1
|
Li B, Liu J, He C, Deng Z, Zhou X, Peng R. Unveiling the Therapeutic Potential of Berberine in Rheumatoid Arthritis: A Comprehensive Study of Network Pharmacology, Metabolomics, and Intestinal Flora. J Inflamm Res 2024; 17:10849-10869. [PMID: 39677295 PMCID: PMC11645930 DOI: 10.2147/jir.s493892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease influenced by environmental triggers, including the commensal microbiota. Recent research has highlighted distinctive features of the gut microbiota in RA patients. This study investigates the therapeutic potential of berberine (BBR), a gut microbiota modulator known for its significant anti-RA effects, and elucidates the underlying mechanisms. Methods Utilizing the collagen-induced arthritis (CIA) rat model, we comprehensively evaluated the anti-rheumatoid arthritis effects of BBR in vivo through various indices, such as paw edema, arthritis index, ankle diameter, inflammatory cytokine levels, pathological conditions, and micro-CT analysis. Employing network pharmacology, we identified potential targets involved in RA alleviation by BBR. To analyze comprehensive metabolic profiles and identify underlying metabolic pathways, we conducted a serum-based widely targeted metabolomics analysis utilizing LC-MS technology. An integrated network encompassing metabolomics and network pharmacology data was constructed using Cytoscape. The potential therapeutic targets and signaling pathways of BBR in the management of RA were predicted using network pharmacology. Key targets and pathways were further validated by molecular docking and immunofluorescent staining, which integrated findings from serum metabolomics and network pharmacology analysis. Additionally, we analyzed the gut microbiota composition in rats employing 16S rDNA sequencing and investigated the effects of BBR on the microbiota of CIA rats through bioinformatics and statistical methods. Results Our results showed that BBR demonstrated significant efficacy in alleviating RA symptoms in CIA rats, as evidenced by improvements in paw redness and swelling, attenuation of bone and cartilage damage, reduction in synovial hyperplasia, inflammatory cell infiltration, and suppression of proinflammatory cytokines IL-1β, IL-6, IL-17A, and TNF-α. KEGG analysis highlighted the PI3K/AKT signaling pathway as a key mediator of BBR's anti-RA effects. Metabolomics profiling via LC-MS revealed 22 potential biomarkers. Arginine and proline metabolism, cutin, suberine and wax biosynthesis, glycine, serine and threonine metabolism and taurine and hypotaurine metabolism are the most related pathways of BBR anti-RA. Molecular docking studies corroborated high affinities between BBR and key targets. Furthermore, 16S analysis demonstrated BBR's capacity to modulate gut bacteria composition, including an increase in the abundance of Lachnoclostridium, Akkermansia, Blautia, Romboutsia, and Faecalibacterium genera, alongside a decrease in Prevotella_9 abundance in genus level. Integrated analysis underscored a strong correlation between serum microbiota and fecal metabolites. Conclusion Our findings elucidate the multifaceted mechanisms underlying BBR's therapeutic efficacy in RA, involving inhibition of the PI3K/AKT pathway, modulation of intestinal flora, and regulation of host metabolites. These insights provide novel perspectives on BBR's role in RA management.
Collapse
Affiliation(s)
- Bocun Li
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| | - Jing Liu
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| | - Chuan He
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| | - Zhou Deng
- Huazhong University of Science and Technology, Union Hospital, Tongji Medical College, Department of Acupuncture, Wuhan, Hubei, People’s Republic of China
| | - Xiaohong Zhou
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| | - Rui Peng
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, People’s Republic of China
| |
Collapse
|
2
|
Alrajeh S, Naveed Khan M, Irhash Putra A, Al-Ugaili DN, Alobaidi KH, Al Dossary O, Al-Obaidi JR, Jamaludin AA, Allawi MY, Al-Taie BS, Abdul Rahman N, Rahmad N. Mapping proteomic response to salinity stress tolerance in oil crops: Towards enhanced plant resilience. J Genet Eng Biotechnol 2024; 22:100432. [PMID: 39674646 PMCID: PMC11555348 DOI: 10.1016/j.jgeb.2024.100432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 12/16/2024]
Abstract
Exposure to saline environments significantly hampers the growth and productivity of oil crops, harmfully affecting their nutritional quality and suitability for biofuel production. This presents a critical challenge, as understanding salt tolerance mechanisms in crops is key to improving their performance in coastal and high-salinity regions. Our content might be read more properly: This review assembles current knowledge on protein-level changes related to salinity resistance in oil crops. From an extensive analysis of proteomic research, featured here are key genes and cellular pathways which react to salt stress. The literature evinces that cutting-edge proteomic approaches - such as 2D-DIGE, IF-MS/MS, and iTRAQ - have been required to reveal protein expression patterns in oil crops under salt conditions. These studies consistently uncover dramatic shifts in protein abundance associated with important physiological activities including antioxidant defence, stress-related signalling pathways, ion homeostasis, and osmotic regulation. Notably, proteins like ion channels (SOS1, NHX), osmolytes (proline, glycine betaine), antioxidant enzymes (SOD, CAT), and stress-related proteins (HSPs, LEA) play central roles in maintaining cellular balance and reducing oxidative stress. These findings underline the complex regulatory networks that govern oil crop salt tolerance. The application of this proteomic information can inform breeding and genetic engineering strategies to enhance salt resistance. Future research should aim to integrate multiple omics data to gain a comprehensive view of salinity responses and identify potential markers for crop improvement.
Collapse
Affiliation(s)
- Sarah Alrajeh
- Department of Biology, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, 35900 Tanjong Malim, Perak, Malaysia
| | - Muhammad Naveed Khan
- Department of Biology, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, 35900 Tanjong Malim, Perak, Malaysia
| | - Aidhya Irhash Putra
- Department of Biology, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, 35900 Tanjong Malim, Perak, Malaysia
| | - Dhafar N Al-Ugaili
- Department of Molecular and Medical Biotechnology, College of Biotechnology, AL-Nahrain University, Jadriya, Baghdad, Iraq
| | - Khalid H Alobaidi
- Department of Plant Biotechnology, College of Biotechnology, AL-Nahrain University, Baghdad, Iraq
| | - Othman Al Dossary
- Agricultural Biotechnology Department, College of Agriculture and Food Sciences, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Jameel R Al-Obaidi
- Department of Biology, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, 35900 Tanjong Malim, Perak, Malaysia; Applied Science Research Center. Applied Science Private University, Amman, Jordan.
| | - Azi Azeyanty Jamaludin
- Department of Biology, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, 35900 Tanjong Malim, Perak, Malaysia; Center of Biodiversity and Conservation, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, 35900 Tanjung Malim, Perak, Malaysia
| | - Mohammed Yahya Allawi
- Environmental Health Department, College of Environmental Sciences, University of Mosul, 41002 Mosul, Iraq
| | - Bilal Salim Al-Taie
- Environmental Health Department, College of Environmental Sciences, University of Mosul, 41002 Mosul, Iraq
| | - Norafizah Abdul Rahman
- Gene Marker Laboratory, Faculty of Agriculture and Life Sciences (AGLS), Science South Building, Lincoln University, Lincoln, 7608 Canterbury, New Zealand
| | - Norasfaliza Rahmad
- Agro-Biotechnology Institute, National Institutes of Biotechnology Malaysia, Jalan Bioteknologi, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
3
|
Padhi B, Liu R, Yang Y, Peng X, Li L, Zhang P, Zhang P. Using multiple drug similarity networks to promote adverse drug event detection. Heliyon 2024; 10:e39728. [PMID: 39748955 PMCID: PMC11693886 DOI: 10.1016/j.heliyon.2024.e39728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 01/04/2025] Open
Abstract
The occurrence of an adverse drug event (ADE) has become a serious social concern of public health. Early detection of ADEs can lower the risk of drug safety as well as the expense of the drug. While post-market spontaneous reports of ADEs remain a cornerstone of pharmacovigilance, most existing signal detection algorithms rely on substantial accumulated data, limiting their applicability to early ADE detection when reports are scarce. To address this issue, we propose a label propagation model for generating enhanced drug safety signals using multiple drug features. We first construct multiple drug similarity networks using a range of drug features. We then calculate initial drug safety signals using conventional signal detection algorithms. These original signals are subsequently propagated across each drug similarity network to obtain enhanced drug safety signals. We evaluate our proposed model using two common signal detection algorithms on data from the FDA Adverse Event Reporting System (FAERS). Results demonstrate that enhanced drug safety signals with pre-clinical information outperform the standard safety signal detection algorithms on early ADE detection. In addition, we systematically evaluate the performance of different drug similarities against different types of ADEs. Furthermore, we have developed a web interface (http://drug-drug-sim.aimedlab.net/) to display our multiple drug similarity scores, facilitating access to this valuable resource for drug safety monitoring.
Collapse
Affiliation(s)
- Biswajit Padhi
- Department of Computer Science and Engineering, The Ohio State University, 2015 Neil Ave, Columbus, OH 43210, USA
| | - Ruoqi Liu
- Department of Computer Science and Engineering, The Ohio State University, 2015 Neil Ave, Columbus, OH 43210, USA
| | - Yuedi Yang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, 410 W. 10th Street HITS 3000, Indianapolis, IN 46202, USA
| | - Xueqiao Peng
- Department of Computer Science and Engineering, The Ohio State University, 2015 Neil Ave, Columbus, OH 43210, USA
| | - Lang Li
- Department of Biomedical Informatics, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, USA
| | - Pengyue Zhang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, 410 W. 10th Street HITS 3000, Indianapolis, IN 46202, USA
| | - Ping Zhang
- Department of Computer Science and Engineering, The Ohio State University, 2015 Neil Ave, Columbus, OH 43210, USA
- Department of Biomedical Informatics, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, USA
- Translational Data Analytics institute, The Ohio State University, 1760 Neil Ave, Columbus, OH 43210, USA
| |
Collapse
|
4
|
Kundu P, Beura S, Mondal S, Das AK, Ghosh A. Machine learning for the advancement of genome-scale metabolic modeling. Biotechnol Adv 2024; 74:108400. [PMID: 38944218 DOI: 10.1016/j.biotechadv.2024.108400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 05/13/2024] [Accepted: 06/23/2024] [Indexed: 07/01/2024]
Abstract
Constraint-based modeling (CBM) has evolved as the core systems biology tool to map the interrelations between genotype, phenotype, and external environment. The recent advancement of high-throughput experimental approaches and multi-omics strategies has generated a plethora of new and precise information from wide-ranging biological domains. On the other hand, the continuously growing field of machine learning (ML) and its specialized branch of deep learning (DL) provide essential computational architectures for decoding complex and heterogeneous biological data. In recent years, both multi-omics and ML have assisted in the escalation of CBM. Condition-specific omics data, such as transcriptomics and proteomics, helped contextualize the model prediction while analyzing a particular phenotypic signature. At the same time, the advanced ML tools have eased the model reconstruction and analysis to increase the accuracy and prediction power. However, the development of these multi-disciplinary methodological frameworks mainly occurs independently, which limits the concatenation of biological knowledge from different domains. Hence, we have reviewed the potential of integrating multi-disciplinary tools and strategies from various fields, such as synthetic biology, CBM, omics, and ML, to explore the biochemical phenomenon beyond the conventional biological dogma. How the integrative knowledge of these intersected domains has improved bioengineering and biomedical applications has also been highlighted. We categorically explained the conventional genome-scale metabolic model (GEM) reconstruction tools and their improvement strategies through ML paradigms. Further, the crucial role of ML and DL in omics data restructuring for GEM development has also been briefly discussed. Finally, the case-study-based assessment of the state-of-the-art method for improving biomedical and metabolic engineering strategies has been elaborated. Therefore, this review demonstrates how integrating experimental and in silico strategies can help map the ever-expanding knowledge of biological systems driven by condition-specific cellular information. This multiview approach will elevate the application of ML-based CBM in the biomedical and bioengineering fields for the betterment of society and the environment.
Collapse
Affiliation(s)
- Pritam Kundu
- School School of Energy Science and Engineering, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Satyajit Beura
- Department of Bioscience and Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Suman Mondal
- P.K. Sinha Centre for Bioenergy and Renewables, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Amit Kumar Das
- Department of Bioscience and Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Amit Ghosh
- School School of Energy Science and Engineering, Indian Institute of Technology Kharagpur, West Bengal 721302, India; P.K. Sinha Centre for Bioenergy and Renewables, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| |
Collapse
|
5
|
Jamal QMS, Ahmad V. Bacterial metabolomics: current applications for human welfare and future aspects. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024:1-24. [PMID: 39078342 DOI: 10.1080/10286020.2024.2385365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
An imbalanced microbiome is linked to several diseases, such as cancer, inflammatory bowel disease, obesity, and even neurological disorders. Bacteria and their by-products are used for various industrial and clinical purposes. The metabolites under discussion were chosen based on their biological impacts on host and gut microbiota interactions as established by metabolome research. The separation of bacterial metabolites by using statistics and machine learning analysis creates new opportunities for applications of bacteria and their metabolites in the environmental and medical sciences. Thus, the metabolite production strategies, methodologies, and importance of bacterial metabolites for human well-being are discussed in this review.
Collapse
Affiliation(s)
- Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Varish Ahmad
- Health Information Technology Department, The Applied College, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
6
|
Feng F, Hu P, Peng L, Xu L, Chen J, Chen Q, Zhang X, Tao X. Integrated network pharmacology and metabolomics to reveal the mechanism of Pinellia ternata inhibiting non-small cell lung cancer cells. BMC Complement Med Ther 2024; 24:263. [PMID: 38992647 PMCID: PMC11238457 DOI: 10.1186/s12906-024-04574-3] [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: 09/13/2023] [Accepted: 07/01/2024] [Indexed: 07/13/2024] Open
Abstract
Lung cancer is a malignant tumor with highly heterogeneous characteristics. A classic Chinese medicine, Pinellia ternata (PT), was shown to exert therapeutic effects on lung cancer cells. However, its chemical and pharmacological profiles are not yet understood. In the present study, we aimed to reveal the mechanism of PT in treating lung cancer cells through metabolomics and network pharmacology. Metabolomic analysis of two strains of lung cancer cells treated with Pinellia ternata extracts (PTE) was used to identify differentially abundant metabolites, and the metabolic pathways associated with the DEGs were identified by MetaboAnalyst. Then, network pharmacology was applied to identify potential targets against PTE-induced lung cancer cells. The integrated network of metabolomics and network pharmacology was constructed based on Cytoscape. PTE obviously inhibited the proliferation, migration and invasion of A549 and NCI-H460 cells. The results of the cellular metabolomics analysis showed that 30 metabolites were differentially expressed in the lung cancer cells of the experimental and control groups. Through pathway enrichment analysis, 5 metabolites were found to be involved in purine metabolism, riboflavin metabolism and the pentose phosphate pathway, including D-ribose 5-phosphate, xanthosine, 5-amino-4-imidazolecarboxyamide, flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD). Combined with network pharmacology, 11 bioactive compounds were found in PT, and networks of bioactive compound-target gene-metabolic enzyme-metabolite interactions were constructed. In conclusion, this study revealed the complicated mechanisms of PT against lung cancer. Our work provides a novel paradigm for identifying the potential mechanisms underlying the pharmacological effects of natural compounds.
Collapse
Affiliation(s)
- Fan Feng
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Ping Hu
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Lei Peng
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Lisheng Xu
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Jun Chen
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Qiong Chen
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Xingtao Zhang
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China
| | - Xingkui Tao
- School of Biological and Food Engineering, Suzhou University, Anhui, 234000, China.
| |
Collapse
|
7
|
Vaparanta K, Merilahti JAM, Ojala VK, Elenius K. De Novo Multi-Omics Pathway Analysis Designed for Prior Data Independent Inference of Cell Signaling Pathways. Mol Cell Proteomics 2024; 23:100780. [PMID: 38703893 PMCID: PMC11259815 DOI: 10.1016/j.mcpro.2024.100780] [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: 07/07/2023] [Revised: 04/07/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024] Open
Abstract
New tools for cell signaling pathway inference from multi-omics data that are independent of previous knowledge are needed. Here, we propose a new de novo method, the de novo multi-omics pathway analysis (DMPA), to model and combine omics data into network modules and pathways. DMPA was validated with published omics data and was found accurate in discovering reported molecular associations in transcriptome, interactome, phosphoproteome, methylome, and metabolomics data, and signaling pathways in multi-omics data. DMPA was benchmarked against module discovery and multi-omics integration methods and outperformed previous methods in module and pathway discovery especially when applied to datasets of relatively low sample sizes. Transcription factor, kinase, subcellular location, and function prediction algorithms were devised for transcriptome, phosphoproteome, and interactome modules and pathways, respectively. To apply DMPA in a biologically relevant context, interactome, phosphoproteome, transcriptome, and proteome data were collected from analyses carried out using melanoma cells to address gamma-secretase cleavage-dependent signaling characteristics of the receptor tyrosine kinase TYRO3. The pathways modeled with DMPA reflected the predicted function and its direction in validation experiments.
Collapse
Affiliation(s)
- Katri Vaparanta
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland; Medicity Research Laboratories, University of Turku, Turku, Finland; Institute of Biomedicine, University of Turku, Turku, Finland.
| | - Johannes A M Merilahti
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland; Medicity Research Laboratories, University of Turku, Turku, Finland; Institute of Biomedicine, University of Turku, Turku, Finland
| | - Veera K Ojala
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland; Medicity Research Laboratories, University of Turku, Turku, Finland; Institute of Biomedicine, University of Turku, Turku, Finland
| | - Klaus Elenius
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland; Medicity Research Laboratories, University of Turku, Turku, Finland; Institute of Biomedicine, University of Turku, Turku, Finland; Department of Oncology, Turku University Hospital, Turku, Finland.
| |
Collapse
|
8
|
Ewald JD, Zhou G, Lu Y, Kolic J, Ellis C, Johnson JD, Macdonald PE, Xia J. Web-based multi-omics integration using the Analyst software suite. Nat Protoc 2024; 19:1467-1497. [PMID: 38355833 DOI: 10.1038/s41596-023-00950-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 11/21/2023] [Indexed: 02/16/2024]
Abstract
The growing number of multi-omics studies demands clear conceptual workflows coupled with easy-to-use software tools to facilitate data analysis and interpretation. This protocol covers three key components involved in multi-omics analysis, including single-omics data analysis, knowledge-driven integration using biological networks and data-driven integration through joint dimensionality reduction. Using the dataset from a recent multi-omics study of human pancreatic islet tissue and plasma samples, the first section introduces how to perform transcriptomics/proteomics data analysis using ExpressAnalyst and lipidomics data analysis using MetaboAnalyst. On the basis of significant features detected in these workflows, the second section demonstrates how to perform knowledge-driven integration using OmicsNet. The last section illustrates how to perform data-driven integration from the normalized omics data and metadata using OmicsAnalyst. The complete protocol can be executed in ~2 h. Compared with other available options for multi-omics integration, the Analyst software suite described in this protocol enables researchers to perform a wide range of omics data analysis tasks via a user-friendly web interface.
Collapse
Affiliation(s)
- Jessica D Ewald
- Institute of Parasitology, McGill University, Montreal, Quebec, Canada
| | - Guangyan Zhou
- Institute of Parasitology, McGill University, Montreal, Quebec, Canada
| | - Yao Lu
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Jelena Kolic
- Life Sciences Institute, Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cara Ellis
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - James D Johnson
- Life Sciences Institute, Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Patrick E Macdonald
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Jianguo Xia
- Institute of Parasitology, McGill University, Montreal, Quebec, Canada.
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
9
|
Muller E, Shiryan I, Borenstein E. Multi-omic integration of microbiome data for identifying disease-associated modules. Nat Commun 2024; 15:2621. [PMID: 38521774 PMCID: PMC10960825 DOI: 10.1038/s41467-024-46888-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/08/2024] [Indexed: 03/25/2024] Open
Abstract
Multi-omic studies of the human gut microbiome are crucial for understanding its role in disease across multiple functional layers. Nevertheless, integrating and analyzing such complex datasets poses significant challenges. Most notably, current analysis methods often yield extensive lists of disease-associated features (e.g., species, pathways, or metabolites), without capturing the multi-layered structure of the data. Here, we address this challenge by introducing "MintTea", an intermediate integration-based approach combining canonical correlation analysis extensions, consensus analysis, and an evaluation protocol. MintTea identifies "disease-associated multi-omic modules", comprising features from multiple omics that shift in concord and that collectively associate with the disease. Applied to diverse cohorts, MintTea captures modules with high predictive power, significant cross-omic correlations, and alignment with known microbiome-disease associations. For example, analyzing samples from a metabolic syndrome study, MintTea identifies a module with serum glutamate- and TCA cycle-related metabolites, along with bacterial species linked to insulin resistance. In another dataset, MintTea identifies a module associated with late-stage colorectal cancer, including Peptostreptococcus and Gemella species and fecal amino acids, in line with these species' metabolic activity and their coordinated gradual increase with cancer development. This work demonstrates the potential of advanced integration methods in generating systems-level, multifaceted hypotheses underlying microbiome-disease interactions.
Collapse
Affiliation(s)
- Efrat Muller
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Itamar Shiryan
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Elhanan Borenstein
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel.
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
- Santa Fe Institute, Santa Fe, NM, USA.
| |
Collapse
|
10
|
Tian D, Xu T, Kang H, Luo H, Wang Y, Chen M, Li R, Ma L, Wang Z, Hao L, Tang B, Zou D, Xiao J, Zhao W, Bao Y, Zhang Z, Song S. Plant genomic resources at National Genomics Data Center: assisting in data-driven breeding applications. ABIOTECH 2024; 5:94-106. [PMID: 38576435 PMCID: PMC10987443 DOI: 10.1007/s42994-023-00134-4] [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: 11/10/2023] [Accepted: 12/18/2023] [Indexed: 04/06/2024]
Abstract
Genomic data serve as an invaluable resource for unraveling the intricacies of the higher plant systems, including the constituent elements within and among species. Through various efforts in genomic data archiving, integrative analysis and value-added curation, the National Genomics Data Center (NGDC), which is a part of the China National Center for Bioinformation (CNCB), has successfully established and currently maintains a vast amount of database resources. This dedicated initiative of the NGDC facilitates a data-rich ecosystem that greatly strengthens and supports genomic research efforts. Here, we present a comprehensive overview of central repositories dedicated to archiving, presenting, and sharing plant omics data, introduce knowledgebases focused on variants or gene-based functional insights, highlight species-specific multiple omics database resources, and briefly review the online application tools. We intend that this review can be used as a guide map for plant researchers wishing to select effective data resources from the NGDC for their specific areas of study. Supplementary Information The online version contains supplementary material available at 10.1007/s42994-023-00134-4.
Collapse
Affiliation(s)
- Dongmei Tian
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
| | - Tianyi Xu
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
| | - Hailong Kang
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Hong Luo
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
| | - Yanqing Wang
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
| | - Meili Chen
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
| | - Rujiao Li
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
| | - Lina Ma
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
| | - Zhonghuang Wang
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Lili Hao
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
| | - Bixia Tang
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
| | - Dong Zou
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
| | - Jingfa Xiao
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Wenming Zhao
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Yiming Bao
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Zhang Zhang
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Shuhui Song
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences & China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
11
|
Costes V, Sellem E, Marthey S, Hoze C, Bonnet A, Schibler L, Kiefer H, Jaffrezic F. Multi-omics data integration for the identification of biomarkers for bull fertility. PLoS One 2024; 19:e0298623. [PMID: 38394258 PMCID: PMC10890740 DOI: 10.1371/journal.pone.0298623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Bull fertility is an important economic trait, and the use of subfertile semen for artificial insemination decreases the global efficiency of the breeding sector. Although the analysis of semen functional parameters can help to identify infertile bulls, no tools are currently available to enable precise predictions and prevent the commercialization of subfertile semen. Because male fertility is a multifactorial phenotype that is dependent on genetic, epigenetic, physiological and environmental factors, we hypothesized that an integrative analysis might help to refine our knowledge and understanding of bull fertility. We combined -omics data (genotypes, sperm DNA methylation at CpGs and sperm small non-coding RNAs) and semen parameters measured on a large cohort of 98 Montbéliarde bulls with contrasting fertility levels. Multiple Factor Analysis was conducted to study the links between the datasets and fertility. Four methodologies were then considered to identify the features linked to bull fertility variation: Logistic Lasso, Random Forest, Gradient Boosting and Neural Networks. Finally, the features selected by these methods were annotated in terms of genes, to conduct functional enrichment analyses. The less relevant features in -omics data were filtered out, and MFA was run on the remaining 12,006 features, including the 11 semen parameters and a balanced proportion of each type of-omics data. The results showed that unlike the semen parameters studied the-omics datasets were related to fertility. Biomarkers related to bull fertility were selected using the four methodologies mentioned above. The most contributory CpGs, SNPs and miRNAs targeted genes were all found to be involved in development. Interestingly, fragments derived from ribosomal RNAs were overrepresented among the selected features, suggesting roles in male fertility. These markers could be used in the future to identify subfertile bulls in order to increase the global efficiency of the breeding sector.
Collapse
Affiliation(s)
- Valentin Costes
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
- R&D Department, ELIANCE, 149 rue de Bercy, Paris, France
- Université Paris-Saclay, AgroParisTech, INRAE, GABI, Jouy-en-Josas, France
| | - Eli Sellem
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
- R&D Department, ELIANCE, 149 rue de Bercy, Paris, France
| | - Sylvain Marthey
- Université Paris-Saclay, AgroParisTech, INRAE, GABI, Jouy-en-Josas, France
- INRAE, MaIAGE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Chris Hoze
- R&D Department, ELIANCE, 149 rue de Bercy, Paris, France
- Université Paris-Saclay, AgroParisTech, INRAE, GABI, Jouy-en-Josas, France
| | - Aurélie Bonnet
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
- R&D Department, ELIANCE, 149 rue de Bercy, Paris, France
| | | | - Hélène Kiefer
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Florence Jaffrezic
- Université Paris-Saclay, AgroParisTech, INRAE, GABI, Jouy-en-Josas, France
| |
Collapse
|
12
|
Shakyawar SK, Sajja BR, Patel JC, Guda C. iCluF: an unsupervised iterative cluster-fusion method for patient stratification using multiomics data. BIOINFORMATICS ADVANCES 2024; 4:vbae015. [PMID: 38698887 PMCID: PMC11063539 DOI: 10.1093/bioadv/vbae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/10/2023] [Accepted: 01/26/2024] [Indexed: 05/05/2024]
Abstract
Motivation Patient stratification is crucial for the effective treatment or management of heterogeneous diseases, including cancers. Multiomic technologies facilitate molecular characterization of human diseases; however, the complexity of data warrants the need for the development of robust data integration tools for patient stratification using machine-learning approaches. Results iCluF iteratively integrates three types of multiomic data (mRNA, miRNA, and DNA methylation) using pairwise patient similarity matrices built from each omic data. The intermediate omic-specific neighborhood matrices implement iterative matrix fusion and message passing among the similarity matrices to derive a final integrated matrix representing all the omics profiles of a patient, which is used to further cluster patients into subtypes. iCluF outperforms other methods with significant differences in the survival profiles of 8581 patients belonging to 30 different cancers in TCGA. iCluF also predicted the four intrinsic subtypes of Breast Invasive Carcinomas with adjusted rand index and Fowlkes-Mallows scores of 0.72 and 0.83, respectively. The Gini importance score showed that methylation features were the primary decisive players, followed by mRNA and miRNA to identify disease subtypes. iCluF can be applied to stratify patients with any disease containing multiomic datasets. Availability and implementation Source code and datasets are available at https://github.com/GudaLab/iCluF_core.
Collapse
Affiliation(s)
- Sushil K Shakyawar
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Balasrinivasa R Sajja
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Jai Chand Patel
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Genetics, Cell Biology and Anatomy, Center for Biomedical Informatics Research and Innovation, University of Nebraska Medical Center, Omaha, NE 68198-5805, United States
| |
Collapse
|
13
|
Mardoc E, Sow MD, Déjean S, Salse J. Genomic data integration tutorial, a plant case study. BMC Genomics 2024; 25:66. [PMID: 38233804 PMCID: PMC10792847 DOI: 10.1186/s12864-023-09833-0] [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: 07/21/2023] [Accepted: 11/22/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND The ongoing evolution of the Next Generation Sequencing (NGS) technologies has led to the production of genomic data on a massive scale. While tools for genomic data integration and analysis are becoming increasingly available, the conceptual and analytical complexities still represent a great challenge in many biological contexts. RESULTS To address this issue, we describe a six-steps tutorial for the best practices in genomic data integration, consisting of (1) designing a data matrix; (2) formulating a specific biological question toward data description, selection and prediction; (3) selecting a tool adapted to the targeted questions; (4) preprocessing of the data; (5) conducting preliminary analysis, and finally (6) executing genomic data integration. CONCLUSION The tutorial has been tested and demonstrated on publicly available genomic data generated from poplar (Populus L.), a woody plant model. We also developed a new graphical output for the unsupervised multi-block analysis, cimDiablo_v2, available at https://forgemia.inra.fr/umr-gdec/omics-integration-on-poplar , and allowing the selection of master drivers in genomic data variation and interplay.
Collapse
Affiliation(s)
- Emile Mardoc
- UCA-INRAE UMR 1095 Genetics, Diversity and Ecophysiology of Cereals (GDEC), 5 Chemin de Beaulieu, 63000, Clermont-Ferrand, France
| | - Mamadou Dia Sow
- UCA-INRAE UMR 1095 Genetics, Diversity and Ecophysiology of Cereals (GDEC), 5 Chemin de Beaulieu, 63000, Clermont-Ferrand, France
| | - Sébastien Déjean
- Institut de Mathématiques de Toulouse, UMR 5219, Université de Toulouse, CNRS, Université Paul Sabatier, Toulouse, France
| | - Jérôme Salse
- UCA-INRAE UMR 1095 Genetics, Diversity and Ecophysiology of Cereals (GDEC), 5 Chemin de Beaulieu, 63000, Clermont-Ferrand, France.
| |
Collapse
|
14
|
Wieder C, Cooke J, Frainay C, Poupin N, Bowler R, Jourdan F, Kechris KJ, Lai RP, Ebbels T. PathIntegrate: Multivariate modelling approaches for pathway-based multi-omics data integration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574780. [PMID: 38260498 PMCID: PMC10802464 DOI: 10.1101/2024.01.09.574780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
As terabytes of multi-omics data are being generated, there is an ever-increasing need for methods facilitating the integration and interpretation of such data. Current multi-omics integration methods typically output lists, clusters, or subnetworks of molecules related to an outcome. Even with expert domain knowledge, discerning the biological processes involved is a time-consuming activity. Here we propose PathIntegrate, a method for integrating multi-omics datasets based on pathways, designed to exploit knowledge of biological systems and thus provide interpretable models for such studies. PathIntegrate employs single-sample pathway analysis to transform multi-omics datasets from the molecular to the pathway-level, and applies a predictive single-view or multi-view model to integrate the data. Model outputs include multi-omics pathways ranked by their contribution to the outcome prediction, the contribution of each omics layer, and the importance of each molecule in a pathway. Using semi-synthetic data we demonstrate the benefit of grouping molecules into pathways to detect signals in low signal-to-noise scenarios, as well as the ability of PathIntegrate to precisely identify important pathways at low effect sizes. Finally, using COPD and COVID-19 data we showcase how PathIntegrate enables convenient integration and interpretation of complex high-dimensional multi-omics datasets. The PathIntegrate Python package is available at https://github.com/cwieder/PathIntegrate.
Collapse
Affiliation(s)
- Cecilia Wieder
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Juliette Cooke
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Clement Frainay
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Nathalie Poupin
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Russell Bowler
- National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA
| | - Fabien Jourdan
- MetaboHUB-Metatoul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Katerina J Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Rachel Pj Lai
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Timothy Ebbels
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
15
|
Grison S, Braga-Tanaka II, Baatout S, Klokov D. In utero exposure to ionizing radiation and metabolic regulation: perspectives for future multi- and trans-generation effects studies. Int J Radiat Biol 2024; 100:1283-1296. [PMID: 38180060 DOI: 10.1080/09553002.2023.2295293] [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: 05/30/2023] [Revised: 11/03/2023] [Accepted: 11/22/2023] [Indexed: 01/06/2024]
Abstract
PURPOSE The radiation protection community has been particularly attentive to the risks of delayed effects on offspring from low dose or low dose-rate exposures to ionizing radiation. Despite this, the current epidemiologic studies and scientific data are still insufficient to provide the necessary evidence for improving risk assessment guidelines. This literature review aims to inform future studies on multigenerational and transgenerational effects. It primarily focuses on animal studies involving in utero exposure and discusses crucial elements for interpreting the results. These elements include in utero exposure scenarios relative to the developmental stages of the embryo/fetus, and the primary biological mechanisms responsible for transmitting heritable or hereditary effects to future generations. The review addresses several issues within the contexts of both multigenerational and transgenerational effects, with a focus on hereditary perspectives. CONCLUSIONS Knowledge consolidation in the field of Developmental Origins of Health and Disease (DOHaD) has led us to propose a new study strategy. This strategy aims to address the transgenerational effects of in utero exposure to low dose and low dose-rate radiation. Within this concept, there is a possibility that disruption of epigenetic programming in embryonic and fetal cells may occur. This disruption could lead to metabolic dysfunction, which in turn may cause abnormal responses to future environmental challenges, consequently increasing disease risk. Lastly, we discuss methodological limitations in our studies. These limitations are related to cohort size, follow-up time, model radiosensitivity, and analytical techniques. We propose scientific and analytical strategies for future research in this field.
Collapse
Affiliation(s)
- Stéphane Grison
- PSE-SANTE, Institut de Radioprotection et de Sûreté Nucléaire, Fontenay-aux-Roses, France
| | - Ignacia Iii Braga-Tanaka
- Department of Radiobiology, Institute for Environmental Sciences (IES), Rokkasho Kamikita, Aomori, Japan
| | - Sarah Baatout
- Belgian Nuclear Research Centre, SCK CEN, Institute of Nuclear Medical Applications, Mol, Belgium
- Department of Molecular Biotechnology (BW25) and Department of Human Structure and Repair (GE38), Ghent University, Ghent, Belgium
| | - Dmitry Klokov
- PSE-SANTE, Institut de Radioprotection et de Sûreté Nucléaire, Fontenay-aux-Roses, France
- Department of Microbiology, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
16
|
Yang B, Meng T, Wang X, Li J, Zhao S, Wang Y, Yi S, Zhou Y, Zhang Y, Li L, Guo L. CAT Bridge: an efficient toolkit for gene-metabolite association mining from multiomics data. Gigascience 2024; 13:giae083. [PMID: 39517109 PMCID: PMC11548955 DOI: 10.1093/gigascience/giae083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/08/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND With advancements in sequencing and mass spectrometry technologies, multiomics data can now be easily acquired for understanding complex biological systems. Nevertheless, substantial challenges remain in determining the association between gene-metabolite pairs due to the nonlinear and multifactorial interactions within cellular networks. The complexity arises from the interplay of multiple genes and metabolites, often involving feedback loops and time-dependent regulatory mechanisms that are not easily captured by traditional analysis methods. FINDINGS Here, we introduce Compounds And Transcripts Bridge (abbreviated as CAT Bridge, available at https://catbridge.work), a free user-friendly platform for longitudinal multiomics analysis to efficiently identify transcripts associated with metabolites using time-series omics data. To evaluate the association of gene-metabolite pairs, CAT Bridge is a pioneering work benchmarking a set of statistical methods spanning causality estimation and correlation coefficient calculation for multiomics analysis. Additionally, CAT Bridge features an artificial intelligence agent to assist users interpreting the association results. CONCLUSIONS We applied CAT Bridge to experimentally obtained Capsicum chinense (chili pepper) and public human and Escherichia coli time-series transcriptome and metabolome datasets. CAT Bridge successfully identified genes involved in the biosynthesis of capsaicin in C. chinense. Furthermore, case study results showed that the convergent cross-mapping method outperforms traditional approaches in longitudinal multiomics analyses. CAT Bridge simplifies access to various established methods for longitudinal multiomics analysis and enables researchers to swiftly identify associated gene-metabolite pairs for further validation.
Collapse
Affiliation(s)
- Bowen Yang
- Shandong Key Laboratory of Precision Molecular Crop Design and Breeding, Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang 261325, China
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Tan Meng
- Shandong Key Laboratory of Precision Molecular Crop Design and Breeding, Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang 261325, China
| | - Xinrui Wang
- Shandong Key Laboratory of Precision Molecular Crop Design and Breeding, Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang 261325, China
| | - Jun Li
- Shandong Key Laboratory of Precision Molecular Crop Design and Breeding, Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang 261325, China
| | - Shuang Zhao
- The Metabolomics Innovation Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Yingheng Wang
- Department of Computer Science, Cornell University, Ithaca, NY 14853, USA
| | - Shu Yi
- Shandong Key Laboratory of Precision Molecular Crop Design and Breeding, Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang 261325, China
| | - Yi Zhou
- Shandong Key Laboratory of Precision Molecular Crop Design and Breeding, Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang 261325, China
| | - Yi Zhang
- Shandong Key Laboratory of Precision Molecular Crop Design and Breeding, Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang 261325, China
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
- The Metabolomics Innovation Centre, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Li Guo
- Shandong Key Laboratory of Precision Molecular Crop Design and Breeding, Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang 261325, China
| |
Collapse
|
17
|
Amente LD, Mills NT, Le TD, Hyppönen E, Lee SH. Unraveling phenotypic variance in metabolic syndrome through multi-omics. Hum Genet 2024; 143:35-47. [PMID: 38095720 DOI: 10.1007/s00439-023-02619-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/18/2023] [Indexed: 01/19/2024]
Abstract
Complex multi-omics effects drive the clustering of cardiometabolic risk factors, underscoring the imperative to comprehend how individual and combined omics shape phenotypic variation. Our study partitions phenotypic variance in metabolic syndrome (MetS), blood glucose (GLU), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), and blood pressure through genome, transcriptome, metabolome, and exposome (i.e., lifestyle exposome) analyses. Our analysis included a cohort of 62,822 unrelated individuals with white British ancestry, sourced from the UK biobank. We employed linear mixed models to partition phenotypic variance using the restricted maximum likelihood (REML) method, implemented in MTG2 (v2.22). We initiated the analysis by individually modeling omics, followed by subsequent integration of pairwise omics in a joint model that also accounted for the covariance and interaction between omics layers. Finally, we estimated the correlations of various omics effects between the phenotypes using bivariate REML. Significant proportions of the MetS variance were attributed to distinct data sources: genome (9.47%), transcriptome (4.24%), metabolome (14.34%), and exposome (3.77%). The phenotypic variances explained by the genome, transcriptome, metabolome, and exposome ranged from 3.28% for GLU to 25.35% for HDL-C, 0% for GLU to 19.34% for HDL-C, 4.29% for systolic blood pressure (SBP) to 35.75% for TG, and 0.89% for GLU to 10.17% for HDL-C, respectively. Significant correlations were found between genomic and transcriptomic effects for TG and HDL-C. Furthermore, significant interaction effects between omics data were detected for both MetS and its components. Interestingly, significant correlation of omics effect between the phenotypes was found. This study underscores omics' roles, interaction effects, and random-effects covariance in unveiling phenotypic variation in multi-omics domains.
Collapse
Affiliation(s)
- Lamessa Dube Amente
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, 5000, Australia.
- UniSA Allied Health and Human Performance, University of South Australia, Adelaide, SA, 5000, Australia.
- South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia.
| | - Natalie T Mills
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Thuc Duy Le
- UniSA STEM, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Elina Hyppönen
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, 5000, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - S Hong Lee
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, 5000, Australia.
- UniSA Allied Health and Human Performance, University of South Australia, Adelaide, SA, 5000, Australia.
- South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia.
| |
Collapse
|
18
|
Mengelkoch S, Miryam Schüssler-Fiorenza Rose S, Lautman Z, Alley JC, Roos LG, Ehlert B, Moriarity DP, Lancaster S, Snyder MP, Slavich GM. Multi-omics approaches in psychoneuroimmunology and health research: Conceptual considerations and methodological recommendations. Brain Behav Immun 2023; 114:475-487. [PMID: 37543247 PMCID: PMC11195542 DOI: 10.1016/j.bbi.2023.07.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/04/2023] [Accepted: 07/30/2023] [Indexed: 08/07/2023] Open
Abstract
The field of psychoneuroimmunology (PNI) has grown substantially in both relevance and prominence over the past 40 years. Notwithstanding its impressive trajectory, a majority of PNI studies are still based on a relatively small number of analytes. To advance this work, we suggest that PNI, and health research in general, can benefit greatly from adopting a multi-omics approach, which involves integrating data across multiple biological levels (e.g., the genome, proteome, transcriptome, metabolome, lipidome, and microbiome/metagenome) to more comprehensively profile biological functions and relate these profiles to clinical and behavioral outcomes. To assist investigators in this endeavor, we provide an overview of multi-omics research, highlight recent landmark multi-omics studies investigating human health and disease risk, and discuss how multi-omics can be applied to better elucidate links between psychological, nervous system, and immune system activity. In doing so, we describe how to design high-quality multi-omics studies, decide which biological samples (e.g., blood, stool, urine, saliva, solid tissue) are most relevant, incorporate behavioral and wearable sensing data into multi-omics research, and understand key data quality, integration, analysis, and interpretation issues. PNI researchers are addressing some of the most interesting and important questions at the intersection of psychology, neuroscience, and immunology. Applying a multi-omics approach to this work will greatly expand the horizon of what is possible in PNI and has the potential to revolutionize our understanding of mind-body medicine.
Collapse
Affiliation(s)
- Summer Mengelkoch
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA.
| | | | - Ziv Lautman
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Jenna C Alley
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Lydia G Roos
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Benjamin Ehlert
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Daniel P Moriarity
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | | | | | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Kwon HJ, Park UH, Goh CJ, Park D, Lim YG, Lee IK, Do WJ, Lee KJ, Kim H, Yun SY, Joo J, Min NY, Lee S, Um SW, Lee MS. Enhancing Lung Cancer Classification through Integration of Liquid Biopsy Multi-Omics Data with Machine Learning Techniques. Cancers (Basel) 2023; 15:4556. [PMID: 37760525 PMCID: PMC10526503 DOI: 10.3390/cancers15184556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/30/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Early detection of lung cancer is crucial for patient survival and treatment. Recent advancements in next-generation sequencing (NGS) analysis enable cell-free DNA (cfDNA) liquid biopsy to detect changes, like chromosomal rearrangements, somatic mutations, and copy number variations (CNVs), in cancer. Machine learning (ML) analysis using cancer markers is a highly promising tool for identifying patterns and anomalies in cancers, making the development of ML-based analysis methods essential. We collected blood samples from 92 lung cancer patients and 80 healthy individuals to analyze the distinction between them. The detection of lung cancer markers Cyfra21 and carcinoembryonic antigen (CEA) in blood revealed significant differences between patients and controls. We performed machine learning analysis to obtain AUC values via Adaptive Boosting (AdaBoost), Multi-Layer Perceptron (MLP), and Logistic Regression (LR) using cancer markers, cfDNA concentrations, and CNV screening. Furthermore, combining the analysis of all multi-omics data for ML showed higher AUC values compared with analyzing each element separately, suggesting the potential for a highly accurate diagnosis of cancer. Overall, our results from ML analysis using multi-omics data obtained from blood demonstrate a remarkable ability of the model to distinguish between lung cancer and healthy individuals, highlighting the potential for a diagnostic model against lung cancer.
Collapse
Affiliation(s)
- Hyuk-Jung Kwon
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
- Department of Computer Science and Engineering, Incheon National University (INU), Incheon 22012, Republic of Korea
| | - Ui-Hyun Park
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
| | - Chul Jun Goh
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
| | - Dabin Park
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
| | - Yu Gyeong Lim
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
| | - Isaac Kise Lee
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
- Department of Computer Science and Engineering, Incheon National University (INU), Incheon 22012, Republic of Korea
- NGENI Foundation, San Diego, CA 92123, USA
| | - Woo-Jung Do
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
| | - Kyoung Joo Lee
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
| | - Hyojung Kim
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
| | - Seon-Young Yun
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
| | - Joungsu Joo
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
| | - Na Young Min
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
| | - Sunghoon Lee
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
| | - Sang-Won Um
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea;
| | - Min-Seob Lee
- Eone-Diagnomics Genome Center, Inc., 143, Gaetbeol-ro, Yeonsu-gu, Incheon 21999, Republic of Korea; (H.-J.K.); (U.-H.P.); (C.J.G.); (D.P.); (Y.G.L.); (I.K.L.); (W.-J.D.); (K.J.L.); (H.K.); (N.Y.M.)
- Diagnomics, Inc., 5795 Kearny Villa Rd., San Diego, CA 92123, USA
| |
Collapse
|
20
|
Sarfraz M, Arafat M, Zaidi SHH, Eltaib L, Siddique MI, Kamal M, Ali A, Asdaq SMB, Khan A, Aaghaz S, Alshammari MS, Imran M. Resveratrol-Laden Nano-Systems in the Cancer Environment: Views and Reviews. Cancers (Basel) 2023; 15:4499. [PMID: 37760469 PMCID: PMC10526844 DOI: 10.3390/cancers15184499] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The genesis of cancer is a precisely organized process in which normal cells undergo genetic alterations that cause the cells to multiply abnormally, colonize, and metastasize to other organs such as the liver, lungs, colon, and brain. Potential drugs that could modify these carcinogenic pathways are the ones that will be used in clinical trials as anti-cancer drugs. Resveratrol (RES) is a polyphenolic natural antitoxin that has been utilized for the treatment of several diseases, owing to its ability to scavenge free radicals, control the expression and activity of antioxidant enzymes, and have effects on inflammation, cancer, aging, diabetes, and cardioprotection. Although RES has a variety of pharmacological uses and shows promising applications in natural medicine, its unpredictable pharmacokinetics compromise its therapeutic efficacy and prevent its use in clinical settings. RES has been encapsulated into various nanocarriers, such as liposomes, polymeric nanoparticles, lipidic nanocarriers, and inorganic nanoparticles, to address these issues. These nanocarriers can modulate drug release, increase bioavailability, and reach therapeutically relevant plasma concentrations. Studies on resveratrol-rich nano-formulations in various cancer types are compiled in the current article. Studies relating to enhanced drug stability, increased therapeutic potential in terms of pharmacokinetics and pharmacodynamics, and reduced toxicity to cells and tissues are the main topics of this research. To keep the readers informed about the current state of resveratrol nano-formulations from an industrial perspective, some recent and significant patent literature has also been provided. Here, the prospects for nano-formulations are briefly discussed, along with machine learning and pharmacometrics methods for resolving resveratrol's pharmacokinetic concerns.
Collapse
Affiliation(s)
- Muhammad Sarfraz
- College of Pharmacy, Al Ain University, Al Ain Campus, Al Ain P.O. Box 64141, United Arab Emirates
| | - Mosab Arafat
- College of Pharmacy, Al Ain University, Al Ain Campus, Al Ain P.O. Box 64141, United Arab Emirates
| | - Syeda Huma H. Zaidi
- Department of Chemistry, Faculty of Science, Northern Border University, Arar 91431, Saudi Arabia
| | - Lina Eltaib
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Muhammad Irfan Siddique
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Abuzer Ali
- Department of Pharmacognosy, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | | | - Abida Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia (M.I.)
| | - Shams Aaghaz
- Department of Pharmacy, School of Medical & Allied Sciences, Galgotias University, Greater Noida 203201, India
| | - Mohammed Sanad Alshammari
- Department of Computer Science, Faculty of Computing and Information Technology, Northern Border University, Rafha 91911, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia (M.I.)
| |
Collapse
|
21
|
Guardado M, Steurer M, Chapin C, Hernandez RD, Ballard PL, Torgerson D. The Urinary Metabolomic Fingerprint in Extremely Preterm Infants on Total Parenteral Nutrition vs. Enteral Feeds. Metabolites 2023; 13:971. [PMID: 37755251 PMCID: PMC10537655 DOI: 10.3390/metabo13090971] [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: 07/28/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023] Open
Abstract
Total Parenteral Nutrition (TPN), which uses intravenous administration of nutrients, minerals and vitamins, is essential for sustaining premature infants until they transition to enteral feeds, but there is limited information on metabolomic differences between infants on TPN and enteral feeds. We performed untargeted global metabolomics on urine samples collected between 23-30 days of life from 314 infants born <29 weeks gestational age from the TOLSURF and PROP cohorts. Principal component analysis across all metabolites showed a separation of infants solely on TPN compared to infants who had transitioned to enteral feeds, indicating global metabolomic differences between infants based on feeding status. Among 913 metabolites that passed quality control filters, 609 varied in abundance between infants on TPN vs. enteral feeds at p < 0.05. Of these, 88% were in the direction of higher abundance in the urine of infants on enteral feeds. In a subset of infants in a longitudinal analysis, both concurrent and delayed changes in metabolite levels were observed with the initiation of enteral feeds. These infants had higher concentrations of essential amino acids, lipids, and vitamins, which are necessary for growth and development, suggesting the nutritional benefit of an enteral feeding regimen.
Collapse
Affiliation(s)
- Miguel Guardado
- Biological and Medical Informatics Graduate Program, School of Medicine, Mission Bay Campus, University of California, San Francisco, CA 94134, USA
- Department of Epidemiology and Biostatistics, School of Medicine, Mission Bay Campus, University of California, San Francisco, CA 94158, USA;
- Department of Bioengineering and Therapeutic Sciences, School of Medicine, Mission Bay Campus, University of California, San Francisco, CA 94134, USA;
| | - Martina Steurer
- Department of Pediatrics, School of Medicine, Mission Bay & Parnassus Campuses, University of California, San Francisco, CA 94158, USA; (M.S.); (C.C.); (P.L.B.)
| | - Cheryl Chapin
- Department of Pediatrics, School of Medicine, Mission Bay & Parnassus Campuses, University of California, San Francisco, CA 94158, USA; (M.S.); (C.C.); (P.L.B.)
| | - Ryan D. Hernandez
- Department of Bioengineering and Therapeutic Sciences, School of Medicine, Mission Bay Campus, University of California, San Francisco, CA 94134, USA;
| | - Philip L. Ballard
- Department of Pediatrics, School of Medicine, Mission Bay & Parnassus Campuses, University of California, San Francisco, CA 94158, USA; (M.S.); (C.C.); (P.L.B.)
| | - Dara Torgerson
- Department of Epidemiology and Biostatistics, School of Medicine, Mission Bay Campus, University of California, San Francisco, CA 94158, USA;
| |
Collapse
|
22
|
Sinha K, Ghosh N, Sil PC. A Review on the Recent Applications of Deep Learning in Predictive Drug Toxicological Studies. Chem Res Toxicol 2023; 36:1174-1205. [PMID: 37561655 DOI: 10.1021/acs.chemrestox.2c00375] [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: 08/12/2023]
Abstract
Drug toxicity prediction is an important step in ensuring patient safety during drug design studies. While traditional preclinical studies have historically relied on animal models to evaluate toxicity, recent advances in deep-learning approaches have shown great promise in advancing drug safety science and reducing animal use in preclinical studies. However, deep-learning-based approaches also face challenges in handling large biological data sets, model interpretability, and regulatory acceptance. In this review, we provide an overview of recent developments in deep-learning-based approaches for predicting drug toxicity, highlighting their potential advantages over traditional methods and the need to address their limitations. Deep-learning models have demonstrated excellent performance in predicting toxicity outcomes from various data sources such as chemical structures, genomic data, and high-throughput screening assays. The potential of deep learning for automated feature engineering is also discussed. This review emphasizes the need to address ethical concerns related to the use of deep learning in drug toxicity studies, including the reduction of animal use and ensuring regulatory acceptance. Furthermore, emerging applications of deep learning in drug toxicity prediction, such as predicting drug-drug interactions and toxicity in rare subpopulations, are highlighted. The integration of deep-learning-based approaches with traditional methods is discussed as a way to develop more reliable and efficient predictive models for drug safety assessment, paving the way for safer and more effective drug discovery and development. Overall, this review highlights the critical role of deep learning in predictive toxicology and drug safety evaluation, emphasizing the need for continued research and development in this rapidly evolving field. By addressing the limitations of traditional methods, leveraging the potential of deep learning for automated feature engineering, and addressing ethical concerns, deep-learning-based approaches have the potential to revolutionize drug toxicity prediction and improve patient safety in drug discovery and development.
Collapse
Affiliation(s)
- Krishnendu Sinha
- Department of Zoology, Jhargram Raj College, Jhargram 721507, West Bengal, India
| | - Nabanita Ghosh
- Department of Zoology, Maulana Azad College, Kolkata 700013, West Bengal, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, West Bengal, India
| |
Collapse
|
23
|
Oh SW, Imran M, Kim EH, Park SY, Lee SG, Park HM, Jung JW, Ryu TH. Approach strategies and application of metabolomics to biotechnology in plants. FRONTIERS IN PLANT SCIENCE 2023; 14:1192235. [PMID: 37636096 PMCID: PMC10451086 DOI: 10.3389/fpls.2023.1192235] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023]
Abstract
Metabolomics refers to the technology for the comprehensive analysis of metabolites and low-molecular-weight compounds in a biological system, such as cells or tissues. Metabolites play an important role in biological phenomena through their direct involvement in the regulation of physiological mechanisms, such as maintaining cell homeostasis or signal transmission through protein-protein interactions. The current review aims provide a framework for how the integrated analysis of metabolites, their functional actions and inherent biological information can be used to understand biological phenomena related to the regulation of metabolites and how this information can be applied to safety assessments of crops created using biotechnology. Advancement in technology and analytical instrumentation have led new ways to examine the convergence between biology and chemistry, which has yielded a deeper understanding of complex biological phenomena. Metabolomics can be utilized and applied to safety assessments of biotechnology products through a systematic approach using metabolite-level data processing algorithms, statistical techniques, and database development. The integration of metabolomics data with sequencing data is a key step towards improving additional phenotypical evidence to elucidate the degree of environmental affects for variants found in genome associated with metabolic processes. Moreover, information analysis technology such as big data, machine learning, and IT investment must be introduced to establish a system for data extraction, selection, and metabolomic data analysis for the interpretation of biological implications of biotechnology innovations. This review outlines the integrity of metabolomics assessments in determining the consequences of genetic engineering and biotechnology in plants.
Collapse
|
24
|
Wehkamp K, Krawczak M, Schreiber S. The Quality and Utility of Artificial Intelligence in Patient Care. DEUTSCHES ARZTEBLATT INTERNATIONAL 2023; 120:463-469. [PMID: 37218054 PMCID: PMC10487679 DOI: 10.3238/arztebl.m2023.0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 11/30/2022] [Accepted: 05/08/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND Artificial intelligence (AI) is increasingly being used in patient care. In the future, physicians will need to understand not only the basic functioning of AI applications, but also their quality, utility, and risks. METHODS This article is based on a selective review of the literature on the principles, quality, limitations, and benefits AI applications in patient care, along with examples of individual applications. RESULTS The number of AI applications in patient care is rising, with more than 500 approvals in the United States to date. Their quality and utility are based on a number of interdependent factors, including the real-life setting, the type and amount of data collected, the choice of variables used by the application, the algorithms used, and the goal and implementation of each application. Bias (which may be hidden) and errors can arise at all these levels. Any evaluation of the quality and utility of an AI application must, therefore, be conducted according to the scientific principles of evidence-based medicine-a requirement that is often hampered by a lack of transparency. CONCLUSION AI has the potential to improve patient care while meeting the challenge of dealing with an ever-increasing surfeit of information and data in medicine with limited human resources. The limitations and risks of AI applications require critical and responsible consideration. This can best be achieved through a combination of scientific.
Collapse
Affiliation(s)
- Kai Wehkamp
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Campus Lübeck, Kiel, Germany
- Department for Medical Management, MSH Medical School Hamburg, Hamburg, Germany
| | - Michael Krawczak
- Institute of Medical Informatics and Statistics, Christian-Albrechts-University of Kiel, University Medical Center Schleswig-Holstein Campus Kiel, Germany
| | - Stefan Schreiber
- Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Campus Lübeck, Kiel, Germany
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, University Medical Center Schleswig-Holstein Campus Kiel, Germany
| |
Collapse
|
25
|
Ebbels TMD, van der Hooft JJJ, Chatelaine H, Broeckling C, Zamboni N, Hassoun S, Mathé EA. Recent advances in mass spectrometry-based computational metabolomics. Curr Opin Chem Biol 2023; 74:102288. [PMID: 36966702 PMCID: PMC11075003 DOI: 10.1016/j.cbpa.2023.102288] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 04/03/2023]
Abstract
The computational metabolomics field brings together computer scientists, bioinformaticians, chemists, clinicians, and biologists to maximize the impact of metabolomics across a wide array of scientific and medical disciplines. The field continues to expand as modern instrumentation produces datasets with increasing complexity, resolution, and sensitivity. These datasets must be processed, annotated, modeled, and interpreted to enable biological insight. Techniques for visualization, integration (within or between omics), and interpretation of metabolomics data have evolved along with innovation in the databases and knowledge resources required to aid understanding. In this review, we highlight recent advances in the field and reflect on opportunities and innovations in response to the most pressing challenges. This review was compiled from discussions from the 2022 Dagstuhl seminar entitled "Computational Metabolomics: From Spectra to Knowledge".
Collapse
Affiliation(s)
- Timothy M D Ebbels
- Section of Bioinformatics, Department of Metabolism, Digestion & Reproduction, Imperial College London, Burlington Danes Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK.
| | - Justin J J van der Hooft
- Bioinformatics Group, Wageningen University & Research, Wageningen 6708 PB, the Netherlands; Department of Biochemistry, University of Johannesburg, Auckland Park, Johannesburg 2006, South Africa
| | - Haley Chatelaine
- Informatics Core, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Corey Broeckling
- Bioanalysis and Omics Center, Analytical Resources Core, Colorado State University, Fort Collins, CO, USA
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Soha Hassoun
- Department of Computer Science, Tufts University, Medford, MA, USA; Department of Chemical and Biological Engineering, Tufts University, Medford, MA, USA
| | - Ewy A Mathé
- Informatics Core, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, USA.
| |
Collapse
|
26
|
Scott-Fordsmand JJ, Amorim MJB. Using Machine Learning to make nanomaterials sustainable. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 859:160303. [PMID: 36410486 DOI: 10.1016/j.scitotenv.2022.160303] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 11/06/2022] [Accepted: 11/15/2022] [Indexed: 06/16/2023]
Abstract
Sustainable development is a key challenge for contemporary human societies; failure to achieve sustainability could threaten human survival. In this review article, we illustrate how Machine Learning (ML) could support more sustainable development, covering the basics of data gathering through each step of the Environmental Risk Assessment (ERA). The literature provides several examples showing how ML can be employed in most steps of a typical ERA.A key observation is that there are currently no clear guidance for using such autonomous technologies in ERAs or which standards/checks are required. Steering thus seems to be the most important task for supporting the use of ML in the ERA of nano- and smart-materials. Resources should be devoted to developing a strategy for implementing ML in ERA with a strong emphasis on data foundations, methodologies, and the related sensitivities/uncertainties. We should recognise historical errors and biases (e.g., in data) to avoid embedding them during ML programming.
Collapse
Affiliation(s)
| | - Mónica J B Amorim
- Department of Biology & CESAM, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
27
|
Mohammed MA, Abdulkareem KH, Dinar AM, Zapirain BG. Rise of Deep Learning Clinical Applications and Challenges in Omics Data: A Systematic Review. Diagnostics (Basel) 2023; 13:diagnostics13040664. [PMID: 36832152 PMCID: PMC9955380 DOI: 10.3390/diagnostics13040664] [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: 12/24/2022] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
This research aims to review and evaluate the most relevant scientific studies about deep learning (DL) models in the omics field. It also aims to realize the potential of DL techniques in omics data analysis fully by demonstrating this potential and identifying the key challenges that must be addressed. Numerous elements are essential for comprehending numerous studies by surveying the existing literature. For example, the clinical applications and datasets from the literature are essential elements. The published literature highlights the difficulties encountered by other researchers. In addition to looking for other studies, such as guidelines, comparative studies, and review papers, a systematic approach is used to search all relevant publications on omics and DL using different keyword variants. From 2018 to 2022, the search procedure was conducted on four Internet search engines: IEEE Xplore, Web of Science, ScienceDirect, and PubMed. These indexes were chosen because they offer enough coverage and linkages to numerous papers in the biological field. A total of 65 articles were added to the final list. The inclusion and exclusion criteria were specified. Of the 65 publications, 42 are clinical applications of DL in omics data. Furthermore, 16 out of 65 articles comprised the review publications based on single- and multi-omics data from the proposed taxonomy. Finally, only a small number of articles (7/65) were included in papers focusing on comparative analysis and guidelines. The use of DL in studying omics data presented several obstacles related to DL itself, preprocessing procedures, datasets, model validation, and testbed applications. Numerous relevant investigations were performed to address these issues. Unlike other review papers, our study distinctly reflects different observations on omics with DL model areas. We believe that the result of this study can be a useful guideline for practitioners who look for a comprehensive view of the role of DL in omics data analysis.
Collapse
Affiliation(s)
- Mazin Abed Mohammed
- College of Computer Science and Information Technology, University of Anbar, Anbar 31001, Iraq
- eVIDA Lab, University of Deusto, 48007 Bilbao, Spain
- Correspondence: (M.A.M.); (B.G.Z.)
| | - Karrar Hameed Abdulkareem
- College of Agriculture, Al-Muthanna University, Samawah 66001, Iraq
- College of Engineering, University of Warith Al-Anbiyaa, Karbala 56001, Iraq
| | - Ahmed M. Dinar
- Computer Engineering Department, University of Technology- Iraq, Baghdad 19006, Iraq
| | | |
Collapse
|
28
|
Eicher T, Spencer KD, Siddiqui JK, Machiraju R, Mathé EA. IntLIM 2.0: identifying multi-omic relationships dependent on discrete or continuous phenotypic measurements. BIOINFORMATICS ADVANCES 2023; 3:vbad009. [PMID: 36922980 PMCID: PMC10010601 DOI: 10.1093/bioadv/vbad009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/01/2023] [Indexed: 02/04/2023]
Abstract
Motivation IntLIM uncovers phenotype-dependent linear associations between two types of analytes (e.g. genes and metabolites) in a multi-omic dataset, which may reflect chemically or biologically relevant relationships. Results The new IntLIM R package includes newly added support for generalized data types, covariate correction, continuous phenotypic measurements, model validation and unit testing. IntLIM analysis uncovered biologically relevant gene-metabolite associations in two separate datasets, and the run time is improved over baseline R functions by multiple orders of magnitude. Availability and implementation IntLIM is available as an R package with a detailed vignette (https://github.com/ncats/IntLIM) and as an R Shiny app (see Supplementary Figs S1-S6) (https://intlim.ncats.io/). Supplementary information Supplementary data are available at Bioinformatics Advances online.
Collapse
Affiliation(s)
- Tara Eicher
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD 20892, USA.,Department of Computer Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Kyle D Spencer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD 20892, USA.,Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Jalal K Siddiqui
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Raghu Machiraju
- Department of Computer Science and Engineering, The Ohio State University, Columbus, OH 43210, USA.,Biomedical Informatics Department, The Ohio State University, Columbus, OH 43210, USA.,Department of Pathology, The Ohio State University, Columbus, OH 43210, USA
| | - Ewy A Mathé
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD 20892, USA
| |
Collapse
|
29
|
Braisted J, Patt A, Tindall C, Sheils T, Neyra J, Spencer K, Eicher T, Mathé EA. RaMP-DB 2.0: a renovated knowledgebase for deriving biological and chemical insight from metabolites, proteins, and genes. Bioinformatics 2023; 39:6827287. [PMID: 36373969 PMCID: PMC9825745 DOI: 10.1093/bioinformatics/btac726] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/19/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
MOTIVATION Functional interpretation of high-throughput metabolomic and transcriptomic results is a crucial step in generating insight from experimental data. However, pathway and functional information for genes and metabolites are distributed among many siloed resources, limiting the scope of analyses that rely on a single knowledge source. RESULTS RaMP-DB 2.0 is a web interface, relational database, API and R package designed for straightforward and comprehensive functional interpretation of metabolomic and multi-omic data. RaMP-DB 2.0 has been upgraded with an expanded breadth and depth of functional and chemical annotations (ClassyFire, LIPID MAPS, SMILES, InChIs, etc.), with new data types related to metabolites and lipids incorporated. To streamline entity resolution across multiple source databases, we have implemented a new semi-automated process, thereby lessening the burden of harmonization and supporting more frequent updates. The associated RaMP-DB 2.0 R package now supports queries on pathways, common reactions (e.g. metabolite-enzyme relationship), chemical functional ontologies, chemical classes and chemical structures, as well as enrichment analyses on pathways (multi-omic) and chemical classes. Lastly, the RaMP-DB web interface has been completely redesigned using the Angular framework. AVAILABILITY AND IMPLEMENTATION The code used to build all components of RaMP-DB 2.0 are freely available on GitHub at https://github.com/ncats/ramp-db, https://github.com/ncats/RaMP-Client/ and https://github.com/ncats/RaMP-Backend. The RaMP-DB web application can be accessed at https://rampdb.nih.gov/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | | | - Cole Tindall
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD 20850, USA
| | - Timothy Sheils
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD 20850, USA
| | | | - Kyle Spencer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD 20850, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Tara Eicher
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD 20850, USA
- Department of Computer Science and Engineering, The Ohio State University, Columbus OH 43210, USA
| | | |
Collapse
|
30
|
Deng P, Liang H, Wang S, Hao R, Han J, Sun X, Pan X, Li D, Wu Y, Huang Z, Xue J, Chen Z. Combined metabolomics and network pharmacology to elucidate the mechanisms of Dracorhodin Perchlorate in treating diabetic foot ulcer rats. Front Pharmacol 2022; 13:1038656. [PMID: 36532755 PMCID: PMC9752146 DOI: 10.3389/fphar.2022.1038656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/31/2022] [Indexed: 10/10/2023] Open
Abstract
Background: Diabetic foot ulcer (DFU) is a severe chronic complication of diabetes, that can result in disability or death. Dracorhodin Perchlorate (DP) is effective for treating DFU, but the potential mechanisms need to be investigated. We aimed to explore the mechanisms underlying the acceleration of wound healing in DFU by the topical application of DP through the combination of metabolomics and network pharmacology. Methods: A DFU rat model was established, and the rate of ulcer wound healing was assessed. Different metabolites were found in the skin tissues of each group, and MetaboAnalyst was performed to analyse metabolic pathways. The candidate targets of DP in the treatment of DFU were screened using network pharmacology. Cytoscape was applied to construct an integrated network of metabolomics and network pharmacology. Moreover, the obtained hub targets were validated using molecular docking. After the topical application of DP, blood glucose, the rate of wound healing and pro-inflammatory cytokine levels were assessed. Results: The levels of IL-1, hs-CRP and TNF-α of the Adm group were significantly downregulated. A total of 114 metabolites were identified. These could be important to the therapeutic effects of DP in the treatment of DFU. Based on the network pharmacology, seven hub genes were found, which were partially consistent with the metabolomics results. We focused on four hub targets by further integrated analysis, namely, PAH, GSTM1, DHFR and CAT, and the crucial metabolites and pathways. Molecular docking results demonstrated that DP was well combined with the hub targets. Conclusion: Our research based on metabolomics and network pharmacology demonstrated that DP improves wound healing in DFU through multiple targets and pathways, and it can potentially be used for DFU treatment.
Collapse
Affiliation(s)
- Pin Deng
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Huan Liang
- Department of Orthopedics, Beijing Longfu Hospital, Beijing, China
| | - Shulong Wang
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Ruinan Hao
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
| | - Jinglu Han
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Xiaojie Sun
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Xuyue Pan
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Dongxiao Li
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Yinwen Wu
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
| | - Zhichao Huang
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Jiajia Xue
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
| | - Zhaojun Chen
- Department of Hand and Foot Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| |
Collapse
|
31
|
Maghsoudi Z, Nguyen H, Tavakkoli A, Nguyen T. A comprehensive survey of the approaches for pathway analysis using multi-omics data integration. Brief Bioinform 2022; 23:6761962. [PMID: 36252928 PMCID: PMC9677478 DOI: 10.1093/bib/bbac435] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/26/2022] [Accepted: 09/08/2022] [Indexed: 02/07/2023] Open
Abstract
Pathway analysis has been widely used to detect pathways and functions associated with complex disease phenotypes. The proliferation of this approach is due to better interpretability of its results and its higher statistical power compared with the gene-level statistics. A plethora of pathway analysis methods that utilize multi-omics setup, rather than just transcriptomics or proteomics, have recently been developed to discover novel pathways and biomarkers. Since multi-omics gives multiple views into the same problem, different approaches are employed in aggregating these views into a comprehensive biological context. As a result, a variety of novel hypotheses regarding disease ideation and treatment targets can be formulated. In this article, we review 32 such pathway analysis methods developed for multi-omics and multi-cohort data. We discuss their availability and implementation, assumptions, supported omics types and databases, pathway analysis techniques and integration strategies. A comprehensive assessment of each method's practicality, and a thorough discussion of the strengths and drawbacks of each technique will be provided. The main objective of this survey is to provide a thorough examination of existing methods to assist potential users and researchers in selecting suitable tools for their data and analysis purposes, while highlighting outstanding challenges in the field that remain to be addressed for future development.
Collapse
Affiliation(s)
- Zeynab Maghsoudi
- Department of Computer Science and Engineering, University of Nevada, Reno, 89557, Nevada, USA
| | - Ha Nguyen
- Department of Computer Science and Engineering, University of Nevada, Reno, 89557, Nevada, USA
| | - Alireza Tavakkoli
- Department of Computer Science and Engineering, University of Nevada, Reno, 89557, Nevada, USA
| | - Tin Nguyen
- Corresponding author: Tin Nguyen, Department of Computer Science and Engineering, University of Nevada, Reno, NV, USA. Tel.: +1-775-784-6619;
| |
Collapse
|
32
|
Innovative Application of Metabolomics on Bioactive Ingredients of Foods. Foods 2022; 11:foods11192974. [PMID: 36230049 PMCID: PMC9562173 DOI: 10.3390/foods11192974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/12/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Metabolomics, as a new omics technology, has been widely accepted by researchers and has shown great potential in the field of nutrition and health in recent years. This review briefly introduces the process of metabolomics analysis, including sample preparation and extraction, derivatization, separation and detection, and data processing. This paper focuses on the application of metabolomics in food-derived bioactive ingredients. For example, metabolomics techniques are used to analyze metabolites in food to find bioactive substances or new metabolites in food materials. Moreover, bioactive substances have been tested in vitro and in vivo, as well as in humans, to investigate the changes of metabolites and the underlying metabolic pathways, among which metabolomics is used to find potential biomarkers and targets. Metabolomics provides a new approach for the prevention and regulation of chronic diseases and the study of the underlying mechanisms. It also provides strong support for the development of functional food or drugs. Although metabolomics has some limitations such as low sensitivity, poor repeatability, and limited detection range, it is developing rapidly in general, and also in the field of nutrition and health. At the end of this paper, we put forward our own insights on the development prospects of metabolomics in the application of bioactive ingredients in food.
Collapse
|
33
|
Robin V, Bodein A, Scott-Boyer MP, Leclercq M, Périn O, Droit A. Overview of methods for characterization and visualization of a protein-protein interaction network in a multi-omics integration context. Front Mol Biosci 2022; 9:962799. [PMID: 36158572 PMCID: PMC9494275 DOI: 10.3389/fmolb.2022.962799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/16/2022] [Indexed: 11/26/2022] Open
Abstract
At the heart of the cellular machinery through the regulation of cellular functions, protein-protein interactions (PPIs) have a significant role. PPIs can be analyzed with network approaches. Construction of a PPI network requires prediction of the interactions. All PPIs form a network. Different biases such as lack of data, recurrence of information, and false interactions make the network unstable. Integrated strategies allow solving these different challenges. These approaches have shown encouraging results for the understanding of molecular mechanisms, drug action mechanisms, and identification of target genes. In order to give more importance to an interaction, it is evaluated by different confidence scores. These scores allow the filtration of the network and thus facilitate the representation of the network, essential steps to the identification and understanding of molecular mechanisms. In this review, we will discuss the main computational methods for predicting PPI, including ones confirming an interaction as well as the integration of PPIs into a network, and we will discuss visualization of these complex data.
Collapse
Affiliation(s)
- Vivian Robin
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Antoine Bodein
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Marie-Pier Scott-Boyer
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Mickaël Leclercq
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Olivier Périn
- Digital Sciences Department, L'Oréal Advanced Research, Aulnay-sous-bois, France
| | - Arnaud Droit
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| |
Collapse
|
34
|
The Future of Biomarkers in Veterinary Medicine: Emerging Approaches and Associated Challenges. Animals (Basel) 2022; 12:ani12172194. [PMID: 36077913 PMCID: PMC9454634 DOI: 10.3390/ani12172194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary In this review we seek to outline the role of new technologies in biomarker discovery, particularly within the veterinary field and with an emphasis on ‘omics’, as well as to examine why many biomarkers-despite much excitement-have not yet made it to clinical practice. Further we emphasise the critical need for close collaboration between clinicians, researchers and funding bodies and the need to set clear goals for biomarker requirements and realistic application in the clinical setting, ensuring that biomarker type, method of detection and clinical utility are compatible, and adequate funding, time and sample size are available for all phases of development. Abstract New biomarkers promise to transform veterinary practice through rapid diagnosis of diseases, effective monitoring of animal health and improved welfare and production efficiency. However, the road from biomarker discovery to translation is not always straightforward. This review focuses on molecular biomarkers under development in the veterinary field, introduces the emerging technological approaches transforming this space and the role of ‘omics platforms in novel biomarker discovery. The vast majority of veterinary biomarkers are at preliminary stages of development and not yet ready to be deployed into clinical translation. Hence, we examine the major challenges encountered in the process of biomarker development from discovery, through validation and translation to clinical practice, including the hurdles specific to veterinary practice and to each of the ‘omics platforms–transcriptomics, proteomics, lipidomics and metabolomics. Finally, recommendations are made for the planning and execution of biomarker studies with a view to assisting the success of novel biomarkers in reaching their full potential.
Collapse
|
35
|
Abstract
"The Primate Malarias" book has been a uniquely important resource for multiple generations of scientists, since its debut in 1971, and remains pertinent to the present day. Indeed, nonhuman primates (NHPs) have been instrumental for major breakthroughs in basic and pre-clinical research on malaria for over 50 years. Research involving NHPs have provided critical insights and data that have been essential for malaria research on many parasite species, drugs, vaccines, pathogenesis, and transmission, leading to improved clinical care and advancing research goals for malaria control, elimination, and eradication. Whilst most malaria scientists over the decades have been studying Plasmodium falciparum, with NHP infections, in clinical studies with humans, or using in vitro culture or rodent model systems, others have been dedicated to advancing research on Plasmodium vivax, as well as on phylogenetically related simian species, including Plasmodium cynomolgi, Plasmodium coatneyi, and Plasmodium knowlesi. In-depth study of these four phylogenetically related species over the years has spawned the design of NHP longitudinal infection strategies for gathering information about ongoing infections, which can be related to human infections. These Plasmodium-NHP infection model systems are reviewed here, with emphasis on modern systems biological approaches to studying longitudinal infections, pathogenesis, immunity, and vaccines. Recent discoveries capitalizing on NHP longitudinal infections include an advanced understanding of chronic infections, relapses, anaemia, and immune memory. With quickly emerging new technological advances, more in-depth research and mechanistic discoveries can be anticipated on these and additional critical topics, including hypnozoite biology, antigenic variation, gametocyte transmission, bone marrow dysfunction, and loss of uninfected RBCs. New strategies and insights published by the Malaria Host-Pathogen Interaction Center (MaHPIC) are recapped here along with a vision that stresses the importance of educating future experts well trained in utilizing NHP infection model systems for the pursuit of innovative, effective interventions against malaria.
Collapse
Affiliation(s)
- Mary R Galinski
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Emory Vaccine Center, Emory University, Atlanta, GA, USA.
- Emory National Primate Research Center (Yerkes National Primate Research Center), Emory University, Atlanta, GA, USA.
| |
Collapse
|
36
|
Yu CT, Chao BN, Barajas R, Haznadar M, Maruvada P, Nicastro HL, Ross SA, Verma M, Rogers S, Zanetti KA. An evaluation of the National Institutes of Health grants portfolio: identifying opportunities and challenges for multi-omics research that leverage metabolomics data. Metabolomics 2022; 18:29. [PMID: 35488937 PMCID: PMC9056487 DOI: 10.1007/s11306-022-01878-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 02/28/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Through the systematic large-scale profiling of metabolites, metabolomics provides a tool for biomarker discovery and improving disease monitoring, diagnosis, prognosis, and treatment response, as well as for delineating disease mechanisms and etiology. As a downstream product of the genome and epigenome, transcriptome, and proteome activity, the metabolome can be considered as being the most proximal correlate to the phenotype. Integration of metabolomics data with other -omics data in multi-omics analyses has the potential to advance understanding of human disease development and treatment. AIM OF REVIEW To understand the current funding and potential research opportunities for when metabolomics is used in human multi-omics studies, we cross-sectionally evaluated National Institutes of Health (NIH)-funded grants to examine the use of metabolomics data when collected with at least one other -omics data type. First, we aimed to determine what types of multi-omics studies included metabolomics data collection. Then, we looked at those multi-omics studies to examine how often grants employed an integrative analysis approach using metabolomics data. KEY SCIENTIFIC CONCEPTS OF REVIEW We observed that the majority of NIH-funded multi-omics studies that include metabolomics data performed integration, but to a limited extent, with integration primarily incorporating only one other -omics data type. Some opportunities to improve data integration may include increasing confidence in metabolite identification, as well as addressing variability between -omics approach requirements and -omics data incompatibility.
Collapse
Affiliation(s)
- Catherine T Yu
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Brittany N Chao
- Office of Workforce Planning and Development, National Cancer Institute, Rockville, MD, USA
| | - Rolando Barajas
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Majda Haznadar
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Rockville, MD, USA
| | - Padma Maruvada
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Holly L Nicastro
- Office of Nutrition Research, National Institutes of Health, Bethesda, MD, USA
| | - Sharon A Ross
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA
| | - Mukesh Verma
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Scott Rogers
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Krista A Zanetti
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA.
| |
Collapse
|
37
|
Yan P, Wei Y, Wang M, Tao J, Ouyang H, Du Z, Li S, Jiang H. Network pharmacology combined with metabolomics and lipidomics to reveal the hypolipidemic mechanism of Alismatis rhizoma in hyperlipidemic mice. Food Funct 2022; 13:4714-4733. [PMID: 35383784 DOI: 10.1039/d1fo04386b] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Alismatis rhizoma (AR), the dried rhizome of Alisma orientale (Sam) Juzep, is effective in treating hyperlipidemia, but the mechanisms involved require further exploration. This study evaluated the hypolipidemic properties of AR using an integrated strategy combining network pharmacology with metabolomics and lipidomics. Firstly, a hyperlipidemia mouse model induced by a high-fat diet was established to evaluate the therapeutic effects of AR. Secondly, plasma metabolomics and lipidomics were used to identify differential metabolites and lipids, and metabolic pathway analysis was performed using MetaboAnalyst. Thirdly, network pharmacology, based on the metabolic profile of AR in vivo, was used to discover potential therapeutic targets. Finally, key targets were obtained through a compound-target-metabolite network, which was verified by molecular docking and quantitative real-time PCR (qPCR). Biochemistry analysis and histological examinations showed that AR exerted hypolipidemic effects on hyperlipidemic mice. Seventy potential biomarkers for the AR treatment of hyperlipidemia were identified by metabolomics and lipidomics, which were mainly involved in lipid metabolism, energy metabolism and amino acid metabolism. Eighteen potentially active compounds were identified in the plasma of mice after oral administration of AR, which were associated with 83 potential therapeutic targets. The PPAR signaling pathway was considered a crucial signaling pathway of AR against hyperlipidemia by KEGG analysis. The joint analysis showed that 6 upstream key targets were regulated by AR, including ALB, TNF, IL1B, MMP9, PPARA and PPARG. Molecular docking showed that active compounds of AR had high binding affinity with these key targets. qPCR further demonstrated that AR could reverse the mRNA expression of these key targets in hyperlipidemic mice. This study integrates network pharmacology with metabolomics and lipidomics to reveal the regulatory effects of AR on endogenous metabolites and validates key therapeutic targets, and represents the most systematic and in-depth study on the hypolipidemic activity of AR.
Collapse
Affiliation(s)
- Pan Yan
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yinyu Wei
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Meiqin Wang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Jianmei Tao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hui Ouyang
- Jiangxi University of Traditional Chinese Medicine, Nanchang 330000, China
| | - Zhifeng Du
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Sen Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hongliang Jiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
38
|
Li X, Ma J, Leng L, Han M, Li M, He F, Zhu Y. MoGCN: A Multi-Omics Integration Method Based on Graph Convolutional Network for Cancer Subtype Analysis. Front Genet 2022; 13:806842. [PMID: 35186034 PMCID: PMC8847688 DOI: 10.3389/fgene.2022.806842] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/14/2022] [Indexed: 12/17/2022] Open
Abstract
In light of the rapid accumulation of large-scale omics datasets, numerous studies have attempted to characterize the molecular and clinical features of cancers from a multi-omics perspective. However, there are great challenges in integrating multi-omics using machine learning methods for cancer subtype classification. In this study, MoGCN, a multi-omics integration model based on graph convolutional network (GCN) was developed for cancer subtype classification and analysis. Genomics, transcriptomics and proteomics datasets for 511 breast invasive carcinoma (BRCA) samples were downloaded from the Cancer Genome Atlas (TCGA). The autoencoder (AE) and the similarity network fusion (SNF) methods were used to reduce dimensionality and construct the patient similarity network (PSN), respectively. Then the vector features and the PSN were input into the GCN for training and testing. Feature extraction and network visualization were used for further biological knowledge discovery and subtype classification. In the analysis of multi-dimensional omics data of the BRCA samples in TCGA, MoGCN achieved the highest accuracy in cancer subtype classification compared with several popular algorithms. Moreover, MoGCN can extract the most significant features of each omics layer and provide candidate functional molecules for further analysis of their biological effects. And network visualization showed that MoGCN could make clinically intuitive diagnosis. The generality of MoGCN was proven on the TCGA pan-kidney cancer datasets. MoGCN and datasets are public available at https://github.com/Lifoof/MoGCN. Our study shows that MoGCN performs well for heterogeneous data integration and the interpretability of classification results, which confers great potential for applications in biomarker identification and clinical diagnosis.
Collapse
Affiliation(s)
- Xiao Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Jie Ma
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Ling Leng
- Stem Cell and Regenerative Medicine Lab, Department of Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Translational Medicine Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Mingfei Han
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Mansheng Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Yunping Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| |
Collapse
|
39
|
Benevenuto RF, Venter HJ, Zanatta CB, Nodari RO, Agapito-Tenfen SZ. Alterations in genetically modified crops assessed by omics studies: Systematic review and meta-analysis. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2022.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
40
|
Debik J, Sangermani M, Wang F, Madssen TS, Giskeødegård GF. Multivariate analysis of NMR-based metabolomic data. NMR IN BIOMEDICINE 2022; 35:e4638. [PMID: 34738674 DOI: 10.1002/nbm.4638] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/08/2021] [Accepted: 09/29/2021] [Indexed: 06/13/2023]
Abstract
Nuclear magnetic resonance (NMR) spectroscopy allows for simultaneous detection of a wide range of metabolites and lipids. As metabolites act together in complex metabolic networks, they are often highly correlated, and optimal biological insight is achieved when using methods that take the correlation into account. For this reason, latent-variable-based methods, such as principal component analysis and partial least-squares discriminant analysis, are widely used in metabolomic studies. However, with increasing availability of larger population cohorts, and a shift from analysis of spectral data to using quantified metabolite levels, both more traditional statistical approaches and alternative machine learning methods have become more widely used. This review aims at providing an overview of the current state-of-the-art multivariate methods for the analysis of NMR-based metabolomic data as well as alternative methods, highlighting their strengths and limitations.
Collapse
Affiliation(s)
- Julia Debik
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology-NTNU, Trondheim, Norway
| | - Matteo Sangermani
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology-NTNU, Trondheim, Norway
| | - Feng Wang
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology-NTNU, Trondheim, Norway
- Clinic of Surgery, St. Olavs Hospital HF, Trondheim, Norway
| | - Torfinn S Madssen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology-NTNU, Trondheim, Norway
| | - Guro F Giskeødegård
- Clinic of Surgery, St. Olavs Hospital HF, Trondheim, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology-NTNU, Trondheim, Norway
| |
Collapse
|
41
|
Metabolic Phenotyping in Prostate Cancer Using Multi-Omics Approaches. Cancers (Basel) 2022; 14:cancers14030596. [PMID: 35158864 PMCID: PMC8833769 DOI: 10.3390/cancers14030596] [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] [Received: 12/28/2021] [Revised: 01/14/2022] [Accepted: 01/20/2022] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer (PCa), one of the most frequently diagnosed cancers among men worldwide, is characterized by a diverse biological heterogeneity. It is well known that PCa cells rewire their cellular metabolism to meet the higher demands required for survival, proliferation, and invasion. In this context, a deeper understanding of metabolic reprogramming, an emerging hallmark of cancer, could provide novel opportunities for cancer diagnosis, prognosis, and treatment. In this setting, multi-omics data integration approaches, including genomics, epigenomics, transcriptomics, proteomics, lipidomics, and metabolomics, could offer unprecedented opportunities for uncovering the molecular changes underlying metabolic rewiring in complex diseases, such as PCa. Recent studies, focused on the integrated analysis of multi-omics data derived from PCa patients, have in fact revealed new insights into specific metabolic reprogramming events and vulnerabilities that have the potential to better guide therapy and improve outcomes for patients. This review aims to provide an up-to-date summary of multi-omics studies focused on the characterization of the metabolomic phenotype of PCa, as well as an in-depth analysis of the correlation between changes identified in the multi-omics studies and the metabolic profile of PCa tumors.
Collapse
|
42
|
Lépine G, Tremblay-Franco M, Bouder S, Dimina L, Fouillet H, Mariotti F, Polakof S. Investigating the Postprandial Metabolome after Challenge Tests to Assess Metabolic Flexibility and Dysregulations Associated with Cardiometabolic Diseases. Nutrients 2022; 14:nu14030472. [PMID: 35276829 PMCID: PMC8840206 DOI: 10.3390/nu14030472] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/16/2022] Open
Abstract
This review focuses on the added value provided by a research strategy applying metabolomics analyses to assess phenotypic flexibility in response to different nutritional challenge tests in the framework of metabolic clinical studies. We discuss findings related to the Oral Glucose Tolerance Test (OGTT) and to mixed meals with varying fat contents and food matrix complexities. Overall, the use of challenge tests combined with metabolomics revealed subtle metabolic dysregulations exacerbated during the postprandial period when comparing healthy and at cardiometabolic risk subjects. In healthy subjects, consistent postprandial metabolic shifts driven by insulin action were reported (e.g., a switch from lipid to glucose oxidation for energy fueling) with similarities between OGTT and mixed meals, especially during the first hours following meal ingestion while differences appeared in a wider timeframe. In populations with expected reduced phenotypic flexibility, often associated with increased cardiometabolic risk, a blunted response on most key postprandial pathways was reported. We also discuss the most suitable statistical tools to analyze the dynamic alterations of the postprandial metabolome while accounting for complexity in study designs and data structure. Overall, the in-depth characterization of the postprandial metabolism and associated phenotypic flexibility appears highly promising for a better understanding of the onset of cardiometabolic diseases.
Collapse
Affiliation(s)
- Gaïa Lépine
- Université Clermont Auvergne, INRAE, UMR 1019, Unité Nutrition Humaine, 63000 Clermont-Ferrand, France; (G.L.); (S.B.); (L.D.)
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 75005 Paris, France; (H.F.); (F.M.)
| | - Marie Tremblay-Franco
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, 31300 Toulouse, France;
- Axiom Platform, MetaToul-MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, 31300 Toulouse, France
| | - Sabrine Bouder
- Université Clermont Auvergne, INRAE, UMR 1019, Unité Nutrition Humaine, 63000 Clermont-Ferrand, France; (G.L.); (S.B.); (L.D.)
| | - Laurianne Dimina
- Université Clermont Auvergne, INRAE, UMR 1019, Unité Nutrition Humaine, 63000 Clermont-Ferrand, France; (G.L.); (S.B.); (L.D.)
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 75005 Paris, France; (H.F.); (F.M.)
| | - Hélène Fouillet
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 75005 Paris, France; (H.F.); (F.M.)
| | - François Mariotti
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 75005 Paris, France; (H.F.); (F.M.)
| | - Sergio Polakof
- Université Clermont Auvergne, INRAE, UMR 1019, Unité Nutrition Humaine, 63000 Clermont-Ferrand, France; (G.L.); (S.B.); (L.D.)
- Correspondence:
| |
Collapse
|
43
|
Lim SY, Selvaraji S, Lau H, Li SFY. Application of omics beyond the central dogma in coronary heart disease research: A bibliometric study and literature review. Comput Biol Med 2022; 140:105069. [PMID: 34847384 DOI: 10.1016/j.compbiomed.2021.105069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022]
Abstract
Despite remarkable progress in disease diagnosis and treatment, coronary heart disease (CHD) remains the number one leading cause of death worldwide. Many practical challenges still faced in clinical settings necessitates the pursuit of omics studies to identify alternative/orthogonal biomarkers, as well as to discover novel insights into disease mechanisms. Albeit relatively nascent as compared to the omics frontrunners (genomics, transcriptomics, and proteomics), omics beyond the central dogma (OBCD; e.g., metabolomics, lipidomics, glycomics, and metallomics) have undeniable contributions and prospects in CHD research. In this bibliometric study, we characterised the global trends in publication/citation outputs, collaborations, and research hotspots concerning OBCD-CHD, with a focus on the more prolific fields of metabolomics and lipidomics. As for glycomics and metallomics, there were insufficient publication records on their applications in CHD research for quantitative bibliometrics analysis. Thus, we reviewed their applications in health/disease research in general, discussed and justified their potential in CHD research, and suggested important/promising research avenues. By summarising evidence obtained both quantitatively and qualitatively, this study offers a first and comprehensive picture of OBCD applications in CHD, facilitating the establishment of future research directions.
Collapse
Affiliation(s)
- Si Ying Lim
- Integrative Sciences & Engineering Programme, NUS Graduate School, National University of Singapore, University Hall, Tan Chin Tuan Wing, Singapore 119077, Singapore; Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore
| | - Sharmelee Selvaraji
- Integrative Sciences & Engineering Programme, NUS Graduate School, National University of Singapore, University Hall, Tan Chin Tuan Wing, Singapore 119077, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, 2 Medical Drive MD9, National University of Singapore, Singapore 117593, Singapore
| | - Hazel Lau
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore
| | - Sam Fong Yau Li
- Integrative Sciences & Engineering Programme, NUS Graduate School, National University of Singapore, University Hall, Tan Chin Tuan Wing, Singapore 119077, Singapore; Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore.
| |
Collapse
|
44
|
Al-Harazi O, Kaya IH, El Allali A, Colak D. A Network-Based Methodology to Identify Subnetwork Markers for Diagnosis and Prognosis of Colorectal Cancer. Front Genet 2021; 12:721949. [PMID: 34790220 PMCID: PMC8591094 DOI: 10.3389/fgene.2021.721949] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/28/2021] [Indexed: 12/30/2022] Open
Abstract
The development of reliable methods for identification of robust biomarkers for complex diseases is critical for disease diagnosis and prognosis efforts. Integrating multi-omics data with protein-protein interaction (PPI) networks to investigate diseases may help better understand disease characteristics at the molecular level. In this study, we developed and tested a novel network-based method to detect subnetwork markers for patients with colorectal cancer (CRC). We performed an integrated omics analysis using whole-genome gene expression profiling and copy number alterations (CNAs) datasets followed by building a gene interaction network for the significantly altered genes. We then clustered the constructed gene network into subnetworks and assigned a score for each significant subnetwork. We developed a support vector machine (SVM) classifier using these scores as feature values and tested the methodology in independent CRC transcriptomic datasets. The network analysis resulted in 15 subnetwork markers that revealed several hub genes that may play a significant role in colorectal cancer, including PTP4A3, FGFR2, PTX3, AURKA, FEN1, INHBA, and YES1. The 15-subnetwork classifier displayed over 98 percent accuracy in detecting patients with CRC. In comparison to individual gene biomarkers, subnetwork markers based on integrated multi-omics and network analyses may lead to better disease classification, diagnosis, and prognosis.
Collapse
Affiliation(s)
- Olfat Al-Harazi
- Biostatistics, Epidemiology and Scientific Computing Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ibrahim H Kaya
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Achraf El Allali
- African Genome Center, Mohammed VI Polytechnic University, Benguerir, Morocco
| | - Dilek Colak
- Biostatistics, Epidemiology and Scientific Computing Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
45
|
Carmona-Bayonas A, Jiménez-Fonseca P, Gallego J, Msaouel P. Causal Considerations Can Inform the Interpretation of Surprising Associations in Medical Registries. Cancer Invest 2021; 40:1-13. [PMID: 34709109 DOI: 10.1080/07357907.2021.1999971] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
An exploratory analysis of registry data from 2437 patients with advanced gastric cancer revealed a surprising association between astrological birth signs and overall survival (OS) with p = 0.01. After dichotomizing or changing the reference sign, p-values <0.05 were observed for several birth signs following adjustments for multiple comparisons. Bayesian models with moderately skeptical priors still pointed to these associations. A more plausible causal model, justified by contextual knowledge, revealed that these associations arose from the astrological sign association with seasonality. This case study illustrates how causal considerations can guide analyses through what would otherwise be a hopeless maze of statistical possibilities.
Collapse
Affiliation(s)
- Alberto Carmona-Bayonas
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, UMU, IMIB, Murcia, Spain
| | - Paula Jiménez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Javier Gallego
- Medical Oncology Department, Hospital General de Elche, Elche, Spain
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
46
|
Wang C, Liang J, Yang W, Wang S, Yu J, Jia P, Du Y, Wang M, Li Y, Zheng X. Ultra-Performance Liquid Chromatography-Q-Exactive Orbitrap-Mass Spectrometry Analysis for Metabolic Communication between Heart and Kidney in Adriamycin-Induced Nephropathy Rats. Kidney Blood Press Res 2021; 47:31-42. [PMID: 34662875 DOI: 10.1159/000519015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 08/12/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Although the adriamycin-induced nephropathy model is frequently employed in the study of nephrotic syndrome and focal segmental glomerulosclerosis, the accompanying myocardial damage has always been a cause for concern. Therefore, there is a great need to study cardiorenal communication in this model. METHODS An adriamycin-induced nephropathy model was established via tail vein injection. The levels of the biochemical indicators serum albumin, serum globulin, serum total protein, serum cholesterol, serum creatinine (SCr), urinary protein, and urinary creatinine (UCr) were measured, and histopathological changes in the heart and kidneys were assessed using hematoxylin-eosin staining. Metabolomic changes in the heart, blood, and kidneys were analyzed using the metabolomics method based on ultra-performance liquid chromatography Q-Exactive Orbitrap mass spectrometry. RESULTS Compared with the control group, the model group showed significant decreases in serum protein and total protein levels, albumin/globulin ratio, and creatinine clearance rate as well as significant increases in serum cholesterol, SCr, urinary protein, and UCr levels. Significant pathological changes were observed in the renal pathology sections in the model group, including diffusely merged glomerular epithelial cells, inflammatory infiltration, and vacuolated glomerular cells. Additionally, thickened myocardial fibers, swollen nuclei, inflammatory infiltration, and partial myocardial necrosis could be seen in the cardiac pathology sections in the model group. Based on multivariate statistical analysis, a total of 20 differential metabolites associated with 15 metabolic pathways were identified in the heart, 7 differential metabolites with 7 metabolic pathways were identified in the blood, and 16 differential metabolites with 21 metabolic pathways were identified in the kidney. Moreover, 6 common metabolic pathways shared by the heart and kidney were identified: arginine and proline metabolism; arginine biosynthesis; glutathione metabolism; alanine, aspartate, and glutamate metabolism; beta-alanine metabolism; and histidine metabolism. Among these metabolic pathways, alanine, aspartate, and glutamate metabolism was shared by the heart, blood, and kidney. Succinic acid was found to be the key regulatory metabolite in cardiorenal metabolic communication. CONCLUSION Six metabolic pathways were found to be involved in cardiorenal metabolic communication in an adriamycin-induced nephropathy model, in which alanine, aspartate, and glutamate metabolism may be the metabolic link between the heart and kidney in the development and maintenance of oxidative stress and inflammation. Succinic acid may serve as a key regulatory metabolic switch or marker of cardiac and renal co-injury, as shown in an adriamycin-induced nephropathy model.
Collapse
Affiliation(s)
- Chunliu Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, China.,Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Jiping Liang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, China
| | - Wenwen Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, China
| | - Shixiang Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, China
| | - Jie Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, China
| | - Pu Jia
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, China
| | - Yapeng Du
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, China
| | - Mei Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, China
| | - Ye Li
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, China
| |
Collapse
|
47
|
Piras C, Noto A, Ibba L, Deidda M, Fanos V, Muntoni S, Leoni VP, Atzori L. Contribution of Metabolomics to the Understanding of NAFLD and NASH Syndromes: A Systematic Review. Metabolites 2021; 11:metabo11100694. [PMID: 34677409 PMCID: PMC8541039 DOI: 10.3390/metabo11100694] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/03/2021] [Accepted: 10/06/2021] [Indexed: 12/20/2022] Open
Abstract
Several differential panels of metabolites have been associated with the presence of metabolic syndrome and its related conditions, namely non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). This study aimed to perform a systematic review to summarize the most recent finding in terms of circulating biomarkers following NAFLD/NASH syndromes. Hence, the research was focused on NAFLD/NASH studies analysed by metabolomics approaches. Following Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines, a systematic search was conducted on the PubMed database. The inclusion criteria were (i) publication date between 2010 and 2021, (ii) presence of the combination of terms: metabolomics and NAFLD/NASH, and (iii) published in a scholarly peer-reviewed journal. Studies were excluded from the review if they were (i) single-case studies, (ii) unpublished thesis and dissertation studies, and (iii) not published in a peer-reviewed journal. Following these procedures, 10 eligible studies among 93 were taken into consideration. The metabolisms of amino acids, fatty acid, and vitamins were significantly different in patients affected by NAFLD and NASH compared to healthy controls. These findings suggest that low weight metabolites are an important indicator for NAFLD/NASH syndrome and there is a strong overlap between NAFLD/NASH and the metabolic syndrome. These findings may lead to new perspectives in early diagnosis, identification of novel biomarkers, and providing novel targets for pharmacological interventions.
Collapse
Affiliation(s)
- Cristina Piras
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (C.P.); (L.I.); (S.M.); (V.P.L.); (L.A.)
| | - Antonio Noto
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (C.P.); (L.I.); (S.M.); (V.P.L.); (L.A.)
- Correspondence:
| | - Luciano Ibba
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (C.P.); (L.I.); (S.M.); (V.P.L.); (L.A.)
| | - Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato, Italy;
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University of Cagliari, 09042 Monserrato, Italy;
| | - Sandro Muntoni
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (C.P.); (L.I.); (S.M.); (V.P.L.); (L.A.)
| | - Vera Piera Leoni
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (C.P.); (L.I.); (S.M.); (V.P.L.); (L.A.)
| | - Luigi Atzori
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy; (C.P.); (L.I.); (S.M.); (V.P.L.); (L.A.)
| |
Collapse
|
48
|
Gómez-Cebrián N, Vázquez Ferreiro P, Carrera Hueso FJ, Poveda Andrés JL, Puchades-Carrasco L, Pineda-Lucena A. Pharmacometabolomics by NMR in Oncology: A Systematic Review. Pharmaceuticals (Basel) 2021; 14:ph14101015. [PMID: 34681239 PMCID: PMC8539252 DOI: 10.3390/ph14101015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
Pharmacometabolomics (PMx) studies aim to predict individual differences in treatment response and in the development of adverse effects associated with specific drug treatments. Overall, these studies inform us about how individuals will respond to a drug treatment based on their metabolic profiles obtained before, during, or after the therapeutic intervention. In the era of precision medicine, metabolic profiles hold great potential to guide patient selection and stratification in clinical trials, with a focus on improving drug efficacy and safety. Metabolomics is closely related to the phenotype as alterations in metabolism reflect changes in the preceding cascade of genomics, transcriptomics, and proteomics changes, thus providing a significant advance over other omics approaches. Nuclear Magnetic Resonance (NMR) is one of the most widely used analytical platforms in metabolomics studies. In fact, since the introduction of PMx studies in 2006, the number of NMR-based PMx studies has been continuously growing and has provided novel insights into the specific metabolic changes associated with different mechanisms of action and/or toxic effects. This review presents an up-to-date summary of NMR-based PMx studies performed over the last 10 years. Our main objective is to discuss the experimental approaches used for the characterization of the metabolic changes associated with specific therapeutic interventions, the most relevant results obtained so far, and some of the remaining challenges in this area.
Collapse
Affiliation(s)
- Nuria Gómez-Cebrián
- Drug Discovery Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain;
| | | | | | | | - Leonor Puchades-Carrasco
- Drug Discovery Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain;
- Correspondence: (L.P.-C.); (A.P.-L.); Tel.: +34-963246713 (L.P.-C.)
| | - Antonio Pineda-Lucena
- Molecular Therapeutics Program, Centro de Investigación Médica Aplicada, 31008 Navarra, Spain
- Correspondence: (L.P.-C.); (A.P.-L.); Tel.: +34-963246713 (L.P.-C.)
| |
Collapse
|
49
|
Elmore JM, Griffin BD, Walley JW. Advances in functional proteomics to study plant-pathogen interactions. CURRENT OPINION IN PLANT BIOLOGY 2021; 63:102061. [PMID: 34102449 DOI: 10.1016/j.pbi.2021.102061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/22/2021] [Accepted: 04/25/2021] [Indexed: 05/20/2023]
Abstract
Pathogen infection triggers complex signaling networks in plant cells that ultimately result in either susceptibility or resistance. We have made substantial progress in dissecting many of these signaling events, and it is becoming clear that changes in proteome composition and protein activity are major drivers of plant-microbe interactions. Here, we highlight different approaches to analyze the functional proteomes of hosts and pathogens and discuss how they have been used to further our understanding of plant disease. Global proteome profiling can quantify the dynamics of proteins, posttranslational modifications, and biological pathways that contribute to immune-related outcomes. In addition, emerging techniques such as enzyme activity-based profiling, proximity labeling, and kinase-substrate profiling are being used to dissect biochemical events that operate during infection. Finally, we discuss how these functional approaches can be integrated with other profiling data to gain a mechanistic, systems-level view of plant and pathogen signaling.
Collapse
Affiliation(s)
- James M Elmore
- Department of Plant Pathology and Microbiology, Iowa State University, Ames, IA, 50014, USA.
| | - Brianna D Griffin
- Department of Plant Pathology and Microbiology, Iowa State University, Ames, IA, 50014, USA
| | - Justin W Walley
- Department of Plant Pathology and Microbiology, Iowa State University, Ames, IA, 50014, USA.
| |
Collapse
|
50
|
Santiago-Rodriguez TM, Hollister EB. Multi 'omic data integration: A review of concepts, considerations, and approaches. Semin Perinatol 2021; 45:151456. [PMID: 34256961 DOI: 10.1016/j.semperi.2021.151456] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The application of 'omic techniques including, but not limited to genomics/metagenomics, transcriptomics/meta-transcriptomics, proteomics/meta-proteomics, and metabolomics to generate multiple datasets from a single sample have facilitated hypothesis generation leading to the identification of biological, molecular and ecological functions and mechanisms, as well as associations and correlations. Despite their power and promise, a variety of challenges must be considered in the successful design and execution of a multi-omics study. In this review, various 'omic technologies applicable to single- and meta-organisms (i.e., host + microbiome) are described, and considerations for sample collection, storage and processing prior to data generation and analysis, as well as approaches to data storage, dissemination and analysis are discussed. Finally, case studies are included as examples of multi-omic applications providing novel insights and a more holistic understanding of biological processes.
Collapse
Affiliation(s)
| | - Emily B Hollister
- Diversigen, Inc, 3 Greenway Plaza, Suite 1575, Houston, TX 77046, USA.
| |
Collapse
|