Microbiome-Metabolomics Analysis of the Impacts of Cryptosporidium muris Infection in BALB/C Mice

ABSTRACT Cryptosporidium is a leading cause of diarrheal disease and mortality in young children worldwide. Cryptosporidium invades small intestinal epithelial cells and forms a unique intracellular niche, a process that may alter gut microbes and their production metabolites. However, the mechanism...

Full description

Bibliographic Details
Main Authors: Luyang Wang, Letian Cao, Yankai Chang, Yin Fu, Yuexin Wang, Kaihui Zhang, Sumei Zhang, Longxian Zhang
Format: Article
Language:English
Published: American Society for Microbiology 2023-02-01
Series:Microbiology Spectrum
Subjects:
Online Access:https://journals.asm.org/doi/10.1128/spectrum.02175-22
_version_ 1811164353657831424
author Luyang Wang
Letian Cao
Yankai Chang
Yin Fu
Yuexin Wang
Kaihui Zhang
Sumei Zhang
Longxian Zhang
author_facet Luyang Wang
Letian Cao
Yankai Chang
Yin Fu
Yuexin Wang
Kaihui Zhang
Sumei Zhang
Longxian Zhang
author_sort Luyang Wang
collection DOAJ
description ABSTRACT Cryptosporidium is a leading cause of diarrheal disease and mortality in young children worldwide. Cryptosporidium invades small intestinal epithelial cells and forms a unique intracellular niche, a process that may alter gut microbes and their production metabolites. However, the mechanism of interactions between gut microbes, metabolites, and parasites is poorly understood. Here, we first characterized the impacts of Cryptosporidium infection on gut microbiota using a microbiome-to-metabolome association study. BALB/c mice were gavaged with Cryptosporidium muris, and fecal samples were collected at 0, 7, 14, 21, and 28 days postinfection (dpi) to observe changes in the intestinal microbes of the body during parasite infection. The infected group had a significantly increased relative abundance of bacterial taxa, such as Lachnospiraceae and Prevotella (P < 0.05), associated with the biosynthesis of short-chain fatty acids (SCFAs). Metabolites related to the metabolic pathways, steroid hormone biosynthesis, and biosynthesis of unsaturated fatty acids pathway were upregulated at 7 dpi, indicating that related metabolites in the biosynthesis of unsaturated fatty acids may be essential for C. muris reproduction in vivo. The metabolites involved in metabolic pathways, bile secretion, and primary bile acid biosynthesis were upregulated at 14 dpi, and we speculate that these metabolites may be critical to the growth and development of Cryptosporidium oocysts in the host. Correlation analysis revealed that Firmicutes bacteria are significantly associated with α-linolenic acid metabolism pathways (P< 0.05). The gut microbiota changes dynamically, and the metabolites involved in fatty acid and bile acid biosynthesis may play important roles during cryptosporidiosis. Details of the gut microbiota and the metabolome after infection with Cryptosporidium may aid in the discovery of specific diagnostic markers and help us understand the changes in parasite metabolic pathways. IMPORTANCE Cryptosporidiosis is a gastrointestinal disease in humans and animals caused by the protozoan parasite Cryptosporidium. Control and treatment of the disease is challenging due to the lack of sensitive diagnostic tools and effective chemotherapy. The dynamics of gut microbiota and metabolites during Cryptosporidium infection may be the key to finding drugs and targets for parasite infection control. Our results indicate that C. muris infection can disrupt gut microbiota and metabolites, resulting in decreased bacterial abundance at the parasitic site. Unsaturated fatty acid pathway biosynthesis-related metabolites are significantly elevated at the patent period. Interestingly, the metabolite pathway that significantly elevated during peak parasite growth was bile acid, the metabolites of which may be important for the circulation of infection of Cryptosporidium oocysts in the host. The enhancing effects of short-chain fatty acid and bile acid metabolism on the growth and development of Cryptosporidium proposed in this study may provide a theoretical basis for future research on novel drugs and vaccines against this intestinal parasite.
first_indexed 2024-04-10T15:20:16Z
format Article
id doaj.art-409bbef74ac44b47a409f22ce5bba692
institution Directory Open Access Journal
issn 2165-0497
language English
last_indexed 2024-04-10T15:20:16Z
publishDate 2023-02-01
publisher American Society for Microbiology
record_format Article
series Microbiology Spectrum
spelling doaj.art-409bbef74ac44b47a409f22ce5bba6922023-02-14T14:15:50ZengAmerican Society for MicrobiologyMicrobiology Spectrum2165-04972023-02-0111110.1128/spectrum.02175-22Microbiome-Metabolomics Analysis of the Impacts of Cryptosporidium muris Infection in BALB/C MiceLuyang Wang0Letian Cao1Yankai Chang2Yin Fu3Yuexin Wang4Kaihui Zhang5Sumei Zhang6Longxian Zhang7College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, ChinaCollege of Veterinary Medicine, Henan Agricultural University, Zhengzhou, ChinaCollege of Veterinary Medicine, Henan Agricultural University, Zhengzhou, ChinaCollege of Veterinary Medicine, Henan Agricultural University, Zhengzhou, ChinaCollege of Veterinary Medicine, Henan Agricultural University, Zhengzhou, ChinaCollege of Veterinary Medicine, Henan Agricultural University, Zhengzhou, ChinaCollege of Veterinary Medicine, Henan Agricultural University, Zhengzhou, ChinaCollege of Veterinary Medicine, Henan Agricultural University, Zhengzhou, ChinaABSTRACT Cryptosporidium is a leading cause of diarrheal disease and mortality in young children worldwide. Cryptosporidium invades small intestinal epithelial cells and forms a unique intracellular niche, a process that may alter gut microbes and their production metabolites. However, the mechanism of interactions between gut microbes, metabolites, and parasites is poorly understood. Here, we first characterized the impacts of Cryptosporidium infection on gut microbiota using a microbiome-to-metabolome association study. BALB/c mice were gavaged with Cryptosporidium muris, and fecal samples were collected at 0, 7, 14, 21, and 28 days postinfection (dpi) to observe changes in the intestinal microbes of the body during parasite infection. The infected group had a significantly increased relative abundance of bacterial taxa, such as Lachnospiraceae and Prevotella (P < 0.05), associated with the biosynthesis of short-chain fatty acids (SCFAs). Metabolites related to the metabolic pathways, steroid hormone biosynthesis, and biosynthesis of unsaturated fatty acids pathway were upregulated at 7 dpi, indicating that related metabolites in the biosynthesis of unsaturated fatty acids may be essential for C. muris reproduction in vivo. The metabolites involved in metabolic pathways, bile secretion, and primary bile acid biosynthesis were upregulated at 14 dpi, and we speculate that these metabolites may be critical to the growth and development of Cryptosporidium oocysts in the host. Correlation analysis revealed that Firmicutes bacteria are significantly associated with α-linolenic acid metabolism pathways (P< 0.05). The gut microbiota changes dynamically, and the metabolites involved in fatty acid and bile acid biosynthesis may play important roles during cryptosporidiosis. Details of the gut microbiota and the metabolome after infection with Cryptosporidium may aid in the discovery of specific diagnostic markers and help us understand the changes in parasite metabolic pathways. IMPORTANCE Cryptosporidiosis is a gastrointestinal disease in humans and animals caused by the protozoan parasite Cryptosporidium. Control and treatment of the disease is challenging due to the lack of sensitive diagnostic tools and effective chemotherapy. The dynamics of gut microbiota and metabolites during Cryptosporidium infection may be the key to finding drugs and targets for parasite infection control. Our results indicate that C. muris infection can disrupt gut microbiota and metabolites, resulting in decreased bacterial abundance at the parasitic site. Unsaturated fatty acid pathway biosynthesis-related metabolites are significantly elevated at the patent period. Interestingly, the metabolite pathway that significantly elevated during peak parasite growth was bile acid, the metabolites of which may be important for the circulation of infection of Cryptosporidium oocysts in the host. The enhancing effects of short-chain fatty acid and bile acid metabolism on the growth and development of Cryptosporidium proposed in this study may provide a theoretical basis for future research on novel drugs and vaccines against this intestinal parasite.https://journals.asm.org/doi/10.1128/spectrum.02175-22Cryptosporidiumgut microbiotametabolitesshort-chain fatty acidbile acid
spellingShingle Luyang Wang
Letian Cao
Yankai Chang
Yin Fu
Yuexin Wang
Kaihui Zhang
Sumei Zhang
Longxian Zhang
Microbiome-Metabolomics Analysis of the Impacts of Cryptosporidium muris Infection in BALB/C Mice
Microbiology Spectrum
Cryptosporidium
gut microbiota
metabolites
short-chain fatty acid
bile acid
title Microbiome-Metabolomics Analysis of the Impacts of Cryptosporidium muris Infection in BALB/C Mice
title_full Microbiome-Metabolomics Analysis of the Impacts of Cryptosporidium muris Infection in BALB/C Mice
title_fullStr Microbiome-Metabolomics Analysis of the Impacts of Cryptosporidium muris Infection in BALB/C Mice
title_full_unstemmed Microbiome-Metabolomics Analysis of the Impacts of Cryptosporidium muris Infection in BALB/C Mice
title_short Microbiome-Metabolomics Analysis of the Impacts of Cryptosporidium muris Infection in BALB/C Mice
title_sort microbiome metabolomics analysis of the impacts of cryptosporidium muris infection in balb c mice
topic Cryptosporidium
gut microbiota
metabolites
short-chain fatty acid
bile acid
url https://journals.asm.org/doi/10.1128/spectrum.02175-22
work_keys_str_mv AT luyangwang microbiomemetabolomicsanalysisoftheimpactsofcryptosporidiummurisinfectioninbalbcmice
AT letiancao microbiomemetabolomicsanalysisoftheimpactsofcryptosporidiummurisinfectioninbalbcmice
AT yankaichang microbiomemetabolomicsanalysisoftheimpactsofcryptosporidiummurisinfectioninbalbcmice
AT yinfu microbiomemetabolomicsanalysisoftheimpactsofcryptosporidiummurisinfectioninbalbcmice
AT yuexinwang microbiomemetabolomicsanalysisoftheimpactsofcryptosporidiummurisinfectioninbalbcmice
AT kaihuizhang microbiomemetabolomicsanalysisoftheimpactsofcryptosporidiummurisinfectioninbalbcmice
AT sumeizhang microbiomemetabolomicsanalysisoftheimpactsofcryptosporidiummurisinfectioninbalbcmice
AT longxianzhang microbiomemetabolomicsanalysisoftheimpactsofcryptosporidiummurisinfectioninbalbcmice