51
|
Alba C, Aparicio M, González-Martínez F, González-Sánchez MI, Pérez-Moreno J, Toledo Del Castillo B, Rodríguez JM, Rodríguez-Fernández R, Fernández L. Nasal and Fecal Microbiota and Immunoprofiling of Infants With and Without RSV Bronchiolitis. Front Microbiol 2021; 12:667832. [PMID: 34140944 PMCID: PMC8203809 DOI: 10.3389/fmicb.2021.667832] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 04/08/2021] [Indexed: 12/21/2022] Open
Abstract
Bronchiolitis associated with the respiratory syncytial virus (RSV) is the leading cause of hospitalization among infants aged < 1 year. The main objective of this work was to assess the nasal and fecal microbiota and immune profiles in infants with RSV bronchiolitis, and to compare them with those of healthy infants. For this purpose, a total of 58 infants with RSV-positive bronchiolitis and 17 healthy infants (aged < 18 months) were recruited in this case-control study, which was approved by the Ethics Committee of the Hospital Gregorio Marañón. Nasal and fecal samples were obtained and submitted to bacterial microbiota analysis by 16S rDNA sequencing and to analysis of several immune factors related to inflammatory processes. Nasal samples in which Haemophilus and/or Moraxella accounted for > 20% of the total sequences were exclusively detected among infants of the bronchiolitis group. In this group, the relative abundances of Staphylococcus and Corynebacterium were significantly lower than in nasal samples from the control group while the opposite was observed for those of Haemophilus and Mannheimia. Fecal bacterial microbiota of infants with bronchiolitis was similar to that of healthy infants. Significant differences were obtained between bronchiolitis and control groups for both the frequency of detection and concentration of BAFF/TNFSF13B and sTNF.R1 in nasal samples. The concentration of BAFF/TNFSF13B was also significantly higher in fecal samples from the bronchiolitis group. In conclusion, signatures of RSV-associated bronchiolitis have been found in this study, including dominance of Haemophilus and a high concentration of BAFF/TNFSF13B, IL-8 and sTNF.R1 in nasal samples, and a high fecal concentration of BAFF/TNFSF13B.
Collapse
Affiliation(s)
- Claudio Alba
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Marina Aparicio
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Felipe González-Martínez
- Department of Pediatrics, Hospital Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - María Isabel González-Sánchez
- Department of Pediatrics, Hospital Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Jimena Pérez-Moreno
- Department of Pediatrics, Hospital Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Blanca Toledo Del Castillo
- Department of Pediatrics, Hospital Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Juan Miguel Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Rosa Rodríguez-Fernández
- Department of Pediatrics, Hospital Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Leonides Fernández
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
52
|
Raita Y, Pérez-Losada M, Freishtat RJ, Harmon B, Mansbach JM, Piedra PA, Zhu Z, Camargo CA, Hasegawa K. Integrated omics endotyping of infants with respiratory syncytial virus bronchiolitis and risk of childhood asthma. Nat Commun 2021; 12:3601. [PMID: 34127671 PMCID: PMC8203688 DOI: 10.1038/s41467-021-23859-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/17/2021] [Indexed: 02/04/2023] Open
Abstract
Respiratory syncytial virus (RSV) bronchiolitis is not only the leading cause of hospitalization in U.S. infants, but also a major risk factor for asthma development. While emerging evidence suggests clinical heterogeneity within RSV bronchiolitis, little is known about its biologically-distinct endotypes. Here, we integrated clinical, virus, airway microbiome (species-level), transcriptome, and metabolome data of 221 infants hospitalized with RSV bronchiolitis in a multicentre prospective cohort study. We identified four biologically- and clinically-meaningful endotypes: A) clinicalclassicmicrobiomeM. nonliquefaciensinflammationIFN-intermediate, B) clinicalatopicmicrobiomeS. pneumoniae/M. catarrhalisinflammationIFN-high, C) clinicalseveremicrobiomemixedinflammationIFN-low, and D) clinicalnon-atopicmicrobiomeM.catarrhalisinflammationIL-6. Particularly, compared with endotype A infants, endotype B infants-who are characterized by a high proportion of IgE sensitization and rhinovirus coinfection, S. pneumoniae/M. catarrhalis codominance, and high IFN-α and -γ response-had a significantly higher risk for developing asthma (9% vs. 38%; OR, 6.00: 95%CI, 2.08-21.9; P = 0.002). Our findings provide an evidence base for the early identification of high-risk children during a critical period of airway development.
Collapse
Affiliation(s)
- Yoshihiko Raita
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Marcos Pérez-Losada
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, The George Washington University, Washington, DC, USA
- CIBIO-InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, Campus Agrário de Vairão, Vairão, Portugal
| | - Robert J Freishtat
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
- Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Brennan Harmon
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
| | - Jonathan M Mansbach
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pedro A Piedra
- Departments of Molecular Virology and Microbiology and Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Zhaozhong Zhu
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
53
|
Abstract
The nasopharyngeal microbiome is a dynamic microbial interface of the aerodigestive tract, and a diagnostic window in the fight against respiratory infections and antimicrobial resistance. As its constituent bacteria, viruses and mycobacteria become better understood and sampling accuracy improves, diagnostics of the nasopharynx could guide more personalized care of infections of surrounding areas including the lungs, ears and sinuses. This review will summarize the current literature from a clinical perspective and highlight its growing importance in diagnostics and infectious disease management.
Collapse
Affiliation(s)
- Matthew Flynn
- School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK
- Otolaryngology Department, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
| | - James Dooley
- School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK
| |
Collapse
|
54
|
Toivonen L, Schuez-Havupalo L, Karppinen S, Waris M, Hoffman KL, Camargo CA, Hasegawa K, Peltola V. Antibiotic Treatments During Infancy, Changes in Nasal Microbiota, and Asthma Development: Population-based Cohort Study. Clin Infect Dis 2021; 72:1546-1554. [PMID: 32170305 PMCID: PMC8096219 DOI: 10.1093/cid/ciaa262] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/10/2020] [Indexed: 12/20/2022] Open
Abstract
Background Early-life exposures to antibiotics may increase the risk of developing childhood asthma. However, little is known about the mechanisms linking antibiotic exposures to asthma. We hypothesized that changes in the nasal airway microbiota serve as a causal mediator in the antibiotics–asthma link. Methods In a population-based birth-cohort study in Finland, we identified longitudinal nasal microbiota profiles during age 2–24 months using 16S rRNA gene sequencing and an unsupervised machine learning approach. We performed a causal mediation analysis to estimate the natural direct effect of systemic antibiotic treatments during age 0–11 months on risks of developing physician-diagnosed asthma by age 7 years and the natural indirect (causal mediation) effect through longitudinal changes in nasal microbiota. Results In our birth cohort of 697 children, 8.0% later developed asthma. Exposure to ≥2 antibiotic treatments during age 0–11 months was associated with a 4.0% increase in the absolute risk of developing asthma (absolute increase, 95% CI, .9–7.2%; P = .006). The unsupervised clustering approach identified 6 longitudinal nasal microbiota profiles. Infants with a larger number of antibiotic treatments had a higher risk of having a profile with early Moraxella sparsity (per each antibiotic treatment, adjusted RRR, 1.38; 95% CI, 1.15–1.66; P < .001). This effect of antibiotics on asthma was partly mediated by longitudinal changes in the nasal microbiota (natural indirect effect, P = .008), accounting for 16% of the total effect. Conclusions Early exposures to antibiotics were associated with increased risk of asthma; the effect was mediated, in part, by longitudinal changes in the nasal airway microbiota.
Collapse
Affiliation(s)
- Laura Toivonen
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Linnea Schuez-Havupalo
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Sinikka Karppinen
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Matti Waris
- Virology Unit, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Kristi L Hoffman
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ville Peltola
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
55
|
Li S, Liu J, Zhou J, Wang Y, Jin F, Chen X, Yang J, Chen Z. Urinary Metabolomic Profiling Reveals Biological Pathways and Predictive Signatures Associated with Childhood Asthma. J Asthma Allergy 2020; 13:713-724. [PMID: 33376359 PMCID: PMC7755329 DOI: 10.2147/jaa.s281198] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/20/2020] [Indexed: 12/16/2022] Open
Abstract
Background Despite considerable efforts, the pathogenic mechanisms of asthma are still incompletely understood, due to its heterogeneous nature. However, metabolomics can offer a global view of a biological system, making it a valuable tool for further elucidation of mechanisms and biomarker discovery in asthma. Methods GC-MS-based metabolomic analysis was conducted for comparison of urine metabolic profiles between asthmatic children (n=30) and healthy controls (n=30). Results An orthogonal projections to latent structures discriminant-analysis model revealed a clear separation of the asthma and control groups (R 2 x =0.137, R 2 y =0.947, Q 2=0.82). A total of 20 differential metabolites were identified as discriminant factors, of which eleven were significantly increased and nine decreased in the asthma group compared to the control group. Pathway-enrichment analysis based on these differential metabolites indicated that sphingolipid metabolism, protein biosynthesis, and citric acid cycle were strongly associated with asthma. Among the identified metabolites, 2-hydroxybutanoic acid showed excellent discriminatory performance for distinguishing asthma from healthy controls, with an AUC of 0.969. Conclusion Our study revealed significant changes in the urine metabolome of asthma patients. Several perturbed pathways (eg, sphingolipid metabolism and citric acid cycle) may be related to asthma pathogenesis, and 2-hydroxybutanoic acid could serve as a potential biomarker for asthma diagnosis.
Collapse
Affiliation(s)
- Shuxian Li
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| | - Jinling Liu
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| | - Junfen Zhou
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China.,Department of Pediatrics, Wenling Maternal and Child Health Care Hospital, Wenling, Zhejiang 317500, China
| | - Yingshuo Wang
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| | - Fang Jin
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| | - Xiaoyang Chen
- Developmental and Behavioral Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| | - Jun Yang
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310013, China.,Department of Toxicology, Hangzhou Normal University School of Public Health, Hangzhou, Zhejiang 310016, China
| | - Zhimin Chen
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| |
Collapse
|
56
|
Raita Y, Camargo CA, Bochkov YA, Celedón JC, Gern JE, Mansbach JM, Rhee EP, Freishtat RJ, Hasegawa K. Integrated-omics endotyping of infants with rhinovirus bronchiolitis and risk of childhood asthma. J Allergy Clin Immunol 2020; 147:2108-2117. [PMID: 33197460 DOI: 10.1016/j.jaci.2020.11.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/24/2020] [Accepted: 11/02/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND Young children with rhinovirus (RV) infection-particularly bronchiolitis-are at high risk for developing childhood asthma. Emerging evidence suggests clinical heterogeneity within RV bronchiolitis. However, little is known about these biologically distinct subgroups (endotypes) and their relations with asthma risk. OBJECTIVE We aimed to identify RV bronchiolitis endotypes and examine their longitudinal relations with asthma risk. METHODS As part of a multicenter prospective cohort study of infants (age <12 months) hospitalized for bronchiolitis, we integrated clinical, RV species (RV-A, RV-B, and RV-C), nasopharyngeal microbiome (16S rRNA gene sequencing), cytokine, and metabolome (liquid chromatography tandem mass spectrometry) data collected at hospitalization. We then applied network and clustering approaches to identify bronchiolitis endotypes. We also examined their longitudinal association with risks of developing recurrent wheeze by age 3 years and asthma by age 5 years. RESULTS Of 122 infants hospitalized for RV bronchiolitis (median age, 4 months), we identified 4 distinct endotypes-mainly characterized by RV species, microbiome, and type 2 cytokine (T2) response: endotype A, virusRV-CmicrobiomemixedT2low; endotype B, virusRV-AmicrobiomeHaemophilusT2low; endotype C, virusRSV/RVmicrobiomeStreptococcusT2low; and endotype D, virusRV-CmicrobiomeMoraxellaT2high. Compared with endotype A infants, endotype D infants had a significantly higher rate of recurrent wheeze (33% vs 64%; hazard ratio, 2.23; 95% CI, 1.00-4.96; P = .049) and a higher risk for developing asthma (28% vs 59%; odds ratio, 3.74: 95% CI, 1.21-12.6; P = .03). CONCLUSIONS Integrated-omics analysis identified biologically meaningful RV bronchiolitis endotypes in infants, such as one characterized by RV-C infection, Moraxella-dominant microbiota, and high T2 cytokine response, at higher risk for developing recurrent wheeze and asthma. This study should facilitate further research toward validating our inferences.
Collapse
Affiliation(s)
- Yoshihiko Raita
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass.
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Yury A Bochkov
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Juan C Celedón
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pa
| | - James E Gern
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Jonathan M Mansbach
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Eugene P Rhee
- Nephrology Division and Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Robert J Freishtat
- Division of Emergency Medicine, Children's National Hospital, Washington, DC; Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC; Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| |
Collapse
|
57
|
Hu Y, Chen J, Xu Y, Zhou H, Huang P, Ma Y, Gao M, Cheng S, Zhou H, Lv Z. Alterations of Gut Microbiome and Metabolite Profiling in Mice Infected by Schistosoma japonicum. Front Immunol 2020; 11:569727. [PMID: 33162984 PMCID: PMC7580221 DOI: 10.3389/fimmu.2020.569727] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
Schistosoma japonicum (S. japonicum) is one of the etiological agents of schistosomiasis, a widespread zoonotic parasitic disease. However, the mechanism of the balanced co-existence between the host immune system and S. japonicum as well as their complex interaction remains unclear. In this study, 16S rRNA gene sequencing, combined with metagenomic sequencing approach as well as ultraperformance liquid chromatography–mass spectrometry metabolic profiling, was applied to demonstrate changes in the gut microbiome community structure during schistosomiasis progression, the functional interactions between the gut bacteria and S. japonicum infection in BALB/c mice, and the dynamic metabolite changes of the host. The results showed that both gut microbiome and the metabolites were significantly altered at different time points after the infection. Decrease in richness and diversity as well as differed composition of the gut microbiota was observed in the infected status when compared with the uninfected status. At the phylum level, the gut microbial communities in all samples were dominated by Firmicutes, Bacteroidetes, Proteobacteria, and Deferribacteres, while at the genus level, Lactobacillus, Lachnospiraceae NK4A136 group, Bacteroides, Staphylococcus, and Alloprevotella were the most abundant. After exposure, Roseburia, and Ruminococcaceae UCG-014 decreased, while Staphylococcus, Alistipes, and Parabacteroides increased, which could raise the risk of infections. Furthermore, LEfSe demonstrated several bacterial taxa that could discriminate between each time point of S. japonicum infection. Besides that, metagenomic analysis illuminated that the AMP-activated protein kinase (AMPK) signaling pathway and the chemokine signaling pathway were significantly perturbed after the infection. Phosphatidylcholine and colfosceril palmitate in serum as well as xanthurenic acid, naphthalenesulfonic acid, and pimelylcarnitine in urine might be metabolic biomarkers due to their promising diagnostic potential at the early stage of the infection. Alterations of glycerophospholipid and purine metabolism were also discovered in the infection. The present study might provide further understanding of the mechanisms during schistosome infection in aspects of gut microbiome and metabolites, and facilitate the discovery of new targets for early diagnosis and prognostic purposes. Further validations of potential biomarkers in human populations are necessary, and the exploration of interactions among S. japonicum, gut microbiome, and metabolites is to be deepened in the future.
Collapse
Affiliation(s)
- Yue Hu
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China.,Joint Program of Pathobiology, Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jiansong Chen
- Instrumental Analysis and Research Center, Sun Yat-sen University, Guangzhou, China
| | - Yiyue Xu
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
| | - Hongli Zhou
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
| | - Ping Huang
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
| | - Yubin Ma
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
| | - Minzhao Gao
- Department of Gastroenterology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Shaoyun Cheng
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
| | - Haiyun Zhou
- Instrumental Analysis and Research Center, Sun Yat-sen University, Guangzhou, China
| | - Zhiyue Lv
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China.,Joint Program of Pathobiology, Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
58
|
Fujiogi M, Camargo CA, Raita Y, Bochkov YA, Gern JE, Mansbach JM, Piedra PA, Hasegawa K. Respiratory viruses are associated with serum metabolome among infants hospitalized for bronchiolitis: A multicenter study. Pediatr Allergy Immunol 2020; 31:755-766. [PMID: 32460384 PMCID: PMC7704725 DOI: 10.1111/pai.13296] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Bronchiolitis is the leading cause of infant hospitalizations in the United States. Growing evidence supports the heterogeneity of bronchiolitis. However, little is known about the interrelationships between major respiratory viruses (and their species), host systemic metabolism, and disease pathobiology. METHODS In an ongoing multicenter prospective cohort study, we profiled the serum metabolome in 113 infants (63 RSV-only, 21 RV-A, and 29 RV-C) hospitalized with bronchiolitis. We identified serum metabolites that are most discriminatory in the RSV-RV-A and RSV-RV-C comparisons using sparse partial least squares discriminant analysis. We then investigated the association between discriminatory metabolites with acute and chronic outcomes. RESULTS In 113 infants with bronchiolitis, we measured 639 metabolites. Serum metabolomic profiles differed in both comparisons (Ppermutation < 0.05). In the RSV-RV-A comparison, we identified 30 discriminatory metabolites, predominantly in lipid metabolism pathways (eg, sphingolipids and carnitines). In multivariable models, these metabolites were significantly associated with the risk of clinical outcomes (eg, tricosanoyl sphingomyelin, OR for recurrent wheezing at age of 3 years = 1.50; 95% CI: 1.05-2.15). In the RSV-RV-C comparison, the discriminatory metabolites were also primarily involved in lipid metabolism (eg, glycerophosphocholines [GPCs], 12,13-diHome). These metabolites were also significantly associated with the risk of outcomes (eg, 1-stearoyl-2-linoleoyl-GPC, OR for positive pressure ventilation use during hospitalization = 0.47; 95% CI: 0.28-0.78). CONCLUSION Respiratory viruses and their species had distinct serum metabolomic signatures that are associated with differential risks of acute and chronic morbidities of bronchiolitis. Our findings advance research into the complex interrelations between viruses, host systemic response, and bronchiolitis pathobiology.
Collapse
Affiliation(s)
- Michimasa Fujiogi
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carlos A. Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yoshihiko Raita
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yury A. Bochkov
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - James E. Gern
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Jonathan M. Mansbach
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Pedro A. Piedra
- Departments of Molecular Virology and Microbiology and Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
59
|
Decoding Susceptibility to Respiratory Viral Infections and Asthma Inception in Children. Int J Mol Sci 2020; 21:ijms21176372. [PMID: 32887352 PMCID: PMC7503410 DOI: 10.3390/ijms21176372] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 01/19/2023] Open
Abstract
Human Respiratory Syncytial Virus and Human Rhinovirus are the most frequent cause of respiratory tract infections in infants and children and are major triggers of acute viral bronchiolitis, wheezing and asthma exacerbations. Here, we will discuss the application of the powerful tools of systems biology to decode the molecular mechanisms that determine risk for infection and subsequent asthma. An important conceptual advance is the understanding that the innate immune system is governed by a Bow-tie architecture, where diverse input signals converge onto a few core pathways (e.g., IRF7), which in turn generate diverse outputs that orchestrate effector and regulatory functions. Molecular profiling studies in children with severe exacerbations of asthma/wheeze have identified two major immunological phenotypes. The IRF7hi phenotype is characterised by robust upregulation of antiviral response networks, and the IRF7lo phenotype is characterised by upregulation of markers of TGFβ signalling and type 2 inflammation. Similar phenotypes have been identified in infants and children with severe viral bronchiolitis. Notably, genome-wide association studies supported by experimental validation have identified key pathways that increase susceptibility to HRV infection (ORMDL3 and CHDR3) and modulate TGFβ signalling (GSDMB, TGFBR1, and SMAD3). Moreover, functional deficiencies in the activation of type I and III interferon responses are already evident at birth in children at risk of developing febrile lower respiratory tract infections and persistent asthma/wheeze, suggesting that the trajectory to asthma begins at birth or in utero. Finally, exposure to microbes and their products reprograms innate immunity and provides protection from the development of allergies and asthma in children, and therefore microbial products are logical candidates for the primary prevention of asthma.
Collapse
|
60
|
Raita Y, Camargo CA, Macias CG, Mansbach JM, Piedra PA, Porter SC, Teach SJ, Hasegawa K. Machine learning-based prediction of acute severity in infants hospitalized for bronchiolitis: a multicenter prospective study. Sci Rep 2020; 10:10979. [PMID: 32620819 PMCID: PMC7335203 DOI: 10.1038/s41598-020-67629-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 05/11/2020] [Indexed: 11/16/2022] Open
Abstract
We aimed to develop machine learning models to accurately predict bronchiolitis severity, and to compare their predictive performance with a conventional scoring (reference) model. In a 17-center prospective study of infants (aged < 1 year) hospitalized for bronchiolitis, by using routinely-available pre-hospitalization data as predictors, we developed four machine learning models: Lasso regression, elastic net regression, random forest, and gradient boosted decision tree. We compared their predictive performance-e.g., area-under-the-curve (AUC), sensitivity, specificity, and net benefit (decision curves)-using a cross-validation method, with that of the reference model. The outcomes were positive pressure ventilation use and intensive treatment (admission to intensive care unit and/or positive pressure ventilation use). Of 1,016 infants, 5.4% underwent positive pressure ventilation and 16.0% had intensive treatment. For the positive pressure ventilation outcome, machine learning models outperformed reference model (e.g., AUC 0.88 [95% CI 0.84-0.93] in gradient boosted decision tree vs 0.62 [95% CI 0.53-0.70] in reference model), with higher sensitivity (0.89 [95% CI 0.80-0.96] vs. 0.62 [95% CI 0.49-0.75]) and specificity (0.77 [95% CI 0.75-0.80] vs. 0.57 [95% CI 0.54-0.60]). The machine learning models also achieved a greater net benefit over ranges of clinical thresholds. Machine learning models consistently demonstrated a superior ability to predict acute severity and achieved greater net benefit.
Collapse
Affiliation(s)
- Yoshihiko Raita
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, 125 Nashua Street, Suite 920, Boston, MA, 02114-1101, USA.
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, 125 Nashua Street, Suite 920, Boston, MA, 02114-1101, USA
| | - Charles G Macias
- Department of Pediatric Emergency Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| | - Jonathan M Mansbach
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pedro A Piedra
- Departments of Molecular Virology and Microbiology and Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Stephen C Porter
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Division of Emergency Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Stephen J Teach
- Division of Emergency Medicine and Department of Pediatrics, Children's National Health System, Washington, DC, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, 125 Nashua Street, Suite 920, Boston, MA, 02114-1101, USA
| |
Collapse
|
61
|
Zheng DX, Mitri EJ, Garg V, Crifase CC, Sullivan AF, Espinola JA, Camargo CA. Socioeconomic Status and Bronchiolitis Severity Among Hospitalized Infants. Acad Pediatr 2020; 20:348-355. [PMID: 31254632 PMCID: PMC6930979 DOI: 10.1016/j.acap.2019.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 03/28/2019] [Accepted: 06/02/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To investigate the relationship between socioeconomic factors and bronchiolitis severity among hospitalized infants. METHODS We performed a 17-center, prospective cohort study from 2011 to 2014. Children <1 year old hospitalized with bronchiolitis were enrolled. Socioeconomic factors included estimated median household income (MHI) per home ZIP code, parent-reported household income, number of adults and children in household, and insurance type. We defined higher bronchiolitis severity as receipt of intensive care treatment. Multivariable logistic regression was used to analyze the association between socioeconomic factors and bronchiolitis severity, with the final model adjusted for potential clustering by site. RESULTS In multivariable models adjusted for demographic and clinical characteristics, estimated MHI was the socioeconomic factor most strongly associated with severity. Compared to infants with an intermediate MHI ($40,000-$79,999), odds of receiving intensive care treatment were significantly higher for those with MHI of ≥$80,000 (aOR 2.05, 95% CI 1.19-3.53). No significant associations were found for the other socioeconomic factors (all P > .30). While there were no significant differences in clinical presentation between income groups (all P > .25) or in receipt of mechanical ventilation alone (P = .98), infants with estimated MHI ≥$80,000 were significantly more likely to specifically have been admitted to the intensive care unit (P = .01). CONCLUSIONS In this multicenter study of infants hospitalized with bronchiolitis, we identified higher median household income as a risk factor for intensive care treatment. This work may yield important biological or nonbiological insights for the future management of infants with bronchiolitis.
Collapse
|
62
|
Stewart CJ, Mansbach JM, Ajami NJ, Petrosino JF, Zhu Z, Liang L, Camargo CA, Hasegawa K. Serum Metabolome Is Associated With the Nasopharyngeal Microbiota and Disease Severity Among Infants With Bronchiolitis. J Infect Dis 2020; 219:2005-2014. [PMID: 30629185 DOI: 10.1093/infdis/jiz021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 01/08/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Emerging evidence suggests relationships between the nasopharyngeal metabolome and both the microbiota and severity of bronchiolitis. However, the influence of host systemic metabolism on disease pathobiology remains unclear. We aimed to examine metabolome profiles and their association with more-severe disease, defined by use of positive pressure ventilation (PPV), in infants hospitalized for bronchiolitis. METHODS In 140 infants with bronchiolitis, metabolomic profiling was performed on serum; samples from 70 were in a training data set, and samples from 70 were in an independent test data set. We also profiled the nasopharyngeal airway microbiota and examined its association with the serum metabolites. RESULTS Serum metabolome profiles differed by bronchiolitis severity (P < .001). In total, 20 metabolites in the training data set were significantly associated with the risk of PPV, of which 18 remained significant following adjustment for confounders (false-discovery rate [FDR], < 0.10). Phosphatidylcholine metabolites were associated with higher risks of PPV use, while metabolites from the plasmalogen subpathway were associated with lower risks. The test data set validated these findings (FDR < 0.05). Streptococcus abundance was positively associated with metabolites that are associated with higher risks of PPV. CONCLUSIONS Serum metabolomic signatures were associated with both the nasopharyngeal microbiota and the severity of bronchiolitis. Our findings advance research into the complex interrelations between the airway microbiome, host systemic response, and pathobiology of bronchiolitis.
Collapse
Affiliation(s)
- Christopher J Stewart
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas.,Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | | | - Nadim J Ajami
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Zhaozhong Zhu
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, United States.,Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, United States
| | - Liming Liang
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, United States.,Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, United States
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School.,Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, United States.,Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, United States
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School
| |
Collapse
|
63
|
Groves HT, Higham SL, Moffatt MF, Cox MJ, Tregoning JS. Respiratory Viral Infection Alters the Gut Microbiota by Inducing Inappetence. mBio 2020; 11:e03236-19. [PMID: 32071269 PMCID: PMC7029140 DOI: 10.1128/mbio.03236-19] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 01/09/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory viral infections are extremely common, but their impacts on the composition and function of the gut microbiota are poorly understood. We previously observed a significant change in the gut microbiota after viral lung infection. Here, we show that weight loss during respiratory syncytial virus (RSV) or influenza virus infection was due to decreased food consumption, and that the fasting of mice altered gut microbiota composition independently of infection. While the acute phase tumor necrosis factor alpha (TNF-α) response drove early weight loss and inappetence during RSV infection, this was not sufficient to induce changes in the gut microbiota. However, the depletion of CD8+ cells increased food intake and prevented weight loss, resulting in a reversal of the gut microbiota changes normally observed during RSV infection. Viral infection also led to changes in the fecal gut metabolome, with a significant shift in lipid metabolism. Sphingolipids, polyunsaturated fatty acids (PUFAs), and the short-chain fatty acid (SCFA) valerate were all increased in abundance in the fecal metabolome following RSV infection. Whether this and the impact of infection-induced anorexia on the gut microbiota are part of a protective anti-inflammatory response during respiratory viral infections remains to be determined.IMPORTANCE The gut microbiota has an important role in health and disease: gut bacteria can generate metabolites that alter the function of immune cells systemically. Understanding the factors that can lead to changes in the gut microbiome may help to inform therapeutic interventions. This is the first study to systematically dissect the pathway of events from viral lung infection to changes in gut microbiota. We show that the cellular immune response to viral lung infection induces inappetence, which in turn alters the gut microbiome and metabolome. Strikingly, there was an increase in lipids that have been associated with the resolution of disease. This opens up new paths of investigation: first, what is the (presumably secreted) factor made by the T cells that can induce inappetence? Second, is inappetence an adaptation that accelerates recovery from infection, and if so, does the microbiome play a role in this?
Collapse
Affiliation(s)
- Helen T Groves
- Mucosal Infection and Immunity Group, Section of Virology, Department of Medicine, St Mary's Campus, Imperial College London, London, United Kingdom
| | - Sophie L Higham
- Mucosal Infection and Immunity Group, Section of Virology, Department of Medicine, St Mary's Campus, Imperial College London, London, United Kingdom
| | - Miriam F Moffatt
- National Heart & Lung Institute, Imperial College London, London, United Kingdom
- Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London, United Kingdom
| | - Michael J Cox
- National Heart & Lung Institute, Imperial College London, London, United Kingdom
- Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London, United Kingdom
| | - John S Tregoning
- Mucosal Infection and Immunity Group, Section of Virology, Department of Medicine, St Mary's Campus, Imperial College London, London, United Kingdom
| |
Collapse
|
64
|
Barlotta A, Pirillo P, Stocchero M, Donato F, Giordano G, Bont L, Zanconato S, Carraro S, Baraldi E. Metabolomic Profiling of Infants With Recurrent Wheezing After Bronchiolitis. J Infect Dis 2020; 219:1216-1223. [PMID: 30445537 PMCID: PMC7107429 DOI: 10.1093/infdis/jiy659] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/12/2018] [Indexed: 12/17/2022] Open
Abstract
Background Bronchiolitis is associated with a greater risk of developing recurrent wheezing, but with currently available tools, it is impossible to know which infants with bronchiolitis will develop this condition. This preliminary prospective study aimed to assess whether urine metabolomic analysis can be used to identify children with bronchiolitis who are at risk of developing recurrent wheezing. Methods Fifty-two infants <1 year old treated in the emergency department at University Hospital of Padova for acute bronchiolitis were enrolled (77% tested positive for respiratory syncytial virus [RSV]). Follow-up visits were conducted for 2 years after the episode of bronchiolitis. Untargeted metabolomic analyses based on mass spectrometry were performed on urine samples collected from infants with acute bronchiolitis. Data modeling was based on univariate and multivariate data analyses. Results We distinguished children with and those without postbronchiolitis recurrent wheeze, defined as ≥3 episodes of physician-diagnosed wheezing. Pathway overrepresentation analysis pointed to a major involvement of the citric acid cycle (P < .001) and some amino acids (lysine, cysteine, and methionine; P ≤ .015) in differentiating between these 2 groups of children. Conclusion This is the first study showing that metabolomic profiling of urine specimens from infants with bronchiolitis can be used to identify children at increased risk of developing recurrent wheezing.
Collapse
Affiliation(s)
- Alessia Barlotta
- Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Paola Pirillo
- Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Matteo Stocchero
- Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Filippo Donato
- Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Giuseppe Giordano
- Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Louis Bont
- Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, the Netherlands.,Department of Pediatrics, University Medical Center Utrecht, the Netherlands
| | - Stefania Zanconato
- Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Silvia Carraro
- Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Eugenio Baraldi
- Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy.,Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Respiratory Syncytial Virus Network (ReSViNET) Foundation, Zeist, the Netherlands
| |
Collapse
|
65
|
Noecker C, Chiu HC, McNally CP, Borenstein E. Defining and Evaluating Microbial Contributions to Metabolite Variation in Microbiome-Metabolome Association Studies. mSystems 2019; 4:e00579-19. [PMID: 31848305 PMCID: PMC6918031 DOI: 10.1128/msystems.00579-19] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 11/20/2019] [Indexed: 01/24/2023] Open
Abstract
Correlation-based analysis of paired microbiome-metabolome data sets is becoming a widespread research approach, aiming to comprehensively identify microbial drivers of metabolic variation. To date, however, the limitations of this approach and other microbiome-metabolome analysis methods have not been comprehensively evaluated. To address this challenge, we have introduced a mathematical framework to quantify the contribution of each taxon to metabolite variation based on uptake and secretion fluxes. We additionally used a multispecies metabolic model to simulate simplified gut communities, generating idealized microbiome-metabolome data sets. We then compared observed taxon-metabolite correlations in these data sets to calculated ground truth taxonomic contribution values. We found that in simulations of both a representative simple 10-species community and complex human gut microbiota, correlation-based analysis poorly identified key contributors, with an extremely low predictive value despite the idealized setting. We further demonstrate that the predictive value of correlation analysis is strongly influenced by both metabolite and taxon properties, as well as by exogenous environmental variation. We finally discuss the practical implications of our findings for interpreting microbiome-metabolome studies.IMPORTANCE Identifying the key microbial taxa responsible for metabolic differences between microbiomes is an important step toward understanding and manipulating microbiome metabolism. To achieve this goal, researchers commonly conduct microbiome-metabolome association studies, comprehensively measuring both the composition of species and the concentration of metabolites across a set of microbial community samples and then testing for correlations between microbes and metabolites. Here, we evaluated the utility of this general approach by first developing a rigorous mathematical definition of the contribution of each microbial taxon to metabolite variation and then examining these contributions in simulated data sets of microbial community metabolism. We found that standard correlation-based analysis of our simulated microbiome-metabolome data sets can identify true contributions with very low predictive value and that its performance depends strongly on specific properties of both metabolites and microbes, as well as on those of the surrounding environment. Combined, our findings can guide future interpretation and validation of microbiome-metabolome studies.
Collapse
Affiliation(s)
- Cecilia Noecker
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Hsuan-Chao Chiu
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Colin P McNally
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Elhanan Borenstein
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Department of Computer Science and Engineering, University of Washington, Seattle, Washington, USA
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Santa Fe Institute, Santa Fe, New Mexico, USA
| |
Collapse
|
66
|
Stewart CJ, Mansbach JM, Piedra PA, Toivonen L, Camargo CA, Hasegawa K. Association of respiratory viruses with serum metabolome in infants with severe bronchiolitis. Pediatr Allergy Immunol 2019; 30:848-851. [PMID: 31231861 PMCID: PMC6906233 DOI: 10.1111/pai.13101] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
| | - Jonathan M Mansbach
- Department of Pediatrics, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology and Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Laura Toivonen
- Department of Emergency Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Carlos A Camargo
- Department of Emergency Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
67
|
Zhu Z, Camargo CA, Hasegawa K. Metabolomics in the prevention and management of asthma. Expert Rev Respir Med 2019; 13:1135-1138. [PMID: 31561725 DOI: 10.1080/17476348.2019.1674650] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Zhaozhong Zhu
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
68
|
Sonawane AR, Tian L, Chu CY, Qiu X, Wang L, Holden-Wiltse J, Grier A, Gill SR, Caserta MT, Falsey AR, Topham DJ, Walsh EE, Mariani TJ, Weiss ST, Silverman EK, Glass K, Liu YY. Microbiome-Transcriptome Interactions Related to Severity of Respiratory Syncytial Virus Infection. Sci Rep 2019; 9:13824. [PMID: 31554845 PMCID: PMC6761288 DOI: 10.1038/s41598-019-50217-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 09/09/2019] [Indexed: 01/07/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections and hospital visits during infancy and childhood. Although risk factors for RSV infection have been identified, the role of microbial species in the respiratory tract is only partially known. We aimed to understand the impact of interactions between the nasal microbiome and host transcriptome on the severity and clinical outcomes of RSV infection. We used 16 S rRNA sequencing to characterize the nasal microbiome of infants with RSV infection. We used RNA sequencing to interrogate the transcriptome of CD4+ T cells obtained from the same set of infants. After dimension reduction through principal component (PC) analysis, we performed an integrative analysis to identify significant co-variation between microbial clade and gene expression PCs. We then employed LIONESS (Linear Interpolation to Obtain Network Estimates for Single Samples) to estimate the clade-gene association patterns for each infant. Our network-based integrative analysis identified several clade-gene associations significantly related to the severity of RSV infection. The microbial taxa with the highest loadings in the implicated clade PCs included Moraxella, Corynebacterium, Streptococcus, Haemophilus influenzae, and Staphylococcus. Interestingly, many of the genes with the highest loadings in the implicated gene PCs are encoded in mitochondrial DNA, while others are involved in the host immune response. This study on microbiome-transcriptome interactions provides insights into how the host immune system mounts a response against RSV and specific infectious agents in nasal microbiota.
Collapse
Affiliation(s)
- Abhijeet R Sonawane
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Liang Tian
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Physics, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Chin-Yi Chu
- Departments of Pediatrics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Lu Wang
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Jeanne Holden-Wiltse
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Alex Grier
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Steven R Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Mary T Caserta
- Departments of Pediatrics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ann R Falsey
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - David J Topham
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Edward E Walsh
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Thomas J Mariani
- Departments of Pediatrics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
- Center for Cancer Systems Biology, Dana Farber Cancer Institute, Boston, MA, 02115, USA.
| |
Collapse
|
69
|
Zheng P, Li Y, Wu J, Zhang H, Huang Y, Tan X, Pan J, Duan J, Liang W, Yin B, Deng F, Perry SW, Wong M, Licinio J, Wei H, Yu G, Xie P. Perturbed Microbial Ecology in Myasthenia Gravis: Evidence from the Gut Microbiome and Fecal Metabolome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901441. [PMID: 31559142 PMCID: PMC6755540 DOI: 10.1002/advs.201901441] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Indexed: 05/27/2023]
Abstract
Myasthenia gravis (MG) is a devastating acquired autoimmune disease. Emerging evidence indicates that the gut microbiome plays a key role in maintaining immune system homeostasis. This work reports that MG is characterized by decreased α-phylogenetic diversity, and significantly disturbed gut microbiome and fecal metabolome. The altered gut microbial composition is associated with fecal metabolome changes, with 38.75% of altered bacterial operational taxonomic units showing significant correlations with a range of metabolite biomarkers. Some microbes are particularly linked with MG severity. Moreover, a combination of microbial makers and their correlated metabolites enable discriminating MG from healthy controls (HCs) with 100% accuracy. To investigate whether disturbed gut mcirobiome might contribute to the onset of MG, germ-free (GF) mice are initially colonized with MG microbiota (MMb) or healthy microbiota (HMb), and then immunized in a classic mouse model of MG. The MMb mice demonstrate substantially impaired locomotion ability compared with the HMb mice. This effect could be reversed by cocolonizing GF mice with both MMb and HMb. The MMb mice also exhibit similar disturbances of fecal metabolic pathways as found in MG. Together these data demonstrate disturbances in microbiome composition and activity that are likely to be relevant to the pathogenesis of MG.
Collapse
Affiliation(s)
- Peng Zheng
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain ScienceChongqing Medical UniversityChongqing400016China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Function and DiseaseChongqing400016China
| | - Yifan Li
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain ScienceChongqing Medical UniversityChongqing400016China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Function and DiseaseChongqing400016China
| | - Jing Wu
- The M.O.E. Key Laboratory of Laboratory Medical Diagnosticsthe College of Laboratory MedicineChongqing Medical UniversityChongqing400016China
| | - Hanping Zhang
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain ScienceChongqing Medical UniversityChongqing400016China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Function and DiseaseChongqing400016China
| | - Yu Huang
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain ScienceChongqing Medical UniversityChongqing400016China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Function and DiseaseChongqing400016China
| | - Xunmin Tan
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain ScienceChongqing Medical UniversityChongqing400016China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Function and DiseaseChongqing400016China
| | - Junxi Pan
- The M.O.E. Key Laboratory of Laboratory Medical Diagnosticsthe College of Laboratory MedicineChongqing Medical UniversityChongqing400016China
| | - Jiajia Duan
- The M.O.E. Key Laboratory of Laboratory Medical Diagnosticsthe College of Laboratory MedicineChongqing Medical UniversityChongqing400016China
| | - Weiwei Liang
- The M.O.E. Key Laboratory of Laboratory Medical Diagnosticsthe College of Laboratory MedicineChongqing Medical UniversityChongqing400016China
| | - Bangmin Yin
- The M.O.E. Key Laboratory of Laboratory Medical Diagnosticsthe College of Laboratory MedicineChongqing Medical UniversityChongqing400016China
| | - Fengli Deng
- Social Medicine and Health ManagementChongqing Medical UniversityChongqing400016China
| | - Seth W. Perry
- Department of PsychiatryCollege of MedicineSUNY Upstate Medical UniversitySyracuseNY13210USA
| | - Ma‐Li Wong
- Department of PsychiatryCollege of MedicineSUNY Upstate Medical UniversitySyracuseNY13210USA
| | - Julio Licinio
- Department of PsychiatryCollege of MedicineSUNY Upstate Medical UniversitySyracuseNY13210USA
| | - Hong Wei
- Precision Medicine InstituteThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Gang Yu
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
| | - Peng Xie
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain ScienceChongqing Medical UniversityChongqing400016China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Function and DiseaseChongqing400016China
| |
Collapse
|
70
|
Forster CS, Tang Girdwood S, Morrison JM, Ambroggio L. Changing the Paradigm of Research in Pediatric Hospital Medicine. Hosp Pediatr 2019; 9:732-735. [PMID: 31444246 DOI: 10.1542/hpeds.2019-0028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Catherine S Forster
- Division of Hospitalist Medicine, Children's National Health System, Washington, District of Columbia;
| | - Sonya Tang Girdwood
- Divisions of Hospital Medicine and.,Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - John M Morrison
- Division of Pediatric Hospital Medicine, Johns Hopkins All Children's Hospital, St Petersburg, Florida; and
| | - Lilliam Ambroggio
- Sections of Emergency Medicine and.,Hospital Medicine, Department of Pediatrics, School of Medicine, University of Colorado and Children's Hospital Colorado, Aurora, Colorado
| |
Collapse
|
71
|
Dai W, Wang H, Zhou Q, Li D, Feng X, Yang Z, Wang W, Qiu C, Lu Z, Xu X, Lyu M, Xie G, Li Y, Bao Y, Liu Y, Shen K, Yao K, Feng X, Yang Y, Zhou K, Li S, Zheng Y. An integrated respiratory microbial gene catalogue to better understand the microbial aetiology of Mycoplasma pneumoniae pneumonia. Gigascience 2019; 8:giz093. [PMID: 31367746 PMCID: PMC6669060 DOI: 10.1093/gigascience/giz093] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/31/2019] [Accepted: 07/10/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The imbalanced respiratory microbiota observed in pneumonia causes high morbidity and mortality in childhood. Respiratory metagenomic analysis demands a comprehensive microbial gene catalogue, which will significantly advance our understanding of host-microorganism interactions. RESULTS We collected 334 respiratory microbial samples from 171 healthy children and 76 children with pneumonia. The respiratory microbial gene catalogue we established comprised 2.25 million non-redundant microbial genes, covering 90.52% of prevalent genes. The major oropharyngeal microbial species found in healthy children were Prevotella and Streptococcus. In children with Mycoplasma pneumoniae pneumonia (MPP), oropharyngeal microbial diversity and associated gene numbers decreased compared with those of healthy children. The concurrence network of oropharyngeal microorganisms in patients predominantly featured Staphylococcus spp. and M. pneumoniae. Functional orthologues, which are associated with the metabolism of various lipids, membrane transport, and signal transduction, accumulated in the oropharyngeal microbiome of children with pneumonia. Several antibiotic resistance genes and virulence factor genes were identified in the genomes of M. pneumoniae and 13 other microorganisms reconstructed via metagenomic data. Although the common macrolide/β-lactam resistance genes were not identified in the assembled M. pneumoniae genome, a single-nucleotide polymorphism (A2063G) related to macrolide resistance was identified in a 23S ribosomal RNA gene. CONCLUSIONS The results of this study will facilitate exploration of unknown microbial components and host-microorganism interactions in studies of the respiratory microbiome. They will also yield further insights into the microbial aetiology of MPP.
Collapse
Affiliation(s)
- Wenkui Dai
- Department of Computer Science, City University of Hong Kong, Hong Kong 999077, China
| | - Heping Wang
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen 518026, China
| | - Qian Zhou
- Department of Microbial Research, WeHealthGene Institute, Shenzhen 518000, China
| | - Dongfang Li
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, No. 1037 Luoyu Road, Wuhan 430074, China
| | - Xin Feng
- Department of Microbial Research, WeHealthGene Institute, Shenzhen 518000, China
| | - Zhenyu Yang
- Department of Microbial Research, WeHealthGene Institute, Shenzhen 518000, China
| | - Wenjian Wang
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen 518026, China
| | - Chuangzhao Qiu
- Department of Microbial Research, WeHealthGene Institute, Shenzhen 518000, China
| | - Zhiwei Lu
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen 518026, China
| | - Ximing Xu
- Institute of Statistics, Nankai University, No. 94 Weijin Road, Tianjin 300071, China
| | - Mengxuan Lyu
- Department of Computer Science, City University of Hong Kong, Hong Kong 999077, China
| | - Gan Xie
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen 518026, China
| | - Yinhu Li
- Department of Microbial Research, WeHealthGene Institute, Shenzhen 518000, China
| | - Yanmin Bao
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen 518026, China
| | - Yanhong Liu
- Department of Microbial Research, WeHealthGene Institute, Shenzhen 518000, China
| | - Kunling Shen
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen 518026, China
- Department of Respiratory Diseases, Beijing Children's Hospital, Beijing 100045, China
| | - Kaihu Yao
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen 518026, China
- Department of Respiratory Diseases, Beijing Children's Hospital, Beijing 100045, China
| | - Xikang Feng
- Department of Computer Science, City University of Hong Kong, Hong Kong 999077, China
| | - Yonghong Yang
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen 518026, China
- Department of Microbial Research, WeHealthGene Institute, Shenzhen 518000, China
- Department of Respiratory Diseases, Beijing Children's Hospital, Beijing 100045, China
| | - Ke Zhou
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, No. 1037 Luoyu Road, Wuhan 430074, China
| | - Shuaicheng Li
- Department of Computer Science, City University of Hong Kong, Hong Kong 999077, China
| | - Yuejie Zheng
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen 518026, China
| |
Collapse
|
72
|
Hasegawa K, Hoptay CE, Harmon B, Celedón JC, Mansbach JM, Piedra PA, Freishtat RJ, Camargo CA. Association of type 2 cytokines in severe rhinovirus bronchiolitis during infancy with risk of developing asthma: A multicenter prospective study. Allergy 2019; 74:1374-1377. [PMID: 30656708 DOI: 10.1111/all.13723] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Kohei Hasegawa
- Department of Emergency Medicine; Massachusetts General Hospital; Harvard Medical School; Boston Massachusetts
| | - Claire E. Hoptay
- Center for Genetic Medicine Research; Children's National Health System; Washington District of Columbia
| | - Brennan Harmon
- Center for Genetic Medicine Research; Children's National Health System; Washington District of Columbia
| | - Juan C. Celedón
- Division of Pulmonary Medicine; Department of Pediatrics; UPMC Children's Hospital of Pittsburgh; University of Pittsburgh; Pittsburgh Pennsylvania
| | | | - Pedro A. Piedra
- Department of Molecular Virology and Microbiology and Pediatrics; Baylor College of Medicine; Houston Texas
| | - Robert J. Freishtat
- Center for Genetic Medicine Research; Children's National Health System; Washington District of Columbia
- Division of Emergency Medicine; Children's National Health System; Washington District of Columbia
- Departments of Pediatrics and Integrative Systems Biology and Pediatrics; George Washington University School of Medicine and Health Sciences; Washington District of Columbia
| | - Carlos A. Camargo
- Department of Emergency Medicine; Massachusetts General Hospital; Harvard Medical School; Boston Massachusetts
| |
Collapse
|
73
|
Roquilly A, Torres A, Villadangos JA, Netea MG, Dickson R, Becher B, Asehnoune K. Pathophysiological role of respiratory dysbiosis in hospital-acquired pneumonia. THE LANCET RESPIRATORY MEDICINE 2019; 7:710-720. [PMID: 31182406 DOI: 10.1016/s2213-2600(19)30140-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/06/2019] [Accepted: 03/08/2019] [Indexed: 12/19/2022]
Abstract
Hospital-acquired pneumonia is a major cause of morbidity and mortality. The incidence of hospital-acquired pneumonia remains high globally and treatment can often be ineffective. Here, we review the available data and unanswered questions surrounding hospital-acquired pneumonia, discuss alterations of the respiratory microbiome and of the mucosal immunity in patients admitted to hospital, and explore potential approaches to stratify patients for tailored treatments. The lungs have been considered a sterile organ for decades because microbiological culture techniques had shown negative results. Culture-independent techniques have shown that healthy lungs harbour a diverse and dynamic ecosystem of bacteria, changing our comprehension of respiratory physiopathology. Understanding dysbiosis of the respiratory microbiome and altered mucosal immunity in patients with critical illness holds great promise to develop targeted host-directed immunotherapy to reduce ineffective treatment, to improve patient outcomes, and to tackle the global threat of resistant bacteria that cause these infections.
Collapse
Affiliation(s)
- A Roquilly
- Department of Anesthesiology and Critical Care, CHU Nantes, Nantes, France; Department of Microbiology and Immunology, Faculty of Medicine, University of Nantes, Nantes, France
| | - A Torres
- Servei de Pneumologia, Hospital Clinic, Universitat de Barcelona Institut d'investigació Biomédica August Pi i Sunyer, Centro de Investigación Biomédica en Red.Enfermedades Respiratorias, Barcelona, Spain
| | - J A Villadangos
- Department of Microbiology and Immunology, Doherty Institute of Infection and Immunity and Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - M G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - R Dickson
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Center for Integrative Research in Critical Care; Ann Arbor, MI, USA
| | - B Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - K Asehnoune
- Department of Anesthesiology and Critical Care, CHU Nantes, Nantes, France; Department of Microbiology and Immunology, Faculty of Medicine, University of Nantes, Nantes, France.
| |
Collapse
|
74
|
Hasegawa K, Mansbach JM, Bochkov YA, Gern JE, Piedra PA, Bauer CS, Teach SJ, Wu S, Sullivan AF, Camargo CA. Association of Rhinovirus C Bronchiolitis and Immunoglobulin E Sensitization During Infancy With Development of Recurrent Wheeze. JAMA Pediatr 2019; 173:544-552. [PMID: 30933255 PMCID: PMC6547078 DOI: 10.1001/jamapediatrics.2019.0384] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
IMPORTANCE Rhinovirus infection in early life, particularly with allergic sensitization, is associated with higher risks of developing recurrent wheeze and asthma. While emerging evidence links different rhinovirus species (eg, rhinovirus C) to a higher severity of infection and asthma exacerbation, to our knowledge, little is known about longitudinal associations of rhinovirus C infection during infancy with subsequent morbidities. OBJECTIVE To examine the association of different viruses (respiratory syncytial virus [RSV], rhinovirus species) in bronchiolitis with risks of developing recurrent wheeze. DESIGN, SETTING, AND PARTICIPANTS This multicenter prospective cohort study of infants younger than 1 year who were hospitalized for bronchiolitis was conducted at 17 hospitals across 14 US states during 3 consecutive fall to winter seasons (2011-2014). EXPOSURES Major causative viruses of bronchiolitis, including RSV (reference group) and 3 rhinovirus species (rhinovirus A, B, and C). MAIN OUTCOMES AND MEASURES Development of recurrent wheeze (as defined in national asthma guidelines) by age 3 years. RESULTS This analytic cohort comprised 716 infants who were hospitalized for RSV-only or rhinovirus bronchiolitis. The median age was 2.9 months (interquartile range, 1.6-3.8 months), 541 (76%) had bronchiolitis with RSV only, 85 (12%) had rhinovirus A, 12 (2%) had rhinovirus B, and 78 (11%) had rhinovirus C infection. Overall, 231 (32%) developed recurrent wheeze by age 3 years. In the multivariable Cox model, compared with infants with RSV-only infection, the risk of recurrent wheeze was not significantly different in those with rhinovirus A or B (rhinovirus A: hazard ratio [HR], 1.27; 95% CI, 0.86-1.88; rhinovirus B: HR, 1.39; 95% CI, 0.51-3.77; both P > .10). By contrast, infants with rhinovirus C had a significantly higher risk (HR, 1.58; 95% CI, 1.08-2.32). There was a significant interaction between virus groups and IgE sensitization on the risk of recurrent wheeze (P for interaction < .01). Only infants with both rhinovirus C infection and IgE sensitization (to food or aeroallergens) during infancy had significantly higher risks of recurrent wheeze (HR, 3.03; 95% CI, 1.20-7.61). Furthermore, compared with RSV-only, rhinovirus C infection with IgE sensitization was associated with significantly higher risks of recurrent wheeze with subsequent development of asthma at age 4 years (HR, 4.06; 95% CI, 1.17-14.1). CONCLUSIONS AND RELEVANCE This multicenter cohort study of infants hospitalized for bronchiolitis demonstrated between-virus differences in the risk of developing recurrent wheeze. Infants with rhinovirus C infection, along with IgE sensitization, had the highest risk. This finding was driven by the association with a subtype of recurrent wheeze: children with subsequent development of asthma.
Collapse
Affiliation(s)
- Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jonathan M. Mansbach
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yury A. Bochkov
- Departments of Pediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Madison
| | - James E. Gern
- Departments of Pediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Madison
| | - Pedro A. Piedra
- Department of Molecular Virology and Microbiology and Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Cindy S. Bauer
- Division of Allergy and Immunology, Phoenix Children's Hospital, Phoenix, Arizona
| | - Stephen J. Teach
- Division of Emergency Medicine and Department of Pediatrics, Children’s National Health System, Washington, DC
| | - Susan Wu
- Division of Hospital Medicine, Children's Hospital of Los Angeles, Los Angeles, California
| | - Ashley F. Sullivan
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Carlos A. Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
75
|
Toivonen L, Hasegawa K, Waris M, Ajami NJ, Petrosino JF, Camargo CA, Peltola V. Early nasal microbiota and acute respiratory infections during the first years of life. Thorax 2019; 74:592-599. [PMID: 31076501 DOI: 10.1136/thoraxjnl-2018-212629] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/16/2019] [Accepted: 04/08/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND Emerging evidence shows that airway microbiota may modulate local immune responses, thereby contributing to the susceptibility and severity of acute respiratory infections (ARIs). However, there are little data on the longitudinal relationships between airway microbiota and susceptibility to ARIs in children. OBJECTIVE We aimed to investigate the association of early nasal microbiota and the subsequent risk of ARIs during the first years of life. METHODS In this prospective population-based birth-cohort study in Finland, we followed 839 healthy infants for ARIs from birth to age 24 months. Nasal microbiota was tested using 16S rRNA gene sequencing at age 2 months. We applied an unsupervised clustering approach to identify early nasal microbiota profiles, and examined the association of profiles with the rate of ARIs during age 2-24 months. RESULTS We identified five nasal microbiota profiles dominated by Moraxella, Streptococcus, Dolosigranulum, Staphylococcus and Corynebacteriaceae, respectively. Incidence rate of ARIs was highest in children with an early Moraxella-dominant profile and lowest in those with a Corynebacteriaceae-dominant profile (738 vs 552/100 children years; unadjusted incidence rate ratio (IRR), 1.34; 95% CI 1.16 to 1.54; p < 0.001). After adjusting for nine potential confounders, the Moraxella-dominant profile-ARI association persisted (adjusted IRR (aIRR), 1.19; 95% CI 1.04 to 1.37; p = 0.01). Similarly, the incidence rate of lower respiratory tract infections (a subset of all ARIs) was significantly higher in children with an early Moraxella-dominant profile (aIRR, 2.79; 95% CI 1.04 to 8.09; p = 0.04). CONCLUSION Moraxella-dominant nasal microbiota profile in early infancy was associated with an increased rate of ARIs during the first 2 years of life.
Collapse
Affiliation(s)
- Laura Toivonen
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA .,Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Matti Waris
- Virology Unit, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Nadim J Ajami
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ville Peltola
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
76
|
Diversity of nasal microbiota and its interaction with surface microbiota among residents in healthcare institutes. Sci Rep 2019; 9:6175. [PMID: 30992494 PMCID: PMC6467918 DOI: 10.1038/s41598-019-42548-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 03/18/2019] [Indexed: 12/17/2022] Open
Abstract
Nasal microbial communities may have crucial implications for human health, including for residents of healthcare institutes (HCIs). Factors that determine the diversity of nasal microbiota in HCIs remain unclear. Herein, we used 16S rRNA amplicon sequencing to investigate the relationship between nasal and surface microbiota in three HCIs. Participants were classified into a hospitalised or nonhospitalised group based on their most recent date of hospitalisation. A total of 88 nasal samples and 83 surface samples were analysed. Dysgonomonas and Corynebacterium were the most abundant taxa in the surface and nasal samples, respectively. Significant differences were discovered in microbiota diversity among HCIs when comparing the surface and nasal samples. Fifteen taxa were identified as present in all the surface and nasal samples. SourceTracker analysis revealed that the ventilation conditions of environment might be associated with the proportion of shared microbial communities between nasal and surface. Additionally, as compared with the nonhospitalised group, the hospitalised group had a higher proportion of surface microbiota in their nasal samples, which might lead to a higher risk of human-related microorganisms or pathogens colonising the nasal cavity. The data suggest that nasal bacterial diversity could be influenced by both health status and living environment. Our results therefore highlight the importance of the indoor environment for HCI residents.
Collapse
|
77
|
Korten I, Ramsey K, Mika M, Usemann J, Frey U, Hilty M, Latzin P. Nasal Microbiota and Respiratory Tract Infections: The Role of Viral Detection. Am J Respir Crit Care Med 2019. [DOI: 10.1164/rccm.201710-2020le] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Insa Korten
- University of BernBern, Switzerland
- University Children’s HospitalBasel, Switzerlandand
| | | | | | - Jakob Usemann
- University of BernBern, Switzerland
- University Children’s HospitalBasel, Switzerlandand
| | - Urs Frey
- University Children’s HospitalBasel, Switzerlandand
| | - Markus Hilty
- University of BernBern, Switzerland
- University HospitalBern, Switzerland
| | | |
Collapse
|
78
|
Abstract
Antenatal and preschool factors are key in determining the progression to pre-school wheeze and eosinophilic school age asthma. The conventional view of eosinophilic asthma is that airway inflammation is the fundamental underlying abnormality, and airway inflammation and hyper-responsiveness are secondary; in fact, these three are parallel processes. Very early structural changes, independent of inflammation and infection, are associated with early airway hyper-responsiveness and later adverse respiratory outcomes. There is a bidirectional relationship between structural airway wall changes and airway inflammation, with airway contraction per se leading to the release of growth factors, and inflammatory pathways promoting airway remodeling. Early viral infection (and increasingly being appreciated, bacterial infection) is important in wheeze outcomes. There is evidence of abnormal immune function including cytokine release before the onset of viral infections. However, viral infections may also have prolonged effects on the host immune system, and the evidence for beneficial and adverse effects of viral infection is conflicting. In older children and adults, asthmatic epithelial cells show impaired interferon responses to viral infection, but only in the presence of uncontrolled type 2 inflammation, implying these are secondary phenomena. There are also compelling data relating the innate immune system to later asthma and atopy, and animal studies suggest that the effects of a high endotoxin, microbiologically diverse environment may be modulated via the epithelial alarmin IL-33. Whereas, previously only viral infection was thought to be important, early bacterial colonization of the upper airway is coming to the fore, associated with a mixed pattern of TH1/TH2/TH17 cytokine secretion, and adverse long term outcomes. Bacterial colonization is probably a marker of a subtle immune deficiency, rather than directly causal. The airway and gut microbiome critically impacts the development of Type 2 inflammatory responses. However, Type 2 inflammatory cytokines, which are critical both to progression from pre-school wheeze to eosinophilic asthma, and sustaining the eosinophilic asthmatic state, are not implicated in the very early development of the disease. Taken together, the evidence is that the earliest cytokine and chemokine signals will come from the study of bronchial epithelial cell function and their interactions with viruses and the microbiome.
Collapse
Affiliation(s)
- Andrew Bush
- Departments of Paediatrics and Paediatric Respiratory Medicine, Royal Brompton Harefield NHS Foundation Trust and Imperial College, London, United Kingdom
| |
Collapse
|
79
|
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: 7.6] [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
|
80
|
Abstract
Respiratory syncytial virus (RSV) is the most common cause of infant hospitalization and causes a high burden of disease in the elderly, too. This enveloped negative-stranded RNA virus has been recently reclassified in the Pneumoviridae family. Infections of the respiratory cells happens when the two major surface glycoproteins, G and F, take contact with the cell receptor CX3CR1 and mediate entry by fusion, respectively. Viral mRNA transcription, genomic RNA synthesis and nucleocapsid formation occur in large cytoplasmic inclusion bodies to avoid recognition by the host innate immune response. Most progeny virions remain associated to the infected cell surface; fusion of infected with adjacent cells results in the formation of large multinucleated syncytia that eventually undergo apoptosis. Desquamated epithelial cells form the plugs that with mucus and fibrin may cause lower airway obstructions. Pathogenetic mechanism of severe RSV disease likely involve both the extent of viral replication and the host immune response. Regarding the latter, single nucleotide polymorphism analysis and genome-wide association studies showed that genetic susceptibility to severe RSV infection is likely a complex trait, in which many different host genetic variants contribute. Recent studies pointed to the fact that bronchiolitis severity depends more on the specific infecting RSV genotypes than on the amount of viral loads. A population-based surveillance system to better define RSV burden of disease would be of valuable help for implementing future vaccination programs.
Collapse
Affiliation(s)
- Alessandra Pierangeli
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, Rome, Italy -
| | - Carolina Scagnolari
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Guido Antonelli
- Laboratory of Virology, Department of Molecular Medicine, Sapienza University, Rome, Italy
| |
Collapse
|
81
|
Lal CV, Kandasamy J, Dolma K, Ramani M, Kumar R, Wilson L, Aghai Z, Barnes S, Blalock JE, Gaggar A, Bhandari V, Ambalavanan N. Early airway microbial metagenomic and metabolomic signatures are associated with development of severe bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2018; 315:L810-L815. [PMID: 30113227 DOI: 10.1152/ajplung.00085.2018] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The pathogenesis of bronchopulmonary dysplasia (BPD) is not well understood. We previously identified differences in the airway microbiome at birth between preterm infants who were BPD predisposed versus those who were BPD resistant. In this study, we attempted to identify mechanisms by which the airway microbiome could modify the risk for BPD. We used a software-based method to predict the metagenome of the tracheal aspirate (TA) microbiome from 16S rRNA sequencing data in preterm infants and to identify functional ortholog genes that were differentially abundant in BPD-predisposed and BPD-resistant infants. We also identified metabolites that were differentially enriched in these samples by use of untargeted mass spectrometry and mummichog to identify the metabolic pathways involved. Microbial metagenome analysis identified specific pathways that were less abundant in the functional metagenome of the microbiota of BPD-predisposed infants compared with BPD-resistant infants. The airway metabolome of BPD-predisposed infants was enriched for metabolites involved in fatty acid activation and androgen and estrogen biosynthesis compared with BPD-resistant infants. These findings suggest that in extremely preterm infants the early airway microbiome may alter the metabolome, thereby modifying the risk of BPD. The differential enrichment of sex steroid metabolic pathways supports previous studies suggesting a role for sexual dimorphism in BPD risk. This study also suggests a role for metabolomic and metagenomic profiles to serve as early biomarkers of BPD risk.
Collapse
Affiliation(s)
| | - Jegen Kandasamy
- Department of Pediatrics, University of Alabama , Birmingham, Alabama
| | - Kalsang Dolma
- Department of Pediatrics, University of Alabama , Birmingham, Alabama
| | - Manimaran Ramani
- Department of Pediatrics, University of Alabama , Birmingham, Alabama
| | - Ranjit Kumar
- Department of Pediatrics, University of Alabama , Birmingham, Alabama
| | - Landon Wilson
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama , Birmingham, Alabama
| | - Zubair Aghai
- Department of Pediatrics, Thomas Jefferson University/Nemours , Philadelphia, Pennsylvania
| | - Stephen Barnes
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama , Birmingham, Alabama
| | - J Edwin Blalock
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama , Birmingham, Alabama
| | - Amit Gaggar
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama , Birmingham, Alabama
| | - Vineet Bhandari
- Department of Pediatrics, Drexel University , Philadelphia, Pennsylvania
| | | |
Collapse
|
82
|
Hasegawa K, Stewart CJ, Celedón JC, Mansbach JM, Tierney C, Camargo CA. Serum 25-hydroxyvitamin D, metabolome, and bronchiolitis severity among infants-A multicenter cohort study. Pediatr Allergy Immunol 2018; 29:441-445. [PMID: 29512857 PMCID: PMC5992049 DOI: 10.1111/pai.12880] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Christopher J. Stewart
- Institute for Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Juan C. Celedón
- Division of Pulmonary Medicine, Allergy, and Immunology, Department of Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA
| | | | - Courtney Tierney
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Carlos A. Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
83
|
Hasegawa K, Stewart CJ, Celedón JC, Mansbach JM, Tierney C, Camargo CA. Circulating 25-hydroxyvitamin D, nasopharyngeal airway metabolome, and bronchiolitis severity. Allergy 2018; 73:1135-1140. [PMID: 29315663 DOI: 10.1111/all.13379] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2017] [Indexed: 12/25/2022]
Abstract
Low circulating 25-hydroxyvitamin D (25OHD) levels are a risk factor for acute respiratory infection (eg, bronchiolitis) in children. However, little is known about the relation of circulating 25OHD with the many downstream functional molecules in target organs-such as the airway-and with clinical outcomes. In this prospective multicenter study of infants (age <1 year) hospitalized with bronchiolitis, we measured serum 25OHD levels and profiled the metabolome of 144 nasopharyngeal airway samples. Among 254 metabolites identified, we defined a set of 20 metabolites that are related to lower serum 25OHD and higher vitamin D-binding protein levels. Of these metabolites, 9 metabolites were associated with a significantly higher risk of positive pressure ventilation use. These metabolites were glycerophosphocholines esterified with proinflammatory fatty acids (palmitate, arachidonate, linoleate, and stearate), sphingomyelins, alpha-hydroxyisovalerate, 2-hydroxybutyrate, and 3-(4-hydroxyphenyl)lactate (all FDR<0.05). Based on the multicenter data, vitamin D-related airway metabolites were associated with risks of bronchiolitis severity.
Collapse
Affiliation(s)
- K. Hasegawa
- Department of Emergency Medicine; Massachusetts General Hospital; Harvard Medical School; Boston MA USA
| | - C. J. Stewart
- Alkek Center for Metagenomics and Microbiome Research; Department of Molecular Virology and Microbiology; Baylor College of Medicine; Houston TX USA
| | - J. C. Celedón
- Division of Pulmonary Medicine, Allergy, and Immunology; Department of Pediatrics; Children's Hospital of Pittsburgh; University of Pittsburgh; Pittsburgh PA USA
| | - J. M. Mansbach
- Department of Medicine; Boston Children's Hospital; Boston MA USA
| | - C. Tierney
- Department of Emergency Medicine; Massachusetts General Hospital; Harvard Medical School; Boston MA USA
| | - C. A. Camargo
- Department of Emergency Medicine; Massachusetts General Hospital; Harvard Medical School; Boston MA USA
| |
Collapse
|
84
|
Stewart CJ, Hasegawa K, Wong MC, Ajami NJ, Petrosino JF, Piedra PA, Espinola JA, Tierney CN, Camargo CA, Mansbach JM. Respiratory Syncytial Virus and Rhinovirus Bronchiolitis Are Associated With Distinct Metabolic Pathways. J Infect Dis 2018; 217:1160-1169. [PMID: 29293990 PMCID: PMC5939849 DOI: 10.1093/infdis/jix680] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/22/2017] [Indexed: 11/12/2022] Open
Abstract
Background Bronchiolitis, the leading cause of hospitalization among infants in the United States, is most commonly caused by respiratory syncytial virus (RSV), followed by rhinovirus (RV). Conventional perception is that bronchiolitis is a single entity, albeit with different viral etiologies and degrees of severity. Methods We conducted a cross-sectional study of nasopharyngeal aspirates from 106 infants hospitalized with bronchiolitis due to either RSV only (80 patients) or RV only (26 patients). We performed metabolomics analysis and 16S ribosomal RNA gene sequencing on all samples and metagenomic sequencing on 58 of 106 samples. Results Infants with RSV-only and RV-only infections had significantly different nasopharyngeal metabolome profiles (P < .001) and bacterial metagenome profiles (P < .05). RSV-only infection was associated with metabolites from a range of pathways and with a microbiome dominated by Streptococcus pneumoniae. By contrast, RV-only infection was associated with increased levels of essential and nonessential N-acetyl amino acids and with a high relative abundance of Haemophilus influenzae. These co-occurring species were associated with driving the bacterially derived metabolic pathways. Multi-omic analysis showed that both the virus and the microbiome were significantly associated with metabolic function in infants hospitalized with bronchiolitis. Conclusion Although replication of these findings is necessary, they highlight that bronchiolitis is not a uniform disease between RSV and RV infections, a result with future implications for prevention and treatment.
Collapse
Affiliation(s)
- Christopher J Stewart
- Alkek Center for Metagenomics and Microbiome Research, Houston, Texas
- Department of Molecular Virology and Microbiology, Houston, Texas
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Matthew C Wong
- Alkek Center for Metagenomics and Microbiome Research, Houston, Texas
- Department of Molecular Virology and Microbiology, Houston, Texas
| | - Nadim J Ajami
- Alkek Center for Metagenomics and Microbiome Research, Houston, Texas
- Department of Molecular Virology and Microbiology, Houston, Texas
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Houston, Texas
- Department of Molecular Virology and Microbiology, Houston, Texas
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Janice A Espinola
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Courtney N Tierney
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jonathan M Mansbach
- Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
85
|
Hasegawa K, Pérez-Losada M, Hoptay CE, Epstein S, Mansbach JM, Teach SJ, Piedra PA, Camargo CA, Freishtat RJ. RSV vs. rhinovirus bronchiolitis: difference in nasal airway microRNA profiles and NFκB signaling. Pediatr Res 2018; 83:606-614. [PMID: 29244796 PMCID: PMC6174252 DOI: 10.1038/pr.2017.309] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 11/25/2017] [Indexed: 01/03/2023]
Abstract
BackgroundAlthough rhinovirus infection is associated with increased risks of acute and chronic respiratory outcomes during childhood compared with respiratory syncytial virus (RSV), the underlying mechanisms remain unclear. We aimed to determine the differences in nasal airway microRNA profiles and their downstream effects between infants with rhinovirus and RSV bronchiolitis.MethodsAs part of a multicenter cohort study of infants hospitalized for bronchiolitis, we examined nasal samples obtained from 16 infants with rhinovirus and 16 infants with RSV. We tested nasal airway samples using microarrays to profile global microRNA expression and determine the predicted regulation of targeted transcripts. We also measured gene expression and cytokines for NFκB pathway components.ResultsBetween the virus groups, 386 microRNAs were differentially expressed (false discovery rate (FDR)<0.05). In infants with rhinovirus, the NFκB pathway was highly ranked as a predicted target for these differentially expressed microRNAs compared with RSV. Pathway analysis using measured mRNA expression data validated that rhinovirus infection had upregulation of NFκB family (RelA and NFκB2) and downregulation of inhibitor κB family. Infants with rhinovirus had higher levels of NFκB-induced type-2 cytokines (IL-10 and IL-13; FDR<0.01).ConclusionIn infants with bronchiolitis, rhinovirus and RSV infections had different nasal airway microRNA profiles associated with NFκB signaling.
Collapse
Affiliation(s)
- Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Marcos Pérez-Losada
- Computational Biology Institute, George Washington University, Ashburn, VA;,Department of Pediatrics, George Washington University School of Medicine and Health Sciences and the Division of Emergency Medicine, Children’s National Health System, Washington, DC;,CIBIO-InBIO, Universidade do Porto, Campus Agrário de Vairão, Vairão, Portugal
| | - Claire E. Hoptay
- Center for Genetic Medicine Research, Children’s National Health System, Washington, DC
| | - Samuel Epstein
- Center for Genetic Medicine Research, Children’s National Health System, Washington, DC
| | | | - Stephen J. Teach
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences and the Division of Emergency Medicine, Children’s National Health System, Washington, DC
| | - Pedro A. Piedra
- Department of Molecular Virology and Microbiology and Pediatrics, Baylor College of Medicine, Houston, TX
| | - Carlos A. Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Robert J. Freishtat
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences and the Division of Emergency Medicine, Children’s National Health System, Washington, DC;,Center for Genetic Medicine Research, Children’s National Health System, Washington, DC;,Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC;,Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
86
|
Ederveen THA, Ferwerda G, Ahout IM, Vissers M, de Groot R, Boekhorst J, Timmerman HM, Huynen MA, van Hijum SAFT, de Jonge MI. Haemophilus is overrepresented in the nasopharynx of infants hospitalized with RSV infection and associated with increased viral load and enhanced mucosal CXCL8 responses. MICROBIOME 2018; 6:10. [PMID: 29325581 PMCID: PMC5765694 DOI: 10.1186/s40168-017-0395-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 12/21/2017] [Indexed: 05/09/2023]
Abstract
BACKGROUND While almost all infants are infected with respiratory syncytial virus (RSV) before the age of 2 years, only a small percentage develops severe disease. Previous studies suggest that the nasopharyngeal microbiome affects disease development. We therefore studied the effect of the nasopharyngeal microbiome on viral load and mucosal cytokine responses, two important factors influencing the pathophysiology of RSV disease. To determine the relation between (i) the microbiome of the upper respiratory tract, (ii) viral load, and (iii) host mucosal inflammation during an RSV infection, nasopharyngeal microbiota profiles of RSV infected infants (< 6 months) with different levels of disease severity and age-matched healthy controls were determined by 16S rRNA marker gene sequencing. The viral load was measured using qPCR. Nasopharyngeal CCL5, CXCL10, MMP9, IL6, and CXCL8 levels were determined with ELISA. RESULTS Viral load in nasopharyngeal aspirates of patients associates significantly to total nasopharyngeal microbiota composition. Healthy infants (n = 21) and RSV patients (n = 54) display very distinct microbial patterns, primarily characterized by a loss in commensals like Veillonella and overrepresentation of opportunistic organisms like Haemophilus and Achromobacter in RSV-infected individuals. Furthermore, nasopharyngeal microbiota profiles are significantly different based on CXCL8 levels. CXCL8 is a chemokine that was previously found to be indicative for disease severity and for which we find Haemophilus abundance as the strongest predictor for CXCL8 levels. CONCLUSIONS The nasopharyngeal microbiota in young infants with RSV infection is marked by an overrepresentation of the genus Haemophilus. We present that this bacterium is associated with viral load and mucosal CXCL8 responses, both which are involved in RSV disease pathogenesis.
Collapse
Affiliation(s)
- Thomas H A Ederveen
- Center for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerben Ferwerda
- Laboratory of Pediatric Infectious Diseases, Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein-Zuid 10 (Route 412), 6525, GA, Nijmegen, The Netherlands.
| | - Inge M Ahout
- Laboratory of Pediatric Infectious Diseases, Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein-Zuid 10 (Route 412), 6525, GA, Nijmegen, The Netherlands
| | - Marloes Vissers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Ronald de Groot
- Laboratory of Pediatric Infectious Diseases, Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein-Zuid 10 (Route 412), 6525, GA, Nijmegen, The Netherlands
| | - Jos Boekhorst
- Center for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- NIZO, Ede, The Netherlands
| | | | - Martijn A Huynen
- Center for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sacha A F T van Hijum
- Center for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- NIZO, Ede, The Netherlands
| | - Marien I de Jonge
- Laboratory of Pediatric Infectious Diseases, Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein-Zuid 10 (Route 412), 6525, GA, Nijmegen, The Netherlands
| |
Collapse
|
87
|
Quinn RA. Integrating Microbiome and Metabolome Data to Understand Infectious Airway Disease. Am J Respir Crit Care Med 2017. [PMID: 28628375 DOI: 10.1164/rccm.201704-0671ed] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Robert A Quinn
- 1 Skaggs School of Pharmacy and Pharmaceutical Sciences and.,2 Center for Microbiome Innovation University of California at San Diego La Jolla, California
| |
Collapse
|
88
|
The lung microbiome. Emerg Top Life Sci 2017; 1:313-324. [PMID: 33525774 DOI: 10.1042/etls20170043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 08/31/2017] [Accepted: 09/29/2017] [Indexed: 12/17/2022]
Abstract
Historically, our understanding of lung microbiology has relied on insight gained through culture-based diagnostic approaches that employ selective culture conditions to isolate specific pathogens. The relatively recent development of culture-independent microbiota-profiling techniques, particularly 16S rRNA (ribosomal ribonucleic acid) gene amplicon sequencing, has enabled more comprehensive characterisation of the microbial content of respiratory samples. The widespread application of such techniques has led to a fundamental shift in our view of respiratory microbiology. Rather than a sterile lung environment that can become colonised by microbes during infection, it appears that a more nuanced balance exists between what we consider respiratory health and disease, mediated by mechanisms that influence the clearance of microbes from the lungs. Where airway defences are compromised, the ongoing transient exposure of the lower airways to microbes can lead to the establishment of complex microbial communities within the lung. Importantly, the characteristics of these communities, and the manner in which they influence lung pathogenesis, can be very different from those of their constituent members when viewed in isolation. The lung microbiome, a construct that incorporates microbes, their genetic material, and the products of microbial genes, is increasingly central to our understanding of the regulation of respiratory physiology and the processes that underlie lung pathogenesis.
Collapse
|
89
|
Hasegawa K, Stewart CJ, Mansbach JM, Linnemann RW, Ajami NJ, Petrosino JF, Camargo CA. Sphingolipid metabolism potential in fecal microbiome and bronchiolitis in infants: a case-control study. BMC Res Notes 2017; 10:325. [PMID: 28747215 PMCID: PMC5530560 DOI: 10.1186/s13104-017-2659-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/21/2017] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Emerging evidence demonstrated that the structure of fecal microbiome is associated with the likelihood of bronchiolitis in infants. However, no study has examined functional profiles of fecal microbiome in infants with bronchiolitis. In this context, we conducted a case-control study. As a part of multicenter prospective study, we collected stool samples from 40 infants hospitalized with bronchiolitis (cases). We concurrently enrolled 115 age-matched healthy controls. RESULTS First, by applying 16S rRNA gene sequencing to these 155 fecal samples, we identified the taxonomic profiles of fecal microbiome. Next, based on the taxonomy data, we inferred the functional capabilities of fecal microbiome and tested for differences in the functional capabilities between cases and controls. Overall, the median age was 3 months and 45% were female. Among 274 metabolic pathways surveyed, there were significant differences between bronchiolitis cases and healthy controls for 37 pathways, including lipid metabolic pathways (false discovery rate [FDR] <0.05). Particularly, the fecal microbiome of bronchiolitis cases had consistently higher abundances of gene function related to the sphingolipid metabolic pathways compared to that of controls (FDR <0.05). These pathways were more abundant in infants with Bacteroides-dominant microbiome profile compared to the others (FDR <0.001). On the basis of the predicted metagenome in this case-control study, we found significant differences in the functional potential of fecal microbiome between infants with bronchiolitis and healthy controls. Although causal inferences remain premature, our data suggest a potential link between the bacteria-derived metabolites, modulations of host immune response, and development of bronchiolitis.
Collapse
Affiliation(s)
- Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, 125 Nashua Street, Suite 920, Boston, MA 02114-1101 USA
| | - Christopher J. Stewart
- Department of Molecular Virology and Microbiology, Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX USA
| | | | - Rachel W. Linnemann
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA USA
| | - Nadim J. Ajami
- Department of Molecular Virology and Microbiology, Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX USA
| | - Joseph F. Petrosino
- Department of Molecular Virology and Microbiology, Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX USA
| | - Carlos A. Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, 125 Nashua Street, Suite 920, Boston, MA 02114-1101 USA
| |
Collapse
|