohort. Several taxa that have been associated with dysbiosis in CF change with age toward a more healthy-like composition; notable exceptions include Akkermansia, which decreases with age, and Blautia, which increases with age. We also examined the relative abundance and prevalence of nine taxa associated with CF lung disease, several of which persist across early life, highlighting the possibility of the lung being seeded directly from the gut early in life. Finally, we applied the Crohn's Dysbiosis Index to each sample, and found that high Crohn's-associated dysbiosis early in life (<2 years) was associated with significantly lower Bacteroides in samples collected from 2 to 4 years of age. Together, these data comprise an observational study that describes the longitudinal development of the CF-associated gut microbiota and suggest that early markers associated with inflammatory bowel disease may shape the later gut microbiota of cwCF. IMPORTANCE Cystic fibrosis is a heritable disease that disrupts ion transport at mucosal surfaces, causing a buildup of mucus and dysregulation of microbial communities in both the lungs and the intestines. Persons with CF are known to have dysbiotic gut microbial communities, but the development of these communities over time beginning at birth has not been thoroughly studied. Here, we describe an observation study following the development of the gut microbiome of cwCF throughout the first 4 years of life, during the critical window of both gut microbiome and immune development. Our findings indicate the possibility of the gut microbiota as a reservoir of airway pathogens and a surprisingly early indication of a microbiota associated with inflammatory bowel disease.
Collapse
Affiliation(s)
- Courtney E. Price
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Thomas H. Hampton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Rebecca A. Valls
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Kaitlyn E. Barrack
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - George A. O’Toole
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Juliette C. Madan
- Department of Pediatrics, Children’s Hospital at Dartmouth, Dartmouth Health, Lebanon, New Hampshire, USA
- Department of Psychiatry, Children’s Hospital at Dartmouth, Dartmouth Health, Lebanon, New Hampshire, USA
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- Department of Quantitative Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Modupe O. Coker
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- Department of Quantitative Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, New Jersey, USA
| |
Collapse
|
17
|
Marsh R, Dos Santos C, Hanson L, Ng C, Major G, Smyth AR, Rivett D, van der Gast C. Tezacaftor/Ivacaftor therapy has negligible effects on the cystic fibrosis gut microbiome. Microbiol Spectr 2023; 11:e0117523. [PMID: 37607068 PMCID: PMC10581179 DOI: 10.1128/spectrum.01175-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/28/2023] [Indexed: 08/24/2023] Open
Abstract
People with cystic fibrosis (pwCF) experience a range of persistent gastrointestinal symptoms throughout life. There is evidence indicating interaction between the microbiota and gut pathophysiology in CF. However, there is a paucity of knowledge on the potential effects of CF transmembrane conductance regulator (CFTR) modulator therapies on the gut microbiome. In a pilot study, we investigated the impact of Tezacaftor/Ivacaftor dual combination CFTR modulator therapy on the gut microbiota and metabolomic functioning in pwCF. Fecal samples from 12 pwCF taken at baseline and following placebo or Tezacaftor/Ivacaftor administration were subjected to microbiota sequencing and to targeted metabolomics to assess the short-chain fatty acid (SCFA) composition. Ten healthy matched controls were included as a comparison. Inflammatory calprotectin levels and patient symptoms were also investigated. No significant differences were observed in overall gut microbiota characteristics between any of the study stages, extended also across intestinal inflammation, gut symptoms, and SCFA-targeted metabolomics. However, microbiota and SCFA metabolomic compositions, in pwCF, were significantly different from controls in all study treatment stages. CFTR modulator therapy with Tezacaftor/Ivacaftor had negligible effects on both the gut microbiota and SCFA composition across the course of the study and did not alter toward compositions observed in healthy controls. Future longitudinal CFTR modulator studies will investigate more effective CFTR modulators and should use prolonged sampling periods, to determine whether longer-term changes occur in the CF gut microbiome. IMPORTANCE People with cystic fibrosis (pwCF) experience persistent gastrointestinal (GI) symptoms throughout life. The research question "how can we relieve gastrointestinal symptoms, such as stomach pain, bloating, and nausea?" remains a top priority for clinical research in CF. While CF transmembrane conductance regulator (CFTR) modulator therapies are understood to correct underlying issues of CF disease and increasing the numbers of pwCF are now receiving some form of CFTR modulator treatment. It is not known how these therapies affect the gut microbiome or GI system. In this pilot study, we investigated, for the first time, effects of the dual combination CFTR modulator medicine, Tezacaftor/Ivacaftor. We found it had negligible effects on patient GI symptoms, intestinal inflammation, or gut microbiome composition and functioning. Our findings are important as they fill important knowledge gaps on the relative effectiveness of these widely used treatments. We are now investigating triple combination CFTR modulators with prolonged sampling periods.
Collapse
Affiliation(s)
- Ryan Marsh
- Department of Applied Sciences, Northumbria University, Newcastle, United Kingdom
| | - Claudio Dos Santos
- Department of Natural Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Liam Hanson
- Department of Natural Sciences, Manchester Metropolitan University, Manchester, United Kingdom
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Christabella Ng
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- NIHR Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Giles Major
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Nestlé Institute of Health Sciences, Société des Produits Nestlé, Lausanne, Switzerland
| | - Alan R. Smyth
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- NIHR Nottingham Biomedical Research Centre, Nottingham, United Kingdom
| | - Damian Rivett
- Department of Natural Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Christopher van der Gast
- Department of Applied Sciences, Northumbria University, Newcastle, United Kingdom
- Department of Respiratory Medicine, Salford Royal NHS Foundation Trust, Salford, United Kingdom
| |
Collapse
|
18
|
de Souza Furtado J, de Almeida Brasiel PG, Luquetti SCPD. Profile of the intestinal microbiota of patients with cystic fibrosis: A systematic review. Clin Nutr ESPEN 2023; 55:400-406. [PMID: 37202074 DOI: 10.1016/j.clnesp.2023.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/28/2023] [Accepted: 04/11/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND & AIMS Cystic fibrosis (CF) is a multisystem disease that can compromise several human body organs. The autosomal recessive genetic disorder is caused by different mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, responsible for chloride ion transport across apical membranes of epithelial cells in tissues and bicarbonate secretion. In this study, we provide a systematic review of the profile of the intestinal microbiota of cystic fibrosis individuals. METHODS The review was conducted according to Preferred Items of Reports for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. PubMed/MEDLINE and Scopus databases were searched for relevant articles until Jully 2022. RESULTS Eighteen studies (1304 participants) met the inclusion criteria. The quality and bias was assessed using the Methodological index for non-randomized studies (MINORS) tool, with the majority of the studies indicating medium to high quality. Results showed significant changes in the composition of the intestinal microbiota of the individuals with CF compared with healthy controls, with increased of Enterococcus, Veillonella, and Streptococcus, and decreased of Bifidobacterium, Roseburia, and Alistipes genus. The intestinal bacterial community of CF patients was marked by a reduction in its richness and diversity. CONCLUSION The systematic review suggests a change in the intestinal microbiota of CF individuals, characterized by a reduction in microbial diversity and abundance of some bacterial markers.
Collapse
|
19
|
Tabassum A, Ali A, Zahedi FD, Ismail NAS. Immunomodulatory Role of Vitamin D on Gut Microbiome in Children. Biomedicines 2023; 11:biomedicines11051441. [PMID: 37239112 DOI: 10.3390/biomedicines11051441] [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: 05/04/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Vitamin D plays a role in regulating the immune system and can be linked to the alteration of the gut microbiome, which leads to several immunological diseases. This systematic review aims to explore the relationship between Vitamin D and children's gut microbiome, as well as its impact towards the immune system. We have systematically collated relevant studies from different databases concerning changes in the gut microbiome of children from infants to 18 years old associated with Vitamin D and the immunological pathways. The studies utilized 16S rRNA sequencing analysis of fecal matter with or without Vitamin D supplementation and Vitamin D levels. Ten studies were selected for the review, among which eight studies showed significant alterations in the gut microbiome related to Vitamin D supplementation or Vitamin D levels. The taxa of the phylum Actinobacteria, Bacteroidetes, Firmicutes, and Proteobacteria are the most altered in these studies. The alteration of the taxa alters the Th1 and Th2 pathways and changes the immune response. We will discuss how Vitamin D may contribute to the activation of immune pathways via its effects on intestinal barrier function, microbiome composition, and/or direct effects on immune responses. In conclusion, the studies examined in this review have provided evidence that Vitamin D levels may have an impact on the composition of children's gut microbiomes.
Collapse
Affiliation(s)
- Anika Tabassum
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Adli Ali
- Department of Pediatric, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Farah Dayana Zahedi
- Department of Otorhinolaryngology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Noor Akmal Shareela Ismail
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
20
|
Ney LM, Wipplinger M, Grossmann M, Engert N, Wegner VD, Mosig AS. Short chain fatty acids: key regulators of the local and systemic immune response in inflammatory diseases and infections. Open Biol 2023; 13:230014. [PMID: 36977462 PMCID: PMC10049789 DOI: 10.1098/rsob.230014] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
The human intestinal microbiome substantially affects human health and resistance to infections in its dynamic composition and varying release of microbial-derived metabolites. Short-chain fatty acids (SCFA) produced by commensal bacteria through fermentation of indigestible fibres are considered key regulators in orchestrating the host immune response to microbial colonization by regulating phagocytosis, chemokine and central signalling pathways of cell growth and apoptosis, thereby shaping the composition and functionality of the intestinal epithelial barrier. Although research of the last decades provided valuable insight into the pleiotropic functions of SCFAs and their capability to maintain human health, mechanistic details on how SCFAs act across different cell types and other organs are not fully understood. In this review, we provide an overview of the various functions of SCFAs in regulating cellular metabolism, emphasizing the orchestration of the immune response along the gut-brain, the gut-lung and the gut-liver axes. We discuss their potential pharmacological use in inflammatory diseases and infections and highlight new options of relevant human three-dimensional organ models to investigate and validate their biological functions in more detail.
Collapse
Affiliation(s)
- Lisa-Marie Ney
- Institute of Biochemistry II, Jena University Hospital, Kastanienallee 1, 07747 Jena, Germany
| | - Maximilian Wipplinger
- Institute of Biochemistry II, Jena University Hospital, Kastanienallee 1, 07747 Jena, Germany
| | - Martha Grossmann
- Institute of Biochemistry II, Jena University Hospital, Kastanienallee 1, 07747 Jena, Germany
| | - Nicole Engert
- Institute of Biochemistry II, Jena University Hospital, Kastanienallee 1, 07747 Jena, Germany
| | - Valentin D Wegner
- Institute of Biochemistry II, Jena University Hospital, Kastanienallee 1, 07747 Jena, Germany
| | - Alexander S Mosig
- Institute of Biochemistry II, Jena University Hospital, Kastanienallee 1, 07747 Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
21
|
Caley LR, White H, de Goffau MC, Floto RA, Parkhill J, Marsland B, Peckham DG. Cystic Fibrosis-Related Gut Dysbiosis: A Systematic Review. Dig Dis Sci 2023; 68:1797-1814. [PMID: 36600119 DOI: 10.1007/s10620-022-07812-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIMS Cystic Fibrosis (CF) is associated with gut dysbiosis, local and systemic inflammation, and impaired immune function. Gut microbiota dysbiosis results from changes in the complex gut milieu in response to CF transmembrane conductance regulator (CFTR) dysfunction, pancreatic malabsorption, diet, medications, and environmental influences. In several diseases, alteration of the gut microbiota influences local and systemic inflammation and disease outcomes. We conducted a systematic review of the gut microbiota in CF and explored factors influencing dysbiosis. METHODS An electronic search of three databases was conducted in January 2019, and re-run in June 2021. Human, animal, and in vitro studies were included. The primary outcome was differences in the gut microbiota between people with CF (pwCF) and healthy controls. Secondary outcomes included the relationship between the gut microbiota and other factors, including diet, medication, inflammation, and pulmonary function in pwCF. RESULTS Thirty-eight studies were identified. The literature confirmed the presence of CF-related gut dysbiosis, characterized by reduced diversity and several taxonomic changes. There was a relative increase of bacteria associated with a pro-inflammatory response coupled with a reduction of those considered anti-inflammatory. However, studies linking gut dysbiosis to systemic and lung inflammation were limited. Causes of gut dysbiosis were multifactorial, and findings were variable. Data on the impact of CFTR modulators on the gut microbiota were limited. CONCLUSIONS CF-related gut dysbiosis is evident in pwCF. Whether this influences local and systemic disease and is amenable to interventions with diet and drugs, such as CFTR modulators, requires further investigation.
Collapse
Affiliation(s)
- L R Caley
- Leeds Institute of Medical Research, St James's University Hospital, Clinical Sciences Building, Leeds, LS9 7TF, UK
| | - H White
- Nutrition, Health & Environment, Leeds Beckett University, Leeds, UK
| | - M C de Goffau
- Wellcome Sanger Institute, Cambridge, UK.,Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - R A Floto
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK.,Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, UK
| | - J Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - B Marsland
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - D G Peckham
- Leeds Institute of Medical Research, St James's University Hospital, Clinical Sciences Building, Leeds, LS9 7TF, UK. .,Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| |
Collapse
|
22
|
Lazzarotto ES, Vasco JFDM, Führ F, Riedi CA, Filho NAR. Systematic review on fecal calprotectin in cystic fibrosis. J Pediatr (Rio J) 2023; 99:4-10. [PMID: 35523321 PMCID: PMC9875247 DOI: 10.1016/j.jped.2022.01.006] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVES Fecal calprotectin is an inflammatory marker used for monitoring intestinal diseases. It has been studied as a marker of intestinal inflammation in cystic fibrosis (CF), a multi-systemic genetic disease caused by alterations to the CFTR gene. Manifestations of the disease favor a systemic inflammation not limited to the respiratory tract, therefore, calprotectin is a non-invasive and effective diagnostic method. The aim of the study was to perform a systematic review of the literature with a qualitative synthesis of studies. SOURCES The articles were selected from PubMed, Web of Science, Scielo and Lilacs. SUMMARY OF THE FINDINGS Nine studies were selected for that qualitative synthesis, one was a randomized clinical trial, and eight were case-control or cohort designs. Most studies have indicated that calprotectin is a marker of systemic inflammation in CF and not just intestinal inflammation. Calprotectin is an aid in monitoring inflammatory bowel conditions in patients with cystic fibrosis. CONCLUSION Further studies should be conducted to investigate the role of this marker in the systemic inflammation of CF.
Collapse
Affiliation(s)
| | | | - Fabiane Führ
- Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Carlos Antônio Riedi
- Universidade Federal do Paraná, Curitiba, PR, Brazil; Universidade Federal do Paraná, Hospital de Clínicas, Curitiba, PR, Brazil
| | - Nelson Augusto Rosário Filho
- Universidade Federal do Paraná, Curitiba, PR, Brazil; Universidade Federal do Paraná, Hospital de Clínicas, Curitiba, PR, Brazil
| |
Collapse
|
23
|
Testa I, Crescenzi O, Esposito S. Gut Dysbiosis in Children with Cystic Fibrosis: Development, Features and the Role of Gut-Lung Axis on Disease Progression. Microorganisms 2022; 11:microorganisms11010009. [PMID: 36677301 PMCID: PMC9865868 DOI: 10.3390/microorganisms11010009] [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: 10/14/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Cystic fibrosis (CF) is the most common autosomal recessive disease among Caucasians. Over the last 20 years, culture-independent analysis, including next-generation sequencing, has paired with culture-based microbiology, offering deeper insight into CF lung and gut microbiota. The aim of this review is to analyse the features of gut microbiota in patients with CF and its possible role in the progression of the disease, establishing the basis for a potential role in microbe-based therapies. The literature analysis showed that the gut environment in CF patients has unique features due to the characteristics of the disease, such as decreased bicarbonate secretion, increased luminal viscosity, and an acidic small intestinal environment, which, due to the treatment, includes regular antibiotic use or a high-energy and fat-dense diet. As a result, the gut microbial composition appears altered, with reduced richness and diversity. Moreover, the population of pro-inflammatory bacteria is higher, while immunomodulatory genera, such as Bacteroides and Bifidobacterium, are scarcer. The imbalanced gut microbial population has a potential role in the development of systemic inflammation and may influence clinical outcomes, such as respiratory exacerbations, spirometry results, and overall growth. Although a better understanding of the pathophysiology behind the gut-lung axis is needed, these findings support the rationale for considering gut microbiota manipulation as a possible intervention to regulate the severity and progression of the disease.
Collapse
Affiliation(s)
- Ilaria Testa
- Respiratory Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1LE, UK
| | - Oliviero Crescenzi
- Department of Anaesthesia, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London WC1N 1LE, UK
| | - Susanna Esposito
- Paediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Correspondence:
| |
Collapse
|
24
|
Williams N, Jayaratnasingam J, Prayle AP, Nevitt SJ, Smyth AR. Prebiotics for people with cystic fibrosis. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2022; 2022:CD015236. [PMCID: PMC9757150 DOI: 10.1002/14651858.cd015236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To evaluate the benefits and harms of prebiotics for improving health outcomes in children and adults with CF.
Collapse
Affiliation(s)
| | - Neil Williams
- Exercise and Health Research Group, Sport Health and Performance Enhancement (SHAPE) Research CentreSchool of Science and Technology, Nottingham Trent UniversityNottinghamUK
| | | | - Andrew P Prayle
- Department of Child Health, School of Clinical SciencesUniversity of NottinghamNottinghamUK
| | - Sarah J Nevitt
- Department of Health Data ScienceUniversity of LiverpoolLiverpoolUK
| | - Alan R Smyth
- Division of Child Health, Obstetrics & Gynaecology (COG)School of Medicine, University of NottinghamNottinghamUK
| |
Collapse
|
25
|
Wrigley-Carr HE, van Dorst JM, Ooi CY. Intestinal dysbiosis and inflammation in cystic fibrosis impacts gut and multi-organ axes. MEDICINE IN MICROECOLOGY 2022; 13:100057. [DOI: 10.1016/j.medmic.2022.100057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
26
|
Esposito S, Testa I, Mariotti Zani E, Cunico D, Torelli L, Grandinetti R, Fainardi V, Pisi G, Principi N. Probiotics Administration in Cystic Fibrosis: What Is the Evidence? Nutrients 2022; 14:3160. [PMID: 35956335 PMCID: PMC9370594 DOI: 10.3390/nu14153160] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 12/25/2022] Open
Abstract
In the last 20 years, gut microbiota in patients with cystic fibrosis (CF) has become an object of interest. It was shown that these patients had gut dysbiosis and this could explain not only the intestinal manifestations of the disease but also part of those involving the respiratory tract. The acquisition of previously unknown information about the importance of some bacteria, i.e., those partially or totally disappeared in the gut of CF patients, in the regulation of the activity and function of the gut and the lung was the base to suggest the use of probiotics in CF patients. The main aim of this paper is to discuss the biological basis for probiotic administration to CF patients and which results could be expected. Literature analysis showed that CF intestinal dysbiosis depends on the same genetic mutations that condition the clinical picture of the diseases and is aggravated by a series of therapeutic interventions, such as dietary modifications, the use of antibiotics, and the administration of antacids. All this translates into a significant worsening of the structure and function of organs, including the lung and intestine, already deeply penalized by the genetic alterations of CF. Probiotics can intervene on dysbiosis, reducing the negative effects derived from it. However, the available data cannot be considered sufficient to indicate that these bacteria are essential elements of CF therapy. Further studies that take into account the still unsolved aspects on how to use probiotics are absolutely necessary.
Collapse
Affiliation(s)
- Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.M.Z.); (D.C.); (L.T.); (R.G.)
| | - Ilaria Testa
- Respiratory Unit, Great Ormond Street Hospital for Children, Foundation Trust, London WC1N 1LE, UK; (I.T.); (V.F.); (G.P.)
| | - Elena Mariotti Zani
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.M.Z.); (D.C.); (L.T.); (R.G.)
| | - Daniela Cunico
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.M.Z.); (D.C.); (L.T.); (R.G.)
| | - Lisa Torelli
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.M.Z.); (D.C.); (L.T.); (R.G.)
| | - Roberto Grandinetti
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.M.Z.); (D.C.); (L.T.); (R.G.)
| | - Valentina Fainardi
- Respiratory Unit, Great Ormond Street Hospital for Children, Foundation Trust, London WC1N 1LE, UK; (I.T.); (V.F.); (G.P.)
| | - Giovanna Pisi
- Respiratory Unit, Great Ormond Street Hospital for Children, Foundation Trust, London WC1N 1LE, UK; (I.T.); (V.F.); (G.P.)
| | | |
Collapse
|
27
|
Changing paradigms in the treatment of gastrointestinal complications of cystic fibrosis in the era of cystic fibrosis transmembrane conductance regulator modulators. Paediatr Respir Rev 2022; 42:9-16. [PMID: 33485777 DOI: 10.1016/j.prrv.2020.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 12/17/2020] [Indexed: 12/17/2022]
Abstract
Cystic fibrosis (CF) - although primarily a lung disease - also causes a variety of gastrointestinal manifestations which are important for diagnosis, prognosis and quality of life. All parts of the gastrointestinal tract can be affected by CF. Besides the well-known pancreatic insufficiency, gastroesophageal reflux disease, liver disease and diseases of the large intestine are important pathologies that impact on prognosis and also impair quality of life. Diagnosis and management of gastrointestinal manifestations will be discussed in this review. Since optimisation of CF therapy is associated with a significantly longer life-span of CF patients nowadays, also gastrointestinal malignancies, which are more common in CF than in the non-CF population need to be considered. Furthermore, novel evidence on the role of the gut microbiome in CF is emerging. The introduction of cystic fibrosis transmembrane conductance regulator (CFTR) protein modulators gives hope for symptom alleviation and even cure of gastrointestinal manifestations of CF.
Collapse
|
28
|
Alagbaoso CA, Mizuno M. LENTINULA EDODES POLYSACCHARIDES SUPPRESSED PRO-INFLAMMATORY CYTOKINES EXPRESSION AND COLITIS IN MICE. ARQUIVOS DE GASTROENTEROLOGIA 2022; 59:288-295. [PMID: 35830043 DOI: 10.1590/s0004-2803.202202000-51] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/28/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Polysaccharides from edible mushrooms possess immunomodulatory, anti-inflammatory, and anti-tumor activities. Recent studies indicated that necroptosis plays a role in the pathogenesis of inflammatory diseases and mediates increased expression of inflammatory cytokines. OBJECTIVE Therefore, it is imperative to determine the impact of polysaccharide extract from Lentinula edodes (L. edodes) on inflammatory cytokines in experimental model of colitis in mice. METHODS Female C57BL/6 mice divided into three or four mice per group were used for this study. Polysaccharide sample was orally administered to mice prior to (7 days) and during colitis induction with 2.5% dextran sodium sulfate (7 days), followed by additional 3 days of administration. Changes in body weight and colon length were used as markers for colitis, and pro-inflammatory cytokines and tumor necrosis factor receptor 1 (TNFR1) expressions, as well as necroptosis were analyzed in the colon of colitis mice. Data obtained were analysed by Tukey-Kramer and two-tailed standard t tests. RESULTS The results indicated that the polysaccharide sample suppressed colitis in mice using effects on the body weight and colon length as markers. Also, it was demonstrated that necrostatin-1, a specific inhibitor of necroptosis, suppressed the expression of interleukin (IL)-8, a pro-inflammatory chemokine, in Caco-2 cells induced necroptosis induced by zVAD and TNF-α, an indication that necroptosis may be involved in the expression of pro-inflammatory cytokines. Moreover, the polysaccharide sample suppressed the expression of pro-inflammatory cytokines such as tumor necrosis factor (TNF)-α, IL-6, IL-1β, and interferon (IFN)-γ in the colon of mice. CONCLUSION These results suggested that the suppressive effects of the polysaccharide sample on inflammatory cytokines expression may contribute to its anti-colitis effect, and so may serve as a potent therapeutic agent against inflammatory bowel disease.
Collapse
Affiliation(s)
| | - Masashi Mizuno
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Japan
| |
Collapse
|
29
|
Tam RY, van Dorst JM, McKay I, Coffey M, Ooi CY. Intestinal Inflammation and Alterations in the Gut Microbiota in Cystic Fibrosis: A Review of the Current Evidence, Pathophysiology and Future Directions. J Clin Med 2022; 11:649. [PMID: 35160099 PMCID: PMC8836727 DOI: 10.3390/jcm11030649] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/22/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a life-limiting autosomal recessive multisystem disease. While its burden of morbidity and mortality is classically associated with pulmonary disease, CF also profoundly affects the gastrointestinal (GI) tract. Chronic low-grade inflammation and alterations to the gut microbiota are hallmarks of the CF intestine. The etiology of these manifestations is likely multifactorial, resulting from cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction, a high-fat CF diet, and the use of antibiotics. There may also be a bidirectional pathophysiological link between intestinal inflammation and changes to the gut microbiome. Additionally, a growing body of evidence suggests that these GI manifestations may have significant clinical associations with growth and nutrition, quality of life, and respiratory function in CF. As such, the potential utility of GI therapies and long-term GI outcomes are areas of interest in CF. Further research involving microbial modulation and multi-omics techniques may reveal novel insights. This article provides an overview of the current evidence, pathophysiology, and future research and therapeutic considerations pertaining to intestinal inflammation and alterations in the gut microbiota in CF.
Collapse
Affiliation(s)
- Rachel Y. Tam
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
| | - Josie M. van Dorst
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
| | - Isabelle McKay
- Wagga Wagga Base Hospital, Wagga Wagga, NSW 2650, Australia;
| | - Michael Coffey
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney, NSW 2031, Australia
| | - Chee Y. Ooi
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney, NSW 2031, Australia
| |
Collapse
|
30
|
van Dorst JM, Tam RY, Ooi CY. What Do We Know about the Microbiome in Cystic Fibrosis? Is There a Role for Probiotics and Prebiotics? Nutrients 2022; 14:480. [PMID: 35276841 PMCID: PMC8840103 DOI: 10.3390/nu14030480] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a life-shortening genetic disorder that affects the cystic fibrosis transmembrane conductance regulator (CFTR) protein. In the gastrointestinal (GI) tract, CFTR dysfunction results in low intestinal pH, thick and inspissated mucus, a lack of endogenous pancreatic enzymes, and reduced motility. These mechanisms, combined with antibiotic therapies, drive GI inflammation and significant alteration of the GI microbiota (dysbiosis). Dysbiosis and inflammation are key factors in systemic inflammation and GI complications including malignancy. The following review examines the potential for probiotic and prebiotic therapies to provide clinical benefits through modulation of the microbiome. Evidence from randomised control trials suggest probiotics are likely to improve GI inflammation and reduce the incidence of CF pulmonary exacerbations. However, the highly variable, low-quality data is a barrier to the implementation of probiotics into routine CF care. Epidemiological studies and clinical trials support the potential of dietary fibre and prebiotic supplements to beneficially modulate the microbiome in gastrointestinal conditions. To date, limited evidence is available on their safety and efficacy in CF. Variable responses to probiotics and prebiotics highlight the need for personalised approaches that consider an individual's underlying microbiota, diet, and existing medications against the backdrop of the complex nutritional needs in CF.
Collapse
Affiliation(s)
- Josie M. van Dorst
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, UNSW, Sydney 2031, Australia; (J.M.v.D.); (R.Y.T.)
| | - Rachel Y. Tam
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, UNSW, Sydney 2031, Australia; (J.M.v.D.); (R.Y.T.)
| | - Chee Y. Ooi
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, UNSW, Sydney 2031, Australia; (J.M.v.D.); (R.Y.T.)
- Molecular and Integrative Cystic Fibrosis (miCF) Research Centre, Sydney 2031, Australia
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney 2031, Australia
| |
Collapse
|
31
|
Talebi S, Day AS, Khadem Rezaiyan M, Ranjbar G, Zarei M, Safarian M, Kianifar HR. Fecal Calprotectin and Phenotype Severity in Patients with Cystic Fibrosis: A Systematic Review and Meta-Analysis. Pediatr Gastroenterol Hepatol Nutr 2022; 25:1-12. [PMID: 35087728 PMCID: PMC8762598 DOI: 10.5223/pghn.2022.25.1.1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/11/2021] [Indexed: 12/27/2022] Open
Abstract
Inflammation plays an important role in the outcome of patients with cystic fibrosis (CF). It may develop due to cystic fibrosis transmembrane conductance regulator protein dysfunction, pancreatic insufficiency, or prolonged pulmonary infection. Fecal calprotectin (FC) has been used as a noninvasive method to detect inflammation. Therefore, the aim of the current meta-analysis was to investigate the relationship between FC and phenotype severity in patients with CF. In this study, searches were conducted in PubMed, Science Direct, Scopus, and Embase databases up to August 2021 using terms such as "cystic fibrosis," "intestine," "calprotectin," and "inflammation." Only articles published in English and human studies were selected. The primary outcome was the level of FC in patients with CF. The secondary outcome was the relationship between FC and clinical severity. Statistical analysis was performed using Comprehensive Meta-Analysis software. Of the initial 303 references, only six articles met the inclusion criteria. The mean (95% confidence interval [CI]) level of FC was 256.5 mg/dL (114.1-398.9). FC levels were significantly associated with pancreatic insufficiency (mean, 243.02; 95% CI, 74.3 to 411.6; p=0.005; I2=0), pulmonary function (r=-0.39; 95% CI, -0.58 to -0.15; p=0.002; I2=60%), body mass index (r=-0.514; 95% CI, 0.26 to 0.69; p<0.001; I2=0%), and Pseudomonas colonization (mean, 174.77; 95% CI, 12.5 to 337.02; p=0.035; I2=71%). While FC is a reliable noninvasive marker for detecting gastrointestinal inflammation, it is also correlated with the severity of the disease in patients with CF.
Collapse
Affiliation(s)
- Saeedeh Talebi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Andrew S. Day
- Department of Pediatrics, University of Otago (Christchurch), Christchurch, New Zealand
| | - Majid Khadem Rezaiyan
- Department of Community Medicine, Clinical Research Development Unit of Akbar Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Golnaz Ranjbar
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mitra Zarei
- Department of Knowledge and Information Science, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahammad Safarian
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Kianifar
- Department of Pediatrics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
32
|
Intestinal function and transit associate with gut microbiota dysbiosis in cystic fibrosis. J Cyst Fibros 2021; 21:506-513. [PMID: 34895838 DOI: 10.1016/j.jcf.2021.11.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/04/2021] [Accepted: 11/23/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Most people with cystic fibrosis (pwCF) suffer from gastrointestinal symptoms and are at risk of gut complications. Gut microbiota dysbiosis is apparent within the CF population across all age groups, with evidence linking dysbiosis to intestinal inflammation and other markers of health. This pilot study aimed to investigate the potential relationships between the gut microbiota and gastrointestinal physiology, transit, and health. STUDY DESIGN Faecal samples from 10 pwCF and matched controls were subject to 16S rRNA sequencing. Results were combined with clinical metadata and MRI metrics of gut function to investigate relationships. RESULTS pwCF had significantly reduced microbiota diversity compared to controls. Microbiota compositions were significantly different, suggesting remodelling of core and rarer satellite taxa in CF. Dissimilarity between groups was driven by a variety of taxa, including Escherichia coli, Bacteroides spp., Clostridium spp., and Faecalibacterium prausnitzii. The core taxa were explained primarily by CF disease, whilst the satellite taxa were associated with pulmonary antibiotic usage, CF disease, and gut function metrics. Species-specific ordination biplots revealed relationships between taxa and the clinical or MRI-based variables observed. CONCLUSIONS Alterations in gut function and transit resultant of CF disease are associated with the gut microbiota composition, notably the satellite taxa. Delayed transit in the small intestine might allow for the expansion of satellite taxa resulting in potential downstream consequences for core community function in the colon.
Collapse
|
33
|
Abstract
Cystic fibrosis (CF) is a heritable, multiorgan disease that impacts all tissues that normally express cystic fibrosis transmembrane conductance regulator (CFTR) protein. While the importance of the airway microbiota has long been recognized, the intestinal microbiota has only recently been recognized as an important player in both intestinal and lung health outcomes for persons with CF (pwCF). Here, we summarize current literature related to the gut-lung axis in CF, with a particular focus on three key ideas: (i) mechanisms through which microbes influence the gut-lung axis, (ii) drivers of microbiota alterations, and (iii) the potential for intestinal microbiota remediation.
Collapse
Affiliation(s)
- Courtney E. Price
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover New Hampshire, USA
| | - George A. O’Toole
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover New Hampshire, USA
| |
Collapse
|
34
|
Coffey MJ, Garg M, Homaira N, Jaffe A, Ooi CY. A systematic cochrane review of probiotics for people with cystic fibrosis. Paediatr Respir Rev 2021; 39:61-64. [PMID: 32917517 DOI: 10.1016/j.prrv.2020.06.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Michael J Coffey
- School of Women's and Children's Health, University of New South Wales, Sydney, Australia; Junior Medical Officers Department, Sydney Children's Hospital, Sydney, Australia.
| | - Millie Garg
- School of Women's and Children's Health, University of New South Wales, Sydney, Australia
| | - Nusrat Homaira
- School of Women's and Children's Health, University of New South Wales, Sydney, Australia; Respiratory Department, Sydney Children's Hospital, Sydney, Australia
| | - Adam Jaffe
- School of Women's and Children's Health, University of New South Wales, Sydney, Australia; Respiratory Department, Sydney Children's Hospital, Sydney, Australia
| | - Chee Y Ooi
- School of Women's and Children's Health, University of New South Wales, Sydney, Australia; Gastroenterology Department, Sydney Children's Hospital, Sydney, Australia
| |
Collapse
|
35
|
Thavamani A, Salem I, Sferra TJ, Sankararaman S. Impact of Altered Gut Microbiota and Its Metabolites in Cystic Fibrosis. Metabolites 2021; 11:123. [PMID: 33671639 PMCID: PMC7926988 DOI: 10.3390/metabo11020123] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 12/14/2022] Open
Abstract
Cystic fibrosis (CF) is the most common lethal, multisystemic genetic disorder in Caucasians. Mutations in the gene encoding the cystic fibrosis transmembrane regulator (CFTR) protein are responsible for impairment of epithelial anionic transport, leading to impaired fluid regulation and pH imbalance across multiple organs. Gastrointestinal (GI) manifestations in CF may begin in utero and continue throughout the life, resulting in a chronic state of an altered intestinal milieu. Inherent dysfunction of CFTR leads to dysbiosis of the gut. This state of dysbiosis is further perpetuated by acquired factors such as use of antibiotics for recurrent pulmonary exacerbations. Since the gastrointestinal microbiome and their metabolites play a vital role in nutrition, metabolic, inflammatory, and immune functions, the gut dysbiosis will in turn impact various manifestations of CF-both GI and extra-GI. This review focuses on the consequences of gut dysbiosis and its metabolic implications on CF disease and possible ways to restore homeostasis.
Collapse
Affiliation(s)
- Aravind Thavamani
- Department of Pediatrics, Division of Pediatric Gastroenterology, UH Rainbow Babies & Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (A.T.); (T.J.S.)
| | - Iman Salem
- Center for Medial Mycology, Case Western Reserve University School of Medicine, UH Cleveland Medical Center, Cleveland, OH 44106, USA;
| | - Thomas J. Sferra
- Department of Pediatrics, Division of Pediatric Gastroenterology, UH Rainbow Babies & Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (A.T.); (T.J.S.)
| | - Senthilkumar Sankararaman
- Department of Pediatrics, Division of Pediatric Gastroenterology, UH Rainbow Babies & Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (A.T.); (T.J.S.)
| |
Collapse
|
36
|
Deane J, Fouhy F, Ronan NJ, Daly M, Fleming C, Eustace JA, Shanahan F, Flanagan ET, Dupont L, Harrison MJ, Haworth CS, Floto A, Rea MC, Ross RP, Stanton C, Plant BJ. A multicentre analysis of Clostridium difficile in persons with Cystic Fibrosis demonstrates that carriage may be transient and highly variable with respect to strain and level. J Infect 2021; 82:363-370. [PMID: 33444699 DOI: 10.1016/j.jinf.2020.12.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 12/15/2020] [Accepted: 12/28/2020] [Indexed: 01/16/2023]
Abstract
PURPOSE Clostridium difficile has been reported to occur in the gastrointestinal tract of 50% of Cystic Fibrosis (CF) subjects, however, clinical C. difficile infection (CDI) is a rare occurrence in this cohort despite the presence of toxigenic and hypervirulent ribotypes. Here, we present the first longitudinal, multicentre analysis of C. difficile prevalence among adult CF subjects. METHODOLOGY Faecal samples were collected from adults with CF (selected based on confirmed Pseudomonas aeruginosa pulmonary colonisation) from Ireland, UK and Belgium as part of the CFMATTERS clinical research trial (grant No. 603038) and from non-CF controls. Faecal samples were collected on enrolment, at three monthly intervals, during pulmonary exacerbation and three months post exacerbation. C. difficile was isolated from faecal samples by ethanol shocking followed by culturing on cycloserine cefoxitin egg yolk agar. Isolates were characterised in terms of ribotype, toxin type and antibiotic susceptibility to antibiotics routinely used in the treatment of CDI (metronidazole and vancomycin) and those implicated in induction of CDI (ciprofloxacin and moxifloxacin). RESULTS Prevalence of C. difficile among CF subjects in the three sites was similar ranging from 47% to 50% at baseline, while the healthy control cohort had a carriage rate of 7.1%. Including subjects who were positive for C. difficile at any time point there was a higher carriage rate of 71.4%, 66.7% and 63.2% in Ireland, UK, and Belgium, respectively. Ribotyping of 80 isolates from 45 CF persons, over multiple time points revealed 23 distinct ribotypes with two ribotypes (046 and 078) shared by all centres. The proportion of toxigenic isolates varied across the sites, ranging from 66.7% in Ireland to 52.9% in Belgium and 100% in the UK. Antibiotic susceptibility rates to vancomycin, metronidazole, ciprofloxacin and moxifloxacin was 100%, 97.5%, 1.3% and 63.8%, respectively. CONCLUSIONS This study demonstrates the highest carriage rate of C. difficile to date in a CF cohort. Longitudinal data show that C. difficile can be a transient inhabitant of the CF gut, changing both in terms of strain and excretion rates.
Collapse
Affiliation(s)
- Jennifer Deane
- Teagasc Food Research Centre, Moorepark, Fermoy, Co., Cork, Ireland; HRB Clinical Research Facility, University College Cork, Cork, Ireland; School of Microbiology, University College Cork, Cork, Ireland
| | - Fiona Fouhy
- Teagasc Food Research Centre, Moorepark, Fermoy, Co., Cork, Ireland; APC Microbiome Ireland, Cork, Ireland
| | - Nicola J Ronan
- Cork Adult Cystic Fibrosis Centre, University College Cork, Cork University Hospital, Wilton, Cork, Ireland
| | - Mary Daly
- Cork Adult Cystic Fibrosis Centre, University College Cork, Cork University Hospital, Wilton, Cork, Ireland
| | - Claire Fleming
- Cork Adult Cystic Fibrosis Centre, University College Cork, Cork University Hospital, Wilton, Cork, Ireland
| | - Joseph A Eustace
- HRB Clinical Research Facility, University College Cork, Cork, Ireland
| | | | - Evelyn T Flanagan
- Cork Adult Cystic Fibrosis Centre, University College Cork, Cork University Hospital, Wilton, Cork, Ireland
| | | | - Michael J Harrison
- Cambridge Centre for Lung Infection, Papworth Hospital, Cambridge United Kingdom
| | - Charles S Haworth
- Cambridge Centre for Lung Infection, Papworth Hospital, Cambridge United Kingdom
| | - Andres Floto
- Cambridge Centre for Lung Infection, Papworth Hospital, Cambridge United Kingdom; Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Mary C Rea
- Teagasc Food Research Centre, Moorepark, Fermoy, Co., Cork, Ireland; APC Microbiome Ireland, Cork, Ireland
| | | | | | - Barry J Plant
- Cork Adult Cystic Fibrosis Centre, University College Cork, Cork University Hospital, Wilton, Cork, Ireland.
| |
Collapse
|
37
|
Beaufils F, Mas E, Mittaine M, Addra M, Fayon M, Delhaes L, Clouzeau H, Galode F, Lamireau T, Bui S, Enaud R. Increased Fecal Calprotectin Is Associated with Worse Gastrointestinal Symptoms and Quality of Life Scores in Children with Cystic Fibrosis. J Clin Med 2020; 9:4080. [PMID: 33348735 PMCID: PMC7766355 DOI: 10.3390/jcm9124080] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 02/06/2023] Open
Abstract
In cystic fibrosis (CF), cystic fibrosis transmembrane regulator (CFTR) dysfunction leads to digestive disorders that promote intestinal inflammation and dysbiosis enhancing gastrointestinal symptoms. In pancreatic insufficiency CF patients, both intestinal inflammation and dysbiosis, are associated with an increase in the fecal calprotectin (FC) level. However, associations between the FC level, gastrointestinal symptoms, and quality of life (QoL) remain poorly studied. We aimed to assess such associations in pancreatic insufficiency CF children. The FC level was measured in pancreatic insufficiency CF children's stool samples. Children and their parents completed two questionnaires: The Gastrointestinal Symptoms Scales 3.0-PedsQLTM and the Quality of Life Pediatric Inventory 4.0-PedsQLTM. Lower scores indicated worse symptomatology or QoL. Thirty-seven CF children were included. A FC level above 250 µg/g was associated with worse gastrointestinal symptoms and QoL scores. The FC level was inversely correlated with several gastrointestinal scores assessed by children (i.e., Total, "Heart Burn Reflux", "Nausea and Vomiting", and "Gas and Bloating"). Several QoL scores were correlated with gastrointestinal scores. The FC level was weakly associated with clinical parameters. Some gastrointestinal and QoL scores were related to disease severity associated parameters. In CF, the FC level, biomarker previously related to intestinal inflammation and dysbiosis, was associated with worse digestive symptoms and QoL scores.
Collapse
Affiliation(s)
- Fabien Beaufils
- CHU Bordeaux, CRCM Pédiatrique, CIC 1401, Place Amélie Raba Léon, F-33000 Bordeaux, France; (M.F.); (L.D.); (H.C.); (F.G.); (T.L.); (S.B.); (R.E.)
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France;
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France
| | - Emmanuel Mas
- CHU Toulouse, CRCM Pédiatrique, F-31300 Toulouse, France; (E.M.); (M.M.)
- INSERM, INRA, ENVT, Université de Toulouse, UPS, F-31000 Toulouse, France
- Unité de Gastroentérologie, Hépatologie, Nutrition, Diabétologie et Maladies Héréditaires du Métabolisme, Hôpital des Enfants, CHU de Toulouse, F-31300 Toulouse, France
| | - Marie Mittaine
- CHU Toulouse, CRCM Pédiatrique, F-31300 Toulouse, France; (E.M.); (M.M.)
| | - Martin Addra
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France;
| | - Michael Fayon
- CHU Bordeaux, CRCM Pédiatrique, CIC 1401, Place Amélie Raba Léon, F-33000 Bordeaux, France; (M.F.); (L.D.); (H.C.); (F.G.); (T.L.); (S.B.); (R.E.)
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France;
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France
| | - Laurence Delhaes
- CHU Bordeaux, CRCM Pédiatrique, CIC 1401, Place Amélie Raba Léon, F-33000 Bordeaux, France; (M.F.); (L.D.); (H.C.); (F.G.); (T.L.); (S.B.); (R.E.)
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France
- CHU Toulouse, CRCM Pédiatrique, F-31300 Toulouse, France; (E.M.); (M.M.)
- CHU Bordeaux, Service de Parasitologie-Mycologie, F-33000 Bordeaux, France
| | - Haude Clouzeau
- CHU Bordeaux, CRCM Pédiatrique, CIC 1401, Place Amélie Raba Léon, F-33000 Bordeaux, France; (M.F.); (L.D.); (H.C.); (F.G.); (T.L.); (S.B.); (R.E.)
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France
| | - François Galode
- CHU Bordeaux, CRCM Pédiatrique, CIC 1401, Place Amélie Raba Léon, F-33000 Bordeaux, France; (M.F.); (L.D.); (H.C.); (F.G.); (T.L.); (S.B.); (R.E.)
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France
| | - Thierry Lamireau
- CHU Bordeaux, CRCM Pédiatrique, CIC 1401, Place Amélie Raba Léon, F-33000 Bordeaux, France; (M.F.); (L.D.); (H.C.); (F.G.); (T.L.); (S.B.); (R.E.)
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France
| | - Stéphanie Bui
- CHU Bordeaux, CRCM Pédiatrique, CIC 1401, Place Amélie Raba Léon, F-33000 Bordeaux, France; (M.F.); (L.D.); (H.C.); (F.G.); (T.L.); (S.B.); (R.E.)
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France;
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France
| | - Raphaël Enaud
- CHU Bordeaux, CRCM Pédiatrique, CIC 1401, Place Amélie Raba Léon, F-33000 Bordeaux, France; (M.F.); (L.D.); (H.C.); (F.G.); (T.L.); (S.B.); (R.E.)
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France;
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France
| |
Collapse
|
38
|
Loman BR, Shrestha CL, Thompson R, Groner JA, Mejias A, Ruoff KL, O'Toole GA, Bailey MT, Kopp BT. Age and environmental exposures influence the fecal bacteriome of young children with cystic fibrosis. Pediatr Pulmonol 2020; 55:1661-1670. [PMID: 32275127 PMCID: PMC7593804 DOI: 10.1002/ppul.24766] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/23/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Mechanisms that facilitate early infection and inflammation in cystic fibrosis (CF) are unclear. We previously showed that young CF children with secondhand smoke exposure (SHSe) have increased susceptibility to respiratory infections. We aimed to define the impact of SHSe and other external factors upon the fecal bacteriome in early CF. METHODS Twenty CF infants and children were enrolled, clinical data recorded, and hair nicotine measured as an objective surrogate of SHSe. Fecal samples were collected at clinic visits and bacteriome 16S rRNA gene sequencing performed. RESULTS SHSe was associated with increased alpha diversity and increased relative abundance of Acinetobacter and Akkermansia, along with decreased Bifidobacterium and Lactobacillus. Recent antibiotic exposure predicted bacterial population structure in children less than 2 years of age and was associated with decreased Bacteroides relative abundance. Age was the strongest predictor of overall fecal bacterial composition and positively associated with Blautia and Parabacteroides. Weight for length was negatively associated with Staphylococcus relative abundance. CONCLUSIONS SHSe and other external factors such as antibiotics appear to alter fecal bacterial composition in young CF children, but the strongest predictor of overall composition was age. These findings have implications for understanding the intestinal microbiome in young CF children.
Collapse
Affiliation(s)
- Brett R Loman
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Center for Microbial Pathogenesis, Columbus, Ohio
| | - Chandra L Shrestha
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Center for Microbial Pathogenesis, Columbus, Ohio
| | - Rohan Thompson
- Division of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Judith A Groner
- Division of Primary Care, Nationwide Children's Hospital, Columbus, Ohio
| | - Asuncion Mejias
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Center for Vaccines and Immunity, Columbus, Ohio.,Division of Infectious Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Kathryn L Ruoff
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - George A O'Toole
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Michael T Bailey
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Center for Microbial Pathogenesis, Columbus, Ohio
| | - Benjamin T Kopp
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Center for Microbial Pathogenesis, Columbus, Ohio.,Division of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, Ohio
| |
Collapse
|
39
|
Kristensen M, Prevaes SMPJ, Kalkman G, Tramper-Stranders GA, Hasrat R, de Winter-de Groot KM, Janssens HM, Tiddens HA, van Westreenen M, Sanders EAM, Arets B, Keijser B, van der Ent CK, Bogaert D. Development of the gut microbiota in early life: The impact of cystic fibrosis and antibiotic treatment. J Cyst Fibros 2020; 19:553-561. [PMID: 32487494 DOI: 10.1016/j.jcf.2020.04.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 03/25/2020] [Accepted: 04/21/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Patients with Cystic Fibrosis (CF) suffer from pancreatic insufficiency, lipid malabsorption and gastrointestinal complaints, next to progressive pulmonary disease. Altered mucosal homoeostasis due to malfunctioning chloride channels results in an adapted microbial composition of the gastrointestinal and the respiratory tract. Additionally, antibiotic treatment has the potential to distort resident microbial communities dramatically. This study aims to investigate early life development of the gut microbial community composition of children with CF compared to healthy infants and to study the independent effects of antibiotics taking into account other clinical and lifestyle factors. STUDY DESIGN Faecal samples from 20 infants with CF and 45 healthy infants were collected regularly during the first 18 months of life and microbial composition was determined using 16S rRNA based sequencing. RESULTS We observed significant differences in the overall microbiota composition between infants with CF and healthy infants (p<0.001). Akkermansia and Anaerostipes were significantly more abundant in control infants, whereas Streptococci and E. coli were significantly more abundant in infants with CF, also after correction for several clinical factors (p<0.05). Antibiotic use in infants with CF was associated with a lower alpha diversity, a reduced abundance of Bifidobacterium and Bacteroides, and a higher abundance of Enterococcus. CONCLUSION Microbial development of the gut is different in infants with CF compared to healthy infants from the first months of life on, and further deviates over time, in part as a result of antibiotic treatment. The resulting dysbiosis may have significant functional consequences for the microbial ecosystem in CF patients.
Collapse
Affiliation(s)
- Maartje Kristensen
- Department of Pediatric pulmonology, Wilhelmina Children's Hospital, University Medical Center Utrecht, the Netherlands.
| | - Sabine M P J Prevaes
- Department of Pediatric pulmonology, Wilhelmina Children's Hospital, University Medical Center Utrecht, the Netherlands
| | - Gino Kalkman
- Microbiology and Systems Biology, TNO, Zeist, the Netherlands
| | | | - Raiza Hasrat
- Department of Pediatric infectious diseases and immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, the Netherlands
| | - Karin M de Winter-de Groot
- Department of Pediatric pulmonology, Wilhelmina Children's Hospital, University Medical Center Utrecht, the Netherlands
| | - Hettie M Janssens
- Department of Pediatric Pulmonology and Allergology, Sophia Children's Hospital, Erasmus University Medical Center, the Netherlands
| | - Harm A Tiddens
- Department of Pediatric Pulmonology and Allergology, Sophia Children's Hospital, Erasmus University Medical Center, the Netherlands
| | - Mireille van Westreenen
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, the Netherlands
| | - Elisabeth A M Sanders
- Department of Pediatric infectious diseases and immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, the Netherlands; Rijksinstituut voor Volksgezondheid en Milieu, Bilthoven, the Netherlands
| | - Bert Arets
- Department of Pediatric pulmonology, Wilhelmina Children's Hospital, University Medical Center Utrecht, the Netherlands
| | - Bart Keijser
- Microbiology and Systems Biology, TNO, Zeist, the Netherlands
| | - Cornelis K van der Ent
- Department of Pediatric pulmonology, Wilhelmina Children's Hospital, University Medical Center Utrecht, the Netherlands
| | - Debby Bogaert
- Department of Pediatric infectious diseases and immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, the Netherlands; The Queen's Medical Research Institute, University of Edinburgh, United Kingdom.
| |
Collapse
|
40
|
The intestinal virome in children with cystic fibrosis differs from healthy controls. PLoS One 2020; 15:e0233557. [PMID: 32442222 PMCID: PMC7244107 DOI: 10.1371/journal.pone.0233557] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/07/2020] [Indexed: 02/06/2023] Open
Abstract
Intestinal bacterial dysbiosis is evident in children with cystic fibrosis (CF) and intestinal viruses may be contributory, given their influence on bacterial species diversity and biochemical cycles. We performed a prospective, case-control study on children with CF and age and gender matched healthy controls (HC), to investigate the composition and function of intestinal viral communities. Stool samples were enriched for viral DNA and RNA by viral extraction, random amplification and purification before sequencing (Illumina MiSeq). Taxonomic assignment of viruses was performed using Vipie. Functional annotation was performed using Virsorter. Inflammation was measured by calprotectin and M2-pyruvate kinase (M2-PK). Eight CF and eight HC subjects were included (50% male, mean age 6.9 ± 3.0 and 6.4 ± 5.3 years, respectively, p = 0.8). All CF subjects were pancreatic insufficient. Regarding the intestinal virome, no difference in Shannon index between CF and HC was identified. Taxonomy-based beta-diversity (presence-absence Bray-Curtis dissimilarity) was significantly different between CF and HC (R2 = 0.12, p = 0.001). Myoviridae, Faecalibacterium phage FP Taranis and unclassified Gokushovirinae were significantly decreased in CF compared with HC (q<0.05). In children with CF (compared to HC), the relative abundance of genes annotated to (i) a peptidoglycan-binding domain of the peptidoglycan hydrolases (COG3409) was significantly increased (q<0.05) and (ii) capsid protein (F protein) (PF02305.16) was significantly decreased (q<0.05). Picornavirales, Picornaviridae, and Enterovirus were found to positively correlate with weight and BMI (r = 0.84, q = 0.01). Single-stranded DNA viruses negatively correlated with M2-PK (r = -0.86, q = 0.048). Children with CF have an altered intestinal virome compared to well-matched HC, with both taxonomic and predicted functional changes. Further exploration of Faecalibacterium phages, Gokushovirinae and phage lysins are warranted. Intestinal viruses and their functions may have important clinical implications for intestinal inflammation and growth in children with CF, potentially providing novel therapeutic targets.
Collapse
|
41
|
Isolation of Anti-Inflammatory and Epithelium Reinforcing Bacteroides and Parabacteroides Spp. from A Healthy Fecal Donor. Nutrients 2020; 12:nu12040935. [PMID: 32230951 PMCID: PMC7230855 DOI: 10.3390/nu12040935] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023] Open
Abstract
Altered intestinal microbiota is associated with systemic and intestinal diseases, such as inflammatory bowel disease (IBD). Dysbiotic microbiota with enhanced proinflammatory capacity is characterized by depletion of anaerobic commensals, increased proportion of facultatively anaerobic bacteria, as well as reduced diversity and stability. In this study, we developed a high-throughput in vitro screening assay to isolate intestinal commensal bacteria with anti-inflammatory capacity from a healthy fecal microbiota transplantation donor. Freshly isolated gut bacteria were screened for their capacity to attenuate Escherichia coli lipopolysaccharide (LPS)-induced interleukin 8 (IL-8) release from HT-29 cells. The screen yielded a number of Bacteroides and Parabacteroides isolates, which were identified as P. distasonis, B. caccae, B. intestinalis, B. uniformis, B. fragilis, B. vulgatus and B. ovatus using whole genome sequencing. We observed that a cell-cell contact with the epithelium was not necessary to alleviate in vitro inflammation as spent culture media from the isolates were also effective and the anti-inflammatory action did not correlate with the enterocyte adherence capacity of the isolates. The anti-inflammatory isolates also exerted enterocyte monolayer reinforcing action and lacked essential genes to synthetize hexa-acylated, proinflammatory lipid A, part of LPS. Yet, the anti-inflammatory effector molecules remain to be identified. The Bacteroides strains isolated and characterized in this study have potential to be used as so-called next-generation probiotics.
Collapse
|
42
|
Łoniewska B, Adamek K, Węgrzyn D, Kaczmarczyk M, Skonieczna-Żydecka K, Clark J, Adler G, Tousty J, Uzar I, Tousty P, Łoniewski I. Analysis of Faecal Zonulin and Calprotectin Concentrations in Healthy Children During the First Two Years of Life. An Observational Prospective Cohort Study. J Clin Med 2020; 9:jcm9030777. [PMID: 32178435 PMCID: PMC7141325 DOI: 10.3390/jcm9030777] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
Factors affecting the intestinal-barrier permeability of newborns, such as body mass index (BMI), nutrition and antibiotics, are assumed to affect intestinal-barrier permeability in the first two years of life. This study assessed 100 healthy, full-term newborns to 24 months old. Faecal zonulin/calprotectin concentrations were measured at 1, 6, 12, 24 months as gut-permeability markers. Zonulin concentrations increased between 1 and 12 months (medians: 114.41, 223.7 ng/mL; respectively), whereas calprotectin concentrations decreased between one and six months (medians: 149. 29, 109.28 µg/mL); both then stabilized (24 months: 256.9 ng/mL zonulin; 59.5 µg/mL calprotectin). In individual children, high levels at one month gave high levels at older ages (correlations: calprotectin: between 1 and 6 or 12 months: correlation coefficient (R) = 0.33, statistical significance (p) = 0.0095; R = 0.28, p = 0.032; zonulin: between 1 and 24 months: R = 0.32; p = 0.022, respectively). Parameters which gave marker increases: antibiotics during pregnancy (calprotectin; six months: by 80%, p = 0.038; 12 months: by 48%, p = 0.028); vaginal birth (calprotectin: 6 months: by 140%, p = 0.005); and > 5.7 pregnancy-BMI increase (zonulin: 12 months: by 74%, p = 0.049). Conclusions: “Closure of the intestines” is spread over time and begins between the sixth and twelfth month of life. Antibiotic therapy, BMI increase > 5.7 during pregnancy and vaginal birth are associated with increased intestinal permeability during the first two years of life. Stool zonulin and calprotectin concentrations were much higher compared with previous measurements at older ages; clinical interpretation and validation are needed (no health associations found).
Collapse
Affiliation(s)
- Beata Łoniewska
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin 70-111, Poland; (K.A.); (D.W.); (J.T.)
- Correspondence: ; Tel.: +48-(91)-466-1375
| | - Karolina Adamek
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin 70-111, Poland; (K.A.); (D.W.); (J.T.)
| | - Dagmara Węgrzyn
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin 70-111, Poland; (K.A.); (D.W.); (J.T.)
| | - Mariusz Kaczmarczyk
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin 70-111, Poland; (M.K.); (J.C.)
| | - Karolina Skonieczna-Żydecka
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, Szczecin 71-460, Poland; (K.S.-Ż.); (I.Ł.)
| | - Jeremy Clark
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin 70-111, Poland; (M.K.); (J.C.)
| | - Grażyna Adler
- Department of Studies in Anthropogenetics and Biogerontology, Pomeranian Medical University, Szczecin 71-210, Poland;
| | - Joanna Tousty
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin 70-111, Poland; (K.A.); (D.W.); (J.T.)
| | - Izabela Uzar
- Department of General Pharmacology and Pharmacoeconomics, Pomeranian Medical University, Szczecin 71-230, Poland;
| | - Piotr Tousty
- Department of Obstetrics and Gynecology, Pomeranian Medical University, Szczecin 70-111, Poland;
| | - Igor Łoniewski
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, Szczecin 71-460, Poland; (K.S.-Ż.); (I.Ł.)
| |
Collapse
|
43
|
Zhang D, Li S, Wang N, Tan HY, Zhang Z, Feng Y. The Cross-Talk Between Gut Microbiota and Lungs in Common Lung Diseases. Front Microbiol 2020; 11:301. [PMID: 32158441 PMCID: PMC7052046 DOI: 10.3389/fmicb.2020.00301] [Citation(s) in RCA: 262] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Emerging findings indicate there is a vital cross-talk between gut microbiota and the lungs, which is known as gut-lung axis. The gut disturbances in lung diseases including allergy, asthma, chronic obstructive pulmonary disease, cystic fibrosis and lung cancer were observed by extensive studies. Investigating how gut microbiota impact other distant organs is of great interest in recent years. Although it has not been fully understood whether the disturbance is the cause or effect of lung diseases, alterations in the gut microbial species and metabolites have been linked to changes in immune responses and inflammation as well as the disease development in the lungs. In this article, we systemically review the role and mechanisms underlying the changes in the constituent of gut microbiota and metabolites in lung diseases. In particular, the roles of gut-lung axis in mediating immune responses and reshaping inflammation are highlighted. Furthermore, we discuss the potential of strategies to manipulate the gut microbiota and metabolites as the therapeutic approach for lung diseases.
Collapse
Affiliation(s)
- Dapeng Zhang
- First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Sha Li
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ning Wang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hor-Yue Tan
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhimin Zhang
- First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yibin Feng
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
44
|
Dayama G, Priya S, Niccum DE, Khoruts A, Blekhman R. Interactions between the gut microbiome and host gene regulation in cystic fibrosis. Genome Med 2020; 12:12. [PMID: 31992345 PMCID: PMC6988342 DOI: 10.1186/s13073-020-0710-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cystic fibrosis is the most common autosomal recessive genetic disease in Caucasians. It is caused by mutations in the CFTR gene, leading to poor hydration of mucus and impairment of the respiratory, digestive, and reproductive organ functions. Advancements in medical care have led to markedly increased longevity of patients with cystic fibrosis, but new complications have emerged, such as early onset of colorectal cancer. Although the pathogenesis of colorectal cancer in cystic fibrosis remains unclear, altered host-microbe interactions might play a critical role. To investigate this, we characterized changes in the microbiome and host gene expression in the colonic mucosa of cystic fibrosis patients relative to healthy controls, and identified host gene-microbiome interactions in the colon of cystic fibrosis patients. METHODS We performed RNA-seq on colonic mucosa samples from cystic fibrosis patients and healthy controls to determine differentially expressed host genes. We also performed 16S rRNA sequencing to characterize the colonic mucosal microbiome and identify gut microbes that are differentially abundant between patients and healthy controls. Lastly, we modeled associations between relative abundances of specific bacterial taxa in the gut mucosa and host gene expression. RESULTS We find that 1543 genes, including CFTR, show differential expression in the colon of cystic fibrosis patients compared to healthy controls. These genes are enriched with functions related to gastrointestinal and colorectal cancer, such as metastasis of colorectal cancer, tumor suppression, p53, and mTOR signaling pathways. In addition, patients with cystic fibrosis show decreased gut microbial diversity, decreased abundance of butyrate producing bacteria, such as Ruminococcaceae and Butyricimonas, and increased abundance of other taxa, such as Actinobacteria and Clostridium. An integrative analysis identified colorectal cancer-related genes, including LCN2 and DUOX2, for which gene expression is correlated with the abundance of colorectal cancer-associated bacteria, such as Ruminococcaceae and Veillonella. CONCLUSIONS In addition to characterizing host gene expression and mucosal microbiome in cystic fibrosis patients, our study explored the potential role of host-microbe interactions in the etiology of colorectal cancer in cystic fibrosis. Our results provide biomarkers that may potentially serve as targets for stratifying risk of colorectal cancer in patients with cystic fibrosis.
Collapse
Affiliation(s)
- Gargi Dayama
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Sambhawa Priya
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - David E Niccum
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Alexander Khoruts
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
- Center for Immunology, BioTechnology Institute, University of Minnesota, Minneapolis, MN, USA.
| | - Ran Blekhman
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA.
- Department of Ecology, Evolution, and Behavior, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
45
|
Abstract
BACKGROUND Cystic fibrosis (CF) is a multisystem disease and the importance of growth and nutrition has been well established, given its implications for lung function and overall survival. It has been established that intestinal dysbiosis (i.e. microbial imbalance) and inflammation is present in people with CF. Probiotics are commercially available (over-the-counter) and may improve both intestinal and overall health. OBJECTIVES To assess the efficacy and safety of probiotics for improving health outcomes in children and adults with CF. SEARCH METHODS We searched the Cochrane Cystic Fibrosis Trials Register, compiled from electronic database searches and handsearching of journals and conference abstract books. Date of last register search: 20 January 2020. We also searched ongoing trials registries and the reference lists of relevant articles and reviews. Date of last search: 29 January 2019. SELECTION CRITERIA Randomised or quasi-randomised controlled trials (RCTs) assessing efficacies and safety of probiotics in children and adults with CF. Cross-over RCTs with a washout phase were included and for those without a washout period, only the first phase of each trial was analysed. DATA COLLECTION AND ANALYSIS We independently extracted data and assessed the risk of bias of the included trials; we used GRADE to assess the certainty of the evidence. We contacted trial authors for additional data. Meta-analyses were undertaken on outcomes at several time points. MAIN RESULTS We identified 17 trials and included 12 RCTs (11 completed and one trial protocol - this trial was terminated early) (464 participants). Eight trials included only children, whilst four trials included both children and adults. Trial duration ranged from one to 12 months. Nine trials compared a probiotic (seven single strain and three multistrain preparations) with a placebo preparation, two trials compared a synbiotic (multistrain) with a placebo preparation and one trial compared two probiotic preparations. Overall we judged the risk of bias in the 12 trials to be low. Three trials had a high risk of performance bias, two trials a high risk of attrition bias and six trials a high risk of reporting bias. Only two trials were judged to have low or unclear risk of bias for all domains. Four trials were sponsored by grants only, two trials by industry only, two trials by both grants and industry and three trials had an unknown funding source. Combined data from four trials (225 participants) suggested probiotics may reduce the number of pulmonary exacerbations during a four to 12 month time-frame, mean difference (MD) -0.32 episodes per participant (95% confidence interval (CI) -0.68 to 0.03; P = 0.07) (low-certainty evidence); however, the 95% CI includes the possibility of both an increased and a reduced number of exacerbations. Additionally, two trials (127 participants) found no evidence of an effect on the duration of antibiotic therapy during the same time period. Combined data from four trials (177 participants) demonstrated probiotics may reduce faecal calprotectin, MD -47.4 µg/g (95% CI -93.28 to -1.54; P = 0.04) (low-certainty evidence), but the results for other biomarkers mainly did not show any difference between probiotics and placebo. Two trials (91 participants) found no evidence of effect on height, weight or body mass index (low-certainty evidence). Combined data from five trials (284 participants) suggested there was no difference in lung function (forced expiratory volume at one second (FEV1) % predicted) during a three- to 12-month time frame, MD 1.36% (95% CI -1.20 to 3.91; P = 0.30) (low-certainty evidence). Combined data from two trials (115 participants) suggested there was no difference in hospitalisation rates during a three- to 12-month time frame, MD -0.44 admissions per participant (95% CI -1.41 to 0.54; P = 0.38) (low-certainty evidence). One trial (37 participants) reported health-related quality of life and while the parent report favoured probiotics, SMD 0.87 (95% CI 0.19 to 1.55) the child self-report did not identify any effect, SMD 0.59 (95% CI -0.07 to 1.26) (low-certainty evidence). There were limited results for gastrointestinal symptoms and intestinal microbial profile which were not analysable. Only four trials and one trial protocol (298 participants) reported adverse events as a priori hypotheses. No trials reported any deaths. One terminated trial (12 participants and available as a protocol only) reported a severe allergic reaction (severe urticaria) for one participant in the probiotic group. Two trials reported a single adverse event each (vomiting in one child and diarrhoea in one child). The estimated number needed to harm for any adverse reaction (serious or not) is 52 people (low-certainty evidence). AUTHORS' CONCLUSIONS Probiotics significantly reduce faecal calprotectin (a marker of intestinal inflammation) in children and adults with CF, however the clinical implications of this require further investigation. Probiotics may make little or no difference to pulmonary exacerbation rates, however, further evidence is required before firm conclusions can be made. Probiotics are associated with a small number of adverse events including vomiting, diarrhoea and allergic reactions. In children and adults with CF, probiotics may be considered by patients and their healthcare providers. Given the variability of probiotic composition and dosage, further adequately-powered multicentre RCTs of at least 12 months duration are required to best assess the efficacy and safety of probiotics for children and adults with CF.
Collapse
Affiliation(s)
- Michael J Coffey
- University of New South WalesSchool of Women's and Children's HealthLevel 8, Centre for Child Health Research & Innovation Bright Alliance Building Cnr Avoca & High StreetsRandwickSydneyNSWAustralia2031
- Sydney Children's HospitalJunior Medical Officers DepartmentHigh StreetRandwickSydneyNSWAustralia2031
| | - Millie Garg
- University of New South WalesSchool of Women's and Children's HealthLevel 8, Centre for Child Health Research & Innovation Bright Alliance Building Cnr Avoca & High StreetsRandwickSydneyNSWAustralia2031
| | - Nusrat Homaira
- University of New South WalesSchool of Women's and Children's HealthLevel 8, Centre for Child Health Research & Innovation Bright Alliance Building Cnr Avoca & High StreetsRandwickSydneyNSWAustralia2031
- Sydney Children's HospitalRespiratory DepartmentHigh StreetRandwickSydneyNew South WalesAustraliaNSW 2031
| | - Adam Jaffe
- University of New South WalesSchool of Women's and Children's HealthLevel 8, Centre for Child Health Research & Innovation Bright Alliance Building Cnr Avoca & High StreetsRandwickSydneyNSWAustralia2031
- Sydney Children's HospitalRespiratory DepartmentHigh StreetRandwickSydneyNew South WalesAustraliaNSW 2031
| | - Chee Y Ooi
- University of New South WalesSchool of Women's and Children's HealthLevel 8, Centre for Child Health Research & Innovation Bright Alliance Building Cnr Avoca & High StreetsRandwickSydneyNSWAustralia2031
- Sydney Children's HospitalGastroenterology DepartmentHigh StreetRandwickSydneyNSWAustralia2031
| | | |
Collapse
|
46
|
Altman K, McDonald CM, Michel SH, Maguiness K. Nutrition in cystic fibrosis: From the past to the present and into the future. Pediatr Pulmonol 2019; 54 Suppl 3:S56-S73. [PMID: 31715089 DOI: 10.1002/ppul.24521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022]
Abstract
Nutritional management is an integral part of multidisciplinary care for persons with cystic fibrosis. This review will look at how nutrition care has evolved over time. In addition, we will look at how some newer therapies impact nutrition care.
Collapse
Affiliation(s)
- Kimberly Altman
- Gunnar Esiason Adult Cystic Fibrosis and Lung Center, New York Presbyterian/Columbia University Medical Center, New York, New York, United States
| | | | - Suzanne H Michel
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Karen Maguiness
- Section of Pediatric Pulmonology, Allergy, and Sleep Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana, United States
| |
Collapse
|
47
|
Leblhuber F, Steiner K, Schuetz B, Fuchs D, Gostner JM. Probiotic Supplementation in Patients with Alzheimer's Dementia - An Explorative Intervention Study. Curr Alzheimer Res 2019; 15:1106-1113. [PMID: 30101706 PMCID: PMC6340155 DOI: 10.2174/1389200219666180813144834] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/17/2018] [Accepted: 08/07/2018] [Indexed: 12/15/2022]
Abstract
Background: Dysbiosis of intestinal microbiota in the elderly can cause a leaky gut, which may result in silent systemic inflammation and promote neuroinflammation - a relevant pathomechanism in the early course of Alzheimer’s disease. Objective: The rebalancing of the microbiome could benefically impact on gut inflammation and immune activation. Methods: In this study, routine laboratory tests in twenty outpatients (9 females, 11 males, aged 76.7 ± 9.6 years) with Alzheimer’s disease were investigated. The mean Mini Mental State Examination score was 18.5 ± 7.7. Biomarkers of immune activation – serum neopterin and tryptophan breakdown - as well as gut inflammation markers and microbiota composition in fecal specimens were analyzed in 18 patients be-fore and after probiotic supplementation for 4 weeks. Results: After treatment a decline of fecal zonulin concentrations and an increase in Faecalibacterium prausnitzii compared to baseline were observed. At the same time, serum kynurenine concentrations in-creased (p <0.05). Delta values (before - after) of neopterin and the kynurenine to tryptophan ratios (Kyn/Trp) correlated significantly (p <0.05). Conclusion: Results show that the supplementation of Alzheimer’s disease patients with a multispecies probiotic influences gut bacteria composition as well as tryptophan metabolism in serum. The correlation between Kyn/Trp and neopterin concentrations points to the activation of macrophages and/or dendritic cells. Further studies are warranted to dissect the potential consequences of Probiotic supplementation in the course of Alzheimer’s disease.
Collapse
Affiliation(s)
- Friedrich Leblhuber
- Department of Gerontology, Neuromed Campus, Kepler University Clinic, Linz, Austria
| | - Kostja Steiner
- Department of Gerontology, Neuromed Campus, Kepler University Clinic, Linz, Austria
| | | | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Johanna M Gostner
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
48
|
Altered Stool Microbiota of Infants with Cystic Fibrosis Shows a Reduction in Genera Associated with Immune Programming from Birth. J Bacteriol 2019; 201:JB.00274-19. [PMID: 31209076 DOI: 10.1128/jb.00274-19] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/24/2019] [Indexed: 12/28/2022] Open
Abstract
Previous work from our group indicated an association between the gastrointestinal microbiota of infants with cystic fibrosis (CF) and airway disease in this population. Here we report that stool microbiota of infants with CF demonstrates an altered but largely unchanging within-individual bacterial diversity (alpha diversity) over the first year of life, in contrast to the infants without CF (control cohort), which showed the expected increase in alpha diversity over the first year. The beta diversity, or between-sample diversity, of these two cohorts was significantly different over the first year of life and was statistically significantly associated with airway exacerbations, confirming our earlier findings. Compared with control infants, infants with CF had reduced levels of Bacteroides, a bacterial genus associated with immune modulation, as early as 6 weeks of life, and this significant reduction of Bacteroides spp. in the cohort with CF persisted over the entire first year of life. Only two other genera were significantly different across the first year of life: Roseburia was significantly reduced and Veillonella was significantly increased. Other genera showed differences between the two cohorts but only at selected time points. In vitro studies demonstrated that exposure of the apical face of polarized intestinal cell lines to Bacteroides species supernatants significantly reduced production of interleukin 8 (IL-8), suggesting a mechanism whereby changes in the intestinal microbiota could impact inflammation in CF. This work further establishes an association between gastrointestinal microbiota, inflammation, and airway disease in infants with CF and presents a potential opportunity for therapeutic interventions beginning in early life.IMPORTANCE There is growing evidence for a link between gastrointestinal bacterial communities and airway disease progression in CF. We demonstrate that infants with CF ≤1 year of age show a distinct stool microbiota versus that of control infants of a comparable age. We detected associations between the gut microbiome and airway exacerbation events in the cohort of infants with CF, and in vitro studies provided one possible mechanism for this observation. These data clarify that current therapeutics do not establish in infants with CF a gastrointestinal microbiota like that in healthy infants, and we suggest that interventions that direct the gastrointestinal microbiota closer to a healthy state may provide systemic benefits to these patients during a critical window of immune programming that might have implications for lifelong health.
Collapse
|
49
|
Enaud R, Hooks KB, Barre A, Barnetche T, Hubert C, Massot M, Bazin T, Clouzeau H, Bui S, Fayon M, Berger P, Lehours P, Bébéar C, Nikolski M, Lamireau T, Delhaes L, Schaeverbeke T. Intestinal Inflammation in Children with Cystic Fibrosis Is Associated with Crohn's-Like Microbiota Disturbances. J Clin Med 2019; 8:jcm8050645. [PMID: 31083321 PMCID: PMC6572243 DOI: 10.3390/jcm8050645] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 02/07/2023] Open
Abstract
Cystic fibrosis (CF) is a systemic genetic disease that leads to pulmonary and digestive disorders. In the majority of CF patients, the intestine is the site of chronic inflammation and microbiota disturbances. The link between gut inflammation and microbiota dysbiosis is still poorly understood. The main objective of this study was to assess gut microbiota composition in CF children depending on their intestinal inflammation. We collected fecal samples from 20 children with CF. Fecal calprotectin levels were measured and fecal microbiota was analyzed by 16S rRNA sequencing. We observed intestinal inflammation was associated with microbiota disturbances characterized mainly by increased abundances of Staphylococcus, Streptococcus, and Veillonella dispar, along with decreased abundances of Bacteroides, Bifidobacterium adolescentis, and Faecalibacterium prausnitzii. Those changes exhibited similarities with that of Crohn's disease (CD), as evidenced by the elevated CD Microbial-Dysbiosis index that we applied for the first time in CF. Furthermore, the significant over-representation of Streptococcus in children with intestinal inflammation appears to be specific to CF and raises the issue of gut-lung axis involvement. Taken together, our results provide new arguments to link gut microbiota and intestinal inflammation in CF and suggest the key role of the gut-lung axis in the CF evolution.
Collapse
Affiliation(s)
- Raphaël Enaud
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France.
- CRCM Pédiatrique, CHU Bordeaux, CIC 1401, F-33000 Bordeaux, France.
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
| | - Katarzyna B Hooks
- Bordeaux Bioinformatics Center, University Bordeaux, F-33000 Bordeaux, France.
- Laboratoire Bordelais de Recherche en Informatique, CNRS, University Bordeaux, UMR 5800, F-33400 Talence, France.
| | - Aurélien Barre
- Bordeaux Bioinformatics Center, University Bordeaux, F-33000 Bordeaux, France.
- Laboratoire Bordelais de Recherche en Informatique, CNRS, University Bordeaux, UMR 5800, F-33400 Talence, France.
| | - Thomas Barnetche
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
- Service de Rhumatologie, CHU Bordeaux, F-33000 Bordeaux, France.
| | - Christophe Hubert
- INSERM, MRGM, University Bordeaux, U1211, F-33000 Bordeaux, France.
- PGTB, University Bordeaux, F-33000 Bordeaux, France.
| | - Marie Massot
- BIOGECO, INRA, University Bordeaux, F-33610 Cestas, France.
| | - Thomas Bazin
- INRA-Bordeaux Aquitaine Centre, University Bordeaux, USC EA 3671, Infections Humaines à Mycoplasmes et à Chlamydiae, CHU Bordeaux, F-33000 Bordeaux, France.
| | - Haude Clouzeau
- CRCM Pédiatrique, CHU Bordeaux, CIC 1401, F-33000 Bordeaux, France.
| | - Stéphanie Bui
- CRCM Pédiatrique, CHU Bordeaux, CIC 1401, F-33000 Bordeaux, France.
| | - Michael Fayon
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France.
- CRCM Pédiatrique, CHU Bordeaux, CIC 1401, F-33000 Bordeaux, France.
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
| | - Patrick Berger
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France.
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
| | - Philippe Lehours
- BaRITOn, INSERM, University Bordeaux, UMR1053, CHU Bordeaux, F-33000 Bordeaux, France.
| | - Cécile Bébéar
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
- INRA-Bordeaux Aquitaine Centre, University Bordeaux, USC EA 3671, Infections Humaines à Mycoplasmes et à Chlamydiae, CHU Bordeaux, F-33000 Bordeaux, France.
| | - Macha Nikolski
- Bordeaux Bioinformatics Center, University Bordeaux, F-33000 Bordeaux, France.
- Laboratoire Bordelais de Recherche en Informatique, CNRS, University Bordeaux, UMR 5800, F-33400 Talence, France.
| | - Thierry Lamireau
- CRCM Pédiatrique, CHU Bordeaux, CIC 1401, F-33000 Bordeaux, France.
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
| | - Laurence Delhaes
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM, University Bordeaux, U1045, F-33000 Bordeaux, France.
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
| | - Thierry Schaeverbeke
- Fédération Hospitalo-Universitaire FHU, ACRONIM, F-33000 Bordeaux, France.
- Service de Rhumatologie, CHU Bordeaux, F-33000 Bordeaux, France.
- INRA-Bordeaux Aquitaine Centre, University Bordeaux, USC EA 3671, Infections Humaines à Mycoplasmes et à Chlamydiae, CHU Bordeaux, F-33000 Bordeaux, France.
| |
Collapse
|
50
|
Knysh OV, Isayenko OY, Voyda YV, Kizimenko OO, Babych YM. Influence of cell-free extracts of Bifidobacterium bifidum and Lactobacillus reuteri on proliferation and biofilm formation by Escherichia coli and Pseudomonas aeruginosa. REGULATORY MECHANISMS IN BIOSYSTEMS 2019. [DOI: 10.15421/021938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The development of new effective preparations for the correction of microecological disorders based on probiotic derivatives requires a comprehensive study of the biological activity of the latter. We studied the proliferative activity and biofilm formation by clinical isolates: Escherichia coli and Pseudomonas aeruginosa under the influence of cell-free extracts containing structural components and metabolites of the Bifidobacterium bifidum and Lactobacillus reuteri probiotic strains. Cell-free extracts were obtained from disintegrates and cultures of probiotics. Disintegrates were prepared by cyclic freezing-thawing of probiotic cell suspensions. The cultures were obtained by cultivating probiotic microorganisms in their own disintegrates. The obtained disintegrates and cultures were filtered. The proliferative activity of the test cultures was studied using the spectrophotometric microtiter plate method after an hour-long exposure in undiluted cell-free extracts and subsequent cultivation in a nutrient medium containing 30%vol of the studied extracts at 37 °C for 24 hours. The biofilm formation of the test cultures was studied with 30% vol content of cell-free extracts in the cultivation medium using the spectrophotometric microtiter plate method. All the studied extracts exerted a similar effect on the proliferative activity and biofilm formation by E. coli and P. aeruginosa. Exposure of the test cultures in all undiluted extracts during an hour led to a significant decrease in the optical density of the test samples: optical density of the test wells ranged from 36.5% to 49.8% of the control wells. The test cultures that were exposed to the extracts: filtrate of L. reuteri disintegrate (L), filtrate of В. bifidum disintegrate (B) and filtrate of В. bifidum culture, grown in В. bifidum disintegrate (MB) after dilution and subsequent cultivation over the next 24 hours completely restored the ability to proliferate. The proliferative activity of the test cultures that were exposed to the extracts: filtrate of L. reuteri culture, grown in L. reuteri disintegrate (ML) and filtrate of L. reuteri culture, grown in L. reuteri disintegrate supplemented with 0.8 M glycerol and 0.4 M glucose (MLG), was significantly inhibited after dilution and subsequent cultivation. The inhibition indices calculated for the ML extract were: 25.9% (E. coli) and 53.0% (P. aeruginosa). Inhibition indices calculated for the MLG extract were: 62.0% (E. coli) and 96.9% (P. aeruginosa). MLG extract had more pronounced inhibitory effect on the proliferation of the test cultures than ML extract. All the studied extracts exerted significant inhibitory effect on the biofilm formation of the test cultures. Analysis of the results of the study shows that cell-free extracts of L. reuteri culture grown in its disintegrate without supplementation or supplemented with glycerol and glucose have the highest antimicrobial activity and can be used as metabiotics to prevent overgrowth of potentially pathogenic bacteria, as well as inoculation and proliferation of pathogenic gram-negative bacteria in the gastrointestinal tract. They can be used alone or in combination with cellular probiotics to enhance their probiotic action. This study encourages further careful investigation of the biochemical composition of cell-free extracts and clarifying the mechanism of their action.
Collapse
|