1
|
Quinn-Bohmann N, Carr AV, Diener C, Gibbons SM. Moving from genome-scale to community-scale metabolic models for the human gut microbiome. Nat Microbiol 2025; 10:1055-1066. [PMID: 40217129 DOI: 10.1038/s41564-025-01972-2] [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: 07/14/2023] [Accepted: 02/26/2025] [Indexed: 05/08/2025]
Abstract
Metabolic models of individual microorganisms or small microbial consortia have become standard research tools in the bioengineering and systems biology fields. However, extending metabolic modelling to diverse microbial communities, such as those in the human gut, remains a practical challenge from both modelling and experimental validation perspectives. In complex communities, metabolic models accounting for community dynamics, or those that consider multiple objectives, may provide optimal predictions over simpler steady-state models, but require a much higher computational cost. Here we describe some of the strengths and limitations of microbial community-scale metabolic models and argue for a robust validation framework for developing personalized, mechanistic and accurate predictions of microbial community metabolic behaviours across environmental contexts. Ultimately, quantitatively accurate microbial community-scale metabolic models could aid in the design and testing of personalized prebiotic, probiotic and dietary interventions that optimize for translationally relevant outcomes.
Collapse
Affiliation(s)
- Nick Quinn-Bohmann
- Institute for Systems Biology, Seattle, WA, USA
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA, USA
| | - Alex V Carr
- Institute for Systems Biology, Seattle, WA, USA
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA, USA
| | - Christian Diener
- Institute for Systems Biology, Seattle, WA, USA.
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria.
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, USA.
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- eScience Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
2
|
Wang Z, Wu Q, Shen W, Wan F, He J, Liu L, Tang S, Tan Z. Cooling redistributed endotoxin across different biofluids via modulating the ruminal microbiota and metabolome without altering quorum sensing signal levels in heat-stressed beef bulls. Anim Microbiome 2025; 7:38. [PMID: 40269989 PMCID: PMC12016233 DOI: 10.1186/s42523-025-00400-4] [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: 11/19/2024] [Accepted: 03/25/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Cooling is one of the most common and economical methods to ameliorate heat stress (HS), and it has been discovered to alter the lipopolysaccharide (LPS) endotoxin level in ruminants. However, whether the endotoxin variation induced by cooling relates to the quorum sensing (QS) within the ruminal microflora remains unknown. The current study was consequently performed to examine whether cooling could influence the endotoxin distribution across different biofluids, ruminal microbiota, and ruminal metabolisms through affecting the QS of rumen microorganisms in beef cattle exposed to HS. Thirty-two Simmental bulls were used as experimental animals and randomly assigned to either the control (CON) group, or the mechanical ventilation and water spray (MVWS) treatment. The temperature-humidity index (THI) was recorded throughout this trial, and samples of the rumen liquid, blood, and urine were collected. RESULTS Cooling significantly lowered (P < 0.05) the temperature-humidity index (THI), ruminal endotoxin, and endotoxin concentration and excretion in urine, and significantly raised endotoxin level in blood (P < 0.05), but did not change the ruminal concentrations of QS signals including 3-OXO-C6-HSL and the AI-2 (P > 0.05). The linear discriminant analysis effect size (LEfSe) analysis revealed that Prevotellaceae, Rikenellaceae, Monoglobales and their affiliated members, as well as other bacterial taxa were significantly differently (P < 0.05) enriched between the two treatments. The Tax4Fun2 prediction suggested that QS function was upregulated in MVWS compared to CON. The metabolomic analysis indicated that cooling altered the ruminal metabolism profile and downregulated the pathways of lysine degradation, phenylalanine, tyrosine and tryptophan biosynthesis, and ubiquinone and other terpenoid-quinone biosynthesis. The significant (P < 0.05) correlations of the differential bacteria and metabolites with endotoxin and QS molecules were also demonstrated through Spearman analysis. CONCLUSIONS Based on the results of this trial, it could be speculated that the cooling reshaped the endotoxin distribution across different biofluids through manipulating ruminal microbiota and metabolome, which might involve the participation of QS. Further investigations are warranted to disclose and verify the mechanisms for those correlations found in this study.
Collapse
Affiliation(s)
- Zuo Wang
- Yuelushan Laboratory, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, People's Republic of China
| | - Qingyang Wu
- Yuelushan Laboratory, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, People's Republic of China
| | - Weijun Shen
- Yuelushan Laboratory, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, People's Republic of China.
| | - Fachun Wan
- Yuelushan Laboratory, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, People's Republic of China
| | - Jianhua He
- Yuelushan Laboratory, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410128, People's Republic of China
| | - Lei Liu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, 410128, People's Republic of China
| | - Shaoxun Tang
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, People's Republic of China
| | - Zhiliang Tan
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, People's Republic of China
| |
Collapse
|
3
|
Zheng L, Xin J, Ye H, Sun N, Gan B, Gong X, Bao S, Xiang M, Wang H, Ni X, Li H, Zhang T. Lactobacillus Johnsonii YH1136 alleviates schizophrenia-like behavior in mice: a gut-microbiota-brain axis hypothesis study. BMC Microbiol 2025; 25:191. [PMID: 40175911 PMCID: PMC11963707 DOI: 10.1186/s12866-025-03893-w] [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: 10/29/2024] [Accepted: 03/14/2025] [Indexed: 04/04/2025] Open
Abstract
Based on the microbiota-gut-brain axis (MGBA) hypothesis, probiotics play an increasingly important role in treating various psychiatric disorders. Schizophrenia (SCZ) is a common mental disease with a complex pathogenesis and is challenging to treat. Although studies have elucidated the mechanisms associated with the interactions between the microbiota-gut-brain axis and SCZ, few have specifically used probiotics as a therapeutic intervention for SCZ. Accordingly, the current study determines whether L. johnsonii YH1136 effectively prevents SCZ-like behavior in mice and identifies the associated key microbes and metabolites. An SCZ mouse model was established by intraperitoneal injection of MK-801; L. johnsonii YH1136 was administered via oral gavage. L. johnsonii YH1136 significantly improves abnormal behaviors, including psychomotor hyperactivity and sociability and alleviates aberrant enzyme expression associated with tryptophan metabolism in SCZ mice. Additionally, L. johnsonii YH1136 upregulates hippocampal brain-derived neurotrophic factor (BDNF) levels while downregulating tryptophan 2,3-dioxygenase (TDO2), indoleamine-pyrrole 2,3-dioxygenase 1 (IDO1), kynurenine aminotransferase 1 (KAT1). Subsequent 16S rRNA sequencing of intestinal contents suggests that L. johnsonii YH1136 modulates the gut flora structure and composition by increasing the relative abundance of Lactobacillus and decreasing Dubosiella in SCZ mice. N-acetylneuraminic acid and hypoxanthine are the key serum metabolites mediating the interaction between the MGBA and SCZ. These results partially reveal the mechanism underlying the effects of L. johnsonii YH1136 on SCZ-like behavior in mice, supporting the development of therapeutic L. johnsonii probiotic formulations against SCZ.
Collapse
Affiliation(s)
- Liqin Zheng
- School of Life Science and Technology, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Lab for Neuroinformation, Sichuan Institute for Brain Science and Brain-Inspired Intelligence, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, West Hi-Tech Zone, Chengdu , Sichuan, 611731, China
| | - Jinge Xin
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huiqian Ye
- The Fourth People's Hospital of Ya'an, 7 Qingxi Road Ya'an 625000, Yucheng ZoneSichuan, China
| | - Ning Sun
- Animal Microecology Institute College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Baoxing Gan
- Animal Microecology Institute College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xuemei Gong
- Animal Microecology Institute College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shusheng Bao
- School of Life Science and Technology, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Lab for Neuroinformation, Sichuan Institute for Brain Science and Brain-Inspired Intelligence, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, West Hi-Tech Zone, Chengdu , Sichuan, 611731, China
| | - Min Xiang
- The Fourth People's Hospital of Ya'an, 7 Qingxi Road Ya'an 625000, Yucheng ZoneSichuan, China
| | - Hesong Wang
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xueqin Ni
- Animal Microecology Institute College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hao Li
- The Fourth People's Hospital of Ya'an, 7 Qingxi Road Ya'an 625000, Yucheng ZoneSichuan, China.
| | - Tao Zhang
- School of Life Science and Technology, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China.
- MOE Key Lab for Neuroinformation, Sichuan Institute for Brain Science and Brain-Inspired Intelligence, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, West Hi-Tech Zone, Chengdu , Sichuan, 611731, China.
| |
Collapse
|
4
|
Yang SY, Han SM, Lee JY, Kim KS, Lee JE, Lee DW. Advancing Gut Microbiome Research: The Shift from Metagenomics to Multi-Omics and Future Perspectives. J Microbiol Biotechnol 2025; 35:e2412001. [PMID: 40223273 PMCID: PMC12010094 DOI: 10.4014/jmb.2412.12001] [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: 12/02/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 04/15/2025]
Abstract
The gut microbiome, a dynamic and integral component of human health, has co-evolved with its host, playing essential roles in metabolism, immunity, and disease prevention. Traditional microbiome studies, primarily focused on microbial composition, have provided limited insights into the functional and mechanistic interactions between microbiota and their host. The advent of multi-omics technologies has transformed microbiome research by integrating genomics, transcriptomics, proteomics, and metabolomics, offering a comprehensive, systems-level understanding of microbial ecology and host-microbiome interactions. These advances have propelled innovations in personalized medicine, enabling more precise diagnostics and targeted therapeutic strategies. This review highlights recent breakthroughs in microbiome research, demonstrating how these approaches have elucidated microbial functions and their implications for health and disease. Additionally, it underscores the necessity of standardizing multi-omics methodologies, conducting large-scale cohort studies, and developing novel platforms for mechanistic studies, which are critical steps toward translating microbiome research into clinical applications and advancing precision medicine.
Collapse
Affiliation(s)
- So-Yeon Yang
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung Min Han
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Ji-Young Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Kyoung Su Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jae-Eun Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Dong-Woo Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
5
|
Hodgkiss R, Acharjee A. Unravelling metabolite-microbiome interactions in inflammatory bowel disease through AI and interaction-based modelling. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167618. [PMID: 39662756 DOI: 10.1016/j.bbadis.2024.167618] [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: 09/20/2024] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
Inflammatory Bowel Diseases (IBDs) are chronic inflammatory disorders of the gastrointestinal tract and colon affecting approximately 7 million individuals worldwide. The knowledge of specific pathology and aetiological mechanisms leading to IBD is limited, however a reduced immune system, antibiotic use and reserved diet may initiate symptoms. Dysbiosis of the gut microbiome, and consequently a varied composition of the metabolome, has been extensively linked to these risk factors and IBD. Metagenomic sequencing and liquid-chromatography mass spectrometry (LC-MS) of N = 220 fecal samples by Fransoza et al., provided abundance data on microbial genera and metabolites for use in this study. Identification of differentially abundant microbes and metabolites was performed using a Wilcoxon test, followed by feature selection of random forest (RF), gradient-boosting (XGBoost) and least absolute shrinkage operator (LASSO) models. The performance of these features was then validated using RF models on the Human Microbiome Project 2 (HMP2) dataset and a microbial community (MICOM) model was utilised to predict and interpret the interactions between key microbes and metabolites. The Flavronifractor genus and microbes of the families Lachnospiraceae and Oscillospiraceae were found differential by all models. Metabolic pathways commonly influenced by such microbes in IBD were CoA biosynthesis, bile acid metabolism and amino acid production and degradation. This study highlights distinct interactive microbiome and metabolome profiles within IBD and the highly potential pathways causing disease pathology. It therefore paves way for future investigation into new therapeutic targets and non-invasive diagnostic tools for IBD.
Collapse
Affiliation(s)
- Rebecca Hodgkiss
- College of Medicine and Health, Cancer and Genomic Sciences, University of Birmingham, B15 2TT Birmingham, UK
| | - Animesh Acharjee
- College of Medicine and Health, Cancer and Genomic Sciences, University of Birmingham, B15 2TT Birmingham, UK; Institute of Translational Medicine, University Hospitals Birmingham NHS Foundation Trust, B15 2TT Birmingham, UK; MRC Health Data Research UK (HDR), Midlands Site, UK; Centre for Health Data Research, University of Birmingham, B15 2TT, UK.
| |
Collapse
|
6
|
Gutierrez MW, van Tilburg Bernardes E, Ren E, Kalbfleisch KN, Day M, Lameu EL, Glatthardt T, Mercer EM, Sharma S, Zhang H, Al-Azawy A, Chleilat F, Hirota SA, Reimer RA, Arrieta MC. Early-life gut mycobiome core species modulate metabolic health in mice. Nat Commun 2025; 16:1467. [PMID: 39922818 PMCID: PMC11807121 DOI: 10.1038/s41467-025-56743-8] [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: 02/12/2024] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
The gut microbiome causally contributes to obesity; however, the role of fungi remains understudied. We previously identified three core species of the infant gut mycobiome (Rhodotorula mucilaginosa, Malassezia restricta and Candida albicans) that correlated with body mass index, however their causal contributions to obesity development are unknown. Here we show the effects of early-life colonization by these fungal species on metabolic health in gnotobiotic mice fed standard (SD) or high-fat-high-sucrose (HFHS) diets. Each species resulted in bacterial microbiome compositional and functional differences. R. mucilaginosa and M. restricta increased adiposity in mice fed SD, while only R. mucilaginosa exacerbated metabolic disease. In contrast, C. albicans resulted in leanness and resistance to diet-induced obesity. Intestinal nutrient transporter expression was unaffected by the presence of fungi in jejunal enteroids, yet the immune landscape in white adipose tissue was distinctly impacted by each fungal species, suggesting that these phenotypes may be a result of fungal immune regulation. This work revealed that three common fungal colonizers have distinct causal influences on obesity and metabolic inflammation and justifies the consideration of fungi in microbiome research on host metabolism.
Collapse
Affiliation(s)
- Mackenzie W Gutierrez
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Erik van Tilburg Bernardes
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Ellen Ren
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Kristen N Kalbfleisch
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Madeline Day
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Ewandson Luiz Lameu
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Thaís Glatthardt
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Emily M Mercer
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Sunita Sharma
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Hong Zhang
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Ali Al-Azawy
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Faye Chleilat
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Simon A Hirota
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Raylene A Reimer
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Marie-Claire Arrieta
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada.
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada.
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
7
|
Pogreba Brown K, Austhof E, McFadden CM, Scranton C, Sun X, Vujkovic-Cviji I, Rodriguez D, Falk L, Heslin KM, Arani G, Obergh V, Bessey K, Cooper K. Determining the incidence, risk factors and biological drivers of irritable bowel syndrome (IBS) as part of the constellation of postacute sequelae of SARS-CoV-2 infection (PASC) outcomes in the Arizona CoVHORT-GI: a longitudinal cohort study. BMJ Open 2025; 15:e095093. [PMID: 39890144 PMCID: PMC11784208 DOI: 10.1136/bmjopen-2024-095093] [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: 10/15/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025] Open
Abstract
INTRODUCTION Postacute sequelae of SARS-CoV-2 infection (PASC) are extensive. Also known as long COVID, primary outcomes reported are neurologic, cardiac and respiratory in nature. However, several studies have also reported an increase in gastrointestinal (GI) symptoms and syndromes following COVID-19. This study of PASC will include extensive analyses of GI symptoms, determine if people with pre-existing irritable bowel syndrome (IBS) are at higher risk of developing PASC generally or PASC-GI, and which biomarkers are impacted and to what degree. This R01 study is being funded by the National Institute of Diabetes and Digestive and Kidney Diseases (1R01DK135483-01) from 2023 to 2028. METHODS AND ANALYSES This study combines a longitudinal epidemiologic cohort study and in-depth, novel biologic analyses. In collaboration with a pre-existing study, the Arizona CoVID-19 Cohort (CoVHORT)-GI will recruit participants based on the history of COVID infection(s), new or ongoing GI symptoms 3-6 months postinfection, and pre-existing or incident IBS diagnosis to represent five study groups for comparison and analyses. A subset (n=1000) of those recruited will submit both stool and blood samples. Both samples will undergo a novel method to quantitate humoral and mucosal immune responses to host-derived faecal communities in conjunction with magnetic bead-based separation and high-depth shotgun microbial sequencing. Stool samples will also undergo traditional microbiome analyses (diversity and abundance) and faecal calprotectin assays. Additional serum analyses will aim to determine if a proteomics-based signature exists that differentiates a unique biomarker compositional signature discriminating PASC-GI versus no PASC. All laboratory data will be linked with in-depth epidemiologic data on demographics, symptoms and chronic conditions. ETHICS AND DISSEMINATION This study involves human participants and was approved by the University of Arizona Institutional Review Board (IRB (#00002332) and has been deemed minimal risk. Participants gave informed consent to participate in the study before taking part. All publications from the study will be shared back to participants along with alternative lay summaries and webinars to communicate key findings. The data management plan has been published and is publicly available online, including protocols for data requests.
Collapse
Affiliation(s)
- Kristen Pogreba Brown
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Erika Austhof
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Caitlyn M McFadden
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Caroline Scranton
- School of Animal and Comparative Biomedical Sciences, The University of Arizona College of Agriculture and Life Sciences, Tucson, Arizona, USA
| | - Xiaoxiao Sun
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Ivan Vujkovic-Cviji
- Department of Biomedical Sciences; Research Division of Immunology, Department of Medicine, Karsh Division of Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dominic Rodriguez
- School of Animal and Comparative Biomedical Sciences, The University of Arizona College of Agriculture and Life Sciences, Tucson, Arizona, USA
| | - Laura Falk
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Kelly M Heslin
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Gayatri Arani
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona, Arizona Health Sciences Center, Tucson, Arizona, USA
| | - Victoria Obergh
- School of Animal and Comparative Biomedical Sciences, The University of Arizona College of Agriculture and Life Sciences, Tucson, Arizona, USA
| | - Kate Bessey
- Mel and Enid Zuckerman College of Public Health; Department of Epidemiology and Biostatistics, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Kerry Cooper
- School of Animal and Comparative Biomedical Sciences, The University of Arizona College of Agriculture and Life Sciences, Tucson, Arizona, USA
| |
Collapse
|
8
|
Jung S. Advances in functional analysis of the microbiome: Integrating metabolic modeling, metabolite prediction, and pathway inference with Next-Generation Sequencing data. J Microbiol 2025; 63:e.2411006. [PMID: 39895076 DOI: 10.71150/jm.2411006] [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: 11/27/2024] [Indexed: 02/04/2025]
Abstract
This review explores current advancements in microbiome functional analysis enabled by next-generation sequencing technologies, which have transformed our understanding of microbial communities from mere taxonomic composition to their functional potential. We examine approaches that move beyond species identification to characterize microbial activities, interactions, and their roles in host health and disease. Genome-scale metabolic models allow for in-depth simulations of metabolic networks, enabling researchers to predict microbial metabolism, growth, and interspecies interactions in diverse environments. Additionally, computational methods for predicting metabolite profiles offer indirect insights into microbial metabolic outputs, which is crucial for identifying biomarkers and potential therapeutic targets. Functional pathway analysis tools further reveal microbial contributions to metabolic pathways, highlighting alterations in response to environmental changes and disease states. Together, these methods offer a powerful framework for understanding the complex metabolic interactions within microbial communities and their impact on host physiology. While significant progress has been made, challenges remain in the accuracy of predictive models and the completeness of reference databases, which limit the applicability of these methods in under-characterized ecosystems. The integration of these computational tools with multi-omic data holds promise for personalized approaches in precision medicine, allowing for targeted interventions that modulate the microbiome to improve health outcomes. This review highlights recent advances in microbiome functional analysis, providing a roadmap for future research and translational applications in human health and environmental microbiology.
Collapse
Affiliation(s)
- Sungwon Jung
- Department of Genome Medicine and Science, Gachon University College of Medicine, Incheon 21565, Republic of Korea
- Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon 21565, Republic of Korea
| |
Collapse
|
9
|
Mildau K, Ehlers H, Meisenburg M, Del Pup E, Koetsier RA, Torres Ortega LR, de Jonge NF, Singh KS, Ferreira D, Othibeng K, Tugizimana F, Huber F, van der Hooft JJJ. Effective data visualization strategies in untargeted metabolomics. Nat Prod Rep 2024. [PMID: 39620439 PMCID: PMC11610048 DOI: 10.1039/d4np00039k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Indexed: 12/11/2024]
Abstract
Covering: 2014 to 2023 for metabolomics, 2002 to 2023 for information visualizationLC-MS/MS-based untargeted metabolomics is a rapidly developing research field spawning increasing numbers of computational metabolomics tools assisting researchers with their complex data processing, analysis, and interpretation tasks. In this article, we review the entire untargeted metabolomics workflow from the perspective of information visualization, visual analytics and visual data integration. Data visualization is a crucial step at every stage of the metabolomics workflow, where it provides core components of data inspection, evaluation, and sharing capabilities. However, due to the large number of available data analysis tools and corresponding visualization components, it is hard for both users and developers to get an overview of what is already available and which tools are suitable for their analysis. In addition, there is little cross-pollination between the fields of data visualization and metabolomics, leaving visual tools to be designed in a secondary and mostly ad hoc fashion. With this review, we aim to bridge the gap between the fields of untargeted metabolomics and data visualization. First, we introduce data visualization to the untargeted metabolomics field as a topic worthy of its own dedicated research, and provide a primer on cutting-edge visualization research into data visualization for both researchers as well as developers active in metabolomics. We extend this primer with a discussion of best practices for data visualization as they have emerged from data visualization studies. Second, we provide a practical roadmap to the visual tool landscape and its use within the untargeted metabolomics field. Here, for several computational analysis stages within the untargeted metabolomics workflow, we provide an overview of commonly used visual strategies with practical examples. In this context, we will also outline promising areas for further research and development. We end the review with a set of recommendations for developers and users on how to make the best use of visualizations for more effective and transparent communication of results.
Collapse
Affiliation(s)
- Kevin Mildau
- Bioinformatics Group, Wageningen University & Research, Wageningen, The Netherlands.
| | - Henry Ehlers
- Visualization Group, Institute of Visual Computing and Human-Centered Technology, TU Wien, Vienna, Austria.
| | - Mara Meisenburg
- Adaptation Physiology Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Elena Del Pup
- Bioinformatics Group, Wageningen University & Research, Wageningen, The Netherlands.
| | - Robert A Koetsier
- Bioinformatics Group, Wageningen University & Research, Wageningen, The Netherlands.
| | | | - Niek F de Jonge
- Bioinformatics Group, Wageningen University & Research, Wageningen, The Netherlands.
| | - Kumar Saurabh Singh
- Bioinformatics Group, Wageningen University & Research, Wageningen, The Netherlands.
- Maastricht University Faculty of Science and Engineering, Plant Functional Genomics Maastricht, Limburg, The Netherlands
- Faculty of Environment, Science and Economy, University of Exeter, Penryl Cornwall, UK
| | | | - Kgalaletso Othibeng
- Department of Biochemistry, University of Johannesburg, Johannesburg, South Africa
| | - Fidele Tugizimana
- Department of Biochemistry, University of Johannesburg, Johannesburg, South Africa
| | - Florian Huber
- Centre for Digitalisation and Digitality, Düsseldorf University of Applied Sciences, Düsseldorf, Germany
| | - Justin J J van der Hooft
- Bioinformatics Group, Wageningen University & Research, Wageningen, The Netherlands.
- Department of Biochemistry, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
10
|
Salihoglu R, Balkenhol J, Dandekar G, Liang C, Dandekar T, Bencurova E. Cat-E: A comprehensive web tool for exploring cancer targeting strategies. Comput Struct Biotechnol J 2024; 23:1376-1386. [PMID: 38596315 PMCID: PMC11001601 DOI: 10.1016/j.csbj.2024.03.024] [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: 01/27/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/11/2024] Open
Abstract
Identifying potential cancer-associated genes and drug targets from omics data is challenging due to its diverse sources and analyses, requiring advanced skills and large amounts of time. To facilitate such analysis, we developed Cat-E (Cancer Target Explorer), a novel R/Shiny web tool designed for comprehensive analysis with evaluation according to cancer-related omics data. Cat-E is accessible at https://cat-e.bioinfo-wuerz.eu/. Cat-E compiles information on oncolytic viruses, cell lines, gene markers, and clinical studies by integrating molecular datasets from key databases such as OvirusTB, TCGA, DrugBANK, and PubChem. Users can use all datasets and upload their data to perform multiple analyses, such as differential gene expression analysis, metabolic pathway exploration, metabolic flux analysis, GO and KEGG enrichment analysis, survival analysis, immune signature analysis, single nucleotide variation analysis, dynamic analysis of gene expression changes and gene regulatory network changes, and protein structure prediction. Cancer target evaluation by Cat-E is demonstrated here on lung adenocarcinoma (LUAD) datasets. By offering a user-friendly interface and detailed user manual, Cat-E eliminates the need for advanced computational expertise, making it accessible to experimental biologists, undergraduate and graduate students, and oncology clinicians. It serves as a valuable tool for investigating genetic variations across diverse cancer types, facilitating the identification of novel diagnostic markers and potential therapeutic targets.
Collapse
Affiliation(s)
- Rana Salihoglu
- Department of Bioinformatics, University of Wurzburg, 97074 Wurzburg, Germany
| | - Johannes Balkenhol
- Department of Bioinformatics, University of Wurzburg, 97074 Wurzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University Hospital of Wurzburg, 97080 Wurzburg, Germany
| | - Gudrun Dandekar
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital of Wurzburg, 97080 Wurzburg, Germany
| | - Chunguang Liang
- Department of Bioinformatics, University of Wurzburg, 97074 Wurzburg, Germany
- Institute of Immunology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, University of Wurzburg, 97074 Wurzburg, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Elena Bencurova
- Department of Bioinformatics, University of Wurzburg, 97074 Wurzburg, Germany
| |
Collapse
|
11
|
Mihailovich M, Tolinački M, Soković Bajić S, Lestarevic S, Pejovic-Milovancevic M, Golić N. The Microbiome-Genetics Axis in Autism Spectrum Disorders: A Probiotic Perspective. Int J Mol Sci 2024; 25:12407. [PMID: 39596472 PMCID: PMC11594817 DOI: 10.3390/ijms252212407] [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: 10/23/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Autism spectrum disorder (commonly known as autism) is a complex and prevalent neurodevelopmental condition characterized by challenges in social behavior, restricted interests, and repetitive behaviors. It is projected that the annual cost of autism spectrum disorder in the US will reach USD 461 billion by 2025. However, despite being a major public health problem, effective treatment for the underlying symptoms remains elusive. As numerous literature data indicate the role of gut microbiota in autism prognosis, particularly in terms of alleviating gastrointestinal (GI) symptoms, high hopes have been placed on probiotics for autism treatment. Approximately twenty clinical studies have been conducted using single or mixed probiotic cultures. However, unequivocal results on the effect of probiotics on people with autism have not been obtained. The small sample sizes, differences in age of participants, choice of probiotics, dose and duration of treatment, outcome measures, and analytical methods used are largely inconsistent, making it challenging to draw distinctive conclusions. Here, we discuss the experimental evidence for specific gut bacteria and their metabolites and how they affect autism in light of the phenotypic and etiological complexity and heterogeneity. We propose a personalized medicine approach for using probiotics to increase the quality of life of individuals with autism by selecting specific probiotics to improve particular features of the condition.
Collapse
Affiliation(s)
- Marija Mihailovich
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, 11042 Belgrade, Serbia; (M.T.); (S.S.B.); (N.G.)
- Human Technopole, 20157 Milan, Italy
| | - Maja Tolinački
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, 11042 Belgrade, Serbia; (M.T.); (S.S.B.); (N.G.)
| | - Svetlana Soković Bajić
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, 11042 Belgrade, Serbia; (M.T.); (S.S.B.); (N.G.)
| | - Sanja Lestarevic
- Institute of Mental Health, 11000 Belgrade, Serbia; (S.L.); (M.P.-M.)
| | - Milica Pejovic-Milovancevic
- Institute of Mental Health, 11000 Belgrade, Serbia; (S.L.); (M.P.-M.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Nataša Golić
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, 11042 Belgrade, Serbia; (M.T.); (S.S.B.); (N.G.)
| |
Collapse
|
12
|
Zulfiqar M, Singh V, Steinbeck C, Sorokina M. Review on computer-assisted biosynthetic capacities elucidation to assess metabolic interactions and communication within microbial communities. Crit Rev Microbiol 2024; 50:1053-1092. [PMID: 38270170 DOI: 10.1080/1040841x.2024.2306465] [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: 03/13/2023] [Revised: 11/17/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Microbial communities thrive through interactions and communication, which are challenging to study as most microorganisms are not cultivable. To address this challenge, researchers focus on the extracellular space where communication events occur. Exometabolomics and interactome analysis provide insights into the molecules involved in communication and the dynamics of their interactions. Advances in sequencing technologies and computational methods enable the reconstruction of taxonomic and functional profiles of microbial communities using high-throughput multi-omics data. Network-based approaches, including community flux balance analysis, aim to model molecular interactions within and between communities. Despite these advances, challenges remain in computer-assisted biosynthetic capacities elucidation, requiring continued innovation and collaboration among diverse scientists. This review provides insights into the current state and future directions of computer-assisted biosynthetic capacities elucidation in studying microbial communities.
Collapse
Affiliation(s)
- Mahnoor Zulfiqar
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Vinay Singh
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University, Jena, Germany
| | - Christoph Steinbeck
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Maria Sorokina
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University, Jena, Germany
- Data Science and Artificial Intelligence, Research and Development, Pharmaceuticals, Bayer, Berlin, Germany
| |
Collapse
|
13
|
Vich Vila A, Zhang J, Liu M, Faber KN, Weersma RK. Untargeted faecal metabolomics for the discovery of biomarkers and treatment targets for inflammatory bowel diseases. Gut 2024; 73:1909-1920. [PMID: 39002973 PMCID: PMC11503092 DOI: 10.1136/gutjnl-2023-329969] [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: 05/11/2024] [Accepted: 06/23/2024] [Indexed: 07/15/2024]
Abstract
The gut microbiome has been recognised as a key component in the pathogenesis of inflammatory bowel diseases (IBD), and the wide range of metabolites produced by gut bacteria are an important mechanism by which the human microbiome interacts with host immunity or host metabolism. High-throughput metabolomic profiling and novel computational approaches now allow for comprehensive assessment of thousands of metabolites in diverse biomaterials, including faecal samples. Several groups of metabolites, including short-chain fatty acids, tryptophan metabolites and bile acids, have been associated with IBD. In this Recent Advances article, we describe the contribution of metabolomics research to the field of IBD, with a focus on faecal metabolomics. We discuss the latest findings on the significance of these metabolites for IBD prognosis and therapeutic interventions and offer insights into the future directions of metabolomics research.
Collapse
Affiliation(s)
- Arnau Vich Vila
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Jingwan Zhang
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong (SAR), People's Republic of China
- Microbiota I-Center (MagIC), Hong Kong (SAR), People's Republic of China
| | - Moting Liu
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
14
|
Lin B, Ye Z, Cao Z, Ye Z, Yu Y, Jiang W, Guo S, Melnikov V, Zhou P, Ji C, Shi C, Wu Z, Chen Z, Xu Y, Zhang Q, Ma Z, Qiao N, Chen L, Shou X, Cao X, Zhou X, Zhang L, He M, Wang Y, Ye H, Li Y, Zhang Z, Wang M, Gao R, Zhang Y. Integrated Microbiome and Metabolome Analysis Reveals Hypothalamic-Comorbidities Related Signatures in Craniopharyngioma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400684. [PMID: 39225628 PMCID: PMC11497089 DOI: 10.1002/advs.202400684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/15/2024] [Indexed: 09/04/2024]
Abstract
Craniopharyngioma (CP) is an intracranial tumor with high mortality and morbidity. Though biologically benign, CP will damage the hypothalamus, inducing comorbidities such as obesity, metabolic syndrome, and cognitive impairments. The roles of gut microbiome and serum metabolome in CP-associated hypothalamic comorbidities are aimed to be explored. Patients with CP are characterized by increased Shannon diversity, Eubacterium, Clostridium, and Roseburia, alongside decreased Alistipes and Bacteroides. CP-enriched taxa are positively correlated with dyslipidemia and cognitive decline, while CP-depleted taxa are negatively associated with fatty liver. Subsequent serum metabolomics identified notably up-regulated purine metabolism, and integrative analysis indicated an association between altered microbiota and elevated hypoxanthine. Phenotypic study and multi-omics analysis in the Rax-CreERT2::BrafV600E/+::PtenFlox/+ mouse model validated potential involvement of increased Clostridium and dysregulated purine metabolism in hypothalamic comorbidities. To further consolidate this, intervention experiments are performed and it is found that hypoxanthine co-variated with the severity of hypothalamic comorbidities and abundance of Clostridium, and induced dysregulated purine metabolism along with redox imbalance in target organs (liver and brain cortex). Overall, the study demonstrated the potential of increased Clostridium and up-regulated purine metabolism as signatures of CP-associated hypothalamic-comorbidities, and unveiled that elevated Clostridium, dysregulated purine metabolism, and redox imbalance may mediate the development and progression of CP-associated hypothalamic-comorbidities.
Collapse
|
15
|
Zhang B, Magnaye KM, Stryker E, Moltzau-Anderson J, Porsche CE, Hertz S, McCauley KE, Smith BJ, Zydek M, Pollard KS, Ma A, El-Nachef N, Lynch SV. Sustained mucosal colonization and fecal metabolic dysfunction by Bacteroides associates with fecal microbial transplant failure in ulcerative colitis patients. Sci Rep 2024; 14:18558. [PMID: 39122767 PMCID: PMC11316000 DOI: 10.1038/s41598-024-62463-8] [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: 09/12/2023] [Accepted: 05/16/2024] [Indexed: 08/12/2024] Open
Abstract
Fecal microbial transplantation (FMT) offers promise for treating ulcerative colitis (UC), though the mechanisms underlying treatment failure are unknown. This study harnessed longitudinally collected colonic biopsies (n = 38) and fecal samples (n = 179) from 19 adults with mild-to-moderate UC undergoing serial FMT in which antimicrobial pre-treatment and delivery mode (capsules versus enema) were assessed for clinical response (≥ 3 points decrease from the pre-treatment Mayo score). Colonic biopsies underwent dual RNA-Seq; fecal samples underwent parallel 16S rRNA and shotgun metagenomic sequencing as well as untargeted metabolomic analyses. Pre-FMT, the colonic mucosa of non-responsive (NR) patients harbored an increased burden of bacteria, including Bacteroides, that expressed more antimicrobial resistance genes compared to responsive (R) patients. NR patients also exhibited muted mucosal expression of innate immune antimicrobial response genes. Post-FMT, NR and R fecal microbiomes and metabolomes exhibited significant divergence. NR metabolomes had elevated concentrations of immunostimulatory compounds including sphingomyelins, lysophospholipids and taurine. NR fecal microbiomes were enriched for Bacteroides fragilis and Bacteroides salyersiae strains that encoded genes capable of taurine production. These findings suggest that both effective mucosal microbial clearance and reintroduction of bacteria that reshape luminal metabolism associate with FMT success and that persistent mucosal and fecal colonization by antimicrobial-resistant Bacteroides species may contribute to FMT failure.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Kevin M Magnaye
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- The Benioff Center for Microbiome Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Emily Stryker
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jacqueline Moltzau-Anderson
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- The Benioff Center for Microbiome Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Cara E Porsche
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Sandra Hertz
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
| | - Kathryn E McCauley
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Byron J Smith
- The Gladstone Institutes, Data Science and Biotechnology, San Francisco, CA, 94158, USA
| | - Martin Zydek
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Katherine S Pollard
- The Gladstone Institutes, Data Science and Biotechnology, San Francisco, CA, 94158, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, 94158, USA
- Chan Zuckerberg Biohub, San Francisco, CA University of California, San Francisco, CA, 94158, USA
| | - Averil Ma
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Najwa El-Nachef
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Division of Gastroenterology, Henry Ford Health System, Detroit, MI, 48208, USA
| | - Susan V Lynch
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.
- The Benioff Center for Microbiome Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
16
|
Han C, Shi C, Liu L, Han J, Yang Q, Wang Y, Li X, Fu W, Gao H, Huang H, Zhang X, Yu K. Majorbio Cloud 2024: Update single-cell and multiomics workflows. IMETA 2024; 3:e217. [PMID: 39135689 PMCID: PMC11316920 DOI: 10.1002/imt2.217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 08/15/2024]
Abstract
Majorbio Cloud (https://cloud.majorbio.com/) is a one-stop online analytic platform aiming at promoting the development of bioinformatics services, narrowing the gap between wet and dry experiments, and accelerating the discoveries for the life sciences community. In 2024, three single-omics workflows, two multiomics workflows, and extensions were newly released to facilitate omics data mining and interpretation.
Collapse
Affiliation(s)
- Chang Han
- Shanghai Majorbio Bio‐Pharm Technology Co. Ltd.ShanghaiChina
| | - Caiping Shi
- Shanghai Majorbio Bio‐Pharm Technology Co. Ltd.ShanghaiChina
| | - Linmeng Liu
- Shanghai Majorbio Bio‐Pharm Technology Co. Ltd.ShanghaiChina
| | - Jichen Han
- Shanghai Majorbio Bio‐Pharm Technology Co. Ltd.ShanghaiChina
| | - Qianqian Yang
- Shanghai Majorbio Bio‐Pharm Technology Co. Ltd.ShanghaiChina
| | - Yan Wang
- Shanghai Majorbio Bio‐Pharm Technology Co. Ltd.ShanghaiChina
| | - Xiaodan Li
- Shanghai Majorbio Bio‐Pharm Technology Co. Ltd.ShanghaiChina
| | - Wenyao Fu
- Shanghai Majorbio Bio‐Pharm Technology Co. Ltd.ShanghaiChina
| | - Hao Gao
- Shanghai Majorbio Bio‐Pharm Technology Co. Ltd.ShanghaiChina
| | - Huasheng Huang
- Shanghai Majorbio Bio‐Pharm Technology Co. Ltd.ShanghaiChina
| | - Xianglin Zhang
- Shanghai Majorbio Bio‐Pharm Technology Co. Ltd.ShanghaiChina
| | - Kegang Yu
- Shanghai Majorbio Bio‐Pharm Technology Co. Ltd.ShanghaiChina
| |
Collapse
|
17
|
Wen Y, Chen B, Huang J, Luo Y, Lv S, Qiu H, Li S, Liu S, He L, He M, Yu Z, Zhao M, Yang Q, Li D, Gu C. Konjac supplementation can alleviate obesity induced by high-fat diet in mice by modulating gut microbiota and its metabolites. Curr Res Food Sci 2024; 9:100805. [PMID: 39131951 PMCID: PMC11315163 DOI: 10.1016/j.crfs.2024.100805] [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: 04/24/2024] [Revised: 06/17/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024] Open
Abstract
As a multi-factorial disease, obesity has become one of the major health problems in the world, and it is still increasing rapidly. Konjac supplementation, as a convenient dietary therapy, has been shown to be able to regulate gut microbiota and improve obesity. However, the specific mechanism by which konjac improves obesity through gut microbiota remains to be studied. In this study, a high-fat diet (HFD) was used to induce a mouse obesity model, and 16S rDNA sequencing and an untargeted metabolomics were used to investigate the impact of konjac on gut microbiota and gut metabolites in HFD-induced obese mice. The results show that konjac can reduce the body weight, adipose tissue weight, and lipid level of high-fat diet induced obese mice by changing the gut microbiota structure and gut metabolic profile. Association analysis revealed that konjac supplementation induced changes in gut microbiota, resulting in the up-regulation of 7-dehydrocholesterol and trehalose 6-phosphate, as well as the down-regulation of glycocholic acid and ursocholic acid within the Secondary bile acid biosynthesis pathway, ultimately leading to improvements in obesity. Among them, g_Acinetobacter (Greengene ID: 911888) can promote the synthesis of 7-dehydrocholesterol by synthesizing ERG3. g_Allobaculum (Greengene ID: 271516) and g_Allobaculum (Greengene ID: 259370) can promote the breakdown of trehalose 6-phosphate by synthesizing glvA. Additionally, the down-regulation of glycocholic acid and ursocholic acid may be influenced by the up-regulation of Lachnospiraceae_NK4A136_group. In conclusion, konjac exerts an influence on gut metabolites through the regulation of gut microbiota, thereby playing a pivotal role in alleviating obesity induced by a high-fat diet.
Collapse
Affiliation(s)
- Yuhang Wen
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| | - Baoting Chen
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| | - Jingrong Huang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| | - Yadan Luo
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| | - Shuya Lv
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| | - Hao Qiu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| | - Shuaibing Li
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Songwei Liu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| | - Lvqin He
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| | - Manli He
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| | - Zehui Yu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| | - Mingde Zhao
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| | - Qian Yang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| | - Dong Li
- College of Bioengineering, Sichuan University of Science and Engineering, Yibin, 643002, China
| | - Congwei Gu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| |
Collapse
|
18
|
Noecker C, Turnbaugh PJ. Emerging tools and best practices for studying gut microbial community metabolism. Nat Metab 2024; 6:1225-1236. [PMID: 38961185 DOI: 10.1038/s42255-024-01074-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/30/2024] [Indexed: 07/05/2024]
Abstract
The human gut microbiome vastly extends the set of metabolic reactions catalysed by our own cells, with far-reaching consequences for host health and disease. However, our knowledge of gut microbial metabolism relies on a handful of model organisms, limiting our ability to interpret and predict the metabolism of complex microbial communities. In this Perspective, we discuss emerging tools for analysing and modelling the metabolism of gut microorganisms and for linking microorganisms, pathways and metabolites at the ecosystem level, highlighting promising best practices for researchers. Continued progress in this area will also require infrastructure development to facilitate cross-disciplinary synthesis of scientific findings. Collectively, these efforts can enable a broader and deeper understanding of the workings of the gut ecosystem and open new possibilities for microbiome manipulation and therapy.
Collapse
Affiliation(s)
- Cecilia Noecker
- Department of Biological Sciences, Minnesota State University, Mankato, Mankato, MN, USA
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Peter J Turnbaugh
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA, USA.
| |
Collapse
|
19
|
Berrios L, Bogar GD, Bogar LM, Venturini AM, Willing CE, Del Rio A, Ansell TB, Zemaitis K, Velickovic M, Velickovic D, Pellitier PT, Yeam J, Hutchinson C, Bloodsworth K, Lipton MS, Peay KG. Ectomycorrhizal fungi alter soil food webs and the functional potential of bacterial communities. mSystems 2024; 9:e0036924. [PMID: 38717159 PMCID: PMC11237468 DOI: 10.1128/msystems.00369-24] [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: 03/22/2024] [Accepted: 04/11/2024] [Indexed: 06/19/2024] Open
Abstract
Most of Earth's trees rely on critical soil nutrients that ectomycorrhizal fungi (EcMF) liberate and provide, and all of Earth's land plants associate with bacteria that help them survive in nature. Yet, our understanding of how the presence of EcMF modifies soil bacterial communities, soil food webs, and root chemistry requires direct experimental evidence to comprehend the effects that EcMF may generate in the belowground plant microbiome. To this end, we grew Pinus muricata plants in soils that were either inoculated with EcMF and native forest bacterial communities or only native bacterial communities. We then profiled the soil bacterial communities, applied metabolomics and lipidomics, and linked omics data sets to understand how the presence of EcMF modifies belowground biogeochemistry, bacterial community structure, and their functional potential. We found that the presence of EcMF (i) enriches soil bacteria linked to enhanced plant growth in nature, (ii) alters the quantity and composition of lipid and non-lipid soil metabolites, and (iii) modifies plant root chemistry toward pathogen suppression, enzymatic conservation, and reactive oxygen species scavenging. Using this multi-omic approach, we therefore show that this widespread fungal symbiosis may be a common factor for structuring soil food webs.IMPORTANCEUnderstanding how soil microbes interact with one another and their host plant will help us combat the negative effects that climate change has on terrestrial ecosystems. Unfortunately, we lack a clear understanding of how the presence of ectomycorrhizal fungi (EcMF)-one of the most dominant soil microbial groups on Earth-shapes belowground organic resources and the composition of bacterial communities. To address this knowledge gap, we profiled lipid and non-lipid metabolites in soils and plant roots, characterized soil bacterial communities, and compared soils amended either with or without EcMF. Our results show that the presence of EcMF changes soil organic resource availability, impacts the proliferation of different bacterial communities (in terms of both type and potential function), and primes plant root chemistry for pathogen suppression and energy conservation. Our findings therefore provide much-needed insight into how two of the most dominant soil microbial groups interact with one another and with their host plant.
Collapse
Affiliation(s)
- Louis Berrios
- Department of Biology, Stanford University, Stanford, California, USA
| | - Glade D. Bogar
- Kellogg Biological Station, Michigan State University, Hickory Corners, Michigan, USA
| | - Laura M. Bogar
- Department of Plant Biology, University of California, Davis, Davis, California, USA
| | | | - Claire E. Willing
- Department of Biology, Stanford University, Stanford, California, USA
- School of Environmental and Forest Sciences, University of Washington, Seattle, Washington, USA
| | - Anastacia Del Rio
- Department of Biology, Stanford University, Stanford, California, USA
| | - T. Bertie Ansell
- Department of Biology, Stanford University, Stanford, California, USA
- Division of CryoEM and Bioimaging, SSRL, SLAC National Accelerator Laboratory, Menlo Park, California, USA
| | - Kevin Zemaitis
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Marija Velickovic
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Dusan Velickovic
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington, USA
| | | | - Jay Yeam
- Department of Biology, Stanford University, Stanford, California, USA
| | - Chelsea Hutchinson
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Kent Bloodsworth
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Mary S. Lipton
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Kabir G. Peay
- Department of Biology, Stanford University, Stanford, California, USA
- Department of Earth System Science, Stanford University, Stanford, California, USA
| |
Collapse
|
20
|
Deek RA, Ma S, Lewis J, Li H. Statistical and computational methods for integrating microbiome, host genomics, and metabolomics data. eLife 2024; 13:e88956. [PMID: 38832759 PMCID: PMC11149933 DOI: 10.7554/elife.88956] [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: 04/26/2023] [Accepted: 05/10/2024] [Indexed: 06/05/2024] Open
Abstract
Large-scale microbiome studies are progressively utilizing multiomics designs, which include the collection of microbiome samples together with host genomics and metabolomics data. Despite the increasing number of data sources, there remains a bottleneck in understanding the relationships between different data modalities due to the limited number of statistical and computational methods for analyzing such data. Furthermore, little is known about the portability of general methods to the metagenomic setting and few specialized techniques have been developed. In this review, we summarize and implement some of the commonly used methods. We apply these methods to real data sets where shotgun metagenomic sequencing and metabolomics data are available for microbiome multiomics data integration analysis. We compare results across methods, highlight strengths and limitations of each, and discuss areas where statistical and computational innovation is needed.
Collapse
Affiliation(s)
- Rebecca A Deek
- Department of Biostatistics, University of PittsburghPittsburghUnited States
| | - Siyuan Ma
- Department of Biostatistics, Vanderbilt School of MedicineNashvilleUnited States
| | - James Lewis
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Hongzhe Li
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
21
|
Cohen A, Turjeman S, Levin R, Tal S, Koren O. Comparison of canine colostrum and milk using a multi-omics approach. Anim Microbiome 2024; 6:19. [PMID: 38650014 PMCID: PMC11034113 DOI: 10.1186/s42523-024-00309-4] [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: 09/04/2023] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND A mother's milk is considered the gold standard of nutrition in neonates and is a source of cytokines, immunoglobulins, growth factors, and other important components, yet little is known about the components of canine milk, specifically colostrum, and the knowledge related to its microbial and metabolic profiles is particularly underwhelming. In this study, we characterized canine colostrum and milk microbiota and metabolome for several breeds of dogs and examined profile shifts as milk matures in the first 8 days post-whelping. RESULTS Through untargeted metabolomics, we identified 63 named metabolites that were significantly differentially abundant between days 1 and 8 of lactation. Surprisingly, the microbial compositions of the colostrum and milk, characterized using 16S rRNA gene sequencing, were largely similar, with only two differentiating genera. The shifts observed, mainly increases in several sugars and amino sugars over time and shifts in amino acid metabolites, align with shifts observed in human milk samples and track with puppy development. CONCLUSION Like human milk, canine milk composition is dynamic, and shifts are well correlated with developing puppies' needs. Such a study of the metabolic profile of canine milk, and its relation to the microbial community, provides insights into the changing needs of the neonate, as well as the ideal nutrition profile for optimal functionality. This information will add to the existing knowledge base of canine milk composition with the prospect of creating a quality, tailored milk substitute or supplement for puppies.
Collapse
Affiliation(s)
- Alisa Cohen
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Sondra Turjeman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Rachel Levin
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Smadar Tal
- Koret School of Veterinary Medicine, The Hebrew University Veterinary Teaching Hospital, Hebrew University of Jerusalem, Rehovot, Israel
- Tel-Hai Academic College, Upper Galilee, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
- Kyung Hee University, Seoul, the Republic of Korea.
| |
Collapse
|
22
|
Liu X, Liu Y, Liu J, Zhang H, Shan C, Guo Y, Gong X, Cui M, Li X, Tang M. Correlation between the gut microbiome and neurodegenerative diseases: a review of metagenomics evidence. Neural Regen Res 2024; 19:833-845. [PMID: 37843219 PMCID: PMC10664138 DOI: 10.4103/1673-5374.382223] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/19/2023] [Accepted: 06/17/2023] [Indexed: 10/17/2023] Open
Abstract
A growing body of evidence suggests that the gut microbiota contributes to the development of neurodegenerative diseases via the microbiota-gut-brain axis. As a contributing factor, microbiota dysbiosis always occurs in pathological changes of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. High-throughput sequencing technology has helped to reveal that the bidirectional communication between the central nervous system and the enteric nervous system is facilitated by the microbiota's diverse microorganisms, and for both neuroimmune and neuroendocrine systems. Here, we summarize the bioinformatics analysis and wet-biology validation for the gut metagenomics in neurodegenerative diseases, with an emphasis on multi-omics studies and the gut virome. The pathogen-associated signaling biomarkers for identifying brain disorders and potential therapeutic targets are also elucidated. Finally, we discuss the role of diet, prebiotics, probiotics, postbiotics and exercise interventions in remodeling the microbiome and reducing the symptoms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaoyan Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Yi Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu Province, China
| | - Junlin Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Hantao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Chaofan Shan
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Yinglu Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Xun Gong
- Department of Rheumatology & Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Mengmeng Cui
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Xiubin Li
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| |
Collapse
|
23
|
Wen Y, Luo Y, Qiu H, Chen B, Huang J, Lv S, Wang Y, Li J, Tao L, Yang B, Li K, He L, He M, Yang Q, Yu Z, Xiao W, Zhao M, Zou X, Lu R, Gu C. Gut microbiota affects obesity susceptibility in mice through gut metabolites. Front Microbiol 2024; 15:1343511. [PMID: 38450171 PMCID: PMC10916699 DOI: 10.3389/fmicb.2024.1343511] [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: 11/23/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction It is well-known that different populations and animals, even experimental animals with the same rearing conditions, differ in their susceptibility to obesity. The disparity in gut microbiota could potentially account for the variation in susceptibility to obesity. However, the precise impact of gut microbiota on gut metabolites and its subsequent influence on susceptibility to obesity remains uncertain. Methods In this study, we established obesity-prone (OP) and obesity-resistant (OR) mouse models by High Fat Diet (HFD). Fecal contents of cecum were examined using 16S rDNA sequencing and untargeted metabolomics. Correlation analysis and MIMOSA2 analysis were used to explore the association between gut microbiota and intestinal metabolites. Results After a HFD, gut microbiota and gut metabolic profiles were significantly different between OP and OR mice. Gut microbiota after a HFD may lead to changes in eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), a variety of branched fatty acid esters of hydroxy fatty acids (FAHFAs) and a variety of phospholipids to promote obesity. The bacteria g_Akkermansia (Greengene ID: 175696) may contribute to the difference in obesity susceptibility through the synthesis of glycerophosphoryl diester phosphodiesterase (glpQ) to promote choline production and the synthesis of valyl-tRNA synthetase (VARS) which promotes L-Valine degradation. In addition, gut microbiota may affect obesity and obesity susceptibility through histidine metabolism, linoleic acid metabolism and protein digestion and absorption pathways.
Collapse
Affiliation(s)
- Yuhang Wen
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Yadan Luo
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Hao Qiu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Baoting Chen
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Jingrong Huang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Shuya Lv
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Yan Wang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Jiabi Li
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Lingling Tao
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Bailin Yang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Ke Li
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Lvqin He
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Manli He
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Qian Yang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Zehui Yu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Wudian Xiao
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Mingde Zhao
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Xiaoxia Zou
- Suining First People's Hospital, Suining, China
| | - Ruilin Lu
- Suining First People's Hospital, Suining, China
| | - Congwei Gu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| |
Collapse
|
24
|
Shtossel O, Koren O, Shai I, Rinott E, Louzoun Y. Gut microbiome-metabolome interactions predict host condition. MICROBIOME 2024; 12:24. [PMID: 38336867 PMCID: PMC10858481 DOI: 10.1186/s40168-023-01737-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 12/10/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND The effect of microbes on their human host is often mediated through changes in metabolite concentrations. As such, multiple tools have been proposed to predict metabolite concentrations from microbial taxa frequencies. Such tools typically fail to capture the dependence of the microbiome-metabolite relation on the environment. RESULTS We propose to treat the microbiome-metabolome relation as the equilibrium of a complex interaction and to relate the host condition to a latent representation of the interaction between the log concentration of the metabolome and the log frequencies of the microbiome. We develop LOCATE (Latent variables Of miCrobiome And meTabolites rElations), a machine learning tool to predict the metabolite concentration from the microbiome composition and produce a latent representation of the interaction. This representation is then used to predict the host condition. LOCATE's accuracy in predicting the metabolome is higher than all current predictors. The metabolite concentration prediction accuracy significantly decreases cross datasets, and cross conditions, especially in 16S data. LOCATE's latent representation predicts the host condition better than either the microbiome or the metabolome. This representation is strongly correlated with host demographics. A significant improvement in accuracy (0.793 vs. 0.724 average accuracy) is obtained even with a small number of metabolite samples ([Formula: see text]). CONCLUSION These results suggest that a latent representation of the microbiome-metabolome interaction leads to a better association with the host condition than any of the two separated or the simple combination of the two. Video Abstract.
Collapse
Affiliation(s)
- Oshrit Shtossel
- Department of Mathematics, Bar-Ilan University, Ramat Gan, 52900, Israel
| | - Omry Koren
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Iris Shai
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ehud Rinott
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yoram Louzoun
- Department of Mathematics, Bar-Ilan University, Ramat Gan, 52900, Israel.
| |
Collapse
|
25
|
Wang Z, Yuan X, Zhu Z, Pang L, Ding S, Li X, Kang Y, Hei G, Zhang L, Zhang X, Wang S, Jian X, Li Z, Zheng C, Fan X, Hu S, Shi Y, Song X. Multiomics Analyses Reveal Microbiome-Gut-Brain Crosstalk Centered on Aberrant Gamma-Aminobutyric Acid and Tryptophan Metabolism in Drug-Naïve Patients with First-Episode Schizophrenia. Schizophr Bull 2024; 50:187-198. [PMID: 37119525 PMCID: PMC10754168 DOI: 10.1093/schbul/sbad026] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
BACKGROUND AND HYPOTHESIS Schizophrenia (SCZ) is associated with complex crosstalk between the gut microbiota and host metabolism, but the underlying mechanism remains elusive. Investigating the aberrant neurotransmitter processes reflected by alterations identified using multiomics analysis is valuable to fully explain the pathogenesis of SCZ. STUDY DESIGN We conducted an integrative analysis of multiomics data, including the serum metabolome, fecal metagenome, single nucleotide polymorphism data, and neuroimaging data obtained from a cohort of 127 drug-naïve, first-episode SCZ patients and 92 healthy controls to characterize the microbiome-gut-brain axis in SCZ patients. We used pathway-based polygenic risk score (PRS) analyses to determine the biological pathways contributing to genetic risk and mediation effect analyses to determine the important neuroimaging features. Additionally, a random forest model was generated for effective SCZ diagnosis. STUDY RESULTS We found that the altered metabolome and dysregulated microbiome were associated with neuroactive metabolites, including gamma-aminobutyric acid (GABA), tryptophan, and short-chain fatty acids. Further structural and functional magnetic resonance imaging analyses highlighted that gray matter volume and functional connectivity disturbances mediate the relationships between Ruminococcus_torgues and Collinsella_aerofaciens and symptom severity and the relationships between species Lactobacillus_ruminis and differential metabolites l-2,4-diaminobutyric acid and N-acetylserotonin and cognitive function. Moreover, analyses of the Polygenic Risk Score (PRS) support that alterations in GABA and tryptophan neurotransmitter pathways are associated with SCZ risk, and GABA might be a more dominant contributor. CONCLUSIONS This study provides new insights into systematic relationships among genes, metabolism, and the gut microbiota that affect brain functional connectivity, thereby affecting SCZ pathogenesis.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University; Henan International Joint Laboratory of Biological Psychiatry; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University; Collaborative Innovation Centre for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Xiuxia Yuan
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University; Henan International Joint Laboratory of Biological Psychiatry; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
| | - Zijia Zhu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University; Collaborative Innovation Centre for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Lijuan Pang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University; Henan International Joint Laboratory of Biological Psychiatry; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
| | - Shizhi Ding
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University; Collaborative Innovation Centre for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Xue Li
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University; Henan International Joint Laboratory of Biological Psychiatry; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
| | - Yulin Kang
- Institute of Environmental Information, Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Gangrui Hei
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University; Henan International Joint Laboratory of Biological Psychiatry; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
| | - Liyuan Zhang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University; Henan International Joint Laboratory of Biological Psychiatry; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
| | - Xiaoyun Zhang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University; Henan International Joint Laboratory of Biological Psychiatry; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
| | - Shuying Wang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University; Henan International Joint Laboratory of Biological Psychiatry; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
| | - Xuemin Jian
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University; Collaborative Innovation Centre for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiqiang Li
- The Affiliated Hospital of Qingdao University and the Biomedical Sciences Institute of Qingdao University, Qingdao Branch of SJTU Bio-X Institutes, Qingdao University, Qingdao, China
| | - Chenxiang Zheng
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University; Collaborative Innovation Centre for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoduo Fan
- Psychotic Disorders Program, UMass Memorial Medical Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Shaohua Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongyong Shi
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University; Henan International Joint Laboratory of Biological Psychiatry; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University; Collaborative Innovation Centre for Brain Science, Shanghai Jiao Tong University, Shanghai, China
- The Affiliated Hospital of Qingdao University and the Biomedical Sciences Institute of Qingdao University, Qingdao Branch of SJTU Bio-X Institutes, Qingdao University, Qingdao, China
| | - Xueqin Song
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University; Henan International Joint Laboratory of Biological Psychiatry; Henan Psychiatric Transformation Research Key Laboratory/Zhengzhou University, Zhengzhou, China
| |
Collapse
|
26
|
Malwe AS, Sharma VK. Application of artificial intelligence approaches to predict the metabolism of xenobiotic molecules by human gut microbiome. Front Microbiol 2023; 14:1254073. [PMID: 38116528 PMCID: PMC10728657 DOI: 10.3389/fmicb.2023.1254073] [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: 07/06/2023] [Accepted: 10/12/2023] [Indexed: 12/21/2023] Open
Abstract
A highly complex, diverse, and dense community of more than 1,000 different gut bacterial species constitutes the human gut microbiome that harbours vast metabolic capabilities encoded by more than 300,000 bacterial enzymes to metabolise complex polysaccharides, orally administered drugs/xenobiotics, nutraceuticals, or prebiotics. One of the implications of gut microbiome mediated biotransformation is the metabolism of xenobiotics such as medicinal drugs, which lead to alteration in their pharmacological properties, loss of drug efficacy, bioavailability, may generate toxic byproducts and sometimes also help in conversion of a prodrug into its active metabolite. Given the diversity of gut microbiome and the complex interplay of the metabolic enzymes and their diverse substrates, the traditional experimental methods have limited ability to identify the gut bacterial species involved in such biotransformation, and to study the bacterial species-metabolite interactions in gut. In this scenario, computational approaches such as machine learning-based tools presents unprecedented opportunities and ability to predict the gut bacteria and enzymes that can potentially metabolise a candidate drug. Here, we have reviewed the need to identify the gut microbiome-based metabolism of xenobiotics and have provided comprehensive information on the available methods, tools, and databases to address it along with their scope and limitations.
Collapse
Affiliation(s)
| | - Vineet K. Sharma
- MetaBioSys Lab, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, India
| |
Collapse
|
27
|
Fernandez ME, Martinez-Romero J, Aon MA, Bernier M, Price NL, de Cabo R. How is Big Data reshaping preclinical aging research? Lab Anim (NY) 2023; 52:289-314. [PMID: 38017182 DOI: 10.1038/s41684-023-01286-y] [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: 05/24/2023] [Accepted: 10/10/2023] [Indexed: 11/30/2023]
Abstract
The exponential scientific and technological progress during the past 30 years has favored the comprehensive characterization of aging processes with their multivariate nature, leading to the advent of Big Data in preclinical aging research. Spanning from molecular omics to organism-level deep phenotyping, Big Data demands large computational resources for storage and analysis, as well as new analytical tools and conceptual frameworks to gain novel insights leading to discovery. Systems biology has emerged as a paradigm that utilizes Big Data to gain insightful information enabling a better understanding of living organisms, visualized as multilayered networks of interacting molecules, cells, tissues and organs at different spatiotemporal scales. In this framework, where aging, health and disease represent emergent states from an evolving dynamic complex system, context given by, for example, strain, sex and feeding times, becomes paramount for defining the biological trajectory of an organism. Using bioinformatics and artificial intelligence, the systems biology approach is leading to remarkable advances in our understanding of the underlying mechanism of aging biology and assisting in creative experimental study designs in animal models. Future in-depth knowledge acquisition will depend on the ability to fully integrate information from different spatiotemporal scales in organisms, which will probably require the adoption of theories and methods from the field of complex systems. Here we review state-of-the-art approaches in preclinical research, with a focus on rodent models, that are leading to conceptual and/or technical advances in leveraging Big Data to understand basic aging biology and its full translational potential.
Collapse
Affiliation(s)
- Maria Emilia Fernandez
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jorge Martinez-Romero
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Miguel A Aon
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michel Bernier
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nathan L Price
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
28
|
Shen Y, An Z, Huyan Z, Shu X, Wu D, Zhang N, Pellegrini N, Rubert J. Lipid complexation reduces rice starch digestibility and boosts short-chain fatty acid production via gut microbiota. NPJ Sci Food 2023; 7:56. [PMID: 37853069 PMCID: PMC10584848 DOI: 10.1038/s41538-023-00230-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 10/02/2023] [Indexed: 10/20/2023] Open
Abstract
In this study, two rice varieties (RS4 and GZ93) with different amylose and lipid contents were studied, and their starch was used to prepare starch-palmitic acid complexes. The RS4 samples showed a significantly higher lipid content in their flour, starch, and complex samples compared to GZ93. The static in vitro digestion highlighted that RS4 samples had significantly lower digestibility than the GZ93 samples. The C∞ of the starch-lipid complex samples was found to be 17.7% and 18.5% lower than that of the starch samples in GZ93 and RS4, respectively. The INFOGEST undigested fractions were subsequently used for in vitro colonic fermentation. Short-chain fatty acids (SCFAs) concentrations, mainly acetate, and propionate were significantly higher in starch-lipid complexes compared to native flour or starch samples. Starch-lipid complexes produced a distinctive microbial composition, which resulted in different gene functions, mainly related to pyruvate, fructose, and mannose metabolism. Using Model-based Integration of Metabolite Observations and Species Abundances 2 (MIMOSA2), SCFA production was predicted and associated with the gut microbiota. These results indicated that incorporating lipids into rice starch promotes SCFA production by modulating the gut microbiota selectively.
Collapse
Affiliation(s)
- Yi Shen
- State Key Laboratory of Rice Biology, Key Laboratory of the Ministry of Agriculture for Nuclear-Agricultural Sciences, Zhejiang University, Hangzhou, 310058, PR China
- Food Quality and Design Group, Wageningen University & Research, P. O. Box 17, 6700 AA, Wageningen, The Netherlands
| | - Zengxu An
- State Key Laboratory of Rice Biology, Key Laboratory of the Ministry of Agriculture for Nuclear-Agricultural Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Zongyao Huyan
- Food Quality and Design Group, Wageningen University & Research, P. O. Box 17, 6700 AA, Wageningen, The Netherlands
| | - Xiaoli Shu
- State Key Laboratory of Rice Biology, Key Laboratory of the Ministry of Agriculture for Nuclear-Agricultural Sciences, Zhejiang University, Hangzhou, 310058, PR China
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Yazhou District, Sanya, 572025, PR China
| | - Dianxing Wu
- State Key Laboratory of Rice Biology, Key Laboratory of the Ministry of Agriculture for Nuclear-Agricultural Sciences, Zhejiang University, Hangzhou, 310058, PR China
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Yazhou District, Sanya, 572025, PR China
| | - Ning Zhang
- State Key Laboratory of Rice Biology, Key Laboratory of the Ministry of Agriculture for Nuclear-Agricultural Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Nicoletta Pellegrini
- Food Quality and Design Group, Wageningen University & Research, P. O. Box 17, 6700 AA, Wageningen, The Netherlands
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, via Sondrio 2/A, Udine, 33100, Italy
| | - Josep Rubert
- Food Quality and Design Group, Wageningen University & Research, P. O. Box 17, 6700 AA, Wageningen, The Netherlands.
| |
Collapse
|
29
|
Gautam A, Bhowmik D, Basu S, Zeng W, Lahiri A, Huson DH, Paul S. Microbiome Metabolome Integration Platform (MMIP): a web-based platform for microbiome and metabolome data integration and feature identification. Brief Bioinform 2023; 24:bbad325. [PMID: 37771003 DOI: 10.1093/bib/bbad325] [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: 04/17/2023] [Revised: 08/12/2023] [Indexed: 09/30/2023] Open
Abstract
A microbial community maintains its ecological dynamics via metabolite crosstalk. Hence, knowledge of the metabolome, alongside its populace, would help us understand the functionality of a community and also predict how it will change in atypical conditions. Methods that employ low-cost metagenomic sequencing data can predict the metabolic potential of a community, that is, its ability to produce or utilize specific metabolites. These, in turn, can potentially serve as markers of biochemical pathways that are associated with different communities. We developed MMIP (Microbiome Metabolome Integration Platform), a web-based analytical and predictive tool that can be used to compare the taxonomic content, diversity variation and the metabolic potential between two sets of microbial communities from targeted amplicon sequencing data. MMIP is capable of highlighting statistically significant taxonomic, enzymatic and metabolic attributes as well as learning-based features associated with one group in comparison with another. Furthermore, MMIP can predict linkages among species or groups of microbes in the community, specific enzyme profiles, compounds or metabolites associated with such a group of organisms. With MMIP, we aim to provide a user-friendly, online web server for performing key microbiome-associated analyses of targeted amplicon sequencing data, predicting metabolite signature, and using learning-based linkage analysis, without the need for initial metabolomic analysis, and thereby helping in hypothesis generation.
Collapse
Affiliation(s)
- Anupam Gautam
- Algorithms in Bioinformatics, Institute for Bioinformatics and Medical Informatics, University of Tübingen, Tübingen, Germany
- International Max Planck Research School "From Molecules to Organisms", Max Planck Institute for Biology Tübingen, Tübingen, Germany
- Cluster of Excellence: EXC 2124: Controlling Microbes to Fight Infection, Tübingen, Germany
| | - Debaleena Bhowmik
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sayantani Basu
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Wenhuan Zeng
- Algorithms in Bioinformatics, Institute for Bioinformatics and Medical Informatics, University of Tübingen, Tübingen, Germany
- Cluster of Excellence: EXC 2064: Machine Learning: New Perspectives for Science, University of Tübingen, Tübingen, Germany
| | - Abhishake Lahiri
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Centre for Health Science and Technology, JIS Institute of Advanced Studies and Research Kolkata, JIS University, West Bengal, India
| | - Daniel H Huson
- Algorithms in Bioinformatics, Institute for Bioinformatics and Medical Informatics, University of Tübingen, Tübingen, Germany
- International Max Planck Research School "From Molecules to Organisms", Max Planck Institute for Biology Tübingen, Tübingen, Germany
- Cluster of Excellence: EXC 2124: Controlling Microbes to Fight Infection, Tübingen, Germany
| | - Sandip Paul
- Centre for Health Science and Technology, JIS Institute of Advanced Studies and Research Kolkata, JIS University, West Bengal, India
| |
Collapse
|
30
|
Meydan Y, Baldini F, Korem T. pymgpipe: microbiome metabolic modeling in Python. JOURNAL OF OPEN SOURCE SOFTWARE 2023; 8:5545. [PMID: 37885608 PMCID: PMC10600976 DOI: 10.21105/joss.05545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Affiliation(s)
- Yoli Meydan
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, United States of America
| | - Federico Baldini
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, United States of America
| | - Tal Korem
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, United States of America
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, United States of America
| |
Collapse
|
31
|
Bartmanski BJ, Rocha M, Zimmermann-Kogadeeva M. Recent advances in data- and knowledge-driven approaches to explore primary microbial metabolism. Curr Opin Chem Biol 2023; 75:102324. [PMID: 37207402 PMCID: PMC10410306 DOI: 10.1016/j.cbpa.2023.102324] [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: 12/28/2022] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 05/21/2023]
Abstract
With the rapid progress in metabolomics and sequencing technologies, more data on the metabolome of single microbes and their communities become available, revealing the potential of microorganisms to metabolize a broad range of chemical compounds. The analysis of microbial metabolomics datasets remains challenging since it inherits the technical challenges of metabolomics analysis, such as compound identification and annotation, while harboring challenges in data interpretation, such as distinguishing metabolite sources in mixed samples. This review outlines the recent advances in computational methods to analyze primary microbial metabolism: knowledge-based approaches that take advantage of metabolic and molecular networks and data-driven approaches that employ machine/deep learning algorithms in combination with large-scale datasets. These methods aim at improving metabolite identification and disentangling reciprocal interactions between microbes and metabolites. We also discuss the perspective of combining these approaches and further developments required to advance the investigation of primary metabolism in mixed microbial samples.
Collapse
Affiliation(s)
| | - Miguel Rocha
- Centre of Biological Engineering, University of Minho, Campus of Gualtar, Braga, Portugal
| | | |
Collapse
|
32
|
Jacobs JP, Lagishetty V, Hauer MC, Labus JS, Dong TS, Toma R, Vuyisich M, Naliboff BD, Lackner JM, Gupta A, Tillisch K, Mayer EA. Multi-omics profiles of the intestinal microbiome in irritable bowel syndrome and its bowel habit subtypes. MICROBIOME 2023; 11:5. [PMID: 36624530 PMCID: PMC9830758 DOI: 10.1186/s40168-022-01450-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 12/14/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is a common gastrointestinal disorder that is thought to involve alterations in the gut microbiome, but robust microbial signatures have been challenging to identify. As prior studies have primarily focused on composition, we hypothesized that multi-omics assessment of microbial function incorporating both metatranscriptomics and metabolomics would further delineate microbial profiles of IBS and its subtypes. METHODS Fecal samples were collected from a racially/ethnically diverse cohort of 495 subjects, including 318 IBS patients and 177 healthy controls, for analysis by 16S rRNA gene sequencing (n = 486), metatranscriptomics (n = 327), and untargeted metabolomics (n = 368). Differentially abundant microbes, predicted genes, transcripts, and metabolites in IBS were identified by multivariate models incorporating age, sex, race/ethnicity, BMI, diet, and HAD-Anxiety. Inter-omic functional relationships were assessed by transcript/gene ratios and microbial metabolic modeling. Differential features were used to construct random forests classifiers. RESULTS IBS was associated with global alterations in microbiome composition by 16S rRNA sequencing and metatranscriptomics, and in microbiome function by predicted metagenomics, metatranscriptomics, and metabolomics. After adjusting for age, sex, race/ethnicity, BMI, diet, and anxiety, IBS was associated with differential abundance of bacterial taxa such as Bacteroides dorei; metabolites including increased tyramine and decreased gentisate and hydrocinnamate; and transcripts related to fructooligosaccharide and polyol utilization. IBS further showed transcriptional upregulation of enzymes involved in fructose and glucan metabolism as well as the succinate pathway of carbohydrate fermentation. A multi-omics classifier for IBS had significantly higher accuracy (AUC 0.82) than classifiers using individual datasets. Diarrhea-predominant IBS (IBS-D) demonstrated shifts in the metatranscriptome and metabolome including increased bile acids, polyamines, succinate pathway intermediates (malate, fumarate), and transcripts involved in fructose, mannose, and polyol metabolism compared to constipation-predominant IBS (IBS-C). A classifier incorporating metabolites and gene-normalized transcripts differentiated IBS-D from IBS-C with high accuracy (AUC 0.86). CONCLUSIONS IBS is characterized by a multi-omics microbial signature indicating increased capacity to utilize fermentable carbohydrates-consistent with the clinical benefit of diets restricting this energy source-that also includes multiple previously unrecognized metabolites and metabolic pathways. These findings support the need for integrative assessment of microbial function to investigate the microbiome in IBS and identify novel microbiome-related therapeutic targets. Video Abstract.
Collapse
Affiliation(s)
- Jonathan P Jacobs
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA.
| | - Venu Lagishetty
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Megan C Hauer
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer S Labus
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Tien S Dong
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Ryan Toma
- Viome Life Sciences, Bellevue, WA, USA
| | | | - Bruce D Naliboff
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jeffrey M Lackner
- Division of Behavioral Medicine, Department of Medicine, Jacobs School of Medicine, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Arpana Gupta
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kirsten Tillisch
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Integrative Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Emeran A Mayer
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
33
|
Agamah FE, Bayjanov JR, Niehues A, Njoku KF, Skelton M, Mazandu GK, Ederveen THA, Mulder N, Chimusa ER, 't Hoen PAC. Computational approaches for network-based integrative multi-omics analysis. Front Mol Biosci 2022; 9:967205. [PMID: 36452456 PMCID: PMC9703081 DOI: 10.3389/fmolb.2022.967205] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 10/20/2022] [Indexed: 08/27/2023] Open
Abstract
Advances in omics technologies allow for holistic studies into biological systems. These studies rely on integrative data analysis techniques to obtain a comprehensive view of the dynamics of cellular processes, and molecular mechanisms. Network-based integrative approaches have revolutionized multi-omics analysis by providing the framework to represent interactions between multiple different omics-layers in a graph, which may faithfully reflect the molecular wiring in a cell. Here we review network-based multi-omics/multi-modal integrative analytical approaches. We classify these approaches according to the type of omics data supported, the methods and/or algorithms implemented, their node and/or edge weighting components, and their ability to identify key nodes and subnetworks. We show how these approaches can be used to identify biomarkers, disease subtypes, crosstalk, causality, and molecular drivers of physiological and pathological mechanisms. We provide insight into the most appropriate methods and tools for research questions as showcased around the aetiology and treatment of COVID-19 that can be informed by multi-omics data integration. We conclude with an overview of challenges associated with multi-omics network-based analysis, such as reproducibility, heterogeneity, (biological) interpretability of the results, and we highlight some future directions for network-based integration.
Collapse
Affiliation(s)
- Francis E. Agamah
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jumamurat R. Bayjanov
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Anna Niehues
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Kelechi F. Njoku
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Michelle Skelton
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gaston K. Mazandu
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- African Institute for Mathematical Sciences, Cape Town, South Africa
| | - Thomas H. A. Ederveen
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nicola Mulder
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Emile R. Chimusa
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, United Kingdom
| | - Peter A. C. 't Hoen
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
34
|
Fortuin S, Soares NC. The Integration of Proteomics and Metabolomics Data Paving the Way for a Better Understanding of the Mechanisms Underlying Microbial Acquired Drug Resistance. Front Med (Lausanne) 2022; 9:849838. [PMID: 35602483 PMCID: PMC9120609 DOI: 10.3389/fmed.2022.849838] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Due to an increase in the overuse of antimicrobials and accelerated incidence of drug resistant pathogens, antimicrobial resistance has become a global health threat. In particular, bacterial antimicrobial resistance, in both hospital and community acquired transmission, have been found to be the leading cause of death due to infectious diseases. Understanding the mechanisms of bacterial drug resistance is of clinical significance irrespective of hospital or community acquired since it plays an important role in the treatment strategy and controlling infectious diseases. Here we highlight the advances in mass spectrometry-based proteomics impact in bacterial proteomics and metabolomics analysis- focus on bacterial drug resistance. Advances in omics technologies over the last few decades now allows multi-omics studies in order to obtain a comprehensive understanding of the biochemical alterations of pathogenic bacteria in the context of antibiotic exposure, identify novel biomarkers to develop new drug targets, develop time-effectively screen for drug susceptibility or resistance using proteomics and metabolomics.
Collapse
Affiliation(s)
- Suereta Fortuin
- African Microbiome Institute, General Internal Medicine, Stellenbosch University, Cape Town, South Africa
- *Correspondence: Suereta Fortuin
| | - Nelson C. Soares
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Nelson C. Soares
| |
Collapse
|
35
|
Beale DJ, Jones OA, Bose U, Broadbent JA, Walsh TK, van de Kamp J, Bissett A. Omics-based ecosurveillance for the assessment of ecosystem function, health, and resilience. Emerg Top Life Sci 2022; 6:185-199. [PMID: 35403668 PMCID: PMC9023019 DOI: 10.1042/etls20210261] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/17/2022] [Accepted: 03/22/2022] [Indexed: 12/15/2022]
Abstract
Current environmental monitoring efforts often focus on known, regulated contaminants ignoring the potential effects of unmeasured compounds and/or environmental factors. These specific, targeted approaches lack broader environmental information and understanding, hindering effective environmental management and policy. Switching to comprehensive, untargeted monitoring of contaminants, organism health, and environmental factors, such as nutrients, temperature, and pH, would provide more effective monitoring with a likely concomitant increase in environmental health. However, even this method would not capture subtle biochemical changes in organisms induced by chronic toxicant exposure. Ecosurveillance is the systematic collection, analysis, and interpretation of ecosystem health-related data that can address this knowledge gap and provide much-needed additional lines of evidence to environmental monitoring programs. Its use would therefore be of great benefit to environmental management and assessment. Unfortunately, the science of 'ecosurveillance', especially omics-based ecosurveillance is not well known. Here, we give an overview of this emerging area and show how it has been beneficially applied in a range of systems. We anticipate this review to be a starting point for further efforts to improve environmental monitoring via the integration of comprehensive chemical assessments and molecular biology-based approaches. Bringing multiple levels of omics technology-based assessment together into a systems-wide ecosurveillance approach will bring a greater understanding of the environment, particularly the microbial communities upon which we ultimately rely to remediate perturbed ecosystems.
Collapse
Affiliation(s)
- David J. Beale
- Land and Water, Commonwealth Scientific and Industrial Research Organisation, Ecosciences Precinct, Dutton Park QLD 4102, Australia
| | - Oliver A.H. Jones
- Australian Centre for Research on Separation Science (ACROSS), School of Science, RMIT University, Bundoora West Campus, PO Box 71, Bundoora, VIC 3083, Australia
| | - Utpal Bose
- Agriculture and Food, Commonwealth Scientific and Industrial Research Organisation, Queensland Bioscience Precinct, St Lucia, QLD 4067, Australia
| | - James A. Broadbent
- Agriculture and Food, Commonwealth Scientific and Industrial Research Organisation, Queensland Bioscience Precinct, St Lucia, QLD 4067, Australia
| | - Thomas K. Walsh
- Land and Water, Commonwealth Scientific and Industrial Research Organisation, Acton, ACT 2601, Australia
| | - Jodie van de Kamp
- Oceans and Atmosphere, Commonwealth Scientific and Industrial Research Organisation, Battery Point, TAS 7004, Australia
| | - Andrew Bissett
- Oceans and Atmosphere, Commonwealth Scientific and Industrial Research Organisation, Battery Point, TAS 7004, Australia
| |
Collapse
|