1
|
Liu L, Zhao W, Zhang H, Shang Y, Huang W, Cheng Q. Relationship between pediatric asthma and respiratory microbiota, intestinal microbiota: a narrative review. Front Microbiol 2025; 16:1550783. [PMID: 40415934 PMCID: PMC12099452 DOI: 10.3389/fmicb.2025.1550783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/21/2025] [Indexed: 05/27/2025] Open
Abstract
Pediatric asthma is a common chronic airway inflammatory disease that begins in childhood and its impact persists throughout all age stages of patients. With the continuous progress of detection technologies, numerous studies have firmly demonstrated that gut microbiota and respiratory microbiota are closely related to the occurrence and development of asthma, and related research is increasing day by day. This article elaborates in detail on the characteristics, composition of normal gut microbiota and lung microbiota at different ages and in different sites, as well as the connection of the gut-lung axis. Subsequently, it deeply analyzes various factors influencing microbiota colonization, including host factor, delivery mode, maternal dietary and infant feeding patterns, environmental microbial exposure and pollutants, and the use of antibiotics in early life. These factors are highly likely to play a crucial role in the onset process and disease progression of asthma. Research shows that obvious changes have occurred in the respiratory and gut microbiota of asthma patients, and these microbiomes exhibit different characteristics according to the phenotypes and endotypes of asthma. Finally, the article summarizes the microbiota-related treatment approaches for asthma carried out in recent years, including the application of probiotics, nutritional interventions, and fecal microbiota transplantation. These treatment modalities are expected to become new directions for future asthma treatment and bring new hope for solving the problem of childhood asthma.
Collapse
Affiliation(s)
- Lian Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wenqi Zhao
- School of Clinical Medicine, Qilu Medical University, Zibo, China
| | - Han Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wanjie Huang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qi Cheng
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
2
|
Zhao JX, Elsheikha HM, Shang KM, Su JW, Wei YJ, Qin Y, Zhao ZY, Ma H, Zhang XX. Investigation of the genetic diversity of gut mycobiota of the wild and laboratory mice. Microbiol Spectr 2025; 13:e0284024. [PMID: 40162766 PMCID: PMC12054021 DOI: 10.1128/spectrum.02840-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/05/2025] [Indexed: 04/02/2025] Open
Abstract
Mice are colonized by diverse gut fungi, known as the mycobiota, which have received much less attention than bacterial microbiota. Here, we studied the diversities and structures of cecal fungal communities in wild (Lasiopodomys brandtii, Apodemus agrarius, and Microtus fortis) vs laboratory C57BL/6J mice to disentangle the contributions of gut fungi to the adaptation of mice to genetic diversity. Using ITS1 gene sequencing, we obtained 2,912 amplicon sequence variants (ASVs) and characterized the composition and diversity of cecal mycobiota in mice. There were significant differences in the composition of cecal fungal communities between wild and C57BL/6J mice, with more species diversity and richness of fungi in wild mice than C57BL/6J mice. We cultured 428 fungal strains from the cecal mycobiota, sequenced the whole genome of 48 selected strains, and identified 500,849 genes. Functional annotation analysis revealed multiple pathways related to energy metabolism, carbohydrate metabolism, fatty acid metabolism, and enzymes involved in the degradation of polysaccharides, lipids, and proteins, and secondary metabolite biosynthesis. The functions and abundance of Hypocreales and Pleosporales, which included the majority of the crucial metabolic pathways, were significantly higher in wild mice than in C57BL/6J mice. The results suggest that variations in the fungal community composition may relate to the adaptability of mice to their environmental habitats. IMPORTANCE In this study, we analyzed the fungal microbiota of three wild mouse species alongside laboratory mice using ITS1 amplicon sequencing. By integrating whole-genome sequencing with culturomics, we sequenced the genomes of 48 fungi isolated from cultured strains and investigated their biological functions to understand the role of intestinal fungi in the environmental adaptability of wild mice. This investigation has expanded the functional gene repository of gut fungi and shed new light on the intricate interplay between mice and their gut fungal communities. The data offer valuable insight into the ecological adaptation in wild mice, emphasizing the complex and dynamic relationship between the murine hosts and their mycobiota.
Collapse
Affiliation(s)
- Ji-Xin Zhao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Hany M. Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Kai-Meng Shang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Jin-Wen Su
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Yong-Jie Wei
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Ya Qin
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Zi-Yu Zhao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - He Ma
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Xiao-Xuan Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| |
Collapse
|
3
|
Wilson L, Nielsen K, Caspasso-Villanueva S, O'Brien T, Hefner LA, Slick P, Petty L, Dienna R, Castillo H, Chavez A. Characterization of virulence-related phenotypes of Candida parapsilosis and Rhodotorula mucilaginosa isolated from the International Space Station (ISS). LIFE SCIENCES IN SPACE RESEARCH 2025; 45:16-24. [PMID: 40280638 DOI: 10.1016/j.lssr.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 04/29/2025]
Abstract
There is increased interest in characterizing and describing the effects of space stressors on human microflora. This study describes virulence-related phenotypes of two human yeast commensals, Rhodotorula mucilaginosa and Candida parapsilosis, isolated from the International Space Station (ISS). The strains were compared with ATCC control strains to provide insights into adaptation and phenotypic switching of fungal species in spacecraft environments. Strains were grown in media that induce filamentation and capsule production. Antimycotic susceptibility was determined after exposure of liquid cultures to fluconazole, amphotericin B, and caspofungin. Biofilm formation was quantified using the crystal violet assay, and autoinducer (AI) production was detected by activation of a reporter fluorescent gene present in biosensor bacterial strains. In vivo infection studies were conducted using a C. elegans killing model. Results indicated increased filamentation production patterns in ISS Candida parapsilosis and increased capsule production in ISS Rhodotorula. Additionally, there was increased resistance to antifungal activity, biofilm formation, long-chain autoinducer production, and heightened nematode virulence detected in the ISS isolates. These results suggest that space conditions might enhance adaptation and phenotypic plasticity in yeast, leading to increased virulence-related phenotypes.
Collapse
Affiliation(s)
- Lauren Wilson
- Department of Human Factors and Behavioral Neurobiology, College of Arts and Sciences, Embry-Riddle Aeronautical University, 1 Aerospace Blvd, Daytona Beach 32114, FL, USA
| | - Kaitlyn Nielsen
- Department of Human Factors and Behavioral Neurobiology, College of Arts and Sciences, Embry-Riddle Aeronautical University, 1 Aerospace Blvd, Daytona Beach 32114, FL, USA
| | - Stefani Caspasso-Villanueva
- Department of Human Factors and Behavioral Neurobiology, College of Arts and Sciences, Embry-Riddle Aeronautical University, 1 Aerospace Blvd, Daytona Beach 32114, FL, USA
| | - Takara O'Brien
- Department of Human Factors and Behavioral Neurobiology, College of Arts and Sciences, Embry-Riddle Aeronautical University, 1 Aerospace Blvd, Daytona Beach 32114, FL, USA
| | - Lily A Hefner
- Westminster College, 501 Westminster Avenue, Fulton, MO 665251, USA
| | - Paulina Slick
- Department of Integrative Biology, College of Arts and Sciences, University of South Florida, 4202 E Fowler Ave, Tampa, Florida 33620, USA
| | - Logan Petty
- Department of Human Factors and Behavioral Neurobiology, College of Arts and Sciences, Embry-Riddle Aeronautical University, 1 Aerospace Blvd, Daytona Beach 32114, FL, USA
| | - Riley Dienna
- Department of Human Factors and Behavioral Neurobiology, College of Arts and Sciences, Embry-Riddle Aeronautical University, 1 Aerospace Blvd, Daytona Beach 32114, FL, USA
| | - Hugo Castillo
- Department of Human Factors and Behavioral Neurobiology, College of Arts and Sciences, Embry-Riddle Aeronautical University, 1 Aerospace Blvd, Daytona Beach 32114, FL, USA
| | - Alba Chavez
- Department of Human Factors and Behavioral Neurobiology, College of Arts and Sciences, Embry-Riddle Aeronautical University, 1 Aerospace Blvd, Daytona Beach 32114, FL, USA.
| |
Collapse
|
4
|
Costa MADC, da Silva Duarte V, Fraiz GM, Cardoso RR, da Silva A, Martino HSD, Dos Santos D'Almeida CT, Ferreira MSL, Corich V, Hamaker BR, Giacomini A, Bressan J, Barros FARD. Regular Consumption of Black Tea Kombucha Modulates the Gut Microbiota in Individuals with and without Obesity. J Nutr 2025; 155:1331-1349. [PMID: 39732435 DOI: 10.1016/j.tjnut.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/22/2024] [Accepted: 12/18/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Kombucha, a fermented beverage obtained from a Symbiotic Culture of Bacteria and Yeast, has shown potential in modulating gut microbiota, although no clinical trials have been done. OBJECTIVES We aimed to evaluate the effects of regular black tea kombucha consumption on intestinal health in individuals with and without obesity. METHODS A pre-post clinical intervention study was conducted lasting 8 wk. Forty-six participants were allocated into 2 groups: normal weight + black tea kombucha (n = 23); and obese + black tea kombucha (n = 23). Blood, urine, and stool samples were collected at baseline (T0) and after 8 wk of intervention (T8). RESULTS A total of 145 phenolic compounds were identified in the kombucha, primarily flavonoids (81%) and phenolic acids (19%). Kombucha favored commensal bacteria such as Bacteroidota and Akkermanciaceae, especially in the obese group. Subdoligranulum, a butyrate producer, also increased in the obese group after kombucha consumption (P = 0.031). Obesity-associated genera Ruminococcus and Dorea were elevated in the obese group at baseline (P < 0.05) and reduced after kombucha consumption, becoming similar to the normal weight group (Ruminococcus: obese T8 × normal weight T8: P = 0.27; Dorea: obese T8 × normal weight T0: P = 0.57; obese T8 × normal weight T8: P = 0.32). Fungal diversity increased, with a greater abundance of Saccharomyces in both groups and reductions in Exophiala and Rhodotorula, particularly in the obese group. Pichia and Dekkera, key microorganisms in kombucha, were identified as biomarkers after the intervention. CONCLUSIONS Regular kombucha consumption positively influenced gut microbiota in both normal and obese groups, with more pronounced effects in the obese group, suggesting that it may be especially beneficial for those individuals. This trial was registered at Brazilian Clinical Trial Registry - ReBEC as UTN code U1111-1263-9550 (https://ensaiosclinicos.gov.br/rg/RBR-9832wsx).
Collapse
Affiliation(s)
- Mirian Aparecida de Campos Costa
- Bioactive Compounds and Carbohydrates (BIOCARB) Research Group, Department of Food Science and Technology, Universidade Federal de Viçosa, Viçosa, MG, Brazil; Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, United States
| | - Vinícius da Silva Duarte
- Faculty of Chemistry, Biotechnology, and Food Science, The Norwegian University of Life Sciences, Ås, Norway
| | - Gabriela Macedo Fraiz
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG, Brazil; Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, Universidad de Navarra, Pamplona, Spain
| | - Rodrigo Rezende Cardoso
- Bioactive Compounds and Carbohydrates (BIOCARB) Research Group, Department of Food Science and Technology, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - Alessandra da Silva
- Public Health Epidemiology Graduate Program, Environmental and Health Education Laboratory, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, RJ, Brazil
| | | | - Carolina Thomaz Dos Santos D'Almeida
- Laboratory of Bioactives, Food and Nutrition Graduate Program, Federal University of State of Rio de Janeiro - UNIRIO, Rio de Janeiro, RJ, Brazil
| | - Mariana Simões Larraz Ferreira
- Laboratory of Bioactives, Food and Nutrition Graduate Program, Federal University of State of Rio de Janeiro - UNIRIO, Rio de Janeiro, RJ, Brazil
| | - Viviana Corich
- Department of Agronomy, Food Natural Resources, Animals, and Environment, Università degli Studi di Padova, Legnaro, Padova, PD, Italy
| | - Bruce R Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, United States
| | - Alessio Giacomini
- Department of Agronomy, Food Natural Resources, Animals, and Environment, Università degli Studi di Padova, Legnaro, Padova, PD, Italy
| | - Josefina Bressan
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - Frederico Augusto Ribeiro de Barros
- Bioactive Compounds and Carbohydrates (BIOCARB) Research Group, Department of Food Science and Technology, Universidade Federal de Viçosa, Viçosa, MG, Brazil.
| |
Collapse
|
5
|
Issa KD, Othman HO, Amin HIM, Omar SE, Jihad SS, Rasool DD, Ahmed AS, Ghazali MF, Hussain FHS. Sustainable Antimicrobial and Anticancer Agents: Eco-Friendly Synthesis of Copper Nanoparticles Using Biebersteinia multifida DC. Chem Biodivers 2025:e202402612. [PMID: 40262138 DOI: 10.1002/cbdv.202402612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 02/12/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
Biebersteinia multifida DC. is a wild therapeutic plant, traditionally used for various medicinal applications. The aim of the present study is to extract bioactive constituents from the plants' roots and synthesize copper nanoparticles (CuNPs). Ethanolic extraction of the plant's roots yielded 19 bioactive compounds, recognized through gas chromatography-mass spectroscopy (GC-MS), mainly including citraconic anhydride, γ-sitosterol, and 2-furancarboxaldehyde. The prepared CuNPs have been fully characterized. The biological activity evaluations revealed these CuNPs possess acceptable antibacterial and antifungal activities. Furthermore, the CuNPs displayed significant cytotoxic potency toward "4T1 breast cancer cells" while showing a relatively low cell death rate against the normal "HEK-293 kidney cell". In conclusion, our findings showed that the CuNPs can be synthesized from B. multifida roots with a simple, fast, and eco-friendly procedure. These CuNPs are efficient in antimicrobial and anticancer activities.
Collapse
Affiliation(s)
- Kovan Dilawer Issa
- Medical Analysis Department, Applied Science Faculty, Tishk International University, Erbil, Iraq
| | - Hazha Omar Othman
- Department of Chemistry, College of Science, Salahaddin University-Erbil, Erbil, Iraq
- Department of Pharmacy, Tishk International University, Erbil, Iraq
| | - Hawraz Ibrahim M Amin
- Department of Chemistry, College of Science, Salahaddin University-Erbil, Erbil, Iraq
- Department of Medical Biochemical Analysis, Cihan University-Erbil, Erbil, Iraq
| | - Shahnaz Erfan Omar
- Medical Analysis Department, Applied Science Faculty, Tishk International University, Erbil, Iraq
| | - Sarya Siraj Jihad
- Medical Analysis Department, Applied Science Faculty, Tishk International University, Erbil, Iraq
| | - Danya Dler Rasool
- Medical Analysis Department, Applied Science Faculty, Tishk International University, Erbil, Iraq
| | - Abdullah Shahab Ahmed
- Medical Analysis Department, Applied Science Faculty, Tishk International University, Erbil, Iraq
| | - Mardin Firsat Ghazali
- Medical Analysis Department, Applied Science Faculty, Tishk International University, Erbil, Iraq
| | - Faiq H S Hussain
- Medical Analysis Department, Applied Science Faculty, Tishk International University, Erbil, Iraq
| |
Collapse
|
6
|
Ongaga EG, Muli JK, Kamau PK, Budambula NL. Endophytic Microflora of Crotalaria: Their Diversity and Role in Plant Growth Promotion. Curr Microbiol 2025; 82:214. [PMID: 40140094 DOI: 10.1007/s00284-025-04181-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 03/09/2025] [Indexed: 03/28/2025]
Abstract
The endophytic microflora of Crotalaria constitute a heterogeneous community of beneficial microorganisms that colonize healthy tissues of the host plant without causing any apparent harm. The microflora play a crucial role in promoting plant growth, nutrient gain, and resilience to various biotic and abiotic stresses. This review highlights the range of endophytic microorganisms that reside in Crotalaria tissues, providing insights on the methods of detection and the role played by endophytes in promoting host plant growth. Diverse groups of endophytes ranging from bacteria, fungi, and actinomycetes colonize internal organs of Crotalaria species. Key findings indicate that Crotalaria-associated endophytes, including species of Bradyrhizobium, Rhizobium, Burkholderia, and Methylobacterium, exhibit plant growth-promoting traits such as nitrogen fixation, phytohormone production, phosphate solubilization, and resistance to abiotic stresses. Additionally, some endophytes produce metabolites that serve as biocontrol agents, protecting Crotalaria against phytopathogens. This review offers valuable insights for future exploitation of endophytic microflora of Crotalaria in enhancing crop productivity and stress tolerance.
Collapse
Affiliation(s)
- Edinah G Ongaga
- Department of Biological Sciences, University of Embu (UoEm), P.O Box 6-60100, Embu, Kenya
| | - Joshua K Muli
- Department of Biological Sciences, University of Embu (UoEm), P.O Box 6-60100, Embu, Kenya
| | - Peter K Kamau
- Department of Life Sciences, South Eastern Kenya University (SEKU), P.O. Box 170-90200, Kitui, Kenya
| | - Nancy L Budambula
- Department of Biological Sciences, University of Embu (UoEm), P.O Box 6-60100, Embu, Kenya.
| |
Collapse
|
7
|
Sun YY, Liu NN. Mycobiome: an underexplored kingdom in cancer. Microbiol Mol Biol Rev 2025:e0026124. [PMID: 40084887 DOI: 10.1128/mmbr.00261-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025] Open
Abstract
SUMMARYThe human microbiome, including bacteria, fungi, archaea, and viruses, is intimately linked to both health and disease. The relationship between bacteria and disease has received much attention and intensive investigation, while that of the fungal microbiome, also known as mycobiome, has lagged far behind bacteria. There is growing evidence showing mycobiome dysbiosis in cancer patients, and certain cancer-specific fungi may contribute to cancer progression by interacting with both host and bacteria. It was also demonstrated that the role of fungi-derived products in cancer should also not be underestimated. Therefore, investigating how fungal pathogenesis contributes to the onset and spread of cancer would yield crucial information for cancer diagnosis, prevention, and anti-cancer therapy.
Collapse
Affiliation(s)
- Yan-Yan Sun
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning-Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Wronowska E, Guevara-Lora I, Brankiewicz A, Bras G, Zawrotniak M, Satala D, Karkowska-Kuleta J, Budziaszek J, Koziel J, Rapala-Kozik M. Synergistic effects of Candida albicans and Porphyromonas gingivalis biofilms on epithelial barrier function in a 3D aspiration pneumonia model. Front Cell Infect Microbiol 2025; 15:1552395. [PMID: 40125517 PMCID: PMC11925950 DOI: 10.3389/fcimb.2025.1552395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Introduction Polymicrobial infections involving Candida albicans and Porphyromonas gingivalis represent a significant challenge in maintaining epithelial barrier integrity. This study explores their synergistic effects on epithelial cells using an air-liquid interface (ALI) model. Methods Mixed-species biofilms were developed and analyzed for their impact on epithelial permeability and tight junction proteins. The effects of biofilm supernatants on IL-8 secretion and oxidative stress markers were also evaluated. The role of P. gingivalis proteases was assessed using a gingipain-null mutant (ΔKΔRAB) compared to the wild-type strain (W83). Survival experiments were conducted using Galleria mellonella larvae to examine the pathogenicity of dual-species biofilms. Results Mixed-species biofilms significantly increased epithelial permeability and disrupted tight junction proteins, as evidenced by reduced levels of ZO-1 and E-cadherin. These changes were accompanied by oxidative stress, characterized by decreased HO-1 expression and enhanced Bax/Bcl-xL ratios, indicating increased pro-apoptotic activity. Supernatants from dual-species biofilms demonstrated a pronounced effect on epithelial cells, modulating IL-8 secretion and exacerbating oxidative damage. C. albicans was identified as the dominant driver of pro-inflammatory responses, while P. gingivalis contributed through immune modulation and enzymatic activity, primarily via gingipains. The ΔKΔRAB mutant biofilms caused less epithelial disruption and oxidative stress compared to the wild-type, highlighting the critical role of gingipains in pathogenesis. Discussion Survival experiments using Galleria mellonella larvae supported these findings, highlighting the reduced survival associated with dual-species biofilms and the potential for high-dose antimicrobial therapies to mitigate this effect. These results emphasize the cooperative mechanisms of C. albicans and P. gingivalis in compromising epithelial barriers and underline the importance of combination therapies targeting both fungal and bacterial components in polymicrobial infections.
Collapse
Affiliation(s)
- Ewelina Wronowska
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Ibeth Guevara-Lora
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Aleksandra Brankiewicz
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Grazyna Bras
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Marcin Zawrotniak
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Dorota Satala
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| | - Joanna Budziaszek
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Joanna Koziel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
9
|
Yan M, Andersen TO, Pope PB, Yu Z. Probing the eukaryotic microbes of ruminants with a deep-learning classifier and comprehensive protein databases. Genome Res 2025; 35:368-378. [PMID: 39730187 PMCID: PMC11874962 DOI: 10.1101/gr.279825.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024]
Abstract
Metagenomics, particularly genome-resolved metagenomics, have significantly deepened our understanding of microbes, illuminating their taxonomic and functional diversity and roles in ecology, physiology, and evolution. However, eukaryotic populations within various microbiomes, including those in the mammalian gastrointestinal (GI) tract, remain relatively underexplored in metagenomic studies owing to the lack of comprehensive reference genome databases and robust bioinformatic tools. The GI tract of ruminants, particularly the rumen, contains a high eukaryotic biomass but a relatively low diversity of ciliates and fungi, which significantly impacts feed digestion, methane emissions, and rumen microbial ecology. In the present study, we developed GutEuk, a bioinformatics tool that improves upon the currently available Tiara and EukRep in accurately identifying eukaryotic sequences from metagenomes. GutEuk is optimized for high precision across different sequence lengths. It can also distinguish fungal and protozoal sequences, further elucidating their unique ecological, physiological, and nutritional impacts. GutEuk was shown to facilitate comprehensive analyses of protozoa and fungi within more than 1000 rumen metagenomes, revealing a greater genomic diversity among protozoa than previously documented. We further curated several ruminant eukaryotic protein databases, significantly enhancing our ability to distinguish the functional roles of ruminant fungi and protozoa from those of prokaryotes. Overall, the newly developed package GutEuk and its associated databases create new opportunities for the in-depth study of GI tract eukaryotes.
Collapse
Affiliation(s)
- Ming Yan
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Center of Microbiome Science, The Ohio State University, Columbus, Ohio 43210, USA
| | - Thea O Andersen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås NO-7491, Norway
- Faculty of Biosciences, Norwegian University of Life Sciences, Ås NO-7491, Norway
| | - Phillip B Pope
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås NO-7491, Norway
- Faculty of Biosciences, Norwegian University of Life Sciences, Ås NO-7491, Norway
- Centre for Microbiome Research, School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, Woolloongabba 4102, Queensland, Australia
| | - Zhongtang Yu
- Department of Animal Sciences, The Ohio State University, Columbus, Ohio 43210, USA;
- Center of Microbiome Science, The Ohio State University, Columbus, Ohio 43210, USA
| |
Collapse
|
10
|
Pu Y, Zhou X, Cai H, Lou T, Liu C, Kong M, Sun Z, Wang Y, Zhang R, Zhu Y, Ye L, Zheng Y, Zhu B, Quan Z, Zhao G, Zheng Y. Impact of DNA Extraction Methods on Gut Microbiome Profiles: A Comparative Metagenomic Study. PHENOMICS (CHAM, SWITZERLAND) 2025; 5:76-90. [PMID: 40313603 PMCID: PMC12040788 DOI: 10.1007/s43657-025-00232-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 05/03/2025]
Abstract
In gut microbial research, DNA extraction remarkably influences study outcomes and biological interpretations. Rapid advancements in the research scale and technological upgrades necessitate evaluating new methods to ensure reliability and precision in microbial community profiling. We systematically evaluated the performance of eight recent and commonly used extraction methods using a microbial mock community (MMC) and fecal samples from two healthy volunteers, incorporating bacterial, archaeal, and fungal constituents. Performance metrics included nucleic acid assessment, microbial profile assessment, and scalability for large-scale studies, leveraging shotgun metagenomics for in-depth analysis. Despite variations in DNA quantity and quality, all methods yielded sufficient DNA for shotgun metagenomic sequencing. In the MMC microbial profile assessment, the QIAamp PowerFecal pro Kit (PF) and DNeasy PowerSoil HTP kit (PS) methods exhibited higher similarity with the theoretical composition and lower variability across technical replicates compared to other methods. For fecal samples, the extraction method accounted for 21.4% of the overall microbiome variation and significantly affected the abundances of 32% of detected microbial species. Methods using mechanical lysis with small beads, such as PF and PS, demonstrated better efficiency, indicated by increased microbial diversity in extracting DNA from Gram-positive bacteria. Furthermore, the PF and PS methods are notably simple to execute and automation-friendly, though relatively costly. Our study underscores the importance of maintaining consistency in DNA extraction methods for reliable comparative metagenomic analyses. We recommend PF and PS methods as optimal for expansive gut metagenomic research, emphasizing the critical role of mechanical lysis in DNA extraction. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-025-00232-x.
Collapse
Affiliation(s)
- Yanni Pu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Room C601, No. 2005 Songhu Road, Yangpu District, Shanghai, 200438 China
| | - Xiaofeng Zhou
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Room C601, No. 2005 Songhu Road, Yangpu District, Shanghai, 200438 China
| | - Hao Cai
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Room C601, No. 2005 Songhu Road, Yangpu District, Shanghai, 200438 China
| | - Tao Lou
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Room C601, No. 2005 Songhu Road, Yangpu District, Shanghai, 200438 China
| | - Chenglin Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Room C601, No. 2005 Songhu Road, Yangpu District, Shanghai, 200438 China
| | - Mengmeng Kong
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Room C601, No. 2005 Songhu Road, Yangpu District, Shanghai, 200438 China
| | - Zhonghan Sun
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Room C601, No. 2005 Songhu Road, Yangpu District, Shanghai, 200438 China
| | - Yanren Wang
- Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Institute of Biodiversity Science, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Ruyi Zhang
- Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Institute of Biodiversity Science, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Yuxuan Zhu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Room C601, No. 2005 Songhu Road, Yangpu District, Shanghai, 200438 China
| | - Lin Ye
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Room C601, No. 2005 Songhu Road, Yangpu District, Shanghai, 200438 China
| | - Yuanting Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute and Shanghai Cancer Center, Fudan University, Shanghai, 200438 China
| | - Baoli Zhu
- Key Laboratory of Agro-Ecological Processes in Subtropical Regions, Taoyuan Agro-Ecosystem Research Station, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125 China
| | - Zhexue Quan
- Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Institute of Biodiversity Science, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Guoping Zhao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Room C601, No. 2005 Songhu Road, Yangpu District, Shanghai, 200438 China
- Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200032 China
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Room C601, No. 2005 Songhu Road, Yangpu District, Shanghai, 200438 China
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, 200032 China
- Yiwu Research Institute, Fudan University, Yiwu, 322000 China
| |
Collapse
|
11
|
Saeed H, Díaz LA, Gil-Gómez A, Burton J, Bajaj JS, Romero-Gomez M, Arrese M, Arab JP, Khan MQ. Microbiome-centered therapies for the management of metabolic dysfunction-associated steatotic liver disease. Clin Mol Hepatol 2025; 31:S94-S111. [PMID: 39604327 PMCID: PMC11925441 DOI: 10.3350/cmh.2024.0811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a significant global health issue, affecting over 30% of the population worldwide due to the rising prevalence of metabolic risk factors such as obesity and type 2 diabetes mellitus. This spectrum of liver disease ranges from isolated steatosis to more severe forms such as steatohepatitis, fibrosis, and cirrhosis. Recent studies highlight the role of gut microbiota in MASLD pathogenesis, showing that dysbiosis significantly impacts metabolic health and the progression of liver disease. This review critically evaluates current microbiome-centered therapies in MASLD management, including prebiotics, probiotics, synbiotics, fecal microbiota transplantation, and emerging therapies such as engineered bacteria and bacteriophage therapy. We explore the scientific rationale, clinical evidence, and potential mechanisms by which these interventions influence MASLD. The gut-liver axis is crucial in MASLD, with notable changes in microbiome composition linked to disease progression. For instance, specific microbial profiles and reduced alpha diversity are associated with MASLD severity. Therapeutic strategies targeting the microbiome could modulate disease progression by improving gut permeability, reducing endotoxin-producing bacteria, and altering bile acid metabolism. Although promising, these therapies require further research to fully understand their mechanisms and optimize their efficacy. This review integrates findings from clinical trials and experimental studies, providing a comprehensive overview of microbiome-centered therapies' potential in managing MASLD. Future research should focus on personalized strategies, utilizing microbiome features, blood metabolites, and customized dietary interventions to enhance the effectiveness of these therapies.
Collapse
Affiliation(s)
- Huma Saeed
- Division of Infectious Diseases, Department of Medicine, University of Western Ontario, London, ON, Canada
| | - Luis Antonio Díaz
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, CA, USA
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Antonio Gil-Gómez
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Jeremy Burton
- Department of Microbiology & Immunology, Western University, London, ON, Canada
| | - Jasmohan S. Bajaj
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Manuel Romero-Gomez
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- UCM Digestive diseases, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Pablo Arab
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Mohammad Qasim Khan
- Division of Gastroenterology, Department of Medicine, University of Western Ontario, London, ON, Canada
- Department of Epidemiology and Biostatistics, University of Western Ontario, London, ON, Canada
| |
Collapse
|
12
|
Taitz JJ, Tan J, Ni D, Potier-Villette C, Grau G, Nanan R, Macia L. Antibiotic-mediated dysbiosis leads to activation of inflammatory pathways. Front Immunol 2025; 15:1493991. [PMID: 39850904 PMCID: PMC11754057 DOI: 10.3389/fimmu.2024.1493991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/19/2024] [Indexed: 01/25/2025] Open
Abstract
Introduction The gut microbiota plays a pivotal role in influencing host health, through the production of metabolites and other key signalling molecules. While the impact of specific metabolites or taxa on host cells is well-documented, the broader impact of a disrupted microbiota on immune homeostasis is less understood, which is particularly important in the context of the increasing overuse of antibiotics. Methods Female C57BL/6 mice were gavaged twice daily for four weeks with Vancomycin, Polymyxin B, or PBS (control). Caecal microbiota composition was assessed via 16S rRNA sequencing and caecal metabolites were quantified with NMR spectroscopy. Immune profiles of spleen and mesenteric lymph nodes (MLNs) were assessed by flow cytometry, and splenocytes assessed for ex vivo cytokine production. A generalised additive model approach was used to examine the relationship between global antibiotic consumption and IBD incidence. Results Antibiotics significantly altered gut microbiota composition, reducing alpha-diversity. Acetate and butyrate were significantly reduced in antibiotic groups, while propionate and succinate increased in Vancomycin and PmB-treated mice, respectively. The MLNs and spleen showed changes only to DC numbers. Splenocytes from antibiotic-treated mice stimulated ex vivo exhibited increased production of TNF. Epidemiological analysis revealed a positive correlation between global antibiotic consumption and IBD incidence. Discussion Our findings demonstrate that antibiotic-mediated dysbiosis results in significantly altered short-chain fatty acid levels but immune homeostasis in spleen and MLNs at steady state is mostly preserved. Non-specific activation of splenocytes ex vivo, however, revealed mice with perturbed microbiota had significantly elevated production of TNF. Thus, this highlights antibiotic-mediated disruption of the gut microbiota may program the host towards dysregulated immune responses, predisposing to the development of TNF-associated autoimmune or chronic inflammatory disease.
Collapse
Affiliation(s)
- Jemma J. Taitz
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jian Tan
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Duan Ni
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Camille Potier-Villette
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Georges Grau
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Vascular Immunology Unit, Discipline of Pathology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Ralph Nanan
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Medical School Nepean, The University of Sydney, Sydney, NSW, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Medical School Nepean, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
13
|
Hadrich I, Turki M, Chaari I, Abdelmoula B, Gargouri R, Khemakhem N, Elatoui D, Abid F, Kammoun S, Rekik M, Aloulou S, Sehli M, Mrad AB, Neji S, Feiguin FM, Aloulou J, Abdelmoula NB, Sellami H. Gut mycobiome and neuropsychiatric disorders: insights and therapeutic potential. Front Cell Neurosci 2025; 18:1495224. [PMID: 39845646 PMCID: PMC11750820 DOI: 10.3389/fncel.2024.1495224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Background The human gut mycobiome, a minor but integral component of the gut microbiome, has emerged as a significant player in host homeostasis and disease development. While bacteria have traditionally been the focus of gut microbiome studies, recent evidence suggests that fungal communities (mycobiota) may also play a crucial role in modulating health, particularly in neuropsychiatric disorders. Objective This review aims to provide a comprehensive overview of current knowledge on the relationship between the gut mycobiome and neuropsychiatric disorders, exploring the potential of targeting fungal communities as a novel therapeutic strategy. Methods We summarized recent findings from metagenomic analyses that characterize the diversity and composition of gut mycobiota and discuss how these communities interact with the host and other microorganisms via the gut-brain axis. Key methodologies for studying mycobiota, such as high-throughout sequencing and bioinformatics approaches, were also reviewed to highlight advances in the field. Results Emerging research links gut mycobiota dysbiosis to conditions such as schizophrenia, Alzheimer's disease, autism spectrum disorders, bipolar disorder, and depression. Studies indicate that specific fungal populations, such as Candida and Saccharomyces, may influence neuroinflammation, gut permeability and immune responses, thereby affecting mental health outcomes. Conclusion Understanding the gut mycobiome's role in neuropsychiatric disorders opens new avenues for therapeutic interventions, including antifungal treatments, probiotics, and dietary modifications. Future research should integrate multi-omics approaches to unravel the complex interkingdom interactions within the gut ecosystem, paving the way for personalized medicine in mental health care.
Collapse
Affiliation(s)
- Ines Hadrich
- Fungal and Parasitic Molecular Biology Laboratory LR 05ES11, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Mariem Turki
- Psychiatry “B” Department, Hedi Chaker University Hospital, Sfax, Tunisia
- Reserach Unit “Drosophila”UR22ES03, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Imen Chaari
- Psychiatry “B” Department, Hedi Chaker University Hospital, Sfax, Tunisia
- Reserach Unit “Drosophila”UR22ES03, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Balkiss Abdelmoula
- Genomics of Signalopathies at the Service of Precision Medicine LR23ES07 FMS, University of Sfax, Sfax, Tunisia
| | - Rahma Gargouri
- Department of Pneumology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Nahed Khemakhem
- Fungal and Parasitic Molecular Biology Laboratory LR 05ES11, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Dhawia Elatoui
- Reserach Unit “Drosophila”UR22ES03, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Fatma Abid
- Genomics of Signalopathies at the Service of Precision Medicine LR23ES07 FMS, University of Sfax, Sfax, Tunisia
| | - Sonda Kammoun
- Genomics of Signalopathies at the Service of Precision Medicine LR23ES07 FMS, University of Sfax, Sfax, Tunisia
- Ophthalmology Department, Habib Bourguiba University Hospital, Faculty of Medicine, Sfax, Tunisia
| | - Mona Rekik
- Genomics of Signalopathies at the Service of Precision Medicine LR23ES07 FMS, University of Sfax, Sfax, Tunisia
- Ophthalmology Department, Habib Bourguiba University Hospital, Faculty of Medicine, Sfax, Tunisia
| | - Samir Aloulou
- Genomics of Signalopathies at the Service of Precision Medicine LR23ES07 FMS, University of Sfax, Sfax, Tunisia
- Medical Carcinology Department, Mohamed Ben Sassi University Hospital of Gabes, Faculty of Medicine, Sfax, Tunisia
| | - Mariem Sehli
- Genomics of Signalopathies at the Service of Precision Medicine LR23ES07 FMS, University of Sfax, Sfax, Tunisia
- Ophthalmology Department, Habib Bourguiba University Hospital, Faculty of Medicine, Sfax, Tunisia
| | - Aymen Ben Mrad
- Genomics of Signalopathies at the Service of Precision Medicine LR23ES07 FMS, University of Sfax, Sfax, Tunisia
- Ophthalmology Department, Habib Bourguiba University Hospital, Faculty of Medicine, Sfax, Tunisia
| | - Sourour Neji
- Fungal and Parasitic Molecular Biology Laboratory LR 05ES11, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Fabian M. Feiguin
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Jihene Aloulou
- Psychiatry “B” Department, Hedi Chaker University Hospital, Sfax, Tunisia
- Reserach Unit “Drosophila”UR22ES03, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Nouha Bouayed Abdelmoula
- Genomics of Signalopathies at the Service of Precision Medicine LR23ES07 FMS, University of Sfax, Sfax, Tunisia
| | - Hayet Sellami
- Reserach Unit “Drosophila”UR22ES03, Faculty of Medicine, University of Sfax, Sfax, Tunisia
- Parasitology and Mycology Laboratory - Habib Bourguiba University Hospital, Sfax, Tunisia
| |
Collapse
|
14
|
Liu HY, Li S, Ogamune KJ, Ahmed AA, Kim IH, Zhang Y, Cai D. Fungi in the Gut Microbiota: Interactions, Homeostasis, and Host Physiology. Microorganisms 2025; 13:70. [PMID: 39858841 PMCID: PMC11767893 DOI: 10.3390/microorganisms13010070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/25/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
The mammalian gastrointestinal tract is a stage for dynamic inter-kingdom interactions among bacteria, fungi, viruses, and protozoa, which collectively shape the gut micro-ecology and influence host physiology. Despite being a modest fraction, the fungal community, also referred to as mycobiota, represents a critical component of the gut microbiota. Emerging evidence suggests that fungi act as early colonizers of the intestine, exerting a lasting influence on gut development. Meanwhile, the composition of the mycobiota is influenced by multiple factors, with diet, nutrition, drug use (e.g., antimicrobials), and physical condition standing as primary drivers. During its establishment, the mycobiota forms both antagonistic and synergistic relationships with bacterial communities within the host. For instance, intestinal fungi can inhibit bacterial colonization by producing alcohol, while certain bacterial pathogens exploit fungal iron carriers to enhance their growth. However, the regulatory mechanisms governing these complex interactions remain poorly understood. In this review, we first introduce the methodologies for studying the microbiota, then address the significance of the mycobiota in the mammalian intestine, especially during weaning when all 'primary drivers' change, and, finally, discuss interactions between fungi and bacteria under various influencing factors. Our review aims to shed light on the complex inter-kingdom dynamics between fungi and bacteria in gut homeostasis and provide insights into how they can be better understood and managed to improve host health and disease outcomes.
Collapse
Affiliation(s)
- Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.)
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Shicheng Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.)
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Kennedy Jerry Ogamune
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.)
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Abdelkareem A. Ahmed
- Department of Veterinary Science, Botswana University of Agriculture and Natural Resources, Private Bag 0027, Gaborone P.O. Box 100, Botswana;
| | - In Ho Kim
- Department of Animal Resource & Science, Dankook University, 119 Dandero, Donnamgu Cheonan, Cheonan-si 31116, Republic of Korea;
| | - Yunzeng Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China;
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.)
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
15
|
Graf MD, Murgueitio N, Vogel SC, Hicks L, Carlson AL, Propper CB, Kimmel M. Maternal Prenatal Stress and the Offspring Gut Microbiome: A Cross-Species Systematic Review. Dev Psychobiol 2025; 67:e70005. [PMID: 39636074 PMCID: PMC12010507 DOI: 10.1002/dev.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/14/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
The prenatal period is a critical developmental juncture with enduring effects on offspring health trajectories. An individual's gut microbiome is associated with health and developmental outcomes across the lifespan. Prenatal stress can disrupt an infant's microbiome, thereby increasing susceptibility to adverse outcomes. This cross-species systematic review investigates whether maternal prenatal stress affects the offspring's gut microbiome. The study analyzes 19 empirical, peer-reviewed research articles, including humans, rodents, and non-human primates, that included prenatal stress as a primary independent variable and offspring gut microbiome characteristics as an outcome variable. Prenatal stress appeared to correlate with differences in beta diversity and specific microbial taxa, but not alpha diversity. Prenatal stress is positively correlated with Proteobacteria, Bacteroidaceae, Lachnospiraceae, Prevotellaceae, Bacteroides, and Serratia. Negative correlations were observed for Actinobacteria, Enterobacteriaceae, Streptococcaceae, Bifidobacteria, Eggerthella, Parabacteroides, and Streptococcus. Evidence for the direction of association between prenatal stress and Lactobacillus was mixed. The synthesis of findings was limited by differences in study design, operationalization and timing of prenatal stress, timing of infant microbiome sampling, and microbiome analysis methods.
Collapse
Affiliation(s)
- Michelle D. Graf
- School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nicolas Murgueitio
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sarah C. Vogel
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, USA
| | - Lauren Hicks
- School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alexander L. Carlson
- Pediatric Physician Scientist Training Program, Department of Pediatrics, Division of Neonatology, University of California San Diego, San Diego, California, USA
| | - Cathi B. Propper
- School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mary Kimmel
- Department of Psychiatry, Washington University in St. Louis., St. Louis, Missouri, USA
| |
Collapse
|
16
|
Malan-Müller S, Martín-Hernández D, Caso JR, Matthijnssens J, Rodríguez-Urrutia A, Lowry CA, Leza JC. Metagenomic symphony of the intestinal ecosystem: How the composition affects the mind. Brain Behav Immun 2025; 123:510-523. [PMID: 39368785 DOI: 10.1016/j.bbi.2024.09.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/04/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024] Open
Abstract
Mental health disorders and neurodegenerative diseases place a heavy burden on patients and societies, and, although great strides have been made to understand the pathophysiology of these conditions, advancement in drug development is lagging. The importance of gastrointestinal health in maintaining overall health and preventing disease is not a new concept. Hundreds of years ago, healers from various cultures and civilizations recognized the crucial role of the gut in sustaining health. More than a century ago, scientists began exploring the restorative effects of probiotics, marking the early recognition of the importance of gut microbes. The omics era brought more enlightenment and enabled researchers to identify the complexity of the microbial ecosystems we harbour, encompassing bacteria, eukaryotes (including fungi), archaea, viruses, and other microorganisms. The extensive genetic capacity of the microbiota is dynamic and influenced by the environment. The microbiota therefore serves as a significant entity within us, with evolutionarily preserved functions in host metabolism, immunity, development, and behavior. The significant role of the bacterial gut microbiome in mental health and neurodegenerative disorders has been realized and described within the framework of the microbiota-gut-brain axis. However, the bacterial members do not function unaccompanied, but rather in concert, and there is a substantial knowledge gap regarding the involvement of non-bacterial microbiome members in these disorders. In this review, we will explore the current literature that implicates a role for the entire metagenomic ensemble, and how their complex interkingdom relationships could influence CNS functioning in mental health disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Stefanie Malan-Müller
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain; Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain.
| | - David Martín-Hernández
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain; Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain
| | - Javier R Caso
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain; Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain
| | - Jelle Matthijnssens
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Division of Clinical and Epidemiological Virology, Laboratory of Viral Metagenomics, Leuven, Belgium
| | - Amanda Rodríguez-Urrutia
- Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain; Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Catalonia, Spain; Group of Psychiatry, Mental Health and Addictions, Vall d'Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain; Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Juan C Leza
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain; Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain
| |
Collapse
|
17
|
Guglietta S, Li X, Saxena D. Role of Fungi in Tumorigenesis: Promises and Challenges. ANNUAL REVIEW OF PATHOLOGY 2025; 20:459-482. [PMID: 39854185 DOI: 10.1146/annurev-pathmechdis-111523-023524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
The mycobiome plays a key role in the host immune responses in homeostasis and inflammation. Recent studies suggest that an imbalance in the gut's fungi contributes to chronic, noninfectious diseases such as obesity, metabolic disorders, and cancers. Pathogenic fungi can colonize specific organs, and the gut mycobiome has been linked to the development and progression of various cancers, including colorectal, breast, head and neck, and pancreatic cancers. Some fungal species can promote tumorigenesis by triggering the complement system. However, in immunocompromised patients, fungi can also inhibit this activation and establish life-threatening infections. Interestingly, the interaction of the fungi and bacteria can also induce unique host immune responses. Recent breakthroughs and advancements in high-throughput sequencing of the gut and tumor mycobiomes are highlighting novel diagnostic and therapeutic opportunities for cancer. We discuss the latest developments in the field of cancer and the mycobiome and the potential benefits and challenges of antifungal therapies.
Collapse
Affiliation(s)
- Silvia Guglietta
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Hollings Cancer Center, Charleston, South Carolina, USA
| | - Xin Li
- Department of Molecular Pathobiology, NYU College of Dentistry, New York, NY, USA;
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
- Department of Urology, NYU Grossman School of Medicine, New York, NY, USA
| | - Deepak Saxena
- Department of Molecular Pathobiology, NYU College of Dentistry, New York, NY, USA;
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
18
|
Wei G. Insights into gut fungi in pigs: A comprehensive review. J Anim Physiol Anim Nutr (Berl) 2025; 109:96-112. [PMID: 39154229 DOI: 10.1111/jpn.14036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 06/17/2024] [Accepted: 08/04/2024] [Indexed: 08/19/2024]
Abstract
Fungi in the gut microbiota of mammals play a crucial role in host physiological regulation, including intestinal homeostasis and host immune regulation. However, our understanding of gut fungi in mammals remains limited, especially in economically valuable animals, such as pigs. Therefore, this review first describes the classification and characterisation of fungi, provides insights into the methods used to study gut fungi, and summarises the recent progress on pig gut fungi. Additionally, it discusses the challenges in the study of pig gut fungi and highlights potential perspectives. The aim of this review is to serve as a valuable reference for advancing our knowledge of gut fungi in animals.
Collapse
Affiliation(s)
- Guanyue Wei
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, China
| |
Collapse
|
19
|
Doan HT, Chiu YL, Cheng LC, Coad RA, Chiang HS. Candida tropicalis Alters Barrier Permeability and Claudin-1 Organization in Intestinal Epithelial Cells. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2025; 68:67-76. [PMID: 39918057 DOI: 10.4103/ejpi.ejpi-d-24-00090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/09/2024] [Indexed: 02/14/2025]
Abstract
ABSTRACT Inflammatory bowel disease (IBD) is an autoimmune disorder characterized by chronic inflammation of the gut and compromised intestinal barrier function, resulting from aberrant immune responses targeting the intestinal microbiota. While the involvement of Candida albicans in IBD pathogenesis is well-documented, the role of non- albicans Candida species in IBD remains less understood. Recent studies have identified a correlation between elevated levels of Candida tropicalis , a notable non- albicans opportunistic fungus, and the development of IBD. However, the precise impact of C. tropicalis on intestinal barrier function is not well elucidated. To address this knowledge gap, we utilized a cell model comprising polarized Caco-2 monolayers, which mimic the intestinal epithelium, to investigate the interaction between C. tropicalis and intestinal barrier function. Our results showed that incubation with C. tropicalis influenced transepithelial electrical resistance and increased permeability to the small molecule lucifer yellow, but did not affect permeability to the larger molecule fluorescein isothiocyanate-dextran. In addition, we observed internalization of the tight junction protein claudin-1 in the Caco-2 monolayers. Further experiments using Caco-2 monolayers exposed to the dectin-1 ligand zymosan induced similar changes in the distribution of claudin-1 but did not alter monolayer permeability. These findings suggest that C. tropicalis specifically affects intestinal barrier integrity and permeability to smaller solutes in intestinal epithelial cells.
Collapse
Affiliation(s)
- Ha The Doan
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yi-Ling Chiu
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Li-Chieh Cheng
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Rae Apaivongse Coad
- Department of Life Science, National Taiwan University, Taipei, Taiwan
- Program in Biological Sciences, Mahidol University International College, Nakorn Pathom, Thailand
| | - Hao-Sen Chiang
- Department of Life Science, National Taiwan University, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
20
|
Naik B, Sasikumar J, Das SP. From Skin and Gut to the Brain: The Infectious Journey of the Human Commensal Fungus Malassezia and Its Neurological Consequences. Mol Neurobiol 2025; 62:533-556. [PMID: 38871941 DOI: 10.1007/s12035-024-04270-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
The human mycobiome encompasses diverse communities of fungal organisms residing within the body and has emerged as a critical player in shaping health and disease. While extensive research has focused on the skin and gut mycobiome, recent investigations have pointed toward the potential role of fungal organisms in neurological disorders. Among those fungal organisms, the presence of the commensal fungus Malassezia in the brain has created curiosity because of its commensal nature and primary association with the human skin and gut. This budding yeast is responsible for several diseases, such as Seborrheic dermatitis, Atopic dermatitis, Pityriasis versicolor, Malassezia folliculitis, dandruff, and others. However recent findings surprisingly show the presence of Malassezia DNA in the brain and have been linked to diseases like Alzheimer's disease, Parkinson's disease, Multiple sclerosis, and Amyotrophic lateral sclerosis. The exact role of Malassezia in these disorders is unknown, but its ability to infect human cells, travel through the bloodstream, cross the blood-brain barrier, and reside along with the lipid-rich neuronal cells are potential mechanisms responsible for pathogenesis. This also includes the induction of pro-inflammatory cytokines, disruption of the blood-brain barrier, gut-microbe interaction, and accumulation of metabolic changes in the brain environment. In this review, we discuss these key findings from studies linking Malassezia to neurological disorders, emphasizing the complex and multifaceted nature of these cases. Furthermore, we discuss potential mechanisms through which Malassezia might contribute to the development of neurological conditions. Future investigations will open up new avenues for our understanding of the fungal gut-brain axis and how it influences human behavior. Collaborative research efforts among microbiologists, neuroscientists, immunologists, and clinicians hold promise for unraveling the enigmatic connections between human commensal Malassezia and neurological disorders.
Collapse
Affiliation(s)
- Bharati Naik
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Jayaprakash Sasikumar
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Shankar Prasad Das
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India.
| |
Collapse
|
21
|
Gong Y, Xue Q, Li J, Zhang S. Antifungal peptides from living organisms. Front Microbiol 2024; 15:1511461. [PMID: 39741586 PMCID: PMC11685209 DOI: 10.3389/fmicb.2024.1511461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 11/29/2024] [Indexed: 01/03/2025] Open
Abstract
In the post-COVID-19 era, people are increasingly concerned about microbial infections, including fungal infections that have risen in recent years. However, the currently available antifungal agents are rather limited. Worse still, the widespread use of the antifungal agents has caused the emergence of antifungal resistance in Candida, Cryptococcus, and Aspergillus species. Therefore, the development of novel antifungals is urgently needed. Antimicrobial peptides (AMPs), as components of the first-line defense of the host, are found to exhibit broad antimicrobial activity against bacteria, fungi, parasites, viruses, and protozoa. AMPs with antifungal activity are specifically referred to as antifungal peptides (AFPs). AFPs are currently regarded as the most promising alternative to conventional antifungal agents due to the fact that they are highly selective and less prone to facilitate the selection of drug resistance. In this review, we present an overview of the origin and classification of natural AFPs as well as their modes of action. Additionally, the production of natural, semisynthetic, and synthetic AFPs with a view to greater levels of exploitation is discussed. Finally, we evaluate the current and potential applications of AFPs in clinics and in the food industry.
Collapse
Affiliation(s)
- Yi Gong
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Qunhang Xue
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Jun Li
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Shicui Zhang
- Key Laboratory of Biological Resources and Ecology of Pamirs Plateau in Xinjiang Uygur Autonomous Region, College of Life and Geographic Sciences, Kashi University, Kashi, China
- Department of Marine Biology, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
22
|
Yang X, Wang X, Zhang M, Shen Y, Teng Y, Li M, Pan H. Gut Mycobiota of Three Rhinopithecus Species Provide New Insights Into the Association Between Diet and Environment. Integr Zool 2024. [PMID: 39690132 DOI: 10.1111/1749-4877.12932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/25/2024] [Accepted: 10/30/2024] [Indexed: 12/19/2024]
Abstract
Gut mycobiota are part of the gut microbiome, typically derived from the host diet and living environment. In this study, we examined the gut mycobiota of three snub-nosed monkeys: Rhinopithecus roxellana, R. bieti, and R. strykeri using next-generation amplicon sequencing targeting the fungal internal transcribed spacer. The alpha diversity indexes of gut mycobiota in R. bieti were significantly higher than R. roxellana and R. strykeri, the beta diversity indicated that R. roxellana and R. bieti had more similar feeding habits. Core mycobiota demonstrated commonalities among the three species and potentially associated with feeding habits. Mycobiota displaying significant differences exhibited the respective characteristics of the host, likely associated with the hosts' living environment. Among them, animal and plant pathogenic fungi and lichen parasites are potential threats to the survival of snub-nosed monkeys for their pathogenicity to both monkeys and their food plants. Functionally, fungal trophic modes and functional guilds revealed a strong association between gut mycobiota and host diet. We found a higher abundance and more significant correlations with lichen parasitic fungi in R. strykeri than the other two species, indicating potential threats to their foods. Accordingly, this study revealed the basic structures of gut mycobiota of three wild Rhinopithecus species and highlighted the associations between gut mycobiota and their feeding habits and living environments. Furthermore, due to the close connection between fungi and the environment, animals could ingest fungi from their diet; thus, we speculate that gut mycobiota may serve a role in environmental monitoring for wildlife.
Collapse
Affiliation(s)
- Xuanyi Yang
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, China
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiaochen Wang
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Mingyi Zhang
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ying Shen
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yang Teng
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ming Li
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Huijuan Pan
- School of Ecology and Nature Conservation, Beijing Forestry University, Beijing, China
| |
Collapse
|
23
|
Zeise KD, Falkowski NR, Metcalf JD, Brown CA, Huffnagle GB. Gene expression profiling reveals host defense strategies for restricting Candida albicans invasion and gastritis to the limiting ridge of the murine stomach. Infect Immun 2024; 92:e0043824. [PMID: 39535200 PMCID: PMC11629626 DOI: 10.1128/iai.00438-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Candida albicans is a fungal constituent of the human gastrointestinal microbiota that can tolerate acidic environments like the stomach, where it can be associated with ulcers and chronic gastritis. In mice, C. albicans induces gastritis without concurrent intestinal inflammation, suggesting that the stomach is particularly prone to fungal infection. We previously showed that C. albicans invasion in the limiting ridge does not extend to or elicit an inflammatory response in the adjacent glandular region, indicating regionalized gastritis in the murine stomach. However, the molecular pathways involved in the host response to C. albicans specifically in the limiting ridge have not been investigated. Here, we found that gastric dysbiosis was associated with C. albicans limiting ridge colonization and gastritis. We isolated the limiting ridge and evaluated the expression of over 90 genes involved in mucosal responses. C. albicans infection triggered a type 3 immune response marked by elevated Il17a, Il17f, Il1b, Tnf, and Il36g, as well as an upregulation of Il12a, Il4, Il10, and l13. Chemokine gene induction (including Ccl2, Ccl3, Ccl4, Ccl1l, Cxcl1, Cxcl2, Cxcl9, and Cxcl10) coincided with an influx of neutrophils, monocytes/macrophages, and eosinophils. Hyphal invasion caused tissue damage, epithelial remodeling, and upregulation of genes linked to epithelium signaling and antimicrobial responses in the limiting ridge. Our findings support a need for continued exploration into the interactions between the immunological milieu, the host microbiota, and clinical interventions such as the use of antibiotics and immunotherapeutic agents and their collective impact on invasive candidiasis risk.
Collapse
Affiliation(s)
- Karen D. Zeise
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicole R. Falkowski
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
- Division of Pulmonary & Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Joseph D. Metcalf
- Division of Pulmonary & Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher A. Brown
- Division of Pulmonary & Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Advanced Research Computing, Information and Technology Services, University of Michigan, Ann Arbor, Michigan, USA
| | - Gary B. Huffnagle
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
- Division of Pulmonary & Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
24
|
Ren Y, Ciwang R, Mehmood K, Li K. Effects of forages on the microbiota of crossed sheep on cold Plateau. Anim Biotechnol 2024; 35:2362639. [PMID: 38856695 DOI: 10.1080/10495398.2024.2362639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Diet is an important component to influence microbiota, there are less data available about the microbiome of Suffolk cross with Tibetan (SCT) animals with different fodders. The current study was conducted for comparing the fungi microbiota in SCT sheep fed with different forages. Sequencing of ileum samples from sheep groups of AH (alfalfa and oat grass), BH (mixture of grass and concentrated feeds), CH (concentrated feed I), DH (concentrated feed II) and EH (concentrated feed III) achieved 3,171,271 raw and 2,719,649 filtered sequences. Concentrated feeds changed fungi microbiota in SCT sheep with three phyla and 47 genera significantly different among the groups. Genera include positive genus of Scytalidium and negative fungi of Sarocladium, Kazachstania, Gibberella, Scytalidium, Candida, Wickerhamomyces. The findings of our study will contribute to efficient feeding of SCT sheep at cold plateau areas.
Collapse
Affiliation(s)
- Yue Ren
- Institute of Livestock Research, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, PR China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, PR China
| | - Renzeng Ciwang
- Institute of Livestock Research, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, PR China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, PR China
| | - Khalid Mehmood
- Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Kun Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, PR China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, PR China
| |
Collapse
|
25
|
Ma X, Li J, Li Z, Chen B, Ling Z, Feng S, Zhong Z, Peng G, Wang Y, Jiang Y, Gu Y. Analysis of fungal diversity in the feces of Arborophila rufipectus. Front Vet Sci 2024; 11:1430518. [PMID: 39469585 PMCID: PMC11514364 DOI: 10.3389/fvets.2024.1430518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
Background Intestinal fungal composition plays a crucial role in modulating host health, and thus is of great significance in the conservation of endangered bird species. However, research on gut fungal composition in birds is limited. Therefore, in this study, we aimed to examine gut fungal community and potential fecal pathogen composition in wild Arborophila rufipectus. Methods Fecal samples were collected from the habitats of wild A. rufipectus and Lophura nycthemera (a widely distributed species belonging to the same family as A. rufipectus) in summer and autumn. Thereafter, RNA was collected and the internal transcribed spacer rRNA gene was sequenced via high-throughput sequencing to investigate seasonal variations in intestinal core fungi, microbial fungi, and potential pathogenic fungi. Results The gut microbiota of A. rufipectus and L. nycthemera were highly similar and mainly consisted of three phyla, Ascomycota (58.46%), Basidiomycota (28.80%), and Zygomycota (3.56%), which accounted for 90.82% of the fungal community in all the samples. Further, the predominant genera were Ascomycota_unclassified (12.24%), Fungi_unclassified (8.37%), Davidiella (5.18%), Helotiales_unclassified (2.76%), Wickerhamomyces (1.84%), and Pleosporales_unclassified (1.14%), and the potential fecal pathogens identified included Candida, Cryptococcus, Trichosporon, and Malassezia. Conclusion Our results provide evidence that the diversity of intestinal fungi in the endangered species, A. rufipectus, is similar to that in the common species, L. nycthemera, and may serve as a basis for monitoring the status of A. rufipectus and for developing conservation measures.
Collapse
Affiliation(s)
- Xiaoping Ma
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Junshu Li
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhiguo Li
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Benping Chen
- Sichuan Laojunshan National Nature Reserve, Yibin, China
| | - Zhenwen Ling
- Sichuan Laojunshan National Nature Reserve, Yibin, China
| | - Shenglin Feng
- Sichuan Laojunshan National Nature Reserve, Yibin, China
| | - Zhijun Zhong
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Guangneng Peng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ya Wang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yaozhang Jiang
- Bioengineering Department, Sichuan Water Conservancy Vocational College, Chengdu, China
| | - Yu Gu
- College of Life Sciences, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
26
|
Sanches Santos Rizzo Zuttion M, Parimon T, Bora SA, Yao C, Lagree K, Gao CA, Wunderink RG, Kitsios GD, Morris A, Zhang Y, McVerry BJ, Modes ME, Marchevsky AM, Stripp BR, Soto CM, Wang Y, Merene K, Cho S, Victor BL, Vujkovic-Cvijin I, Gupta S, Cassel SL, Sutterwala FS, Devkota S, Underhill DM, Chen P. Antibiotic use during influenza infection augments lung eosinophils that impair immunity against secondary bacterial pneumonia. J Clin Invest 2024; 134:e180986. [PMID: 39255040 PMCID: PMC11527449 DOI: 10.1172/jci180986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024] Open
Abstract
A leading cause of mortality after influenza infection is the development of a secondary bacterial pneumonia. In the absence of a bacterial superinfection, prescribing antibacterial therapies is not indicated but has become a common clinical practice for those presenting with a respiratory viral illness. In a murine model, we found that antibiotic use during influenza infection impaired the lung innate immunologic defenses toward a secondary challenge with methicillin-resistant Staphylococcus aureus (MRSA). Antibiotics augment lung eosinophils, which have inhibitory effects on macrophage function through the release of major basic protein. Moreover, we demonstrated that antibiotic treatment during influenza infection caused a fungal dysbiosis that drove lung eosinophilia and impaired MRSA clearance. Finally, we evaluated 3 cohorts of hospitalized patients and found that eosinophils positively correlated with antibiotic use, systemic inflammation, and worsened outcomes. Altogether, our work demonstrates a detrimental effect of antibiotic treatment during influenza infection that has harmful immunologic consequences via recruitment of eosinophils to the lungs, thereby increasing the risk of developing a secondary bacterial infection.
Collapse
Affiliation(s)
| | | | | | - Changfu Yao
- Department of Medicine
- Women’s Guild Lung Institute
| | - Katherine Lagree
- Department of Biomedical Sciences
- Widjaja Foundation Inflammatory Bowel Disease Institute, and
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Catherine A. Gao
- Division of Pulmonary and Critical Care, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Richard G. Wunderink
- Division of Pulmonary and Critical Care, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Georgios D. Kitsios
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alison Morris
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yingze Zhang
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and
| | - Bryan J. McVerry
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | - Barry R. Stripp
- Department of Medicine
- Women’s Guild Lung Institute
- Department of Biomedical Sciences
| | | | - Ying Wang
- Department of Medicine
- Women’s Guild Lung Institute
| | | | - Silvia Cho
- Department of Medicine
- Women’s Guild Lung Institute
| | | | - Ivan Vujkovic-Cvijin
- Department of Medicine
- Department of Biomedical Sciences
- Widjaja Foundation Inflammatory Bowel Disease Institute, and
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Suman Gupta
- Department of Medicine
- Women’s Guild Lung Institute
| | | | | | - Suzanne Devkota
- Department of Medicine
- Department of Biomedical Sciences
- Widjaja Foundation Inflammatory Bowel Disease Institute, and
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Human Microbiome Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - David M. Underhill
- Department of Biomedical Sciences
- Widjaja Foundation Inflammatory Bowel Disease Institute, and
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Peter Chen
- Department of Medicine
- Women’s Guild Lung Institute
- Department of Biomedical Sciences
| |
Collapse
|
27
|
Wang D, Wang F, Huang Z, Li A, Dai W, Leng H, Jin L, Li Z, Sun K, Feng J. Structure and assembly process of skin fungal communities among bat species in northern China. Front Microbiol 2024; 15:1458258. [PMID: 39309528 PMCID: PMC11414763 DOI: 10.3389/fmicb.2024.1458258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/23/2024] [Indexed: 09/25/2024] Open
Abstract
Background The skin fungal communities of animals play a crucial role in maintaining host health and defending against pathogens. Because fungal infections can affect the skin microbiota of bats, gaining a comprehensive understanding of the characteristics of healthy bat skin fungal communities and the ecological processes driving them provides valuable insights into the interactions between pathogens and fungi. Methods We used Kruskal-Wallis tests and Permutational Multivariate Analysis of Variance (PERMANOVA) to clarify differences in skin fungal community structure among bat species. A Generalized Linear Model (GLM) based on a quasi-Poisson distribution and partial distance-based redundancy analysis (db-RDA) was performed to assess the influence of variables on skin fungal communities. Using community construction models to explore the ecological processes driving fungal community changes, t-tests and Wilcoxon tests were used to compare the alpha diversity and species abundance differences between the fungal structure on bat species' skin and the environmental fungal pool. Results We found significant differences in the composition and diversity of skin fungal communities among bat species influenced by temperature, sampling site, and body mass index. Trophic modes and skin fungal community complexity also varied among bat species. Null model and neutral model analysis demonstrated that deterministic processes dominated the assembly of skin fungal communities, with homogeneous selection as the predominant process. Skin fungal communities on bat species were impacted by the environmental fungal reservoir, and actively selected certain amplicon sequence variants (ASVs) from the environmental reservoir to adhere to the skin. Conclusion In this study, we revealed the structure and the ecological process driving the skin fungal community across bat species in northern China. Overall, these results broaden our knowledge of skin fungal communities among bat species, which may be beneficial to potential strategies for the protection of bats in China.
Collapse
Affiliation(s)
- Denghui Wang
- College of Life Science, Jilin Agricultural University, Changchun, China
| | - Fan Wang
- College of Life Science, Jilin Agricultural University, Changchun, China
| | - Zihao Huang
- College of Life Science, Jilin Agricultural University, Changchun, China
| | - Aoqiang Li
- School of Life Sciences, Central China Normal University, Wuhan, China
| | - Wentao Dai
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
| | - Haixia Leng
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
| | - Longru Jin
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
| | - Zhongle Li
- College of Life Science, Jilin Agricultural University, Changchun, China
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun, China
| | - Keping Sun
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
| | - Jiang Feng
- College of Life Science, Jilin Agricultural University, Changchun, China
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun, China
| |
Collapse
|
28
|
Katsoulis O, Pitts OR, Singanayagam A. The airway mycobiome and interactions with immunity in health and chronic lung disease. OXFORD OPEN IMMUNOLOGY 2024; 5:iqae009. [PMID: 39206335 PMCID: PMC11357796 DOI: 10.1093/oxfimm/iqae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/07/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
The existence of commensal fungi that reside within the respiratory tract, termed the airway mycobiome, has only recently been discovered. Studies are beginning to characterize the spectrum of fungi that inhabit the human upper and lower respiratory tract but heterogeneous sampling and analysis techniques have limited the generalizability of findings to date. In this review, we discuss existing studies that have examined the respiratory mycobiota in healthy individuals and in those with inflammatory lung conditions such as asthma, chronic obstructive pulmonary disease and cystic fibrosis. Associations between specific fungi and features of disease pathogenesis are emerging but the precise functional consequences imparted by mycobiota upon the immune system remain poorly understood. It is imperative that further research is conducted in this important area as a more detailed understanding could facilitate the development of novel approaches to manipulating the mycobiome for therapeutic benefit.
Collapse
Affiliation(s)
- Orestis Katsoulis
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2DD, UK
| | - Oliver R Pitts
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2DD, UK
| | - Aran Singanayagam
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2DD, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2DD, UK
| |
Collapse
|
29
|
Cheng KO, Montaño DE, Zelante T, Dietschmann A, Gresnigt MS. Inflammatory cytokine signalling in vulvovaginal candidiasis: a hot mess driving immunopathology. OXFORD OPEN IMMUNOLOGY 2024; 5:iqae010. [PMID: 39234208 PMCID: PMC11374039 DOI: 10.1093/oxfimm/iqae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 09/06/2024] Open
Abstract
Protective immunity to opportunistic fungal infections consists of tightly regulated innate and adaptive immune responses that clear the infection. Immune responses to infections of the vaginal mucosa by Candida species are, however, an exception. In the case of vulvovaginal candidiasis (VVC), the inflammatory response is associated with symptomatic disease, rather than that it results in pathogen clearance. As such VVC can be considered an inflammatory disease, which is a significant public health problem due to its predominance as a female-specific fungal infection. Particularly, women with recurrent VVC (RVVC) suffer from a significant negative impact on their quality of life and mental health. Knowledge of the inflammatory pathogenesis of (R)VVC may guide more effective diagnostic and therapeutic options to improve the quality of life of women with (R)VVC. Here, we review the immunopathogenesis of (R)VVC describing several elements that induce an inflammatory arson, starting with the activation threshold established by vaginal epithelial cells that prevent unnecessary ignition of inflammatory responses, epithelial and inflammasome-dependent immune responses. These inflammatory responses will drive neutrophil recruitment and dysfunctional neutrophil-mediated inflammation. We also review the, sometimes controversial, findings on the involvement of adaptive and systemic responses. Finally, we provide future perspectives on the potential of some unexplored cytokine axes and discuss whether VVC needs to be subdivided into subgroups to improve diagnosis and treatment.
Collapse
Affiliation(s)
- Kar On Cheng
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Beutenbergstraße 11a, Jena, 07749, Germany
| | - Dolly E Montaño
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Beutenbergstraße 11a, Jena, 07749, Germany
| | - Teresa Zelante
- Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi 1, Perugia, 06132, Italy
| | - Axel Dietschmann
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Beutenbergstraße 11a, Jena, 07749, Germany
| | - Mark S Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Beutenbergstraße 11a, Jena, 07749, Germany
| |
Collapse
|
30
|
Pu L, Pang S, Mu W, Chen X, Zou Y, Wang Y, Ding Y, Yan Q, Huang Y, Chen X, Peng T, Luo W, Wang S. The gut mycobiome signatures in long-lived populations. iScience 2024; 27:110412. [PMID: 39081291 PMCID: PMC11284699 DOI: 10.1016/j.isci.2024.110412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/22/2024] [Accepted: 06/26/2024] [Indexed: 08/02/2024] Open
Abstract
Long-lived individuals have been extensively studied as a model to investigate the role of the gut microbiota in aging, but their gut fungi remain almost unexplored. Here, we recruited a community-dwelling cohort of 251 participants (24-108 years, including 47 centenarians) from Guangxi in China to characterize the gut mycobiome signatures. We found gut mycobiome markedly varied during aging and determined aging as a predominant factor driving these variations. For long-lived individuals, core taxa, including Penicillium and Aspergillus, were maintained and Candida enterotype was enriched when compared with old counterparts. Individuals with this enterotype were more likely to possess Bacteroides enterotype enriched in young and centenarians. Moreover, the drivers from Candida enterotype were positively linked with the bacteria components dominated in Bacteroides enterotype. We also identified potentially beneficial yeasts-enriched features to differentiate long-lived individuals from others. Our findings suggest that the gut mycobiome develops with aging, and long-lived individuals possess unique fungal signatures.
Collapse
Affiliation(s)
- Lixia Pu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Shifu Pang
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
| | - Wenjie Mu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xiaodong Chen
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
- Guangxi Key Laboratory of Longevity Science and Technology, AIage Life Science Corporation Ltd., Nanning, Guangxi, China
| | - Yang Zou
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yugui Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yingying Ding
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Qi Yan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yu Huang
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
| | - Xiaochun Chen
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
- Guangxi Key Laboratory of Longevity Science and Technology, AIage Life Science Corporation Ltd., Nanning, Guangxi, China
| | - Tao Peng
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Weifei Luo
- AIage Life Science Corporation Ltd., Guangxi Free Trade Zone Aisheng Biotechnology Corporation Ltd., Nanning, Guangxi, China
- Guangxi Key Laboratory of Longevity Science and Technology, AIage Life Science Corporation Ltd., Nanning, Guangxi, China
| | - Shuai Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
31
|
Miranda FM, Azevedo VC, Ramos RJ, Renard BY, Piro VC. Hitac: a hierarchical taxonomic classifier for fungal ITS sequences compatible with QIIME2. BMC Bioinformatics 2024; 25:228. [PMID: 38956506 PMCID: PMC11220968 DOI: 10.1186/s12859-024-05839-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/11/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Fungi play a key role in several important ecological functions, ranging from organic matter decomposition to symbiotic associations with plants. Moreover, fungi naturally inhabit the human body and can be beneficial when administered as probiotics. In mycology, the internal transcribed spacer (ITS) region was adopted as the universal marker for classifying fungi. Hence, an accurate and robust method for ITS classification is not only desired for the purpose of better diversity estimation, but it can also help us gain a deeper insight into the dynamics of environmental communities and ultimately comprehend whether the abundance of certain species correlate with health and disease. Although many methods have been proposed for taxonomic classification, to the best of our knowledge, none of them fully explore the taxonomic tree hierarchy when building their models. This in turn, leads to lower generalization power and higher risk of committing classification errors. RESULTS Here we introduce HiTaC, a robust hierarchical machine learning model for accurate ITS classification, which requires a small amount of data for training and can handle imbalanced datasets. HiTaC was thoroughly evaluated with the established TAXXI benchmark and could correctly classify fungal ITS sequences of varying lengths and a range of identity differences between the training and test data. HiTaC outperforms state-of-the-art methods when trained over noisy data, consistently achieving higher F1-score and sensitivity across different taxonomic ranks, improving sensitivity by 6.9 percentage points over top methods in the most noisy dataset available on TAXXI. CONCLUSIONS HiTaC is publicly available at the Python package index, BIOCONDA and Docker Hub. It is released under the new BSD license, allowing free use in academia and industry. Source code and documentation, which includes installation and usage instructions, are available at https://gitlab.com/dacs-hpi/hitac .
Collapse
Affiliation(s)
- Fábio M Miranda
- Data Analytics and Computational Statistics, Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam, Germany
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Vasco C Azevedo
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rommel J Ramos
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Centro de Computação de Alto Desempenho, Universidade Federal do Pará, Belém, Brazil
| | - Bernhard Y Renard
- Data Analytics and Computational Statistics, Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam, Germany
| | - Vitor C Piro
- Data Analytics and Computational Statistics, Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Potsdam, Germany.
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
32
|
Stindt KR, McClean MN. Tuning interdomain conjugation to enable in situ population modification in yeasts. mSystems 2024; 9:e0005024. [PMID: 38747597 PMCID: PMC11326116 DOI: 10.1128/msystems.00050-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/15/2024] [Indexed: 05/28/2024] Open
Abstract
The ability to modify and control natural and engineered microbiomes is essential for biotechnology and biomedicine. Fungi are critical members of most microbiomes, yet technology for modifying the fungal members of a microbiome has lagged far behind that for bacteria. Interdomain conjugation (IDC) is a promising approach, as DNA transfer from bacterial cells to yeast enables in situ modification. While such genetic transfers have been known to naturally occur in a wide range of eukaryotes and are thought to contribute to their evolution, IDC has been understudied as a technique to control fungal or fungal-bacterial consortia. One major obstacle to the widespread use of IDC is its limited efficiency. In this work, we manipulated metabolic and physical interactions between genetically tractable Escherichia coli and Saccharomyces cerevisiae to control the incidence of IDC. We test the landscape of population interactions between the bacterial donors and yeast recipients to find that bacterial commensalism leads to maximized IDC, both in culture and in mixed colonies. We demonstrate the capacity of cell-to-cell binding via mannoproteins to assist both IDC incidence and bacterial commensalism in culture and model how these tunable controls can predictably yield a range of IDC outcomes. Furthermore, we demonstrate that these controls can be utilized to irreversibly alter a recipient yeast population, by both "rescuing" a poor-growing recipient population and collapsing a stable population via a novel IDC-mediated CRISPR/Cas9 system.IMPORTANCEFungi are important but often unaddressed members of most natural and synthetic microbial communities. This work highlights opportunities for modifying yeast microbiome populations through bacterial conjugation. While conjugation has been recognized for its capacity to deliver engineerable DNA to a range of cells, its dependence on cell contact has limited its efficiency. Here, we find "knobs" to control DNA transfer, by engineering the metabolic dependence between bacterial donors and yeast recipients and by changing their ability to physically adhere to each other. Importantly, we functionally validate these "knobs" by irreversibly altering yeast populations. We use these controls to "rescue" a failing yeast population, demonstrate the capacity of conjugated CRISPR/Cas9 to depress or collapse populations, and show that conjugation can be easily interrupted by disrupting cell-to-cell binding. These results offer building blocks toward in situ mycobiome editing, with significant implications for clinical treatments of fungal pathogens and other fungal system engineering.
Collapse
Affiliation(s)
- Kevin R Stindt
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Doctoral Program in Biophysics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Megan N McClean
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
33
|
Van Syoc E, Nixon MP, Silverman JD, Luo Y, Gonzalez FJ, Elbere I, Klovins J, Patterson AD, Rogers CJ, Ganda E. Changes in the type 2 diabetes gut mycobiome associate with metformin treatment across populations. mBio 2024; 15:e0016924. [PMID: 38767350 PMCID: PMC11237675 DOI: 10.1128/mbio.00169-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/08/2024] [Indexed: 05/22/2024] Open
Abstract
The human gut teems with a diverse ecosystem of microbes, yet non-bacterial portions of that community are overlooked in studies of metabolic diseases firmly linked to gut bacteria. Type 2 diabetes mellitus (T2D) is associated with compositional shifts in the gut bacterial microbiome and the mycobiome, the fungal portion of the microbiome. However, whether T2D and/or metformin treatment underpins fungal community changes is unresolved. To differentiate these effects, we curated a gut mycobiome cohort spanning 1,000 human samples across five countries and validated our findings in a murine experimental model. We use Bayesian multinomial logistic normal models to show that T2D and metformin both associate with shifts in the relative abundance of distinct gut fungi. T2D is associated with shifts in the Saccharomycetes and Sordariomycetes fungal classes, while the genera Fusarium and Tetrapisipora most consistently associate with metformin treatment. We confirmed the impact of metformin on individual gut fungi by administering metformin to healthy mice. Thus, metformin and T2D account for subtle, but significant and distinct variation in the gut mycobiome across human populations. This work highlights for the first time that metformin can confound associations of gut fungi with T2D and warrants the need to consider pharmaceutical interventions in investigations of linkages between metabolic diseases and gut microbial inhabitants. IMPORTANCE This is the largest to-date multi-country cohort characterizing the human gut mycobiome, and the first to investigate potential perturbations in gut fungi from oral pharmaceutical treatment. We demonstrate the reproducible effects of metformin treatment on the human and murine gut mycobiome and highlight a need to consider metformin as a confounding factor in investigations between type 2 diabetes mellitus and the gut microbial ecosystem.
Collapse
Affiliation(s)
- Emily Van Syoc
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Animal Science, The Pennsylvania State University, University Park, Pennsylvania, USA
- One Health Microbiome Center, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Michelle Pistner Nixon
- College of Information Sciences and Technology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Justin D. Silverman
- One Health Microbiome Center, The Pennsylvania State University, University Park, Pennsylvania, USA
- College of Information Sciences and Technology, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Medicine, The Pennsylvania State University, Hershey, Pennsylvania, USA
| | - Yuhong Luo
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Frank J. Gonzalez
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ilze Elbere
- Latvian Biomedical Research and Study Center, Riga, Latvia
| | - Janis Klovins
- Latvian Biomedical Research and Study Center, Riga, Latvia
| | - Andrew D. Patterson
- One Health Microbiome Center, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Connie J. Rogers
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia, USA
| | - Erika Ganda
- Department of Animal Science, The Pennsylvania State University, University Park, Pennsylvania, USA
- One Health Microbiome Center, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
34
|
Yan Q, Li S, Yan Q, Huo X, Wang C, Wang X, Sun Y, Zhao W, Yu Z, Zhang Y, Guo R, Lv Q, He X, Yao C, Li Z, Chen F, Ji Q, Zhang A, Jin H, Wang G, Feng X, Feng L, Wu F, Ning J, Deng S, An Y, Guo DA, Martin FM, Ma X. A genomic compendium of cultivated human gut fungi characterizes the gut mycobiome and its relevance to common diseases. Cell 2024; 187:2969-2989.e24. [PMID: 38776919 DOI: 10.1016/j.cell.2024.04.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 02/17/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
The gut fungal community represents an essential element of human health, yet its functional and metabolic potential remains insufficiently elucidated, largely due to the limited availability of reference genomes. To address this gap, we presented the cultivated gut fungi (CGF) catalog, encompassing 760 fungal genomes derived from the feces of healthy individuals. This catalog comprises 206 species spanning 48 families, including 69 species previously unidentified. We explored the functional and metabolic attributes of the CGF species and utilized this catalog to construct a phylogenetic representation of the gut mycobiome by analyzing over 11,000 fecal metagenomes from Chinese and non-Chinese populations. Moreover, we identified significant common disease-related variations in gut mycobiome composition and corroborated the associations between fungal signatures and inflammatory bowel disease (IBD) through animal experimentation. These resources and findings substantially enrich our understanding of the biological diversity and disease relevance of the human gut mycobiome.
Collapse
Affiliation(s)
- Qiulong Yan
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, School of Pharmacy, Dalian Medical University, Dalian 116044, China; College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Shenghui Li
- Puensum Genetech Institute, Wuhan 430076, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100091, China
| | - Qingsong Yan
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Xiaokui Huo
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Chao Wang
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, School of Pharmacy, Dalian Medical University, Dalian 116044, China; First Affiliated Hospital, Dalian Medical University, Dalian 116044, China.
| | - Xifan Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100091, China; Department of Obstetrics and Gynecology, Columbia University, New York, NY 10027, USA
| | - Yan Sun
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Wenyu Zhao
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, School of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Zhenlong Yu
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, School of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yue Zhang
- Puensum Genetech Institute, Wuhan 430076, China
| | - Ruochun Guo
- Puensum Genetech Institute, Wuhan 430076, China
| | - Qingbo Lv
- Puensum Genetech Institute, Wuhan 430076, China
| | - Xin He
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, School of Pharmacy, Dalian Medical University, Dalian 116044, China; Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Changliang Yao
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | | | - Fang Chen
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Qianru Ji
- Puensum Genetech Institute, Wuhan 430076, China
| | - Aiqin Zhang
- Puensum Genetech Institute, Wuhan 430076, China
| | - Hao Jin
- Puensum Genetech Institute, Wuhan 430076, China
| | - Guangyang Wang
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Xiaoying Feng
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Lei Feng
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Fan Wu
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Jing Ning
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, School of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Sa Deng
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, School of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yue An
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China
| | - De-An Guo
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Francis M Martin
- Université de Lorraine, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement, UMR Interactions Arbres/Microorganismes, Centre INRAE Grand Est-Nancy, Champenoux 54280, France; Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, Beijing Forestry University, Beijing 100091, China.
| | - Xiaochi Ma
- Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, School of Pharmacy, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
35
|
Nagpal S, Mande SS, Hooda H, Dutta U, Taneja B. EnsembleSeq: a workflow towards real-time, rapid, and simultaneous multi-kingdom-amplicon sequencing for holistic and resource-effective microbiome research at scale. Microbiol Spectr 2024; 12:e0415023. [PMID: 38687072 PMCID: PMC11237516 DOI: 10.1128/spectrum.04150-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/30/2024] [Indexed: 05/02/2024] Open
Abstract
Bacterial communities are often concomitantly present with numerous microorganisms in the human body and other natural environments. Amplicon-based microbiome studies have generally paid skewed attention, that too at a rather shallow genus level resolution, to the highly abundant bacteriome, with interest now forking toward the other microorganisms, particularly fungi. Given the generally sparse abundance of other microbes in the total microbiome, simultaneous sequencing of amplicons targeting multiple microbial kingdoms could be possible even with full multiplexing. Guiding studies are currently needed for performing and monitoring multi-kingdom-amplicon sequencing and data capture at scale. Aiming to address these gaps, amplification of full-length bacterial 16S rRNA gene and entire fungal internal-transcribed spacer (ITS) region was performed for human saliva samples (n = 96, including negative and positive controls). Combined amplicon DNA libraries were prepared for nanopore sequencing using a major fraction of 16S molecules and a minor fraction of ITS amplicons. Sequencing was performed in a single run of an R10.4.1 flow cell employing the latest V14 chemistry. An approach for real-time monitoring of the species saturation using dynamic rarefaction was designed as a guiding determinant of optimal run time. Real-time saturation monitoring for both bacterial and fungal species enabled the completion of sequencing within 30 hours, utilizing less than 60% of the total nanopores. Approximately 5 million high quality (HQ) taxonomically assigned reads were generated (~4.2 million bacterial and 0.7 million fungal), providing a wider (beyond bacteriome) snapshot of human oral microbiota at species-level resolution. Among the more than 400 bacterial and 240 fungal species identified in the studied samples, the species of Streptococcus (e.g., Streptococcus mitis and Streptococcus oralis) and Candida (e.g., Candida albicans and Candida tropicalis) were observed to be the dominating microbes in the oral cavity, respectively. This conformed well with the previous reports of the human oral microbiota. EnsembleSeq provides a proof-of-concept toward the identification of both fungal and bacterial species simultaneously in a single fully multiplexed nanopore sequencing run in a time- and resource-effective manner. Details of this workflow, along with the associated codebase, are provided to enable large-scale application for a holistic species-level microbiome study. IMPORTANCE Human microbiome is a sum total of a variety of microbial genomes (including bacteria, fungi, protists, viruses, etc.) present in and on the human body. Yet, a majority of amplicon-based microbiome studies have largely remained skewed toward bacteriome as an assumed proxy of the total microbiome, primarily at a shallow genus level. Cost, time, effort, data quality/management, and importantly lack of guiding studies often limit progress in the direction of moving beyond bacteriome. Here, EnsembleSeq presents a proof-of-concept toward concomitantly capturing multiple-kingdoms of microorganisms (bacteriome and mycobiome) in a fully multiplexed (96-sample) single run of long-read amplicon sequencing. In addition, the workflow captures dynamic tracking of species-level saturation in a time- and resource-effective manner.
Collapse
Affiliation(s)
- Sunil Nagpal
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- TCS Research, Tata Consultancy Services Ltd, Pune, India
| | | | - Harish Hooda
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Usha Dutta
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Bhupesh Taneja
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
36
|
Pereira H, Chakarov N, Hoffman JI, Rinaud T, Ottensmann M, Gladow KP, Tobias B, Caspers BA, Maraci Ö, Krüger O. Early-life factors shaping the gut microbiota of Common buzzard nestlings. Anim Microbiome 2024; 6:27. [PMID: 38745254 DOI: 10.1186/s42523-024-00313-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/01/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Exploring the dynamics of gut microbiome colonisation during early-life stages is important for understanding the potential impact of microbes on host development and fitness. Evidence from model organisms suggests a crucial early-life phase when shifts in gut microbiota can lead to immune dysregulation and reduced host condition. However, our understanding of gut microbiota colonisation in long-lived vertebrates, especially during early development, remains limited. We therefore used a wild population of common buzzard nestlings (Buteo buteo) to investigate connections between the early-life gut microbiota colonisation, environmental and host factors. RESULTS We targeted both bacterial and eukaryotic microbiota using the 16S and 28S rRNA genes. We sampled the individuals during early developmental stages in a longitudinal design. Our data revealed that age significantly affected microbial diversity and composition. Nest environment was a notable predictor of microbiota composition, with particularly eukaryotic communities differing between habitats occupied by the hosts. Nestling condition and infection with the blood parasite Leucocytozoon predicted microbial community composition. CONCLUSION Our findings emphasise the importance of studying microbiome dynamics to capture changes occurring during ontogeny. They highlight the role of microbial communities in reflecting host health and the importance of the nest environment for the developing nestling microbiome. Overall, this study contributes to understanding the complex interplay between microbial communities, host factors, and environmental variables, and sheds light on the ecological processes governing gut microbial colonisation during early-life stages.
Collapse
Affiliation(s)
- Hugo Pereira
- Department of Animal Behaviour, Bielefeld University, Konsequenz 45, 33615, Bielefeld, NRW, Germany.
| | - Nayden Chakarov
- Department of Animal Behaviour, Bielefeld University, Konsequenz 45, 33615, Bielefeld, NRW, Germany
- Joint Institute for Individualisation in a Changing Environment (JICE), Bielefeld University and University of Münster, Konsequenz 45, 33615, Bielefeld, NRW, Germany
| | - Joseph I Hoffman
- Department of Animal Behaviour, Bielefeld University, Konsequenz 45, 33615, Bielefeld, NRW, Germany
- Department of Evolutionary Population Genetics, Bielefeld University, Konsequenz 45, 33615, Bielefeld, NRW, Germany
- Joint Institute for Individualisation in a Changing Environment (JICE), Bielefeld University and University of Münster, Konsequenz 45, 33615, Bielefeld, NRW, Germany
- British Antarctic Survey, High Cross, Madingley Road, Cambridge, CB3 OET, UK
| | - Tony Rinaud
- Department of Animal Behaviour, Bielefeld University, Konsequenz 45, 33615, Bielefeld, NRW, Germany
| | - Meinolf Ottensmann
- Department of Animal Behaviour, Bielefeld University, Konsequenz 45, 33615, Bielefeld, NRW, Germany
| | - Kai-Philipp Gladow
- Department of Animal Behaviour, Bielefeld University, Konsequenz 45, 33615, Bielefeld, NRW, Germany
| | - Busche Tobias
- Medical School East Westphalia-Lippe & Center for Biotechnology (CeBiTec), Bielefeld University, Universitätsstraße 27, 33615, Bielefeld, NRW, Germany
| | - Barbara A Caspers
- Department of Behavioural Ecology, Bielefeld University, Konsequenz 45, 33615, Bielefeld, NRW, Germany
- Joint Institute for Individualisation in a Changing Environment (JICE), Bielefeld University and University of Münster, Konsequenz 45, 33615, Bielefeld, NRW, Germany
| | - Öncü Maraci
- Department of Behavioural Ecology, Bielefeld University, Konsequenz 45, 33615, Bielefeld, NRW, Germany
- Joint Institute for Individualisation in a Changing Environment (JICE), Bielefeld University and University of Münster, Konsequenz 45, 33615, Bielefeld, NRW, Germany
| | - Oliver Krüger
- Department of Animal Behaviour, Bielefeld University, Konsequenz 45, 33615, Bielefeld, NRW, Germany
- Joint Institute for Individualisation in a Changing Environment (JICE), Bielefeld University and University of Münster, Konsequenz 45, 33615, Bielefeld, NRW, Germany
| |
Collapse
|
37
|
Nenciarini S, Renzi S, di Paola M, Meriggi N, Cavalieri D. Ascomycetes yeasts: The hidden part of human microbiome. WIREs Mech Dis 2024; 16:e1641. [PMID: 38228159 DOI: 10.1002/wsbm.1641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 01/18/2024]
Abstract
The fungal component of the microbiota, the mycobiota, has been neglected for a long time due to its poor richness compared to bacteria. Limitations in fungal detection and taxonomic identification arise from using metagenomic approaches, often borrowed from bacteriome analyses. However, the relatively recent discoveries of the ability of fungi to modulate the host immune response and their involvement in human diseases have made mycobiota a fundamental component of the microbial communities inhabiting the human host, deserving some consideration in host-microbe interaction studies and in metagenomics. Here, we reviewed recent data on the identification of yeasts of the Ascomycota phylum across human body districts, focusing on the most representative genera, that is, Saccharomyces and Candida. Then, we explored the key factors involved in shaping the human mycobiota across the lifespan, ranging from host genetics to environment, diet, and lifestyle habits. Finally, we discussed the strengths and weaknesses of culture-dependent and independent methods for mycobiota characterization. Overall, there is still room for some improvements, especially regarding fungal-specific methodological approaches and bioinformatics challenges, which are still critical steps in mycobiota analysis, and to advance our knowledge on the role of the gut mycobiota in human health and disease. This article is categorized under: Immune System Diseases > Genetics/Genomics/Epigenetics Immune System Diseases > Environmental Factors Infectious Diseases > Environmental Factors.
Collapse
Affiliation(s)
| | - Sonia Renzi
- Department of Biology, University of Florence, Florence, Italy
| | - Monica di Paola
- Department of Biology, University of Florence, Florence, Italy
| | - Niccolò Meriggi
- Department of Biology, University of Florence, Florence, Italy
| | | |
Collapse
|
38
|
Liu T, Asif IM, Chen Y, Zhang M, Li B, Wang L. The Relationship between Diet, Gut Mycobiome, and Functional Gastrointestinal Disorders: Evidence, Doubts, and Prospects. Mol Nutr Food Res 2024; 68:e2300382. [PMID: 38659179 DOI: 10.1002/mnfr.202300382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 12/11/2023] [Indexed: 04/26/2024]
Abstract
Gut fungi are important parts of intestinal microbes. Dietary ingredients have the potential to regulate the structure of gut fungi in different directions and modulate mycobiome composition by changing dietary patterns, which have been applied to neurological disorders. Emerging pieces of evidence have revealed the regulatory functions of gut mycobiome in gastrointestinal diseases, but the relationships between gut fungi and functional gastrointestinal disorders (FGIDs) are ignored in the past. This review discusses the impact of dietary nutrients and patterns on mycobiome, and the possible ways in which gut fungi are involved in the pathogenesis of FGIDs. Besides affecting host immunity, intestinal fungi can be involved in the pathogenesis of FGIDs by endosymbiosis or bidirectional regulation with gut bacteria as well. In addition, the Mediterranean diet may be the most appropriate dietary pattern for subjects with FGIDs. A full understanding of these associations may have important implications for the pathogenesis and treatment of FGIDs.
Collapse
Affiliation(s)
- Tianxu Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, 430070, China
| | - Ismail Muhammad Asif
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, 430070, China
| | - Yan Chen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, 430070, China
| | - Meixue Zhang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, 430070, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, 430070, China
| | - Ling Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan, Hubei, 430070, China
| |
Collapse
|
39
|
Han M, Wang X, Zhang J, Su L, Ishaq HM, Li D, Cui J, Zhao H, Yang F. Gut bacterial and fungal dysbiosis in tuberculosis patients. BMC Microbiol 2024; 24:141. [PMID: 38658829 PMCID: PMC11044546 DOI: 10.1186/s12866-024-03275-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/24/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Recent studies have more focused on gut microbial alteration in tuberculosis (TB) patients. However, no detailed study on gut fungi modification has been reported till now. So, current research explores the characteristics of gut microbiota (bacteria)- and mycobiota (fungi)-dysbiosis in TB patients and also assesses the correlation between the gut microbiome and serum cytokines. It may help to screen the potential diagnostic biomarker for TB. RESULTS The results show that the alpha diversity of the gut microbiome (including bacteria and fungi) decreased and altered the gut microbiome composition of TB patients. The bacterial genera Bacteroides and Prevotella were significantly increased, and Blautia and Bifidobacterium decreased in the TB patients group. The fungi genus Saccharomyces was increased while decreased levels of Aspergillus in TB patients. It indicates that gut microbial equilibrium between bacteria and fungi has been altered in TB patients. The fungal-to-bacterial species ratio was significantly decreased, and the bacterial-fungal trans-kingdom interactions have been reduced in TB patients. A set model including Bacteroides, Blautia, Eubacterium_hallii_group, Apiotrichum, Penicillium, and Saccharomyces may provide a better TB diagnostics option than using single bacterial or fungi sets. Also, gut microbial dysbiosis has a strong correlation with the alteration of IL-17 and IFN-γ. CONCLUSIONS Our results demonstrate that TB patients exhibit the gut bacterial and fungal dysbiosis. In the clinics, some gut microbes may be considered as potential biomarkers for auxiliary TB diagnosis.
Collapse
Affiliation(s)
- MeiQing Han
- Department Four of Tuberculosis Medicine, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Xia Wang
- Department Four of Tuberculosis Medicine, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - JiaMin Zhang
- Department Four of Tuberculosis Medicine, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Lin Su
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Hafiz Muhammad Ishaq
- Department of Pathobiology, Faculty of Veterinary and Animal Sciences, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Duan Li
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - JunWei Cui
- Department Four of Tuberculosis Medicine, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - HuaJie Zhao
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China.
| | - Fan Yang
- Department Four of Tuberculosis Medicine, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang, China.
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
40
|
Bharti S, Singh B, Kumar S, Kumar R, Kumar J. Synthesis of bio-stabilized silver nanoparticles using Roccella montagnei, their anticandidal capacities & potential to inhibit the virulence factors in fluconazole-resistant Candida albicans. World J Microbiol Biotechnol 2024; 40:158. [PMID: 38592601 DOI: 10.1007/s11274-024-03928-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 02/15/2024] [Indexed: 04/10/2024]
Abstract
Candida species is the causative agent in approximately 80% of invasive mycoses and drug-resistant Candida albicans is among the four strains of 'critical priority group' framed by WHO. Lichens are endowed with some rare phytochemicals and a plethora of therapeutics viz. antifungal capacities of Roccella montagnei. Biosynthesis of silver nanoparticles (AgNPs) using lichen could offer an eco-friendly, and cost-effective alternative against emerging 'microbial resistance.' Therefore, the objective was to biosynthesize silver nanoparticles (Rm-AgNPs) using a Hydro-alcoholic (1:1) extract of R. montagnei to develop a potent anticandidal agent against Fluconazole-resistant C. albicans NBC099. UV-Spectroscopy identified AgNPs specific-peak of Rm-AgNPs at 420-440 nm and FTIR revealed the presence of amines, alcohol, aromatic compounds, and acids. SEM and TEM analysis indicated that Rm-AgNPs are spherical shaped with a size range of 10-50 nm. Zetasizer analysis indicated that particles are highly stable and have a mean hydrodynamic diameter of 116 nm with a zeta potential charge of - 41 mV. XRD analysis suggested face centered cubic crystal lattice structure. Results indicated that Rm-AgNPs strongly inhibited the growth of NBC099 at a minimum inhibitory concentration (IC50) of ≤ 15 µg. C. albicans culture treated with Rm-AgNPs at concentrations below IC50, down-regulates the production of different virulence factors in NBC099, viz. hyphal formation (> 85%), biofilms production (> 80%), phospholipase, esterase, proteinase activity. The apoptosis assay demonstrated the Rm-AgNPs induced apoptosis in NBC099 cells via oxidative stress. Interestingly, Rm-AgNPs showed negligible cytotoxicity (< 6%) in murine RAW 246.7 macrophage cells at a concentration above 15 µg/mL. Therefore, Rm-AgNPs have been offered as an anti-candida alternative that can be utilized to improve the efficacy of already available medications.
Collapse
Affiliation(s)
- Shweta Bharti
- Department of Environmental Microbiology, Babasaheb Bhimrao Ambedkar University, Lucknow, Lucknow, India
| | - Balwant Singh
- Department of Biotechnology, Bundelkhand University, Jhansi, India
| | - Sanket Kumar
- Department of Botany, School of Sciences, IFTM University, Moradabad, 244102, India
| | - Rajesh Kumar
- Department of Environmental Microbiology, Babasaheb Bhimrao Ambedkar University, Lucknow, Lucknow, India
| | - Jatinder Kumar
- CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu & Kashmir, India.
| |
Collapse
|
41
|
Guo J, Shi W, Li X, Yang B, Qin C, Su L. Comparative Analysis of Gut Microbiomes in Laboratory Chinchillas, Ferrets, and Marmots: Implications for Pathogen Infection Research. Microorganisms 2024; 12:646. [PMID: 38674591 PMCID: PMC11051751 DOI: 10.3390/microorganisms12040646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Gut microbes play a vital role in the health and disease of animals, especially in relation to pathogen infections. Chinchillas, ferrets, and marmots are commonly used as important laboratory animals for infectious disease research. Here, we studied the bacterial and fungal microbiota and discovered that chinchillas had higher alpha diversity and a higher abundance of bacteria compared to marmots and ferrets by using the metabarcoding of 16S rRNA genes and ITS2, coupled with co-occurrence network analysis. The dominant microbes varied significantly among the three animal species, particularly in the gut mycobiota. In the ferrets, the feces were dominated by yeast such as Rhodotorula and Kurtzmaniella, while in the chinchillas, we found Teunomyces and Penicillium dominating, and Acaulium, Piromyces, and Kernia in the marmots. Nevertheless, the dominant bacterial genera shared some similarities, such as Clostridium and Pseudomonas across the three animal species. However, there were significant differences observed, such as Vagococcus and Ignatzschineria in the ferrets, Acinetobacter and Bacteroides in the chinchillas, and Bacteroides and Cellvibrio in the marmots. Additionally, our differential analysis revealed significant differences in classification levels among the three different animal species, as well as variations in feeding habitats that resulted in distinct contributions from the host microbiome. Therefore, our data are valuable for monitoring and evaluating the impacts of the microbiome, as well as considering potential applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Lei Su
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing 100021, China; (J.G.); (W.S.); (X.L.); (B.Y.); (C.Q.)
| |
Collapse
|
42
|
Monteiro JS, Kaushik K, de Arruda JAA, Georgakopoulou E, Vieira AT, Silva TA, Devadiga D, Anyanechi CE, Shetty S. Fungal footprints in oral cancer: unveiling the oral mycobiome. FRONTIERS IN ORAL HEALTH 2024; 5:1360340. [PMID: 38550775 PMCID: PMC10973146 DOI: 10.3389/froh.2024.1360340] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/14/2024] [Indexed: 11/12/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common type of head and neck cancer, with a high mortality rate. There is growing evidence supporting a link between oral cancer and the microbiome. The microbiome can impact various aspects of cancer, such as pathogenesis, diagnosis, treatment, and prognosis. While there is existing information on bacteria and its connection to oral cancer, the fungi residing in the oral cavity represent a significant component of the microbiome that remains in its early stages of exploration and understanding. Fungi comprise a minuscule part of the human microbiome called the mycobiome. Mycobiome is ubiquitous in the human body but a weakened immune system offers a leeway space for fungi to showcase its virulence. The role of mycobiome as a colonizer, facilitator, or driver of carcinogenesis is still ambiguous. Reactivating the mycobiome that undergoes collateral damage associated with cancer treatment can be watershed event in cancer research. The coordinated, virulent, non-virulent behavior of the fungi once they reach a critical density must be hacked, considering its diagnostic, prognostic and therapeutic implications in cancer. This review highlights the diversity of the mycobiome and its potential role in oral cancer.
Collapse
Affiliation(s)
- Jessica Sonal Monteiro
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Kriti Kaushik
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Mangalore, India
| | - José Alcides Almeida de Arruda
- Department of Oral Diagnosis and Pathology, School of Dentistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eleni Georgakopoulou
- Laboratory of Histology-Embryology, Molecular Carcinogenesis Group, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Angelica Thomaz Vieira
- Laboratory of Microbiota and Immunomodulation, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tarcilia A. Silva
- Department of Oral Surgery, Pathology and Clinical Dentistry, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Darshana Devadiga
- Department of Conservative Dentistry and Endodontics, AB Shetty Memorial Institute of Dental Sciences, NITTE (Deemed to be University), Mangalore, India
| | - Charles E. Anyanechi
- Department of Oral and Maxillofacial Surgery, University of Calabar/University of Calabar Teaching Hospital, Calabar, Nigeria
| | - Sameep Shetty
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Mangalore, India
| |
Collapse
|
43
|
Mouzan ME, Hussaini AA, Sarkhy AA, Assiri A. Intestinal fungal profile in healthy Saudi children. Arab J Gastroenterol 2024; 25:18-21. [PMID: 37993375 DOI: 10.1016/j.ajg.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 11/11/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND AND STUDY AIM Fungi have a well-established role in medicine. Herein, we describe the fungal profile and abundance in the gut of healthy Saudi children. PATIENTS AND METHODS Fecal samples from a random sample of 20 school-age Saudi children were collected, stored at -80 °C, and dispatched to the laboratory in the USA where fungal DNAs were isolated and shotgun metagenomic sequencing was performed. Abundance was presented as average percentage of fungal taxa. RESULTS The median age of the participants was 12.5 years (range: 7-16 years), and 35 % were male. Ascomycota were the most abundant phyla and Eurotiomycetes, Saccharomycetes, were the most abundant class. The average abundance of fungal genera were Histoplasma (36 %) and Saccharomyces (31 %). The most abundant species were Histoplasma capsulatum (36 %) and Saccharomyces pastorianus (23 %). Other less abundant but may be functionally important genera and species included Candida (2.6 %) and Saccharomycescerevisiae (8 %). CONCLUSION The profile and abundance of the gut fungi in healthy Saudi children reveals important differences compared to Western literature. Accordingly, this report represents a more appropriate reference than Western data to use as controls for regional studies aiming to identify fungi associated with disease.
Collapse
Affiliation(s)
- Mohammad El Mouzan
- Department of Pediatrics, Gastroenterology Division, King Saud University, Riyadh, Kingdom of Saudi Arabia.
| | | | - Ahmed Al Sarkhy
- Department of Pediatrics, Gastroenterology Division, King Saud University, Riyadh, Kingdom of Saudi Arabia.
| | - Asaad Assiri
- Department of Pediatrics, Gastroenterology Division, King Saud University, Riyadh, Kingdom of Saudi Arabia.
| |
Collapse
|
44
|
Chen Y, Meng L, Yuan W, Gao Z, Zhang X, Xie B, Song J, Li J, Zhong J, Liu X. Gut Fungal Microbiota Alterations in Pulmonary Arterial Hypertensive Rats. Biomedicines 2024; 12:298. [PMID: 38397900 PMCID: PMC10886911 DOI: 10.3390/biomedicines12020298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
The gut microbiome's imbalance has been implicated in the pathogenesis of pulmonary arterial hypertension (PAH), yet the contribution of the gut mycobiome remains largely unclear. This study delineates the gut mycobiome profile in PAH and examines its interplay with the bacterial microbiome alterations. Fecal samples from monocrotaline-induced PAH rats and matched controls were subjected to internal transcribed spacer 1 (ITS1) sequencing for fungal community assessment and 16S ribosomal RNA (rRNA) gene sequencing for bacterial community characterization. Comparative analysis revealed no significant disparities in the overall mycobiome diversity between the PAH and control groups. However, taxonomic profiling identified differential mycobiome compositions, with the PAH group exhibiting a significant enrichment of genera such as Wallemia, unidentified_Branch02, Postia, Malassezia, Epicoccum, Cercospora, and Alternaria. Conversely, genera Xeromyces, unidentified_Plectosphaerellaceae, and Monilia were more abundant in the controls. Correlations of Malassezia and Wallemia abundance with hemodynamic parameters were observed. Indications of bidirectional fungal-bacterial community interactions were also noted. This investigation reveals distinct gut mycobiome alterations in PAH, which are intricately associated with concurrent bacterial microbiome changes, suggesting a possible contributory role of gut fungi in PAH pathophysiology. These findings underscore the potential for novel gut mycobiome-targeted therapeutic interventions in PAH management.
Collapse
Affiliation(s)
- Yihang Chen
- Department of Cardiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; (Y.C.); (B.X.); (J.S.)
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Liukun Meng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100032, China;
| | - Wen Yuan
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China;
| | - Zehan Gao
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; (Z.G.); (J.L.)
| | - Xun Zhang
- Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China;
| | - Boqia Xie
- Department of Cardiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; (Y.C.); (B.X.); (J.S.)
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jiawei Song
- Department of Cardiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; (Y.C.); (B.X.); (J.S.)
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jifeng Li
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; (Z.G.); (J.L.)
| | - Jiuchang Zhong
- Department of Cardiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; (Y.C.); (B.X.); (J.S.)
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaoyan Liu
- Department of Cardiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; (Y.C.); (B.X.); (J.S.)
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China;
| |
Collapse
|
45
|
Li R, Li J, Zhou X. Lung microbiome: new insights into the pathogenesis of respiratory diseases. Signal Transduct Target Ther 2024; 9:19. [PMID: 38228603 PMCID: PMC10791971 DOI: 10.1038/s41392-023-01722-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/25/2023] [Accepted: 11/22/2023] [Indexed: 01/18/2024] Open
Abstract
The lungs were long thought to be sterile until technical advances uncovered the presence of the lung microbial community. The microbiome of healthy lungs is mainly derived from the upper respiratory tract (URT) microbiome but also has its own characteristic flora. The selection mechanisms in the lung, including clearance by coughing, pulmonary macrophages, the oscillation of respiratory cilia, and bacterial inhibition by alveolar surfactant, keep the microbiome transient and mobile, which is different from the microbiome in other organs. The pulmonary bacteriome has been intensively studied recently, but relatively little research has focused on the mycobiome and virome. This up-to-date review retrospectively summarizes the lung microbiome's history, composition, and function. We focus on the interaction of the lung microbiome with the oropharynx and gut microbiome and emphasize the role it plays in the innate and adaptive immune responses. More importantly, we focus on multiple respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), fibrosis, bronchiectasis, and pneumonia. The impact of the lung microbiome on coronavirus disease 2019 (COVID-19) and lung cancer has also been comprehensively studied. Furthermore, by summarizing the therapeutic potential of the lung microbiome in lung diseases and examining the shortcomings of the field, we propose an outlook of the direction of lung microbiome research.
Collapse
Affiliation(s)
- Ruomeng Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Xikun Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
46
|
Borrego-Ruiz A, Borrego JJ. An updated overview on the relationship between human gut microbiome dysbiosis and psychiatric and psychological disorders. Prog Neuropsychopharmacol Biol Psychiatry 2024; 128:110861. [PMID: 37690584 DOI: 10.1016/j.pnpbp.2023.110861] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/06/2023] [Accepted: 09/06/2023] [Indexed: 09/12/2023]
Abstract
There is a lot of evidence establishing that nervous system development is related to the composition and functions of the gut microbiome. In addition, the central nervous system (CNS) controls the imbalance of the intestinal microbiota, constituting a bidirectional communication system. At present, various gut-brain crosstalk routes have been described, including immune, endocrine and neural circuits via the vagal pathway. Several empirical data have associated gut microbiota alterations (dysbiosis) with neuropsychiatric diseases, such as Alzheimer's disease, autism and Parkinson's disease, and with other psychological disorders, like anxiety and depression. Fecal microbiota transplantation (FMT) therapy has shown that the gut microbiota can transfer behavioral features to recipient animals, which provides strong evidence to establish a causal-effect relationship. Interventions, based on prebiotics, probiotics or synbiotics, have demonstrated an important influence of microbiota on neurological disorders by the synthesis of neuroactive compounds that interact with the nervous system and by the regulation of inflammatory and endocrine processes. Further research is needed to demonstrate the influence of gut microbiota dysbiosis on psychiatric and psychological disorders, and how microbiota-based interventions may be used as potential therapeutic tools.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Facultad de Psicología, UNED, Madrid, Spain
| | - Juan J Borrego
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain.
| |
Collapse
|
47
|
Doan HT, Cheng LC, Chiu YL, Cheng YK, Hsu CC, Chen YC, Lo HJ, Chiang HS. Candida tropicalis-derived vitamin B3 exerts protective effects against intestinal inflammation by promoting IL-17A/IL-22-dependent epithelial barrier function. Gut Microbes 2024; 16:2416922. [PMID: 39462273 PMCID: PMC11524206 DOI: 10.1080/19490976.2024.2416922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/16/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024] Open
Abstract
Candida tropicalis-a prevalent gut commensal fungus in healthy individuals - contributes to intestinal health and disease. However, how commensal C. tropicalis influences intestinal homeostasis and barrier function is poorly understood. Here, we demonstrated that the reference strain of C. tropicalis (MYA-3404) ameliorated intestinal inflammation in murine models of chemically induced colitis and bacterial infection. Intestinal colonization of C. tropicalis robustly upregulated the expression of IL-17A and IL-22 to increase barrier function and promote proliferation of intestinal epithelial cells in the mouse colon. Metabolomics analysis of fecal samples from mice colonized with C. tropicalis revealed alterations in vitamin B3 metabolism, promoting conversion of nicotinamide to nicotinic acid. Although nicotinamide worsened colitis, treatment with nicotinic acid alleviated disease symptoms and enhanced epithelial proliferation and Th17 cell differentiation. Oral gavage of C. tropicalis mitigated nicotinamide-induced intestinal dysfunction in experimental colitis. Blockade of nicotinic acid production with nicotinamidase inhibitors lowered the protective effects against colitis in mice treated with C. tropicalis. Notably, a clinical C. tropicalis strain isolated from patients with candidemia lacked the protective effects against murine colitis observed with the reference strain. Together, our results highlight a novel role for C. tropicalis in resolving intestinal inflammation through the modulation of vitamin B3 metabolism.
Collapse
Affiliation(s)
- Ha T Doan
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Li-Chieh Cheng
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yi-Ling Chiu
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yuan-Kai Cheng
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
- Leeuwenhoek Laboratories Co. Ltd, Taipei, Taiwan
| | - Yee-Chun Chen
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsiu-Jung Lo
- National Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Hao-Sen Chiang
- Department of Life Science, National Taiwan University, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
48
|
Özdirik B, Schnabl B. Microbial Players in Primary Sclerosing Cholangitis: Current Evidence and Concepts. Cell Mol Gastroenterol Hepatol 2023; 17:423-438. [PMID: 38109970 PMCID: PMC10837305 DOI: 10.1016/j.jcmgh.2023.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023]
Abstract
Primary sclerosing cholangitis (PSC) is a rare cholestatic liver disease with progressive biliary inflammation, destruction of the biliary tract, and fibrosis, resulting in liver cirrhosis and end-stage liver disease. To date, liver transplantation is the only definitive treatment option for PSC. The precise etiology of PSC remains elusive, but it is widely accepted to involve a complex interplay between genetic predisposition, immunologic dysfunction, and environmental influence. In recent years, the gut-liver axis has emerged as a crucial pathway contributing to the pathogenesis of PSC, with particular focus on the role of gut microbiota. However, the role of the fungal microbiome or mycobiome has been overlooked for years, resulting in a lack of comprehensive studies on its involvement in PSC. In this review, we clarify the present clinical and mechanistic data and concepts concerning the gut bacterial and fungal microbiota in the context of PSC. This review sheds light on the role of specific microbes and elucidates the dynamics of bacterial and fungal populations. Moreover, we discuss the latest insights into microbe-altering therapeutic approaches involving the gut-liver axis and bile acid metabolism.
Collapse
Affiliation(s)
- Burcin Özdirik
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California; Department of Medicine, VA San Diego Healthcare System, San Diego, California.
| |
Collapse
|
49
|
Zheng R, Xiang X, Shi Y, Qiu A, Luo X, Xie J, Russell R, Zhang D. Chronic jet lag alters gut microbiome and mycobiome and promotes the progression of MAFLD in HFHFD-fed mice. Front Microbiol 2023; 14:1295869. [PMID: 38130943 PMCID: PMC10733492 DOI: 10.3389/fmicb.2023.1295869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is the most common chronic liver disease worldwide. Circadian disruptors, such as chronic jet lag (CJ), may be new risk factors for MAFLD development. However, the roles of CJ on MAFLD are insufficiently understood, with mechanisms remaining elusive. Studies suggest a link between gut microbiome dysbiosis and MAFLD, but most of the studies are mainly focused on gut bacteria, ignoring other components of gut microbes, such as gut fungi (mycobiome), and few studies have addressed the rhythm of the gut fungi. This study explored the effects of CJ on MAFLD and its related microbiotic and mycobiotic mechanisms in mice fed a high fat and high fructose diet (HFHFD). Forty-eight C57BL6J male mice were divided into four groups: mice on a normal diet exposed to a normal circadian cycle (ND-NC), mice on a normal diet subjected to CJ (ND-CJ), mice on a HFHFD exposed to a normal circadian cycle (HFHFD-NC), and mice on a HFHFD subjected to CJ (HFHFD-CJ). After 16 weeks, the composition and rhythm of microbiota and mycobiome in colon contents were compared among groups. The results showed that CJ exacerbated hepatic steatohepatitis in the HFHFD-fed mice. Compared with HFHFD-NC mice, HFHFD-CJ mice had increases in Aspergillus, Blumeria and lower abundances of Akkermansia, Lactococcus, Prevotella, Clostridium, Bifidobacterium, Wickerhamomyces, and Saccharomycopsis genera. The fungi-bacterial interaction network became more complex after HFHFD and/or CJ interventions. The study revealed that CJ altered the composition and structure of the gut bacteria and fungi, disrupted the rhythmic oscillation of the gut microbiota and mycobiome, affected interactions among the gut microbiome, and promoted the progression of MAFLD in HFHFD mice.
Collapse
Affiliation(s)
- Ruoyi Zheng
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Changsha, China
| | - Xingwei Xiang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Shi
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Anqi Qiu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Luo
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junyan Xie
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ryan Russell
- Department of Health and Human Performance, College of Health Professions, University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Dongmei Zhang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center for Obesity and its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
50
|
Van Syoc E, Nixon MP, Silverman JD, Luo Y, Gonzalez FJ, Elbere I, Klovins J, Patterson AD, Rogers CJ, Ganda E. Changes in the Type 2 diabetes gut mycobiome associate with metformin treatment across populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.25.542255. [PMID: 37398234 PMCID: PMC10312434 DOI: 10.1101/2023.05.25.542255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The human gut teems with a diverse ecosystem of microbes, yet non-bacterial portions of that community are overlooked in studies of metabolic diseases firmly linked to gut bacteria. Type 2 diabetes mellitus (T2D) associates with compositional shifts in the gut bacterial microbiome and fungal mycobiome, but whether T2D and/or pharmaceutical treatments underpin the community change is unresolved. To differentiate these effects, we curated a gut mycobiome cohort to-date spanning 1,000 human samples across 5 countries and a murine experimental model. We use Bayesian multinomial logistic normal models to show that metformin and T2D both associate with shifts in the relative abundance of distinct gut fungi. T2D associates with shifts in the Saccharomycetes and Sordariomycetes fungal classes, while the genera Fusarium and Tetrapisipora most consistently associate with metformin treatment. We confirmed the impact of metformin on individual gut fungi by administering metformin to healthy mice. Thus, metformin and T2D account for subtle, but significant and distinct variation in the gut mycobiome across human populations. This work highlights for the first time that oral pharmaceuticals can confound associations of gut fungi with T2D and warrants the need to consider pharmaceutical interventions in investigations of linkages between metabolic diseases and gut microbial inhabitants.
Collapse
Affiliation(s)
- Emily Van Syoc
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Animal Science, The Pennsylvania State University, University Park, PA 16802, USA
- One Health Microbiome Center, The Pennsylvania State University, University Park, PA 16802, USA
| | - Michelle Pistner Nixon
- College of Information Sciences and Technology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Justin D. Silverman
- One Health Microbiome Center, The Pennsylvania State University, University Park, PA 16802, USA
- College of Information Sciences and Technology, The Pennsylvania State University, University Park, PA 16802, USA
- Departments of Statistics and Medicine, The Pennsylvania State University, University Park, PA 16802, USA
| | - Yuhong Luo
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Frank J. Gonzalez
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ilze Elbere
- Latvian Biomedical Research and Study Center, Riga, Latvia
| | - Janis Klovins
- Latvian Biomedical Research and Study Center, Riga, Latvia
| | - Andrew D. Patterson
- One Health Microbiome Center, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Connie J. Rogers
- Department of Nutritional Sciences, University of Georgia, Athens, GA 30602, USA
| | - Erika Ganda
- Department of Animal Science, The Pennsylvania State University, University Park, PA 16802, USA
- One Health Microbiome Center, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|