Microbiome-Transcriptome Interactions Related to Severity of Respiratory Syncytial Virus Infection

Abhijeet R Sonawane, Liang Tian, Chin-Yi Chu, Xing Qiu, Lu Wang, Jeanne Holden-Wiltse, Alex Grier, Steven R Gill, Mary T Caserta, Ann R Falsey, David J Topham, Edward E Walsh, Thomas J Mariani, Scott T Weiss, Edwin K Silverman, Kimberly Glass, Yang-Yu Liu, Abhijeet R Sonawane, Liang Tian, Chin-Yi Chu, Xing Qiu, Lu Wang, Jeanne Holden-Wiltse, Alex Grier, Steven R Gill, Mary T Caserta, Ann R Falsey, David J Topham, Edward E Walsh, Thomas J Mariani, Scott T Weiss, Edwin K Silverman, Kimberly Glass, Yang-Yu Liu

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.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
An overview of the data and analyses performed in this study.
Figure 2
Figure 2
(A) Overview of study timelines, including when each type of data was collected. All 58 infants included in this study had both transcriptomic and microbiome data collected for at least one time point. (B) Venn diagrams showing the overlap in the infants that had either transcriptomic or microbiome data collected during the acute visit (57 of the 58 infants), the overlap in the transcriptomic data collected at either the initial (Visit 1) or follow-up visit (Visit 3), and the overlap in the infants microbiome data collected at either the initial (Visit 1) or follow-up visit (Visit 2). Numbers are shown separately for mild and severe groups, with the total number in each category indicated below the Venn diagram. (C) Differential gene expression analysis comparing Visit 1 versus Visit 2 RNA-Seq samples yields 27 significantly differentially expressed genes (FDR < 0.05). The log2 of the expression levels of these genes are shown as boxplots combining mild and severe samples. The genes on the left side of the panel have higher mean expression during the acute visit and those on the right side of the panel have higher expression levels in the follow-up visit (D,E) The log2 expression levels for EZH1 comparing (D) data from infants with severe versus mild RSV infection and (E) data from the acute and follow-up visit. The reported FDR p-values are based on limma analysis. (F,G) Abundance profiles of nasal microbiota comparing groups of mild and severe infants at each of the two visits, including (F) observed diversity and (G) Shannon diversity index.
Figure 3
Figure 3
(A) Principal component analysis was performed on Visit 1 samples for which we had both transcriptome and microbiome data to reduce the transcriptome into gPCs (top) and the microbiome into cPCs (bottom). (B) Cumulative distribution of the amount of variance explained by the top gPCs and cPCs. The vertical and horizontal lines indicate the number of PCs which explain 95% of variance and which were included in our network analysis. (C) The coordinates of the gene and clade principal components across the 40 analyzed samples from Visit 1. (D) Spearman correlation across the loadings of the top 13 gPCs and the top 10 cPCs.
Figure 4
Figure 4
(A) The weights of edges predicted for each of the 40 sample-specific networks obtained from the LIONESS analysis. Columns are grouped based on each infant’s infection severity, sex, and race. Rows are sorted based on the significance of edges. (B) The significance of each edge as defined from multivariate linear analysis comparing edge-weights between two groups, mild and severe, and corrected for sex and race. The top significant edges with nominal p-value less than 0.05 are labeled.
Figure 5
Figure 5
GO term enrichment analysis for the top 100 genes based on the loadings of (A) gPC1, (B) gPC8, and (C) gPC3. These bubble plots include the 25 most significant GO terms identified in each analysis based on the FDR significance. All terms shown are significant at an FDR < 0.05. Size of each bubble indicates the number of genes annotated to the respective GO term and the color indicates the percentage of the top 100 genes annotated to that term. The top 25 GO terms enriched in the other identified gPCs are shown in Supplemental Fig. 6A,B and all GO terms enriched at an FDR < 0.05 are shown in Supplemental Fig. 6C–G.

References

    1. Shi T, et al. Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in young children in 2015: a systematic review and modelling study. Lancet. 2017;390:946–958. doi: 10.1016/S0140-6736(17)30938-8.
    1. Karron RA, Black RE. Determining the burden of respiratory syncytial virus disease: the known and the unknown. Lancet. 2017;390:917–918. doi: 10.1016/S0140-6736(17)31476-9.
    1. Svensson C, Berg K, Sigurs N, Trollfors B. Incidence, risk factors and hospital burden in children under five years of age hospitalised with respiratory syncytial virus infections. Acta paediatrica. 2015;104:922–926. doi: 10.1111/apa.13061.
    1. Sonego M, Pellegrin MC, Becker G, Lazzerini M. Risk factors for mortality from acute lower respiratory infections (ALRI) in children under five years of age in low and middle-income countries: a systematic review and meta-analysis of observational studies. PloS one. 2015;10:e0116380. doi: 10.1371/journal.pone.0116380.
    1. Hasegawa K, Tsugawa Y, Brown DF, Mansbach JM, Camargo CA., Jr. Trends in bronchiolitis hospitalizations in the United States, 2000–2009. Pediatrics. 2013;132:28–36. doi: 10.1542/peds.2012-3877.
    1. Shi T, et al. Risk factors for respiratory syncytial virus associated with acute lower respiratory infection in children under five years: Systematic review and meta-analysis. Journal of global health. 2015;5:020416. doi: 10.7189/jogh.05.020416.
    1. Welliver RC, Sr., et al. Fatality rates in published reports of RSV hospitalizations among high-risk and otherwise healthy children. Current medical research and opinion. 2010;26:2175–2181. doi: 10.1185/03007995.2010.505126.
    1. Meissner HC. Viral Bronchiolitis in Children. The New England journal of medicine. 2016;374:62–72. doi: 10.1056/NEJMra1413456.
    1. Blanken MO, et al. Respiratory syncytial virus and recurrent wheeze in healthy preterm infants. The New England journal of medicine. 2013;368:1791–1799. doi: 10.1056/NEJMoa1211917.
    1. Sigurs N, et al. Asthma and allergy patterns over 18 years after severe RSV bronchiolitis in the first year of life. Thorax. 2010;65:1045–1052. doi: 10.1136/thx.2009.121582.
    1. Backman K, Piippo-Savolainen E, Ollikainen H, Koskela H, Korppi M. Adults face increased asthma risk after infant RSV bronchiolitis and reduced respiratory health-related quality of life after RSV pneumonia. Acta paediatrica. 2014;103:850–855. doi: 10.1111/apa.12662.
    1. Mariani TJ, et al. Association of Dynamic Changes in the CD4 T-Cell Transcriptome With Disease Severity During Primary Respiratory Syncytial Virus Infection in Young Infants. The Journal of infectious diseases. 2017;216:1027–1037. doi: 10.1093/infdis/jix400.
    1. Mejias A, et al. Whole blood gene expression profiles to assess pathogenesis and disease severity in infants with respiratory syncytial virus infection. PLoS medicine. 2013;10:e1001549. doi: 10.1371/journal.pmed.1001549.
    1. Fjaerli HO, et al. Whole blood gene expression in infants with respiratory syncytial virus bronchiolitis. BMC infectious diseases. 2006;6:175. doi: 10.1186/1471-2334-6-175.
    1. Christiaansen AF, Knudson CJ, Weiss KA, Varga SM. The CD4 T cell response to respiratory syncytial virus infection. Immunologic research. 2014;59:109–117. doi: 10.1007/s12026-014-8540-1.
    1. Hacking D, Hull J. Respiratory syncytial virus–viral biology and the host response. The Journal of infection. 2002;45:18–24. doi: 10.1053/jinf.2002.1015.
    1. Openshaw PJ, Chiu C. Protective and dysregulated T cell immunity in RSV infection. Current opinion in virology. 2013;3:468–474. doi: 10.1016/j.coviro.2013.05.005.
    1. Raiden S, et al. Respiratory Syncytial Virus (RSV) Infects CD4+ T Cells: Frequency of Circulating CD4+ RSV+ T Cells as a Marker of Disease Severity in Young Children. The Journal of infectious diseases. 2017;215:1049–1058. doi: 10.1093/infdis/jix070.
    1. Bennett, J. E., Dolin, R. & Blaser, M. J. Infectious Disease Essentials. (2017).
    1. Stewart CJ, et al. Associations of Nasopharyngeal Metabolome and Microbiome with Severity among Infants with Bronchiolitis. A Multiomic Analysis. American journal of respiratory and critical care medicine. 2017;196:882–891. doi: 10.1164/rccm.201701-0071OC.
    1. Teo SM, et al. The infant nasopharyngeal microbiome impacts severity of lower respiratory infection and risk of asthma development. Cell host & microbe. 2015;17:704–715. doi: 10.1016/j.chom.2015.03.008.
    1. de Steenhuijsen Piters WA, et al. Nasopharyngeal Microbiota, Host Transcriptome, and Disease Severity in Children with Respiratory Syncytial Virus Infection. American journal of respiratory and critical care medicine. 2016;194:1104–1115. doi: 10.1164/rccm.201602-0220OC.
    1. Silverman EK, Loscalzo J. Network medicine approaches to the genetics of complex diseases. Discovery medicine. 2012;14:143–152.
    1. Fodha I, et al. Respiratory syncytial virus infections in hospitalized infants: association between viral load, virus subgroup, and disease severity. J Med Virol. 2007;79:1951–1958. doi: 10.1002/jmv.21026.
    1. Garcia-Maurino C, et al. Viral Load Dynamics and Clinical Disease Severity in Infants With Respiratory Syncytial Virus Infection. J Infect Dis. 2019;219:1207–1215. doi: 10.1093/infdis/jiy655.
    1. Walsh EE, et al. Virus-Specific Antibody, Viral Load, and Disease Severity in Respiratory Syncytial Virus Infection. J Infect Dis. 2018;218:208–217. doi: 10.1093/infdis/jiy106.
    1. Zhang W, et al. Human respiratory syncytial virus infection is inhibited by IFN-induced transmembrane proteins. J Gen Virol. 2015;96:170–182. doi: 10.1099/vir.0.066555-0.
    1. Smith, S. E. et al. Interferon-induced Transmembrane Protein 1 restricts replication of virus that enter cells via the plasma membrane. J Virol, 10.1128/JVI.02003-18 (2018).
    1. Ioannidis I, et al. Plasticity and virus specificity of the airway epithelial cell immune response during respiratory virus infection. J Virol. 2012;86:5422–5436. doi: 10.1128/JVI.06757-11.
    1. Goswami R, et al. Viral degradasome hijacks mitochondria to suppress innate immunity. Cell Res. 2013;23:1025–1042. doi: 10.1038/cr.2013.98.
    1. Bakre A, et al. Respiratory syncytial virus modifies microRNAs regulating host genes that affect virus replication. J Gen Virol. 2012;93:2346–2356. doi: 10.1099/vir.0.044255-0.
    1. Ampuero S, et al. Time-course of transcriptome response to respiratory syncytial virus infection in lung epithelium cells. Acta Virol. 2018;62:310–325. doi: 10.4149/av_2018_225.
    1. Gonzalez-Sanz R, et al. ISG15 Is Upregulated in Respiratory Syncytial Virus Infection and Reduces Virus Growth through Protein ISGylation. J Virol. 2016;90:3428–3438. doi: 10.1128/JVI.02695-15.
    1. Reed JL, et al. Innate immune signals modulate antiviral and polyreactive antibody responses during severe respiratory syncytial virus infection. J Infect Dis. 2009;199:1128–1138. doi: 10.1086/597386.
    1. Russell CD, Unger SA, Walton M, Schwarze J. The Human Immune Response to Respiratory Syncytial Virus Infection. Clin Microbiol Rev. 2017;30:481–502. doi: 10.1128/CMR.00090-16.
    1. Panapasa JA, Cox RJ, Mohn KG, Aqrawi LA, Brokstad KA. The expression of B & T cell activation markers in children’s tonsils following live attenuated influenza vaccine. Hum Vaccin Immunother. 2015;11:1663–1672. doi: 10.1080/21645515.2015.1032486.
    1. Li X, et al. Mucolipin 1 positively regulates TLR7 responses in dendritic cells by facilitating RNA transportation to lysosomes. Int Immunol. 2015;27:83–94. doi: 10.1093/intimm/dxu086.
    1. Rinkenberger, N. & Schoggins, J. W. Mucolipin-2 Cation Channel Increases Trafficking Efficiency of Endocytosed Viruses. MBio9, 10.1128/mBio.02314-17 (2018).
    1. Zhang X, et al. MCOLN1 is a ROS sensor in lysosomes that regulates autophagy. Nat Commun. 2016;7:12109. doi: 10.1038/ncomms12109.
    1. Gardinassi LG. A Cross-Study Biomarker Signature of Human Bronchial Epithelial Cells Infected with Respiratory Syncytial Virus. Advances in virology. 2016;2016:3605302. doi: 10.1155/2016/3605302.
    1. Alexa A, Rahnenfuhrer J, Lengauer T. Improved scoring of functional groups from gene expression data by decorrelating GO graph structure. Bioinformatics. 2006;22:1600–1607. doi: 10.1093/bioinformatics/btl140.
    1. Österreicher F, Vajda I. {A new class of metric divergences on probability spaces and its applicability in statistics} Annals of the Institute of Statistical Mathematics. 2003;55:639–653. doi: 10.1007/BF02517812.
    1. Lozupone C, Knight R. UniFrac: a new phylogenetic method for comparing microbial communities. Applied and environmental microbiology. 2005;71:8228–8235. doi: 10.1128/AEM.71.12.8228-8235.2005.
    1. Koren O, et al. A guide to enterotypes across the human body: meta-analysis of microbial community structures in human microbiome datasets. PLoS computational biology. 2013;9:e1002863. doi: 10.1371/journal.pcbi.1002863.
    1. Morgan XC, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome biology. 2012;13:R79. doi: 10.1186/gb-2012-13-9-r79.
    1. Morgan XC, Huttenhower C. Chapter 12: Human microbiome analysis. PLoS computational biology. 2012;8:e1002808. doi: 10.1371/journal.pcbi.1002808.
    1. Morgan XC, et al. Associations between host gene expression, the mucosal microbiome, and clinical outcome in the pelvic pouch of patients with inflammatory bowel disease. Genome biology. 2015;16:67. doi: 10.1186/s13059-015-0637-x.
    1. Chen C-Y, et al. Sexual dimorphism in gene expression and regulatory networks across human tissues. bioRxiv. 2016 doi: 10.1101/082289.
    1. Kuijjer ML, Tung MG, Yuan G, Quackenbush J, Glass K. Estimating Sample-Specific Regulatory Networks. iScience. 2019;14:226–240. doi: 10.1016/j.isci.2019.03.021.
    1. Inchley CS, Osterholt HC, Sonerud T, Fjaerli HO, Nakstad B. Downregulation of IL7R, CCR7, and TLR4 in the cord blood of children with respiratory syncytial virus disease. The Journal of infectious diseases. 2013;208:1431–1435. doi: 10.1093/infdis/jit336.
    1. McNamara PS, Smyth RL. The pathogenesis of respiratory syncytial virus disease in childhood. Br Med Bull. 2002;61:13–28. doi: 10.1093/bmb/61.1.13.
    1. Wang SZ, et al. Shedding of L-selectin and PECAM-1 and upregulation of Mac-1 and ICAM-1 on neutrophils in RSV bronchiolitis. Am J Physiol. 1998;275:L983–989.
    1. Hishiki H, et al. Incidence of bacterial coinfection with respiratory syncytial virus bronchopulmonary infection in pediatric inpatients. Journal of infection and chemotherapy: official journal of the Japan Society of Chemotherapy. 2011;17:87–90. doi: 10.1007/s10156-010-0097-x.
    1. Thorburn K, Harigopal S, Reddy V, Taylor N, van Saene HK. High incidence of pulmonary bacterial co-infection in children with severe respiratory syncytial virus (RSV) bronchiolitis. Thorax. 2006;61:611–615. doi: 10.1136/thx.2005.048397.
    1. Resch B, Gusenleitner W, Mueller WD. Risk of concurrent bacterial infection in preterm infants hospitalized due to respiratory syncytial virus infection. Acta paediatrica. 2007;96:495–498. doi: 10.1111/j.1651-2227.2007.00226.x.
    1. Durbin JE, et al. The role of IFN in respiratory syncytial virus pathogenesis. Journal of immunology. 2002;168:2944–2952. doi: 10.4049/jimmunol.168.6.2944.
    1. Diamond MS, Farzan M. The broad-spectrum antiviral functions of IFIT and IFITM proteins. Nature reviews. Immunology. 2013;13:46–57. doi: 10.1038/nri3344.
    1. Huang IC, et al. Distinct patterns of IFITM-mediated restriction of filoviruses, SARS coronavirus, and influenza A virus. PLoS pathogens. 2011;7:e1001258. doi: 10.1371/journal.ppat.1001258.
    1. Brass AL, et al. The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and dengue virus. Cell. 2009;139:1243–1254. doi: 10.1016/j.cell.2009.12.017.
    1. Anand SK, Tikoo SK. Viruses as modulators of mitochondrial functions. Adv Virol. 2013;2013:738794. doi: 10.1155/2013/738794.
    1. Ashton-Rickardt PG. Mitochondria Apply the Brake to Viral Immunity. Cell Metab. 2016;23:967–968. doi: 10.1016/j.cmet.2016.05.018.
    1. Garaude J, et al. Mitochondrial respiratory-chain adaptations in macrophages contribute to antibacterial host defense. Nat Immunol. 2016;17:1037–1045. doi: 10.1038/ni.3509.
    1. Weinberg SE, Sena LA, Chandel NS. Mitochondria in the regulation of innate and adaptive immunity. Immunity. 2015;42:406–417. doi: 10.1016/j.immuni.2015.02.002.
    1. Franco-Obregon, A. & Gilbert, J. A. The Microbiome-Mitochondrion Connection: Common Ancestries, Common Mechanisms, Common Goals. mSystems2, 10.1128/mSystems.00018-17 (2017).
    1. Gruber J, Kennedy BK. Microbiome and Longevity: Gut Microbes Send Signals to Host Mitochondria. Cell. 2017;169:1168–1169. doi: 10.1016/j.cell.2017.05.048.
    1. Ichinohe T, et al. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci USA. 2011;108:5354–5359. doi: 10.1073/pnas.1019378108.
    1. Caserta MT, et al. Development of a Global Respiratory Severity Score for Respiratory Syncytial Virus Infection in Infants. J Infect Dis. 2017;215:750–756. doi: 10.1093/infdis/jiw624.
    1. Misra RS, et al. Flow-based sorting of neonatal lymphocyte populations for transcriptomics analysis. Journal of immunological methods. 2016;437:13–20. doi: 10.1016/j.jim.2016.07.001.
    1. Chu CY, et al. The Healthy Infant Nasal Transcriptome: A Benchmark Study. Scientific reports. 2016;6:33994. doi: 10.1038/srep33994.
    1. Huber W, et al. Orchestrating high-throughput genomic analysis with Bioconductor. Nature methods. 2015;12:115–121. doi: 10.1038/nmeth.3252.
    1. Ritchie ME, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic acids research. 2015;43:e47. doi: 10.1093/nar/gkv007.
    1. Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics. 2007;8:118–127. doi: 10.1093/biostatistics/kxj037.
    1. Leek JT, Storey JD. A general framework for multiple testing dependence. Proc Natl Acad Sci USA. 2008;105:18718–18723. doi: 10.1073/pnas.0808709105.
    1. Leek JT, Storey JD. Capturing heterogeneity in gene expression studies by surrogate variable analysis. PLoS Genet. 2007;3:1724–1735. doi: 10.1371/journal.pgen.0030161.
    1. Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28:882–883. doi: 10.1093/bioinformatics/bts034.
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nature methods. 2010;7:335–336. doi: 10.1038/nmeth.f.303.
    1. Gibbons SM, Duvallet C, Alm EJ. Correcting for batch effects in case-control microbiome studies. PLoS Comput Biol. 2018;14:e1006102. doi: 10.1371/journal.pcbi.1006102.

Source: PubMed

Подписаться