101
|
Rocchetti G, Bhumireddy SR, Giuberti G, Mandal R, Lucini L, Wishart DS. Edible nuts deliver polyphenols and their transformation products to the large intestine: An in vitro fermentation model combining targeted/untargeted metabolomics. Food Res Int 2018; 116:786-794. [PMID: 30717008 DOI: 10.1016/j.foodres.2018.09.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/28/2018] [Accepted: 09/08/2018] [Indexed: 01/22/2023]
Abstract
The fate of polyphenols from edible tree nuts was investigated using a simulated in vitro intestinal fermentation system. The digested food matrix was fermented for 48 h and the changes in the phenolic profiles were evaluated by both untargeted UHPLC-QTOF and targeted UHPLC-Orbitrap mass spectrometry. The untargeted metabolomics approach allowed us to monitor the comprehensive changes in phenolic profiles from 0 up to 48 h of in vitro fermentation. Multivariate statistics (i.e., orthogonal projection to latent structures discriminant analysis) applied to this untargeted data allowed us to identify the most discriminating phenolic metabolites and to further understand the colonic transformation pathways involved. In particular, 13 putatively identified compounds derived from flavonoids, lignans and phenolic acids were found to have the highest discrimination potential. Six phenolic metabolites were then quantified by means of targeted metabolomics (using a UHPLC-Orbitrap). These metabolites included 3,4-dihydroxyphenylacetic acid, 4-hydroxybenzoic acid, hippuric acid, caffeic acid, protocatechuic acid and protocatechuic aldehyde. Using the targeted data, a clear matrix effect was observed over time, with an increase of some phenolic metabolites moving from 8 to 48 h of in vitro fermentation. Based on these data, catabolic pathways for colonic microbial degradation of flavonoids, hydroxycinnamic acids, tyrosols and lignans are proposed. Our findings show that edible tree nuts deliver polyphenols to the colon, where several microbial transformations occur that lead to smaller phenolic metabolites being observed. Furthermore, we found that the combined use of targeted and untargeted metabolomics can be particularly effective for investigating the fate of polyphenols in the large intestine.
Collapse
Affiliation(s)
- Gabriele Rocchetti
- Department of Animal Science, Food and Nutrition, Università Cattolica del Sacro Cuore, Piacenza 29122, Italy; Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza 29122, Italy; Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada.
| | | | - Gianluca Giuberti
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza 29122, Italy
| | - Rupasri Mandal
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Luigi Lucini
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza 29122, Italy.
| | - David S Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada; Department of Computing Science, University of Alberta, Edmonton, AB T6G 2E8, Canada
| |
Collapse
|
102
|
Li L, Somerset S. Associations between Flavonoid Intakes and Gut Microbiota in a Group of Adults with Cystic Fibrosis. Nutrients 2018; 10:nu10091264. [PMID: 30205496 PMCID: PMC6164979 DOI: 10.3390/nu10091264] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 08/31/2018] [Accepted: 09/03/2018] [Indexed: 02/06/2023] Open
Abstract
Dietary flavonoid intakes can influence gut microbiota (GM), which in turn can affect immune function and host metabolism, both vital considerations in cystic fibrosis (CF) management. In CF, GM may be altered and link to CF respiratory events. This study explored the relationship between flavonoid intakes and GM in free-living adults with CF. Associations between the overall GM variations (unweighted and weighted UniFrac distances between pyrosequencing results of bacterial 16-ss rDNA from frozen faecal samples of sixteen CF adults) and standardised dietary flavonoid intakes (a validated flavonoid-specific food frequency questionnaire) were analysed using adonis tests. Flavonoid intakes that were significant at a false discovery rate (FDR) < 0.3 were subjected to Spearman correlation tests with standardised bacterial relative abundances (FDR < 0.3). Gallocatechin intakes (p = 0.047, q = 0.285) were associated with unweighted UniFrac distances. Intakes of apigenin (p = 0.028, q = 0.227) and kaempferol (p = 0.029, q = 0.227), and % flavonoid intake as flavones (p = 0.013, q = 0.227) and flavonols (p = 0.016, q = 0.227) (both excluding contribution of tea) were associated with weighted UniFrac distances. Among these, gallocatechin correlated with the genus Actinomyces and family Actinomycetaceae (Actinobacteria). Gallocatechin correlated negatively with class Coriobacteriia (Actinobacteria). Intakes of some flavonoids may be associated with GM variations with potential consequences for metabolism, immune function, and inflammation, which are important in CF lung disease and co-morbidity management.
Collapse
Affiliation(s)
- Li Li
- School of Medicine, Menzies Health Institute Queensland, Griffith University, 68 University Drive, Meadowbrook, QLD 4131, Australia.
- Faculty of Health, University of Canberra, University Drive, Bruce, ACT 2617, Australia.
| | - Shawn Somerset
- Faculty of Health, University of Canberra, University Drive, Bruce, ACT 2617, Australia.
| |
Collapse
|
103
|
van de Peppel IP, Bodewes FAJA, Verkade HJ, Jonker JW. Bile acid homeostasis in gastrointestinal and metabolic complications of cystic fibrosis. J Cyst Fibros 2018; 18:313-320. [PMID: 30201330 DOI: 10.1016/j.jcf.2018.08.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022]
Abstract
With the improved treatment of the pulmonary complications of cystic fibrosis (CF), gastrointestinal problems have become more important in the morbidity in CF. A hallmark of the gastrointestinal phenotype of CF, apart from pancreatic insufficiency, is a disruption of bile acid homeostasis. Bile acid homeostasis is important for many gastrointestinal processes including fat absorption, inflammation, microbial composition, as well as regulation of whole body energy metabolism. This review describes the impairment of bile acid homeostasis in CF, its possible consequences for gastrointestinal and metabolic complications and its potential as a target for therapy.
Collapse
Affiliation(s)
- Ivo P van de Peppel
- Pediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, the Netherlands; Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands.
| | - Frank A J A Bodewes
- Pediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Henkjan J Verkade
- Pediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, the Netherlands; Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Johan W Jonker
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, the Netherlands.
| |
Collapse
|
104
|
Van Biervliet S, Hauser B, Verhulst S, Stepman H, Delanghe J, Warzee JP, Pot B, Vandewiele T, Wilschanski M. Probiotics in cystic fibrosis patients: A double blind crossover placebo controlled study: Pilot study from the ESPGHAN Working Group on Pancreas/CF. Clin Nutr ESPEN 2018; 27:59-65. [PMID: 30144894 DOI: 10.1016/j.clnesp.2018.06.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/25/2018] [Accepted: 06/22/2018] [Indexed: 01/15/2023]
Abstract
BACKGROUND A potential positive effect of probiotics in cystic fibrosis (CF) on fecal calprotectin (FCP), pulmonary exacerbations and weight has been described in small controlled trials. METHODS A double-blind multicenter cross-over study (2 × 4 m) was performed looking at abdominal pain, nutritional status, pulmonary function, pulmonary exacerbation, FCP and lactulose/mannitol gut permeability test. Patients kept a diary with daily scoring of abdominal pain, stool frequency and consistency as well as treatment changes. RESULTS 31 CF patients entered the study of which 25 finished it. At start patients aged 9.3yrs (6.9-12.2), had a median BMI z-score of -0.5 (-1.5-0.08), height z-score of -0.4 (-1.1-0.05) and FEV1% of 100% (87.2-106.6). Median FCP at start was 61 μg/g (17-108) and gut permeability 0.079 (0.051-0.122). No significant changes were observed in the clinical parameters (BMI, FEV1%, abdominal pain, exacerbations). Despite being frequently abnormal (17/28 (61%) >50 mg/kg), FCP did not change significantly with probiotics. The proportion of patients with normal permeability was 8% during placebo and 32% during probiotic treatment (p = 0.031). FCP correlated to BMI z-score (p = 0.043) and gut permeability to abdominal pain (p = 0.015). The microbiome revealed a high predominance of Actinobacteria and Proteobacteriae. Probiotic supplementation did not result in a shift at the phylum nor at phylogenetic level. CONCLUSION Normalization of gut permeability was observed in 13% of patients during probiotic treatment. However, none of the previously described effects could be confirmed.
Collapse
Affiliation(s)
| | - Bruno Hauser
- Vrije Universiteit Brussel, Pediatric Gastroenterology, CF Centre, Brussels, Belgium
| | - Stijn Verhulst
- Antwerp University, Pediatric Pulmonology, CF Centre, Antwerp, Belgium
| | - Hedwig Stepman
- Ghent University Hospital, Laboratory of Clinical Chemistry, Microbiology and Immunology, Ghent, Belgium
| | - Joris Delanghe
- Ghent University Hospital, Laboratory of Clinical Chemistry, Microbiology and Immunology, Ghent, Belgium
| | | | - Bruno Pot
- Vrije Universiteit Brussel, Research Group of Industrial Microbiology and Food Biotechnology, Brussels, Belgium
| | | | - Michael Wilschanski
- Hadassah University Hospitals, Pediatric Gastroenterology, CF Centre, Jerusalem, Israel
| |
Collapse
|
105
|
Bacterial overgrowth, dysbiosis, inflammation, and dysmotility in the Cystic Fibrosis intestine. J Cyst Fibros 2018; 16 Suppl 2:S14-S23. [PMID: 28986022 DOI: 10.1016/j.jcf.2017.07.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/15/2017] [Accepted: 07/17/2017] [Indexed: 02/08/2023]
Abstract
Gastrointestinal disease in Cystic Fibrosis (CF) is caused by defective chloride and bicarbonate transport in intestinal cells leading to reduced intraluminal fluidity, increased mucous viscosity and consequently development of intestinal inflammation, dysbiosis and often times dysmotility. This triad is also referred to as the "CF gut". A diagnosis is mainly based on clinical observation and treatment is often times decided empirically. This review of the literature should provide CF caregivers with some tools to identify intestinal inflammation, dysbiosis and dysmotility as possible cause for their patient's gastrointestinal complaints and provide an overview of our current approach to its management.
Collapse
|
106
|
de Freitas MB, Moreira EAM, Tomio C, Moreno YMF, Daltoe FP, Barbosa E, Ludwig Neto N, Buccigrossi V, Guarino A. Altered intestinal microbiota composition, antibiotic therapy and intestinal inflammation in children and adolescents with cystic fibrosis. PLoS One 2018; 13:e0198457. [PMID: 29933382 PMCID: PMC6014676 DOI: 10.1371/journal.pone.0198457] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 05/18/2018] [Indexed: 12/27/2022] Open
Abstract
The aim of the present study was to evaluate the effect of cystic fibrosis and antibiotic therapy on intestinal microbiota composition and intestinal inflammation in children and adolescents. A cross-sectional controlled study was conducted with 36 children and adolescents: 19 in the cystic fibrosis group (CFG) and 17 in the control group (CG) matched for age and sex. The CFG was subdivided based on the use of antibiotic therapy (CFAB group) and non-use of antibiotic therapy (CFnAB group). The following data were evaluated: colonization, antibiotic therapy, mutation, breastfeeding, use of infant formula, type of delivery, introduction of solid foods, body mass index, fecal calprotectin and intestinal microbiota composition (fluorescence in situ hybridization). Intestinal inflammation evaluated by fecal calprotectin was significantly higher in the CFG (median: 40.80 µg/g, IQR: 19.80-87.10, p = 0.040) and CFAB group (median: 62.95 µg/g, IQR: 21.80-136.62, p = 0.045) compared to the CG (median: 20.15 µg/g, IQR: 16.20-31.00), and the Bacteroides, Firmicutes, Eubacterium rectale and Faecalibacterium prausnitzii were significantly decreased (p < 0.05) in the CFG compared to the CG, whereas the bacteria Clostridium difficile, Escherichia coli and Pseudomonas aeruginosa were significantly increased in the CFG (p < 0.05). The main differences were found between the CG and CFAB group for Eubacterium rectale (p = 0.006), Bifidobacterium (p = 0.017), Escherichia coli (p = 0.030), Firmicutes (p = 0.002), Pseudomonas aeruginosa (p < 0.001) and Clostridium difficile (p = 0.006). The results of this study confirm intestinal inflammation in patients with CF, which may be related to changes in the composition of the intestinal microbiota.
Collapse
Affiliation(s)
| | | | - Camila Tomio
- Graduate Program in Nutrition, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Yara Maria Franco Moreno
- Department of Nutrition, Graduate Program in Nutrition, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Felipe Perozzo Daltoe
- Department of Pathology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Eliana Barbosa
- Joana de Gusmão Children´s Hospital, Santa Catarina, Florianópolis, Brazil
| | | | - Vittoria Buccigrossi
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Alfredo Guarino
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| |
Collapse
|
107
|
Lack of efficacy of Lactobacillus GG in reducing pulmonary exacerbations and hospital admissions in children with cystic fibrosis: A randomised placebo controlled trial. J Cyst Fibros 2018; 17:375-382. [DOI: 10.1016/j.jcf.2017.10.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 10/22/2017] [Accepted: 10/22/2017] [Indexed: 12/15/2022]
|
108
|
Harnessing the Power of Microbiome Assessment Tools as Part of Neuroprotective Nutrition and Lifestyle Medicine Interventions. Microorganisms 2018; 6:microorganisms6020035. [PMID: 29693607 PMCID: PMC6027349 DOI: 10.3390/microorganisms6020035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/02/2018] [Accepted: 04/20/2018] [Indexed: 12/11/2022] Open
Abstract
An extensive body of evidence documents the importance of the gut microbiome both in health and in a variety of human diseases. Cell and animal studies describing this relationship abound, whilst clinical studies exploring the associations between changes in gut microbiota and the corresponding metabolites with neurodegeneration in the human brain have only begun to emerge more recently. Further, the findings of such studies are often difficult to translate into simple clinical applications that result in measurable health outcomes. The purpose of this paper is to appraise the literature on a select set of faecal biomarkers from a clinician’s perspective. This practical review aims to examine key physiological processes that influence both gastrointestinal, as well as brain health, and to discuss how tools such as the characterisation of commensal bacteria, the identification of potential opportunistic, pathogenic and parasitic organisms and the quantification of gut microbiome biomarkers and metabolites can help inform clinical decisions of nutrition and lifestyle medicine practitioners.
Collapse
|
109
|
Gupta N, Kumar R, Agrawal B. New Players in Immunity to Tuberculosis: The Host Microbiome, Lung Epithelium, and Innate Immune Cells. Front Immunol 2018; 9:709. [PMID: 29692778 PMCID: PMC5902499 DOI: 10.3389/fimmu.2018.00709] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/22/2018] [Indexed: 12/31/2022] Open
Abstract
Tuberculosis (TB) is a highly contagious infection and devastating chronic disease, causing 10.4 million new infections and 1.8 million deaths every year globally. Efforts to control and eradicate TB are hampered by the rapid emergence of drug resistance and limited efficacy of the only available vaccine, BCG. Immunological events in the airways and lungs are of major importance in determining whether exposure to Mycobacterium tuberculosis (Mtb) results in successful infection or protective immunity. Several studies have demonstrated that the host microbiota is in constant contact with the immune system, and thus continually directs the nature of immune responses occurring during new infections. However, little is known about its role in the eventual outcome of the mycobacterial infection. In this review, we highlight the changes in microbial composition in the respiratory tract and gut that have been linked to the alteration of immune responses, and to the risk, prevention, and treatment of TB. In addition, we summarize our current understanding of alveolar epithelial cells and the innate immune system, and their interaction with Mtb during early infection. Extensive studies are warranted to fully understand the all-inclusive role of the lung microbiota, its interaction with epithelium and innate immune responses and resulting adaptive immune responses, and in the pathogenesis and/or protection from Mtb infection. Novel interventions aimed at influencing the microbiota, the alveolar immune system and innate immunity will shape future strategies of prevention and treatment for TB.
Collapse
Affiliation(s)
- Nancy Gupta
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Rakesh Kumar
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
110
|
Rocchetti G, Chiodelli G, Giuberti G, Lucini L. Bioaccessibility of phenolic compounds following in vitro large intestine fermentation of nuts for human consumption. Food Chem 2018; 245:633-640. [DOI: 10.1016/j.foodchem.2017.10.146] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 10/13/2017] [Accepted: 10/31/2017] [Indexed: 10/18/2022]
|
111
|
Jayashree S, Karthikeyan R, Nithyalakshmi S, Ranjani J, Gunasekaran P, Rajendhran J. Anti-adhesion Property of the Potential Probiotic Strain Lactobacillus fermentum 8711 Against Methicillin-Resistant Staphylococcus aureus (MRSA). Front Microbiol 2018; 9:411. [PMID: 29568290 PMCID: PMC5852077 DOI: 10.3389/fmicb.2018.00411] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 02/21/2018] [Indexed: 01/01/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a multidrug-resistant pathogen and one of the leading causes of nosocomial infection worldwide. Probiotic bacteria play a significant role in preventive or therapeutic interventions of gastrointestinal infections in human as well as animals. In this study, we have investigated the adhesion property of the probiotic strain Lactobacillus fermentum MTCC 8711 and its ability to prevent the adhesion of MRSA to human colon adenocarcinoma cells, Caco-2. We have shown that L. fermentum could efficiently adhere to the Caco-2 cells. Also, we have shown that L. fermentum significantly reduced MRSA adhesion to Caco-2 cells. Three types of experiments were performed to assess the anti-adhesion property of L. fermentum against MRSA. Inhibition (Caco-2 cells were pre-treated with L. fermentum, and subsequently MRSA was added), competition (both L. fermentum and MRSA were added to Caco-2 cells simultaneously), and displacement or exclusion (Caco-2 cells were pre-treated with MRSA, and subsequently L. fermentum was added). In all three experiments, adhesion of MRSA was significantly reduced. Interestingly, L. fermentum could efficiently displace the adhered MRSA, and hence this probiotic can be used for therapeutic applications also. In cytotoxicity assay, we found that L. fermentum per se was not cytotoxic, and also significantly reduced the MRSA-induced cytotoxicity. The protective effect occurred without affecting Caco-2 cell morphology and viability.
Collapse
Affiliation(s)
| | - Raman Karthikeyan
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Sampath Nithyalakshmi
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Jothi Ranjani
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | | | - Jeyaprakash Rajendhran
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| |
Collapse
|
112
|
Galacto-Oligosaccharide/Polidextrose Enriched Formula Protects against Respiratory Infections in Infants at High Risk of Atopy: A Randomized Clinical Trial. Nutrients 2018; 10:nu10030286. [PMID: 29494489 PMCID: PMC5872704 DOI: 10.3390/nu10030286] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/20/2018] [Accepted: 02/24/2018] [Indexed: 01/06/2023] Open
Abstract
Background: Early nutrition affects the risk of atopy and infections through modifications of intestinal microbiota. The Prebiotics in the Prevention of Atopy (PIPA) study was a 24-month randomised, double-blind, placebo-controlled trial. It aimed to evaluate the effects of a galacto-oligosaccharide/polydextrose (GOS/PDX)-formula (PF) on atopic dermatitis (AD) and common infections in infants who were born to atopic parents and to investigate the relationship among early nutrition, gut microbiota and clinical outcomes. Methods: A total of 201 and 199 infants were randomized to receive a PF and standard formula (SF), respectively; 140 infants remained on exclusive breastfeeding (BF). Results: The cumulative incidence of AD and its intensity and duration were not statistically different among the three groups. The number of infants with at least one episode of respiratory infection (RI) and the mean number of episodes until 48 weeks of age were significantly lower in the PF group than in the SF group. The number of patients with recurrent RIs and incidence of wheezing lower RIs until 96 weeks were lower in the PF group than the SF group, but similar to the BF group. Bifidobacteria and Clostridium cluster I colonization increased over time in the PF group but decreased in the SF and BF groups. Bifidobacteria had a protective role in RIs, whereas Clostridium cluster I was associated with atopy protection. Conclusion: The early administration of PF protects against RIs and mediates a species-specific modulation of the intestinal microbiota. Trial registration: clinicaltrial.gov Identifier: NCT02116452.
Collapse
|
113
|
Coffey MJ, Garg M, Homaira N, Jaffe A, Ooi CY. Probiotics for people with cystic fibrosis. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2018. [DOI: 10.1002/14651858.cd012949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Michael J Coffey
- University of New South Wales; School of Women's and Children's Health; Level 8, Centre for Child Health Research & Innovation Bright Alliance Building Cnr Avoca & High Streets Randwick Sydney NSW Australia 2031
- Sydney Children's Hospital; Junior Medical Officers Department; High Street Randwick Sydney NSW Australia 2031
| | - Millie Garg
- University of New South Wales; School of Women's and Children's Health; Level 8, Centre for Child Health Research & Innovation Bright Alliance Building Cnr Avoca & High Streets Randwick Sydney NSW Australia 2031
| | - Nusrat Homaira
- University of New South Wales; School of Women's and Children's Health; Level 8, Centre for Child Health Research & Innovation Bright Alliance Building Cnr Avoca & High Streets Randwick Sydney NSW Australia 2031
| | - Adam Jaffe
- University of New South Wales; School of Women's and Children's Health; Level 8, Centre for Child Health Research & Innovation Bright Alliance Building Cnr Avoca & High Streets Randwick Sydney NSW Australia 2031
- Sydney Children's Hospital; Respiratory Department; High Street Randwick Sydney NSW Australia 2031
| | - Chee Y Ooi
- University of New South Wales; School of Women's and Children's Health; Level 8, Centre for Child Health Research & Innovation Bright Alliance Building Cnr Avoca & High Streets Randwick Sydney NSW Australia 2031
- Sydney Children's Hospital; Gastroenterology Department; High Street Randwick Sydney NSW Australia 2031
| |
Collapse
|
114
|
Garg M, Leach ST, Pang T, Needham B, Coffey MJ, Katz T, Strachan R, Widger J, Field P, Belessis Y, Chuang S, Day AS, Jaffe A, Ooi CY. Age-related levels of fecal M2-pyruvate kinase in children with cystic fibrosis and healthy children 0 to 10years old. J Cyst Fibros 2018; 17:109-113. [PMID: 28754328 DOI: 10.1016/j.jcf.2017.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/11/2017] [Accepted: 07/11/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND The pathogenesis of gut inflammation, bacterial dysbiosis and increased rates of malignancy in CF is unclear. Fecal M2-pyruvate kinase (M2-PK) is a biomarker indicative of cellular proliferation that may be raised in intestinal malignancy and inflammation. Biomarkers, including M2-PK, may be useful in assessing effects of novel therapies on the gastrointestinal tract. METHODS M2-PK was measured in stools collected from patients with CF and HC (0-10years). Linear mixed model analysis was used. RESULTS M2-PK levels did not significantly change in children with CF (36 patients, 77 samples) (P=0.998) or HC (45 patients, 45 samples) (P=0.21), over the age range 0-10years. Patients with CF had elevated M2-PK compared to HC (median [IQR; range]: 10.7 [5.7-28.6; 1.0-239.1] (n=77) vs. 1.0 [1.0-1.0; 1.0-50.0] (n=45) U/mL, respectively; P=0.001). CONCLUSIONS Fecal M2-PK was elevated in children with CF compared with HC during infancy and throughout childhood suggesting abnormalities in the CF gut exist in early life.
Collapse
Affiliation(s)
- Millie Garg
- School of Women and Children's Health, Medicine, The University of New South Wales, High Street, Sydney Children's Hospital, Randwick 2031, New South Wales, Australia
| | - Steven T Leach
- School of Women and Children's Health, Medicine, The University of New South Wales, High Street, Sydney Children's Hospital, Randwick 2031, New South Wales, Australia
| | - Tamara Pang
- School of Women and Children's Health, Medicine, The University of New South Wales, High Street, Sydney Children's Hospital, Randwick 2031, New South Wales, Australia
| | - Bronwen Needham
- Sydney Medical Program, The University of Sydney, Camperdown 2050, New South Wales, Australia
| | - Michael J Coffey
- School of Women and Children's Health, Medicine, The University of New South Wales, High Street, Sydney Children's Hospital, Randwick 2031, New South Wales, Australia
| | - Tamarah Katz
- Department of Nutrition and Dietetics, Sydney Children's Hospital, High Street, Randwick 2031, New South Wales, Australia
| | - Roxanne Strachan
- Department of Respiratory Medicine, Sydney Children's Hospital, High Street, Randwick 2031, New South Wales, Australia
| | - John Widger
- School of Women and Children's Health, Medicine, The University of New South Wales, High Street, Sydney Children's Hospital, Randwick 2031, New South Wales, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, High Street, Randwick 2031, New South Wales, Australia
| | - Penelope Field
- School of Women and Children's Health, Medicine, The University of New South Wales, High Street, Sydney Children's Hospital, Randwick 2031, New South Wales, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, High Street, Randwick 2031, New South Wales, Australia
| | - Yvonne Belessis
- School of Women and Children's Health, Medicine, The University of New South Wales, High Street, Sydney Children's Hospital, Randwick 2031, New South Wales, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, High Street, Randwick 2031, New South Wales, Australia
| | - Sandra Chuang
- School of Women and Children's Health, Medicine, The University of New South Wales, High Street, Sydney Children's Hospital, Randwick 2031, New South Wales, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, High Street, Randwick 2031, New South Wales, Australia
| | - Andrew S Day
- Department of Paediatrics, University of Otago, Riccarton Ave, Christchurch 8011, Canterbury, New Zealand
| | - Adam Jaffe
- School of Women and Children's Health, Medicine, The University of New South Wales, High Street, Sydney Children's Hospital, Randwick 2031, New South Wales, Australia; Department of Respiratory Medicine, Sydney Children's Hospital, High Street, Randwick 2031, New South Wales, Australia
| | - Chee Y Ooi
- School of Women and Children's Health, Medicine, The University of New South Wales, High Street, Sydney Children's Hospital, Randwick 2031, New South Wales, Australia; Department of Gastroenterology, Sydney Children's Hospital, High Street, Randwick 2031, New South Wales, Australia.
| |
Collapse
|
115
|
Rocchetti G, Chiodelli G, Giuberti G, Ghisoni S, Baccolo G, Blasi F, Montesano D, Trevisan M, Lucini L. UHPLC-ESI-QTOF-MS profile of polyphenols in Goji berries (Lycium barbarum L.) and its dynamics during in vitro gastrointestinal digestion and fermentation. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.11.042] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
|
116
|
Takahashi J, Rindfleisch JA. Prescribing Probiotics. Integr Med (Encinitas) 2018. [DOI: 10.1016/b978-0-323-35868-2.00105-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
117
|
Kanhere M, Chassaing B, Gewirtz AT, Tangpricha V. Role of vitamin D on gut microbiota in cystic fibrosis. J Steroid Biochem Mol Biol 2018; 175:82-87. [PMID: 27818276 PMCID: PMC5415426 DOI: 10.1016/j.jsbmb.2016.11.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 10/24/2016] [Accepted: 11/01/2016] [Indexed: 02/07/2023]
Abstract
This review explores the potential for vitamin D to favorably alter the gut microbiota, given emerging evidence of the role of vitamin D in controlling mucosal inflammation in the gut. It will focus on cystic fibrosis (CF) patients, a population with both vitamin D deficiency due to gut malabsorption and an altered gut microbiota composition. Recent evidence shows that vitamin D acts to maintain the integrity of the gut mucosal barrier by enhancement of intercellular junctions that control mucosal permeability and reduction of pro-inflammatory cytokines such as IL-8. In addition, vitamin D receptor-mediated signaling has been shown to inhibit inflammation-induced apoptosis of intestinal epithelial cells. As a result of these effects on the intestinal mucosa, maintenance of sufficient vitamin D status may be essential for the development of a healthy gut microbiota, particularly in conditions defined by chronic mucosal inflammation such as CF. We hypothesize here that high dose vitamin D may be used to favorably manipulate the aberrant mucosa seen in patients with CF. This may result in improved clinical outcomes in association with a low inflammatory environment that allows beneficial bacteria to outcompete opportunistic pathogens. Current evidence is sparse but encouraging, and additional evidence is needed to establish vitamin D as a therapeutic approach for gut microbiota modification.
Collapse
Affiliation(s)
- Mansi Kanhere
- Division of Endocrinology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Benoit Chassaing
- Center for Inflammation, Immunity, & Infection, Institute for Biomedical Sciences, Georgia State University, USA
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity, & Infection, Institute for Biomedical Sciences, Georgia State University, 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
|
118
|
Ojha UC, Singh DP, Choudhari OK, Gothi D, Singh S. Correlation of Severity of Functional Gastrointestinal Disease Symptoms with that of Asthma and Chronic Obstructive Pulmonary Disease: A Multicenter Study. Int J Appl Basic Med Res 2018; 8:83-88. [PMID: 29744319 PMCID: PMC5932929 DOI: 10.4103/ijabmr.ijabmr_258_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Introduction There is a growing clinical awareness about the influence of gut-lung axis on lung injury and coexisting manifestations of disease processes in both the intestine and lungs. Patients of chronic lung diseases such as chronic obstructive pulmonary disease (COPD) and asthma very often present with coexistent gut symptoms. In the present study, we have tried to establish the correlation of severity of pulmonary pathology of COPD and asthma patients with functional gastrointestinal (GI) symptoms of the patients. Materials and Methods This is a prospective, questionnaire-based study comprising patients with asthma and COPD. After following strict inclusion and exclusion criteria, a total of 200 patients (100 patients of bronchial asthma and 100 patients of COPD) were included in the study. Functional GI symptom questionnaire [Annexure 1-Bowel Disease Questionnaire] is based on ROME III diagnostic criteria. On the basis of GOLD (Global Initiative for Obstructive Lung Disease) guidelines, COPD patients were divided into 4 categories (mild - GOLD 1, moderate - GOLD2, severe - GOLD3 and very severe - GOLD4). Asthma patients were divided into three categories (well controlled, partly controlled, uncontrolled) on the basis of GINA (Global Initiative for Asthma) guidelines. Results Highest percentage of patients with maximum GI symptoms was found in "GOLD-4" group among COPD patients and "uncontrolled" group among asthma patients. Highest percentage of patients with least GI symptoms was found in "GOLD-1" group among COPD patients and "well controlled" group among asthma patients. Conclusion We can conclude from our study that the phenomenon of gut-lung axis not only exists but also the severity of symptoms of one system (gut) carries a high degree of concordance with severity of other (lung).
Collapse
Affiliation(s)
- Umesh Chandra Ojha
- Department of Pulmonary Medicine, ESI Post Graduate Institute of Medical Sciences and Research, New Delhi, India
| | - Devesh Pratap Singh
- Department of TB and Respiratory Diseases, Hind Institute of Medical Sciences, Barabanki, Uttar Pradesh, India
| | - Omkar Kalidasrao Choudhari
- Department of Pulmonary Medicine, ESI Post Graduate Institute of Medical Sciences and Research, New Delhi, India
| | - Dipti Gothi
- Department of Pulmonary Medicine, ESI Post Graduate Institute of Medical Sciences and Research, New Delhi, India
| | - Shweta Singh
- Department of Obstetrics and Gynecology, Hind Institute of Medical Sciences, Barabanki, Uttar Pradesh, India
| |
Collapse
|
119
|
A Metagenomic and in Silico Functional Prediction of Gut Microbiota Profiles May Concur in Discovering New Cystic Fibrosis Patient-Targeted Probiotics. Nutrients 2017; 9:nu9121342. [PMID: 29232848 PMCID: PMC5748792 DOI: 10.3390/nu9121342] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 12/15/2022] Open
Abstract
Cystic fibrosis (CF) is a life-limiting hereditary disorder that results in aberrant mucosa in the lungs and digestive tract, chronic respiratory infections, chronic inflammation, and the need for repeated antibiotic treatments. Probiotics have been demonstrated to improve the quality of life of CF patients. We investigated the distribution of gut microbiota (GM) bacteria to identify new potential probiotics for CF patients on the basis of GM patterns. Fecal samples of 28 CF patients and 31 healthy controls (HC) were collected and analyzed by 16S rRNA-based pyrosequencing analysis of GM, to produce CF-HC paired maps of the distribution of operational taxonomic units (OTUs), and by Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) for Kyoto Encyclopedia of Genes and Genomes (KEGG) biomarker prediction. The maps were scanned to highlight the distribution of bacteria commonly claimed as probiotics, such as bifidobacteria and lactobacilli, and of butyrate-producing colon bacteria, such as Eubacterium spp. and Faecalibacterium prausnitzii. The analyses highlighted 24 OTUs eligible as putative probiotics. Eleven and nine species were prevalently associated with the GM of CF and HC subjects, respectively. Their KEGG prediction provided differential CF and HC pathways, indeed associated with health-promoting biochemical activities in the latter case. GM profiling and KEGG biomarkers concurred in the evaluation of nine bacterial species as novel putative probiotics that could be investigated for the nutritional management of CF patients.
Collapse
|
120
|
Costantini L, Molinari R, Farinon B, Merendino N. Impact of Omega-3 Fatty Acids on the Gut Microbiota. Int J Mol Sci 2017; 18:ijms18122645. [PMID: 29215589 PMCID: PMC5751248 DOI: 10.3390/ijms18122645] [Citation(s) in RCA: 452] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/14/2017] [Accepted: 12/01/2017] [Indexed: 12/12/2022] Open
Abstract
Long-term dietary habits play a crucial role in creating a host-specific gut microbiota community in humans. Despite the many publications about the effects of carbohydrates (prebiotic fibers), the impact of dietary fats, such as omega-3 polyunsaturated fatty acids (PUFAs), on the gut microbiota is less well defined. The few studies completed in adults showed some common changes in the gut microbiota after omega-3 PUFA supplementation. In particular, a decrease in Faecalibacterium, often associated with an increase in the Bacteroidetes and butyrate-producing bacteria belonging to the Lachnospiraceae family, has been observed. Coincidentally, a dysbiosis of these taxa is found in patients with inflammatory bowel disease. Omega-3 PUFAs can exert a positive action by reverting the microbiota composition in these diseases, and increase the production of anti-inflammatory compounds, like short-chain fatty acids. In addition, accumulating evidence in animal model studies indicates that the interplay between gut microbiota, omega-3 fatty acids, and immunity helps to maintain the intestinal wall integrity and interacts with host immune cells. Finally, human and animal studies have highlighted the ability of omega-3 PUFAs to influence the gut–brain axis, acting through gut microbiota composition. From these findings, the importance of the omega-3 connection to the microbiota emerges, encouraging further studies.
Collapse
Affiliation(s)
- Lara Costantini
- Department of Ecological and Biological Sciences (DEB), Tuscia University, Largo dell'Università snc, 01100 Viterbo, Italy.
| | - Romina Molinari
- Department of Ecological and Biological Sciences (DEB), Tuscia University, Largo dell'Università snc, 01100 Viterbo, Italy.
| | - Barbara Farinon
- Department of Ecological and Biological Sciences (DEB), Tuscia University, Largo dell'Università snc, 01100 Viterbo, Italy.
| | - Nicolò Merendino
- Department of Ecological and Biological Sciences (DEB), Tuscia University, Largo dell'Università snc, 01100 Viterbo, Italy.
| |
Collapse
|
121
|
Cockburn DW, Suh C, Medina KP, Duvall RM, Wawrzak Z, Henrissat B, Koropatkin NM. Novel carbohydrate binding modules in the surface anchored α-amylase of Eubacterium rectale provide a molecular rationale for the range of starches used by this organism in the human gut. Mol Microbiol 2017; 107:249-264. [PMID: 29139580 PMCID: PMC6939882 DOI: 10.1111/mmi.13881] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2017] [Indexed: 12/30/2022]
Abstract
Gut bacteria recognize accessible glycan substrates within a complex environment. Carbohydrate binding modules (CBMs) of cell surface glycoside hydrolases often drive binding to the target substrate. Eubacterium rectale, an important butyrate-producing organism in the gut, consumes a limited range of substrates, including starch. Host consumption of resistant starch increases the abundance of E. rectale in the intestine, likely because it successfully captures the products of resistant starch degradation by other bacteria. Here, we demonstrate that the cell wall anchored starch-degrading α-amylase, Amy13K of E. rectale harbors five CBMs that all target starch with differing specificities. Intriguingly these CBMs efficiently bind to both regular and high amylose corn starch (a type of resistant starch), but have almost no affinity for potato starch (another type of resistant starch). Removal of these CBMs from Amy13K reduces the activity level of the enzyme toward corn starches by ∼40-fold, down to the level of activity toward potato starch, suggesting that the CBMs facilitate activity on corn starch and allow its utilization in vivo. The specificity of the Amy13K CBMs provides a molecular rationale for why E. rectale is able to only use certain starch types without the aid of other organisms.
Collapse
Affiliation(s)
- Darrell W Cockburn
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Food Science, Pennsylvania State University, University Park, PA 16802, USA
| | - Carolyn Suh
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Krizia Perez Medina
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Rebecca M Duvall
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Zdzislaw Wawrzak
- Life Sciences Collaborative Access Team (LS-CAT), Advanced Photon Source, Argonne National Laboratory, 9700 S. Cass Avenue, Argonne, IL, 60439, USA
| | - Bernard Henrissat
- Architecture et Fonction des Macromolécules Biologiques, CNRS, Aix-Marseille University, Marseille, F-13288, France.,Institut National de la Recherche Agronomique, USC1408 Architecture et Fonction des Macromolécules Biologiques, Marseille, F-13288, France.,Department of Biological Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Nicole M Koropatkin
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
122
|
Chotirmall SH, Gellatly SL, Budden KF, Mac Aogain M, Shukla SD, Wood DLA, Hugenholtz P, Pethe K, Hansbro PM. Microbiomes in respiratory health and disease: An Asia-Pacific perspective. Respirology 2017; 22:240-250. [PMID: 28102970 DOI: 10.1111/resp.12971] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/30/2016] [Accepted: 11/17/2016] [Indexed: 02/06/2023]
Abstract
There is currently enormous interest in studying the role of the microbiome in health and disease. Microbiome's role is increasingly being applied to respiratory diseases, in particular COPD, asthma, cystic fibrosis and bronchiectasis. The changes in respiratory microbiomes that occur in these diseases and how they are modified by environmental challenges such as cigarette smoke, air pollution and infection are being elucidated. There is also emerging evidence that gut microbiomes play a role in lung diseases through the modulation of systemic immune responses and can be modified by diet and antibiotic treatment. There are issues that are particular to the Asia-Pacific region involving diet and prevalence of specific respiratory diseases. Each of these issues is further complicated by the effects of ageing. The challenges now are to elucidate the cause and effect relationships between changes in microbiomes and respiratory diseases and how to translate these into new treatments and clinical care. Here we review the current understanding and progression in these areas.
Collapse
Affiliation(s)
- Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Shaan L Gellatly
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Kurtis F Budden
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Micheál Mac Aogain
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - David L A Wood
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Kevin Pethe
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| |
Collapse
|
123
|
Effects of new and emerging therapies on gastrointestinal outcomes in cystic fibrosis. Curr Opin Pulm Med 2017; 23:551-555. [DOI: 10.1097/mcp.0000000000000423] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
124
|
Fecal Calprotectin in Cystic Fibrosis and Its Relation to Disease Parameters: A Longitudinal Analysis for 12 Years. J Pediatr Gastroenterol Nutr 2017; 65:438-442. [PMID: 28207476 DOI: 10.1097/mpg.0000000000001544] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Fecal calprotectin (FC) is a marker of inflammation in the intestinal tract. We assessed FC levels longitudinally in patients with cystic fibrosis (CF) and evaluated the relation between FC results and relevant markers of disease. METHODS Calprotectin was measured in fecal samples starting in 2003 and values were stored in the center's patient database. In this retrospective analysis, we searched for associations of FC concentrations with disease severity and progression. Linear mixed effects models were used to model the logarithm of FC levels. RESULTS A total of 171 patients (0-61 years) had 2434 FC measurements between 2003 and 2015, with a total observation period of 1686 patient-years. Median (interquartile range) FC concentrations were 60.9 (75.9) μg/g and 61% of the samples showed elevated FC concentrations (>50 μg/g). Despite some statistically significant effects, there was no clinically relevant association among FC and sex, age, forced expiratory volume in 1 second z score, or body mass index z score. Pancreatic insufficiency (ie, fecal elastase <100 μg/g stool) was associated with considerably higher FC values compared to normal pancreatic function (median FC 68 vs 29 μg/g, P < 0.0001). F508del homozygous subjects showed a trend to higher FC values than heterozygous patients (median 71 vs 62 μg/g, P = 0.173). In addition, a significant association with increasing serum C-reactive protein concentrations (P < 0.0001) was observed. CONCLUSIONS FC was elevated in two-thirds of stool specimens. Increased FC was more common in patients with pancreatic insufficiency. Whether increased FC reflects intestinal inflammation in patients with CF remains to be determined.
Collapse
|
125
|
Reid G, Abrahamsson T, Bailey M, Bindels LB, Bubnov R, Ganguli K, Martoni C, O'Neill C, Savignac HM, Stanton C, Ship N, Surette M, Tuohy K, van Hemert S. How do probiotics and prebiotics function at distant sites? Benef Microbes 2017; 8:521-533. [PMID: 28726511 DOI: 10.3920/bm2016.0222] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2025]
Abstract
The realisation that microbes regarded as beneficial to the host can impart effects at sites distant from their habitat, has raised many possibilities for treatment of diseases. The objective of a workshop hosted in Turku, Finland, by the International Scientific Association for Probiotics and Prebiotics, was to assess the evidence for these effects and the extent to which early life microbiome programming influences how the gut microbiota communicates with distant sites. In addition, we examined how probiotics and prebiotics might affect the skin, airways, heart, brain and metabolism. The growing levels of scientific and clinical evidence showing how microbes influence the physiology of many body sites, leads us to call for more funding to advance a potentially exciting avenue for novel therapies for many chronic diseases.
Collapse
Affiliation(s)
- G Reid
- 1 Lawson Health Research Institute, F3-106, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
- 2 Departments of Microbiology & Immunology and Surgery, The University of Western Ontario, London, Canada
| | - T Abrahamsson
- 3 Department of Clinical and Experimental Medicine, Division of Paediatrics, Linköping University, Linköping, Sweden
| | - M Bailey
- 4 Department of Pediatrics, Ohio State University, College of Medicine, Columbus, OH, USA
- 5 Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - L B Bindels
- 6 Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Avenue Mounier 73, P.O. Box B1.73.11, 1200 Brussels, Belgium
| | - R Bubnov
- 7 Zabolotny Institute of Microbiology and Virology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - K Ganguli
- 8 Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children and Harvard Medical School, Charlestown, MA, 02114, USA
| | - C Martoni
- 9 UAS Laboratories, 4027 Owl Creek Drive, Madison, WI 53718, USA
| | - C O'Neill
- 10 Centre for Dermatology, Faculty of Biology Medicine and Health, The University of Manchester, UK
| | - H M Savignac
- 11 Former (during ISAPP): Clasado Research Services Ltd, Reading, United Kingdom; present: 4D Pharma PLC, Life Sciences Innovation Building, Cornhill Road, Aberdeen, AB25 2ZS, United Kingdom
| | - C Stanton
- 12 APC Microbiome Institute, Biosciences Building, University College Cork, Ireland
| | - N Ship
- 13 Bio-K+ Pharma Inc., 495 Boulevard Armand-Frappier, Laval QC, H7V 4B3 Canada
| | - M Surette
- 14 Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton ON, L8S 4K1 Canada
| | - K Tuohy
- 15 Nutrition and Nutrigenomics Unit, Department of Food Quality and Nutrition, Fondazione Edmund Mach, San Michele all'Adige, Trento, 38010, Italy
| | - S van Hemert
- 16 Winclove Probiotics, Hulstweg 11, 1032 LB Amsterdam, the Netherlands
| |
Collapse
|
126
|
O'Dwyer DN, Dickson RP, Moore BB. The Lung Microbiome, Immunity, and the Pathogenesis of Chronic Lung Disease. THE JOURNAL OF IMMUNOLOGY 2017; 196:4839-47. [PMID: 27260767 DOI: 10.4049/jimmunol.1600279] [Citation(s) in RCA: 245] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/24/2016] [Indexed: 12/17/2022]
Abstract
The development of culture-independent techniques for microbiological analysis has uncovered the previously unappreciated complexity of the bacterial microbiome at various anatomic sites. The microbiome of the lung has relatively less bacterial biomass when compared with the lower gastrointestinal tract yet displays considerable diversity. The composition of the lung microbiome is determined by elimination, immigration, and relative growth within its communities. Chronic lung disease alters these factors. Many forms of chronic lung disease demonstrate exacerbations that drive disease progression and are poorly understood. Mounting evidence supports ways in which microbiota dysbiosis can influence host defense and immunity, and in turn may contribute to disease exacerbations. Thus, the key to understanding the pathogenesis of chronic lung disease may reside in deciphering the complex interactions between the host, pathogen, and resident microbiota during stable disease and exacerbations. In this brief review we discuss new insights into these labyrinthine relationships.
Collapse
Affiliation(s)
- David N O'Dwyer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109; and
| | - Robert P Dickson
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109; and
| | - Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109; and Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
127
|
Nikniaz Z, Nikniaz L, Bilan N, Somi MH, Faramarzi E. Does probiotic supplementation affect pulmonary exacerbation and intestinal inflammation in cystic fibrosis: a systematic review of randomized clinical trials. World J Pediatr 2017; 13:307-313. [PMID: 28470579 DOI: 10.1007/s12519-017-0033-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 10/31/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Patients with cystic fibrosis (CF) usually have abnormal intestinal microbiota due to massive exposure to antibiotics. Probiotics could modify the gut microbiota and hence may affect CF management. So the aim of present systematic review was evaluation of the efficacy and safety of probiotic supplementation for the management of cystic fibrosis. DATA SOURCES We searched PubMed, Science Direct, Google Scholar, Springer Cochrane Library Databases until January 2016 for randomized controlled trials (RCTs) performed in pediatric or adult populations related to the study aim. Key words were selected based on Mesh terms. Based on the Critical Appraisal Skills Programme checklist, eligibility of included articles was evaluated. RESULTS Five studies included in this review represent 188 participants with a follow up period ranging from 1 month to 6 months. The results of the included studies supporting the use of probiotics in management of pulmonary exacerbation and intestinal calprotectin in patients with cystic fibrosis. However the level of evidence was limited. CONCLUSIONS The lack of high quality RCTs makes it impossible to support a general recommendation about the use of probiotics in the treatment of CF pulmonary exacerbation and intestinal inflammation.
Collapse
Affiliation(s)
- Zeinab Nikniaz
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Nikniaz
- Tabriz Health Services Management Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Nemat Bilan
- Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hossein Somi
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elnaz Faramarzi
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
128
|
Buford TW. (Dis)Trust your gut: the gut microbiome in age-related inflammation, health, and disease. MICROBIOME 2017; 5:80. [PMID: 28709450 PMCID: PMC5512975 DOI: 10.1186/s40168-017-0296-0] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/04/2017] [Indexed: 05/08/2023]
Abstract
Chronic inflammation represents one of the most consistent biologic features of aging. However, the precise etiology of persistent low-grade increases in inflammation remains unclear. Recent evidence suggests that the gut microbiome may play a key role in age-related inflammation. Indeed, several studies have indicated that older adults display an altered composition of the gut microbiota, and early evidence indicates that this dysbiosis is associated with the presence of several key circulating inflammatory analytes. The present review summarizes knowledge on age-related inflammation and discusses how potential relationships with gut dysbiosis may lead to novel treatment strategies in the future."The pattern of disease is an expression of the response of man to his total environment (physical, biological, and social); this response is, therefore, determined by anything that affects man himself or his environment." - Rene Dubos, 1961.
Collapse
Affiliation(s)
- Thomas W Buford
- Department of Medicine, University of Alabama at Birmingham, 933 19th Street South, Birmingham, AL, 35294, USA.
| |
Collapse
|
129
|
Rocchetti G, Lucini L, Chiodelli G, Giuberti G, Gallo A, Masoero F, Trevisan M. Phenolic profile and fermentation patterns of different commercial gluten-free pasta during in vitro large intestine fermentation. Food Res Int 2017; 97:78-86. [DOI: 10.1016/j.foodres.2017.03.035] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/12/2017] [Accepted: 03/19/2017] [Indexed: 12/12/2022]
|
130
|
Gasta MG, Gossard CM, Williamson CB, Dolan KE, Finley HJ, Burns CM, Parker EC, Pizano JM, Lipski EA. Probiotics and Disease: A Comprehensive Summary-Part 5, Respiratory Conditions of the Ears, Nose, and Throat. Integr Med (Encinitas) 2017; 16:28-40. [PMID: 30881245 PMCID: PMC6419786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
This article series provides a literature review of the disease-specific probiotic strains studied in published clinical trials in humans and animals. The goal of the series is to provide clinically useful tools. The table design allows for quick access to supportive data and will be helpful as a guide for both researchers and clinicians. The first article (part 1) focused on mental health and neurological conditions and the second article (part 2) explored cultured and fermented foods that are commonly available in the United States. The third article (part 3) explored the relationship between bacterial strains and 2 of the most prevalent diseases we have in modern society, cardiometabolic disease and fatigue syndromes. The fourth article (part 4) elucidated the role of the microbiome in infectious diseases, and this fifth article (part 5) investigates probiotic strains on respiratory conditions that affect the ears, nose, and throat. Future articles will review conditions related to autoimmunity and dermatological conditions; the influence of the microbiome on cancer development and prognosis; gastrointestinal and genitourinary diseases associated with dysbiosis conditions; followed by an article focused on probiotic supplements. This literature review is specific to disease condition, probiotic classification, and individual strain.
Collapse
|
131
|
Tabori H, Arnold C, Jaudszus A, Mentzel HJ, Renz DM, Reinsch S, Lorenz M, Michl R, Gerber A, Lehmann T, Mainz JG. Abdominal symptoms in cystic fibrosis and their relation to genotype, history, clinical and laboratory findings. PLoS One 2017; 12:e0174463. [PMID: 28472055 PMCID: PMC5417419 DOI: 10.1371/journal.pone.0174463] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 03/09/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND & AIMS Abdominal symptoms (AS) are a hallmark of the multiorgan-disease cystic fibrosis (CF). However, the abdominal involvement in CF is insufficiently understood and, compared to the pulmonary manifestation, still receives little scientific attention. Aims were to assess and quantify AS and to relate them to laboratory parameters, clinical findings, and medical history. METHODS A total of 131 patients with CF of all ages were assessed with a new CF-specific questionnaire (JenAbdomen-CF score 1.0) on abdominal pain and non-pain symptoms, disorders of appetite, eating, and bowel movements as well as symptom-related quality of life. Results were metrically dimensioned and related to abdominal manifestations, history of surgery, P. aeruginosa and S. aureus colonization, genotype, liver enzymes, antibiotic therapy, lung function, and nutritional status. RESULTS AS during the preceding 3 months were reported by all of our patients. Most common were lack of appetite (130/131) and loss of taste (119/131) followed by abdominal pain (104/131), flatulence (102/131), and distention (83/131). Significantly increased AS were found in patients with history of rectal prolapse (p = 0.013), distal intestinal obstruction syndrome (p = 0.013), laparotomy (p = 0.022), meconium ileus (p = 0.037), pancreas insufficiency (p = 0.042), or small bowel resection (p = 0.048) as well as in patients who have been intermittently colonized with P. aeruginosa (p = 0.006) compared to patients without history of these events. In contrast, no statistically significant associations were found to CF-associated liver disease, chronic pathogen colonization, lung function, CF-related diabetes, and nutritional status. CONCLUSION As the complex abdominal involvement in CF is still not fully understood, the assessment of the common AS is of major interest. In this regard, symptom questionnaires like the herein presented are meaningful and practical tools facilitating a wider understanding of the abdominal symptoms in CF. Furthermore, they render to evaluate possible abdominal effects of novel modulators of the underlying cystic fibrosis transmembrane (conductance) regulator (CFTR) defect.
Collapse
Affiliation(s)
- Harold Tabori
- Jena University Hospital, Cystic Fibrosis Center, Jena, Germany
| | - Christin Arnold
- Jena University Hospital, Cystic Fibrosis Center, Jena, Germany
| | - Anke Jaudszus
- Jena University Hospital, Cystic Fibrosis Center, Jena, Germany
| | | | - Diane M. Renz
- Jena University Hospital, Pediatric Radiology, Jena, Germany
| | - Steffen Reinsch
- Jena University Hospital, Pediatric Gastroenterology, Jena, Germany
| | - Michael Lorenz
- Jena University Hospital, Cystic Fibrosis Center, Jena, Germany
| | - Ruth Michl
- Jena University Hospital, Cystic Fibrosis Center, Jena, Germany
| | - Andrea Gerber
- Jena University Hospital, Cystic Fibrosis Center, Jena, Germany
| | - Thomas Lehmann
- Jena University Hospital, Institute of Medical Statistics, Jena, Germany
| | - Jochen G. Mainz
- Jena University Hospital, Cystic Fibrosis Center, Jena, Germany
| |
Collapse
|
132
|
Mirza A, Mao-Draayer Y. The gut microbiome and microbial translocation in multiple sclerosis. Clin Immunol 2017; 183:213-224. [PMID: 28286112 DOI: 10.1016/j.clim.2017.03.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/03/2017] [Accepted: 03/07/2017] [Indexed: 02/07/2023]
Abstract
Individuals with multiple sclerosis (MS) have a distinct intestinal microbial community (microbiota) and increased low-grade translocation of bacteria from the intestines into the circulation. The observed change of intestinal bacteria in MS patients regulate immune functions involved in MS pathogenesis. These functions include: systemic and central nervous system (CNS) immunity (including peripheral regulatory T cell function), the blood-brain barrier (BBB) permeability and CNS-resident cell activity. This review discusses the MS intestinal microbiota implication on MS systemic- and CNS-immunopathology. We introduce the possible contributions of MS low-grade microbial translocation (LG-MT) to the development of MS, and end on a discussion on microbiota therapies for MS patients.
Collapse
Affiliation(s)
- Ali Mirza
- Department of Microbiology and Immunology, University of Michigan School of Medicine, 4258 Alfred Taubman Biomedical Sciences Research Bldg. 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States; Department of Neurology, University of Michigan School of Medicine, 4258 Alfred Taubman Biomedical Sciences Research Bldg. 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan School of Medicine, 4015 Alfred Taubman Biomedical Sciences Research Bldg. 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States.
| |
Collapse
|
133
|
Van Biervliet S, Declercq D, Somerset S. Clinical effects of probiotics in cystic fibrosis patients: A systematic review. Clin Nutr ESPEN 2017; 18:37-43. [PMID: 29132736 DOI: 10.1016/j.clnesp.2017.01.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 01/27/2017] [Indexed: 02/08/2023]
Abstract
Cystic fibrosis (CF) is characterised by a build-up of thick, intransient mucus linings of the digestive and respiratory mucosa, which disrupts digestive system functioning and microbiota composition. In view of the potential for probiotics to enhance microbiota composition in other contexts, this study investigated the current evidence for probiotics as an adjunct to usual therapy for CF. Electronic clinical databases were interrogated for human randomised, controlled, intervention trials (1985-2015) testing the effects of probiotics on clinical endpoints in CF were reviewed. From 191 articles identified in initial searches, six studies met the critical inclusion criteria, and were reviewed in detail. These studies varied in size (n = 22 to 61) but were generally small and showed substantial diversity in protocol, specific probiotic species used and range of clinical outcomes measured. Probiotic administration showed beneficial effects on fecal calprotectin levels, pulmonary exacerbation risk, and quality of life indicators. In one study, such changes were associated with variations in gut microbiota composition. Despite encouraging preliminary results, the limited number of small and highly varied studies to date do not justify the addition of probiotics as an adjunct to current CF treatment protocols. Importantly, very minimal adverse effects of probiotics have been reported.
Collapse
Affiliation(s)
| | | | - Shawn Somerset
- School of Allied Health, Faculty of Health Sciences, Australian Catholic University, Brisbane, Queensland, Australia.
| |
Collapse
|
134
|
Garg M, Ooi CY. The Enigmatic Gut in Cystic Fibrosis: Linking Inflammation, Dysbiosis, and the Increased Risk of Malignancy. Curr Gastroenterol Rep 2017; 19:6. [PMID: 28155088 DOI: 10.1007/s11894-017-0546-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Intestinal inflammation, dysbiosis, and increased gastrointestinal malignancy risks are well-described in patients with cystic fibrosis (CF). However, there is limited understanding of their pathophysiology. This review aims to discuss these issues and assess potential links between them. RECENT FINDINGS Evidence of links between intestinal inflammation and dysbiosis (an imbalance in intestinal microbial populations) exist. Recent studies have demonstrated reduction in intestinal inflammation with probiotic administration. Both bacterial dysbiosis and gut inflammation contribute to the suboptimal nutritional status seen in children with CF. Short-chain fatty acids may be reduced in the gut lumen as a result of bacterial imbalances and may promote inflammation. Inflammation and bacterial dysbiosis in CF may also contribute to emerging adult complications such as gastrointestinal malignancy. An increase in carcinogenic microbes and reduction in microbes protective against cancer have been found in CF, linking bacterial dysbiosis and cancer. Murine studies suggest the CF gene, cystic fibrosis transmembrane conductance regulator (CFTR) gene, itself may be a tumour suppressor gene. The pathophysiology of interactions among intestinal inflammation, dysbiosis, and malignancy in CF is not clearly understood and requires further research.
Collapse
Affiliation(s)
- Millie Garg
- School of Women's and Children's Health, Medicine, University of New South Wales, Randwick, NSW, 2031, Australia
| | - Chee Y Ooi
- School of Women's and Children's Health, Medicine, University of New South Wales, Randwick, NSW, 2031, Australia.
- Department of Paediatric Gastroenterology, Sydney Children's Hospital, Randwick, NSW, 2031, Australia.
| |
Collapse
|
135
|
Normative Fecal Calprotectin Concentrations in Guatemalan Preschoolers Are High Relative to Children Reported Elsewhere. J Pediatr Gastroenterol Nutr 2017; 64:238-244. [PMID: 27101538 DOI: 10.1097/mpg.0000000000001241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND Calprotectin is a fecal marker of intraintestinal inflammation derived from activated enteric neutrophils and macrophages. It is useful as a clinical marker in inflammatory bowel diseases; furthermore, it may have a role in public health epidemiology. OBJECTIVES The aim of the study was to describe the distribution of fecal calprotectin in Guatemalan preschool children sharing a common institutional diet; to relate it collectively to pediatric distributions in other geographic settings, and individually to concomitant indicators of intestinal infection or colonization and other descriptive features of the child. METHODS Fecal samples were collected in 87 subjects, ages 2 to 7 years across 3 daycare centers sharing a common institutional menu, but from different ecological settings. Stools were examined, variously by routine light microscopy, quantitative egg counts, and a Giardia antigen test, for microbiological diagnosis, and an ELISA assay for fecal calprotectin (CalproLab). RESULTS The median fecal calprotectin value was 58 mg/kg, with a mean of 98 ± 136 mg/kg and a range from 10 to 950 mg/kg; 61% of values were above the manufacturer's cut-off for elevated concentration and 51% exceeded an age-adjusted criterion. There were no associations between sex, age, growth indicators, or fecal microbiological findings by microscopy or ELISA assays, alone or in combination. The central tendency (mean or median) and distribution were generally shifted to the right in relation to comparable reports from children across the world literature. CONCLUSIONS Although specific, low-grade intestinal infections do not define calprotectin subgroups, right-shifted fecal calprotectin status in this population may reflect a general and diffuse stress of adverse environmental sanitation.
Collapse
|
136
|
Boutin S, Dalpke AH. Acquisition and adaptation of the airway microbiota in the early life of cystic fibrosis patients. Mol Cell Pediatr 2017; 4:1. [PMID: 28097632 PMCID: PMC5241261 DOI: 10.1186/s40348-016-0067-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/15/2016] [Indexed: 12/26/2022] Open
Abstract
Cystic fibrosis (CF) is a genetic disease in which bacterial infections of the airways play a major role in the long-term clinical outcome. In recent years, a number of next-generation sequencing (NGS)-based studies aimed at deciphering the structure and composition of the airways’ microbiota. It was shown that the nasal cavity of CF patients displays dysbiosis early in life indicating a failure in the first establishment of a healthy microbiota. In contrast, within the conducting and lower airways, the establishment occurs normally first, but is sensitive to future dysbiosis including chronic infections with classical pathogens in later life. The objective of this mini-review is to give an update on the current knowledge about the development of the microbiota in the early life of CF patients. Microbial acquisition in the human airways can be described by the island model: Microbes found in the lower airways of CF patients represent “islands” that are at first populated from the upper airways reflecting the “mainland.” Colonization can be modeled following the neutral theory in which the most abundant bacteria in the mainland are also frequently found in the lower airways initially. At later times, however, the colonization process of the lower airways segregates by active selection of specific microbes. Future research should focus on those processes of microbial and host interactions to understand how microbial communities are shaped on short- and long-term scales. We point out what therapeutic consequences arise from the microbiome data obtained within ecological framework models.
Collapse
Affiliation(s)
- Sébastien Boutin
- Department of Infectious Disease, Medical Microbiology and Hygiene, University Hospital Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Alexander H Dalpke
- Department of Infectious Disease, Medical Microbiology and Hygiene, University Hospital Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany. .,Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany.
| |
Collapse
|
137
|
Miragoli F, Federici S, Ferrari S, Minuti A, Rebecchi A, Bruzzese E, Buccigrossi V, Guarino A, Callegari ML. Impact of cystic fibrosis disease on archaea and bacteria composition of gut microbiota. FEMS Microbiol Ecol 2016; 93:fiw230. [PMID: 27810876 PMCID: PMC5155554 DOI: 10.1093/femsec/fiw230] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/08/2016] [Accepted: 11/02/2016] [Indexed: 12/21/2022] Open
Abstract
Cystic fibrosis is often associated with intestinal inflammation due to several factors, including altered gut microbiota composition. In this study, we analyzed the fecal microbiota among patients with cystic fibrosis of 10–22 years of age, and compared the findings with age-matched healthy subjects. The participating patients included 14 homozygotes and 14 heterozygotes with the delF508 mutation, and 2 heterozygotes presenting non-delF508 mutations. We used PCR-DGGE and qPCR to analyze the presence of bacteria, archaea and sulfate-reducing bacteria. Overall, our findings confirmed disruption of the cystic fibrosis gut microbiota. Principal component analysis of the qPCR data revealed no differences between homozygotes and heterozygotes, while both groups were distinct from healthy subjects who showed higher biodiversity. Archaea were under the detection limit in all homozygotes subjects, whereas methanogens were detected in 62% of both cystic fibrosis heterozygotes and healthy subjects. Our qPCR results revealed a low frequency of sulfate-reducing bacteria in the homozygote (13%) and heterozygote (13%) patients with cystic fibrosis compared with healthy subjects (87.5%). This is a pioneer study showing that patients with cystic fibrosis exhibit significant reduction of H2-consuming microorganisms, which could increase hydrogen accumulation in the colon and the expulsion of this gas through non-microbial routes.
Collapse
Affiliation(s)
- Francesco Miragoli
- Centro Ricerche Biotecnologiche, Università Cattolica del Sacro Cuore, Cremona 26100, Italy
| | - Sara Federici
- Centro Ricerche Biotecnologiche, Università Cattolica del Sacro Cuore, Cremona 26100, Italy
| | - Susanna Ferrari
- Centro Ricerche Biotecnologiche, Università Cattolica del Sacro Cuore, Cremona 26100, Italy
| | - Andrea Minuti
- Istituto di Zootecnica, Università Cattolica del Sacro Cuore, Piacenza 29122, Italy
| | - Annalisa Rebecchi
- Centro Ricerche Biotecnologiche, Università Cattolica del Sacro Cuore, Cremona 26100, Italy
| | - Eugenia Bruzzese
- Department of Translational Medical Sciences, Section of Pediatrics, University Federico II, Naples 80131, Italy
| | - Vittoria Buccigrossi
- Department of Translational Medical Sciences, Section of Pediatrics, University Federico II, Naples 80131, Italy
| | - Alfredo Guarino
- Department of Translational Medical Sciences, Section of Pediatrics, University Federico II, Naples 80131, Italy
| | - Maria Luisa Callegari
- Centro Ricerche Biotecnologiche, Università Cattolica del Sacro Cuore, Cremona 26100, Italy
| |
Collapse
|
138
|
Bayes HK, Ritchie ND, Ward C, Corris PA, Brodlie M, Evans TJ. IL-22 exacerbates weight loss in a murine model of chronic pulmonary Pseudomonas aeruginosa infection. J Cyst Fibros 2016; 15:759-768. [PMID: 27375092 PMCID: PMC5154339 DOI: 10.1016/j.jcf.2016.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 01/20/2023]
Abstract
BACKGROUND Interleukin (IL)-22 is a critical mediator of mucosal immunity and tissue regeneration, protecting against a number of respiratory pathogens. Whether IL-22 confers protection against chronic Pseudomonas aeruginosa (PA) infection in cystic fibrosis (CF) is unknown. METHODS Explanted CF lungs were examined for IL-22 production and immune-localization. A murine model of persistent pulmonary PA infection was used to examine production of IL-22 following infective challenge. The role of IL-22 was examined using IL-22 knockout (KO) animals. RESULTS IL-22 is produced within the adult CF lung and localizes to the airway epithelium. IL-22 is produced by murine pulmonary lymph node cells following lung infection. The absence of IL-22 resulted in no significant difference in acute mortality, bacterial burden, chronic infection rates, histological changes or neutrophilic inflammation in the chronic PA infection model. However, IL-22 KO animals lost less weight following infection. CONCLUSION IL-22 is produced in the CF lung and in response to PA infection yet is dispensable in protection against chronic pulmonary P. aeruginosa infection in a murine model. However, we identified a novel role for the cytokine in promoting infection-related weight-loss, a significant prognostic factor in the CF population.
Collapse
Affiliation(s)
- Hannah K Bayes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA, United Kingdom.
| | - Neil D Ritchie
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Christopher Ward
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Paul A Corris
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK; Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne NE7 7DN, UK
| | - Malcolm Brodlie
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK; Paediatric Respiratory Medicine, Great North Children's Hospital, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK
| | - Thomas J Evans
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA, United Kingdom
| |
Collapse
|
139
|
Hodgkinson AJ, Young W, Cakebread JA, Haigh BJ. Feeding bovine milks with low or high IgA levels is associated with altered re-establishment of murine intestinal microbiota after antibiotic treatment. PeerJ 2016; 4:e2518. [PMID: 27703861 PMCID: PMC5047218 DOI: 10.7717/peerj.2518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/02/2016] [Indexed: 11/30/2022] Open
Abstract
Antibiotics are a vital and commonly used therapeutic tool, but their use also results in profound changes in the intestinal microbiota that can, in turn, have significant health consequences. Understanding how the microbiota recovers after antibiotic treatment will help to devise strategies for mitigating the adverse effects of antibiotics. Using a mouse model, we have characterized the changes occurring in the intestinal microbiota immediately after five days exposure to ampicillin, and then at three and fourteen days thereafter. During the fourteen day period of antibiotic recovery, groups of mice were fed either water, cows’ milk containing high levels of IgA, or cows’ milk containing low levels of IgA as their sole source of liquid. Effects on microbiota of feeding milks for 14 days were also assessed in groups of mice that had no ampicillin exposure. Changes in microbiota were measured by high throughput sequencing of the V4 to V6 variable regions of the 16S ribosomal RNA gene. As expected, exposure to ampicillin led to profound changes to the types and abundance of bacteria present, along with a loss of diversity. At 14 days following antibiotic exposure, mice fed water had recovered microbiota compositions similar to that prior to antibiotics. However, feeding High-IgA milk to mice that has been exposed to antibiotics was associated with altered microbiota compositions, including increased relative abundance of Lactobacillus and Barnesiella compared to the start of the study. Mice exposed to antibiotics then fed Low-IgA milk also showed increased Barnesiella at day 14. Mice without antibiotic perturbation, showed no change in their microbiota after 14 days of milk feeding. Overall, these findings add to a knowledge platform for optimizing intestinal function after treatment with antibiotics in the human population.
Collapse
Affiliation(s)
| | - Wayne Young
- Food & Bio-based Products, AgResearch , Palmerston North , New Zealand
| | | | - Brendan J Haigh
- Food & Bio-based Products, AgResearch , Hamilton , New Zealand
| |
Collapse
|
140
|
Rogers GB, Narkewicz MR, Hoffman LR. The CF gastrointestinal microbiome: Structure and clinical impact. Pediatr Pulmonol 2016; 51:S35-S44. [PMID: 27662102 PMCID: PMC5303757 DOI: 10.1002/ppul.23544] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 02/06/2023]
Abstract
The gastrointestinal (GI) microbiome is shaped by host diet, immunity, and other physicochemical characteristics of the GI tract, and perturbations such as antibiotic treatments can lead to persistent changes in microbial constituency and function. These GI microbes also play critical roles in host nutrition and health. A growing body of evidence suggests that the GI microbiome in people with CF is altered, and that these dysbioses contribute to disease manifestations in many organs, both within and beyond the GI tract. Therapies that people with CF receive, even those targeting the respiratory tract, may impact the CF GI microbiome in ways that can influence the outcome of treatment. These new perspectives on the microbial contents of the CF intestine offer new opportunities for preventing a variety of CF-associated disorders. Pediatr Pulmonol. 2016;51:S35-S44. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Geraint B Rogers
- SAHMRI Infection and Immunity Theme, School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Michael R Narkewicz
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado.,Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado
| | - Lucas R Hoffman
- Departments of Pediatrics and Microbiology, University of Washington, Seattle, Washington. .,Seattle Children's Hospital, Seattle, Washington.
| |
Collapse
|
141
|
Ananthan A, Balasubramanian H, Rao S, Patole S. Probiotic supplementation in children with cystic fibrosis-a systematic review. Eur J Pediatr 2016; 175:1255-66. [PMID: 27576473 DOI: 10.1007/s00431-016-2769-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 08/09/2016] [Accepted: 08/19/2016] [Indexed: 12/17/2022]
Abstract
UNLABELLED Probiotics may benefit in cystic fibrosis (CF) as gut dysbiosis is associated with gastrointestinal symptoms and exacerbation of respiratory symptoms in CF. We conducted a systematic review of randomized controlled trials (RCTs) and non-RCTs of probiotic supplementation in children with CF, using the Cochrane methodology, preferred reporting items for systematic reviews (PRISMA) statement, and meta-analysis of observational studies in epidemiology (MOOSE) guidelines. Primary outcomes were pulmonary exacerbations, duration of hospitalization and antibiotics, and all-cause mortality. Secondary outcomes included gastrointestinal symptoms, markers of gut inflammation, and intestinal microbial balance. A total of nine studies (RCTs, 6, non-RCTs, 3; N = 275) with some methodological weaknesses were included in the review. The pooled estimate showed significant reduction in the rate of pulmonary exacerbation (fixed effects model, two parallel group RCTs and one cross-over trial: relative risk (RR) 0.25, (95 % confidence interval (95 % CI) 0.15,0.41); p < 0.00001; level of evidence: low) and decrease in fecal calprotectin (FCLP) levels (fixed effect model, three RCTs: mean difference (MD) -16.71, 95 % CI -27.30,-6.13); p = 0.002; level of evidence: low) after probiotic supplementation. Probiotic supplementation significantly improved gastrointestinal symptoms (one RCT, one non-RCT) and gut microbial balance (decreased Proteobacteria, increased Firmicutes, and Bacteroides in one RCT, one non-RCT). CONCLUSION Limited low-quality evidence exists on the effects of probiotics in children with CF. Well-designed adequately powered RCTs assessing clinically meaningful outcomes are required to study this important issue. WHAT IS KNOWN • Gut dysbiosis is frequent in children with cystic fibrosis due to frequent exposure to pathogens and antibiotics. • Probiotics decrease gut dysbiosis and improve gut maturity and function. What is New: • This comprehensive systematic review shows that current evidence on the safety and efficacy of probiotics in children with cystic fibrosis is limited and of low quality. • Well-designed and adequately powered trials assessing clinically important outcomes are required considering the health burden of cystic fibrosis and the potential benefits of probiotics.
Collapse
Affiliation(s)
- Anitha Ananthan
- Department of Neonatal Paediatrics, King Edward Memorial Hospital for Women, 378 Bagot Road, Subiaco, Perth, WA, 6008, Australia.
| | - Haribalakrishna Balasubramanian
- Department of Neonatal Paediatrics, King Edward Memorial Hospital for Women, 378 Bagot Road, Subiaco, Perth, WA, 6008, Australia
| | - Shripada Rao
- Department of Neonatal Paediatrics, Princess Margaret Hospital for Children, Perth, WA, Australia.,Centre for Neonatal Research and Education, University of Western Australia, Crawley, Australia
| | - Sanjay Patole
- Department of Neonatal Paediatrics, King Edward Memorial Hospital for Women, 378 Bagot Road, Subiaco, Perth, WA, 6008, Australia.,Centre for Neonatal Research and Education, University of Western Australia, Crawley, Australia
| |
Collapse
|
142
|
Anderson JL, Miles C, Tierney AC. Effect of probiotics on respiratory, gastrointestinal and nutritional outcomes in patients with cystic fibrosis: A systematic review. J Cyst Fibros 2016; 16:186-197. [PMID: 27693010 DOI: 10.1016/j.jcf.2016.09.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 08/12/2016] [Accepted: 09/13/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND An increasing body of research investigating the use of probiotics to improve health outcomes in patients with cystic fibrosis (CF) prompted the need to systematically assess and summarise the relevant literature. METHODS An electronic search of five databases and three trial databases was conducted. Studies describing the administration of probiotics to patients with CF older than 2years, with a comparator group on respiratory, gastrointestinal and nutritional outcomes were included. RESULTS Three pre-post studies and six randomised controlled trials met the inclusion criteria. Overall studies showed a positive effect of probiotics on reducing the number of pulmonary exacerbations and decreasing gastrointestinal inflammation. There was limited effect of probiotics on other outcomes and inadequate evidence for the effects of specific probiotic species and strains. CONCLUSION The findings suggest that probiotics may improve respiratory and gastrointestinal outcomes in a stable CF clinic population with no reported evidence of harm. There is inadequate evidence at this time to recommend a specific species, strain or dose of probiotic as likely to be of significant benefit.
Collapse
Affiliation(s)
- Jacqueline L Anderson
- Dietetics and Human Nutrition, School of Allied Health, La Trobe University, Bundoora, Melbourne, Australia.
| | - Caitlin Miles
- Nutrition and Dietetics, Monash Health, Clayton, Victoria, Australia
| | - Audrey C Tierney
- Dietetics and Human Nutrition, School of Allied Health, La Trobe University, Bundoora, Melbourne, Australia; Department of Nutrition and Dietetics, Alfred Health, Prahran, Melbourne, Australia
| |
Collapse
|
143
|
Abstract
Host-microorganism interactions shape local cell functionality, immune responses, and can influence disease development. Evidence indicates that the impact of host-microbe interactions reaches far beyond the local environment, thus influencing responses in peripheral tissues. There is a vital cross-talk between the mucosal tissues of our body, as exemplified by intestinal complications during respiratory disease and vice versa. Although, mechanistically, this phenomenon remains poorly defined, the existence of the gut-lung axis and its implications in both health and disease could be profoundly important for both disease etiology and treatment. In this review, we highlight how changes in the intestinal microenvironment, with a particular focus on the intestinal microbiota, impact upon respiratory disease.
Collapse
|
144
|
Cattaneo A, Cattane N, Galluzzi S, Provasi S, Lopizzo N, Festari C, Ferrari C, Guerra UP, Paghera B, Muscio C, Bianchetti A, Volta GD, Turla M, Cotelli MS, Gennuso M, Prelle A, Zanetti O, Lussignoli G, Mirabile D, Bellandi D, Gentile S, Belotti G, Villani D, Harach T, Bolmont T, Padovani A, Boccardi M, Frisoni GB. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging 2016; 49:60-68. [PMID: 27776263 DOI: 10.1016/j.neurobiolaging.2016.08.019] [Citation(s) in RCA: 932] [Impact Index Per Article: 103.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 08/23/2016] [Accepted: 08/24/2016] [Indexed: 12/14/2022]
Abstract
The pathway leading from amyloid-β deposition to cognitive impairment is believed to be a cornerstone of the pathogenesis of Alzheimer's disease (AD). However, what drives amyloid buildup in sporadic nongenetic cases of AD is still unknown. AD brains feature an inflammatory reaction around amyloid plaques, and a specific subset of the gut microbiota (GMB) may promote brain inflammation. We investigated the possible role of the GMB in AD pathogenesis by studying the association of brain amyloidosis with (1) GMB taxa with pro- and anti-inflammatory activity; and (2) peripheral inflammation in cognitively impaired patients. We measured the stool abundance of selected bacterial GMB taxa (Escherichia/Shigella, Pseudomonas aeruginosa, Eubacterium rectale, Eubacterium hallii, Faecalibacterium prausnitzii, and Bacteroides fragilis) and the blood expression levels of cytokines (pro-inflammatory cytokines: CXCL2, CXCL10, interleukin [IL]-1β, IL-6, IL-18, IL-8, inflammasome complex (NLRP3), tumor necrosis factor-alpha [TNF-α]; anti-inflammatory cytokines: IL-4, IL-10, IL-13) in cognitively impaired patients with (n = 40, Amy+) and with no brain amyloidosis (n = 33, Amy-) and also in a group of controls (n = 10, no brain amyloidosis and no cognitive impairment). Amy+ patients showed higher levels of pro-inflammatory cytokines (IL-6, CXCL2, NLRP3, and IL-1β) compared with both controls and with Amy- patients. A reduction of the anti-inflammatory cytokine IL-10 was observed in Amy+ versus Amy-. Amy+ showed lower abundance of E. rectale and higher abundance of Escherichia/Shigella compared with both healthy controls (fold change, FC = -9.6, p < 0.001 and FC = +12.8, p < 0.001, respectively) and to Amy- (FC = -7.7, p < 0.001 and FC = +7.4, p = 0.003). A positive correlation was observed between pro-inflammatory cytokines IL-1β, NLRP3, and CXCL2 with abundance of the inflammatory bacteria taxon Escherichia/Shigella (rho = 0.60, p < 0.001; rho = 0.57, p < 0.001; and rho = 0.30, p = 0.007, respectively) and a negative correlation with the anti-inflammatory E. rectale (rho = -0.48, p < 0.001; rho = -0.25, p = 0.024; rho = -0.49, p < 0.001). Our data indicate that an increase in the abundance of a pro-inflammatory GMB taxon, Escherichia/Shigella, and a reduction in the abundance of an anti-inflammatory taxon, E. rectale, are possibly associated with a peripheral inflammatory state in patients with cognitive impairment and brain amyloidosis. A possible causal relation between GMB-related inflammation and amyloidosis deserves further investigation.
Collapse
Affiliation(s)
- Annamaria Cattaneo
- Biological Psychiatry Laboratory, IRCCS Fatebenefratelli, Brescia, Italy; King's College London, Institute of Psychiatry, London, UK.
| | - Nadia Cattane
- Biological Psychiatry Laboratory, IRCCS Fatebenefratelli, Brescia, Italy
| | - Samantha Galluzzi
- Laboratory of Neuroimaging and Alzheimer's Epidemiology, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Stefania Provasi
- Biological Psychiatry Laboratory, IRCCS Fatebenefratelli, Brescia, Italy
| | - Nicola Lopizzo
- Biological Psychiatry Laboratory, IRCCS Fatebenefratelli, Brescia, Italy
| | - Cristina Festari
- Laboratory of Neuroimaging and Alzheimer's Epidemiology, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Clarissa Ferrari
- Biological Psychiatry Laboratory, IRCCS Fatebenefratelli, Brescia, Italy
| | - Ugo Paolo Guerra
- Nuclear Medicine, Poliambulanza Foundation, Istituto Ospedaliero, Brescia, Italy
| | - Barbara Paghera
- Nuclear Medicine, University of Brescia and Spedali Civili di Brescia, Brescia, Italy
| | - Cristina Muscio
- Biological Psychiatry Laboratory, IRCCS Fatebenefratelli, Brescia, Italy; European Foundation Biomedical Research (FERB), Center of Excellence Alzheimer, Ospedale Briolini, Gazzaniga, Bergamo, Italy
| | - Angelo Bianchetti
- Department of Medicine and Rehabilitation, Istituto Clinico Sant'Anna, Brescia, Italy
| | | | - Marinella Turla
- Neurology Unit, Ospedale di Vallecamonica Esine, Brescia, Italy
| | | | | | | | - Orazio Zanetti
- Biological Psychiatry Laboratory, IRCCS Fatebenefratelli, Brescia, Italy
| | - Giulia Lussignoli
- Biological Psychiatry Laboratory, IRCCS Fatebenefratelli, Brescia, Italy
| | - Dario Mirabile
- European Foundation Biomedical Research (FERB), Center of Excellence Alzheimer, Ospedale Briolini, Gazzaniga, Bergamo, Italy
| | - Daniele Bellandi
- Alzheimer Evaluation Unit, Istituto Ospedaliero di Sospiro ONLUS Foundation, Cremona, Italy
| | - Simona Gentile
- Department of Rehabilitation, Casa di Cura Ancelle della Carità, Cremona, Italy
| | - Gloria Belotti
- Alzheimer Evaluation Unit, Hospice Santa Maria Ausiliatrice ONLUS Foundation, Bergamo, Italy
| | - Daniele Villani
- Alzheimer Evaluation Unit, Casa di Cura Figlie di S. Camillo, Cremona, Italy
| | - Taoufiq Harach
- Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | | | - Alessandro Padovani
- Neurology Unit, Department of Medical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Marina Boccardi
- Laboratory of Neuroimaging and Alzheimer's Epidemiology, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Giovanni B Frisoni
- Laboratory of Neuroimaging and Alzheimer's Epidemiology, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; LANVIE-Laboratory of Neuroimaging of Aging, University Hospitals, Geneva, Switzerland; Memory Clinic, University Hospitals and University of Geneva, Geneva, Switzerland
| |
Collapse
|
145
|
Li L, Krause L, Somerset S. Associations between micronutrient intakes and gut microbiota in a group of adults with cystic fibrosis. Clin Nutr 2016; 36:1097-1104. [PMID: 27595636 DOI: 10.1016/j.clnu.2016.06.029] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 06/23/2016] [Accepted: 06/30/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Cystic fibrosis (CF) involves chronic inflammation and oxidative stress affecting mainly the respiratory and digestive systems. Survival rates for CF have improved with advances in treatment including nutritional interventions such as micronutrient supplementation. Diet can modulate gut microbiota in the general population with consequences on local and systemic immunity, and inflammation. The gut microbiota appears disrupted and may associate with pulmonary status in CF. This study investigated associations between micronutrient intakes and gut microbiota variations in a group of adults with CF. METHODS Faecal microbiota of sixteen free-living adults with CF was profiled by 16ss rDNA sequencing on the GS-FLX platform. Associations were tested between UniFrac distances of faecal microbiota and time-corresponding micronutrient intakes. Associations between relative abundances of bacterial taxa and micronutrient intakes (those showing significant associations with UniFrac distances) were examined by Spearman correlation. RESULTS Unweighted UniFrac distances were associated with intakes of potassium and antioxidant vitamins C, E and beta-carotene equivalents, whereas weighted UniFrac distances were associated with antioxidant vitamins riboflavin, niacin equivalents, beta-carotene equivalents and vitamin A equivalents. Intakes of beta-carotene equivalents, vitamin C, vitamin E, niacin equivalents and riboflavin correlated negatively with Bacteroides and/or its corresponding higher level taxa. Intakes of beta-carotene equivalents and vitamin E also positively correlated with Firmicutes and specific taxa belonging to Firmicutes. CONCLUSION Some micronutrients, particularly antioxidant vitamins, correlated with gut microbiota variations in the studied cohort. Further research is required to clarify whether antioxidant vitamin intakes can influence CF gut microbiota and potential clinical/therapeutic implications in CF.
Collapse
Affiliation(s)
- Li Li
- School of Medicine, Griffith University, Brisbane, Queensland, Australia
| | - Lutz Krause
- The University of Queensland, Diamantina Institute, Translational Research Institute, Woolloongabba 4102, Brisbane, Queensland, Australia
| | - Shawn Somerset
- School of Medicine, Griffith University, Brisbane, Queensland, Australia; School of Allied Health, Faculty of Health Sciences, Australian Catholic University, Brisbane, Queensland, Australia.
| |
Collapse
|
146
|
Quality of Reporting Nutritional Randomized Controlled Trials in Patients With Cystic Fibrosis. J Pediatr Gastroenterol Nutr 2016; 63:265-9. [PMID: 26881412 DOI: 10.1097/mpg.0000000000001164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVE Randomized controlled trials (RCTs) have a major role in the making of evidence-based guidelines. The aim of the present study was to critically appraise the RCTs that addressed nutritional interventions in patients with cystic fibrosis. METHODS Embase, PubMed, and the Cochrane Library were systematically searched until July 2015. Methodology and reporting of nutritional RCTs were evaluated by the Consolidated Standards of Reporting Trials (CONSORT) checklist and additional dimensions relevant to patients with CF. RESULTS Fifty-one RCTs were included. Full details on methods were provided in a minority of studies. The mean duration of intervention was <6 months. 56.9% of the RCTs did not define a primary outcome; 70.6% of studies did not provide details on sample size calculation; and only 31.4% reported on the subgroup or separated between important subgroups. CONCLUSIONS The examined RCTs were characterized by a weak methodology, a small number of patients with no sample size calculations, a relatively short intervention, and many times did not examine the outcomes that are important to the patient. Improvement over the years has been minor.
Collapse
|
147
|
Sathe MN, Freeman AJ. Gastrointestinal, Pancreatic, and Hepatobiliary Manifestations of Cystic Fibrosis. Pediatr Clin North Am 2016; 63:679-98. [PMID: 27469182 DOI: 10.1016/j.pcl.2016.04.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pulmonary disease is the primary cause of morbidity and mortality in people with cystic fibrosis (CF), but significant involvement within gastrointestinal, pancreatic, and hepatobiliary systems occurs as well. As in the airways, defects in CFTR alter epithelial surface fluid, mucus viscosity, and pH, increasing risk of stasis through the various hollow epithelial-lined structures of the gastrointestinal tract. This exerts secondary influences that are responsible for most gastrointestinal, pancreatic, and hepatobiliary manifestations of CF. Understanding these gastrointestinal morbidities of CF is essential in understanding and treating CF as a multisystem disease process and improving overall patient care.
Collapse
Affiliation(s)
- Meghana Nitin Sathe
- Division of Pediatric Gastroenterology and Nutrition, Children's Health, University of Texas Southwestern, F4.06, 1935 Medical District Drive, Dallas, TX 75235, USA
| | - Alvin Jay Freeman
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Healthcare of Atlanta, Emory University, 2015 Uppergate Drive, Northeast, Atlanta, GA 30322, USA.
| |
Collapse
|
148
|
Hasegawa K, Linnemann RW, Mansbach JM, Ajami NJ, Espinola JA, Petrosino JF, Piedra PA, Stevenson MD, Sullivan AF, Thompson AD, Camargo CA. The Fecal Microbiota Profile and Bronchiolitis in Infants. Pediatrics 2016; 138:peds.2016-0218. [PMID: 27354456 PMCID: PMC4925084 DOI: 10.1542/peds.2016-0218] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/12/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Little is known about the association of gut microbiota, a potentially modifiable factor, with bronchiolitis in infants. We aimed to determine the association of fecal microbiota with bronchiolitis in infants. METHODS We conducted a case-control study. As a part of multicenter prospective study, we collected stool samples from 40 infants hospitalized with bronchiolitis. We concurrently enrolled 115 age-matched healthy controls. By applying 16S rRNA gene sequencing and an unbiased clustering approach to these 155 fecal samples, we identified microbiota profiles and determined the association of microbiota profiles with likelihood of bronchiolitis. RESULTS Overall, the median age was 3 months, 55% were male, and 54% were non-Hispanic white. Unbiased clustering of fecal microbiota identified 4 distinct profiles: Escherichia-dominant profile (30%), Bifidobacterium-dominant profile (21%), Enterobacter/Veillonella-dominant profile (22%), and Bacteroides-dominant profile (28%). The proportion of bronchiolitis was lowest in infants with the Enterobacter/Veillonella-dominant profile (15%) and highest in the Bacteroides-dominant profile (44%), corresponding to an odds ratio of 4.59 (95% confidence interval, 1.58-15.5; P = .008). In the multivariable model, the significant association between the Bacteroides-dominant profile and a greater likelihood of bronchiolitis persisted (odds ratio for comparison with the Enterobacter/Veillonella-dominant profile, 4.24; 95% confidence interval, 1.56-12.0; P = .005). In contrast, the likelihood of bronchiolitis in infants with the Escherichia-dominant or Bifidobacterium-dominant profile was not significantly different compared with those with the Enterobacter/Veillonella-dominant profile. CONCLUSIONS In this case-control study, we identified 4 distinct fecal microbiota profiles in infants. The Bacteroides-dominant profile was associated with a higher likelihood of bronchiolitis.
Collapse
Affiliation(s)
- Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts;
| | - Rachel W. Linnemann
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | | | - Nadim J. Ajami
- Alkek Center for Metagenomics and Microbiome Research and
| | - Janice A. Espinola
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Pedro A. Piedra
- Department of Molecular Virology and Microbiology, and Pediatrics, Baylor College of Medicine, Houston, Texas
| | | | - Ashley F. Sullivan
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amy D. Thompson
- Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Carlos A. Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
149
|
Demeyer S, De Boeck K, Witters P, Cosaert K. Beyond pancreatic insufficiency and liver disease in cystic fibrosis. Eur J Pediatr 2016; 175:881-94. [PMID: 27055450 DOI: 10.1007/s00431-016-2719-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/17/2016] [Accepted: 03/22/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED Cystic fibrosis is a life shortening hereditary disease, primarily leading to progressive pulmonary infection and exocrine pancreatic dysfunction. Several gastrointestinal complications other than malabsorption can arise during the disease course and with the progressively increasing life span of patients with CF; new and more rare complications are being recognized. We review the literature on gastrointestinal manifestations in CF, excluding the liver and pancreas. CONCLUSION We describe the clinical presentation and treatment of more common conditions like gastroesophageal reflux, small intestinal bacterial overgrowth, intussusception, meconium ileus, distal intestinal obstruction syndrome, and constipation, and we also discuss what is known on celiac disease, appendicitis, fibrosing colonopathy, inflammation and inflammatory bowel disease and gastrointestinal cancer. WHAT IS KNOWN • Gastrointestinal complications arise early in the course of the disease and have a severe impact on the quality of life of the patients. What is New: • This review is a concise summary of the current literature on gastrointestinal complications of cystic fibrosis. • We focused on clinical presentation and diagnostic investigations and provide a comprehensive resume of the current treatment options.
Collapse
Affiliation(s)
- Stephanie Demeyer
- Universitaire Ziekenhuizen Leuven, Leuven, Vlaams-Brabant, Belgium. .,Department of Pediatrics, University Hospital Gasthuisberg, Herestraat 49, Leuven, 3000, Belgium.
| | - Kris De Boeck
- Department of Pediatrics, University Hospital Gasthuisberg, Herestraat 49, Leuven, 3000, Belgium
| | - Peter Witters
- Department of Pediatrics, University Hospital Gasthuisberg, Herestraat 49, Leuven, 3000, Belgium
| | - Katrien Cosaert
- Department of Pharmocology, University Hospital Gasthuisberg, Herestraat 49, Leuven, 3000, Belgium
| |
Collapse
|
150
|
Nielsen S, Needham B, Leach ST, Day AS, Jaffe A, Thomas T, Ooi CY. Disrupted progression of the intestinal microbiota with age in children with cystic fibrosis. Sci Rep 2016; 6:24857. [PMID: 27143104 PMCID: PMC4855157 DOI: 10.1038/srep24857] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 04/01/2016] [Indexed: 12/18/2022] Open
Abstract
Cystic fibrosis (CF) is a genetic disorder that leads to formation of thick epithelial secretions in affected organs. Chronic microbial infections associated with thick mucus secretions are the hallmarks of lung disease in CF. Despite similar conditions existing in the gastrointestinal tract, it is much less studied. We therefore examined the microbial communities within the gastrointestinal tract of children with and without CF (either pancreatic sufficient or insufficient) across a range of childhood ages (0.87-17 years). We observed a substantial reduction in the richness and diversity of gut bacteria associated with CF from early childhood (2 years) until late adolescence (17 years). A number of bacteria that establish themselves in the gut of healthy children were unable to do so in children with CF. In contrast, a few bacteria dominated the gut microbiota in children with CF and are unlikely to be beneficial for the metabolic function of the gut. A functioning pancreas (pancreatic sufficient) under a CF lifestyle showed little effect on microbial communities. Our results argue that any attempts to rectify the loss of bacterial diversity and provide normal bacterial function in the gut of CF patients should be conducted no later than early childhood.
Collapse
Affiliation(s)
- Shaun Nielsen
- Centre for Marine Bio-innovation, University of New South Wales, Sydney, 2052, Australia
- School of Biotechnology and Biomolecular Sciences, and University of New South Wales, Sydney, 2052, Australia
| | - Bronwen Needham
- School of Biotechnology and Biomolecular Sciences, and University of New South Wales, Sydney, 2052, Australia
| | - Steven T. Leach
- Discipline of Paediatrics, School of Women’s and Children’s Health, Medicine, University of New South Wales, Sydney, 2052, Australia
| | - Andrew S. Day
- Department of Paediatrics, University of Otago, Christchurch, 8011, New Zealand
| | - Adam Jaffe
- Discipline of Paediatrics, School of Women’s and Children’s Health, Medicine, University of New South Wales, Sydney, 2052, Australia
- Department of Paediatric Respiratory and Sydney Children’s Hospital, Randwick, 2031, Australia
| | - Torsten Thomas
- Centre for Marine Bio-innovation, University of New South Wales, Sydney, 2052, Australia
- School of Biotechnology and Biomolecular Sciences, and University of New South Wales, Sydney, 2052, Australia
| | - Chee Y. Ooi
- Discipline of Paediatrics, School of Women’s and Children’s Health, Medicine, University of New South Wales, Sydney, 2052, Australia
- Department of Paediatric Gastroenterology, Sydney Children’s Hospital, Randwick, 2031, Australia
| |
Collapse
|