A shotgun metagenomic analysis of the fecal microbiome in humans infected with Giardia duodenalis

Abstract Background The mechanisms underlying the clinical outcome disparity during human infection with Giardia duodenalis are still unclear. In recent years, evidence has pointed to the roles of host factors as well as parasite’s genetic heterogeneity as major contributing factors in the developme...

Full description

Bibliographic Details
Main Authors: Brett A. McGregor, Elham Razmjou, Hossein Hooshyar, Drew R. Seeger, Svetlana A. Golovko, Mikhail Y. Golovko, Steven M. Singer, Junguk Hur, Shahram Solaymani-Mohammadi
Format: Article
Language:English
Published: BMC 2023-07-01
Series:Parasites & Vectors
Subjects:
Online Access:https://doi.org/10.1186/s13071-023-05821-1
_version_ 1797774363823439872
author Brett A. McGregor
Elham Razmjou
Hossein Hooshyar
Drew R. Seeger
Svetlana A. Golovko
Mikhail Y. Golovko
Steven M. Singer
Junguk Hur
Shahram Solaymani-Mohammadi
author_facet Brett A. McGregor
Elham Razmjou
Hossein Hooshyar
Drew R. Seeger
Svetlana A. Golovko
Mikhail Y. Golovko
Steven M. Singer
Junguk Hur
Shahram Solaymani-Mohammadi
author_sort Brett A. McGregor
collection DOAJ
description Abstract Background The mechanisms underlying the clinical outcome disparity during human infection with Giardia duodenalis are still unclear. In recent years, evidence has pointed to the roles of host factors as well as parasite’s genetic heterogeneity as major contributing factors in the development of symptomatic human giardiasis. However, it remains contested as to how only a small fraction of individuals infected with G. duodenalis develop clinical gastrointestinal manifestations, whereas the majority of infected individuals remain asymptomatic. Here, we demonstrate that diversity in the fecal microbiome correlates with the clinical outcome of human giardiasis. Methods The genetic heterogeneity of G. duodenalis clinical isolates from human subjects with asymptomatic and symptomatic giardiasis was determined using a multilocus analysis approach. We also assessed the genetic proximity of G. duodenalis isolates by constructing phylogenetic trees using the maximum likelihood. Total genomic DNA (gDNA) from fecal specimens was utilized to construct DNA libraries, followed by performing paired-end sequencing using the HiSeq X platform. The Kraken2-generated, filtered FASTQ files were assigned to microbial metabolic pathways and functions using HUMAnN 3.04 and the UniRef90 diamond annotated full reference database (version 201901b). Results from HUMAnN for each sample were evaluated for differences among the biological groups using the Kruskal–Wallis non-parametric test with a post hoc Dunn test. Results We found that a total of 8/11 (72.73%) human subjects were infected with assemblage A (sub-assemblage AII) of G. duodenalis, whereas 3/11 (27.27%) human subjects in the current study were infected with assemblage B of the parasite. We also found that the parasite’s genetic diversity was not associated with the clinical outcome of the infection. Further phylogenetic analysis based on the tpi and gdh loci indicated that those clinical isolates belonging to assemblage A of G. duodenalis subjects clustered compactly together in a monophyletic clade despite being isolated from human subjects with asymptomatic and symptomatic human giardiasis. Using a metagenomic shotgun sequencing approach, we observed that infected individuals with asymptomatic and symptomatic giardiasis represented distinctive microbial diversity profiles, and that both were distinguishable from the profiles of healthy volunteers. Conclusions These findings identify a potential association between host microbiome disparity with the development of clinical disease during human giardiasis, and may provide insights into the mechanisms by which the parasite induces pathological changes in the gut. These observations may also lead to the development of novel selective therapeutic targets for preventing human enteric microbial infections. Graphical Abstract
first_indexed 2024-03-12T22:19:57Z
format Article
id doaj.art-95b79fddc8524a69b6e268fa44451f35
institution Directory Open Access Journal
issn 1756-3305
language English
last_indexed 2024-03-12T22:19:57Z
publishDate 2023-07-01
publisher BMC
record_format Article
series Parasites & Vectors
spelling doaj.art-95b79fddc8524a69b6e268fa44451f352023-07-23T11:08:02ZengBMCParasites & Vectors1756-33052023-07-0116111910.1186/s13071-023-05821-1A shotgun metagenomic analysis of the fecal microbiome in humans infected with Giardia duodenalisBrett A. McGregor0Elham Razmjou1Hossein Hooshyar2Drew R. Seeger3Svetlana A. Golovko4Mikhail Y. Golovko5Steven M. Singer6Junguk Hur7Shahram Solaymani-Mohammadi8Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North DakotaDepartment of Parasitology and Mycology, School of Medicine, Iran University of Medical SciencesDepartment of Medical Parasitology and Mycology, School of Medicine, Kashan University of Medical SciencesDepartment of Biomedical Sciences, School of Medicine and Health Sciences, University of North DakotaDepartment of Biomedical Sciences, School of Medicine and Health Sciences, University of North DakotaDepartment of Biomedical Sciences, School of Medicine and Health Sciences, University of North DakotaDepartment of Biology, Georgetown UniversityDepartment of Biomedical Sciences, School of Medicine and Health Sciences, University of North DakotaDepartment of Biomedical Sciences, School of Medicine and Health Sciences, University of North DakotaAbstract Background The mechanisms underlying the clinical outcome disparity during human infection with Giardia duodenalis are still unclear. In recent years, evidence has pointed to the roles of host factors as well as parasite’s genetic heterogeneity as major contributing factors in the development of symptomatic human giardiasis. However, it remains contested as to how only a small fraction of individuals infected with G. duodenalis develop clinical gastrointestinal manifestations, whereas the majority of infected individuals remain asymptomatic. Here, we demonstrate that diversity in the fecal microbiome correlates with the clinical outcome of human giardiasis. Methods The genetic heterogeneity of G. duodenalis clinical isolates from human subjects with asymptomatic and symptomatic giardiasis was determined using a multilocus analysis approach. We also assessed the genetic proximity of G. duodenalis isolates by constructing phylogenetic trees using the maximum likelihood. Total genomic DNA (gDNA) from fecal specimens was utilized to construct DNA libraries, followed by performing paired-end sequencing using the HiSeq X platform. The Kraken2-generated, filtered FASTQ files were assigned to microbial metabolic pathways and functions using HUMAnN 3.04 and the UniRef90 diamond annotated full reference database (version 201901b). Results from HUMAnN for each sample were evaluated for differences among the biological groups using the Kruskal–Wallis non-parametric test with a post hoc Dunn test. Results We found that a total of 8/11 (72.73%) human subjects were infected with assemblage A (sub-assemblage AII) of G. duodenalis, whereas 3/11 (27.27%) human subjects in the current study were infected with assemblage B of the parasite. We also found that the parasite’s genetic diversity was not associated with the clinical outcome of the infection. Further phylogenetic analysis based on the tpi and gdh loci indicated that those clinical isolates belonging to assemblage A of G. duodenalis subjects clustered compactly together in a monophyletic clade despite being isolated from human subjects with asymptomatic and symptomatic human giardiasis. Using a metagenomic shotgun sequencing approach, we observed that infected individuals with asymptomatic and symptomatic giardiasis represented distinctive microbial diversity profiles, and that both were distinguishable from the profiles of healthy volunteers. Conclusions These findings identify a potential association between host microbiome disparity with the development of clinical disease during human giardiasis, and may provide insights into the mechanisms by which the parasite induces pathological changes in the gut. These observations may also lead to the development of novel selective therapeutic targets for preventing human enteric microbial infections. Graphical Abstracthttps://doi.org/10.1186/s13071-023-05821-1HumanGiardiasisGutMicrobiomeInfectionMucosal
spellingShingle Brett A. McGregor
Elham Razmjou
Hossein Hooshyar
Drew R. Seeger
Svetlana A. Golovko
Mikhail Y. Golovko
Steven M. Singer
Junguk Hur
Shahram Solaymani-Mohammadi
A shotgun metagenomic analysis of the fecal microbiome in humans infected with Giardia duodenalis
Parasites & Vectors
Human
Giardiasis
Gut
Microbiome
Infection
Mucosal
title A shotgun metagenomic analysis of the fecal microbiome in humans infected with Giardia duodenalis
title_full A shotgun metagenomic analysis of the fecal microbiome in humans infected with Giardia duodenalis
title_fullStr A shotgun metagenomic analysis of the fecal microbiome in humans infected with Giardia duodenalis
title_full_unstemmed A shotgun metagenomic analysis of the fecal microbiome in humans infected with Giardia duodenalis
title_short A shotgun metagenomic analysis of the fecal microbiome in humans infected with Giardia duodenalis
title_sort shotgun metagenomic analysis of the fecal microbiome in humans infected with giardia duodenalis
topic Human
Giardiasis
Gut
Microbiome
Infection
Mucosal
url https://doi.org/10.1186/s13071-023-05821-1
work_keys_str_mv AT brettamcgregor ashotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT elhamrazmjou ashotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT hosseinhooshyar ashotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT drewrseeger ashotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT svetlanaagolovko ashotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT mikhailygolovko ashotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT stevenmsinger ashotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT jungukhur ashotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT shahramsolaymanimohammadi ashotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT brettamcgregor shotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT elhamrazmjou shotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT hosseinhooshyar shotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT drewrseeger shotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT svetlanaagolovko shotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT mikhailygolovko shotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT stevenmsinger shotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT jungukhur shotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis
AT shahramsolaymanimohammadi shotgunmetagenomicanalysisofthefecalmicrobiomeinhumansinfectedwithgiardiaduodenalis