1
|
Huang C, Ma Q, Zeng X, He J, You F, Fu X, Ren Y. Exploring non-invasive biomarkers for pulmonary nodule detection based on salivary microbiomics and machine learning algorithms. Sci Rep 2025; 15:10848. [PMID: 40155734 PMCID: PMC11953406 DOI: 10.1038/s41598-025-95692-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Microorganisms are one of the most promising biomarkers for cancer, and the relationship between microorganisms and lung cancer occurrence and development provides significant potential for pulmonary nodule (PN) diagnosis from a microbiological perspective. This study aimed to analyze the salivary microbiota features of patients with PN and assess the potential of the salivary microbiota as a non-invasive PN biomarker. We collected saliva smples from 153 patients with PN and 40 controls. Using 16 S rRNA gene sequencing, differences in α- and β-diversity and community composition between the group with PN and controls were analyzed. Subsequently, specific microbial variables were selected using six models were trained on the selected salivary microbial features. The models were evaluated using metrics, such as the area under the receiver operating characteristic curve (AUC), to identify the best-performing model. Furthermore, the Bayesian optimization algorithm was used to optimize this best-performing model. Finally, the SHapley Additive exPlanations (SHAP) interpretability framework was used to interpret the output of the optimal model and identify potential PN biomarkers. Significant differences in α- and β-diversity were observed between the group with PN and controls. Although the predominant genera were consistent between the groups, significant disparities were observed in their relative abundances. By leveraging the random forest algorithm, ten characteristic microbial variables were identified and incorporated into six models, which effectively facilitated PN diagnosis. The XGBoost model demonstrated the best performance. Further optimization of the XGBoost model resulted in a Bayesian Optimization-based XGBoost (BOXGB) model. Based on the BOXGB model, an online saliva microbiota-based PN prediction platform was developed. Lastly, SHAP analysis suggested Defluviitaleaceae_UCG-011, Aggregatibacter, Oribacterium, Bacillus, and Prevotalla are promising non-invasive PN biomarkers. This study proved salivary microbiota as a non-invasive PN biomarker, expanding the clinical diagnostic approaches for PN.
Collapse
Affiliation(s)
- Chunxia Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Qiong Ma
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Xiao Zeng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Jiawei He
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
| | - Xi Fu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
| | - Yifeng Ren
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
| |
Collapse
|
2
|
Bai D, Ma C, Xun J, Luo H, Yang H, Lyu H, Zhu Z, Gai A, Yousuf S, Peng K, Xu S, Gao Y, Wang Y, Liu Y. MicrobiomeStatPlots: Microbiome statistics plotting gallery for meta-omics and bioinformatics. IMETA 2025; 4:e70002. [PMID: 40027478 PMCID: PMC11865346 DOI: 10.1002/imt2.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 03/05/2025]
Abstract
The rapid growth of microbiome research has generated an unprecedented amount of multi-omics data, presenting challenges in data analysis and visualization. To address these issues, we present MicrobiomeStatPlots, a comprehensive platform offering streamlined, reproducible tools for microbiome data analysis and visualization. This platform integrates essential bioinformatics workflows with multi-omics pipelines and provides 82 distinct visualization cases for interpreting microbiome datasets. By incorporating basic tutorials and advanced R-based visualization strategies, MicrobiomeStatPlots enhances accessibility and usability for researchers. Users can customize plots, contribute to the platform's expansion, and access a wealth of bioinformatics knowledge freely on GitHub (https://github.com/YongxinLiu/MicrobiomeStatPlot). Future plans include extending support for metabolomics, viromics, and metatranscriptomics, along with seamless integration of visualization tools into omics workflows. MicrobiomeStatPlots bridges gaps in microbiome data analysis and visualization, paving the way for more efficient, impactful microbiome research.
Collapse
Affiliation(s)
- Defeng Bai
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| | - Chuang Ma
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
- School of HorticultureAnhui Agricultural UniversityHefeiChina
| | - Jiani Xun
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| | - Hao Luo
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| | - Haifei Yang
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
- College of Life SciencesQingdao Agricultural UniversityQingdaoChina
| | - Hujie Lyu
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
- Department of Food Science and NutritionThe Hong Kong Polytechnic UniversityHong KongSARChina
| | - Zhihao Zhu
- Zhanjiang Key Laboratory of Human Microecology and Clinical Translation Research, the Marine Biomedical Research Institute, College of Basic MedicineGuangdong Medical UniversityZhanjiangChina
| | - Anran Gai
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
- School of Agricultural SciencesZhengzhou UniversityZhengzhouChina
| | - Salsabeel Yousuf
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| | - Kai Peng
- Jiangsu Co‐Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary MedicineYangzhou UniversityYangzhouChina
| | - Shanshan Xu
- School of Food and Biological EngineeringHefei University of TechnologyHefeiChina
| | - Yunyun Gao
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
- School of Ecology and Nature ConservationBeijing Forestry UniversityBeijingChina
| | - Yao Wang
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| | - Yong‐Xin Liu
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhenChina
| |
Collapse
|
3
|
Ghosh S, Zhao X, Alim M, Brudno M, Bhat M. Artificial intelligence applied to 'omics data in liver disease: towards a personalised approach for diagnosis, prognosis and treatment. Gut 2025; 74:295-311. [PMID: 39174307 PMCID: PMC11874365 DOI: 10.1136/gutjnl-2023-331740] [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: 04/15/2024] [Accepted: 07/24/2024] [Indexed: 08/24/2024]
Abstract
Advancements in omics technologies and artificial intelligence (AI) methodologies are fuelling our progress towards personalised diagnosis, prognosis and treatment strategies in hepatology. This review provides a comprehensive overview of the current landscape of AI methods used for analysis of omics data in liver diseases. We present an overview of the prevalence of different omics levels across various liver diseases, as well as categorise the AI methodology used across the studies. Specifically, we highlight the predominance of transcriptomic and genomic profiling and the relatively sparse exploration of other levels such as the proteome and methylome, which represent untapped potential for novel insights. Publicly available database initiatives such as The Cancer Genome Atlas and The International Cancer Genome Consortium have paved the way for advancements in the diagnosis and treatment of hepatocellular carcinoma. However, the same availability of large omics datasets remains limited for other liver diseases. Furthermore, the application of sophisticated AI methods to handle the complexities of multiomics datasets requires substantial data to train and validate the models and faces challenges in achieving bias-free results with clinical utility. Strategies to address the paucity of data and capitalise on opportunities are discussed. Given the substantial global burden of chronic liver diseases, it is imperative that multicentre collaborations be established to generate large-scale omics data for early disease recognition and intervention. Exploring advanced AI methods is also necessary to maximise the potential of these datasets and improve early detection and personalised treatment strategies.
Collapse
Affiliation(s)
- Soumita Ghosh
- Transplant AI Initiative, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Xun Zhao
- Transplant AI Initiative, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Mouaid Alim
- Transplant AI Initiative, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Michael Brudno
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Vector Institute of Artificial Intelligence, Toronto, Ontario, Canada
| | - Mamatha Bhat
- Transplant AI Initiative, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Gastroenterology, University of Toronto Faculty of Medicine, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Nychas E, Marfil-Sánchez A, Chen X, Mirhakkak M, Li H, Jia W, Xu A, Nielsen HB, Nieuwdorp M, Loomba R, Ni Y, Panagiotou G. Discovery of robust and highly specific microbiome signatures of non-alcoholic fatty liver disease. MICROBIOME 2025; 13:10. [PMID: 39810263 PMCID: PMC11730835 DOI: 10.1186/s40168-024-01990-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND The pathogenesis of non-alcoholic fatty liver disease (NAFLD) with a global prevalence of 30% is multifactorial and the involvement of gut bacteria has been recently proposed. However, finding robust bacterial signatures of NAFLD has been a great challenge, mainly due to its co-occurrence with other metabolic diseases. RESULTS Here, we collected public metagenomic data and integrated the taxonomy profiles with in silico generated community metabolic outputs, and detailed clinical data, of 1206 Chinese subjects w/wo metabolic diseases, including NAFLD (obese and lean), obesity, T2D, hypertension, and atherosclerosis. We identified highly specific microbiome signatures through building accurate machine learning models (accuracy = 0.845-0.917) for NAFLD with high portability (generalizable) and low prediction rate (specific) when applied to other metabolic diseases, as well as through a community approach involving differential co-abundance ecological networks. Moreover, using these signatures coupled with further mediation analysis and metabolic dependency modeling, we propose synergistic defined microbial consortia associated with NAFLD phenotype in overweight and lean individuals, respectively. CONCLUSION Our study reveals robust and highly specific NAFLD signatures and offers a more realistic microbiome-therapeutics approach over individual species for this complex disease. Video Abstract.
Collapse
Affiliation(s)
- Emmanouil Nychas
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany
| | - Andrea Marfil-Sánchez
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany
| | - Xiuqiang Chen
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany
| | - Mohammad Mirhakkak
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany
| | - Huating Li
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai, 200233, China
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai, 200233, China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | | | - Max Nieuwdorp
- Amsterdam UMC, Location AMC, Department of Vascular Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Rohit Loomba
- Department of Medicine, MASLD Research Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yueqiong Ni
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany.
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai, 200233, China.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany.
- Faculty of Biological Sciences, Friedrich Schiller University, Jena, 07745, Germany.
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
5
|
Jiang C, Wang L, Yu CQ, You ZH, Wang XF, Wei MM, Shi TL, Liang SZ, Wang DW. Hither-CMI: Prediction of circRNA-miRNA Interactions Based on a Hybrid Multimodal Network and Higher-Order Neighborhood Information via a Graph Convolutional Network. J Chem Inf Model 2025; 65:446-459. [PMID: 39686716 DOI: 10.1021/acs.jcim.4c01991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Numerous studies show that circular RNA (circRNA) functions as a sponge for microRNA (miRNA), significantly regulating gene expression by interacting with miRNA, which in turn affects the progression of human diseases. Traditional experimental approaches for investigating circRNA-miRNA interactions (CMI) are both time-consuming and costly, making computational methods a valuable alternative. Hence, we propose a computational model for predicting CMI, leveraging a hybrid multimodal network and higher-order neighborhood information (Hither-CMI). Specifically, Hither-CMI employs Multiple Kernel Learning (MKL) to integrate sequence, structure, and expression similarity networks of circRNA and miRNA, resulting in a hybrid multimodal network. Next, an enhanced Graph Convolutional Network (GCN) is utilized to combine the circRNA-miRNA hybrid multimodal network with the CMI association network, producing a hybrid higher-order embedding representation. Finally, the XGBoost classifier is applied for training and prediction. The Hither-CMI model achieved a predicted AUC value of 0.9134. In case studies, 25 out of the top 30 predicted CMI were confirmed by recent literature. These extensive experimental results further validate the effectiveness of Hither-CMI in predicting potential CMI, making it a promising prescreening tool for further biological research.
Collapse
Affiliation(s)
- Chen Jiang
- School of Information Engineering, Xijing Univerity, Xi'an 710123, China
| | - Lei Wang
- Guangxi Academy of Science, Nanning 530007, China
- School of Computer Science and Technology, China University of Mining and Technology, Xuzhou 221116, China
| | - Chang-Qing Yu
- School of Information Engineering, Xijing Univerity, Xi'an 710123, China
| | - Zhu-Hong You
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710129, China
| | - Xin-Fei Wang
- College of Computer Science and Technology, Jilin University, Changchun 130012, China
| | - Meng-Meng Wei
- School of Computer Science and Technology, China University of Mining and Technology, Xuzhou 221116, China
| | - Tai-Long Shi
- School of Information Engineering, Xijing Univerity, Xi'an 710123, China
| | - Si-Zhe Liang
- School of Information Engineering, Xijing Univerity, Xi'an 710123, China
| | - Deng-Wu Wang
- School of Information Engineering, Xijing Univerity, Xi'an 710123, China
| |
Collapse
|
6
|
Karwowska Z, Aasmets O, Kosciolek T, Org E. Effects of data transformation and model selection on feature importance in microbiome classification data. MICROBIOME 2025; 13:2. [PMID: 39754220 PMCID: PMC11699698 DOI: 10.1186/s40168-024-01996-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 12/04/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Accurate classification of host phenotypes from microbiome data is crucial for advancing microbiome-based therapies, with machine learning offering effective solutions. However, the complexity of the gut microbiome, data sparsity, compositionality, and population-specificity present significant challenges. Microbiome data transformations can alleviate some of the aforementioned challenges, but their usage in machine learning tasks has largely been unexplored. RESULTS Our analysis of over 8500 samples from 24 shotgun metagenomic datasets showed that it is possible to classify healthy and diseased individuals using microbiome data with minimal dependence on the choice of algorithm or transformation. Presence-absence transformations performed comparably to abundance-based transformations, and only a small subset of predictors is necessary for accurate classification. However, while different transformations resulted in comparable classification performance, the most important features varied significantly, which highlights the need to reevaluate machine learning-based biomarker detection. CONCLUSIONS Microbiome data transformations can significantly influence feature selection but have a limited effect on classification accuracy. Our findings suggest that while classification is robust across different transformations, the variation in feature selection necessitates caution when using machine learning for biomarker identification. This research provides valuable insights for applying machine learning to microbiome data and identifies important directions for future work.
Collapse
Affiliation(s)
- Zuzanna Karwowska
- Małopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
- Sano Centre for Computational Medicine, Krakow, Poland
| | - Oliver Aasmets
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tomasz Kosciolek
- Małopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland.
- Sano Centre for Computational Medicine, Krakow, Poland.
| | - Elin Org
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia.
| |
Collapse
|
7
|
Beck KL, Haiminen N, Agarwal A, Carrieri AP, Madgwick M, Kelly J, Pylro V, Kawas B, Wiedmann M, Ganda E. Development and evaluation of statistical and artificial intelligence approaches with microbial shotgun metagenomics data as an untargeted screening tool for use in food production. mSystems 2024; 9:e0084024. [PMID: 39387577 PMCID: PMC11575248 DOI: 10.1128/msystems.00840-24] [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: 07/01/2024] [Accepted: 07/26/2024] [Indexed: 10/15/2024] Open
Abstract
The increasing knowledge of microbial ecology in food products relating to quality and safety and the established usefulness of machine learning algorithms for anomaly detection in multiple scenarios suggests that the application of microbiome data in food production systems for anomaly detection could be a valuable approach to be used in food systems. These methods could be used to identify ingredients that deviate from their typical microbial composition, which could indicate food fraud or safety issues. The objective of this study was to assess the feasibility of using shotgun sequencing data as input into anomaly detection algorithms using fluid milk as a model system. Contrastive principal component analysis (PCA), cluster-based methods, and explainable artificial intelligence (AI) were evaluated for the detection of two anomalous sample classes using longitudinal metagenomic profiling of fluid milk compared to baseline (BL) samples collected under comparable circumstances. Traditional methods (alpha and beta diversity, clustering-based contrastive PCA, multidimensional scaling, and dendrograms) failed to differentiate anomalous sample classes; however, explainable AI was able to classify anomalous vs baseline samples and indicate microbial drivers in association with antibiotic use. We validated the potential for explainable AI to classify different milk sources using larger publicly available fluid milk 16S rDNA sequencing data sets and demonstrated that explainable AI is able to differentiate between milk storage methods, processing stages, and seasons. Our results indicate that the application of artificial intelligence continues to hold promise in the realm of microbiome data analysis and could present further opportunities for downstream analytic automation to aid in food safety and quality. IMPORTANCE We evaluated the feasibility of using untargeted metagenomic sequencing of raw milk for detecting anomalous food ingredient content with artificial intelligence methods in a study specifically designed to test this hypothesis. We also show through analysis of publicly available fluid milk microbial data that our artificial intelligence approach is able to successfully predict milk in different stages of processing. The approach could potentially be applied in the food industry for safety and quality control.
Collapse
Affiliation(s)
| | - Niina Haiminen
- IBM T.J. Watson Research Center, New York, New York, USA
| | | | | | | | - Jennifer Kelly
- IBM Research Europe-Daresbury, Warrington, United Kingdom
| | - Victor Pylro
- Department of Biology, Federal University of Lavras, Lavras, Minas Gerais, Brazil
| | - Ban Kawas
- IBM Almaden Research Center, San Jose, California, USA
| | - Martin Wiedmann
- Cornell University, College of Agriculture and Life Sciences, Ithaca, New York, USA
| | - Erika Ganda
- Department of Animal Science, Pennsylvania State University, University Park, Pennsylvania, USA
- The One Health Microbiome Center, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
8
|
Dey P, Choubey S. An urgent need for longitudinal microbiome profiling coupled with machine learning interventions. Front Microbiol 2024; 15:1487841. [PMID: 39588102 PMCID: PMC11586336 DOI: 10.3389/fmicb.2024.1487841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/18/2024] [Indexed: 11/27/2024] Open
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Sandeep Choubey
- The Institute of Mathematical Sciences, Chennai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
9
|
Wang Q, Tang X, Qiao W, Sun L, Shi H, Chen D, Xu B, Liu Y, Zhao J, Huang C, Jin R. Machine learning-based characterization of the gut microbiome associated with the progression of primary biliary cholangitis to cirrhosis. Microbes Infect 2024; 26:105368. [PMID: 38797428 DOI: 10.1016/j.micinf.2024.105368] [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: 11/18/2023] [Revised: 04/20/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Primary biliary cholangitis (PBC) is associated closely with the gut microbiota. This study aimed to explore the characteristics of the gut microbiota after the progress of PBC to cirrhosis. METHOD This study focuses on utilizing the 16S rRNA gene sequencing method to screen for differences in gut microbiota in PBC patients who progress to cirrhosis. Then, we divided the data into training and verification sets and used seven different machine learning (ML) models to validate them respectively, calculating and comparing the accuracy, F1 score, precision, and recall, and screening the dominant intestinal flora affecting PBC cirrhosis. RESULT PBC cirrhosis patients showed decreased diversity and richness of gut microbiota. Additionally, there are alterations in the composition of gut microbiota in PBC cirrhosis patients. The abundance of Faecalibacterium and Gemmiger bacteria significantly decreases, while the abundance of Veillonella and Streptococcus significantly increases. Furthermore, machine learning methods identify Streptococcus and Gemmiger as the predominant gut microbiota in PBC patients with cirrhosis, serving as non-invasive biomarkers (AUC = 0.902). CONCLUSION Our study revealed that PBC cirrhosis patients gut microbiota composition and function have significantly changed. Streptococcus and Gemmiger may become a non-invasive biomarker for predicting the progression of PBC progress to cirrhosis.
Collapse
Affiliation(s)
- Qi Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China; Beijing Institute of Infectious Diseases, Beijing, PR China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China
| | - Xiaomeng Tang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China; Beijing Institute of Infectious Diseases, Beijing, PR China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China
| | - Wenying Qiao
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China; Beijing Institute of Infectious Diseases, Beijing, PR China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China; Changping Laboratory, Beijing, PR China
| | - Lina Sun
- Beijing Institute of Hepatology, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China
| | - Han Shi
- Beijing Institute of Hepatology, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China
| | - Dexi Chen
- Beijing Institute of Hepatology, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China
| | - Bin Xu
- Second Department of Liver Disease Center, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China
| | - Yanmin Liu
- Second Department of Liver Disease Center, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China
| | - Juan Zhao
- Second Department of Liver Disease Center, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China
| | - Chunyang Huang
- Second Department of Liver Disease Center, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China.
| | - Ronghua Jin
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China; Beijing Institute of Infectious Diseases, Beijing, PR China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China; Changping Laboratory, Beijing, PR China.
| |
Collapse
|
10
|
Liu M, Li Y, Sun L, Sun M, Hu X, Li Q, Yu M, Wang C, Ren X, Ma J. Integrating Multi-Organ Imaging-Derived Phenotypes and Genomic Information for Predicting the Occurrence of Common Diseases. Bioengineering (Basel) 2024; 11:872. [PMID: 39329614 PMCID: PMC11428582 DOI: 10.3390/bioengineering11090872] [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: 06/12/2024] [Revised: 07/17/2024] [Accepted: 08/16/2024] [Indexed: 09/28/2024] Open
Abstract
As medical imaging technologies advance, these tools are playing a more and more important role in assisting clinical disease diagnosis. The fusion of biomedical imaging and multi-modal information is profound, as it significantly enhances diagnostic precision and comprehensiveness. Integrating multi-organ imaging with genomic information can significantly enhance the accuracy of disease prediction because many diseases involve both environmental and genetic determinants. In the present study, we focused on the fusion of imaging-derived phenotypes (IDPs) and polygenic risk score (PRS) of diseases from different organs including the brain, heart, lung, liver, spleen, pancreas, and kidney for the prediction of the occurrence of nine common diseases, namely atrial fibrillation, heart failure (HF), hypertension, myocardial infarction, asthma, type 2 diabetes, chronic kidney disease, coronary artery disease (CAD), and chronic obstructive pulmonary disease, in the UK Biobank (UKBB) dataset. For each disease, three prediction models were developed utilizing imaging features, genomic data, and a fusion of both, respectively, and their performances were compared. The results indicated that for seven diseases, the model integrating both imaging and genomic data achieved superior predictive performance compared to models that used only imaging features or only genomic data. For instance, the Area Under Curve (AUC) of HF risk prediction was increased from 0.68 ± 0.15 to 0.79 ± 0.12, and the AUC of CAD diagnosis was increased from 0.76 ± 0.05 to 0.81 ± 0.06.
Collapse
Affiliation(s)
- Meng Liu
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Yan Li
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China;
| | - Longyu Sun
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Mengting Sun
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Xumei Hu
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Qing Li
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Mengyao Yu
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Chengyan Wang
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Xinping Ren
- Ultrasound Department, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Jinlian Ma
- Radiology Department, Jiangyin Affiliated Hospital of Nanjing University of Chinese Medicine, 130 Renmin Middle Road, Jiangyin 214400, China
| |
Collapse
|
11
|
Park IG, Yoon SJ, Won SM, Oh KK, Hyun JY, Suk KT, Lee U. Gut microbiota-based machine-learning signature for the diagnosis of alcohol-associated and metabolic dysfunction-associated steatotic liver disease. Sci Rep 2024; 14:16122. [PMID: 38997279 PMCID: PMC11245548 DOI: 10.1038/s41598-024-60768-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/26/2024] [Indexed: 07/14/2024] Open
Abstract
Alcoholic-associated liver disease (ALD) and metabolic dysfunction-associated steatotic liver disease (MASLD) show a high prevalence rate worldwide. As gut microbiota represents current state of ALD and MASLD via gut-liver axis, typical characteristics of gut microbiota can be used as a potential diagnostic marker in ALD and MASLD. Machine learning (ML) algorithms improve diagnostic performance in various diseases. Using gut microbiota-based ML algorithms, we evaluated the diagnostic index for ALD and MASLD. Fecal 16S rRNA sequencing data of 263 ALD (control, elevated liver enzyme [ELE], cirrhosis, and hepatocellular carcinoma [HCC]) and 201 MASLD (control and ELE) subjects were collected. For external validation, 126 ALD and 84 MASLD subjects were recruited. Four supervised ML algorithms (support vector machine, random forest, multilevel perceptron, and convolutional neural network) were used for classification with 20, 40, 60, and 80 features, in which three nonsupervised ML algorithms (independent component analysis, principal component analysis, linear discriminant analysis, and random projection) were used for feature reduction. A total of 52 combinations of ML algorithms for each pair of subgroups were performed with 60 hyperparameter variations and Stratified ShuffleSplit tenfold cross validation. The ML models of the convolutional neural network combined with principal component analysis achieved areas under the receiver operating characteristic curve (AUCs) > 0.90. In ALD, the diagnostic AUC values of the ML strategy (vs. control) were 0.94, 0.97, and 0.96 for ELE, cirrhosis, and liver cancer, respectively. The AUC value (vs. control) for MASLD (ELE) was 0.93. In the external validation, the AUC values of ALD and MASLD (vs control) were > 0.90 and 0.88, respectively. The gut microbiota-based ML strategy can be used for the diagnosis of ALD and MASLD.ClinicalTrials.gov NCT04339725.
Collapse
Affiliation(s)
- In-Gyu Park
- Department of Electrical Engineering, Hallym University, Gyo-dong, Chuncheon, 24253, Republic of Korea
| | - Sang Jun Yoon
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, 24253, Republic of Korea
| | - Sung-Min Won
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, 24253, Republic of Korea
| | - Ki-Kwang Oh
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, 24253, Republic of Korea
| | - Ji Ye Hyun
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, 24253, Republic of Korea
| | - Ki Tae Suk
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, 24253, Republic of Korea.
- Department of Internal Medicine, Hallym University Chuncheon Sacred Heart Hospital, Hallym University, Gyo-dong, Chuncheon, 24253, Republic of Korea.
| | - Unjoo Lee
- Department of Electrical Engineering, Hallym University, Gyo-dong, Chuncheon, 24253, Republic of Korea.
| |
Collapse
|
12
|
Liu Y, Fachrul M, Inouye M, Méric G. Harnessing human microbiomes for disease prediction. Trends Microbiol 2024; 32:707-719. [PMID: 38246848 DOI: 10.1016/j.tim.2023.12.004] [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] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024]
Abstract
The human microbiome has been increasingly recognized as having potential use for disease prediction. Predicting the risk, progression, and severity of diseases holds promise to transform clinical practice, empower patient decisions, and reduce the burden of various common diseases, as has been demonstrated for cardiovascular disease or breast cancer. Combining multiple modifiable and non-modifiable risk factors, including high-dimensional genomic data, has been traditionally favored, but few studies have incorporated the human microbiome into models for predicting the prospective risk of disease. Here, we review research into the use of the human microbiome for disease prediction with a particular focus on prospective studies as well as the modulation and engineering of the microbiome as a therapeutic strategy.
Collapse
Affiliation(s)
- Yang Liu
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK; Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia; Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK; British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Muhamad Fachrul
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia; Human Genomics and Evolution Unit, St Vincent's Institute of Medical Research, Victoria, Australia; Melbourne Integrative Genomics, University of Melbourne, Parkville, Victoria, Australia; School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK; Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK; British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK; Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK; British Heart Foundation Cambridge Centre of Research Excellence, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Guillaume Méric
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Medical Science, Molecular Epidemiology, Uppsala University, Uppsala, Sweden; Department of Cardiovascular Research, Translation, and Implementation, La Trobe University, Melbourne, Victoria, Australia.
| |
Collapse
|
13
|
Chen B, Zeng G, Sun L, Jiang C. When smoke meets gut: deciphering the interactions between tobacco smoking and gut microbiota in disease development. SCIENCE CHINA. LIFE SCIENCES 2024; 67:854-864. [PMID: 38265598 DOI: 10.1007/s11427-023-2446-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/09/2023] [Indexed: 01/25/2024]
Abstract
Tobacco smoking is a prevalent and detrimental habit practiced worldwide, increasing the risk of various diseases, including chronic obstructive pulmonary disease (COPD), cardiovascular disease, liver disease, and cancer. Although previous research has explored the detrimental health effects of tobacco smoking, recent studies suggest that gut microbiota dysbiosis may play a critical role in these outcomes. Numerous tobacco smoke components, such as nicotine, are found in the gastrointestinal tract and interact with gut microbiota, leading to lasting impacts on host health and diseases. This review delves into the ways tobacco smoking and its various constituents influence gut microbiota composition and functionality. We also summarize recent advancements in understanding how tobacco smoking-induced gut microbiota dysbiosis affects host health. Furthermore, this review introduces a novel perspective on how changes in gut microbiota following smoking cessation may contribute to withdrawal syndrome and the degree of health improvements in smokers.
Collapse
Affiliation(s)
- Bo Chen
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Guangyi Zeng
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Lulu Sun
- State Key Laboratory of Women's Reproductive Health and Fertility Promotion, Peking University, Beijing, 100191, China.
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, 100191, China.
| | - Changtao Jiang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China.
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
- Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Women's Reproductive Health and Fertility Promotion, Peking University, Beijing, 100191, China.
| |
Collapse
|
14
|
Zheng X, Zheng Y, Chen T, Hou C, Zhou L, Liu C, Zheng J, Hu R. Effect of Laryngopharyngeal Reflux and Potassium-Competitive Acid Blocker (P-CAB) on the Microbiological Comprise of the Laryngopharynx. Otolaryngol Head Neck Surg 2024; 170:1380-1390. [PMID: 38385787 DOI: 10.1002/ohn.682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/27/2023] [Accepted: 01/21/2024] [Indexed: 02/23/2024]
Abstract
OBJECTIVE To probe the microbiota composition progressing from healthy individuals to those with laryngopharyngeal reflux disease (LPRD) and subsequently undergoing potassium-competitive acid inhibitor (P-CAB) therapy. STUDY DESIGN Prospective case-control study. SETTING Academic Medical Center. METHODS Forty patients with LPRD and 51 patients without LPRD were recruited. An 8-week P-CAB therapy was initiated (post-T-LPRD), and 39 had return visits. In total, 130 laryngopharyngeal saliva samples were collected and sequenced by targeting the V3-V4 region of the 16S ribosomal RNA (rRNA) gene using an Illumina MiSeq. Amplicon sequence variants (ASVs) and clinical indices were analyzed. RESULTS Alpha and beta diversities were compared among the non-LPRD, LPRD, and post-T-LPRD groups, and the Observed_ASVs were not significantly different. At the same time, the Shannon and Simpson indices, unweighted Unifrac, weighted Unifrac, and binary Jaccard distance were significantly different between non-LPRD and LPRD groups. In addition, significant differences were found in the abundance of Streptococcus, Prevotella, and Prevotellaceae in the LPRD versus non-LPRD groups, and Neisseria, Leptotrichia, and Allprevotella in the LPRD versus post-T-LPRD groups. The genera model was used to distinguish patients with LPRD from those without, and a better receiver operating characteristic curve was formed after combining the clinical indices of reflux symptom index, reflux finding score, and pepsin, with an area under the curve of 0.960. CONCLUSION Laryngopharyngeal microbial communities changed after laryngopharyngeal reflux and were modified further after P-CAB treatment, which provides a potential diagnostic value for LPRD, especially when combined with clinical indices.
Collapse
Affiliation(s)
- Xiaowei Zheng
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yujin Zheng
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Ting Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Chenjie Hou
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Liqun Zhou
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Chaofeng Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jingyi Zheng
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Renyou Hu
- Chongqing Jinshan Science & Technology (Group) Co. Ltd., Chongqing, China
| |
Collapse
|
15
|
Liu Y, Ritchie SC, Teo SM, Ruuskanen MO, Kambur O, Zhu Q, Sanders J, Vázquez-Baeza Y, Verspoor K, Jousilahti P, Lahti L, Niiranen T, Salomaa V, Havulinna AS, Knight R, Méric G, Inouye M. Integration of polygenic and gut metagenomic risk prediction for common diseases. NATURE AGING 2024; 4:584-594. [PMID: 38528230 PMCID: PMC11031402 DOI: 10.1038/s43587-024-00590-7] [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: 08/11/2023] [Accepted: 02/13/2024] [Indexed: 03/27/2024]
Abstract
Multiomics has shown promise in noninvasive risk profiling and early detection of various common diseases. In the present study, in a prospective population-based cohort with ~18 years of e-health record follow-up, we investigated the incremental and combined value of genomic and gut metagenomic risk assessment compared with conventional risk factors for predicting incident coronary artery disease (CAD), type 2 diabetes (T2D), Alzheimer disease and prostate cancer. We found that polygenic risk scores (PRSs) improved prediction over conventional risk factors for all diseases. Gut microbiome scores improved predictive capacity over baseline age for CAD, T2D and prostate cancer. Integrated risk models of PRSs, gut microbiome scores and conventional risk factors achieved the highest predictive performance for all diseases studied compared with models based on conventional risk factors alone. The present study demonstrates that integrated PRSs and gut metagenomic risk models improve the predictive value over conventional risk factors for common chronic diseases.
Collapse
Affiliation(s)
- Yang Liu
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- Department of Clinical Pathology, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| | - Scott C Ritchie
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cambridge Centre of Research Excellence, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Shu Mei Teo
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Matti O Ruuskanen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Computing, University of Turku, Turku, Finland
| | - Oleg Kambur
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Qiyun Zhu
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, USA
| | - Jon Sanders
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY, USA
| | - Yoshiki Vázquez-Baeza
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - Karin Verspoor
- School of Computing Technologies, RMIT University, Melbourne, Victoria, Australia
- School of Computing and Information Systems, University of Melbourne, Melbourne, Victoria, Australia
| | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Teemu Niiranen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Division of Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Aki S Havulinna
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Institute for Molecular Medicine Finland, FIMM-HiLIFE, University of Helsinki, Helsinki, Finland
| | - Rob Knight
- Center for Microbiome Innovation, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Guillaume Méric
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- Department of Clinical Pathology, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cambridge Centre of Research Excellence, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK.
- The Alan Turing Institute, London, UK.
| |
Collapse
|
16
|
Nehmi‐Filho V, de Freitas JA, Franco LA, Martins RC, Turri JAO, Santamarina AB, Fonseca JVDS, Sabino EC, Moraes BC, Souza E, Murata GM, Costa SF, Alcântara PS, Otoch JP, Pessoa AFM. Modulation of the gut microbiome and Firmicutes phylum reduction by a nutraceutical blend in the obesity mouse model and overweight humans: A double-blind clinical trial. Food Sci Nutr 2024; 12:2436-2454. [PMID: 38628220 PMCID: PMC11016419 DOI: 10.1002/fsn3.3927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/12/2023] [Accepted: 12/17/2023] [Indexed: 04/19/2024] Open
Abstract
Overweight and obesity are closely linked to gut dysbiosis/dysmetabolism and disrupted De-Ritis ratio [aspartate aminotransferase (AST)/alanine aminotransferase (ALT) ratio], which may contribute to chronic noncommunicable diseases onset. Concurrently, extensive research explores nutraceuticals, and health-enhancing supplements, for disease prevention or treatment. Thus, sedentary overweight volunteers were double-blind randomized into two groups: Novel Nutraceutical_(S) (without silymarin) and Novel Nutraceutical (with silymarin). Experimental formulations were orally administered twice daily over 180 consecutive days. We evaluated fecal gut microbiota, based on partial 16S rRNA sequences, biochemistry and endocrine markers, steatosis biomarker (AST/ALT ratio), and anthropometric parameters. Post-supplementation, only the Novel Nutraceutical group reduced Clostridium clostridioforme (Firmicutes), Firmicutes/Bacteroidetes ratio (F/B ratio), and De-Ritis ratio, while elevating Bacteroides caccae and Bacteroides uniformis (Bacteroidetes) in Brazilian sedentary overweight volunteers after 180 days. In summary, the results presented here allow us to suggest the gut microbiota as the action mechanism of the Novel Nutraceutical promoting metabolic hepatic recovery in obesity/overweight non-drug interventions.
Collapse
Affiliation(s)
- Victor Nehmi‐Filho
- Laboratório de Investigação Médica (LIM‐26), Laboratório de Produtos e Derivados Naturais, Departamento de CirurgiaUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
| | - Jessica Alves de Freitas
- Laboratório de Investigação Médica (LIM‐26), Laboratório de Produtos e Derivados Naturais, Departamento de CirurgiaUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
| | - Lucas Augusto Franco
- Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Parasitologia Médica (LIM‐46)Universidade de São Paulo Instituto de Medicina Tropical de São PauloJardim AmericaBrazil
| | - Roberta Cristina Martins
- Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Parasitologia Médica (LIM‐46)Universidade de São Paulo Instituto de Medicina Tropical de São PauloJardim AmericaBrazil
| | - José Antônio Orellana Turri
- Departamento de Ginecologia e Obstetrícia, Grupo de Pesquisa em Economia da SaúdeUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
| | - Aline Boveto Santamarina
- Laboratório de Investigação Médica (LIM‐26), Laboratório de Produtos e Derivados Naturais, Departamento de CirurgiaUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
| | - Joyce Vanessa da Silva Fonseca
- Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Investigação Médica em Protozoologia, Bacteriologia e Resistência Antimicrobiana (LIM‐49)Universidade de São Paulo Instituto de Medicina Tropical de São PauloJardim AmericaBrazil
| | - Ester Cerdeira Sabino
- Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Parasitologia Médica (LIM‐46)Universidade de São Paulo Instituto de Medicina Tropical de São PauloJardim AmericaBrazil
| | - Bruna Carvalho Moraes
- Laboratório de Investigação Médica (LIM‐31), Laboratório Investigação Médica em Patogênese e Terapia dirigida em Onco‐Imuno‐HematologiaUniversidade de São Paulo Faculdade de Medicina, Universidade de São Paulo Hospital das ClínicasCerqueira CésarBrazil
| | | | - Gilson Masahiro Murata
- Laboratório de Investigação Médica (LIM‐29), Laboratório de Nefrologia Celular, Genética e Molecular, Departamento de Clínica MédicaUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
| | - Silvia Figueiredo Costa
- Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Investigação Médica em Protozoologia, Bacteriologia e Resistência Antimicrobiana (LIM‐49)Universidade de São Paulo Instituto de Medicina Tropical de São PauloJardim AmericaBrazil
| | - Paulo Sérgio Alcântara
- Departamento de CirurgiaUniversidade de São Paulo Hospital Universitário de São PauloButantãBrazil
| | - José Pinhata Otoch
- Laboratório de Investigação Médica (LIM‐26), Laboratório de Produtos e Derivados Naturais, Departamento de CirurgiaUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
- Departamento de CirurgiaUniversidade de São Paulo Hospital Universitário de São PauloButantãBrazil
| | - Ana Flávia Marçal Pessoa
- Laboratório de Investigação Médica (LIM‐26), Laboratório de Produtos e Derivados Naturais, Departamento de CirurgiaUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
- Efeom NutritionUniversidade de São Paulo Faculdade de MedicinaPacaembuBrazil
| |
Collapse
|
17
|
Yuan M, Wang Y, Tian X, Zheng W, Zuo H, Zhang X, Song H. Ferrostatin-1 improves prognosis and regulates gut microbiota of steatotic liver transplantation recipients in rats. Future Microbiol 2024; 19:413-429. [PMID: 38305222 DOI: 10.2217/fmb-2023-0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/15/2023] [Indexed: 02/03/2024] Open
Abstract
Aims: To investigate the effects of Ferrostatin-1 (Fer-1) on improving the prognosis of liver transplant recipients with steatotic liver grafts and regulating gut microbiota in rats. Methods: We obtained steatotic liver grafts and established a liver transplantation model. Recipients were divided into sham, liver transplantation and Fer-1 treatment groups, which were assessed 1 and 7 days after surgery (n = 6). Results & conclusion: Fer-1 promotes recovery of the histological structure and function of steatotic liver grafts and the intestinal tract, and improves inflammatory responses of recipients following liver transplantation. Fer-1 reduces gut microbiota pathogenicity, and lowers iron absorption and improves fat metabolism of recipients, thereby protecting steatotic liver grafts.
Collapse
Affiliation(s)
- Mengshu Yuan
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China
| | - Yuxin Wang
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China
| | - Xiaorong Tian
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China
| | - Weiping Zheng
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin, 300192, PR China
- NHC Key Laboratory of Critical Care Medicine, Tianjin, 300192, PR China
| | - Huaiwen Zuo
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China
| | - Xinru Zhang
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, PR China
| | - Hongli Song
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin, 300192, PR China
- Tianjin Key Laboratory of Organ Transplantation, Tianjin, PR China
| |
Collapse
|
18
|
Walsh C, Stallard-Olivera E, Fierer N. Nine (not so simple) steps: a practical guide to using machine learning in microbial ecology. mBio 2024; 15:e0205023. [PMID: 38126787 PMCID: PMC10865974 DOI: 10.1128/mbio.02050-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Due to the complex nature of microbiome data, the field of microbial ecology has many current and potential uses for machine learning (ML) modeling. With the increased use of predictive ML models across many disciplines, including microbial ecology, there is extensive published information on the specific ML algorithms available and how those algorithms have been applied. Thus, our goal is not to summarize the breadth of ML models available or compare their performances. Rather, our goal is to provide more concrete and actionable information to guide microbial ecologists in how to select, run, and interpret ML algorithms to predict the taxa or genes associated with particular sample categories or environmental gradients of interest. Such microbial data often have unique characteristics that require careful consideration of how to apply ML models and how to interpret the associated results. This review is intended for practicing microbial ecologists who may be unfamiliar with some of the intricacies of ML models. We provide examples and discuss common opportunities and pitfalls specific to applying ML models to the types of data sets most frequently collected by microbial ecologists.
Collapse
Affiliation(s)
- Corinne Walsh
- Cooperative Institute of Research in Environmental Sciences, CU Boulder, Boulder, Colorado, USA
- Ecology and Evolutionary Biology Department, CU Boulder, Boulder, Colorado, USA
| | - Elías Stallard-Olivera
- Cooperative Institute of Research in Environmental Sciences, CU Boulder, Boulder, Colorado, USA
- Ecology and Evolutionary Biology Department, CU Boulder, Boulder, Colorado, USA
| | - Noah Fierer
- Cooperative Institute of Research in Environmental Sciences, CU Boulder, Boulder, Colorado, USA
- Ecology and Evolutionary Biology Department, CU Boulder, Boulder, Colorado, USA
| |
Collapse
|
19
|
Zhou D, Chen Y, Wang Z, Zhu S, Zhang L, Song J, Bai T, Hou X. Integrating clinical and cross-cohort metagenomic features: a stable and non-invasive colorectal cancer and adenoma diagnostic model. Front Mol Biosci 2024; 10:1298679. [PMID: 38455360 PMCID: PMC10919151 DOI: 10.3389/fmolb.2023.1298679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/24/2023] [Indexed: 03/09/2024] Open
Abstract
Background: Dysbiosis is associated with colorectal cancer (CRC) and adenomas (CRA). However, the robustness of diagnostic models based on microbial signatures in multiple cohorts remains unsatisfactory. Materials and Methods: In this study, we used machine learning models to screen metagenomic signatures from the respective cross-cohort datasets of CRC and CRA (selected from CuratedMetagenomicData, each disease included 4 datasets). Then select a CRC and CRA data set from the CuratedMetagenomicData database and meet the requirements of having both metagenomic data and clinical data. This data set will be used to verify the inference that integrating clinical features can improve the performance of microbial disease prediction models. Results: After repeated verification, we selected 20 metagenomic features that performed well and were stably expressed within cross-cohorts to represent the diagnostic role of bacterial communities in CRC/CRA. The performance of the selected cross-cohort metagenomic features was stable for multi-regional and multi-ethnic populations (CRC, AUC: 0.817-0.867; CRA, AUC: 0.766-0.833). After clinical feature combination, AUC of our integrated CRC diagnostic model reached 0.939 (95% CI: 0.932-0.947, NRI=30%), and that of the CRA integrated model reached 0.925 (95%CI: 0.917-0.935, NRI=18%). Conclusion: In conclusion, the integrated model performed significantly better than single microbiome or clinical feature models in all cohorts. Integrating cross-cohort common discriminative microbial features with clinical features could help construct stable diagnostic models for early non-invasive screening for CRC and CRA.
Collapse
Affiliation(s)
- Dan Zhou
- Division of Gastroenterology, Union Hospital, Tongji Medical College Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Youli Chen
- State Key Laboratory for Oncogenes and Related Genes, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zehao Wang
- School of Management, Huazhong University of Science and Technology, Wuhan, China
| | - Siran Zhu
- Division of Gastroenterology, Union Hospital, Tongji Medical College Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Song
- Division of Gastroenterology, Union Hospital, Tongji Medical College Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Bai
- Division of Gastroenterology, Union Hospital, Tongji Medical College Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Wu J, Singleton SS, Bhuiyan U, Krammer L, Mazumder R. Multi-omics approaches to studying gastrointestinal microbiome in the context of precision medicine and machine learning. Front Mol Biosci 2024; 10:1337373. [PMID: 38313584 PMCID: PMC10834744 DOI: 10.3389/fmolb.2023.1337373] [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: 11/15/2023] [Accepted: 12/27/2023] [Indexed: 02/06/2024] Open
Abstract
The human gastrointestinal (gut) microbiome plays a critical role in maintaining host health and has been increasingly recognized as an important factor in precision medicine. High-throughput sequencing technologies have revolutionized -omics data generation, facilitating the characterization of the human gut microbiome with exceptional resolution. The analysis of various -omics data, including metatranscriptomics, metagenomics, glycomics, and metabolomics, holds potential for personalized therapies by revealing information about functional genes, microbial composition, glycans, and metabolites. This multi-omics approach has not only provided insights into the role of the gut microbiome in various diseases but has also facilitated the identification of microbial biomarkers for diagnosis, prognosis, and treatment. Machine learning algorithms have emerged as powerful tools for extracting meaningful insights from complex datasets, and more recently have been applied to metagenomics data via efficiently identifying microbial signatures, predicting disease states, and determining potential therapeutic targets. Despite these rapid advancements, several challenges remain, such as key knowledge gaps, algorithm selection, and bioinformatics software parametrization. In this mini-review, our primary focus is metagenomics, while recognizing that other -omics can enhance our understanding of the functional diversity of organisms and how they interact with the host. We aim to explore the current intersection of multi-omics, precision medicine, and machine learning in advancing our understanding of the gut microbiome. A multidisciplinary approach holds promise for improving patient outcomes in the era of precision medicine, as we unravel the intricate interactions between the microbiome and human health.
Collapse
Affiliation(s)
- Jingyue Wu
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Stephanie S. Singleton
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Urnisha Bhuiyan
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Lori Krammer
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
- Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Raja Mazumder
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
- The McCormick Genomic and Proteomic Center, The George Washington University, Washington, DC, United States
| |
Collapse
|
21
|
Effenberger M, Grander C, Grabherr F, Tilg H. Nonalcoholic Fatty Liver Disease and the Intestinal Microbiome: An Inseparable Link. J Clin Transl Hepatol 2023; 11:1498-1507. [PMID: 38161503 PMCID: PMC10752805 DOI: 10.14218/jcth.2023.00069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/21/2023] [Accepted: 07/18/2023] [Indexed: 01/03/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) particularly affects patients with type 2 diabetes and obesity. The incidence of NAFLD has increased significantly over the last decades and is now pandemically across the globe. It is a complex systemic disease comprising hepatic lipid accumulation, inflammation, lipotoxicity, gut dysbiosis, and insulin resistance as main features and with the potential to progress to cirrhosis and hepatocellular carcinoma (HCC). In numerous animal and human studies the gut microbiota plays a key role in the pathogenesis of NAFLD, NAFLD-cirrhosis and NAFLD-associated HCC. Lipotoxicity is the driver of inflammation, insulin resistance, and liver injury. Likewise, western diet, obesity, and metabolic disorders may alter the gut microbiota, which activates innate and adaptive immune responses and fuels hereby hepatic and systemic inflammation. Indigestible carbohydrates are fermented by the gut microbiota to produce important metabolites, such as short-chain fatty acids and succinate. Numerous animal and human studies suggested a pivotal role of these metabolites in the progression of NAFLD and its comorbidities. Though, modification of the gut microbiota and/or the metabolites could even be beneficial in patients with NAFLD, NAFLD-cirrhosis, and NAFLD-associated HCC. In this review we collect the evidence that exogenous and endogenous hits drive liver injury in NAFLD and propel liver fibrosis and the progressing to advanced disease stages. NAFLD can be seen as the product of a complex interplay between gut microbiota, the immune response and metabolism. Thus, the challenge will be to understand its pathogenesis and to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Maria Effenberger
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
22
|
Díaz LA, Arab JP, Louvet A, Bataller R, Arrese M. The intersection between alcohol-related liver disease and nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol 2023; 20:764-783. [PMID: 37582985 DOI: 10.1038/s41575-023-00822-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 08/17/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and alcohol-related liver disease (ALD) are the leading causes of chronic liver disease worldwide. NAFLD and ALD share pathophysiological, histological and genetic features and both alcohol and metabolic dysfunction coexist as aetiological factors in many patients with hepatic steatosis. A diagnosis of NAFLD requires the exclusion of significant alcohol consumption and other causes of liver disease. However, data suggest that significant alcohol consumption is often under-reported in patients classified as having NAFLD and that alcohol and metabolic factors interact to exacerbate the progression of liver disease. In this Review, we analyse existing data on the interaction between alcohol consumption and metabolic syndrome as well as the overlapping features and differences in the pathogenesis of ALD and NAFLD. We also discuss the clinical implications of the coexistence of alcohol consumption, of any degree, in patients with evidence of metabolic derangement as well as the use of alcohol biomarkers to detect alcohol intake. Finally, we summarize the evolving nomenclature of fatty liver disease and describe a recent proposal to classify patients at the intersection of NAFLD and ALD. We propose that, regardless of the presumed aetiology, patients with fatty liver disease should be evaluated for both metabolic syndrome and alcohol consumption to enable better prognostication and a personalized medicine approach.
Collapse
Affiliation(s)
- Luis Antonio Díaz
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Pablo Arab
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University & London Health Sciences Centre, London, Ontario, Canada
- Department of Epidemiology and Biostatistics, Schulich School of Medicine, Western University, London, Ontario, Canada
| | - Alexandre Louvet
- Service des Maladies de l'Appareil Digestif, Hôpital Huriez, Lille Cedex, France
- Université Lille Nord de France, Lille, France
- Unité INSERM INFINITE 1286, Lille, France
| | - Ramón Bataller
- Liver Unit, Hospital Clinic, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
23
|
Nie D, Li C, Zhang Y. PitNETs and the gut microbiota: potential connections, future directions. Front Endocrinol (Lausanne) 2023; 14:1255911. [PMID: 38027221 PMCID: PMC10657991 DOI: 10.3389/fendo.2023.1255911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
The role of the gut microbiome has been widely discussed in numerous works of literature. The biggest concern is the association of the gut microbiome with the central nervous system through the microbiome-brain-gut axis in the past ten years. As more and more research has been done on the relationship between the disease of the central nervous system and gut microbes. This fact is being revealed that gut microbes seem to play an important role from the onset and progression of the disease to clinical symptoms, and new treatments. As a special tumor of the central nervous system, pituitary neuroendocrine tumors (PitNETs)are closely related to metabolism, endocrinology, and immunity. These factors are the vectors through which intestinal microbes interact with the central nervous system. However, little is known about the effects of gut microbes on the PitNET. In this review, the relationship of gut microbiota in PitNETs is introduced, the potential effects of the gut-brain axis in this relationship are analyzed, and future research directions are presented.
Collapse
Affiliation(s)
| | | | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Prince DS, Nash E, Liu K. Alcohol-Associated Liver Disease: Evolving Concepts and Treatments. Drugs 2023; 83:1459-1474. [PMID: 37747685 PMCID: PMC10624727 DOI: 10.1007/s40265-023-01939-9] [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] [Accepted: 09/03/2023] [Indexed: 09/26/2023]
Abstract
Alcohol is a prominent cause of liver disease worldwide with higher prevalence in developed nations. The spectrum of alcohol-associated liver disease (ALD) encompasses a diverse range of clinical entities, from asymptomatic isolated steatosis to decompensated cirrhosis, and in some cases, acute or chronic liver failure. Consequently, it is important for healthcare practitioners to maintain awareness and systematically screen for ALD. The optimal evaluation and management of ALD necessitates a collaborative approach, incorporating a multidisciplinary team and accounting for concurrent medical conditions. A repertoire of therapeutic interventions exists to support patients in achieving alcohol cessation and sustaining remission, with complete abstinence being the ultimate objective. This review explores the existing therapeutic options for ALD acknowledging geographical discrepancies in accessibility. Recent innovations, including the inclusion of alcohol consumption biomarkers into clinical protocols and the expansion of liver transplantation eligibility to encompass severe alcohol-associated hepatitis, are explored.
Collapse
Affiliation(s)
- David Stephen Prince
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
- Department of Gastroenterology and Liver, Liverpool Hospital, Sydney, NSW, Australia.
- Liver Injury and Cancer Program, Centenary Institute, Sydney, NSW, Australia.
- The Ingham Institute for Applied Medical Research, Sydney, NSW, Australia.
- Faculty of Medicine and Health, The University of New South Wales, Sydney, NSW, Australia.
| | - Emily Nash
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Ken Liu
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Liver Injury and Cancer Program, Centenary Institute, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
25
|
Korobeinikova AV, Zlobovskaya OA, Sheptulina AF, Ashniev GA, Bobrova MM, Yafarova AA, Akasheva DU, Kabieva SS, Bakoev SY, Zagaynova AV, Lukashina MV, Abramov IA, Pokrovskaya MS, Doludin YV, Tolkacheva LR, Kurnosov AS, Zyatenkova EV, Lavrenova EA, Efimova IA, Glazunova EV, Kiselev AR, Shipulin GA, Kontsevaya AV, Keskinov AA, Yudin VS, Makarov VV, Drapkina OM, Yudin SM. Gut Microbiota Patterns in Patients with Non-Alcoholic Fatty Liver Disease: A Comprehensive Assessment Using Three Analysis Methods. Int J Mol Sci 2023; 24:15272. [PMID: 37894951 PMCID: PMC10607775 DOI: 10.3390/ijms242015272] [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: 08/17/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 10/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered the most common chronic liver disease worldwide, affecting nearly 25% of the global adult population. Increasing evidence suggests that functional and compositional changes in the gut microbiota may contribute to the development and promote the progression of NAFLD. 16S rRNA gene next-generation sequencing is widely used to determine specific features of the NAFLD microbiome, but a complex system such as the gut microbiota requires a comprehensive approach. We used three different approaches: MALDI-TOF-MS of bacterial cultures, qPCR, and 16S NGS sequencing, as well as a wide variety of statistical methods to assess the differences in gut microbiota composition between NAFLD patients without significant fibrosis and the control group. The listed methods showed enrichment in Collinsella sp. and Oscillospiraceae for the control samples and enrichment in Lachnospiraceae (and in particular Dorea sp.) and Veillonellaceae in NAFLD. The families, Bifidobacteriaceae, Lactobacillaceae, and Enterococcaceae (particularly Enterococcus faecium and Enterococcus faecalis), were also found to be important taxa for NAFLD microbiome evaluation. Considering individual method observations, an increase in Candida krusei and a decrease in Bacteroides uniformis for NAFLD patients were detected using MALDI-TOF-MS. An increase in Gracilibacteraceae, Chitinophagaceae, Pirellulaceae, Erysipelatoclostridiaceae, Muribaculaceae, and Comamonadaceae, and a decrease in Acidaminococcaceae in NAFLD were observed with 16S NGS, and enrichment in Fusobacterium nucleatum was shown using qPCR analysis. These findings confirm that NAFLD is associated with changes in gut microbiota composition. Further investigations are required to determine the cause-and-effect relationships and the impact of microbiota-derived compounds on the development and progression of NAFLD.
Collapse
Affiliation(s)
- Anna V. Korobeinikova
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Olga A. Zlobovskaya
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Anna F. Sheptulina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - German A. Ashniev
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Maria M. Bobrova
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Adel A. Yafarova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Dariga U. Akasheva
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Shuanat Sh. Kabieva
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Siroj Yu. Bakoev
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Anjelica V. Zagaynova
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Maria V. Lukashina
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Ivan A. Abramov
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Mariya S. Pokrovskaya
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Yurii V. Doludin
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Larisa R. Tolkacheva
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Alexander S. Kurnosov
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Elena V. Zyatenkova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Evgeniya A. Lavrenova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Irina A. Efimova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Evgeniya V. Glazunova
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Anton R. Kiselev
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - German A. Shipulin
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Anna V. Kontsevaya
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Anton A. Keskinov
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Vladimir S. Yudin
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Valentin V. Makarov
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| | - Oxana M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, bld.3, 101990 Moscow, Russia; (A.F.S.); (A.A.Y.); (D.U.A.)
| | - Sergey M. Yudin
- Centre for Strategic Planning and Management of Biomedical Health Risks of Federal Medical Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia; (A.V.K.); (S.S.K.); (S.Y.B.); (M.V.L.); (A.S.K.)
| |
Collapse
|
26
|
Ibrahimi E, Lopes MB, Dhamo X, Simeon A, Shigdel R, Hron K, Stres B, D’Elia D, Berland M, Marcos-Zambrano LJ. Overview of data preprocessing for machine learning applications in human microbiome research. Front Microbiol 2023; 14:1250909. [PMID: 37869650 PMCID: PMC10588656 DOI: 10.3389/fmicb.2023.1250909] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Although metagenomic sequencing is now the preferred technique to study microbiome-host interactions, analyzing and interpreting microbiome sequencing data presents challenges primarily attributed to the statistical specificities of the data (e.g., sparse, over-dispersed, compositional, inter-variable dependency). This mini review explores preprocessing and transformation methods applied in recent human microbiome studies to address microbiome data analysis challenges. Our results indicate a limited adoption of transformation methods targeting the statistical characteristics of microbiome sequencing data. Instead, there is a prevalent usage of relative and normalization-based transformations that do not specifically account for the specific attributes of microbiome data. The information on preprocessing and transformations applied to the data before analysis was incomplete or missing in many publications, leading to reproducibility concerns, comparability issues, and questionable results. We hope this mini review will provide researchers and newcomers to the field of human microbiome research with an up-to-date point of reference for various data transformation tools and assist them in choosing the most suitable transformation method based on their research questions, objectives, and data characteristics.
Collapse
Affiliation(s)
- Eliana Ibrahimi
- Department of Biology, Faculty of Natural Sciences, University of Tirana, Tirana, Albania
| | - Marta B. Lopes
- Department of Mathematics, Center for Mathematics and Applications (NOVA Math), NOVA School of Science and Technology, Caparica, Portugal
- UNIDEMI, Department of Mechanical and Industrial Engineering, NOVA School of Science and Technology, Caparica, Portugal
| | - Xhilda Dhamo
- Department of Applied Mathematics, Faculty of Natural Sciences, University of Tirana, Tirana, Albania
| | - Andrea Simeon
- BioSense Institute, University of Novi Sad, Novi Sad, Serbia
| | - Rajesh Shigdel
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Karel Hron
- Department of Mathematical Analysis and Applications of Mathematics, Faculty of Science, Palacký University Olomouc, Olomouc, Czechia
| | - Blaž Stres
- Department of Catalysis and Chemical Reaction Engineering, National Institute of Chemistry, Ljubljana, Slovenia
- Faculty of Civil and Geodetic Engineering, Institute of Sanitary Engineering, Ljubljana, Slovenia
- Department of Automation, Biocybernetics and Robotics, Jožef Stefan Institute, Ljubljana, Slovenia
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Domenica D’Elia
- Department of Biomedical Sciences, National Research Council, Institute for Biomedical Technologies, Bari, Italy
| | - Magali Berland
- INRAE, MetaGenoPolis, Université Paris-Saclay, Jouy-en-Josas, France
| | - Laura Judith Marcos-Zambrano
- Computational Biology Group, Precision Nutrition and Cancer Research Program, IMDEA Food Institute, Madrid, Spain
| |
Collapse
|
27
|
Muralitharan RR, Snelson M, Meric G, Coughlan MT, Marques FZ. Guidelines for microbiome studies in renal physiology. Am J Physiol Renal Physiol 2023; 325:F345-F362. [PMID: 37440367 DOI: 10.1152/ajprenal.00072.2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/28/2023] [Accepted: 07/07/2023] [Indexed: 07/15/2023] Open
Abstract
Gut microbiome research has increased dramatically in the last decade, including in renal health and disease. The field is moving from experiments showing mere association to causation using both forward and reverse microbiome approaches, leveraging tools such as germ-free animals, treatment with antibiotics, and fecal microbiota transplantations. However, we are still seeing a gap between discovery and translation that needs to be addressed, so that patients can benefit from microbiome-based therapies. In this guideline paper, we discuss the key considerations that affect the gut microbiome of animals and clinical studies assessing renal function, many of which are often overlooked, resulting in false-positive results. For animal studies, these include suppliers, acclimatization, baseline microbiota and its normalization, littermates and cohort/cage effects, diet, sex differences, age, circadian differences, antibiotics and sweeteners, and models used. Clinical studies have some unique considerations, which include sampling, gut transit time, dietary records, medication, and renal phenotypes. We provide best-practice guidance on sampling, storage, DNA extraction, and methods for microbial DNA sequencing (both 16S rRNA and shotgun metagenome). Finally, we discuss follow-up analyses, including tools available, metrics, and their interpretation, and the key challenges ahead in the microbiome field. By standardizing study designs, methods, and reporting, we will accelerate the findings from discovery to translation and result in new microbiome-based therapies that may improve renal health.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Victoria, Australia
- Institute for Medical Research, Ministry of Health Malaysia, Kuala Lumpur, Malaysia
| | - Matthew Snelson
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Guillaume Meric
- Cambridge-Baker Systems Genomics Initiative, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Melinda T Coughlan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Victoria, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
28
|
Xu W, Wang T, Wang N, Zhang H, Zha Y, Ji L, Chu Y, Ning K. Artificial intelligence-enabled microbiome-based diagnosis models for a broad spectrum of cancer types. Brief Bioinform 2023; 24:7152257. [PMID: 37141141 DOI: 10.1093/bib/bbad178] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 04/07/2023] [Accepted: 04/18/2023] [Indexed: 05/05/2023] Open
Abstract
Microbiome-based diagnosis of cancer is an increasingly important supplement for the genomics approach in cancer diagnosis, yet current models for microbiome-based diagnosis of cancer face difficulties in generality: not only diagnosis models could not be adapted from one cancer to another, but models built based on microbes from tissues could not be adapted for diagnosis based on microbes from blood. Therefore, a microbiome-based model suitable for a broad spectrum of cancer types is urgently needed. Here we have introduced DeepMicroCancer, a diagnosis model using artificial intelligence techniques for a broad spectrum of cancer types. Built based on the random forest models it has enabled superior performances on more than twenty types of cancers' tissue samples. And by using the transfer learning techniques, improved accuracies could be obtained, especially for cancer types with only a few samples, which could satisfy the requirement in clinical scenarios. Moreover, transfer learning techniques have enabled high diagnosis accuracy that could also be achieved for blood samples. These results indicated that certain sets of microbes could, if excavated using advanced artificial techniques, reveal the intricate differences among cancers and healthy individuals. Collectively, DeepMicroCancer has provided a new venue for accurate diagnosis of cancer based on tissue and blood materials, which could potentially be used in clinics.
Collapse
Affiliation(s)
- Wei Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Teng Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Nan Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Haohong Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yuguo Zha
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lei Ji
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Geneis Beijing Co., Ltd., Beijing, 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
| | - Yuwen Chu
- Geneis Beijing Co., Ltd., Beijing, 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
- School of Electrical & Information Engineering, Anhui University of Technology, Anhui, 243002, China
| | - Kang Ning
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
29
|
Zhu CH, Li YX, Xu YC, Wang NN, Yan QJ, Jiang ZQ. Tamarind Xyloglucan Oligosaccharides Attenuate Metabolic Disorders via the Gut-Liver Axis in Mice with High-Fat-Diet-Induced Obesity. Foods 2023; 12:foods12071382. [PMID: 37048202 PMCID: PMC10093524 DOI: 10.3390/foods12071382] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 04/14/2023] Open
Abstract
Functional oligosaccharides exert obesity-reducing effects by acting at various pathological sites responsible for the development of obesity. In this study, tamarind xyloglucan oligosaccharides (TXOS) were used to attenuate metabolic disorders via the gut-liver axis in mice with high-fat-diet (HFD)-induced obesity, as determined through LC/MS-MS and 16S rRNA sequencing technology. A TXOS dose equivalent to 0.39 g/kg/day in humans restored the gut microbiota in obese mice, which was in part supported by the key microflora, particularly Bifidobacterium pseudolongum. Moreover, TXOS reduced the abundance of opportunistic pathogen species, such as Klebsiella variicola and Romboutsia ilealis. The bodyweight and weight gain of TXOS-treated (4.8 g/kg per day) mice began to decrease at the 14th week, decreasing by 12.8% and 23.3%, respectively. Sixteen fatty acids were identified as potential biomarkers in the liver, and B. pseudolongum and caprylic acid were found to tightly regulate each other. This was associated with reduced inflammation in the liver, circulation, and adipose tissue and protection from metabolic disorders. The findings of this study indicate that TXOS can significantly increase the gut microbiota diversity of obese mice and restore the HFD-induced dysbiosis of gut microbiota.
Collapse
Affiliation(s)
- Chun-Hua Zhu
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yan-Xiao Li
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Engineering, China Agricultural University, Beijing 100083, China
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Yun-Cong Xu
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Nan-Nan Wang
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Qiao-Juan Yan
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Engineering, China Agricultural University, Beijing 100083, China
| | - Zheng-Qiang Jiang
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| |
Collapse
|
30
|
Nie C, Xie X, Liu H, Yuan X, Ma Q, Tu A, Zhang M, Chen Z, Li J. Galactooligosaccharides ameliorate dietary advanced glycation end product-induced intestinal barrier damage in C57BL/6 mice by modulation of the intestinal microbiome. Food Funct 2023; 14:845-856. [PMID: 36537141 DOI: 10.1039/d2fo02959f] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Advanced glycation end products (AGEs) are increasingly recognized as potentially pathogenic components of processed foods, and long-term consumption of dietary AGEs triggers disruption of the intestinal barrier integrity and increases the risk of chronic diseases. Galactooligosaccharides (GOS) as prebiotics can modulate the intestinal microbiota and improve the intestinal barrier integrity. In this study, we aimed to investigate whether GOS could ameliorate the intestinal barrier damage induced by AGEs. The results showed an increased number of goblet cells (AGEs vs. H-GOS, 133.4 vs. 174.7, p < 0.05) and neutral mucin area (PAS positive area, 7.29% vs. 10.05%, p < 0.05). Upregulated expressions of occludin and claudin-1 and improved intestinal barrier integrity were observed in the H-GOS group. Using 16S rRNA sequencing analysis, we found that GOS significantly reduced the high enrichment of Akkermansia (16.95% vs. 1.29%, p < 0.05) induced by dietary AGEs while increasing the content of short-chain fatty acids. Fecal microbiota transplantation (FMT) showed that AGE-induced damage to the intestinal mucus barrier was reversed in the H-GOS transplanted group. Collectively, GOS ameliorated dietary AGE-induced intestinal barrier damage by reversing the dysregulated state of the intestinal microbiota. Our study lays the foundation for further research on dietary guidelines for populations with high AGE diets.
Collapse
Affiliation(s)
- Chenxi Nie
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Xiaoqing Xie
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Huicui Liu
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Xiaojin Yuan
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Qingyu Ma
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Aobai Tu
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Min Zhang
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Zhifei Chen
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| | - Juxiu Li
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi Province 712100, China.
| |
Collapse
|
31
|
Barbieri E, Santoro N, Umano GR. Clinical features and metabolic complications for non-alcoholic fatty liver disease (NAFLD) in youth with obesity. Front Endocrinol (Lausanne) 2023; 14:1062341. [PMID: 36733529 PMCID: PMC9887046 DOI: 10.3389/fendo.2023.1062341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 01/02/2023] [Indexed: 01/18/2023] Open
Abstract
Pediatric obesity has become in the last forty years the most common metabolic disease in children and adolescents affecting about 25% of the pediatric population in the western world. As obesity worsens, a whole-body insulin resistance (IR) occurs. This phenomenon is more pronounced during adolescence, when youth experience a high degree of insulin resistance due the production of growth hormone. As IR progresses, the blunted control of insulin on adipose tissue lipolysis causes an increased flux of fatty acids with FFA deposition in ectopic tissues and organs such as the liver, leading to the development of NAFLD. In this brief review, we will discuss the clinical implications of IR and NAFLD in the context of pediatric obesity. We will review the pathogenesis and the link between these two entities, the major pathophysiologic underpinnings, including the role of genetics and metagenomics, how these two entities lead to the development of type 2 diabetes, and which are the therapeutic options for NAFLD in youth.
Collapse
Affiliation(s)
| | - Nicola Santoro
- Department of Pediatrics, Kansas University Medical Center, Kansas City, KS, United States
- Department of Medicine and Health Sciences, “V. Tiberio” University of Molise, Campobasso, Italy
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, United States
| | - Giuseppina Rosaria Umano
- Department of the Woman, the Child, and General and Specialized Surgery, University of Campania, Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
32
|
Ekundayo TC, Ijabadeniyi OA, Igbinosa EO, Okoh AI. Using machine learning models to predict the effects of seasonal fluxes on Plesiomonas shigelloides population density. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 317:120734. [PMID: 36455774 DOI: 10.1016/j.envpol.2022.120734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 06/17/2023]
Abstract
Seasonal variations (SVs) affect the population density (PD), fate, and fitness of pathogens in environmental water resources and the public health impacts. Therefore, this study is aimed at applying machine learning intelligence (MLI) to predict the impacts of SVs on P. shigelloides population density (PDP) in the aquatic milieu. Physicochemical events (PEs) and PDP from three rivers acquired via standard microbiological and instrumental techniques across seasons were fitted to MLI algorithms (linear regression (LR), multiple linear regression (MR), random forest (RF), gradient boosted machine (GBM), neural network (NN), K-nearest neighbour (KNN), boosted regression tree (BRT), extreme gradient boosting (XGB) regression, support vector regression (SVR), decision tree regression (DTR), M5 pruned regression (M5P), artificial neural network (ANN) regression (with one 10-node hidden layer (ANN10), two 6- and 4-node hidden layers (ANN64), and two 5- and 5-node hidden layers (ANN55)), and elastic net regression (ENR)) to assess the implications of the SVs of PEs on aquatic PDP. The results showed that SVs significantly influenced PDP and PEs in the water (p < 0.0001), exhibiting a site-specific pattern. While MLI algorithms predicted PDP with differing absolute flux magnitudes for the contributing variables, DTR predicted the highest PDP value of 1.707 log unit, followed by XGB (1.637 log unit), but XGB (mean-squared-error (MSE) = 0.0025; root-mean-squared-error (RMSE) = 0.0501; R2 =0.998; medium absolute deviation (MAD) = 0.0275) outperformed other models in terms of regression metrics. Temperature and total suspended solids (TSS) ranked first and second as significant factors in predicting PDP in 53.3% (8/15) and 40% (6/15), respectively, of the models, based on the RMSE loss after permutations. Additionally, season ranked third among the 7 models, and turbidity (TBS) ranked fourth at 26.7% (4/15), as the primary significant factor for predicting PDP in the aquatic milieu. The results of this investigation demonstrated that MLI predictive modelling techniques can promisingly be exploited to complement the repetitive laboratory-based monitoring of PDP and other pathogens, especially in low-resource settings, in response to seasonal fluxes and can provide insights into the potential public health risks of emerging pathogens and TSS pollution (e.g., nanoparticles and micro- and nanoplastics) in the aquatic milieu. The model outputs provide low-cost and effective early warning information to assist watershed managers and fish farmers in making appropriate decisions about water resource protection, aquaculture management, and sustainable public health protection.
Collapse
Affiliation(s)
- Temitope C Ekundayo
- SAMRC Microbial Water Quality Monitoring Centre, University of Fort Hare, Alice, Eastern Cape, South Africa; Department of Biotechnology and Food Science, Durban University of Technology, Steve Biko Campus, Steve Biko Rd, Musgrave, Berea, 4001, Durban, South Africa; Department of Microbiology, University of Medical Sciences, Ondo City, Ondo State, Nigeria.
| | - Oluwatosin A Ijabadeniyi
- Department of Biotechnology and Food Science, Durban University of Technology, Steve Biko Campus, Steve Biko Rd, Musgrave, Berea, 4001, Durban, South Africa
| | - Etinosa O Igbinosa
- SAMRC Microbial Water Quality Monitoring Centre, University of Fort Hare, Alice, Eastern Cape, South Africa; Department of Microbiology, Faculty of Life Sciences University of Benin, Private Mail Bag 1154, Benin City, 300283, Nigeria
| | - Anthony I Okoh
- SAMRC Microbial Water Quality Monitoring Centre, University of Fort Hare, Alice, Eastern Cape, South Africa; Department of Environmental Health Sciences, College of Health Sciences, University of Sharjah, Sharjah, P.O. Box 27272, United Arab Emirates
| |
Collapse
|
33
|
Alcohol consumption and metabolic syndrome: Clinical and epidemiological impact on liver disease. J Hepatol 2023; 78:191-206. [PMID: 36063967 DOI: 10.1016/j.jhep.2022.08.030] [Citation(s) in RCA: 126] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/04/2022] [Accepted: 08/19/2022] [Indexed: 02/01/2023]
Abstract
Alcohol use and metabolic syndrome are highly prevalent in the population and frequently co-exist. Both are implicated in a large range of health problems, including chronic liver disease, hepatocellular carcinoma, and liver-related outcomes (i.e. decompensation or liver transplantation). Studies have yielded mixed results regarding the effects of mild-moderate alcohol consumption on the risk of metabolic syndrome and fatty liver disease, possibly due to methodological differences. The few available prospective studies have indicated that mild-moderate alcohol use is associated with an increase in liver-related outcomes. This conclusion was substantiated by systems biology analyses suggesting that alcohol and metabolic syndrome may play a similar role in fatty liver disease, potentiating an already existing dysregulation of common vital homeostatic pathways. Alcohol and metabolic factors are independently and jointly associated with liver-related outcomes. Indeed, metabolic syndrome increases the risk of liver-related outcomes, regardless of alcohol intake. Moreover, the components of metabolic syndrome appear to have additive effects when it comes to the risk of liver-related outcomes. A number of population studies have implied that measures of central/abdominal obesity, such as the waist-to-hip ratio, can predict liver-related outcomes more accurately than BMI, including in individuals who consume harmful quantities of alcohol. Many studies even point to synergistic interactions between harmful alcohol use and many metabolic components. This accumulating evidence showing independent, combined, and modifying effects of alcohol and metabolic factors on the onset and progression of chronic liver disease highlights the multifactorial background of liver disease in the population. The available evidence suggests that more holistic approaches could be useful for risk prediction, diagnostics and treatment planning.
Collapse
|
34
|
Petri BJ, Piell KM, Wahlang B, Head KZ, Andreeva K, Rouchka EC, Cave MC, Klinge CM. Polychlorinated biphenyls alter hepatic m6A mRNA methylation in a mouse model of environmental liver disease. ENVIRONMENTAL RESEARCH 2023; 216:114686. [PMID: 36341798 PMCID: PMC10120843 DOI: 10.1016/j.envres.2022.114686] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/30/2022] [Accepted: 10/25/2022] [Indexed: 05/21/2023]
Abstract
Exposure to polychlorinated biphenyls (PCBs) has been associated with liver injury in human cohorts and with nonalcoholic steatohepatitis (NASH) in mice fed a high fat diet (HFD). N (6)-methyladenosine (m6A) modification of mRNA regulates transcript fate, but the contribution of m6A modification on the regulation of transcripts in PCB-induced steatosis and fibrosis is unknown. This study tested the hypothesis that PCB and HFD exposure alters the levels of m6A modification in transcripts that play a role in NASH in vivo. Male C57Bl6/J mice were fed a HFD (12 wks) and administered a single oral dose of Aroclor1260, PCB126, or Aroclor1260 + PCB126. Genome-wide identification of m6A peaks was accomplished by m6A mRNA immunoprecipitation sequencing (m6A-RIP) and the mRNA transcriptome identified by RNA-seq. Exposure of HFD-fed mice to Aroclor1260 decreased the number of m6A peaks and m6A-containing genes relative to PCB vehicle control whereas PCB126 or the combination of Aroclor1260 + PCB126 increased m6A modification frequency. ∼41% of genes had one m6A peak and ∼49% had 2-4 m6A peaks. 117 m6A peaks were common in the four experimental groups. The Aroclor1260 + PCB126 exposure group showed the highest number (52) of m6A-peaks. qRT-PCR confirmed enrichment of m6A-containing fragments of the Apob transcript with PCB exposure. A1cf transcript abundance, m6A peak count, and protein abundance was increased with Aroclor1260 + PCB126 co-exposure. Irrespective of the PCB type, all PCB groups exhibited enriched pathways related to lipid/lipoprotein metabolism and inflammation through the m6A modification. Integrated analysis of m6A-RIP-seq and mRNA-seq identified 242 differentially expressed genes (DEGs) with increased or reduced number of m6A peaks. These data show that PCB exposure in HFD-fed mice alters the m6A landscape offering an additional layer of regulation of gene expression affecting a subset of gene responses in NASH.
Collapse
Affiliation(s)
- Belinda J Petri
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Kellianne M Piell
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Banrida Wahlang
- University of Louisville Center for Integrative Environmental Health Sciences (CIEHS), USA; University of Louisville Hepatobiology and Toxicology Center, USA; The University of Louisville Superfund Research Center, USA; Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine, University of Louisville School of Medicine, USA
| | - Kimberly Z Head
- University of Louisville Hepatobiology and Toxicology Center, USA
| | - Kalina Andreeva
- KY INBRE Bioinformatics Core, University of Louisville, USA; Department of Genetics, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Eric C Rouchka
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA; KY INBRE Bioinformatics Core, University of Louisville, USA
| | - Matthew C Cave
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA; Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine, University of Louisville School of Medicine, USA
| | - Carolyn M Klinge
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA; University of Louisville Center for Integrative Environmental Health Sciences (CIEHS), USA.
| |
Collapse
|
35
|
Dalal S, Onyema EM, Malik A. Hybrid XGBoost model with hyperparameter tuning for prediction of liver disease with better accuracy. World J Gastroenterol 2022; 28:6551-6563. [PMID: 36569269 PMCID: PMC9782838 DOI: 10.3748/wjg.v28.i46.6551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/27/2022] [Accepted: 11/21/2022] [Indexed: 12/08/2022] Open
Abstract
BACKGROUND Liver disease indicates any pathology that can harm or destroy the liver or prevent it from normal functioning. The global community has recently witnessed an increase in the mortality rate due to liver disease. This could be attributed to many factors, among which are human habits, awareness issues, poor healthcare, and late detection. To curb the growing threats from liver disease, early detection is critical to help reduce the risks and improve treatment outcome. Emerging technologies such as machine learning, as shown in this study, could be deployed to assist in enhancing its prediction and treatment.
AIM To present a more efficient system for timely prediction of liver disease using a hybrid eXtreme Gradient Boosting model with hyperparameter tuning with a view to assist in early detection, diagnosis, and reduction of risks and mortality associated with the disease.
METHODS The dataset used in this study consisted of 416 people with liver problems and 167 with no such history. The data were collected from the state of Andhra Pradesh, India, through https://www.kaggle.com/datasets/uciml/indian-liver-patient-records. The population was divided into two sets depending on the disease state of the patient. This binary information was recorded in the attribute "is_patient".
RESULTS The results indicated that the chi-square automated interaction detection and classification and regression trees models achieved an accuracy level of 71.36% and 73.24%, respectively, which was much better than the conventional method. The proposed solution would assist patients and physicians in tackling the problem of liver disease and ensuring that cases are detected early to prevent it from developing into cirrhosis (scarring) and to enhance the survival of patients. The study showed the potential of machine learning in health care, especially as it concerns disease prediction and monitoring.
CONCLUSION This study contributed to the knowledge of machine learning application to health and to the efforts toward combating the problem of liver disease. However, relevant authorities have to invest more into machine learning research and other health technologies to maximize their potential.
Collapse
Affiliation(s)
- Surjeet Dalal
- Department of CSE, Amity University, Gurugram 122413, Haryana, India
| | - Edeh Michael Onyema
- Department of Mathematics and Computer Science, Coal City University, Enugu 400102, Nigeria
| | - Amit Malik
- Department of CSE, SRM University, Delhi-NCR, Sonipat 131001, Haryana, India
| |
Collapse
|
36
|
Machine Learning Approach for Metabolic Syndrome Diagnosis Using Explainable Data-Augmentation-Based Classification. Diagnostics (Basel) 2022; 12:diagnostics12123117. [PMID: 36553124 PMCID: PMC9777696 DOI: 10.3390/diagnostics12123117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022] Open
Abstract
Metabolic syndrome (MetS) is a cluster of risk factors including hypertension, hyperglycemia, dyslipidemia, and abdominal obesity. Metabolism-related risk factors include diabetes and heart disease. MetS is also linked to numerous cancers and chronic kidney disease. All of these variables raise medical costs. Developing a prediction model that can quickly identify persons at high risk of MetS and offer them a treatment plan is crucial. Early prediction of metabolic syndrome will highly impact the quality of life of patients as it gives them a chance for making a change to the bad habit and preventing a serious illness in the future. In this paper, we aimed to assess the performance of various algorithms of machine learning in order to decrease the cost of predictive diagnoses of metabolic syndrome. We employed ten machine learning algorithms along with different metaheuristics for feature selection. Moreover, we examined the effects of data augmentation in the prediction accuracy. The statistics show that the augmentation of data after applying feature selection on the data highly improves the performance of the classifiers.
Collapse
|
37
|
Huang Y, Lin X, Yu S, Chen R, Chen W. Intestinal Engineered Probiotics as Living Therapeutics: Chassis Selection, Colonization Enhancement, Gene Circuit Design, and Biocontainment. ACS Synth Biol 2022; 11:3134-3153. [PMID: 36094344 DOI: 10.1021/acssynbio.2c00314] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Intestinal probiotics are often used for the in situ treatment of diseases, such as metabolic disorders, tumors, and chronic inflammatory infections. Recently, there has been an increased emphasis on intelligent, customized treatments with a focus on long-term efficacy; however, traditional probiotic therapy has not kept up with this trend. The use of synthetic biology to construct gut-engineered probiotics as live therapeutics is a promising avenue in the treatment of specific diseases, such as phenylketonuria and inflammatory bowel disease. These studies generally involve a series of fundamental design issues: choosing an engineered chassis, improving the colonization ability of engineered probiotics, designing functional gene circuits, and ensuring the safety of engineered probiotics. In this review, we summarize the relevant past research, the progress of current research, and discuss the key issues that restrict the widespread application of intestinal engineered probiotic living therapeutics.
Collapse
Affiliation(s)
- Yan Huang
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Xiaojun Lin
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Siyang Yu
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Ruiyue Chen
- Team SZU-China at iGEM 2021, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Weizhao Chen
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.,Shenzhen Key Laboratory for Microbial Gene Engineering, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
38
|
Aasmets O, Krigul KL, Org E. Evaluating the clinical relevance of the enterotypes in the Estonian microbiome cohort. Front Genet 2022; 13:917926. [PMID: 36061192 PMCID: PMC9428584 DOI: 10.3389/fgene.2022.917926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Human gut microbiome is subject to high inter-individual and temporal variability, which complicates building microbiome-based applications, including applications that can be used to improve public health. Categorizing the microbiome profiles into a small number of distinct clusters, such as enterotyping, has been proposed as a solution that can ameliorate these shortcomings. However, the clinical relevance of the enterotypes is poorly characterized despite a few studies marking the potential for using the enterotypes for disease diagnostics and personalized nutrition. To gain a further understanding of the clinical relevance of the enterotypes, we used the Estonian microbiome cohort dataset (n = 2,506) supplemented with diagnoses and drug usage information from electronic health records to assess the possibility of using enterotypes for disease diagnostics, detecting disease subtypes, and evaluating the susceptibility for developing a condition. In addition to the previously established 3-cluster enterotype model, we propose a 5-cluster community type model based on our data, which further separates the samples with extremely high Bacteroides and Prevotella abundances. Collectively, our systematic analysis including 231 phenotypic factors, 62 prevalent diseases, and 33 incident diseases greatly expands the knowledge about the enterotype-specific characteristics; however, the evidence suggesting the practical use of enterotypes in clinical practice remains scarce.
Collapse
|
39
|
Gao Y, Ma L, Su J. Host and microbial-derived metabolites for Clostridioides difficile infection: Contributions, mechanisms and potential applications. Microbiol Res 2022; 263:127113. [PMID: 35841835 DOI: 10.1016/j.micres.2022.127113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/01/2022] [Accepted: 07/03/2022] [Indexed: 12/23/2022]
Abstract
Clostridioides difficile infection (CDI), which mostly occurs in hospitalized patients, is the most common and costly health care-associated disease. However, the biology of C. difficile remains incompletely understood. Current therapeutics are still challenged by the frequent recurrence of CDI. Advances in metabolomics facilitate our understanding of the etiology of CDI, which is not merely an alteration in the structure of the gut microbial community but also a dysbiosis metabolic setting promoting the germination, expansion and virulence of C. difficile. Therefore, we summarized the gut microbial and metabolic profiles for CDI under different conditions, such as those of postantibiotic treatment and postfecal microbiota transplantation. The current understanding of the role of host and gut microbial-derived metabolites as well as other nutrients in preventing or alleviating the disease symptoms of CDI will also be provided in this review. We hope that a specific nutrient-centric dietary strategy or the administration of certain nutrients to the colon could serve as an alternate line of investigation for the prophylaxis and mitigation of CDI in the future. Nevertheless, rigorously designed basic studies and randomized controlled trials need to be conducted to assess the functional mechanisms and effects of such therapeutics.
Collapse
Affiliation(s)
- Yan Gao
- Department of Clinical Laboratory Diagnostics, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Liyan Ma
- Department of Clinical Laboratory Diagnostics, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jianrong Su
- Department of Clinical Laboratory Diagnostics, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
40
|
Ma Y, Liu X, Wang J. Small molecules in the big picture of gut microbiome-host cross-talk. EBioMedicine 2022; 81:104085. [PMID: 35636316 PMCID: PMC9156878 DOI: 10.1016/j.ebiom.2022.104085] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/06/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022] Open
Abstract
Research on the gut microbiome and related diseases is rapidly growing with the development of sequencing technologies. An increasing number of studies offer new perspectives on disease development or treatment. Among these, the mechanisms of gut microbial metabolite-mediated effects merit better understanding. In this review, we first summarize the shifts in gut microbial metabolites within complex diseases, in which metabolites have correlational and occasionally causal effects on diseases and discuss the reported mechanisms. We further investigate the interactions between gut microbes and drugs, providing insights for precision medication as well as limitations of current research. Finally, we provide new research directions and research strategies for the development of drugs from gut microbial metabolites. FUNDING STATEMENT: None.
Collapse
Affiliation(s)
- Yue Ma
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaolin Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|