151
|
Enaud R, Prevel R, Ciarlo E, Beaufils F, Wieërs G, Guery B, Delhaes L. The Gut-Lung Axis in Health and Respiratory Diseases: A Place for Inter-Organ and Inter-Kingdom Crosstalks. Front Cell Infect Microbiol 2020; 10:9. [PMID: 32140452 PMCID: PMC7042389 DOI: 10.3389/fcimb.2020.00009] [Citation(s) in RCA: 394] [Impact Index Per Article: 98.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022] Open
Abstract
The gut and lungs are anatomically distinct, but potential anatomic communications and complex pathways involving their respective microbiota have reinforced the existence of a gut-lung axis (GLA). Compared to the better-studied gut microbiota, the lung microbiota, only considered in recent years, represents a more discreet part of the whole microbiota associated to human hosts. While the vast majority of studies focused on the bacterial component of the microbiota in healthy and pathological conditions, recent works have highlighted the contribution of fungal and viral kingdoms at both digestive and respiratory levels. Moreover, growing evidence indicates the key role of inter-kingdom crosstalks in maintaining host homeostasis and in disease evolution. In fact, the recently emerged GLA concept involves host-microbe as well as microbe-microbe interactions, based both on localized and long-reaching effects. GLA can shape immune responses and interfere with the course of respiratory diseases. In this review, we aim to analyze how the lung and gut microbiota influence each other and may impact on respiratory diseases. Due to the limited knowledge on the human virobiota, we focused on gut and lung bacteriobiota and mycobiota, with a specific attention on inter-kingdom microbial crosstalks which are able to shape local or long-reached host responses within the GLA.
Collapse
Affiliation(s)
- Raphaël Enaud
- CHU de Bordeaux, CRCM Pédiatrique, CIC 1401, Bordeaux, France
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- CHU de Bordeaux, Univ. Bordeaux, FHU ACRONIM, Bordeaux, France
| | - Renaud Prevel
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- CHU de Bordeaux, Univ. Bordeaux, FHU ACRONIM, Bordeaux, France
- CHU de Bordeaux, Médecine Intensive Réanimation, Bordeaux, France
| | - Eleonora Ciarlo
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Fabien Beaufils
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- CHU de Bordeaux, Univ. Bordeaux, FHU ACRONIM, Bordeaux, France
- CHU de Bordeaux, Service d'Explorations Fonctionnelles Respiratoires, Bordeaux, France
| | - Gregoire Wieërs
- Clinique Saint Pierre, Department of Internal Medicine, Ottignies, Belgium
| | - Benoit Guery
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laurence Delhaes
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Bordeaux, France
- CHU de Bordeaux, Univ. Bordeaux, FHU ACRONIM, Bordeaux, France
- CHU de Bordeaux: Laboratoire de Parasitologie-Mycologie, Univ. Bordeaux, Bordeaux, France
| |
Collapse
|
152
|
Hufnagl K, Pali-Schöll I, Roth-Walter F, Jensen-Jarolim E. Dysbiosis of the gut and lung microbiome has a role in asthma. Semin Immunopathol 2020; 42:75-93. [PMID: 32072252 PMCID: PMC7066092 DOI: 10.1007/s00281-019-00775-y] [Citation(s) in RCA: 204] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/15/2019] [Indexed: 02/07/2023]
Abstract
Worldwide 300 million children and adults are affected by asthma. The development of asthma is influenced by environmental and other exogenous factors synergizing with genetic predisposition, and shaping the lung microbiome especially during birth and in very early life. The healthy lung microbial composition is characterized by a prevalence of bacteria belonging to the phyla Bacteroidetes, Actinobacteria, and Firmicutes. However, viral respiratory infections are associated with an abundance of Proteobacteria with genera Haemophilus and Moraxella in young children and adult asthmatics. This dysbiosis supports the activation of inflammatory pathways and contributes to bronchoconstriction and bronchial hyperresponsiveness. Exogenous factors can affect the natural lung microbiota composition positively (farming environment) or negatively (allergens, air pollutants). It is evident that also gut microbiota dysbiosis has a high influence on asthma pathogenesis. Antibiotics, antiulcer medications, and other drugs severely impair gut as well as lung microbiota. Resulting dysbiosis and reduced microbial diversity dysregulate the bidirectional crosstalk across the gut-lung axis, resulting in hypersensitivity and hyperreactivity to respiratory and food allergens. Efforts are undertaken to reconstitute the microbiota and immune balance by probiotics and engineered bacteria, but results from human studies do not yet support their efficacy in asthma prevention or treatment. Overall, dysbiosis of gut and lung seem to be critical causes of the increased emergence of asthma.
Collapse
Affiliation(s)
- Karin Hufnagl
- The Interuniversity Messerli Research Institute, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna, Austria
| | - Isabella Pali-Schöll
- The Interuniversity Messerli Research Institute, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna, Austria
| | - Franziska Roth-Walter
- The Interuniversity Messerli Research Institute, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna, Austria
| | - Erika Jensen-Jarolim
- The Interuniversity Messerli Research Institute, Medical University Vienna and University of Veterinary Medicine Vienna, Vienna, Austria. .,Center for Pathophysiology, Infectiology and Immunology, Institute of Pathophysiology and Allergy Research, Medical University Vienna, Währinger G. 18-20, 1090, Vienna, Austria.
| |
Collapse
|
153
|
Leszczyńska J, Stryjewska-Makuch G, Ścierski W, Lisowska G. Bacterial Flora of the Nose and Paranasal Sinuses Among Patients Over 65 Years Old with Chronic Rhinosinusitis Who Underwent Endoscopic Sinus Surgery. Clin Interv Aging 2020; 15:207-215. [PMID: 32103923 PMCID: PMC7027888 DOI: 10.2147/cia.s215917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 01/04/2020] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Chronic rhinosinusitis (CRS) is one of the most common chronic diseases in the geriatric population. However, CRS inflammatory mechanisms in older people have not been thoroughly investigated. Our work aimed to analyze the bacterial flora of the nose and paranasal sinuses in patients with CRS over 65 years of age, including comorbidities, previously performed endoscopic sinus surgery (ESS), presence or absence of polyps and the extent of the inflammatory process. PATIENTS AND METHODS The study involved 529 patients between 18 and 84 years of age with chronic rhinosinusitis who underwent endoscopic sinus surgery. There were 101 patients separated over 65 years of age (M = 52, K = 49, mean age 69 ± 0.7 years). The control group consisted of 168 patients aged 18-40 years with CRS. The bacterial culture of material collected from the patients during ESS was analyzed. RESULTS In the group of patients over 65 years of age, more frequent occurrence of Proteus spp. and Pseudomonas aeruginosa was found in comparison to younger patients. In older patients with bronchial asthma, the occurrence of S. aureus, Escherichia coli, and Citrobacter spp. was more frequent than in control group. Multiple sinus surgical procedures in older patients were associated with the dominance of Staphylococcus aureus and Escherichia coli, which was not demonstrated in the control group. There were no statistically significant differences between the occurrence of bacterial strain and the presence of polyps, both in the group of patients over 65 years of age as well as in the control group. CONCLUSION The bacterial flora of patients with CRS is different in older and younger patients. A different therapeutic approach should be considered in older patients with CRS, but this problem requires further studies.
Collapse
Affiliation(s)
- Joanna Leszczyńska
- Department of Laryngology and Laryngological Oncology, Upper Silesian Medical Centre of Silesian Medical University, Katowice, Poland
| | - Grażyna Stryjewska-Makuch
- Department of Laryngology and Laryngological Oncology, Upper Silesian Medical Centre of Silesian Medical University, Katowice, Poland
| | - Wojciech Ścierski
- Department of Otorhinolaryngology and Laryngological Oncology, Medical University of Silesia, Zabrze, Poland
| | - Grażyna Lisowska
- Department of Otorhinolaryngology and Laryngological Oncology, Medical University of Silesia, Zabrze, Poland
| |
Collapse
|
154
|
Asthma Case Cluster during Renovation of a Water-Damaged and Toxic Building. Microorganisms 2019; 7:microorganisms7120642. [PMID: 31816917 PMCID: PMC6956061 DOI: 10.3390/microorganisms7120642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 11/28/2019] [Accepted: 11/30/2019] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND An association between fungal exposure at work and asthma onset has been shown, but a causal relationship between them has not beTanle en established. METHODS The study describes an asthma cluster in workers in a building under renovation. Before renovation the work site had significant water damage, technical deficiencies, and ventilation problems. Worker protection was insufficient during renovation. In the building, toxicity was determined from dust as well as from cultured dust. Toxicity analysis was conducted in vitro using the boar spermatozoa motility assay. RESULTS During the 8-month renovation period, among 290 workers, 21 (7.2%) experienced new-onset asthma (9 women, 42.9%; 12 men, 57.1%; median age, 43 years (range, 30-60 years)). At the renovation site, they had been exposed to areas where remarkable toxicity was demonstrated in vitro. One year later, 13 (61.9%) of them still had moderate disease, and three (14.8%) had severe disease. Most patients had a poor response to inhaled corticosteroids. CONCLUSIONS This study documents a clear temporal association between occupational exposure during renovation of a water-damaged building and a cluster of 21 new occupational asthma cases. In addition, dust and cultured dust from their work spaces showed remarkable toxicity based on inhibition of boar sperm motility in vitro.
Collapse
|
155
|
Abstract
Over the last few decades, advances in our understanding of microbial ecology have allowed us to appreciate the important role of microbial communities in maintaining human health. While much of this research has focused on gut microbes, microbial communities in other body sites and from the environment are increasingly recognized in human disease. Here, we discuss recent advances in our understanding of host-microbiota interactions in the development and manifestation of asthma focusing on three distinct microbial compartments. First, environmental microbes originating from house dust, pets, and farm animals have been linked to asthma pathogenesis, which is often connected to their production of bioactive molecules such as lipopolysaccharide. Second, respiratory microbial communities, including newly appreciated populations of microbes in the lung have been associated with allergic airway inflammation. Current evidence suggests that the presence of particular microbes, especially Streptococcus, Haemophilus, and Morexella species within the airway may shape local immune responses and alter the severity and manifestations of airway inflammation. Third, the gut microbiota has been implicated in both experimental models and clinical studies in predisposing to asthma. There appears to be a "critical window" of colonization that occurs during early infancy in which gut microbial communities shape immune maturation and confer susceptibility to allergic airway inflammation. The mechanisms by which gut microbial communities influence lung immune responses and physiology, the "gut-lung axis," are still being defined but include the altered differentiation of immune cell populations important in asthma and the local production of metabolites that affect distal sites. Together, these findings suggest an intimate association of microbial communities with host immune development and the development of allergic airway inflammation. Improved understanding of these relationships raises the possibility of microbiota-directed therapies to improve or prevent asthma.
Collapse
Affiliation(s)
- Aaron Ver Heul
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Joseph Planer
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Andrew L Kau
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
156
|
Abstract
The human microbiome has been identified as having a key role in health and numerous diseases. Trillions of microbial cells and viral particles comprise the microbiome, each representing modifiable working elements of an intricate bioactive ecosystem. The significance of the human microbiome as it relates to human biology has progressed through culture-dependent (for example, media-based methods) and, more recently, molecular (for example, genetic sequencing and metabolomic analysis) techniques. The latter have become increasingly popular and evolved from being used for taxonomic identification of microbiota to elucidation of functional capacity (sequencing) and metabolic activity (metabolomics). This review summarises key elements of the human microbiome and its metabolic capabilities within the context of health and disease.
Collapse
Affiliation(s)
- Wiley Barton
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, P61C996, Ireland.,APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, T12YT20, Ireland.,VistaMilk SFI Research Centre, Teagasc, Moorepark, Fermoy, Cork, P61C996, Ireland
| | - Orla O'Sullivan
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, P61C996, Ireland.,APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, T12YT20, Ireland.,VistaMilk SFI Research Centre, Teagasc, Moorepark, Fermoy, Cork, P61C996, Ireland
| | - Paul D Cotter
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, P61C996, Ireland.,APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, T12YT20, Ireland.,VistaMilk SFI Research Centre, Teagasc, Moorepark, Fermoy, Cork, P61C996, Ireland
| |
Collapse
|
157
|
Lung Microbiome in Asthma: Current Perspectives. J Clin Med 2019; 8:jcm8111967. [PMID: 31739446 PMCID: PMC6912699 DOI: 10.3390/jcm8111967] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 12/20/2022] Open
Abstract
A growing body of evidence implicates the human microbiome as a potentially influential player actively engaged in shaping the pathogenetic processes underlying the endotypes and phenotypes of chronic respiratory diseases, particularly of the airways. In this article, we specifically review current evidence on the characteristics of lung microbiome, and specifically the bacteriome, the modes of interaction between lung microbiota and host immune system, the role of the “lung–gut axis”, and the functional effects thereof on asthma pathogenesis. We also attempt to explore the possibilities of therapeutic manipulation of the microbiome, aiming at the establishment of asthma prevention strategies and the optimization of asthma treatment.
Collapse
|
158
|
Sharma A, Laxman B, Naureckas ET, Hogarth DK, Sperling AI, Solway J, Ober C, Gilbert JA, White SR. Associations between fungal and bacterial microbiota of airways and asthma endotypes. J Allergy Clin Immunol 2019; 144:1214-1227.e7. [PMID: 31279011 PMCID: PMC6842419 DOI: 10.1016/j.jaci.2019.06.025] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/12/2019] [Accepted: 06/19/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND The relationship between asthma, atopy, and underlying type 2 (T2) airway inflammation is complex. Although the bacterial airway microbiota is known to differ in asthmatic patients, the fungal and bacterial markers that discriminate T2-high (eosinophilic) and T2-low (neutrophilic/mixed-inflammation) asthma and atopy are still incompletely identified. OBJECTIVES The aim of this study was to demonstrate the fungal microbiota structure of airways in asthmatic patients associated with T2 inflammation, atopy, and key clinical parameters. METHODS We collected endobronchial brush (EB) and bronchoalveolar lavage (BAL) samples from 39 asthmatic patients and 19 healthy subjects followed by 16S gene and internal transcribed spacer-based microbiota sequencing. The microbial sequences were classified into exact sequence variants. The T2 phenotype was defined by using a blood eosinophil count with a threshold of 300 cells/μL. RESULTS Fungal diversity was significantly lower in EB samples from patients with T2-high compared with T2-low inflammation; key fungal genera enriched in patients with T2-high inflammation included Trichoderma species, whereas Penicillium species was enriched in patients with atopy. In BAL fluid samples the dominant genera were Cladosporium, Fusarium, Aspergillus, and Alternaria. Using generalized linear models, we identified significant associations between specific fungal exact sequence variants and FEV1, fraction of exhaled nitric oxide values, BAL fluid cell counts, and corticosteroid use. Investigation of interkingdom (bacterial-fungal) co-occurrence patterns revealed different topologies between asthmatic patients and healthy control subjects. Random forest models with fungal classifiers predicted asthma status with 75% accuracy for BAL fluid samples and 80% accuracy for EB samples. CONCLUSIONS We demonstrate clear differences in bacterial and fungal microbiota in asthma-associated phenotypes. Our study provides additional support for considering microbial signatures in delineating asthma phenotypes.
Collapse
Affiliation(s)
- Anukriti Sharma
- Department of Surgery, University of Chicago, Chicago, Ill; Biosciences Division (BIO), Argonne National Laboratory, Argonne, Ill; Department of Pediatrics and Scripps Institution of Oceanography, University of California San Diego, La Jolla, Calif
| | - Bharathi Laxman
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Ill
| | - Edward T Naureckas
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Ill
| | - D Kyle Hogarth
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Ill
| | - Anne I Sperling
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Ill
| | - Julian Solway
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Ill
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, Ill
| | - Jack A Gilbert
- Department of Surgery, University of Chicago, Chicago, Ill; Biosciences Division (BIO), Argonne National Laboratory, Argonne, Ill; Department of Pediatrics and Scripps Institution of Oceanography, University of California San Diego, La Jolla, Calif
| | - Steven R White
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Ill.
| |
Collapse
|
159
|
Budden KF, Shukla SD, Rehman SF, Bowerman KL, Keely S, Hugenholtz P, Armstrong-James DPH, Adcock IM, Chotirmall SH, Chung KF, Hansbro PM. Functional effects of the microbiota in chronic respiratory disease. THE LANCET. RESPIRATORY MEDICINE 2019; 7:907-920. [PMID: 30975495 DOI: 10.1016/s2213-2600(18)30510-1] [Citation(s) in RCA: 251] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/29/2018] [Accepted: 11/29/2018] [Indexed: 01/19/2023]
Abstract
The composition of the lung microbiome is increasingly well characterised, with changes in microbial diversity or abundance observed in association with several chronic respiratory diseases such as asthma, cystic fibrosis, bronchiectasis, and chronic obstructive pulmonary disease. However, the precise effects of the microbiome on pulmonary health and the functional mechanisms by which it regulates host immunity are only now beginning to be elucidated. Bacteria, viruses, and fungi from both the upper and lower respiratory tract produce structural ligands and metabolites that interact with the host and alter the development and progression of chronic respiratory diseases. Here, we review recent advances in our understanding of the composition of the lung microbiome, including the virome and mycobiome, the mechanisms by which these microbes interact with host immunity, and their functional effects on the pathogenesis, exacerbations, and comorbidities of chronic respiratory diseases. We also describe the present understanding of how respiratory microbiota can influence the efficacy of common therapies for chronic respiratory disease, and the potential of manipulation of the microbiome as a therapeutic strategy. Finally, we highlight some of the limitations in the field and propose how these could be addressed in future research.
Collapse
Affiliation(s)
- Kurtis F Budden
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Saima Firdous Rehman
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Kate L Bowerman
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biology, The University of Queensland, QLD, Australia
| | - Simon Keely
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biology, The University of Queensland, QLD, Australia
| | | | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia; Centre for Inflammation, Centenary Institute, and University of Technology Sydney, NSW, Australia.
| |
Collapse
|
160
|
Wypych TP, Wickramasinghe LC, Marsland BJ. The influence of the microbiome on respiratory health. Nat Immunol 2019; 20:1279-1290. [PMID: 31501577 DOI: 10.1038/s41590-019-0451-9] [Citation(s) in RCA: 313] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/18/2019] [Indexed: 02/06/2023]
Abstract
The revolution in microbiota research over the past decade has provided invaluable knowledge about the function of the microbial species that inhabit the human body. It has become widely accepted that these microorganisms, collectively called 'the microbiota', engage in networks of interactions with each other and with the host that aim to benefit both the microbial members and the mammalian members of this unique ecosystem. The lungs, previously thought to be sterile, are now known to harbor a unique microbiota and, additionally, to be influenced by microbial signals from distal body sites, such as the intestine. Here we review the role of the lung and gut microbiotas in respiratory health and disease and highlight the main pathways of communication that underlie the gut-lung axis.
Collapse
Affiliation(s)
- Tomasz P Wypych
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Lakshanie C Wickramasinghe
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Benjamin J Marsland
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
161
|
Janson C, Malinovschi A, Amaral AFS, Accordini S, Bousquet J, Buist AS, Canonica GW, Dahlén B, Garcia-Aymerich J, Gnatiuc L, Kowalski ML, Patel J, Tan W, Torén K, Zuberbier T, Burney P, Jarvis D. Bronchodilator reversibility in asthma and COPD: findings from three large population studies. Eur Respir J 2019; 54:13993003.00561-2019. [PMID: 31221806 DOI: 10.1183/13993003.00561-2019] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/28/2019] [Indexed: 12/27/2022]
Abstract
Bronchodilator response (BDR) testing is used as a diagnostic method in obstructive airway diseases. The aim of this investigation was to compare different methods for measuring BDR in participants with asthma and chronic obstructive pulmonary disease (COPD) and to study to the extent to which BDR was related to symptom burden and phenotypic characteristics.Forced expiratory volume in 1 s (FEV1) and forced vital capacity (FVC) were measured before and 15 min after 200 μg of salbutamol in 35 628 subjects aged ≥16 years from three large international population studies. The subjects were categorised in three groups: current asthma (n=2833), COPD (n=1146) and no airway disease (n=31 649). Three definitions for flow-related reversibility (increase in FEV1) and three for volume-related reversibility (increase in FVC) were used.The prevalence of bronchodilator reversibility expressed as increase FEV1 ≥12% and 200 mL was 17.3% and 18.4% in participants with asthma and COPD, respectively, while the corresponding prevalence was 5.1% in those with no airway disease. In asthma, bronchodilator reversibility was associated with wheeze (OR 1.36, 95% CI 1.04-1.79), atopy (OR 1.36, 95% CI 1.04-1.79) and higher exhaled nitric oxide fraction, while in COPD neither flow- nor volume-related bronchodilator reversibility was associated with symptom burden, exacerbations or health status after adjusting for pre-bronchodilator FEV1Bronchodilator reversibility was at least as common in participants with COPD as those with asthma. This indicates that measures of reversibility are of limited value for distinguishing asthma from COPD in population studies. However, in asthma, bronchodilator reversibility may be a phenotypic marker.
Collapse
Affiliation(s)
- Christer Janson
- Dept of Medical Sciences, Respiratory Allergy and Sleep Research, Uppsala University, Uppsala, Sweden .,Population Health and Occupational Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Andrei Malinovschi
- Dept of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Andre F S Amaral
- Population Health and Occupational Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Simone Accordini
- Unit of Epidemiology and Medical Statistics, Dept of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Jean Bousquet
- Fondation FMC VIA-LR, Montpellier, France.,Euforea, Brussels, Belgium.,Allergy Centre Charité, Dept of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - A Sonia Buist
- Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, OR, USA
| | | | - Barbro Dahlén
- Dept of Medicine, Unit for Heart and Lung disease, Karolinska Institutet, Stockholm, Sweden
| | - Judith Garcia-Aymerich
- ISGlobal, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Louisa Gnatiuc
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Dept of Population Health, University of Oxford, Oxford, UK
| | - Marek L Kowalski
- Dept of Immunology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Jaymini Patel
- Population Health and Occupational Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Wan Tan
- Centre for Heart Lung Innovation (Tan, Sin), University of British Columbia, St. Paul's Hospital Vancouver, Vancouver, BC, Canada
| | - Kjell Torén
- Dept of Occupational and Environmental Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Torsten Zuberbier
- Allergy Centre Charité, Dept of Dermatology and Allergy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Burney
- Population Health and Occupational Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Deborah Jarvis
- Population Health and Occupational Disease, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
162
|
McGinniss JE, Collman RG. Of Mice and Men . . . and Microbes: Conclusions and Cautions from a Murine Study of the Lung Microbiome and Microbiome-Immune Interactions. Am J Respir Crit Care Med 2019; 198:419-422. [PMID: 29995432 DOI: 10.1164/rccm.201803-0586ed] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- John E McGinniss
- 1 Department of Medicine University of Pennsylvania School of Medicine Philadelphia, Pennsylvania
| | - Ronald G Collman
- 1 Department of Medicine University of Pennsylvania School of Medicine Philadelphia, Pennsylvania
| |
Collapse
|
163
|
Abstract
Current management of severe asthma relying either on guidelines (bulk approach) or on disease phenotypes (stratified approach) did not improve the burden of the disease. Several severe phenotypes are described: clinical, functional, morphological, inflammatory, molecular and microbiome-related. However, phenotypes do not necessarily relate to or give insights into the underlying pathogenetic mechanisms which are described by the disease endotypes. Based on the major immune-inflammatory pathway involved type-2 high, type-2 low and mixed endotypes are described for severe asthma, with several shared pathogenetic pathways such as genetic and epigenetic, metabolic, neurogenic and remodelling subtypes. The concept of multidimensional endotyping as un unbiased approach to severe asthma is discussed, together with new tools and targets facilitating the shift from the stratified to the precision medicine approach.
Collapse
|
164
|
Wang G, Pang Z, Chen-Yu Hsu A, Guan X, Ran N, Yuan Y, Wang Z, Guo Y, Zheng R, Wang F. Combined treatment with SB203580 and dexamethasone suppresses non-typeable Haemophilus influenzae-induced Th17 inflammation response in murine allergic asthma. Eur J Pharmacol 2019; 862:172623. [PMID: 31445014 DOI: 10.1016/j.ejphar.2019.172623] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/14/2019] [Accepted: 08/20/2019] [Indexed: 12/20/2022]
Abstract
Accumulating evidence suggests that non-typeable Haemophilus influenzae (NTHi) infection drives the development of steroid-resistant allergic airway disease (SRAAD), exacerbates clinical symptoms, worsens quality of life, and accounts for most of the related healthcare burden. The poor understanding of the pathogenesis of SRAAD deters the development of more effective therapeutic strategies. Here, we established a murine model of NTHi infection-induced exacerbation of allergic airway disease. We showed that NTHi infection drove Th 17-mediated pulmonary neutrophilic inflammation, aggravated airway hyper-responsiveness, and upset the balance of MUC5AC and MUC5B expression. Dexamethasone treatment effectively inhibited the features of allergic airway disease but failed to reduce NTHi-induced exacerbation, which was associated with the hyper-phosphorylation of p38 mitogen-activated protein kinase (MAPK). Interestingly, inhibition of p38 using a specific inhibitor (SB203580) only partly suppressed the airway hyper-responsiveness and mucus hyper-secretion but failed to abrogate the infection-induced neutrophilic inflammatory response in SRAAD. However, SB203580 and dexamethasone co-treatment substantially suppressed all the features of NTHi-induced SRAAD. Our findings highlight the importance of p38 MAPK in the pathogenesis of NTHi-induced steroid resistance, and this combined treatment approach may be a novel strategy against steroid-resistant asthma.
Collapse
Affiliation(s)
- Guoqiang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Zhiqiang Pang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Alan Chen-Yu Hsu
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute and the University of Newcastle, NSW, 2305, Australia
| | - Xuewa Guan
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Nan Ran
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Yuze Yuan
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Ziyan Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Yingqiao Guo
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Ruipeng Zheng
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China; Department of Invasive Technology, First Hospital of Jilin University, Changchun, 130021, China
| | - Fang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China.
| |
Collapse
|
165
|
Caverly LJ, Huang YJ, Sze MA. Past, Present, and Future Research on the Lung Microbiome in Inflammatory Airway Disease. Chest 2019; 156:376-382. [PMID: 31154042 PMCID: PMC6945648 DOI: 10.1016/j.chest.2019.05.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/07/2019] [Accepted: 05/10/2019] [Indexed: 02/08/2023] Open
Abstract
COPD, asthma, and cystic fibrosis (CF) are obstructive lung diseases with distinct pathophysiologies and clinical phenotypes. In this paper, we highlight recent advances in our understanding of relationships between clinical phenotypes, host inflammatory response, and lung microbiota in these diseases. Although COPD, asthma, and CF largely have distinct lung microbiota and inflammatory profiles, certain commonalities exist. In all three of these lung diseases, and in healthy persons, anaerobic taxa that are typically associated with oral microbiota (eg, Prevotella species, Veillonella species) are present in the airways and associated with increased host inflammatory response. Similarly, across all three diseases, members of the Proteobacteria phylum are associated with more advanced disease. Finally, we highlight challenges in translating these findings into advances in clinical care, including continued knowledge gaps regarding the causal relationships between host inflammatory response, lung microbiota, medication effects, and clinical phenotypes.
Collapse
Affiliation(s)
| | - Yvonne J Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Ann Arbor, MI
| | - Marc A Sze
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI.
| |
Collapse
|
166
|
Shukla SD, Shastri MD, Chong WC, Dua K, Budden KF, Mahmood MQ, Hansbro NG, Keely S, Eri R, Patel RP, Peterson GM, Hansbro PM. Microbiome-focused asthma management strategies. Curr Opin Pharmacol 2019; 46:143-149. [PMID: 31357048 DOI: 10.1016/j.coph.2019.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022]
Abstract
Asthma is a common, heterogeneous and serious disease with high prevalence globally. Poorly controlled, steroid-resistant asthma is particularly important as there are no effective therapies and it exerts substantial healthcare and societal burden. The role of microbiomes, particularly in chronic diseases has generated considerable interest in recent times. Existing evidence clearly demonstrates an association between asthma initiation and the microbiome, both respiratory and gastro-intestinal, although its' roles are poorly understood when assessing the asthma progression or heterogeneity (i.e. phenotypes/endotypes) across different geographical locations. Moreover, modulating microbiomes could be preventive and/or therapeutic in patients with asthma warrants urgent attention. Here, we review recent advances in assessing the role of microbiomes in asthma and present the challenges associated with the potential therapeutic utility of modifying microbiomes in management.
Collapse
Affiliation(s)
- Shakti D Shukla
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia
| | - Madhur D Shastri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia
| | - Wai Chin Chong
- Department of Molecular and Translational Science, Monash University, Clayton, Australia; Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Australia
| | - Kamal Dua
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia
| | - Kurtis F Budden
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia
| | - Malik Quasir Mahmood
- Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia; Centre for inflammation, Centenary Institute, Sydney, and School of Life Sciences, University of Technology, Ultimo, NSW, Australia
| | - Simon Keely
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia
| | - Rajaraman Eri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia
| | - Rahul P Patel
- Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Gregory M Peterson
- Pharmacy, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute & University of Newcastle, Callaghan, NSW, Australia; Centre for inflammation, Centenary Institute, Sydney, and School of Life Sciences, University of Technology, Ultimo, NSW, Australia.
| |
Collapse
|
167
|
O'Byrne P, Fabbri LM, Pavord ID, Papi A, Petruzzelli S, Lange P. Asthma progression and mortality: the role of inhaled corticosteroids. Eur Respir J 2019; 54:1900491. [PMID: 31048346 PMCID: PMC6637285 DOI: 10.1183/13993003.00491-2019] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/24/2019] [Indexed: 01/22/2023]
Abstract
Overall, asthma mortality rates have declined dramatically in the last 30 years, due to improved diagnosis and to better treatment, particularly in the 1990s following the more widespread use of inhaled corticosteroids (ICSs). The impact of ICS on other long-term outcomes, such as lung function decline, is less certain, in part because the factors associated with these outcomes are incompletely understood. The purpose of this review is to evaluate the effect of pharmacological interventions, particularly ICS, on asthma progression and mortality. Furthermore, we review the potential mechanisms of action of pharmacotherapy on asthma progression and mortality, the effects of ICS on long-term changes in lung function, and the role of ICS in various asthma phenotypes.Overall, there is compelling evidence of the value of ICS in improving asthma control, as measured by improved symptoms, pulmonary function and reduced exacerbations. There is, however, less convincing evidence that ICS prevents the decline in pulmonary function that occurs in some, although not all, patients with asthma. Severe exacerbations are associated with a more rapid decline in pulmonary function, and by reducing the risk of severe exacerbations, it is likely that ICS will, at least partially, prevent this decline. Studies using administrative databases also support an important role for ICS in reducing asthma mortality, but the fact that asthma mortality is, fortunately, an uncommon event makes it highly improbable that this will be demonstrated in prospective trials.
Collapse
Affiliation(s)
- Paul O'Byrne
- Faculty of Health Sciences, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - Leonardo M Fabbri
- Section of Cardiorespiratory and Internal Medicine, Dept of Medical Sciences, University of Ferrara, Ferrara, Italy
- COPD Center, Institute of Medicine, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Ian D Pavord
- Respiratory Medicine Unit and Oxford Respiratory NIHR BRC, Nuffield Dept of Medicine, University of Oxford, Oxford, UK
| | - Alberto Papi
- Section of Cardiorespiratory and Internal Medicine, Dept of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Peter Lange
- Section of Epidemiology, Dept of Public Health, University of Copenhagen, Copenhagen, Denmark
- Medical Dept, Respiratory Section, Herlev and Gentofte Hospital, Herlev, Denmark
| |
Collapse
|
168
|
Abdel-Aziz MI, Vijverberg SJH, Neerincx AH, Kraneveld AD, Maitland-van der Zee AH. The crosstalk between microbiome and asthma: Exploring associations and challenges. Clin Exp Allergy 2019; 49:1067-1086. [PMID: 31148278 PMCID: PMC6852296 DOI: 10.1111/cea.13444] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/17/2022]
Abstract
With the advancement of high‐throughput DNA/RNA sequencing and computational analysis techniques, commensal bacteria are now considered almost as important as pathological ones. Understanding the interaction between these bacterial microbiota, host and asthma is crucial to reveal their role in asthma pathophysiology. Several airway and/or gut microbiome studies have shown associations between certain bacterial taxa and asthma. However, challenges remain before gained knowledge from these studies can be implemented into clinical practice, such as inconsistency between studies in choosing sampling compartments and/or sequencing approaches, variability of results in asthma studies, and not taking into account medication intake and diet composition especially when investigating gut microbiome. Overcoming those challenges will help to better understand the complex asthma disease process. The therapeutic potential of using pro‐ and prebiotics to prevent or reduce risk of asthma exacerbations requires further investigation. This review will focus on methodological issues regarding setting up a microbiome study, recent developments in asthma bacterial microbiome studies, challenges and future therapeutic potential.
Collapse
Affiliation(s)
- Mahmoud I Abdel-Aziz
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Susanne J H Vijverberg
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne H Neerincx
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Anke H Maitland-van der Zee
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatric Respiratory Medicine, Amsterdam UMC, Emma Children's Hospital, Amsterdam, The Netherlands
| |
Collapse
|
169
|
Huang YJ. Nasopharyngeal Microbiota: Gatekeepers or Fortune Tellers of Susceptibility to Respiratory Tract Infections? Am J Respir Crit Care Med 2019; 196:1504-1505. [PMID: 28800258 DOI: 10.1164/rccm.201707-1470ed] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Yvonne J Huang
- 1 Division of Pulmonary/Critical Care Medicine University of Michigan, Ann Arbor Ann Arbor, Michigan
| |
Collapse
|
170
|
Abstract
The airways are under continuous assault from aerosolized bacteria and oral flora. The bacteria present in the airways and gastrointestinal tract of neonates promote immune maturation and protect against asthma pathogenesis. Later bacterial infections and perturbations to the microbiome can contribute to asthma pathogenesis, persistence, and severity.
Collapse
Affiliation(s)
- Michael Insel
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of Arizona Health Sciences, University of Arizona College of Medicine - Tucson, 1501 North Campbell Avenue, PO Box 245017, Tucson, AZ 85724, USA
| | - Monica Kraft
- Department of Medicine, College of Medicine Tucson, Asthma and Airway Disease Research Center, University of Arizona Health Sciences, University of Arizona College of Medicine - Tucson, 1501 North Campbell Avenue, PO Box 245017, Tucson, AZ 85724, USA.
| |
Collapse
|
171
|
Affiliation(s)
- Naomi G. Wilson
- Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ariel Hernandez-Leyva
- Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Andrew L. Kau
- Department of Medicine and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
172
|
Abstract
PURPOSE OF REVIEW The precision medicine concept is both appealing and challenging. We review here the recent findings in the endotype-driven approach for major allergic diseases. RECENT FINDINGS Stratified medicine for different allergic diseases can identify patients who are more likely to benefit or experience an adverse reaction in response to a given therapy and anticipate their long-term outcome and vital risk. In addition, this approach potentially facilitates drug development and prevention strategies. SUMMARY The endotype-driven approach in allergic diseases has tremendous potential, but there are notable barriers in reaching the new world of precision medicine. Multidimensional endotyping integrating visible properties with multiple biomarkers is recommended for both type 2 and nontype 2 allergic diseases to provide evidence that a certain pathway is the key driver for a given patient. Significant healthcare system changes are required to achieve the expected targets.
Collapse
|
173
|
Lee J, Kim S, Lee M, Kim B, Song W, Park H, Cho S, Hong S, Chang Y, Kim B. Different upper airway microbiome and their functional genes associated with asthma in young adults and elderly individuals. Allergy 2019; 74:709-719. [PMID: 30242844 DOI: 10.1111/all.13608] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/19/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Microbes in the airway have been shown to be associated with the pathogenesis of asthma. The upper airway microbiome influences the dysbiosis of the lower airway microbiome. However, to date, the influence of upper airway microbiome for adult and elderly asthma has not been fully elucidated. Here, the metagenome of upper airway microbiome of young adults and elderly was analyzed to identify their association with adult asthma. METHODS Nasopharyngeal swabs were collected from young adult and elderly asthma patients and non-asthmatic subjects. The compositions and functional genes of airway microbiome were analyzed by high-throughput sequencing. RESULTS The composition of microbiota differed between young adult and elderly, and it was different between asthmatics and non-asthmatics in each age group. Different bacteria were related to FEV1% predicted in each age group. Genes related to lysine degradation, N-glycan biosynthesis, caprolactam degradation, and PPAR signaling pathway, which could be related to the reduction in inflammation and degradation of air pollutants, were higher in non-asthmatics. Genes related to pentose phosphate pathway, lipopolysaccharide biosynthesis, flagella assembly, and bacterial chemotaxis-which may all be related to increased inflammation and colonization of pathogenic bacteria-were higher in young adult asthmatic patients. However, the functional genes of airway microbiome in elderly patients were not significantly different according to asthma morbidity. CONCLUSIONS These results suggest that the composition and function of upper airway microbiome could influence asthma pathogenesis, and the microbiome could play various roles depending on the age group.
Collapse
Affiliation(s)
- Jin‐Jae Lee
- Department of Life Science Multidisciplinary Genome Institute Hallym University Chuncheon Korea
| | - Sae‐Hoon Kim
- Department of Internal Medicine Seoul National University Bundang Hospital Seongnam Korea
- Department of Internal Medicine College of Medicine Seoul National University Seoul Korea
| | - Min‐Jung Lee
- Department of Life Science Multidisciplinary Genome Institute Hallym University Chuncheon Korea
| | - Byung‐Keun Kim
- Department of Internal Medicine Seoul National University Bundang Hospital Seongnam Korea
| | - Woo‐Jung Song
- Department of Internal Medicine College of Medicine Seoul National University Seoul Korea
| | - Heung‐Woo Park
- Department of Internal Medicine College of Medicine Seoul National University Seoul Korea
| | - Sang‐Heon Cho
- Department of Internal Medicine College of Medicine Seoul National University Seoul Korea
| | - Soo‐Jong Hong
- Department of Pediatrics Childhood Asthma Atopy Center Environmental Health Center Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - Yoon‐Seok Chang
- Department of Internal Medicine Seoul National University Bundang Hospital Seongnam Korea
- Department of Internal Medicine College of Medicine Seoul National University Seoul Korea
| | - Bong‐Soo Kim
- Department of Life Science Multidisciplinary Genome Institute Hallym University Chuncheon Korea
| |
Collapse
|
174
|
Goleva E, Berdyshev E, Leung DY. Epithelial barrier repair and prevention of allergy. J Clin Invest 2019; 129:1463-1474. [PMID: 30776025 DOI: 10.1172/jci124608] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Allergic diseases have in common a dysfunctional epithelial barrier, which allows the penetration of allergens and microbes, leading to the release of type 2 cytokines that drive allergic inflammation. The accessibility of skin, compared with lung or gastrointestinal tissue, has facilitated detailed investigations into mechanisms underlying epithelial barrier dysfunction in atopic dermatitis (AD). This Review describes the formation of the skin barrier and analyzes the link between altered skin barrier formation and the pathogenesis of AD. The keratinocyte differentiation process is under tight regulation. During epidermal differentiation, keratinocytes sequentially switch gene expression programs, resulting in terminal differentiation and the formation of a mature stratum corneum, which is essential for the skin to prevent allergen or microbial invasion. Abnormalities in keratinocyte differentiation in AD skin result in hyperproliferation of the basal layer of epidermis, inhibition of markers of terminal differentiation, and barrier lipid abnormalities, compromising skin barrier and antimicrobial function. There is also compelling evidence for epithelial dysregulation in asthma, food allergy, eosinophilic esophagitis, and allergic rhinosinusitis. This Review examines current epithelial barrier repair strategies as an approach for allergy prevention or intervention.
Collapse
Affiliation(s)
- Elena Goleva
- Division of Pediatric Allergy and Clinical Immunology, Department of Pediatrics, and
| | - Evgeny Berdyshev
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Donald Ym Leung
- Division of Pediatric Allergy and Clinical Immunology, Department of Pediatrics, and.,Department of Pediatrics, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
175
|
Pulvirenti G, Parisi GF, Giallongo A, Papale M, Manti S, Savasta S, Licari A, Marseglia GL, Leonardi S. Lower Airway Microbiota. Front Pediatr 2019; 7:393. [PMID: 31612122 PMCID: PMC6776601 DOI: 10.3389/fped.2019.00393] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 09/12/2019] [Indexed: 12/14/2022] Open
Abstract
During the last several years, the interest in the role of microbiota in human health has grown significantly. For many years, the lung was considered a sterile environment, and only recently, with the use of more sophisticated techniques, has it been demonstrated that colonization by a complex population of microorganisms in lower airways also occurs in healthy subjects; a predominance of some species of Proteobacteria, Firmicutes, and Bacteroidetes phyla and with a peculiar composition in some disease conditions, such as asthma, have been noted. Lung microbiota derives mainly from the higher airways microbiota. Although we have some information about the role of gut microbiota in modulation of immune system, less it is known about the connection between lung microbiota and local and systemic immunity. There is a correlation between altered microbiota composition and some diseases or chronic states; however, despite this correlation, it has not been clearly demonstrated whether the lung microbiota dysbiosis could be a consequence or a cause of these diseases. We are far from a scientific approach to the therapeutic use of probiotics in airway diseases, but we are only at the starting point of a knowledge process in this fascinating field that could reveal important surprises, and randomized prospective studies in future could reveal more about the clinical possibilities for controlling lung microbiota. This review was aimed at updating the current knowledge in the field of airway microbiota.
Collapse
Affiliation(s)
- Giulio Pulvirenti
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giuseppe Fabio Parisi
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Alessandro Giallongo
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Maria Papale
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sara Manti
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.,Unit of Pediatric Emergency, Department of Human Pathology of the Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - Salvatore Savasta
- Department of Pediatrics, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Amelia Licari
- Department of Pediatrics, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Gian Luigi Marseglia
- Department of Pediatrics, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Salvatore Leonardi
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
176
|
Hernandez-Pacheco N, Pino-Yanes M, Flores C. Genomic Predictors of Asthma Phenotypes and Treatment Response. Front Pediatr 2019; 7:6. [PMID: 30805318 PMCID: PMC6370703 DOI: 10.3389/fped.2019.00006] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/10/2019] [Indexed: 12/11/2022] Open
Abstract
Asthma is a complex respiratory disease considered as the most common chronic condition in children. A large genetic contribution to asthma susceptibility is predicted by the clustering of asthma and allergy symptoms among relatives and the large disease heritability estimated from twin studies, ranging from 55 to 90%. Genetic basis of asthma has been extensively investigated in the past 40 years using linkage analysis and candidate-gene association studies. However, the development of dense arrays for polymorphism genotyping has enabled the transition toward genome-wide association studies (GWAS), which have led the discovery of several unanticipated asthma genes in the last 11 years. Despite this, currently known risk variants identified using many thousand samples from distinct ethnicities only explain a small proportion of asthma heritability. This review examines the main findings of the last 2 years in genomic studies of asthma using GWAS and admixture mapping studies, as well as the direction of studies fostering integrative perspectives involving omics data. Additionally, we discuss the need for assessing the whole spectrum of genetic variation in association studies of asthma susceptibility, severity, and treatment response in order to further improve our knowledge of asthma genes and predictive biomarkers. Leveraging the individual's genetic information will allow a better understanding of asthma pathogenesis and will facilitate the transition toward a more precise diagnosis and treatment.
Collapse
Affiliation(s)
- Natalia Hernandez-Pacheco
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Maria Pino-Yanes
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
| |
Collapse
|
177
|
The Role of the Microbiome in Asthma: The Gut⁻Lung Axis. Int J Mol Sci 2018; 20:ijms20010123. [PMID: 30598019 PMCID: PMC6337651 DOI: 10.3390/ijms20010123] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/22/2018] [Accepted: 12/27/2018] [Indexed: 12/14/2022] Open
Abstract
Asthma is one of the most common chronic respiratory diseases worldwide. It affects all ages but frequently begins in childhood. Initiation and exacerbations may depend on individual susceptibility, viral infections, allergen exposure, tobacco smoke exposure, and outdoor air pollution. The aim of this review was to analyze the role of the gut⁻lung axis in asthma development, considering all asthma phenotypes, and to evaluate whether microbe-based therapies may be used for asthma prevention. Several studies have confirmed the role of microbiota in the regulation of immune function and the development of atopy and asthma. These clinical conditions have apparent roots in an insufficiency of early life exposure to the diverse environmental microbiota necessary to ensure colonization of the gastrointestinal and/or respiratory tracts. Commensal microbes are necessary for the induction of a balanced, tolerogenic immune system. The identification of commensal bacteria in both the gastroenteric and respiratory tracts could be an innovative and important issue. In conclusion, the function of microbiota in healthy immune response is generally acknowledged, and gut dysbacteriosis might result in chronic inflammatory respiratory disorders, particularly asthma. Further investigations are needed to improve our understanding of the role of the microbiome in inflammation and its influence on important risk factors for asthma, including tobacco smoke and host genetic features.
Collapse
|
178
|
Abstract
The lung and gut microbiome are factors in asthma risk or protection. Relevant elements of the microbiome within both niches include the importance of the early life window for microbiome establishment, the diversity of bacteria, richness of bacteria, and effect of those bacteria on the local epithelium and immune system. Mechanisms of protection include direct anti-inflammatory action or induction of non-type 2 inflammation by certain bacterial colonies. The gut microbiome further impacts asthma risk through the contribution of metabolic products. This article reviews the mechanisms that connect the lung and gut microbiota to asthma development and severity.
Collapse
Affiliation(s)
- Tara F Carr
- Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ 85724-5030, USA.
| | - Rhonda Alkatib
- Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ 85724-5030, USA
| | - Monica Kraft
- Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ 85724-5030, USA
| |
Collapse
|
179
|
Kozik AJ, Huang YJ. The microbiome in asthma: Role in pathogenesis, phenotype, and response to treatment. Ann Allergy Asthma Immunol 2018; 122:270-275. [PMID: 30552986 DOI: 10.1016/j.anai.2018.12.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To synthesize evidence on the role of microbiota in asthma pathogenesis, phenotype, and treatment outcomes, and to provide perspective on future research directions and challenges. DATA SOURCES Studies identified from a PubMed search, including all or some of the terms "asthma," "microbiome," "microbiota," "gut," "airway," "respiratory," "lung," "viral," and "fungal". STUDY SELECTIONS Studies included and referenced based on the authors' opinion of the study design and methods, value of the research questions, and the relevance of the results to the objective of the article. RESULTS Many studies have demonstrated an important role for intestinal or upper airway microbiota in mediating the pathogenesis of childhood asthma. Fewer but robust studies have implicated a role for lower respiratory tract microbiota in adult asthma phenotype, including effects of treatments. Bacterial and fungal members of the respiratory microbiota are associated with and may drive specific molecular phenotypes of asthma in adults. CONCLUSION Current evidence supports the role of human microbiota changes in shaping asthma risk, pathogenesis, and clinical presentation. Further understanding of how microbiota functionally mediate these aspects in clinically relevant contexts will require better integration of advanced scientific tools, analytic methods, and well-designed clinical studies. These efforts should be pursued with a systems-level perspective of the complex interactions between human hosts and their microbiomes, and the impact on these interactions of changes in environmental and lifestyle factors across the lifespan.
Collapse
Affiliation(s)
- Ariangela J Kozik
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan
| | - Yvonne J Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
180
|
Mezouar S, Chantran Y, Michel J, Fabre A, Dubus JC, Leone M, Sereme Y, Mège JL, Ranque S, Desnues B, Chanez P, Vitte J. Microbiome and the immune system: From a healthy steady-state to allergy associated disruption. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.humic.2018.10.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
181
|
Katsoulis K, Ismailos G, Kipourou M, Kostikas K. Microbiota and asthma: Clinical implications. Respir Med 2018; 146:28-35. [PMID: 30665515 DOI: 10.1016/j.rmed.2018.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 10/01/2018] [Accepted: 11/20/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Konstantinos Katsoulis
- Pulmonary Department, 424 Army General Hospital, Periferiaki Odos, 56429, Efkarpia, Thessaloniki, Greece
| | - Georgios Ismailos
- Experimental-Research Center ELPEN, ELPEN Pharmaceuticals, Leoforos Marathonos 95, 19009, Pikermi, Attika, Greece
| | - Maria Kipourou
- Pulmonary Department, 424 Army General Hospital, Periferiaki Odos, 56429, Efkarpia, Thessaloniki, Greece.
| | - Konstantinos Kostikas
- 2nd Respiratory Medicine Department, University of Athens Medical School, Attikon Hospital, Athens, Greece
| |
Collapse
|
182
|
Samra M, Nam SK, Lim DH, Kim DH, Yang J, Kim YK, Kim JH. Urine Bacteria-Derived Extracellular Vesicles and Allergic Airway Diseases in Children. Int Arch Allergy Immunol 2018; 178:150-158. [PMID: 30415264 DOI: 10.1159/000492677] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/03/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Microbiota and human allergic airway diseases have been proven to be interrelated. Bacteria-derived extracellular vesicle (EV)s are known to play important roles in interbacterial and human-bacteria communications, but their relationship with allergies has not been examined yet. Urine EVs were investigated to determine whether they could be used as biomarkers for monitoring allergic airway diseases in children. METHODS Subjects were 4 groups of chronic rhinitis (CR), allergic rhinitis (AR), atopic asthma (AS) and healthy controls. Single voided urine samples were collected. Urine EVs were isolated and their DNA was extracted for 16S-rDNA pyrosequencing. RESULTS A total of 118 children participated in this study; 27, 39, 19, and 33 were in the CR, AR, AS, and control group, respectively. The AR had a significantly high Chao-1 index than that of controls. Principal component analysis revealed dysbiosis in the CR, AR, and AS compared to the controls. One phylum and 19 families and genera were significantly enriched or depleted in the disease groups compared to the controls; the Actinobacteria phylum and the Sphingomonadaceae family were more abundant in the AS and CR, the Comamonadaceae family, the Propionibacteraceae family, Propionibacterium and Enhydrobacter were more enriched in the CR, and the Methylobacteriaceae family and Methylobacterium were more abundant in each disease group, while the Enterobacteriaceae family was depleted in each disease group. CONCLUSIONS CR, AR, and AS had a distinct composition of urine EVs. Urine EVs could be an indicator for assessing allergic airway diseases in children.
Collapse
Affiliation(s)
- Mona Samra
- Department of Pediatrics, Inha University School of Medicine, Incheon, Republic of Korea.,Environmental Health Center of Allergic Rhinitis, Inha University Hospital, Incheon, Republic of Korea
| | - Soo Kyung Nam
- Department of Pediatrics, Inha University Hospital, Incheon, Republic of Korea
| | - Dae Hyun Lim
- Department of Pediatrics, Inha University School of Medicine, Incheon, Republic of Korea.,Environmental Health Center of Allergic Rhinitis, Inha University Hospital, Incheon, Republic of Korea.,Department of Pediatrics, Inha University Hospital, Incheon, Republic of Korea
| | - Dong Hyun Kim
- Environmental Health Center of Allergic Rhinitis, Inha University Hospital, Incheon, Republic of Korea.,Department of Pediatrics, Inha University Hospital, Incheon, Republic of Korea
| | - Jinho Yang
- MD Healthcare Inc., Seoul, Republic of Korea
| | | | - Jeong Hee Kim
- Department of Pediatrics, Inha University School of Medicine, Incheon, Republic of Korea, .,Environmental Health Center of Allergic Rhinitis, Inha University Hospital, Incheon, Republic of Korea, .,Department of Pediatrics, Inha University Hospital, Incheon, Republic of Korea,
| |
Collapse
|
183
|
van Dijkhuizen EHP, Aidonopoulos O, Ter Haar NM, Pires Marafon D, Magni-Manzoni S, Ioannidis YE, Putignani L, Vastert SJ, Malattia C, De Benedetti F, Martini A. Prediction of inactive disease in juvenile idiopathic arthritis: a multicentre observational cohort study. Rheumatology (Oxford) 2018; 57:1752-1760. [PMID: 29931340 DOI: 10.1093/rheumatology/key148] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Indexed: 12/13/2022] Open
Abstract
Objectives To predict the occurrence of inactive disease in JIA in the first 2 years of disease. Methods An inception cohort of 152 treatment-naïve JIA patients with disease duration <6 months was analysed. Potential predictors were baseline clinical variables, joint US, gut microbiota composition and a panel of inflammation-related compounds in blood plasma. Various algorithms were employed to predict inactive disease according to Wallace criteria at 6-month intervals in the first 2 years. Performance of the models was evaluated using the split-cohort technique. The cohort was analysed in its entirety, and separate models were developed for oligoarticular patients, polyarticular RF negative patients and ANA positive patients. Results All models analysing the cohort as a whole showed poor performance in test data [area under the curve (AUC): <0.65]. The subgroup models performed better. Inactive disease was predicted by lower baseline juvenile arthritis DAS (JADAS)-71 and lower relative abundance of the operational taxonomic unit Mogibacteriaceae for oligoarticular patients (AUC in test data: 0.69); shorter duration of morning stiffness, higher haemoglobin and lower CXCL-9 levels at baseline for polyarticular RF negative patients (AUC in test data: 0.69); and shorter duration of morning stiffness and higher baseline haemoglobin for ANA positive patients (AUC in test data: 0.72). Conclusion Inactive disease could not be predicted with satisfactory accuracy in the whole cohort, likely due to disease heterogeneity. Interesting predictors were found in more homogeneous subgroups. These need to be validated in future studies.
Collapse
Affiliation(s)
- Evert H Pieter van Dijkhuizen
- Wilhelmina Children's Hospital, Department of Paediatric Rheumatology, University Medical Centre Utrecht, Utrecht, The Netherlands.,Paediatric Rheumatology, Istituto Giannina Gaslini, Genoa, Italy
| | - Orfeas Aidonopoulos
- Institute for the Management of Information Systems, Athena Research and Innovation Centre, Athens, Greece
| | - Nienke M Ter Haar
- Wilhelmina Children's Hospital, Department of Paediatric Rheumatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | | | - Yannis E Ioannidis
- Department of Informatics and Telecommunications, University of Athens, Athens, Greece
| | | | - Sebastiaan J Vastert
- Wilhelmina Children's Hospital, Department of Paediatric Rheumatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Clara Malattia
- Paediatric Rheumatology, Istituto Giannina Gaslini, Genoa, Italy.,DINOGMI (Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili), Università degli studi di Genova, Genoa, Italy
| | | | - Alberto Martini
- Paediatric Rheumatology, Istituto Giannina Gaslini, Genoa, Italy.,DINOGMI (Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili), Università degli studi di Genova, Genoa, Italy
| |
Collapse
|
184
|
Begley L, Madapoosi S, Opron K, Ndum O, Baptist A, Rysso K, Erb-Downward JR, Huang YJ. Gut microbiota relationships to lung function and adult asthma phenotype: a pilot study. BMJ Open Respir Res 2018; 5:e000324. [PMID: 30271607 PMCID: PMC6157510 DOI: 10.1136/bmjresp-2018-000324] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/06/2018] [Indexed: 12/31/2022] Open
Abstract
Introduction Despite strong evidence that maturation patterns of the gut microbiome in early life influence the risk for childhood asthma, very little is known about gut microbiota patterns in adults with established asthma, and of greater interest relationships to phenotypic features that characterise asthma heterogeneity. Methods Fifty-eight faecal samples from 32 adults with (n=24) and without (n=8) asthma were analysed using 16S ribosomal RNA gene sequencing methods to characterise intestinal bacterial composition. Compositional stability of paired samples was evaluated and features of gut bacterial community structure analysed in relation to extensive clinical characterisation data collected from subjects, who were enrolled in a prospective observational cohort study at the University of Michigan. Results Differences in gut bacterial community structure were associated with aeroallergen sensitisation and lung function as assessed by forced expiratory volume in 1 s (FEV1) %predicted. Associations with FEV1 were consistently observed across independent analytic approaches. k-means clustering of the gut microbiota data in subjects with asthma revealed three different clusters, distinguished most strongly by FEV1 (p<0.05) and trends in differences in other clinical and inflammatory features. Conclusion In this pilot study of asthmatic and non-asthmatic subjects, significant relationships between gut microbiota composition, aeroallergen sensitisation and lung function were observed. These preliminary findings merit further study in larger cohorts to explore possible mechanistic links to asthma phenotype.
Collapse
Affiliation(s)
- Lesa Begley
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Siddharth Madapoosi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kristopher Opron
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Bioinformatics Core, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ogechukwu Ndum
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alan Baptist
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kelly Rysso
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - John R Erb-Downward
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yvonne Jean Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
185
|
Kazachkov M, Kapoor BC, Malecha PW, Wu BG, Li Y, Levine J, Erkman J, Fitzgerald K, Moy L, Segal LN. Aerodigestive dysbiosis in children with chronic cough. Pediatr Pulmonol 2018; 53:1288-1298. [PMID: 29984544 DOI: 10.1002/ppul.24115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 06/11/2018] [Indexed: 12/31/2022]
Abstract
UNLABELLED In pediatric patients with chronic cough, respiratory culture techniques commonly yield negative results. Studies using culture-independent methods have found a high relative abundance of oral microbes in the lower airways, suggesting that the topographical continuity, and dynamics of the intraluminal contents of the aerodigestive system likely influence the lower airway microbiota. We hypothesize that in subjects with chronic cough, clinical diagnosis will correlate with distinct microbial signatures detected using culture-independent methods. STUDY DESIGN AND METHODS We enrolled 36 pediatric subjects with chronic cough in a cross-sectional study. Subjects were categorized into four clinical groups: asthma, bacterial bronchitis, neurologically impaired-orally fed, and neurologically impaired enterally fed. Samples from the aerodigestive tract were obtained through bronchoscopy and upper endoscopy. 16S rRNA gene sequencing compared the microbiota from bronchoalveolar lavage (BAL), tracheal, supraglottic, esophageal, gastric, and duodenal samples. RESULTS We observed that the lower airway microbiota of asthma subjects had higher α diversity as compared with the other groups. β diversity analysis of BAL samples revealed significant differences between the groups. Among the taxonomic differences found, most differentially enriched taxa were upper airway organisms such as Rothia, Gemellaceae (u.g. or uncharacterized genus), and Granulicatella in asthma, Prevotella in bacterial bronchitis, and Veillonella in neurologically impaired orally fed subjects. Greater dissimilarity between the upper airway and lower airway microbiota was associated with increased neutrophilic airway inflammation. CONCLUSIONS Distinct dysbiotic signatures can be identified in the lower airway microbiota of pediatric subjects with chronic cough that relates to the degree and type of inflammation.
Collapse
Affiliation(s)
- Mikhail Kazachkov
- Division of Pediatric Pulmonary Medicine, New York University School of Medicine, New York, New York
| | - Bianca C Kapoor
- Division of Pediatric Pulmonary Medicine, New York University School of Medicine, New York, New York
| | - Patrick W Malecha
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Benjamin G Wu
- Division of Pulmonary and Critical Care Medicine, New York University School of Medicine, New York, New York
| | - Yonghua Li
- Division of Pulmonary and Critical Care Medicine, New York University School of Medicine, New York, New York
| | - Jeremiah Levine
- Division of Pediatric Gastroenterology, New York University School of Medicine, New York, New York
| | - Jessica Erkman
- Division of Pediatric Pulmonary Medicine, New York University School of Medicine, New York, New York
| | - Kathryn Fitzgerald
- Division of Pediatric Pulmonary Medicine, New York University School of Medicine, New York, New York
| | - Libia Moy
- Division of Pediatric Gastroenterology, New York University School of Medicine, New York, New York
| | - Leopoldo N Segal
- Division of Pulmonary and Critical Care Medicine, New York University School of Medicine, New York, New York
| |
Collapse
|
186
|
Wang L, de Ángel Solá D, Mao Y, Bielecki P, Zhu Y, Sun Z, Shan L, Flavell RA, Bazzy-Asaad A, DeWan A. Family-based study reveals decreased abundance of sputum Granulicatella in asthmatics. Allergy 2018; 73:1918-1921. [PMID: 29862523 DOI: 10.1111/all.13493] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- L. Wang
- Division of Pulmonary and Critical Care Medicine; Department of Medicine; Washington University School of Medicine; St. Louis MO USA
- Department of Chronic Disease Epidemiology; Yale University School of Public Health; New Haven CT USA
| | - D. de Ángel Solá
- Department of Pediatrics; Section of Pulmonology, Allergy, Immunology and Sleep Medicine; Yale University School of Medicine; New Haven CT USA
| | - Y. Mao
- National Institute of Environmental Health; Chinese Center for Disease Control and Prevention; Beijing China
- Department of Environmental Health Sciences; Yale University School of Public Health; New Haven CT USA
| | - P. Bielecki
- Department of Immunobiology; Yale University School of Medicine; New Haven CT USA
| | - Y. Zhu
- Department of Environmental Health Sciences; Yale University School of Public Health; New Haven CT USA
| | - Z. Sun
- National Institute of Environmental Health; Chinese Center for Disease Control and Prevention; Beijing China
| | - L. Shan
- Division of Infectious Diseases; Department of Medicine; Washington University School of Medicine; St. Louis MO USA
| | - R. A. Flavell
- Department of Immunobiology; Yale University School of Medicine; New Haven CT USA
- Howard Hughes Medical Institute; Yale University; New Haven CT USA
| | - A. Bazzy-Asaad
- Department of Pediatrics; Section of Pulmonology, Allergy, Immunology and Sleep Medicine; Yale University School of Medicine; New Haven CT USA
| | - A. DeWan
- Department of Chronic Disease Epidemiology; Yale University School of Public Health; New Haven CT USA
| |
Collapse
|
187
|
Ramakrishnan VR, Holt J, Nelson LF, Ir D, Robertson CE, Frank DN. Determinants of the Nasal Microbiome: Pilot Study of Effects of Intranasal Medication Use. ALLERGY & RHINOLOGY 2018; 9:2152656718789519. [PMID: 30128169 PMCID: PMC6088474 DOI: 10.1177/2152656718789519] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction A role for bacteria and other microbes has long been suspected in the chronic
inflammatory sinonasal diseases. Recent studies utilizing
culture-independent, sequence-based identification have demonstrated
aberrant shifts in the sinus microbiota of chronic rhinosinusitis subjects,
compared with ostensibly healthy controls. Examining how such microbiota
shifts occur and the potential for physician-prescribed interventions to
influence microbiota dynamics are the topics of the current article. Methods The nasal cavity microbiota of 5 subjects was serially examined over an
8-week period using pan-bacterial 16S rRNA gene sequencing. Four of the
subjects were administered topical mometasone furoate spray, while 1 subject
underwent a mupirocin decolonization procedure in anticipation of orthopedic
surgery. Results Measures of microbial diversity were unaffected by intranasal treatment in 2
patients and were markedly increased in the remaining 3. The increase in
microbial diversity was related to clearance of Moraxella
spp. and a simultaneous increase in members of the phylum Actinobacteria.
Both effects persisted at least 2 weeks beyond cessation of treatment.
Transient changes in the relative abundance of several bacterial genera,
including Staphylococcus and
Priopionibacteria, were also observed during
treatment. Conclusions The effects of intranasal steroids on the sinonasal microbiome are poorly
understood, despite their widespread use in treating chronic sinonasal
inflammatory disorders. In this longitudinal study, administration of
intranasal mometasone furoate or mupirocin resulted in shifts in microbial
diversity that persisted to some degree following treatment cessation.
Further characterization of these effects as well as elucidation of the
mechanism(s) underlying these changes is needed.
Collapse
Affiliation(s)
- Vijay R Ramakrishnan
- Department of Otolaryngology-Head and Neck Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Justin Holt
- Department of Otolaryngology-Head and Neck Surgery, University of Colorado School of Medicine, Aurora, Colorado.,Department of Otolaryngology-Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon
| | - Leah F Nelson
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado
| | - Diana Ir
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado
| | - Charles E Robertson
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado
| | - Daniel N Frank
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
188
|
Costa AN, Costa FMD, Campos SV, Salles RK, Athanazio RA. The pulmonary microbiome: challenges of a new paradigm. ACTA ACUST UNITED AC 2018; 44:424-432. [PMID: 30066739 PMCID: PMC6467588 DOI: 10.1590/s1806-37562017000000209] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/16/2017] [Indexed: 01/06/2023]
Abstract
The study of the human microbiome-and, more recently, that of the respiratory system-by means of sophisticated molecular biology techniques, has revealed the immense diversity of microbial colonization in humans, in human health, and in various diseases. Apparently, contrary to what has been believed, there can be nonpathogenic colonization of the lungs by microorganisms such as bacteria, fungi, and viruses. Although this physiological lung microbiome presents low colony density, it presents high diversity. However, some pathological conditions lead to a loss of that diversity, with increasing concentrations of some bacterial genera, to the detriment of others. Although we possess qualitative knowledge of the bacteria present in the lungs in different states of health or disease, that knowledge has advanced to an understanding of the interaction of this microbiota with the local and systemic immune systems, through which it modulates the immune response. Given this intrinsic relationship between the microbiota and the lungs, studies have put forth new concepts about the pathophysiological mechanisms of homeostasis in the respiratory system and the potential dysbiosis in some diseases, such as cystic fibrosis, COPD, asthma, and interstitial lung disease. This departure from the paradigm regarding knowledge of the lung microbiota has made it imperative to improve understanding of the role of the microbiome, in order to identify possible therapeutic targets and to develop innovative clinical approaches. Through this new leap of knowledge, the results of preliminary studies could translate to benefits for our patients.
Collapse
Affiliation(s)
- André Nathan Costa
- . Divisão de Pneumologia, Instituto do Coração - InCor - Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Felipe Marques da Costa
- . Divisão de Pneumologia, Instituto do Coração - InCor - Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Silvia Vidal Campos
- . Divisão de Pneumologia, Instituto do Coração - InCor - Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Roberta Karla Salles
- . Divisão de Pneumologia, Instituto do Coração - InCor - Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Rodrigo Abensur Athanazio
- . Divisão de Pneumologia, Instituto do Coração - InCor - Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| |
Collapse
|
189
|
Abstract
The introduction of 16s ribosomal RNA sequencing as a nonculture technique has led to the discovery of the presence of microbiota in the lower airways of healthy individuals. These bacterial communities may originate from the mouth and nasopharynx or from the environment by inhalation. The microbial composition of the lower airways may be modulated by dietary factors, antibiotic therapy, and microbial infections, particularly in early life. In addition, circulatory products from gut microbiota may influence the lung microbiota to maintain mucosal immunity. Recent studies have revealed that, in asthma, the lower airway microbiota show reduced diversity and community composition that is linked to severity and inflammatory phenotype. There is also a greater prevalence of proteobacteria, including Haemophilus, in symptomatic asthma. Microbial dysbiosis may contribute to both the inception and progression of asthma in infants and children, and to corticosteroid resistance in asthma. A better understanding of the regulation of the lung and gut microbiota in asthma may pave the way for targeting microbiota to prevent and treat asthma.
Collapse
|
190
|
Sitarik A, Havstad S, Levin A, Lynch SV, Fujimura K, Ownby D, Johnson C, Wegienka G. Dog introduction alters the home dust microbiota. INDOOR AIR 2018; 28:539-547. [PMID: 29468742 PMCID: PMC6003855 DOI: 10.1111/ina.12456] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 02/13/2018] [Indexed: 05/13/2023]
Abstract
Research has largely reported that dog exposure is associated with reduced allergic disease risk. Responsible mechanism(s) are not understood. The goal was to investigate whether introducing a dog into the home changes the home dust microbiota. Families without dogs or cats planning to adopt a dog and those who were not were recruited. Dust samples were collected from the homes at recruitment and 12 months later. Microbiota composition and taxa (V4 region of the 16S rRNA gene) were compared between homes that did and did not adopt a dog. A total of 91 dust samples from 54 families (27 each, dog and no dog; 17 dog and 20 no dog homes with paired samples) were analyzed. A significant dog effect was seen across time in both unweighted UniFrac and Canberra metrics (both P = .008), indicating dog introduction may result in rapid establishment of rarer and phylogenetically related taxa. A significant dog-time interaction was seen in both weighted UniFrac (P < .001) and Bray-Curtis (P = .002) metrics, suggesting that while there may not initially be large relative abundance shifts following dog introduction, differences can be seen within a year. Therefore, dog introduction into the home has both immediate effects and effects that emerge over time.
Collapse
Affiliation(s)
- Alexandra Sitarik
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI
- The in-FLAME Global Network, an affiliate of the World Universities Network (WUN), West New York, NJ 07093 USA
| | - Suzanne Havstad
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI
- The in-FLAME Global Network, an affiliate of the World Universities Network (WUN), West New York, NJ 07093 USA
| | - Albert Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI
| | - Susan V. Lynch
- Division of Gastroenterology, University of California, San Francisco, California
| | - Kei Fujimura
- Division of Gastroenterology, University of California, San Francisco, California
| | - Dennis Ownby
- Department of Pediatrics, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Christine Johnson
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI
- The in-FLAME Global Network, an affiliate of the World Universities Network (WUN), West New York, NJ 07093 USA
| | - Ganesa Wegienka
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI
- The in-FLAME Global Network, an affiliate of the World Universities Network (WUN), West New York, NJ 07093 USA
| |
Collapse
|
191
|
Durack J, Huang YJ, Nariya S, Christian LS, Ansel KM, Beigelman A, Castro M, Dyer AM, Israel E, Kraft M, Martin RJ, Mauger DT, Rosenberg SR, King TS, White SR, Denlinger LC, Holguin F, Lazarus SC, Lugogo N, Peters SP, Smith LJ, Wechsler ME, Lynch SV, Boushey HA. Bacterial biogeography of adult airways in atopic asthma. MICROBIOME 2018; 6:104. [PMID: 29885665 PMCID: PMC5994066 DOI: 10.1186/s40168-018-0487-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/25/2018] [Indexed: 05/24/2023]
Abstract
BACKGROUND Perturbations to the composition and function of bronchial bacterial communities appear to contribute to the pathophysiology of asthma. Unraveling the nature and mechanisms of these complex associations will require large longitudinal studies, for which bronchoscopy is poorly suited. Studies of samples obtained by sputum induction and nasopharyngeal brushing or lavage have also reported asthma-associated microbiota characteristics. It remains unknown, however, whether the microbiota detected in these less-invasive sample types reflect the composition of bronchial microbiota in asthma. RESULTS Bacterial microbiota in paired protected bronchial brushings (BB; n = 45), induced sputum (IS; n = 45), oral wash (OW; n = 45), and nasal brushings (NB; n = 27) from adults with mild atopic asthma (AA), atopy without asthma (ANA), and healthy controls (HC) were profiled using 16S rRNA gene sequencing. Though microbiota composition varied with sample type (p < 0.001), compositional similarity was greatest for BB-IS, particularly in AAs and ANAs. The abundance of genera detected in BB correlated with those detected in IS and OW (r median [IQR] 0.869 [0.748-0.942] and 0.822 [0.687-0.909] respectively), but not with those in NB (r = 0.004 [- 0.003-0.011]). The number of taxa shared between IS-BB and NB-BB was greater in AAs than in HCs (p < 0.05) and included taxa previously associated with asthma. Of the genera abundant in NB, only Moraxella correlated positively with abundance in BB; specific members of this genus were shared between the two compartments only in AAs. Relative abundance of Moraxella in NB of AAs correlated negatively with that of Corynebacterium but positively with markers of eosinophilic inflammation in the blood and BAL fluid. The genus, Corynebacterium, trended to dominate all NB samples of HCs but only half of AAs (p = 0.07), in whom abundance of this genus was negatively associated with markers of eosinophilic inflammation. CONCLUSIONS Induced sputum is superior to nasal brush or oral wash for assessing bronchial microbiota composition in asthmatic adults. Although compositionally similar to the bronchial microbiota, the microbiota in induced sputum are distinct, reflecting enrichment of oral bacteria. Specific bacterial genera are shared between the nasal and the bronchial mucosa which are associated with markers of systemic and bronchial inflammation.
Collapse
Affiliation(s)
- Juliana Durack
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Yvonne J Huang
- Department of Internal Medicine, Division of Pulmonary/Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Snehal Nariya
- Department of Medicine, Division of Pulmonary/Critical Care Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Laura S Christian
- Department Microbiology/Immunology and Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA, USA
| | - K Mark Ansel
- Department Microbiology/Immunology and Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA, USA
| | - Avraham Beigelman
- Division of Pediatric Allergy, Immunology and Pulmonary Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Mario Castro
- Division of Pediatric Allergy, Immunology and Pulmonary Medicine, Washington University School of Medicine, St Louis, MO, USA
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Anne-Marie Dyer
- Department of Public Health Sciences, Penn State University, Hershey, PA, USA
| | - Elliot Israel
- Department of Medicine, Brigham & Women's Hospital, Boston, MA, USA
| | - Monica Kraft
- University of Arizona, Health Sciences, Tucson, AZ, USA
| | - Richard J Martin
- Department of Medicine, National Jewish Hospital, Denver, CO, USA
| | - David T Mauger
- Department of Public Health Sciences, Penn State University, Hershey, PA, USA
| | | | - Tonya S King
- Department of Public Health Sciences, Penn State University, Hershey, PA, USA
| | - Steven R White
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Loren C Denlinger
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Fernando Holguin
- The University of Pittsburgh Asthma Institute at UPMC/UPSOM, Pittsburgh, PA, USA
| | - Stephen C Lazarus
- Department of Medicine, Division of Pulmonary/Critical Care Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Njira Lugogo
- Duke Asthma, Allergy & Airway Center, Duke University School of Medicine, Durham, NC, USA
| | | | - Lewis J Smith
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Susan V Lynch
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Homer A Boushey
- Department of Medicine, Division of Pulmonary/Critical Care Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
192
|
Lee K, Pletcher SD, Lynch SV, Goldberg AN, Cope EK. Heterogeneity of Microbiota Dysbiosis in Chronic Rhinosinusitis: Potential Clinical Implications and Microbial Community Mechanisms Contributing to Sinonasal Inflammation. Front Cell Infect Microbiol 2018; 8:168. [PMID: 29876323 PMCID: PMC5974464 DOI: 10.3389/fcimb.2018.00168] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/02/2018] [Indexed: 12/27/2022] Open
Abstract
Recent studies leveraging next-generation sequencing and functional approaches to understand the human microbiota have demonstrated the presence of diverse, niche-specific microbial communities at nearly every mucosal surface. These microbes contribute to the development and function of physiologic and immunological features that are key to host health status. Not surprisingly, several chronic inflammatory diseases have been attributed to dysbiosis of microbiota composition or function, including chronic rhinosinusitis (CRS). CRS is a heterogeneous disease characterized by inflammation of the sinonasal cavity and mucosal microbiota dysbiosis. Inflammatory phenotypes and bacterial community compositions vary considerably across individuals with CRS, complicating current studies that seek to address causality of a dysbiotic microbiome as a driver or initiator of persistent sinonasal inflammation. Murine models have provided some experimental evidence that alterations in local microbial communities and microbially-produced metabolites influence health status. In this perspective, we will discuss the clinical implications of distinct microbial compositions and community-level functions in CRS and how mucosal microbiota relate to the diverse inflammatory endotypes that are frequently observed. We will also describe specific microbial interactions that can deterministically shape the pattern of co-colonizers and the resulting metabolic products that drive or exacerbate host inflammation. These findings are discussed in the context of CRS-associated inflammation and in other chronic inflammatory diseases that share features observed in CRS. An improved understanding of CRS patient stratification offers the opportunity to personalize therapeutic regimens and to design novel treatments aimed at manipulation of the disease-associated microbiota to restore sinus health.
Collapse
Affiliation(s)
- Keehoon Lee
- Department of Biological Sciences, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, United States
| | - Steven D Pletcher
- Department of Otolaryngology Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Susan V Lynch
- Division of Medicine, Department of Gastroenterology, University of California, San Francisco, San Francisco, CA, United States
| | - Andrew N Goldberg
- Department of Otolaryngology Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Emily K Cope
- Department of Biological Sciences, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, United States
| |
Collapse
|
193
|
Novel human microbe-disease associations inference based on network consistency projection. Sci Rep 2018; 8:8034. [PMID: 29795313 PMCID: PMC5966389 DOI: 10.1038/s41598-018-26448-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 01/09/2018] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence shows that microbes are closely related to various human diseases. Obtaining a comprehensive and detailed understanding of the relationships between microbes and diseases would not only be beneficial to disease prevention, diagnosis and prognosis, but also would lead to the discovery of new drugs. However, because of a lack of data, little effort has been made to predict novel microbe-disease associations. To date, few methods have been proposed to solve the problem. In this study, we developed a new computational model based on network consistency projection to infer novel human microbe-disease associations (NCPHMDA) by integrating Gaussian interaction profile kernel similarity of microbes and diseases, and symptom-based disease similarity. NCPHMDA is a non-parametric and global network based model that combines microbe space projection and disease space projection to achieve the final prediction. Experimental results demonstrated that the integrated space projection of microbes and diseases, and symptom-based disease similarity played roles in the model performance. Cross validation frameworks and case studies further illustrated the superior predictive performance over other methods.
Collapse
|
194
|
Yang X, Wang Y, Zhao S, Wang R, Wang C. Long-term exposure to low-dose Haemophilus influenzae during allergic airway disease drives a steroid-resistant neutrophilic inflammation and promotes airway remodeling. Oncotarget 2018; 9:24898-24913. [PMID: 29861841 PMCID: PMC5982741 DOI: 10.18632/oncotarget.24653] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 01/14/2018] [Indexed: 12/19/2022] Open
Abstract
Growing evidences indicate that bacteria are associated with pathogenesis of neutrophilic asthma. However, the long-term effect of airway bacterial colonization remains unclear. We sought to establish a murine model to simulate the airway inflammation of long-term bacterial colonization, and to assess the effects of bacteria on allergic airway disease (AAD). BALB/c mice were sensitized twice and subsequently challenged with ovalbumin (OVA) and exposed to low-dose Haemophilus influenzae for approximately 2 months. Mice in treatment groups inhaled budesonide for consecutively 6 days in the last week. Airway inflammatory phenotype, immune response, phagocytic capacity, mucus production, airway remodeling and steroid sensitivity were assessed. Long-term exposure to low-dose H. influenzae during AAD did not cause serious infection but only a slightly increased airway inflammation, which resembled the colonization. Inflammatory phenotype was converted from a steroid-sensitive T helper (Th) 2-associated eosinophilic inflammation to a steroid-resistant Th17-associated neutrophilic inflammation. The increased neutrophilic inflammation was accompanied by defects in regulatory T cell (Treg)-associated immunosuppression and macrophage phagocytosis, and finally promoted mucus hypersecretion and airway remodeling. These features resembled those of refractory neutrophilic asthma in humans. These findings indicate that in asthmatic patients, airway bacterial colonization may be a potential therapeutic target. Minimizing the pathogen burden in airway, such as Haemophilus influenzae, may be beneficial.
Collapse
Affiliation(s)
- Xu Yang
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Yijie Wang
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China.,Department of Respiratory Medicine, The 305 Hospital of PLA, Beijing, 100017, China
| | - Shengtao Zhao
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China.,Department of Respiratory Medicine, Kunming General Hospital of Chengdu Military Region, Kunming, 650032, China
| | - Ran Wang
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Changzheng Wang
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| |
Collapse
|
195
|
Abstract
BACKGROUND Nasal microbiota may influence asthma pathobiology. OBJECTIVE We sought to characterize the nasal microbiome of subjects with exacerbated asthma, nonexacerbated asthma, and healthy controls to identify nasal microbiota associated with asthma activity. METHODS We performed 16S ribosomal RNA sequencing on nasal swabs obtained from 72 primarily adult subjects with exacerbated asthma (n = 20), nonexacerbated asthma (n = 31), and healthy controls (n = 21). Analyses were performed using Quantitative Insights into Microbial (QIIME); linear discriminant analysis effect size (LEfSe); Phylogenetic Investigation of Communities by Reconstruction of Unobserved States; and Statistical Analysis of Metagenomic Profiles (PICRUSt); and Statistical Analysis of Metagenomic Profiles (STAMP). Species found to be associated with asthma activity were validated using quantitative PCR. Metabolic pathways associated with differentially abundant nasal taxa were inferred through metagenomic functional prediction. RESULTS Nasal bacterial composition significantly differed among subjects with exacerbated asthma, nonexacerbated asthma, and healthy controls (permutational multivariate ANOVA, P = 2.2 × 10-2). Relative to controls, the nasal microbiota of subjects with asthma were enriched with taxa from Bacteroidetes (Wilcoxon-Mann-Whitney, r = 0.33, P = 5.1 × 10-3) and Proteobacteria (r = 0.29, P = 1.4 × 10-2). Four species were differentially abundant based on asthma status after correction for multiple comparisons: Prevotella buccalis, Padj = 1.0 × 10-2; Dialister invisus, Padj = 9.1 × 10-3; Gardnerella vaginalis, Padj = 2.8 × 10-3; Alkanindiges hongkongensis, Padj = 2.6 × 10-3. These phyla and species were also differentially abundant based on asthma activity (exacerbated asthma vs nonexacerbated asthma vs controls). Quantitative PCR confirmed species overrepresentation in asthma relative to controls for Prevotella buccalis (fold change = 130, P = 2.1 × 10-4) and Gardnerella vaginalis (fold change = 160, P = 6.8 × 10-4). Metagenomic inference revealed differential glycerolipid metabolism (Kruskal-Wallis, P = 1.9 × 10-4) based on asthma activity. CONCLUSIONS Nasal microbiome composition differs in subjects with exacerbated asthma, nonexacerbated asthma, and healthy controls. The identified nasal taxa could be further investigated for potential mechanistic roles in asthma and as possible biomarkers of asthma activity.
Collapse
|
196
|
Sokolowska M, Frei R, Lunjani N, Akdis CA, O'Mahony L. Microbiome and asthma. Asthma Res Pract 2018; 4:1. [PMID: 29318023 PMCID: PMC5755449 DOI: 10.1186/s40733-017-0037-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/18/2017] [Indexed: 12/14/2022] Open
Abstract
The mucosal immune system is in constant communication with the vast diversity of microbes present on body surfaces. The discovery of novel molecular mechanisms, which mediate host-microbe communication, have highlighted the important roles played by microbes in influencing mucosal immune responses. Dendritic cells, epithelial cells, ILCs, T regulatory cells, effector lymphocytes, NKT cells and B cells can all be influenced by the microbiome. Many of the mechanisms being described are bacterial strain- or metabolite-specific. Microbial dysbiosis in the gut and the lung is increasingly being associated with the incidence and severity of asthma. More accurate endotyping of patients with asthma may be assisted by further analysis of the composition and metabolic activity of an individual’s microbiome. In addition, the efficacy of specific therapeutics may be influenced by the microbiome and novel bacterial-based therapeutics should be considered in future clinical studies.
Collapse
Affiliation(s)
- Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Obere Strasse 22, 7270 Davos, Switzerland.,Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Remo Frei
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Obere Strasse 22, 7270 Davos, Switzerland.,Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Nonhlanhla Lunjani
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Obere Strasse 22, 7270 Davos, Switzerland.,Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland.,University of Cape Town, Cape Town, South Africa
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Obere Strasse 22, 7270 Davos, Switzerland.,Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Liam O'Mahony
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Obere Strasse 22, 7270 Davos, Switzerland
| |
Collapse
|
197
|
Durack J, Boushey HA, Huang YJ. Incorporating the airway microbiome into asthma phenotyping: Moving toward personalized medicine for noneosinophilic asthma. J Allergy Clin Immunol 2018; 141:82-83. [PMID: 28627361 PMCID: PMC5889616 DOI: 10.1016/j.jaci.2017.05.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/03/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Juliana Durack
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, Calif
| | - Homer A Boushey
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, University of California, San Francisco, Calif.
| | - Yvonne J Huang
- Division of Pulmonary & Critical Care Medicine, University of Michigan, Ann Arbor, Mich
| |
Collapse
|
198
|
Inflammatory phenotypes in patients with severe asthma are associated with distinct airway microbiology. J Allergy Clin Immunol 2018; 141:94-103.e15. [DOI: 10.1016/j.jaci.2017.03.044] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 02/28/2017] [Accepted: 03/15/2017] [Indexed: 12/31/2022]
|
199
|
Zhang I, Pletcher SD, Goldberg AN, Barker BM, Cope EK. Fungal Microbiota in Chronic Airway Inflammatory Disease and Emerging Relationships with the Host Immune Response. Front Microbiol 2017; 8:2477. [PMID: 29312187 PMCID: PMC5733051 DOI: 10.3389/fmicb.2017.02477] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 11/29/2017] [Indexed: 12/22/2022] Open
Abstract
The respiratory tract is a complex system that is inhabited by niche-specific communities of microbes including bacteria, fungi, and viruses. These complex microbial assemblages are in constant contact with the mucosal immune system and play a critical role in airway health and immune homeostasis. Changes in the composition and diversity of airway microbiota are frequently observed in patients with chronic inflammatory diseases including chronic rhinosinusitis (CRS), cystic fibrosis, allergy, and asthma. While the bacterial microbiome of the upper and lower airways has been the focus of many recent studies, the contribution of fungal microbiota to inflammation is an emerging research interest. Within the context of allergic airway disease, fungal products are important allergens and fungi are potent inducers of inflammation. In addition, murine models have provided experimental evidence that fungal microbiota in peripheral organs, notably the gastrointestinal (GI) tract, influence pulmonary health. In this review, we explore the role of the respiratory and GI microbial communities in chronic airway inflammatory disease development with a specific focus on fungal microbiome interactions with the airway immune system and fungal-bacterial interactions that likely contribute to inflammatory disease. These findings are discussed in the context of clinical and immunological features of fungal-mediated disease in CRS, allergy, and asthmatic patients. While this field is still nascent, emerging evidence suggests that dysbiotic fungal and bacterial microbiota interact to drive or exacerbate chronic airway inflammatory disease.
Collapse
Affiliation(s)
- Irene Zhang
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, United States
| | - Steven D. Pletcher
- Department of Otolaryngology Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Andrew N. Goldberg
- Department of Otolaryngology Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Bridget M. Barker
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, United States
| | - Emily K. Cope
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, United States
| |
Collapse
|
200
|
Mao Q, Jiang F, Yin R, Wang J, Xia W, Dong G, Ma W, Yang Y, Xu L, Hu J. Interplay between the lung microbiome and lung cancer. Cancer Lett 2017; 415:40-48. [PMID: 29197615 DOI: 10.1016/j.canlet.2017.11.036] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/23/2017] [Accepted: 11/27/2017] [Indexed: 12/31/2022]
Abstract
The human microbiome confers benefits or disease susceptibility to the human body through multiple pathways. Disruption of the symbiotic balance of the human microbiome is commonly found in systematic diseases such as diabetes, obesity, and chronic gastric diseases. Emerging evidence has suggested that dysbiosis of the microbiota may also play vital roles in carcinogenesis at multiple levels, e.g., by affecting metabolic, inflammatory, or immune pathways. Although the impact of the gut microbiome on the digestive cancer has been widely explored, few studies have investigated the interplay between the microbiome and lung cancer. Some recent studies have shown that certain microbes and microbiota dysbiosis are correlated with development of lung cancer. In this mini-review, we briefly summarize current research findings describing the relationship between the lung microbiome and lung cancer. We further discuss the potential mechanisms through which the lung microbiome may play a role in lung carcinogenesis and impact lung cancer treatment. A better knowledge of the interplay between the lung microbiome and lung cancer may promote the development of innovative strategies for early prevention and personalized treatment in lung cancer.
Collapse
Affiliation(s)
- Qixing Mao
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York NY10029, USA; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China; The Fourth Clinical College of Nanjing Medical University, Graduated College of Nanjing Medical University, Nanjing 210000, PR China
| | - Feng Jiang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China
| | - Rong Yin
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China
| | - Jie Wang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China
| | - Wenjie Xia
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China; The Fourth Clinical College of Nanjing Medical University, Graduated College of Nanjing Medical University, Nanjing 210000, PR China
| | - Gaochao Dong
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China
| | - Weidong Ma
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China; The Fourth Clinical College of Nanjing Medical University, Graduated College of Nanjing Medical University, Nanjing 210000, PR China
| | - Yao Yang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York NY10029, USA
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, PR China.
| | - Jianzhong Hu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York NY10029, USA.
| |
Collapse
|