1
|
Zhang Z, Niu H, Qu Q, Guo D, Wan X, Yang Q, Mo Z, Tan S, Xiang Q, Tian X, Yang H, Liu Z. Advancements in Lactiplantibacillus plantarum: probiotic characteristics, gene editing technologies and applications. Crit Rev Food Sci Nutr 2025:1-22. [PMID: 39745813 DOI: 10.1080/10408398.2024.2448562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The exploration of microorganisms in fermented products has become a pivotal area of scientific research, primarily due to their widespread availability and profound potential to improve human health. Among these, Lactiplantibacillus plantarum (formerly known as Lactobacillus plantarum) stands out as a versatile lactic acid bacterium, prevalent across diverse ecological niches. Its appeal extends beyond its well-documented probiotic benefits to include the remarkable plasticity of its genome, which has captivated both scientific and industrial stakeholders. Despite this interest, substantial challenges persist in fully understanding and harnessing the potential of L. plantarum. This review aims to illuminate the probiotic attributes of L. plantarum, consolidate current advancements in gene editing technologies, and explore the multifaceted applications of both wild-type and genetically engineered strains.
Collapse
Affiliation(s)
- Zhiqi Zhang
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Haorui Niu
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Qiu Qu
- Division of geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Dingming Guo
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xuchun Wan
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Qianqian Yang
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zihao Mo
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Siyu Tan
- Department of Biotechnology, Wuhan No. 2 High School, Wuhan, China
| | - Qian Xiang
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xue Tian
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Hongju Yang
- Division of geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Zhi Liu
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Lv Y, Xian Y, Lei X, Xie S, Zhang B. The role of the microbiota-gut-brain axis and artificial intelligence in cognitive health of pediatric obstructive sleep apnea: A narrative review. Medicine (Baltimore) 2024; 103:e40900. [PMID: 39686454 DOI: 10.1097/md.0000000000040900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
Pediatric obstructive sleep apnea (OSA) is a prevalent sleep-related breathing disorder associated with significant neurocognitive and behavioral impairments. Recent studies have highlighted the role of gut microbiota and the microbiota-gut-brain axis (MGBA) in influencing cognitive health in children with OSA. This narrative review aims to summarize current knowledge on the relationship between gut microbiota, MGBA, and cognitive function in pediatric OSA. It also explores the potential of artificial intelligence and machine learning in advancing this field and identifying novel therapeutic strategies. Pediatric OSA is associated with gut dysbiosis, reduced microbial diversity, and metabolic disruptions. MGBA mechanisms, such as endocrine, immune, and neural pathways, link gut microbiota to cognitive outcomes. Artificial intelligence and machine learning methodologies offer promising tools to uncover microbial markers and mechanisms associated with cognitive deficits in OSA. Future research should focus on validating these findings through clinical trials and developing personalized therapeutic approaches targeting the gut microbiota.
Collapse
Affiliation(s)
- Yunjiao Lv
- Department of First Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Yongtao Xian
- Department of First Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Xinye Lei
- Department of First Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Siqi Xie
- Department of First Clinical College, Guangzhou Medical University, Guangzhou, China
| | - Biyun Zhang
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Zhai Y, Kim M, Fan P, Rajeev S, Kim SA, Driver JD, Galvão KN, Boucher C, Jeong KC. Machine learning-enhanced assessment of potential probiotics from healthy calves for the treatment of neonatal calf diarrhea. Front Microbiol 2024; 15:1507537. [PMID: 39717273 PMCID: PMC11663915 DOI: 10.3389/fmicb.2024.1507537] [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: 10/07/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Abstract
Neonatal calf diarrhea (NCD) remains a significant contributor to calf mortality within the first 3 weeks of life, prompting widespread antibiotic use with associated concerns about antimicrobial resistance and disruption of the calf gut microbiota. Recent research exploring NCD treatments targeting gut microbiota dysbiosis has highlighted probiotic supplementation as a promising and safe strategy for gut homeostasis. However, varying treatment outcomes across studies suggest the need for efficient treatment options. In this study, we evaluated the potential of probiotics Limosilactobacillus reuteri, formally known as Lactobacillus reuteri, isolated from healthy neonatal calves to treat NCD. Through in silico whole genome analysis and in vitro assays, we identified nine L. reuteri strains, which were then administered to calves with NCD. Calves treated with L. reuteri strains shed healthy feces and demonstrated restored gut microbiota and normal animal behavior. Leveraging a machine learning model, we evaluated microbiota profiles and identified bacterial taxa associated with calf gut health that were elevated by L. reuteri administration. These findings represent a crucial advancement towards sustainable antibiotic alternatives for managing NCD, contributing significantly to global efforts in mitigating antimicrobial resistance and promoting overall animal health and welfare.
Collapse
Affiliation(s)
- Yuting Zhai
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Miju Kim
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Food Science and Biotechnology, Kyung Hee University, Seoul, Republic of Korea
| | - Peixin Fan
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Sharath Rajeev
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Sun Ae Kim
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul, Republic of Korea
| | - J. Danny Driver
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Klibs N. Galvão
- Department of Large Animal Clinical Sciences, University of Florida, Gainesville, FL, United States
| | - Christina Boucher
- Department of Computer and Information Science and Engineering, University of Florida, Gainesville, FL, United States
| | - Kwangcheol C. Jeong
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
4
|
Charizani E, Dushku E, Kyritsi M, Metallinou ET, Karathodorou A, Amanetidou E, Kokkaleniou MM, Passalis N, Tefas A, Staikou A, Yiangou M. Predicting the immunomodulatory activity of probiotic lactic acid bacteria using supervised machine learning in a Cornu aspersum snail model. FISH & SHELLFISH IMMUNOLOGY 2024; 152:109788. [PMID: 39053586 DOI: 10.1016/j.fsi.2024.109788] [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: 04/29/2024] [Revised: 07/09/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
In the process of screening for probiotic strains, there are no clearly established bacterial phenotypic markers which could be used for the prediction of their in vivo mechanism of action. In this work, we demonstrate for the first time that Machine Learning (ML) methods can be used for accurately predicting the in vivo immunomodulatory activity of probiotic strains based on their cell surface phenotypic features using a snail host-microbe interaction model. A broad range of snail gut presumptive probiotics, including 240 new lactic acid bacterial strains (Lactobacillus, Leuconostoc, Lactococcus, and Enterococcus), were isolated and characterized based on their capacity to withstand snails' gastrointestinal defense barriers, such as the pedal mucus, gastric mucus, gastric juices, and acidic pH, in association with their cell surface hydrophobicity, autoaggregation, and biofilm formation ability. The implemented ML pipeline predicted with high accuracy (88 %) strains with a strong capacity to enhance chemotaxis and phagocytic activity of snails' hemolymph cells, while also revealed bacterial autoaggregation and cell surface hydrophobicity as the most important parameters that significantly affect host immune responses. The results show that ML approaches may be useful to derive a predictive understanding of host-probiotic interactions, while also highlighted the use of snails as an efficient animal model for screening presumptive probiotic strains in the light of their interaction with cellular innate immune responses.
Collapse
Affiliation(s)
- Elissavet Charizani
- Department of Genetics, Development & Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Esmeralda Dushku
- Department of Genetics, Development & Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Maria Kyritsi
- Department of Genetics, Development & Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Eleftheria Theodora Metallinou
- Department of Genetics, Development & Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Argyro Karathodorou
- Department of Genetics, Development & Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Eleni Amanetidou
- Department of Genetics, Development & Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Marianthi-Maria Kokkaleniou
- Department of Genetics, Development & Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Nikolaos Passalis
- Computational Intelligence and Deep Learning Group, Artificial Intelligence and Information Analysis Laboratory, School of Informatics, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Anastasios Tefas
- Computational Intelligence and Deep Learning Group, Artificial Intelligence and Information Analysis Laboratory, School of Informatics, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Alexandra Staikou
- Department of Zoology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Minas Yiangou
- Department of Genetics, Development & Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.
| |
Collapse
|
5
|
Mosquera FEC, Guevara-Montoya MC, Serna-Ramirez V, Liscano Y. Neuroinflammation and Schizophrenia: New Therapeutic Strategies through Psychobiotics, Nanotechnology, and Artificial Intelligence (AI). J Pers Med 2024; 14:391. [PMID: 38673018 PMCID: PMC11051547 DOI: 10.3390/jpm14040391] [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: 03/13/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
The prevalence of schizophrenia, affecting approximately 1% of the global population, underscores the urgency for innovative therapeutic strategies. Recent insights into the role of neuroinflammation, the gut-brain axis, and the microbiota in schizophrenia pathogenesis have paved the way for the exploration of psychobiotics as a novel treatment avenue. These interventions, targeting the gut microbiome, offer a promising approach to ameliorating psychiatric symptoms. Furthermore, advancements in artificial intelligence and nanotechnology are set to revolutionize psychobiotic development and application, promising to enhance their production, precision, and effectiveness. This interdisciplinary approach heralds a new era in schizophrenia management, potentially transforming patient outcomes and offering a beacon of hope for those afflicted by this complex disorder.
Collapse
Affiliation(s)
| | | | | | - Yamil Liscano
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia; (F.E.C.M.); (M.C.G.-M.); (V.S.-R.)
| |
Collapse
|
6
|
Grandmont A, Rhouma M, Létourneau-Montminy MP, Thériault W, Mainville I, Arcand Y, Leduc R, Demers B, Thibodeau A. Characterization of the Effects of a Novel Probiotic on Salmonella Colonization of a Piglet-Derived Intestinal Microbiota Using Improved Bioreactor. Animals (Basel) 2024; 14:787. [PMID: 38473172 DOI: 10.3390/ani14050787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The carriage of Salmonella in pigs is a major concern for the agri-food industry and for global healthcare systems. Humans could develop salmonellosis when consuming contaminated pig products. On the other hand, some Salmonella serotypes could cause disease in swine, leading to economic losses on farms. The purpose of the present study was to characterize the anti-Salmonella activity of a novel Bacillus-based probiotic using a bioreactor containing a piglet-derived intestinal microbiota. Two methods of probiotic administration were tested: a single daily and a continuous dose. Salmonella enumeration was performed using selective agar at T24h, T48h, T72h, T96h and T120h. The DNA was extracted from bioreactor samples to perform microbiome profiling by targeted 16S rRNA gene sequencing on Illumina Miseq. The quantification of short-chain fatty acids (SCFAs) was also assessed at T120h. The probiotic decreased Salmonella counts at T96 for the daily dose and at T120 for the continuous one. Both probiotic doses affected the alpha and beta diversity of the piglet-derived microbiota (p < 0.05). A decrease in acetate concentration and an increase in propionate proportion were observed in the continuous condition. In conclusion, the tested Bacillus-based product showed a potential to modulate microbiota and reduce Salmonella colonization in a piglet-derived intestinal microbiota and could therefore be used in vivo.
Collapse
Affiliation(s)
- Amely Grandmont
- Chaire de Recherche en Salubrité des Viandes, Département de Microbiologie et Pathologie, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
- Groupe de Recherche et d'Enseignement en Salubrité Alimentaire, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
| | - Mohamed Rhouma
- Chaire de Recherche en Salubrité des Viandes, Département de Microbiologie et Pathologie, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
- Groupe de Recherche et d'Enseignement en Salubrité Alimentaire, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
| | - Marie-Pierre Létourneau-Montminy
- Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
- Chaire de Recherche sur les Stratégies Alternatives d'Alimentation des Porcs et des Volailles: Approche Systémique pour un Développement Durable, Faculté des Sciences de l'Agriculture et de l'Alimentation, Université Laval, Quebec, QC G1V 0A6, Canada
| | - William Thériault
- Chaire de Recherche en Salubrité des Viandes, Département de Microbiologie et Pathologie, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
- Groupe de Recherche et d'Enseignement en Salubrité Alimentaire, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
| | | | - Yves Arcand
- Agriculture et Agroalimentaire Canada, St-Hyacinthe, QC J2S 8E3, Canada
| | - Roland Leduc
- NUVAC Éco-Sciences, Valcourt, QC J0E 2L0, Canada
| | - Bruno Demers
- NUVAC Éco-Sciences, Valcourt, QC J0E 2L0, Canada
| | - Alexandre Thibodeau
- Chaire de Recherche en Salubrité des Viandes, Département de Microbiologie et Pathologie, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
- Groupe de Recherche et d'Enseignement en Salubrité Alimentaire, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, QC J2S 2M2, Canada
| |
Collapse
|
7
|
Das A, Behera RN, Kapoor A, Ambatipudi K. The Potential of Meta-Proteomics and Artificial Intelligence to Establish the Next Generation of Probiotics for Personalized Healthcare. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:17528-17542. [PMID: 37955263 DOI: 10.1021/acs.jafc.3c03834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The symbiosis of probiotic bacteria with humans has rendered various health benefits while providing nutrition and a suitable environment for their survival. However, the probiotics must survive unfavorable gut conditions to exert beneficial effects. The intrinsic resistance of probiotics to survive harsh conditions results from a myriad of proteins. Interaction of microbial proteins with the host is indispensable for modulating the gut microbiome, such as interaction with cell receptors and protective action against pathogens. The complex interplay of proteins should be unraveled by utilizing metaproteomic strategies. The contribution of probiotics to health is now widely accepted. However, due to the inconsistency of generalized probiotics, contemporary research toward precision probiotics has gained momentum for customized treatment. This review explores the application of metaproteomics and AI/ML algorithms in resolving multiomics data analysis and in silico prediction of microbial features for screening specific beneficial probiotic organisms. Implementing these integrative strategies could augment the potential of precision probiotics for personalized healthcare.
Collapse
Affiliation(s)
- Arpita Das
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Rama N Behera
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Ayushi Kapoor
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Kiran Ambatipudi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| |
Collapse
|
8
|
Liu S, Zhao Y, Feng X, Xu H. SARS-CoV-2 infection threatening intestinal health: A review of potential mechanisms and treatment strategies. Crit Rev Food Sci Nutr 2023; 63:12578-12596. [PMID: 35894645 DOI: 10.1080/10408398.2022.2103090] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The outbreak of the COVID-19 pandemic has brought great problems to mankind, including economic recession and poor health. COVID-19 patients are frequently reported with gastrointestinal symptoms such as diarrhea and vomiting in clinical diagnosis. Maintaining intestinal health is the key guarantee to maintain the normal function of multiple organs, otherwise it will be a disaster. Therefore, the purpose of this review was deeply understanded the potential mechanism of SARS-CoV-2 infection threatening intestinal health and put forward reasonable treatment strategies. Combined with the existing researches, we summarized the mechanism of SARS-CoV-2 infection threatening intestinal health, including intestinal microbiome disruption, intestinal barrier dysfunction, intestinal oxidative stress and intestinal cytokine storm. These adverse intestinal events may affect other organs through the circulatory system or aggravate the course of the disease. Typically, intestinal disadvantage may promote the progression of SARS-CoV-2 through the gut-lung axis and increase the disease degree of COVID-19 patients. In view of the lack of specific drugs to inhibit SARS-CoV-2 replication, the current review described new strategies of probiotics, prebiotics, postbiotics and nutrients to combat SARS-CoV-2 infection and maintain intestinal health. To provide new insights for the prevention and treatment of gastrointestinal symptoms and pneumonia in patients with COVID-19.
Collapse
Affiliation(s)
- Shanji Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yu Zhao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Xiaoyan Feng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
9
|
Kwoji ID, Aiyegoro OA, Okpeku M, Adeleke MA. 'Multi-omics' data integration: applications in probiotics studies. NPJ Sci Food 2023; 7:25. [PMID: 37277356 DOI: 10.1038/s41538-023-00199-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 05/22/2023] [Indexed: 06/07/2023] Open
Abstract
The concept of probiotics is witnessing increasing attention due to its benefits in influencing the host microbiome and the modulation of host immunity through the strengthening of the gut barrier and stimulation of antibodies. These benefits, combined with the need for improved nutraceuticals, have resulted in the extensive characterization of probiotics leading to an outburst of data generated using several 'omics' technologies. The recent development in system biology approaches to microbial science is paving the way for integrating data generated from different omics techniques for understanding the flow of molecular information from one 'omics' level to the other with clear information on regulatory features and phenotypes. The limitations and tendencies of a 'single omics' application to ignore the influence of other molecular processes justify the need for 'multi-omics' application in probiotics selections and understanding its action on the host. Different omics techniques, including genomics, transcriptomics, proteomics, metabolomics and lipidomics, used for studying probiotics and their influence on the host and the microbiome are discussed in this review. Furthermore, the rationale for 'multi-omics' and multi-omics data integration platforms supporting probiotics and microbiome analyses was also elucidated. This review showed that multi-omics application is useful in selecting probiotics and understanding their functions on the host microbiome. Hence, recommend a multi-omics approach for holistically understanding probiotics and the microbiome.
Collapse
Affiliation(s)
- Iliya Dauda Kwoji
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090, Durban, South Africa
| | - Olayinka Ayobami Aiyegoro
- Unit for Environmental Sciences and Management, North-West University, Potchefstroom, Northwest, South Africa
| | - Moses Okpeku
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090, Durban, South Africa
| | - Matthew Adekunle Adeleke
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090, Durban, South Africa.
| |
Collapse
|
10
|
Johnson D, Letchumanan V, Thum CC, Thurairajasingam S, Lee LH. A Microbial-Based Approach to Mental Health: The Potential of Probiotics in the Treatment of Depression. Nutrients 2023; 15:nu15061382. [PMID: 36986112 PMCID: PMC10053794 DOI: 10.3390/nu15061382] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
Probiotics are currently the subject of intensive research pursuits and also represent a multi-billion-dollar global industry given their vast potential to improve human health. In addition, mental health represents a key domain of healthcare, which currently has limited, adverse-effect prone treatment options, and probiotics may hold the potential to be a novel, customizable treatment for depression. Clinical depression is a common, potentially debilitating condition that may be amenable to a precision psychiatry-based approach utilizing probiotics. Although our understanding has not yet reached a sufficient level, this could be a therapeutic approach that can be tailored for specific individuals with their own unique set of characteristics and health issues. Scientifically, the use of probiotics as a treatment for depression has a valid basis rooted in the microbiota-gut-brain axis (MGBA) mechanisms, which play a role in the pathophysiology of depression. In theory, probiotics appear to be ideal as adjunct therapeutics for major depressive disorder (MDD) and as stand-alone therapeutics for mild MDD and may potentially revolutionize the treatment of depressive disorders. Although there is a wide range of probiotics and an almost limitless range of therapeutic combinations, this review aims to narrow the focus to the most widely commercialized and studied strains, namely Lactobacillus and Bifidobacterium, and to bring together the arguments for their usage in patients with major depressive disorder (MDD). Clinicians, scientists, and industrialists are critical stakeholders in exploring this groundbreaking concept.
Collapse
Affiliation(s)
- Dinyadarshini Johnson
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Vengadesh Letchumanan
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Pathogen Resistome Virulome and Diagnostic Research Group (PathRiD), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Chern Choong Thum
- Department of Psychiatry, Hospital Sultan Abdul Aziz Shah, Persiaran Mardi-UPM, Serdang 43400, Malaysia
| | - Sivakumar Thurairajasingam
- Clinical School Johor Bahru, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Johor Bahru 80100, Malaysia
- Correspondence: (S.T.); or (L.-H.L.)
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Pathogen Resistome Virulome and Diagnostic Research Group (PathRiD), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Correspondence: (S.T.); or (L.-H.L.)
| |
Collapse
|
11
|
de Deus C, Eduardo de Souza Brener C, Marques da Silva T, Somacal S, Queiroz Zepka L, Jacob Lopes E, de Bona da Silva C, Teixeira Barcia M, Lozano Sanchez J, Ragagnin de Menezes C. Co-encapsulation of Lactobacillus plantarum and bioactive compounds extracted from red beet stem (Beta vulgaris L.) by spray dryer. Food Res Int 2023; 167:112607. [PMID: 37087225 DOI: 10.1016/j.foodres.2023.112607] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/21/2023]
Abstract
Probiotic bacteria and bioactive compounds obtained from plant origin stand out as ingredients with the potential to increase the healthiness of functional foods, as there is currently a recurrent search for them. Probiotics and bioactive compounds are sensitive to intrinsic and extrinsic factors in the processing and packaging of the finished product. In this sense, the present study aims to evaluate the co-encapsulation by spray dryer (inlet air temperature 120 °C, air flow 40 L / min, pressure of 0.6 MPa and 1.5 mm nozzle diameter) of probiotic bacteria (L.plantarum) and compounds extracted from red beet stems (betalains) in order to verify the interaction between both and achieve better viability and resistance of the encapsulated material. When studying the co-encapsulation of L.plantarum and betalains extracted from beet stems, an unexpected influence was observed with a decrease in probiotic viability in the highest concentration of extract (100 %), on the other hand, the concentration of 50 % was the best enabled and maintained the survival of L.plantarum in conditions of 25 °C (63.06 %), 8 °C (88.80 %) and -18 °C (89.28 %). The viability of the betalains and the probiotic was better preserved in storage at 8 and -18 °C, where the encapsulated stability for 120 days was successfully achieved. Thus, the polyfunctional formulation developed in this study proved to be promising, as it expands the possibilities of application and development of new foods.
Collapse
|
12
|
Krittanawong C, Singh NK, Scheuring RA, Urquieta E, Bershad EM, Macaulay TR, Kaplin S, Dunn C, Kry SF, Russomano T, Shepanek M, Stowe RP, Kirkpatrick AW, Broderick TJ, Sibonga JD, Lee AG, Crucian BE. Human Health during Space Travel: State-of-the-Art Review. Cells 2022; 12:cells12010040. [PMID: 36611835 PMCID: PMC9818606 DOI: 10.3390/cells12010040] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
The field of human space travel is in the midst of a dramatic revolution. Upcoming missions are looking to push the boundaries of space travel, with plans to travel for longer distances and durations than ever before. Both the National Aeronautics and Space Administration (NASA) and several commercial space companies (e.g., Blue Origin, SpaceX, Virgin Galactic) have already started the process of preparing for long-distance, long-duration space exploration and currently plan to explore inner solar planets (e.g., Mars) by the 2030s. With the emergence of space tourism, space travel has materialized as a potential new, exciting frontier of business, hospitality, medicine, and technology in the coming years. However, current evidence regarding human health in space is very limited, particularly pertaining to short-term and long-term space travel. This review synthesizes developments across the continuum of space health including prior studies and unpublished data from NASA related to each individual organ system, and medical screening prior to space travel. We categorized the extraterrestrial environment into exogenous (e.g., space radiation and microgravity) and endogenous processes (e.g., alteration of humans' natural circadian rhythm and mental health due to confinement, isolation, immobilization, and lack of social interaction) and their various effects on human health. The aim of this review is to explore the potential health challenges associated with space travel and how they may be overcome in order to enable new paradigms for space health, as well as the use of emerging Artificial Intelligence based (AI) technology to propel future space health research.
Collapse
Affiliation(s)
- Chayakrit Krittanawong
- Department of Medicine and Center for Space Medicine, Section of Cardiology, Baylor College of Medicine, Houston, TX 77030, USA
- Translational Research Institute for Space Health, Houston, TX 77030, USA
- Department of Cardiovascular Diseases, New York University School of Medicine, New York, NY 10016, USA
- Correspondence: or (C.K.); (B.E.C.); Tel.: +1-713-798-4951 (C.K.); +1-281-483-0123 (B.E.C.)
| | - Nitin Kumar Singh
- Biotechnology and Planetary Protection Group, Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA 91109, USA
| | | | - Emmanuel Urquieta
- Translational Research Institute for Space Health, Houston, TX 77030, USA
- Department of Emergency Medicine and Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Eric M. Bershad
- Department of Neurology, Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Scott Kaplin
- Department of Cardiovascular Diseases, New York University School of Medicine, New York, NY 10016, USA
| | - Carly Dunn
- Department of Dermatology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephen F. Kry
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Marc Shepanek
- Office of the Chief Health and Medical Officer, NASA, Washington, DC 20546, USA
| | | | - Andrew W. Kirkpatrick
- Department of Surgery and Critical Care Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | | | - Jean D. Sibonga
- Division of Biomedical Research and Environmental Sciences, NASA Lyndon B. Johnson Space Center, Houston, TX 77058, USA
| | - Andrew G. Lee
- Department of Ophthalmology, University of Texas Medical Branch School of Medicine, Galveston, TX 77555, USA
- Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Ophthalmology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Ophthalmology, Texas A and M College of Medicine, College Station, TX 77807, USA
- Department of Ophthalmology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
- Departments of Ophthalmology, Neurology, and Neurosurgery, Weill Cornell Medicine, New York, NY 10021, USA
| | - Brian E. Crucian
- National Aeronautics and Space Administration (NASA) Johnson Space Center, Human Health and Performance Directorate, Houston, TX 77058, USA
- Correspondence: or (C.K.); (B.E.C.); Tel.: +1-713-798-4951 (C.K.); +1-281-483-0123 (B.E.C.)
| |
Collapse
|
13
|
Zhong Y, Fu D, Deng Z, Tang W, Mao J, Zhu T, Zhang Y, Liu J, Wang H. Lactic Acid Bacteria Mixture Isolated From Wild Pig Alleviated the Gut Inflammation of Mice Challenged by Escherichia coli. Front Immunol 2022; 13:822754. [PMID: 35154141 PMCID: PMC8825813 DOI: 10.3389/fimmu.2022.822754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Wild pigs usually showed high tolerance and resistance to several diseases in the wild environment, suggesting that the gut bacteria of wild pigs could be a good source for discovering potential probiotic strains. In our study, wild pig feces were sequenced and showed a higher relative abundance of the genus Lactobacillus (43.61% vs. 2.01%) than that in the domestic pig. A total of 11 lactic acid bacteria (LAB) strains including two L. rhamnosus, six L. mucosae, one L. fermentum, one L. delbrueckii, and one Enterococcus faecalis species were isolated. To investigate the synergistic effects of mixed probiotics strains, the mixture of 11 LAB strains from an intestinal ecology system was orally administrated in mice for 3 weeks, then the mice were challenged with Escherichia coli ATCC 25922 (2 × 109 CFU) and euthanized after challenge. Mice administrated with LAB strains showed higher (p < 0.05) LAB counts in feces and ileum. Moreover, alterations of specific bacterial genera occurred, including the higher (p < 0.05) relative abundance of Butyricicoccus and Clostridium IV and the lower (p < 0.05) abundance of Enterorhabdus in mice fed with mixed LAB strains. Mice challenged with Escherichia coli showed vacuolization of the liver, lower GSH in serum, and lower villus to the crypt proportion and Claudin-3 level in the gut. In contrast, administration of mixed LAB strains attenuated inflammation of the liver and gut, especially the lowered IL-6 and IL-1β levels (p < 0.05) in the gut. Our study highlighted the importance of gut bacterial diversity and the immunomodulation effects of LAB strains mixture from wild pig in gut health.
Collapse
Affiliation(s)
- Yifan Zhong
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Dongyan Fu
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Zhaoxi Deng
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Wenjie Tang
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Jiangdi Mao
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Tao Zhu
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Yu Zhang
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Jianxin Liu
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Haifeng Wang
- College of Animal Science, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| |
Collapse
|
14
|
McCoubrey LE, Seegobin N, Elbadawi M, Hu Y, Orlu M, Gaisford S, Basit AW. Active Machine learning for formulation of precision probiotics. Int J Pharm 2022; 616:121568. [PMID: 35150845 DOI: 10.1016/j.ijpharm.2022.121568] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 12/15/2022]
Abstract
It is becoming clear that the human gut microbiome is critical to health and well-being, with increasing evidence demonstrating that dysbiosis can promote disease. Increasingly, precision probiotics are being investigated as investigational drug products for restoration of healthy microbiome balance. To reach the distal gut alive where the density of microbiota is highest, oral probiotics should be protected from harsh conditions during transit through the stomach and small intestines. At present, few probiotic formulations are designed with this delivery strategy in mind. This study employs an emerging machine learning (ML) technique, known as active ML, to predict how excipients at pharmaceutically relevant concentrations affect the intestinal proliferation of a common probiotic, Lactobacillus paracasei. Starting with a labelled dataset of just 6 bacteria-excipient interactions, active ML was able to predict the effects of a further 111 excipients using uncertainty sampling. The average certainty of the final model was 67.70% and experimental validation demonstrated that 3/4 excipient-probiotic interactions could be correctly predicted. The model can be used to enable superior probiotic delivery to maximise proliferation in vivo and marks the first use of active ML in microbiome science.
Collapse
Affiliation(s)
- Laura E McCoubrey
- UCL School of Pharmacy, University College London, Brunswick Square, London WC1N 1AX, United Kingdom
| | - Nidhi Seegobin
- UCL School of Pharmacy, University College London, Brunswick Square, London WC1N 1AX, United Kingdom
| | - Moe Elbadawi
- UCL School of Pharmacy, University College London, Brunswick Square, London WC1N 1AX, United Kingdom
| | - Yiling Hu
- UCL School of Pharmacy, University College London, Brunswick Square, London WC1N 1AX, United Kingdom
| | - Mine Orlu
- UCL School of Pharmacy, University College London, Brunswick Square, London WC1N 1AX, United Kingdom
| | - Simon Gaisford
- UCL School of Pharmacy, University College London, Brunswick Square, London WC1N 1AX, United Kingdom
| | - Abdul W Basit
- UCL School of Pharmacy, University College London, Brunswick Square, London WC1N 1AX, United Kingdom.
| |
Collapse
|
15
|
Sabater C, Calvete-Torre I, Villamiel M, Moreno FJ, Margolles A, Ruiz L. Vegetable waste and by-products to feed a healthy gut microbiota: Current evidence, machine learning and computational tools to design novel microbiome-targeted foods. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
16
|
Rajha HN, Paule A, Aragonès G, Barbosa M, Caddeo C, Debs E, Dinkova R, Eckert GP, Fontana A, Gebrayel P, Maroun RG, Napolitano A, Panzella L, Pasinetti GM, Stevens JF, Schieber A, Edeas M. Recent Advances in Research on Polyphenols: Effects on Microbiota, Metabolism, and Health. Mol Nutr Food Res 2021; 66:e2100670. [PMID: 34806294 DOI: 10.1002/mnfr.202100670] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/23/2021] [Indexed: 01/02/2023]
Abstract
Polyphenols have attracted huge interest among researchers of various disciplines because of their numerous biological activities, such as antioxidative, antiinflammatory, antiapoptotic, cancer chemopreventive, anticarcinogenic, and antimicrobial properties, and their promising applications in many fields, mainly in the medical, cosmetics, dietary supplement and food industries. In this review, the latest scientific findings in the research on polyphenols interaction with the microbiome and mitochondria, their metabolism and health beneficial effects, their involvement in cognitive diseases and obesity development, as well as some innovations in their analysis, extraction methods, development of cosmetic formulations and functional food are summarized based on the papers presented at the 13th World Congress on Polyphenol Applications. Future implications of polyphenols in disease prevention and their strategic use as prophylactic measures are specifically addressed. Polyphenols may play a key role in our tomorrow´s food and nutrition to prevent many diseases.
Collapse
Affiliation(s)
| | - Armelle Paule
- International Society of Antioxidants in Nutrition and Health, Paris, France
| | | | | | | | | | - Rada Dinkova
- University of Food Technologies, Plovdiv, Bulgaria
| | | | | | - Prisca Gebrayel
- International Society of Antioxidants in Nutrition and Health, Paris, France
| | | | | | | | | | | | | | - Marvin Edeas
- University de Paris, Institut Cochin, Inserm, Paris, 1016, France
| |
Collapse
|
17
|
Pandey M, Bhati A, Priya K, Sharma KK, Singhal B. Precision Postbiotics and Mental Health: the Management of Post-COVID-19 Complications. Probiotics Antimicrob Proteins 2021; 14:426-448. [PMID: 34806151 PMCID: PMC8606251 DOI: 10.1007/s12602-021-09875-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2021] [Indexed: 01/14/2023]
Abstract
The health catastrophe originated by COVID-19 pandemic construed profound impact on a global scale. However, a plethora of research studies corroborated convincing evidence conferring severity of infection of SARS-CoV-2 with the aberrant gut microbiome that strongly speculated its importance for development of novel therapeutic modalities. The intense exploration of probiotics has been envisaged to promote the healthy growth of the host, and restore intestinal microecological balance through various metabolic and physiological processes. The demystifying effect of probiotics cannot be defied, but there exists a strong skepticism related to their safety and efficacy. Therefore, molecular signature of probiotics termed as "postbiotics" are of paramount importance and there is continuous surge of utilizing postbiotics for enhancing health benefits, but little is explicit about their antiviral effects. Therefore, it is worth considering their prospective role in post-COVID regime that pave the way for exploring the pastoral vistas of postbiotics. Based on previous research investigations, the present article advocates prospective role of postbiotics in alleviating the health burden of viral infections, especially SARS-CoV-2. The article also posits current challenges and proposes a futuristic model describing the concept of "precision postbiotics" for effective therapeutic and preventive interventions that can be used for management of this deadly disease.
Collapse
Affiliation(s)
- Muskan Pandey
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India
| | - Archana Bhati
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India
| | - Kumari Priya
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India
| | - K K Sharma
- Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Barkha Singhal
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India.
| |
Collapse
|
18
|
McCoubrey LE, Gaisford S, Orlu M, Basit AW. Predicting drug-microbiome interactions with machine learning. Biotechnol Adv 2021; 54:107797. [PMID: 34260950 DOI: 10.1016/j.biotechadv.2021.107797] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Pivotal work in recent years has cast light on the importance of the human microbiome in maintenance of health and physiological response to drugs. It is now clear that gastrointestinal microbiota have the metabolic power to promote, inactivate, or even toxify the efficacy of a drug to a level of clinically relevant significance. At the same time, it appears that drug intake has the propensity to alter gut microbiome composition, potentially affecting health and response to other drugs. Since the precise composition of an individual's microbiome is unique, one's drug-microbiome relationship is similarly unique. Thus, in the age of evermore personalised medicine, the ability to predict individuals' drug-microbiome interactions is highly sought. Machine learning (ML) offers a powerful toolkit capable of characterising and predicting drug-microbiota interactions at the individual patient level. ML techniques have the potential to learn the mechanisms operating drug-microbiome activities and measure patients' risk of such occurrences. This review will outline current knowledge at the drug-microbiota interface, and present ML as a technique for examining and forecasting personalised drug-microbiome interactions. When harnessed effectively, ML could alter how the pharmaceutical industry and healthcare professionals consider the drug-microbiome axis in patient care.
Collapse
Affiliation(s)
| | | | - Mine Orlu
- University College London, London, United Kingdom
| | | |
Collapse
|
19
|
Sabater C, Ruiz L, Margolles A. A Machine Learning Approach to Study Glycosidase Activities from Bifidobacterium. Microorganisms 2021; 9:1034. [PMID: 34064844 PMCID: PMC8151561 DOI: 10.3390/microorganisms9051034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/27/2022] Open
Abstract
This study aimed to recover metagenome-assembled genomes (MAGs) from human fecal samples to characterize the glycosidase profiles of Bifidobacterium species exposed to different prebiotic oligosaccharides (galacto-oligosaccharides, fructo-oligosaccharides and human milk oligosaccharides, HMOs) as well as high-fiber diets. A total of 1806 MAGs were recovered from 487 infant and adult metagenomes. Unsupervised and supervised classification of glycosidases codified in MAGs using machine-learning algorithms allowed establishing characteristic hydrolytic profiles for B. adolescentis, B. bifidum, B. breve, B. longum and B. pseudocatenulatum, yielding classification rates above 90%. Glycosidase families GH5 44, GH32, and GH110 were characteristic of B. bifidum. The presence or absence of GH1, GH2, GH5 and GH20 was characteristic of B. adolescentis, B. breve and B. pseudocatenulatum, while families GH1 and GH30 were relevant in MAGs from B. longum. These characteristic profiles allowed discriminating bifidobacteria regardless of prebiotic exposure. Correlation analysis of glycosidase activities suggests strong associations between glycosidase families comprising HMOs-degrading enzymes, which are often found in MAGs from the same species. Mathematical models here proposed may contribute to a better understanding of the carbohydrate metabolism of some common bifidobacteria species and could be extrapolated to other microorganisms of interest in future studies.
Collapse
Affiliation(s)
- Carlos Sabater
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA), Consejo Superior de Investigaciones Científicas (CSIC), Paseo Río Linares S/N, 33300 Villaviciosa, Asturias, Spain; (L.R.); (A.M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Lorena Ruiz
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA), Consejo Superior de Investigaciones Científicas (CSIC), Paseo Río Linares S/N, 33300 Villaviciosa, Asturias, Spain; (L.R.); (A.M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Abelardo Margolles
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA), Consejo Superior de Investigaciones Científicas (CSIC), Paseo Río Linares S/N, 33300 Villaviciosa, Asturias, Spain; (L.R.); (A.M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| |
Collapse
|