Alterations in Fecal Fungal Microbiome of Patients With COVID-19 During Time of Hospitalization until Discharge

Tao Zuo, Hui Zhan, Fen Zhang, Qin Liu, Eugene Y K Tso, Grace C Y Lui, Nan Chen, Amy Li, Wenqi Lu, Francis K L Chan, Paul K S Chan, Siew C Ng, Tao Zuo, Hui Zhan, Fen Zhang, Qin Liu, Eugene Y K Tso, Grace C Y Lui, Nan Chen, Amy Li, Wenqi Lu, Francis K L Chan, Paul K S Chan, Siew C Ng

Abstract

Background & aims: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects intestinal cells, and might affect the intestinal microbiota. We investigated changes in the fecal fungal microbiomes (mycobiome) of patients with SARS-CoV-2 infection during hospitalization and on recovery.

Methods: We performed deep shotgun metagenomic sequencing analysis of fecal samples from 30 patients with coronavirus disease 2019 (COVID-19) in Hong Kong, from February 5 through May 12, 2020. Fecal samples were collected 2 to 3 times per week from time of hospitalization until discharge. We compared fecal mycobiome compositions of patients with COVID-19 with those from 9 subjects with community-acquired pneumonia and 30 healthy individuals (controls). We assessed fecal mycobiome profiles throughout time of hospitalization until clearance of SARS-CoV-2 from nasopharyngeal samples.

Results: Patients with COVID-19 had significant alterations in their fecal mycobiomes compared with controls, characterized by enrichment of Candia albicans and a highly heterogeneous mycobiome configuration, at time of hospitalization. Although fecal mycobiomes of 22 patients with COVID-19 did not differ significantly from those of controls during times of hospitalization, 8 of 30 patients with COVID-19 had continued significant differences in fecal mycobiome composition, through the last sample collected. The diversity of the fecal mycobiome of the last sample collected from patients with COVID-19 was 2.5-fold higher than that of controls (P < .05). Samples collected at all timepoints from patients with COVID-19 had increased proportions of opportunistic fungal pathogens, Candida albicans, Candida auris, and Aspergillus flavus compared with controls. Two respiratory-associated fungal pathogens, A. flavus and Aspergillus niger, were detected in fecal samples from a subset of patients with COVID-19, even after clearance of SARS-CoV-2 from nasopharyngeal samples and resolution of respiratory symptoms.

Conclusions: In a pilot study, we found heterogeneous configurations of the fecal mycobiome, with enrichment of fungal pathogens from the genera Candida and Aspergillus, during hospitalization of 30 patients with COVID-19 compared with controls. Unstable gut mycobiomes and prolonged dysbiosis persisted in a subset of patients with COVID-19 up to 12 days after nasopharyngeal clearance of SARS-CoV-2. Studies are needed to determine whether alterations in intestinal fungi contribute to or result from SARS-CoV-2 infection, and the effects of these changes in disease progression.

Keywords: Coronovirus; Intestine; Microbe; Yeast.

Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Schematic diagram of stool specimen collection and duration of hospitalization in patients with COVID-19 (n = 30). “CoV” denotes patient with COVID-19. Stool specimens were serially collected for shotgun metagenomics sequencing. “D0” denotes baseline date when the first stool was collected after hospitalization; the following time points starting with “D” represent days since baseline stool collection. “-ve nasopharyngeal/throat swab”: the first negative result for SARS-CoV-2 virus in 2 consecutive negative nasopharyngeal/throat/pooled swab tests, on which patient was then discharged.
Figure 2
Figure 2
Gut mycobiome (fungal community) alterations in patients with COVID-19. (A) Fecal mycobiome alterations in COVID-19, viewed by NMDS (nonmetric multidimensional scaling) plot based on Bray-Curtis dissimilarities. The fecal mycobiome was compared among healthy controls (n = 30), COVID-19 (n = 30), and pneumonia patient controls (n = 9). (B) Interindividual dissimilarities between fecal mycobiomes within each group. The mycobiome dissimilarity was calculated as Bray-Curtis dissimilarity. Between-group comparison was conducted by t test. (C) The relative abundance of Candida albicans in the fecal mycobiome. Between-group comparison was conducted by Wilcoxon rank sum test.
Figure 3
Figure 3
Gut mycobiome (fungal community) alterations in patients with COVID-19 and longitudinal changes during time of hospitalization. Temporal compositional changes in fecal mycobiome with respect to each COVID-19 case were viewed by NMDS (nonmetric multidimensional scaling) plot based upon Bray-Curtis dissimilarities. The aqua cluster denotes the fecal mycobiome cluster of healthy controls. “CoV” denotes patient with COVID-19. “Day0” denotes baseline date when the first stool was collected after hospitalization; the following time points starting with “Day” represents days since baseline stool collection.
Figure 4
Figure 4
Bloom of gut fungi in patients with COVID-19 during time of hospitalization. (A) The diversity of fecal mycobiome (fungi) in patients with COVID-19 over time of hospitalization (plotted as the baseline time point and the last follow-up time point after hospitalization), compared with healthy controls (n = 30) and pneumonia patient controls (n = 9). Between-group comparison was conducted by t test, last follow-up versus baseline comparison for the hospitalized patients with COVID-19 were conducted by paired t test. (B) The richness of fecal mycobiome (fungi) in patients with COVID-19 over time of hospitalization (plotted as the baseline time point and the last follow-up time point after hospitalization), compared with healthy controls (n = 30) and pneumonia patient controls (n = 9). Between-group comparison was conducted by t test, last follow-up versus baseline comparison for the hospitalized patients with COVID-19 were conducted by paired t test. (C) Overrepresented fungal species in feces in patients with COVID-19 during time of hospitalization, compared with healthy controls. LefSE analysis was conducted to identify differential species, only species with LDA effect size > 2 and false discovery rate P < .1 were plotted.
Figure 5
Figure 5
The presence of Aspergillus flavus (A) and Aspergillus niger (B) in patients with COVID-19 over time of hospitalization. “CoV” denotes patient with COVID-19. “Controls” denotes healthy controls (n = 30). “Pneumonia” denotes patient controls with pneumonia (n = 9). “Day0” denotes baseline date when the first stool was collected after hospitalization; the following time points starting with “Day” represent days since baseline stool collection. “throat swab -ve” indicates the first negative result for SARS-CoV-2 virus in 2 consecutive negative nasopharyngeal/throat/pooled swab tests, on which patient was then discharged.
Supplementary Figure 1
Supplementary Figure 1
Temporal changes in the diversity of gut mycobiome (fungal) in COVID-19 patients over time of hospitalization.
Supplementary Figure 2
Supplementary Figure 2
Temporal changes in the relative abundance of Candida albicans in the gut mycobiome of COVID-19 patients over time of hospitalization. (A) Longitudinal changes of C. albicans in each COVID-19 case. (B) Alteration in the relative abundance of C. albicans compared between baseline and the last follow-up; statistical comparison was conducted by paired Wilcoxon rank sum test. (C) The relative abundance of C. albicans across all time points in all patients with COVID-19 during hospitalization, as compared with healthy controls. Statistical comparison was conducted by paired Wilcoxon rank sum test.
Supplementary Figure 3
Supplementary Figure 3
Temporal changes in the relative abundance of Candida auris in the gut mycobiome of COVID-19 patients over time of hospitalization. (A) Longitudinal changes of C. auris in each COVID-19 case. (B) Alteration in the relative abundance of C. auris compared between baseline and the last follow-up; statistical comparison was conducted by paired Wilcoxon rank sum test. (C) The relative abundance of C. auris across all time points in all patients with COVID-19 during hospitalization, as compared with healthy controls. Statistical comparison was conducted by paired Wilcoxon rank sum test.

References

    1. Onder G., Rezza G., Brusaferro S. Case-fatality rate and characteristics of patients dying in relation to COVID-19 in Italy [printed online ahead of print March 23, 2020]. JAMA.
    1. Mao R., Qiu Y., He J.-S. Manifestations and prognosis of gastrointestinal and liver involvement in patients with COVID-19: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol. 2020;5:667–678.
    1. Wölfel R., Corman V.M., Guggemos W. Virological assessment of hospitalized patients with COVID-2019. Nature. 2020;581:465–469.
    1. Xiao F., Tang M., Zheng X. Evidence for gastrointestinal infection of SARS-CoV-2. Gastroenterology. 2020;158:1831–1833.e3.
    1. Zhou J., Li C., Liu X. Infection of bat and human intestinal organoids by SARS-CoV-2. Nat Med. 2020;26:1077–1083.
    1. Zhou P., Liu Z., Chen Y. Bacterial and fungal infections in COVID-19 patients: a matter of concern [published online ahead ofprint April 22, 2020]. Infect Control Hosp Epidemiol.
    1. Zuo T., Zhang F., Lui G.C.Y. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization [published online ahead of print May 19, 2020]. Gastroenterology.
    1. van Tilburg Bernardes E., Pettersen V.K., Gutierrez M.W. Intestinal fungi are causally implicated in microbiome assembly and immune development in mice. Nat Commun. 2020;11:2577.
    1. Zhang I., Pletcher S.D., Goldberg A.N. Fungal microbiota in chronic airway inflammatory disease and emerging relationships with the host immune response. Front Microbiol. 2017;8:2477.
    1. Fan D., Coughlin L.A., Neubauer M.M. Activation of HIF-1α and LL-37 by commensal bacteria inhibits Candida albicans colonization. Nat Med. 2015;21:808.
    1. Richard M.L., Sokol H. The gut mycobiota: insights into analysis, environmental interactions and role in gastrointestinal diseases. Nat Rev Gastroenterol Hepatol. 2019;16:331–345.
    1. Wu J., Liu J., Zhao X. Clinical characteristics of imported cases of COVID-19 in Jiangsu province: a multicenter descriptive study. Clin Infect Dis. 2020;71:706–712.
    1. Bolger A.M., Lohse M., Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30:2114–2120.
    1. LaPierre N., Mangul S., Alser M. MiCoP: microbial community profiling method for detecting viral and fungal organisms in metagenomic samples. BMC Genomics. 2019;20:423.
    1. Segata N., Izard J., Waldron L. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12:R60.
    1. Baba R, Takaoka H, Kamo T, et al. Clinical interpretations and therapeutic significance of isolating aspergillus species from respiratory specimens. A58. clinical studies in fungal infections: American Thoracic Society, 2020:A2117–A2117.
    1. Kosmidis C., Denning D.W. The clinical spectrum of pulmonary aspergillosis. Thorax. 2015;70:270–277.
    1. Zaneveld J.R., McMinds R., Thurber R.V. Stress and stability: applying the Anna Karenina principle to animal microbiomes. Nat Microbiol. 2017;2:1–8.
    1. Zuo T., Lu X.-J., Zhang Y. Gut mucosal virome alterations in ulcerative colitis. Gut. 2019;68:1169–1179.
    1. Masur H., Brooks J.T., Benson C.A. Prevention and treatment of opportunistic infections in HIV-infected adults and adolescents: Updated Guidelines from the Centers for Disease Control and Prevention, National Institutes of Health, and HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis. 2014;58:1308–1311.
    1. Williams B., Landay A., Presti R.M. Microbiome alterations in HIV infection: a review. Cellular microbiology. 2016;18:645–651.
    1. Qin C., Zhou L., Hu Z. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin Infect Dis. 2020;71:762–768.
    1. Diao B., Wang C., Tan Y. Reduction and functional exhaustion of T cells in patients with coronavirus disease 2019 (COVID-19) Front Immunol. 2020;11:827.
    1. Cox M.J., Loman N., Bogaert D. Co-infections: potentially lethal and unexplored in COVID-19. Lancet Microbe. 2020;1:e11.
    1. Zuo T., Wong S.H., Cheung C.P. Gut fungal dysbiosis correlates with reduced efficacy of fecal microbiota transplantation in Clostridium difficile infection. Nat Commun. 2018;9:3663.
    1. Downward J.R.E., Falkowski N.R., Mason K.L. Modulation of post-antibiotic bacterial community reassembly and host response by Candida albicans. Sci Rep. 2013;3:2191.
    1. Sonoyama K., Miki A., Sugita R. Gut colonization by Candida albicans aggravates inflammation in the gut and extra-gut tissues in mice. Med Mycol. 2011;49:237–247.
    1. Kumamoto C.A. Inflammation and gastrointestinal Candida colonization. Curr Opin Microbiol. 2011;14:386–391.
    1. Yang X., Yu Y., Xu J., Liu H. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. 2020;8:475–481.
    1. Lescure F.-X., Bouadma L., Nguyen D. Clinical and virological data of the first cases of COVID-19 in Europe: a case series. Lancet Infect Dis. 2020;20:697–706.
    1. van Arkel A.L., Rijpstra T.A., Belderbos H.N. COVID-19 associated pulmonary aspergillosis. Am J Respir Crit Care Med. 2020;202:132–135.
    1. Alanio A., Dellière S., Fodil S. Prevalence of putative invasive pulmonary aspergillosis in critically ill patients with COVID-19. Lancet Respir Med. 2020;8:e48–e49.
    1. Koehler P., Cornely O.A., Böttiger B.W. COVID-19 associated pulmonary aspergillosis. Mycoses. 2020;63:528–534.
    1. Mortensen E.M., Coley C.M., Singer D.E. Causes of death for patients with community-acquired pneumonia: results from the Pneumonia Patient Outcomes Research Team cohort study. Arch Intern Med. 2002;162:1059–1064.
    1. Nguyen L.D.N., Viscogliosi E., Delhaes L. The lung mycobiome: an emerging field of the human respiratory microbiome. Front Microbiol. 2015;6:89.
    1. Li Z., Lu G., Meng G. Pathogenic fungal infection in the lung. Front Immunol. 2019;10:1524.

Source: PubMed

3
Abonner