Applications of Artificial Intelligence in Microbiome Analysis and Probiotic Interventions—An Overview and Perspective Based on the Current State of the Art
Abstract
:1. Introduction
2. AI-Driven Analysis of Microbiota Functionality
2.1. The Impact of AI on Microbial Analysis
2.2. Machine Learning (ML) and Deep Learning Approaches for Microbiome Data Analysis
3. Dysbiosis and Clinical Implications
3.1. Clinical Evidence for Probiotic Mixtures in CIDs
- (a)
- Data quality and standardization: Ensuring consistent and high-quality data across different studies and platforms is crucial for developing robust AI models [27].
- (b)
- Interpretability: Many AI models, particularly deep learning approaches, operate as “black boxes,” making it challenging to interpret their decision-making processes [35].
- (c)
- Integration of multi-omics data: Developing AI models that can effectively integrate diverse data types (e.g., metagenomics, metabolomics, and host genomics) remains a significant challenge [36].
- (d)
- Ethical considerations: The use of AI in healthcare raises important ethical questions regarding data privacy, algorithmic bias, and clinical decision-making [37]. Future research directions should focus on addressing these challenges and exploring new AI paradigms. The development of explainable AI models, federated learning approaches for privacy-preserving analysis, and the integration of AI with other emerging technologies (e.g., single-cell sequencing and microfluidics) hold great promise for advancing our understanding of the microbiome and combating antibiotic resistance.
3.2. Microbiome Profiling and Targeted Interventions
4. Current and Potential Solutions to Improve Microbiome Analysis and Probiotic Interventions
5. Conclusions and Future Perspective
Author Contributions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. The human microbiome project. Nature 2007, 449, 804–810. [Google Scholar] [CrossRef] [PubMed]
- Ursell, L.K.; Metcalf, J.L.; Parfrey, L.W.; Knight, R. Defining the human microbiome. Nutr. Rev. 2012, 70 (Suppl. 1), S38–S44. [Google Scholar] [CrossRef] [PubMed]
- Dekaboruah, E.; Suryavanshi, M.V.; Chettri, D.; Verma, A.K. Human microbiome: An academic update on human body site specific surveillance and its possible role. Arch. Microbiol. 2020, 202, 2147–2167. [Google Scholar] [CrossRef] [PubMed]
- Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef]
- Lynch, S.V.; Pedersen, O. The Human Intestinal Microbiome in Health and Disease. N. Engl. J. Med. 2016, 375, 2369–2379. [Google Scholar] [CrossRef] [PubMed]
- Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef]
- Hoffman, D.E.; Fraser, C.M.; Palumbo, F.B.; Ravel, J.; Rothenberg, K.; Rowthorn, V.; Schwartz, J. Probiotics: Finding the right regulatory balance. Science 2013, 342, 314–315. [Google Scholar] [CrossRef]
- Suez, J.; Zmora, N.; Segal, E.; Elinav, E. The pros, cons, and many unknowns of probiotics. Nat. Med. 2019, 25, 716–729. [Google Scholar] [CrossRef]
- O’Toole, P.W.; Marchesi, J.R.; Hill, C. Next-generation probiotics: The spectrum from probiotics to live biotherapeutics. Nat. Microbiol. 2017, 2, 17057. [Google Scholar] [CrossRef]
- Abavisani, M.; Khoshrou, A.; Foroushan, S.K.; Ebadpour, N.; Sahebkar, A. Deciphering the gut microbiome: The revolution of artificial intelligence in microbiota analysis and intervention. Curr. Res. Biotechnol. 2024, 7, 100211. [Google Scholar] [CrossRef]
- Zhang, C.; Wang, J.; Li, J.; Liu, X.; Wang, X. Artificial Intelligence in Microbiome Research: Opportunities and Challenges. Front. Microbiol. 2020, 11, 527458. [Google Scholar]
- Oh, M.; Zhang, L. DeepMicro: Deep representation learning for disease prediction based on microbiome data. Sci. Rep. 2020, 10, 6026. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Zhang, L. Artificial Intelligence for Remote Sensing Data Analysis: A review of challenges and opportunities. IEEE Geosci. Remote Sens. 2022, 10, 270–294. [Google Scholar] [CrossRef]
- Kumar, S.; Sharma, D.; Rao, S.; Lim, W.M.; Mangla, S.K. Past, present, and future of sustainable finance: Insights from big data analytics through machine learning of scholarly research. Ann. Oper. Res. 2022, 1–44. [Google Scholar] [CrossRef]
- Heshiki, Y.; Vazquez-Uribe, R.; Li, J.; Ni, Y.; Quainoo, S.; Imamovic, L.; Li, J.; Sørensen, M.; Chow, B.K.C.; Weiss, G.J.; et al. Predictable modulation of cancer treatment outcomes by the gut microbiota. Microbiome 2020, 8, 28. [Google Scholar] [CrossRef] [PubMed]
- Knights, D.; Costello, E.K.; Knight, R. Supervised classification of human microbiota. FEMS Microbiol. Rev. 2011, 35, 343–359. [Google Scholar] [CrossRef]
- Pasolli, E.; Truong, D.T.; Malik, F.; Waldron, L.; Segata, N. Machine Learning Meta-analysis of Large Metagenomic Datasets: Tools and Biological Insights. PLoS Comput. Biol. 2016, 12, e1004977. [Google Scholar] [CrossRef]
- Knights, D.; Ward, T.L.; McKinlay, C.E.; Miller, H.; Gonzalez, A.; McDonald, D.; Knight, R. Rethinking “enterotypes”. Cell Host Microbe 2014, 16, 433–437. [Google Scholar] [CrossRef]
- Mallick, H.; Franzosa, E.A.; Mclver, L.J.; Banerjee, S.; Sirota-Madi, A.; Kostic, A.D.; Clish, C.B.; Vlamakis, H.; Xavier, R.J.; Huttenhower, C. Predictive metabolomic profiling of microbial communities using amplicon or metagenomic sequences. Nat. Commun. 2019, 10, 3136. [Google Scholar] [CrossRef]
- Fioravanti, A.; Fustaino, V.; Orsini, M.; Corsi, C.; Arrigo, P.; Patarnello, T.; Carbonneau, M.A.; Mauri, M.; Biondi, E.G. Phylogenetic convolutional neural networks in metagenomics. BMC Bioinform. 2018, 19, 49. [Google Scholar] [CrossRef]
- Marcos-Zambrano, L.J.; Karaduzovic-Hadziabdic, K.; Loncar Turukalo, T.; Przymus, P.; Trajkovik, V.; Aasmets, O.; Berland, M.; Gruca, A.; Hasic, J.; Hron, K.; et al. Applications of Machine Learning in Human Microbiome Studies: A Review on Feature Selection, Biomarker Identification, Disease Prediction and Treatment. Front. Microbiol. 2021, 12, 634511. [Google Scholar] [CrossRef]
- Zhu, Q.; Pan, M.; Liu, J.; Wang, H.; Wei, Y.; Liu, Y.; Xu, Z.; Yang, S.; Wei, H.; Huang, Y.; et al. MicrobioGraph: A Graph Neural Network-Based Framework for Inferring Microbial Interactions from Metagenomic Data. bioRxiv 2021. bioRxiv:2021.02.11.430831. [Google Scholar]
- Arango-Argoty, G.; Garner, E.; Pruden, A.; Heath, L.S.; Vikesland, P.; Zhang, L. DeepARG: A deep learning approach for predicting antibiotic resistance genes from metagenomic data. Microbiome 2018, 6, 23. [Google Scholar] [CrossRef] [PubMed]
- Stokes, J.M.; Yang, K.; Swanson, K.; Jin, W.; Cubillos-Ruiz, A.; Donghia, N.M.; MacNair, C.R.; French, S.; Carfrae, L.A.; Bloom-Ackermann, Z.; et al. A Deep Learning Approach to Antibiotic Discovery. Cell 2020, 180, 688–702.e13. [Google Scholar] [CrossRef]
- Yang, K.K.; Wu, Z.; Bedbrook, C.N.; Arnold, F.H. Learned protein embeddings for machine learning. Bioinformatics 2018, 34, 2642–2648. [Google Scholar] [CrossRef] [PubMed]
- Yelin, I.; Snitser, O.; Novich, G.; Katz, R.; Tal, O.; Parizade, M.; Chodick, G.; Koren, G.; Shalev, V.; Kishony, R. Personal clinical history predicts antibiotic resistance of urinary tract infections. Nat. Med. 2019, 25, 1143–1152. [Google Scholar] [CrossRef] [PubMed]
- Sinha, R.; Abnet, C.C.; White, O.; Knight, R.; Huttenhower, C. The microbiome quality control project: Baseline study design and future directions. Genome Biol. 2015, 16, 276. [Google Scholar] [CrossRef]
- Tursi, A.; Brandimarte, G.; Papa, A.; Giglio, A.; Elisei, W.; Giorgetti, G.M.; Forti, G.; Morini, S.; Hassan, C.; Pistoia, M.; et al. Treatment of relapsing mild-to-moderate ulcerative colitis with the probiotic VSL#3 as adjunctive to a standard pharmaceutical treatment: A double-blind, randomized, placebo-controlled study. Am. J. Gastroenterol. 2010, 105, 2218–2227. [Google Scholar]
- Sabico, S.; Al-Mashharawi, A.; Al-Daghri, N.M.; Yakout, S.; Alnaami, A.M.; Alokail, M.S.; McTernan, P.G. Effects of a multi-strain probiotic supplement for 12 weeks in circulating endotoxin levels and cardiometabolic profiles of medication naïve T2DM patients: A randomized clinical trial. J. Transl. Med. 2019, 17, 286. [Google Scholar] [CrossRef]
- Zamani, B.; Golkar, H.R.; Farshbaf, S.; Emadi-Baygi, M.; Tajabadi-Ebrahimi, M.; Jafari, P.; Akhavan, R.; Taghizadeh, M.; Memarzadeh, M.R.; Asemi, Z. Clinical and metabolic response to probiotic administration in patients with rheumatoid arthritis: A randomized, double-blind, placebo-controlled trial. Int. J. Rheum. Dis. 2016, 19, 869–879. [Google Scholar] [CrossRef]
- Tamtaji, O.R.; Taghizadeh, M.; Daneshvar Kakhaki, R.; Kouchaki, E.; Bahmani, F.; Borzabadi, S.; Oryan, S.; Mafi, A.; Asemi, Z. Clinical and metabolic response to probiotic administration in people with Parkinson’s disease: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2019, 38, 1031–1035. [Google Scholar] [CrossRef]
- Zeevi, D.; Korem, T.; Zmora, N.; Israeli, D.; Rothschild, D.; Weinberger, A.; Ben-Yacov, O.; Lador, D.; Avnit-Sagi, T.; Lotan-Pompan, M. Personalized Nutrition by Prediction of Glycemic Responses. Cell 2015, 163, 1079–1094. [Google Scholar] [CrossRef] [PubMed]
- Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef] [PubMed]
- Rothschild, D.; Weissbrod, O.; Barkan, E.; Kurilshikov, A.; Korem, T.; Zeevi, D.; Costea, P.I.; Godneva, A.; Kalka, I.N.; Bar, N. Environment dominates over host genetics in shaping human gut microbiota. Nature 2018, 555, 210–215. [Google Scholar] [CrossRef]
- Lipton, Z.C. The Mythos of Model Interpretability. Queue 2018, 16, 31–57. [Google Scholar] [CrossRef]
- Heintz-Buschart, A.; Wilmes, P. Human Gut Microbiome: Function Matters. Trends Microbiol. 2018, 26, 563–574. [Google Scholar] [CrossRef]
- Char, D.S.; Shah, N.H.; Magnus, D. Implementing Machine Learning in Health Care—Addressing Ethical Challenges. N. Engl. J. Med. 2018, 378, 981–983. [Google Scholar] [CrossRef]
- Franzosa, E.A.; McIver, L.J.; Rahnavard, G.; Thompson, L.R.; Schirmer, M.; Weingart, G.; Lipson, K.S.; Knight, R.; Caporaso, J.G.; Segata, N.; et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat. Methods 2018, 15, 962–968. [Google Scholar] [CrossRef]
Microbiome Analysis | Explanation | AI Application | Example |
---|---|---|---|
1. Metabolomics | Metabolomics involves the comprehensive analysis of metabolites in a biological system. These metabolites can provide insights into metabolic changes and microbial interactions. | AI algorithms, particularly machine learning models, can analyze complex metabolomic data to identify patterns and correlations between metabolites and microbial communities. For instance, AI can use clustering techniques to categorize different metabolic profiles associated with specific microbiome compositions or health conditions. | A study might use AI to analyze urine and blood samples from patients with metabolic disorders to identify specific metabolite patterns linked to gut microbiome composition. This could help in identifying biomarkers for early diagnosis or personalized treatment plans. |
2. Transcriptomics | Transcriptomics focuses on the RNA transcripts produced by the genome under specific circumstances. It helps in understanding gene expression patterns within the microbiome. | AI can be used to process and interpret large-scale transcriptomic data to discern gene expression changes in microbial communities. For example, deep learning models can analyze RNA sequencing data to identify differentially expressed genes that are influenced by or influence the microbiome. | AI-driven tools can analyze transcriptomic data from gut microbiome samples to identify how microbial gene expression changes in response to dietary interventions, helping to understand which microbial genes are linked to improved health outcomes. |
3. Proteomics | Proteomics involves studying the entire set of proteins produced by an organism. It provides insights into the functional aspects of the microbiome. | AI can facilitate the analysis of proteomic data by identifying protein expression patterns and interactions within the microbiome. Machine learning algorithms can predict the functional impact of specific proteins or protein interactions. | AI models can be used to analyze protein expression profiles in the gut microbiome to identify proteins that correlate with CIDs, potentially leading to the discovery of new therapeutic targets. |
4. Genomics | Genomics involves studying the complete DNA sequence of an organism, including the microbiome. It helps in understanding genetic variations and their impact on microbial function. Genomics focuses on the genome of an individual organism, while metagenomics studies the combined genetic material from an entire community of organisms. Similarly, microbiota refers to the organisms themselves, while microbiome emphasizes the genes and functional capacities of these organisms. | AI can assist in analyzing genomic data to map microbial genomes, identify genetic variations, and predict their functional consequences. For instance, AI models can analyze metagenomic data to link specific microbial genes with health outcomes. | AI-driven genomic analysis can identify specific microbial genes associated with antibiotic resistance. This information can be used to predict resistance patterns and inform treatment strategies. |
Explanation | Application | Example | |
---|---|---|---|
1. Predictive Modeling | AI can predict how different probiotic strains will interact with the microbiome and influence health outcomes, based on historical data and clinical trials. | By analyzing data from previous studies and clinical trials, AI can identify which probiotic strains are most effective for specific conditions or patient demographics. | An AI model might predict that a certain combination of Lactobacillus and Bifidobacterium strains is particularly effective for managing irritable bowel syndrome (IBS) in adults, based on a comprehensive analysis of patient data and clinical outcomes. |
2. Personalized Probiotic Recommendations | AI can be used to tailor probiotic recommendations based on individual microbiome profiles and health conditions. | By integrating data from genomic, metabolomic, and clinical sources, AI can recommend personalized probiotic mixtures that are more likely to be effective for individual patients | For a patient with a disrupted gut microbiome and specific metabolic abnormalities, an AI system might suggest a custom probiotic blend that targets the imbalances observed in their microbiome. |
3. Clinical Decision Support | AI can support clinicians in selecting appropriate probiotic treatments by analyzing patient data and predicting potential outcomes. | AI systems can provide evidence-based recommendations for probiotic use in various clinical settings, such as managing chronic diseases or supporting recovery after antibiotic treatment. | In a hospital setting, AI could analyze data from patients undergoing antibiotic therapy to recommend specific probiotics that could help prevent antibiotic-associated diarrhea. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
D’Urso, F.; Broccolo, F. Applications of Artificial Intelligence in Microbiome Analysis and Probiotic Interventions—An Overview and Perspective Based on the Current State of the Art. Appl. Sci. 2024, 14, 8627. https://doi.org/10.3390/app14198627
D’Urso F, Broccolo F. Applications of Artificial Intelligence in Microbiome Analysis and Probiotic Interventions—An Overview and Perspective Based on the Current State of the Art. Applied Sciences. 2024; 14(19):8627. https://doi.org/10.3390/app14198627
Chicago/Turabian StyleD’Urso, Fabiana, and Francesco Broccolo. 2024. "Applications of Artificial Intelligence in Microbiome Analysis and Probiotic Interventions—An Overview and Perspective Based on the Current State of the Art" Applied Sciences 14, no. 19: 8627. https://doi.org/10.3390/app14198627
APA StyleD’Urso, F., & Broccolo, F. (2024). Applications of Artificial Intelligence in Microbiome Analysis and Probiotic Interventions—An Overview and Perspective Based on the Current State of the Art. Applied Sciences, 14(19), 8627. https://doi.org/10.3390/app14198627