1
|
Liu N, Kattan WE, Mead BE, Kummerlowe C, Cheng T, Ingabire S, Cheah JH, Soule CK, Vrcic A, McIninch JK, Triana S, Guzman M, Dao TT, Peters JM, Lowder KE, Crawford L, Amini AP, Blainey PC, Hahn WC, Cleary B, Bryson B, Winter PS, Raghavan S, Shalek AK. Scalable, compressed phenotypic screening using pooled perturbations. Nat Biotechnol 2024:10.1038/s41587-024-02403-z. [PMID: 39375446 DOI: 10.1038/s41587-024-02403-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 08/26/2024] [Indexed: 10/09/2024]
Abstract
High-throughput phenotypic screens using biochemical perturbations and high-content readouts are constrained by limitations of scale. To address this, we establish a method of pooling exogenous perturbations followed by computational deconvolution to reduce required sample size, labor and cost. We demonstrate the increased efficiency of compressed experimental designs compared to conventional approaches through benchmarking with a bioactive small-molecule library and a high-content imaging readout. We then apply compressed screening in two biological discovery campaigns. In the first, we use early-passage pancreatic cancer organoids to map transcriptional responses to a library of recombinant tumor microenvironment protein ligands, uncovering reproducible phenotypic shifts induced by specific ligands distinct from canonical reference signatures and correlated with clinical outcome. In the second, we identify the pleotropic modulatory effects of a chemical compound library with known mechanisms of action on primary human peripheral blood mononuclear cell immune responses. In sum, our approach empowers phenotypic screens with information-rich readouts to advance drug discovery efforts and basic biological inquiry.
Collapse
Affiliation(s)
- Nuo Liu
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Walaa E Kattan
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Benjamin E Mead
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Conner Kummerlowe
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Thomas Cheng
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Sarah Ingabire
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Jaime H Cheah
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christian K Soule
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anita Vrcic
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jane K McIninch
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sergio Triana
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Manuel Guzman
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Tyler T Dao
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joshua M Peters
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kristen E Lowder
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana Farber Cancer Institute, Boston, MA, USA
| | - Lorin Crawford
- Microsoft Research, Cambridge, MA, USA
- Center for Computational Biology, Brown University, Providence, RI, USA
- Department of Biostatistics, Brown University, Providence, RI, USA
| | | | - Paul C Blainey
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - William C Hahn
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Brian Cleary
- Faculty of Computing and Data Sciences, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Department of Biology, Boston University, Boston, MA, USA
| | - Bryan Bryson
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Srivatsan Raghavan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Alex K Shalek
- Institute for Medical Engineering and Science (IMES) and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Program in Immunology, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
2
|
Duan X, Wan JMF, Yu ACH. The molecular impact of sonoporation: A transcriptomic analysis of gene regulation profile. ULTRASONICS SONOCHEMISTRY 2024:107077. [PMID: 39368882 DOI: 10.1016/j.ultsonch.2024.107077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/17/2024] [Accepted: 09/17/2024] [Indexed: 10/07/2024]
Abstract
Sonoporation has long been known to disrupt intracellular signaling, yet the involved molecules and pathways have not been identified with clarity. In this study, we employed whole transcriptome shotgun sequencing (RNA-seq) to profile sonoporation-induced gene responses after membrane resealing has taken place. Sonoporation was achieved by microbubble-mediated ultrasound (MB-US) exposure in the form of 1 MHz ultrasound pulsing (0.50 MPa peak negative pressure, 10 % duty cycle, 30 s exposure period) in the presence of microbubbles (1:1 cell-to-bubble ratio). Using propidium iodide (PI) and calcein respectively as cell viability and cytoplasmic uptake labels, post-exposure flow cytometry was performed to identify three viable cell populations: 1) unsonoporated cells, 2) sonoporated cells with low uptake, and 3) sonoporated cells with high uptake. Fluorescence-activated cell sorting was then conducted to separate the different groups followed by RNA-seq analysis of the gene expressions in each group of cells. We found that sonoporated cells with low or high calcein uptake showed high similarity in the gene responses, including the activation of multiple heat shock protein (HSP) genes and immediate early response genes mediating apoptosis and transcriptional regulation. In contrast, unsonoporated cells exhibited a more extensive gene expression alteration that included the activation of more HSP genes and the upregulation of diverse apoptotic mediators. Four oxidative stress-related and three immune-related genes were also differentially expressed in unsonoporated cells. Our results provided new information for understanding the intracellular mobilization in response to sonoporation at the molecular level, including the identification of new molecules in the sonoporation-induced apoptosis regulatory network. Our data also shed light on the innovative therapeutic strategy which could potentially leverage the responses of viable unsonoporated cells as a synergistic effector in the microenvironment to favor tumor treatment.
Collapse
Affiliation(s)
- Xinxing Duan
- Schlegel Research Institute for Aging and Department of Electrical & Computer Engineering, University of Waterloo, Waterloo, ON N2L3G1, Canada; School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China; State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Jennifer M F Wan
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Alfred C H Yu
- Schlegel Research Institute for Aging and Department of Electrical & Computer Engineering, University of Waterloo, Waterloo, ON N2L3G1, Canada.
| |
Collapse
|
3
|
Bartzis G, Peeters CFW, Ligterink W, Van Eeuwijk FA. A guided network estimation approach using multi-omic information. BMC Bioinformatics 2024; 25:202. [PMID: 38816801 PMCID: PMC11137963 DOI: 10.1186/s12859-024-05778-7] [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: 10/24/2023] [Accepted: 04/11/2024] [Indexed: 06/01/2024] Open
Abstract
INTODUCTION In systems biology, an organism is viewed as a system of interconnected molecular entities. To understand the functioning of organisms it is essential to integrate information about the variations in the concentrations of those molecular entities. This information can be structured as a set of networks with interconnections and with some hierarchical relations between them. Few methods exist for the reconstruction of integrative networks. OBJECTIVE In this work, we propose an integrative network reconstruction method in which the network organization for a particular type of omics data is guided by the network structure of a related type of omics data upstream in the omic cascade. The structure of these guiding data can be either already known or be estimated from the guiding data themselves. METHODS The method consists of three steps. First a network structure for the guiding data should be provided. Next, responses in the target set are regressed on the full set of predictors in the guiding data with a Lasso penalty to reduce the number of predictors and an L2 penalty on the differences between coefficients for predictors that share edges in the network for the guiding data. Finally, a network is reconstructed on the fitted target responses as functions of the predictors in the guiding data. This way we condition the target network on the network of the guiding data. CONCLUSIONS We illustrate our approach on two examples in Arabidopsis. The method detects groups of metabolites that have a similar genetic or transcriptomic basis.
Collapse
Affiliation(s)
- Georgios Bartzis
- Mathematical and Statistical Methods Group - Biometris, Wageningen University and Research, Wageningen, The Netherlands
| | - Carel F W Peeters
- Mathematical and Statistical Methods Group - Biometris, Wageningen University and Research, Wageningen, The Netherlands.
| | - Wilco Ligterink
- Laboratory of Plant Physiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Fred A Van Eeuwijk
- Mathematical and Statistical Methods Group - Biometris, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
4
|
Zhou W, Yan Z, Zhang L. A comparative study of 11 non-linear regression models highlighting autoencoder, DBN, and SVR, enhanced by SHAP importance analysis in soybean branching prediction. Sci Rep 2024; 14:5905. [PMID: 38467662 PMCID: PMC10928191 DOI: 10.1038/s41598-024-55243-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/21/2024] [Indexed: 03/13/2024] Open
Abstract
To explore a robust tool for advancing digital breeding practices through an artificial intelligence-driven phenotype prediction expert system, we undertook a thorough analysis of 11 non-linear regression models. Our investigation specifically emphasized the significance of Support Vector Regression (SVR) and SHapley Additive exPlanations (SHAP) in predicting soybean branching. By using branching data (phenotype) of 1918 soybean accessions and 42 k SNP (Single Nucleotide Polymorphism) polymorphic data (genotype), this study systematically compared 11 non-linear regression AI models, including four deep learning models (DBN (deep belief network) regression, ANN (artificial neural network) regression, Autoencoders regression, and MLP (multilayer perceptron) regression) and seven machine learning models (e.g., SVR (support vector regression), XGBoost (eXtreme Gradient Boosting) regression, Random Forest regression, LightGBM regression, GPs (Gaussian processes) regression, Decision Tree regression, and Polynomial regression). After being evaluated by four valuation metrics: R2 (R-squared), MAE (Mean Absolute Error), MSE (Mean Squared Error), and MAPE (Mean Absolute Percentage Error), it was found that the SVR, Polynomial Regression, DBN, and Autoencoder outperformed other models and could obtain a better prediction accuracy when they were used for phenotype prediction. In the assessment of deep learning approaches, we exemplified the SVR model, conducting analyses on feature importance and gene ontology (GO) enrichment to provide comprehensive support. After comprehensively comparing four feature importance algorithms, no notable distinction was observed in the feature importance ranking scores across the four algorithms, namely Variable Ranking, Permutation, SHAP, and Correlation Matrix, but the SHAP value could provide rich information on genes with negative contributions, and SHAP importance was chosen for feature selection. The results of this study offer valuable insights into AI-mediated plant breeding, addressing challenges faced by traditional breeding programs. The method developed has broad applicability in phenotype prediction, minor QTL (quantitative trait loci) mining, and plant smart-breeding systems, contributing significantly to the advancement of AI-based breeding practices and transitioning from experience-based to data-based breeding.
Collapse
Affiliation(s)
- Wei Zhou
- Florida Agricultural and Mechanical University, Tallahassee, FL, 32307, USA.
| | - Zhengxiao Yan
- Florida State University, Tallahassee, FL, 32306, USA
| | - Liting Zhang
- Florida State University, Tallahassee, FL, 32306, USA
| |
Collapse
|
5
|
Murmu S, Sinha D, Chaurasia H, Sharma S, Das R, Jha GK, Archak S. A review of artificial intelligence-assisted omics techniques in plant defense: current trends and future directions. FRONTIERS IN PLANT SCIENCE 2024; 15:1292054. [PMID: 38504888 PMCID: PMC10948452 DOI: 10.3389/fpls.2024.1292054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 01/24/2024] [Indexed: 03/21/2024]
Abstract
Plants intricately deploy defense systems to counter diverse biotic and abiotic stresses. Omics technologies, spanning genomics, transcriptomics, proteomics, and metabolomics, have revolutionized the exploration of plant defense mechanisms, unraveling molecular intricacies in response to various stressors. However, the complexity and scale of omics data necessitate sophisticated analytical tools for meaningful insights. This review delves into the application of artificial intelligence algorithms, particularly machine learning and deep learning, as promising approaches for deciphering complex omics data in plant defense research. The overview encompasses key omics techniques and addresses the challenges and limitations inherent in current AI-assisted omics approaches. Moreover, it contemplates potential future directions in this dynamic field. In summary, AI-assisted omics techniques present a robust toolkit, enabling a profound understanding of the molecular foundations of plant defense and paving the way for more effective crop protection strategies amidst climate change and emerging diseases.
Collapse
Affiliation(s)
- Sneha Murmu
- Indian Agricultural Statistics Research Institute, Indian Council of Agricultural Research (ICAR), New Delhi, India
| | - Dipro Sinha
- Indian Agricultural Statistics Research Institute, Indian Council of Agricultural Research (ICAR), New Delhi, India
| | - Himanshushekhar Chaurasia
- Central Institute for Research on Cotton Technology, Indian Council of Agricultural Research (ICAR), Mumbai, India
| | - Soumya Sharma
- Indian Agricultural Statistics Research Institute, Indian Council of Agricultural Research (ICAR), New Delhi, India
| | - Ritwika Das
- Indian Agricultural Statistics Research Institute, Indian Council of Agricultural Research (ICAR), New Delhi, India
| | - Girish Kumar Jha
- Indian Agricultural Statistics Research Institute, Indian Council of Agricultural Research (ICAR), New Delhi, India
| | - Sunil Archak
- National Bureau of Plant Genetic Resources, Indian Council of Agricultural Research (ICAR), New Delhi, India
| |
Collapse
|
6
|
Jang WJ, Lee S, Jeong CH. Uncovering transcriptomic biomarkers for enhanced diagnosis of methamphetamine use disorder: a comprehensive review. Front Psychiatry 2024; 14:1302994. [PMID: 38260797 PMCID: PMC10800441 DOI: 10.3389/fpsyt.2023.1302994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction Methamphetamine use disorder (MUD) is a chronic relapsing disorder characterized by compulsive Methamphetamine (MA) use despite its detrimental effects on physical, psychological, and social well-being. The development of MUD is a complex process that involves the interplay of genetic, epigenetic, and environmental factors. The treatment of MUD remains a significant challenge, with no FDA-approved pharmacotherapies currently available. Current diagnostic criteria for MUD rely primarily on self-reporting and behavioral assessments, which have inherent limitations owing to their subjective nature. This lack of objective biomarkers and unidimensional approaches may not fully capture the unique features and consequences of MA addiction. Methods We performed a literature search for this review using the Boolean search in the PubMed database. Results This review explores existing technologies for identifying transcriptomic biomarkers for MUD diagnosis. We examined non-invasive tissues and scrutinized transcriptomic biomarkers relevant to MUD. Additionally, we investigated transcriptomic biomarkers identified for diagnosing, predicting, and monitoring MUD in non-invasive tissues. Discussion Developing and validating non-invasive MUD biomarkers could address these limitations, foster more precise and reliable diagnostic approaches, and ultimately enhance the quality of care for individuals with MA addiction.
Collapse
Affiliation(s)
| | | | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| |
Collapse
|
7
|
Adarshan S, Sree VSS, Muthuramalingam P, Nambiar KS, Sevanan M, Satish L, Venkidasamy B, Jeelani PG, Shin H. Understanding Macroalgae: A Comprehensive Exploration of Nutraceutical, Pharmaceutical, and Omics Dimensions. PLANTS (BASEL, SWITZERLAND) 2023; 13:113. [PMID: 38202421 PMCID: PMC10780804 DOI: 10.3390/plants13010113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/17/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Driven by a surge in global interest in natural products, macroalgae or seaweed, has emerged as a prime source for nutraceuticals and pharmaceutical applications. Characterized by remarkable genetic diversity and a crucial role in marine ecosystems, these organisms offer not only substantial nutritional value in proteins, fibers, vitamins, and minerals, but also a diverse array of bioactive molecules with promising pharmaceutical properties. Furthermore, macroalgae produce approximately 80% of the oxygen in the atmosphere, highlighting their ecological significance. The unique combination of nutritional and bioactive attributes positions macroalgae as an ideal resource for food and medicine in various regions worldwide. This comprehensive review consolidates the latest advancements in the field, elucidating the potential applications of macroalgae in developing nutraceuticals and therapeutics. The review emphasizes the pivotal role of omics approaches in deepening our understanding of macroalgae's physiological and molecular characteristics. By highlighting the importance of omics, this review also advocates for continued exploration and utilization of these extraordinary marine organisms in diverse domains, including drug discovery, functional foods, and other industrial applications. The multifaceted potential of macroalgae warrants further research and development to unlock their full benefits and contribute to advancing global health and sustainable industries.
Collapse
Affiliation(s)
- Sivakumar Adarshan
- Department of Biotechnology, Alagappa University, Karaikudi 630003, Tamil Nadu, India;
| | - Vairavel Sivaranjani Sivani Sree
- Department of Biotechnology, Karunya Institute of Technology and Sciences, Coimbatore 641114, Tamil Nadu, India; (V.S.S.S.); (K.S.N.); (M.S.)
| | - Pandiyan Muthuramalingam
- Division of Horticultural Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52725, Republic of Korea;
- Department of Oral and Maxillofacial Surgery, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha Dental College and Hospitals, Saveetha University, Chennai 600077, Tamil Nadu, India;
| | - Krishnanjana S Nambiar
- Department of Biotechnology, Karunya Institute of Technology and Sciences, Coimbatore 641114, Tamil Nadu, India; (V.S.S.S.); (K.S.N.); (M.S.)
| | - Murugan Sevanan
- Department of Biotechnology, Karunya Institute of Technology and Sciences, Coimbatore 641114, Tamil Nadu, India; (V.S.S.S.); (K.S.N.); (M.S.)
| | - Lakkakula Satish
- Applied Phycology and Biotechnology Division, Marine Algal Research Station, CSIR—Central Salt and Marine Chemicals Research Institute, Mandapam 623519, Tamil Nadu, India;
| | - Baskar Venkidasamy
- Department of Oral and Maxillofacial Surgery, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha Dental College and Hospitals, Saveetha University, Chennai 600077, Tamil Nadu, India;
| | - Peerzada Gh Jeelani
- Department of Biotechnology, Microbiology & Bioinformatics, National College Trichy, Tiruchirapalli 620001, Tamil Nadu, India;
| | - Hyunsuk Shin
- Division of Horticultural Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52725, Republic of Korea;
| |
Collapse
|
8
|
Miano A, Rychel K, Lezia A, Sastry A, Palsson B, Hasty J. High-resolution temporal profiling of E. coli transcriptional response. Nat Commun 2023; 14:7606. [PMID: 37993418 PMCID: PMC10665441 DOI: 10.1038/s41467-023-43173-7] [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: 07/22/2023] [Accepted: 11/02/2023] [Indexed: 11/24/2023] Open
Abstract
Understanding how cells dynamically adapt to their environment is a primary focus of biology research. Temporal information about cellular behavior is often limited by both small numbers of data time-points and the methods used to analyze this data. Here, we apply unsupervised machine learning to a data set containing the activity of 1805 native promoters in E. coli measured every 10 minutes in a high-throughput microfluidic device via fluorescence time-lapse microscopy. Specifically, this data set reveals E. coli transcriptome dynamics when exposed to different heavy metal ions. We use a bioinformatics pipeline based on Independent Component Analysis (ICA) to generate insights and hypotheses from this data. We discovered three primary, time-dependent stages of promoter activation to heavy metal stress (fast, intermediate, and steady). Furthermore, we uncovered a global strategy E. coli uses to reallocate resources from stress-related promoters to growth-related promoters following exposure to heavy metal stress.
Collapse
Affiliation(s)
- Arianna Miano
- Department of Bioengineering, University of California San Diego, 9500 Gliman Dr, La Jolla, CA, USA.
| | - Kevin Rychel
- Department of Bioengineering, University of California San Diego, 9500 Gliman Dr, La Jolla, CA, USA
| | - Andrew Lezia
- Department of Bioengineering, University of California San Diego, 9500 Gliman Dr, La Jolla, CA, USA
| | - Anand Sastry
- Department of Bioengineering, University of California San Diego, 9500 Gliman Dr, La Jolla, CA, USA
| | - Bernhard Palsson
- Department of Bioengineering, University of California San Diego, 9500 Gliman Dr, La Jolla, CA, USA
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs, Lyngby, Denmark
| | - Jeff Hasty
- Department of Bioengineering, University of California San Diego, 9500 Gliman Dr, La Jolla, CA, USA
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, 9500 Gliman Dr, La Jolla, CA, USA
- Synthetic Biology Institute, University of California San Diego, 9500 Gliman Dr, La Jolla, CA, USA
| |
Collapse
|
9
|
Mattei G, Gan Z, Ramazzotti M, Palsson BO, Zielinski DC. Differential Expression Analysis Utilizing Condition-Specific Metabolic Pathways. Metabolites 2023; 13:1127. [PMID: 37999223 PMCID: PMC10672963 DOI: 10.3390/metabo13111127] [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: 07/01/2023] [Revised: 10/19/2023] [Accepted: 11/01/2023] [Indexed: 11/25/2023] Open
Abstract
Pathway analysis is ubiquitous in biological data analysis due to the ability to integrate small simultaneous changes in functionally related components. While pathways are often defined based on either manual curation or network topological properties, an attractive alternative is to generate pathways around specific functions, in which metabolism can be defined as the production and consumption of specific metabolites. In this work, we present an algorithm, termed MetPath, that calculates pathways for condition-specific production and consumption of specific metabolites. We demonstrate that these pathways have several useful properties. Pathways calculated in this manner (1) take into account the condition-specific metabolic role of a gene product, (2) are localized around defined metabolic functions, and (3) quantitatively weigh the importance of expression to a function based on the flux contribution of the gene product. We demonstrate how these pathways elucidate network interactions between genes across different growth conditions and between cell types. Furthermore, the calculated pathways compare favorably to manually curated pathways in predicting the expression correlation between genes. To facilitate the use of these pathways, we have generated a large compendium of pathways under different growth conditions for E. coli. The MetPath algorithm provides a useful tool for metabolic network-based statistical analyses of high-throughput data.
Collapse
Affiliation(s)
- Gianluca Mattei
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50121 Florence, Italy; (G.M.)
| | - Zhuohui Gan
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, China;
| | - Matteo Ramazzotti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50121 Florence, Italy; (G.M.)
| | - Bernhard O. Palsson
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093-0412, USA
| | - Daniel C. Zielinski
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093-0412, USA
| |
Collapse
|
10
|
Hao T, Fang W, Xu D, Chen Q, Liu Q, Cui K, Cao X, Li Y, Mai K, Ai Q. Phosphatidylethanolamine alleviates OX-LDL-induced macrophage inflammation by upregulating autophagy and inhibiting NLRP1 inflammasome activation. Free Radic Biol Med 2023; 208:402-417. [PMID: 37660837 DOI: 10.1016/j.freeradbiomed.2023.08.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
Oxidized low-density lipoprotein (OX-LDL)-induced inflammation and autophagy dysregulation are important events in the progression of atherosclerosis. Phosphatidylethanolamine (PE), a multifunctional phospholipid that is enriched in cells, has been proven to be directly involved in autophagy which is closely associated with inflammation. However, whether PE can influence OX-LDL-induced autophagy dysregulation and inflammation has not been reported. In the present study, we revealed that OX-LDL significantly induced macrophage inflammation through the CD36-NLRP1-caspase-1 signaling pathway in fish. Meanwhile, cellular PE levels were significantly decreased in response to OX-LDL induction. Based on the relationship between PE and autophagy, we then examined the effect of PE supplementation on OX-LDL-mediated autophagy impairment and inflammation induction in macrophages. As expected, exogenous PE restored impaired autophagy and alleviated inflammation in OX-LDL-stimulated cells. Notably, autophagy inhibitors reversed the inhibitory effect of PE on OX-LDL-induced maturation of IL-1β, indicating that the regulation of PE on OX-LDL-induced inflammation is dependent on autophagy. Furthermore, the positive effect of PE on OX-LDL-induced inflammation was relatively conserved in mouse and fish macrophages. In conclusion, we elucidated the role of the CD36-NLRP1-caspase-1 signaling pathway in OX-LDL-induced inflammation in fish and revealed for the first time that altering PE abundance in OX-LDL-treated cells could alleviate inflammasome-mediated inflammation by inducing autophagy. Given the relationship between OX-LDL-induced inflammation and atherosclerosis, this study prompts that the use of PE-rich foods promises to be a new strategy for atherosclerosis treatment in vertebrates.
Collapse
Affiliation(s)
- Tingting Hao
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Wei Fang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Dan Xu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Qiang Chen
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Qiangde Liu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Kun Cui
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Xiufei Cao
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Yueru Li
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, 266237, Qingdao, Shandong, People's Republic of China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs), Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, 266237, Qingdao, Shandong, People's Republic of China.
| |
Collapse
|
11
|
Bertile F, Matallana-Surget S, Tholey A, Cristobal S, Armengaud J. Diversifying the concept of model organisms in the age of -omics. Commun Biol 2023; 6:1062. [PMID: 37857885 PMCID: PMC10587087 DOI: 10.1038/s42003-023-05458-x] [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: 06/21/2023] [Accepted: 10/13/2023] [Indexed: 10/21/2023] Open
Abstract
In today's post-genomic era, it is crucial to rethink the concept of model organisms. While a few historically well-established organisms, e.g. laboratory rodents, have enabled significant scientific breakthroughs, there is now a pressing need for broader inclusion. Indeed, new organisms and models, from complex microbial communities to holobionts, are essential to fully grasp the complexity of biological principles across the breadth of biodiversity. By fostering collaboration between biology, advanced molecular science and omics communities, we can collectively adopt new models, unraveling their molecular functioning, and uncovering fundamental mechanisms. This concerted effort will undoubtedly enhance human health, environmental quality, and biodiversity conservation.
Collapse
Affiliation(s)
- Fabrice Bertile
- Université de Strasbourg, CNRS, IPHC UMR 7178, 23 rue du Loess, 67037, Strasbourg Cedex 2, France.
| | - Sabine Matallana-Surget
- Division of Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - Andreas Tholey
- Systematic Proteome Research & Bioanalytics, Institute for Experimental Medicine, Christian-Albrechts-Universität zu Kiel, 24105, Kiel, Germany
| | - Susana Cristobal
- Department of Biomedical and Clinical Sciences, Cell Biology, Medical Faculty, Linköping University, Linköping, 581 85, Sweden
- Ikerbasque, Basque Foundation for Science, Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Barrio Sarriena, s/n, Leioa, 48940, Spain
| | - Jean Armengaud
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 30200, Bagnols-sur-Cèze, France
| |
Collapse
|
12
|
Toufiq M, Rinchai D, Bettacchioli E, Kabeer BSA, Khan T, Subba B, White O, Yurieva M, George J, Jourde-Chiche N, Chiche L, Palucka K, Chaussabel D. Harnessing large language models (LLMs) for candidate gene prioritization and selection. J Transl Med 2023; 21:728. [PMID: 37845713 PMCID: PMC10580627 DOI: 10.1186/s12967-023-04576-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 09/28/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Feature selection is a critical step for translating advances afforded by systems-scale molecular profiling into actionable clinical insights. While data-driven methods are commonly utilized for selecting candidate genes, knowledge-driven methods must contend with the challenge of efficiently sifting through extensive volumes of biomedical information. This work aimed to assess the utility of large language models (LLMs) for knowledge-driven gene prioritization and selection. METHODS In this proof of concept, we focused on 11 blood transcriptional modules associated with an Erythroid cells signature. We evaluated four leading LLMs across multiple tasks. Next, we established a workflow leveraging LLMs. The steps consisted of: (1) Selecting one of the 11 modules; (2) Identifying functional convergences among constituent genes using the LLMs; (3) Scoring candidate genes across six criteria capturing the gene's biological and clinical relevance; (4) Prioritizing candidate genes and summarizing justifications; (5) Fact-checking justifications and identifying supporting references; (6) Selecting a top candidate gene based on validated scoring justifications; and (7) Factoring in transcriptome profiling data to finalize the selection of the top candidate gene. RESULTS Of the four LLMs evaluated, OpenAI's GPT-4 and Anthropic's Claude demonstrated the best performance and were chosen for the implementation of the candidate gene prioritization and selection workflow. This workflow was run in parallel for each of the 11 erythroid cell modules by participants in a data mining workshop. Module M9.2 served as an illustrative use case. The 30 candidate genes forming this module were assessed, and the top five scoring genes were identified as BCL2L1, ALAS2, SLC4A1, CA1, and FECH. Researchers carefully fact-checked the summarized scoring justifications, after which the LLMs were prompted to select a top candidate based on this information. GPT-4 initially chose BCL2L1, while Claude selected ALAS2. When transcriptional profiling data from three reference datasets were provided for additional context, GPT-4 revised its initial choice to ALAS2, whereas Claude reaffirmed its original selection for this module. CONCLUSIONS Taken together, our findings highlight the ability of LLMs to prioritize candidate genes with minimal human intervention. This suggests the potential of this technology to boost productivity, especially for tasks that require leveraging extensive biomedical knowledge.
Collapse
Affiliation(s)
- Mohammed Toufiq
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Eleonore Bettacchioli
- INSERM UMR1227, Lymphocytes B et Autoimmunité, Université de Bretagne Occidentale, Brest, France
- Service de Rhumatologie, CHU de Brest, Brest, France
| | | | - Taushif Khan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Bishesh Subba
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Olivia White
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Marina Yurieva
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Joshy George
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Laurent Chiche
- Service de Médecine Interne, Hôpital Européen, Marseille, France
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | |
Collapse
|
13
|
Way GP, Sailem H, Shave S, Kasprowicz R, Carragher NO. Evolution and impact of high content imaging. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:292-305. [PMID: 37666456 DOI: 10.1016/j.slasd.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/09/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023]
Abstract
The field of high content imaging has steadily evolved and expanded substantially across many industry and academic research institutions since it was first described in the early 1990's. High content imaging refers to the automated acquisition and analysis of microscopic images from a variety of biological sample types. Integration of high content imaging microscopes with multiwell plate handling robotics enables high content imaging to be performed at scale and support medium- to high-throughput screening of pharmacological, genetic and diverse environmental perturbations upon complex biological systems ranging from 2D cell cultures to 3D tissue organoids to small model organisms. In this perspective article the authors provide a collective view on the following key discussion points relevant to the evolution of high content imaging: • Evolution and impact of high content imaging: An academic perspective • Evolution and impact of high content imaging: An industry perspective • Evolution of high content image analysis • Evolution of high content data analysis pipelines towards multiparametric and phenotypic profiling applications • The role of data integration and multiomics • The role and evolution of image data repositories and sharing standards • Future perspective of high content imaging hardware and software.
Collapse
Affiliation(s)
- Gregory P Way
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Heba Sailem
- School of Cancer and Pharmaceutical Sciences, King's College London, UK
| | - Steven Shave
- GlaxoSmithKline Medicines Research Centre, Gunnels Wood Rd, Stevenage SG1 2NY, UK; Edinburgh Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, UK
| | - Richard Kasprowicz
- GlaxoSmithKline Medicines Research Centre, Gunnels Wood Rd, Stevenage SG1 2NY, UK
| | - Neil O Carragher
- Edinburgh Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, UK.
| |
Collapse
|
14
|
Gant TW, Auerbach SS, Von Bergen M, Bouhifd M, Botham PA, Caiment F, Currie RA, Harrill J, Johnson K, Li D, Rouquie D, van Ravenzwaay B, Sistare F, Tralau T, Viant MR, van de Laan JW, Yauk C. Applying genomics in regulatory toxicology: a report of the ECETOC workshop on omics threshold on non-adversity. Arch Toxicol 2023; 97:2291-2302. [PMID: 37296313 PMCID: PMC10322787 DOI: 10.1007/s00204-023-03522-3] [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: 01/23/2023] [Accepted: 05/11/2023] [Indexed: 06/12/2023]
Abstract
In a joint effort involving scientists from academia, industry and regulatory agencies, ECETOC's activities in Omics have led to conceptual proposals for: (1) A framework that assures data quality for reporting and inclusion of Omics data in regulatory assessments; and (2) an approach to robustly quantify these data, prior to interpretation for regulatory use. In continuation of these activities this workshop explored and identified areas of need to facilitate robust interpretation of such data in the context of deriving points of departure (POD) for risk assessment and determining an adverse change from normal variation. ECETOC was amongst the first to systematically explore the application of Omics methods, now incorporated into the group of methods known as New Approach Methodologies (NAMs), to regulatory toxicology. This support has been in the form of both projects (primarily with CEFIC/LRI) and workshops. Outputs have led to projects included in the workplan of the Extended Advisory Group on Molecular Screening and Toxicogenomics (EAGMST) group of the Organisation for Economic Co-operation and Development (OECD) and to the drafting of OECD Guidance Documents for Omics data reporting, with potentially more to follow on data transformation and interpretation. The current workshop was the last in a series of technical methods development workshops, with a sub-focus on the derivation of a POD from Omics data. Workshop presentations demonstrated that Omics data developed within robust frameworks for both scientific data generation and analysis can be used to derive a POD. The issue of noise in the data was discussed as an important consideration for identifying robust Omics changes and deriving a POD. Such variability or "noise" can comprise technical or biological variation within a dataset and should clearly be distinguished from homeostatic responses. Adverse outcome pathways (AOPs) were considered a useful framework on which to assemble Omics methods, and a number of case examples were presented in illustration of this point. What is apparent is that high dimension data will always be subject to varying processing pipelines and hence interpretation, depending on the context they are used in. Yet, they can provide valuable input for regulatory toxicology, with the pre-condition being robust methods for the collection and processing of data together with a comprehensive description how the data were interpreted, and conclusions reached.
Collapse
Affiliation(s)
- Timothy W Gant
- United Kingdom Health Security Agency, Harwell Science Campus, Didcot, Oxfordshire, United Kingdom.
- Imperial College London School of Public Health, London, United Kingdom.
| | - Scott S Auerbach
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, RTP, Durham, NC, USA
| | - Martin Von Bergen
- Department for Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | | | | | - Florian Caiment
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | | | - Joshua Harrill
- Cellular and Molecular Toxicologist, Center for Computational Toxicology and Exposure (CCTE), U.S. Environmental Protection Agency, Durham, NC, USA
| | - Kamin Johnson
- Predictive Safety Center, Corteva Agriscience, Indianapolis, IN, USA
| | - Dongying Li
- National Center for Toxicological Research, U.S. Food and Drug Administration (FDA), 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - David Rouquie
- Bayer SAS, Bayer Crop Science, 355 Rue Dostoïevski, CS 90153, 06906, Valbonne Sophia-Antipolis, France
| | | | | | - Tewes Tralau
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Mark R Viant
- School of Biosciences, University of Birmingham, Birmingham, UK
| | | | - Carole Yauk
- Department of Biology, University of Ottawa, Ottawa, Canada
| |
Collapse
|
15
|
Reitz C, Pericak-Vance MA, Foroud T, Mayeux R. A global view of the genetic basis of Alzheimer disease. Nat Rev Neurol 2023; 19:261-277. [PMID: 37024647 PMCID: PMC10686263 DOI: 10.1038/s41582-023-00789-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 04/08/2023]
Abstract
The risk of Alzheimer disease (AD) increases with age, family history and informative genetic variants. Sadly, there is still no cure or means of prevention. As in other complex diseases, uncovering genetic causes of AD could identify underlying pathological mechanisms and lead to potential treatments. Rare, autosomal dominant forms of AD occur in middle age as a result of highly penetrant genetic mutations, but the most common form of AD occurs later in life. Large-scale, genome-wide analyses indicate that 70 or more genes or loci contribute to AD. One of the major factors limiting progress is that most genetic data have been obtained from non-Hispanic white individuals in Europe and North America, preventing the development of personalized approaches to AD in individuals of other ethnicities. Fortunately, emerging genetic data from other regions - including Africa, Asia, India and South America - are now providing information on the disease from a broader range of ethnicities. Here, we summarize the current knowledge on AD genetics in populations across the world. We predominantly focus on replicated genetic discoveries but also include studies in ethnic groups where replication might not be feasible. We attempt to identify gaps that need to be addressed to achieve a complete picture of the genetic and molecular factors that drive AD in individuals across the globe.
Collapse
Affiliation(s)
- Christiane Reitz
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- The Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Margaret A Pericak-Vance
- The John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- National Centralized Repository for Alzheimer's Disease and Related Dementias, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Richard Mayeux
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
- The Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA.
- Department of Neurology, Columbia University, New York, NY, USA.
- Department of Epidemiology, Columbia University, New York, NY, USA.
| |
Collapse
|
16
|
Pensotti A, Bertolaso M, Bizzarri M. Is Cancer Reversible? Rethinking Carcinogenesis Models-A New Epistemological Tool. Biomolecules 2023; 13:733. [PMID: 37238604 PMCID: PMC10216038 DOI: 10.3390/biom13050733] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
A growing number of studies shows that it is possible to induce a phenotypic transformation of cancer cells from malignant to benign. This process is currently known as "tumor reversion". However, the concept of reversibility hardly fits the current cancer models, according to which gene mutations are considered the primary cause of cancer. Indeed, if gene mutations are causative carcinogenic factors, and if gene mutations are irreversible, how long should cancer be considered as an irreversible process? In fact, there is some evidence that intrinsic plasticity of cancerous cells may be therapeutically exploited to promote a phenotypic reprogramming, both in vitro and in vivo. Not only are studies on tumor reversion highlighting a new, exciting research approach, but they are also pushing science to look for new epistemological tools capable of better modeling cancer.
Collapse
Affiliation(s)
- Andrea Pensotti
- Research Unit of Philosophy of Science and Human Development, University Campus Bio-Medico of Rome, 00128 Rome, Italy
- Systems Biology Group Lab, Department of Experimental Medicine, Sapienza University, 00185 Rome, Italy
| | - Marta Bertolaso
- Research Unit of Philosophy of Science and Human Development, University Campus Bio-Medico of Rome, 00128 Rome, Italy
| | - Mariano Bizzarri
- Systems Biology Group Lab, Department of Experimental Medicine, Sapienza University, 00185 Rome, Italy
| |
Collapse
|
17
|
Pandey D, Onkara Perumal P. A scoping review on deep learning for next-generation RNA-Seq. data analysis. Funct Integr Genomics 2023; 23:134. [PMID: 37084004 DOI: 10.1007/s10142-023-01064-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/24/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
In the last decade, transcriptome research adopting next-generation sequencing (NGS) technologies has gathered incredible momentum amongst functional genomics scientists, particularly amongst clinical/biomedical research groups. The progressive enfoldment/adoption of NGS technologies has incited an abundance of next-generation transcriptomic data harbouring an opulence of new knowledge in public databases. Nevertheless, knowledge discovery from these next-generation RNA-Seq. data analysis necessitates extensive bioinformatics know-how besides elaborate data analysis software packages consistent with the type and context of data analysis. Several reliability and reproducibility concerns continue to impede RNA-Seq. data analysis. Characteristic challenges comprise of data quality, hardware and networking provisions, selection and prioritisation of data analysis tools, and yet significantly implementing of robust machine learning algorithms for maximised exploitation of these experimental transcriptomic data. Over the years, numerous machine learning algorithms have been implemented for improved transcriptomic data analysis executing predominantly shallow learning approaches. More recently, deep learning algorithms are becoming more mainstream, and enactment for next-generation RNA-Seq. data analysis could be revolutionary in the coming years in the biomedical domain. In this scoping review, we attempt to determine the existing literature's size and potential nature in deep learning and NGS RNA-Seq. data analysis. An analysis of the contemporary topics of next-generation RNA-Seq. data analysis based on deep learning algorithms is critically reviewed, emphasising open-source resources.
Collapse
Affiliation(s)
- Diksha Pandey
- Department of Biotechnology, National Institute of Technology, Warangal, Telanga na, 506004, India
| | - P Onkara Perumal
- Department of Biotechnology, National Institute of Technology, Warangal, Telanga na, 506004, India.
| |
Collapse
|
18
|
Kumar A, Das SK, Emdad L, Fisher PB. Applications of tissue-specific and cancer-selective gene promoters for cancer diagnosis and therapy. Adv Cancer Res 2023; 160:253-315. [PMID: 37704290 DOI: 10.1016/bs.acr.2023.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Current treatment of solid tumors with standard of care chemotherapies, radiation therapy and/or immunotherapies are often limited by severe adverse toxic effects, resulting in a narrow therapeutic index. Cancer gene therapy represents a targeted approach that in principle could significantly reduce undesirable side effects in normal tissues while significantly inhibiting tumor growth and progression. To be effective, this strategy requires a clear understanding of the molecular biology of cancer development and evolution and developing biological vectors that can serve as vehicles to target cancer cells. The advent and fine tuning of omics technologies that permit the collective and spatial recognition of genes (genomics), mRNAs (transcriptomics), proteins (proteomics), metabolites (metabolomics), epiomics (epigenomics, epitranscriptomics, and epiproteomics), and their interactomics in defined complex biological samples provide a roadmap for identifying crucial targets of relevance to the cancer paradigm. Combining these strategies with identified genetic elements that control target gene expression uncovers significant opportunities for developing guided gene-based therapeutics for cancer. The purpose of this review is to overview the current state and potential limitations in developing gene promoter-directed targeted expression of key genes and highlights their potential applications in cancer gene therapy.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
19
|
Contributions of Adaptive Laboratory Evolution towards the Enhancement of the Biotechnological Potential of Non-Conventional Yeast Species. J Fungi (Basel) 2023; 9:jof9020186. [PMID: 36836301 PMCID: PMC9964053 DOI: 10.3390/jof9020186] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/19/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Changes in biological properties over several generations, induced by controlling short-term evolutionary processes in the laboratory through selective pressure, and whole-genome re-sequencing, help determine the genetic basis of microorganism's adaptive laboratory evolution (ALE). Due to the versatility of this technique and the imminent urgency for alternatives to petroleum-based strategies, ALE has been actively conducted for several yeasts, primarily using the conventional species Saccharomyces cerevisiae, but also non-conventional yeasts. As a hot topic at the moment since genetically modified organisms are a debatable subject and a global consensus on their employment has not yet been attained, a panoply of new studies employing ALE approaches have emerged and many different applications have been exploited in this context. In the present review, we gathered, for the first time, relevant studies showing the ALE of non-conventional yeast species towards their biotechnological improvement, cataloging them according to the aim of the study, and comparing them considering the species used, the outcome of the experiment, and the employed methodology. This review sheds light on the applicability of ALE as a powerful tool to enhance species features and improve their performance in biotechnology, with emphasis on the non-conventional yeast species, as an alternative or in combination with genome editing approaches.
Collapse
|
20
|
Mead BE, Kummerlowe C, Liu N, Kattan WE, Cheng T, Cheah JH, Soule CK, Peters J, Lowder KE, Blainey PC, Hahn WC, Cleary B, Bryson B, Winter PS, Raghavan S, Shalek AK. Compressed phenotypic screens for complex multicellular models and high-content assays. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.525189. [PMID: 36747859 PMCID: PMC9900857 DOI: 10.1101/2023.01.23.525189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
High-throughput phenotypic screens leveraging biochemical perturbations, high-content readouts, and complex multicellular models could advance therapeutic discovery yet remain constrained by limitations of scale. To address this, we establish a method for compressing screens by pooling perturbations followed by computational deconvolution. Conducting controlled benchmarks with a highly bioactive small molecule library and a high-content imaging readout, we demonstrate increased efficiency for compressed experimental designs compared to conventional approaches. To prove generalizability, we apply compressed screening to examine transcriptional responses of patient-derived pancreatic cancer organoids to a library of tumor-microenvironment (TME)-nominated recombinant protein ligands. Using single-cell RNA-seq as a readout, we uncover reproducible phenotypic shifts induced by ligands that correlate with clinical features in larger datasets and are distinct from reference signatures available in public databases. In sum, our approach enables phenotypic screens that interrogate complex multicellular models with rich phenotypic readouts to advance translatable drug discovery as well as basic biology.
Collapse
Affiliation(s)
- Benjamin E Mead
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
| | - Conner Kummerlowe
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - Nuo Liu
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - Walaa E Kattan
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
| | - Thomas Cheng
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
| | - Jaime H Cheah
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Christian K Soule
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
| | - Josh Peters
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Harvard Medical School; Boston, MA, 02115, USA
| | - Kristen E Lowder
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Paul C Blainey
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - William C Hahn
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Dana Farber Cancer Institute, Boston, MA, 02215, USA
- Harvard Medical School; Boston, MA, 02115, USA
| | - Brian Cleary
- Faculty of Computing and Data Sciences, Department of Biomedical Engineering, Department of Biology, Boston University; Boston, MA, 02215, USA
| | - Bryan Bryson
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - Peter S Winter
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | - Srivatsan Raghavan
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Dana Farber Cancer Institute, Boston, MA, 02215, USA
- Harvard Medical School; Boston, MA, 02115, USA
| | - Alex K Shalek
- Institute for Medical Engineering and Science (IMES), Department of Chemistry, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02142, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA, 02139, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Program in Immunology, Harvard Medical School; Boston, MA, 02115, USA
- Harvard Stem Cell Institute; Cambridge, MA, 02138, USA
| |
Collapse
|
21
|
Campesi I, Ruoppolo M, Franconi F, Caterino M, Costanzo M. Sex-Gender-Based Differences in Metabolic Diseases. Handb Exp Pharmacol 2023; 282:241-257. [PMID: 37528324 DOI: 10.1007/164_2023_683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Sexual dimorphism creates different biological and cellular activities and selective regulation mechanisms in males and females, thus generating differential responses in health and disease. In this scenario, the sex itself is a source of physiologic metabolic disparities that depend on constitutive genetic and epigenetic features that characterize in a specific manner one sex or the other. This has as a direct consequence a huge impact on the metabolic routes that drive the phenotype of an individual. The impact of sex is being clearly recognized also in disease, whereas male and females are more prone to the development of some disorders, or have selective responses to drugs and therapeutic treatments. Actually, very less is known regarding the probable differences guided by sex in the context of inherited metabolic disorders, owing to the scarce consideration of sex in such restricted field, accompanied by an intrinsic bias connected with the rarity of such diseases. Metabolomics technologies have been ultimately developed and adopted for being excellent tools for the investigation of metabolic mechanisms, for marker discovery or monitoring, and for supporting diagnostic procedures of metabolic disorders. Hence, metabolomic approaches can excellently embrace the discovery of sex differences, especially when associated to the outcome or the management of certain inborn errors of the metabolism.
Collapse
Affiliation(s)
- Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
- CEINGE - Biotecnologie Avanzate Franco Salvatore s.c.ar.l., Naples, Italy
| | - Flavia Franconi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, Sassari, Italy
| | - Marianna Caterino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
- CEINGE - Biotecnologie Avanzate Franco Salvatore s.c.ar.l., Naples, Italy
| | - Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.
- CEINGE - Biotecnologie Avanzate Franco Salvatore s.c.ar.l., Naples, Italy.
| |
Collapse
|
22
|
Abstract
The high-dimensional nature of proteomics data presents challenges for statistical analysis and biological interpretation. Multivariate analysis, combined with insightful visualization can help to reveal the underlying patterns in complex biological data. This chapter introduces the R package mixOmics which focuses on data exploration and integration. We first introduce methods for single data sets: both Principal Component Analysis, which can identify the patterns of variance present in data, and sparse Partial Least Squares Discriminant Analysis, which aims to identify variables that can classify samples into known groups. We then present integrative methods with Projection to Latent Structures and further extensions for discriminant analysis. We illustrate each technique on a breast cancer multi-omics study and provide the R code and data as online supplementary material for readers interested in reproducing these analyses.
Collapse
Affiliation(s)
- Zoe Welham
- Kolling Institute, St Leonards, NSW, Australia
| | - Sébastien Déjean
- Institut de Mathématiques de Toulouse, UMR 5219, Université de Toulouse, CNRS, Université Paul Sabatier, Toulouse, France
| | - Kim-Anh Lê Cao
- Melbourne Integrative Genomics, School of Mathematics and Statistics, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
23
|
Systems Biology of Aromatic Compound Catabolism in Facultative Anaerobic Aromatoleum aromaticum EbN1 T. mSystems 2022; 7:e0068522. [PMID: 36445109 PMCID: PMC9765128 DOI: 10.1128/msystems.00685-22] [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] [Indexed: 12/03/2022] Open
Abstract
Members of the genus Aromatoleum thrive in diverse habitats and use a broad range of recalcitrant organic molecules coupled to denitrification or O2 respiration. To gain a holistic understanding of the model organism A. aromaticum EbN1T, we studied its catabolic network dynamics in response to 3-(4-hydroxyphenyl)propanoate, phenylalanine, 3-hydroxybenzoate, benzoate, and acetate utilized under nitrate-reducing versus oxic conditions. Integrated multi-omics (transcriptome, proteome, and metabolome) covered most of the catabolic network (199 genes) and allowed for the refining of knowledge of the degradation modules studied. Their substrate-dependent regulation showed differing degrees of specificity, ranging from high with 3-(4-hydroxyphenyl)propanoate to mostly relaxed with benzoate. For benzoate, the transcript and protein formation were essentially constitutive, contrasted by that of anoxia-specific versus oxia-specific metabolite profiles. The matrix factorization of transcriptomic data revealed that the anaerobic modules accounted for most of the variance across the degradation network. The respiration network appeared to be constitutive, both on the transcript and protein levels, except for nitrate reductase (with narGHI expression occurring only under nitrate-reducing conditions). The anoxia/nitrate-dependent transcription of denitrification genes is apparently controlled by three FNR-type regulators as well as by NarXL (all constitutively formed). The resequencing and functional reannotation of the genome fostered a genome-scale metabolic model, which is comprised of 655 enzyme-catalyzed reactions and 731 distinct metabolites. The model predictions for growth rates and biomass yields agreed well with experimental stoichiometric data, except for 3-(4-hydroxyphenyl)propanoate, with which 4-hydroxybenzoate was exported. Taken together, the combination of multi-omics, growth physiology, and a metabolic model advanced our knowledge of an environmentally relevant microorganism that differs significantly from other bacterial model strains. IMPORTANCE Aromatic compounds are abundant constituents not only of natural organic matter but also of bulk industrial chemicals and fuel components of environmental concern. Considering the widespread occurrence of redox gradients in the biosphere, facultative anaerobic degradation specialists can be assumed to play a prominent role in the natural mineralization of organic matter and in bioremediation at contaminated sites. Surprisingly, differential multi-omics profiling of the A. aromaticum EbN1T studied here revealed relaxed regulatory stringency across its four main physiological modi operandi (i.e., O2-independent and O2-dependent degradation reactions versus denitrification and O2 respiration). Combining multi-omics analyses with a genome-scale metabolic model aligned with measured growth performances establishes A. aromaticum EbN1T as a systems-biology model organism and provides unprecedented insights into how this bacterium functions on a holistic level. Moreover, this experimental platform invites future studies on eco-systems and synthetic biology of the environmentally relevant betaproteobacterial Aromatoleum/Azoarcus/Thauera cluster.
Collapse
|
24
|
Romo E, Torres M, Martin-Solano S. Current situation of snakebites envenomation in the Neotropics: Biotechnology, a versatile tool in the production of antivenoms. BIONATURA 2022. [DOI: 10.21931/rb/2022.07.04.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Snakebite envenomation is a neglected tropical disease that affects millions of people around the world with a great impact on health and the economy. Unfortunately, public health programs do not include this kind of disease as a priority in their social programs. Cases of snakebite envenomations in the Neotropics are inaccurate due to inadequate disease management from medical records to the choice of treatments. Victims of snakebite envenomation are primarily found in impoverished agricultural areas where remote conditions limit the availability of antivenom. Antivenom serum is the only Food and Drug Administration-approved treatment used up to date. However, it has several disadvantages in terms of safety and effectiveness. This review provides a comprehensive insight dealing with the current epidemiological status of snakebites in the Neotropics and technologies employed in antivenom production. Also, modern biotechnological tools such as transcriptomic, proteomic, immunogenic, high-density peptide microarray and epitope mapping are highlighted for producing new-generation antivenom sera. These results allow us to propose strategic solutions in the Public Health Sector for managing this disease.
Keywords: antivenom, biotechnology, neglected tropical disease, omics, recombinant antibody.
Collapse
Affiliation(s)
- Elizabeth Romo
- Carrera de Ingeniería en Biotecnología, Departamento de Ciencias de la Vida y la Agricultura, Universidad de las Fuerzas Armadas-ESPE, Sangolquí, Ecuador
| | - Marbel Torres
- Carrera de Ingeniería en Biotecnología, Departamento de Ciencias de la Vida y la Agricultura, Universidad de las Fuerzas Armadas-ESPE, Sangolquí, Ecuador, Grupo de Investigación en Sanidad Animal y Humana (GISAH), Carrera de Ingeniería en Biotecnología, Departamento de Ciencias de la Vida y la Agricultura, Universidad de las Fuerzas Armadas-ESPE, Immunology and Virology Laboratory, Nanoscience and Nanotechnology Center, Universidad de las Fuerzas Armadas, ESPE, Sangolquí, Ecuador
| | - Sarah Martin-Solano
- Carrera de Ingeniería en Biotecnología, Departamento de Ciencias de la Vida y la Agricultura, Universidad de las Fuerzas Armadas-ESPE, Sangolquí, Ecuador, Grupo de Investigación en Sanidad Animal y Humana (GISAH), Carrera de Ingeniería en Biotecnología, Departamento de Ciencias de la Vida y la Agricultura, Universidad de las Fuerzas Armadas-ESPE, Grupo de Investigación en Biodiversidad, Zoonosis y Salud Pública, Universidad Central del Ecuador
| |
Collapse
|
25
|
Franks AM. Reducing subspace models for large-scale covariance regression. Biometrics 2022; 78:1604-1613. [PMID: 34458980 DOI: 10.1111/biom.13531] [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: 08/05/2020] [Revised: 06/29/2021] [Accepted: 07/08/2021] [Indexed: 12/30/2022]
Abstract
We develop an envelope model for joint mean and covariance regression in the large p, small n setting. In contrast to existing envelope methods, which improve mean estimates by incorporating estimates of the covariance structure, we focus on identifying covariance heterogeneity by incorporating information about mean-level differences. We use a Monte Carlo EM algorithm to identify a low-dimensional subspace that explains differences in both means and covariances as a function of covariates, and then use MCMC to estimate the posterior uncertainty conditional on the inferred low-dimensional subspace. We demonstrate the utility of our model on a motivating application on the metabolomics of aging. We also provide R code that can be used to develop and test other generalizations of the response envelope model.
Collapse
Affiliation(s)
- Alexander M Franks
- Department of Statistics and Applied Probability, University of California Santa Barbara, Santa Barbara, California, USA
| |
Collapse
|
26
|
Du YH, Wang MY, Yang LH, Tong LL, Guo DS, Ji XJ. Optimization and Scale-Up of Fermentation Processes Driven by Models. Bioengineering (Basel) 2022; 9:bioengineering9090473. [PMID: 36135019 PMCID: PMC9495923 DOI: 10.3390/bioengineering9090473] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/05/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
In the era of sustainable development, the use of cell factories to produce various compounds by fermentation has attracted extensive attention; however, industrial fermentation requires not only efficient production strains, but also suitable extracellular conditions and medium components, as well as scaling-up. In this regard, the use of biological models has received much attention, and this review will provide guidance for the rapid selection of biological models. This paper first introduces two mechanistic modeling methods, kinetic modeling and constraint-based modeling (CBM), and generalizes their applications in practice. Next, we review data-driven modeling based on machine learning (ML), and highlight the application scope of different learning algorithms. The combined use of ML and CBM for constructing hybrid models is further discussed. At the end, we also discuss the recent strategies for predicting bioreactor scale-up and culture behavior through a combination of biological models and computational fluid dynamics (CFD) models.
Collapse
Affiliation(s)
- Yuan-Hang Du
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Min-Yu Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Lin-Hui Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Ling-Ling Tong
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Dong-Sheng Guo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
- Correspondence: (D.-S.G.); (X.-J.J.)
| | - Xiao-Jun Ji
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
- Correspondence: (D.-S.G.); (X.-J.J.)
| |
Collapse
|
27
|
Nam SE, Bae DY, Ki JS, Ahn CY, Rhee JS. The importance of multi-omics approaches for the health assessment of freshwater ecosystems. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00286-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
28
|
Dutta B, Lahiri D, Nag M, Abukhader R, Sarkar T, Pati S, Upadhye V, Pandit S, Amin MFM, Al Tawaha ARMS, Kumar M, Ray RR. Multi-Omics Approach in Amelioration of Food Products. Front Microbiol 2022; 13:955683. [PMID: 35903478 PMCID: PMC9315205 DOI: 10.3389/fmicb.2022.955683] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Determination of the quality of food products is an essential key factor needed for safe-guarding the quality of food for the interest of the consumers, along with the nutritional and sensory improvements that are necessary for delivering better quality products. Bacteriocins are a group of ribosomally synthesized antimicrobial peptides that help in maintaining the quality of food. The implementation of multi-omics approach has been important for the overall enhancement of the quality of the food. This review uses various recent technologies like proteomics, transcriptomics, and metabolomics for the overall enhancement of the quality of food products. The matrix associated with the food products requires the use of sophisticated technologies that help in the extraction of a large amount of information necessary for the amelioration of the food products. This review would provide a wholesome view of how various recent technologies can be used for improving the quality food products and for enhancing their shelf-life.
Collapse
Affiliation(s)
- Bandita Dutta
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, India
| | - Dibyajit Lahiri
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Moupriya Nag
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Rose Abukhader
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Tanmay Sarkar
- Department of Food Processing Technology, Malda Polytechnic, West Bengal State Council of Technical Education, Government of West Bengal, Malda, India
| | - Siddhartha Pati
- NatNov Bioscience Private Limited, Balasore, India
- Skills Innovation & Academic Network (SIAN) Institute, Association for Biodiversity Conservation & Research (ABC), Balasore, India
| | - Vijay Upadhye
- Center of Research for Development (CR4D), Parul Institute of Applied Sciences (PIAS), Parul University, Vadodara, India
| | - Soumya Pandit
- Department of Life Sciences, Sharda University, Noida, India
| | | | | | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR-Central Institute for Research on Cotton Technology, Mumbai, India
| | - Rina Rani Ray
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, India
| |
Collapse
|
29
|
Régimbeau A, Budinich M, Larhlimi A, Pierella Karlusich JJ, Aumont O, Memery L, Bowler C, Eveillard D. Contribution of genome-scale metabolic modelling to niche theory. Ecol Lett 2022; 25:1352-1364. [PMID: 35384214 PMCID: PMC9324083 DOI: 10.1111/ele.13954] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/22/2021] [Accepted: 11/28/2021] [Indexed: 12/22/2022]
Abstract
Standard niche modelling is based on probabilistic inference from organismal occurrence data but does not benefit yet from genome‐scale descriptions of these organisms. This study overcomes this shortcoming by proposing a new conceptual niche that resumes the whole metabolic capabilities of an organism. The so‐called metabolic niche resumes well‐known traits such as nutrient needs and their dependencies for survival. Despite the computational challenge, its implementation allows the detection of traits and the formal comparison of niches of different organisms, emphasising that the presence–absence of functional genes is not enough to approximate the phenotype. Further statistical exploration of an organism's niche sheds light on genes essential for the metabolic niche and their role in understanding various biological experiments, such as transcriptomics, paving the way for incorporating better genome‐scale description in ecological studies.
Collapse
Affiliation(s)
| | | | | | - Juan José Pierella Karlusich
- Département de Biologie, Institut de Biologie de l'ENS, École Normale supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Olivier Aumont
- Laboratoire d'Océanographie et du Climat: Expérimentations et Approches Numériques (LOCEAN), IRD-IPSL, Paris, France
| | - Laurent Memery
- Université de Brest (UBO), CNRS, IRD, Ifremer, Laboratoire des Sciences de l'Environnement Marin, Plouzané, France
| | - Chris Bowler
- Département de Biologie, Institut de Biologie de l'ENS, École Normale supérieure, CNRS, INSERM, Université PSL, Paris, France.,Research Federation for the study of Global Ocean Systems Ecology and Evolution, FR2022/Tara GO-SEE, Paris, France
| | - Damien Eveillard
- Université de Nantes, CNRS, LS2N, Nantes, France.,Research Federation for the study of Global Ocean Systems Ecology and Evolution, FR2022/Tara GO-SEE, Paris, France
| |
Collapse
|
30
|
Burks DJ, Sengupta S, De R, Mittler R, Azad RK. The Arabidopsis gene co-expression network. PLANT DIRECT 2022; 6:e396. [PMID: 35492683 PMCID: PMC9039629 DOI: 10.1002/pld3.396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 06/14/2023]
Abstract
Identifying genes that interact to confer a biological function to an organism is one of the main goals of functional genomics. High-throughput technologies for assessment and quantification of genome-wide gene expression patterns have enabled systems-level analyses to infer pathways or networks of genes involved in different functions under many different conditions. Here, we leveraged the publicly available, information-rich RNA-Seq datasets of the model plant Arabidopsis thaliana to construct a gene co-expression network, which was partitioned into clusters or modules that harbor genes correlated by expression. Gene ontology and pathway enrichment analyses were performed to assess functional terms and pathways that were enriched within the different gene modules. By interrogating the co-expression network for genes in different modules that associate with a gene of interest, diverse functional roles of the gene can be deciphered. By mapping genes differentially expressing under a certain condition in Arabidopsis onto the co-expression network, we demonstrate the ability of the network to uncover novel genes that are likely transcriptionally active but prone to be missed by standard statistical approaches due to their falling outside of the confidence zone of detection. To our knowledge, this is the first A. thaliana co-expression network constructed using the entire mRNA-Seq datasets (>20,000) available at the NCBI SRA database. The developed network can serve as a useful resource for the Arabidopsis research community to interrogate specific genes of interest within the network, retrieve the respective interactomes, decipher gene modules that are transcriptionally altered under certain condition or stage, and gain understanding of gene functions.
Collapse
Affiliation(s)
- David J. Burks
- Department of Biological Sciences and BioDiscovery Institute, College of ScienceUniversity of North TexasDentonTexasUSA
| | - Soham Sengupta
- Department of Biological Sciences and BioDiscovery Institute, College of ScienceUniversity of North TexasDentonTexasUSA
| | - Ronika De
- Department of Biological Sciences and BioDiscovery Institute, College of ScienceUniversity of North TexasDentonTexasUSA
| | - Ron Mittler
- The Division of Plant Sciences and Interdisciplinary Plant Group, College of Agriculture, Food and Natural ResourcesChristopher S. Bond Life Sciences Center University of MissouriColumbiaMissouriUSA
- Department of SurgeryUniversity of Missouri School of MedicineColumbiaMissouriUSA
| | - Rajeev K. Azad
- Department of Biological Sciences and BioDiscovery Institute, College of ScienceUniversity of North TexasDentonTexasUSA
- Department of MathematicsUniversity of North TexasDentonTexasUSA
| |
Collapse
|
31
|
OUP accepted manuscript. Brief Funct Genomics 2022; 21:243-269. [DOI: 10.1093/bfgp/elac007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/14/2022] Open
|
32
|
Alzubaidi A, Tepper J. Deep Mining from Omics Data. Methods Mol Biol 2022; 2449:349-386. [PMID: 35507271 DOI: 10.1007/978-1-0716-2095-3_15] [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] [Indexed: 06/14/2023]
Abstract
Since the advent of high-throughput omics technologies, various molecular data such as genes, transcripts, proteins, and metabolites have been made widely available to researchers. This has afforded clinicians, bioinformaticians, statisticians, and data scientists the opportunity to apply their innovations in feature mining and predictive modeling to a rich data resource to develop a wide range of generalizable prediction models. What has become apparent over the last 10 years is that researchers have adopted deep neural networks (or "deep nets") as their preferred paradigm of choice for complex data modeling due to the superiority of performance over more traditional statistical machine learning approaches, such as support vector machines. A key stumbling block, however, is that deep nets inherently lack transparency and are considered to be a "black box" approach. This naturally makes it very difficult for clinicians and other stakeholders to trust their deep learning models even though the model predictions appear to be highly accurate. In this chapter, we therefore provide a detailed summary of the deep net architectures typically used in omics research, together with a comprehensive summary of the notable "deep feature mining" techniques researchers have applied to open up this black box and provide some insights into the salient input features and why these models behave as they do. We group these techniques into the following three categories: (a) hidden layer visualization and interpretation; (b) input feature importance and impact evaluation; and (c) output layer gradient analysis. While we find that omics researchers have made some considerable gains in opening up the black box through interpretation of the hidden layer weights and node activations to identify salient input features, we highlight other approaches for omics researchers, such as employing deconvolutional network-based approaches and development of bespoke attribute impact measures to enable researchers to better understand the relationships between the input data and hidden layer representations formed and thus the output behavior of their deep nets.
Collapse
Affiliation(s)
- Abeer Alzubaidi
- School of Science and Technology, Department of Computer Science, Nottingham Trent University, Nottingham, UK.
| | | |
Collapse
|
33
|
Begum N, Harzandi A, Lee S, Uhlen M, Moyes DL, Shoaie S. Host-mycobiome metabolic interactions in health and disease. Gut Microbes 2022; 14:2121576. [PMID: 36151873 PMCID: PMC9519009 DOI: 10.1080/19490976.2022.2121576] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/31/2022] [Accepted: 08/31/2022] [Indexed: 02/04/2023] Open
Abstract
Fungal communities (mycobiome) have an important role in sustaining the resilience of complex microbial communities and maintenance of homeostasis. The mycobiome remains relatively unexplored compared to the bacteriome despite increasing evidence highlighting their contribution to host-microbiome interactions in health and disease. Despite being a small proportion of the total species, fungi constitute a large proportion of the biomass within the human microbiome and thus serve as a potential target for metabolic reprogramming in pathogenesis and disease mechanism. Metabolites produced by fungi shape host niches, induce immune tolerance and changes in their levels prelude changes associated with metabolic diseases and cancer. Given the complexity of microbial interactions, studying the metabolic interplay of the mycobiome with both host and microbiome is a demanding but crucial task. However, genome-scale modelling and synthetic biology can provide an integrative platform that allows elucidation of the multifaceted interactions between mycobiome, microbiome and host. The inferences gained from understanding mycobiome interplay with other organisms can delineate the key role of the mycobiome in pathophysiology and reveal its role in human disease.
Collapse
Affiliation(s)
- Neelu Begum
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Azadeh Harzandi
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Sunjae Lee
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Mathias Uhlen
- Science for Life Laboratory, KTH–Royal Institute of Technology, Stockholm, Sweden
| | - David L. Moyes
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Saeed Shoaie
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
- Science for Life Laboratory, KTH–Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
34
|
Vijayakumar S, Magazzù G, Moon P, Occhipinti A, Angione C. A Practical Guide to Integrating Multimodal Machine Learning and Metabolic Modeling. Methods Mol Biol 2022; 2399:87-122. [PMID: 35604554 DOI: 10.1007/978-1-0716-1831-8_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Complex, distributed, and dynamic sets of clinical biomedical data are collectively referred to as multimodal clinical data. In order to accommodate the volume and heterogeneity of such diverse data types and aid in their interpretation when they are combined with a multi-scale predictive model, machine learning is a useful tool that can be wielded to deconstruct biological complexity and extract relevant outputs. Additionally, genome-scale metabolic models (GSMMs) are one of the main frameworks striving to bridge the gap between genotype and phenotype by incorporating prior biological knowledge into mechanistic models. Consequently, the utilization of GSMMs as a foundation for the integration of multi-omic data originating from different domains is a valuable pursuit towards refining predictions. In this chapter, we show how cancer multi-omic data can be analyzed via multimodal machine learning and metabolic modeling. Firstly, we focus on the merits of adopting an integrative systems biology led approach to biomedical data mining. Following this, we propose how constraint-based metabolic models can provide a stable yet adaptable foundation for the integration of multimodal data with machine learning. Finally, we provide a step-by-step tutorial for the combination of machine learning and GSMMs, which includes: (i) tissue-specific constraint-based modeling; (ii) survival analysis using time-to-event prediction for cancer; and (iii) classification and regression approaches for multimodal machine learning. The code associated with the tutorial can be found at https://github.com/Angione-Lab/Tutorials_Combining_ML_and_GSMM .
Collapse
Affiliation(s)
- Supreeta Vijayakumar
- Computational Systems Biology and Data Analytics Research Group, Teesside University, Middlebrough, UK
| | - Giuseppe Magazzù
- Computational Systems Biology and Data Analytics Research Group, Teesside University, Middlebrough, UK
| | - Pradip Moon
- Computational Systems Biology and Data Analytics Research Group, Teesside University, Middlebrough, UK
| | - Annalisa Occhipinti
- Computational Systems Biology and Data Analytics Research Group, Middlebrough, UK
- Centre for Digital Innovation, Teesside University, Middlesbrough, UK
| | - Claudio Angione
- Computational Systems Biology and Data Analytics Research Group, Teesside University, Middlebrough, UK.
- Centre for Digital Innovation, Teesside University, Middlesbrough, UK.
- Healthcare Innovation Centre, Teesside University, Middlesbrough, UK.
| |
Collapse
|
35
|
Francisco FR, Aono AH, da Silva CC, Gonçalves PS, Scaloppi Junior EJ, Le Guen V, Fritsche-Neto R, Souza LM, de Souza AP. Unravelling Rubber Tree Growth by Integrating GWAS and Biological Network-Based Approaches. FRONTIERS IN PLANT SCIENCE 2021; 12:768589. [PMID: 34992619 PMCID: PMC8724537 DOI: 10.3389/fpls.2021.768589] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/02/2021] [Indexed: 06/08/2023]
Abstract
Hevea brasiliensis (rubber tree) is a large tree species of the Euphorbiaceae family with inestimable economic importance. Rubber tree breeding programs currently aim to improve growth and production, and the use of early genotype selection technologies can accelerate such processes, mainly with the incorporation of genomic tools, such as marker-assisted selection (MAS). However, few quantitative trait loci (QTLs) have been used successfully in MAS for complex characteristics. Recent research shows the efficiency of genome-wide association studies (GWAS) for locating QTL regions in different populations. In this way, the integration of GWAS, RNA-sequencing (RNA-Seq) methodologies, coexpression networks and enzyme networks can provide a better understanding of the molecular relationships involved in the definition of the phenotypes of interest, supplying research support for the development of appropriate genomic based strategies for breeding. In this context, this work presents the potential of using combined multiomics to decipher the mechanisms of genotype and phenotype associations involved in the growth of rubber trees. Using GWAS from a genotyping-by-sequencing (GBS) Hevea population, we were able to identify molecular markers in QTL regions with a main effect on rubber tree plant growth under constant water stress. The underlying genes were evaluated and incorporated into a gene coexpression network modelled with an assembled RNA-Seq-based transcriptome of the species, where novel gene relationships were estimated and evaluated through in silico methodologies, including an estimated enzymatic network. From all these analyses, we were able to estimate not only the main genes involved in defining the phenotype but also the interactions between a core of genes related to rubber tree growth at the transcriptional and translational levels. This work was the first to integrate multiomics analysis into the in-depth investigation of rubber tree plant growth, producing useful data for future genetic studies in the species and enhancing the efficiency of the species improvement programs.
Collapse
Affiliation(s)
- Felipe Roberto Francisco
- Molecular Biology and Genetic Engineering Center (CBMEG), University of Campinas (UNICAMP), Campinas, Brazil
| | - Alexandre Hild Aono
- Molecular Biology and Genetic Engineering Center (CBMEG), University of Campinas (UNICAMP), Campinas, Brazil
| | - Carla Cristina da Silva
- Molecular Biology and Genetic Engineering Center (CBMEG), University of Campinas (UNICAMP), Campinas, Brazil
| | - Paulo S. Gonçalves
- Center of Rubber Tree and Agroforestry Systems, Agronomic Institute (IAC), Votuporanga, Brazil
| | | | - Vincent Le Guen
- Centre de Coopération Internationale en Recherche Agronomique pour le Développement (CIRAD), UMR AGAP, Montpellier, France
- AGAP, Univ Montpellier, CIRAD, INRAE, Institut Agro, Montpellier, France
| | - Roberto Fritsche-Neto
- Department of Genetics, Luiz de Queiroz College of Agriculture (ESALQ), University of São Paulo (USP), Piracicaba, Brazil
| | - Livia Moura Souza
- Molecular Biology and Genetic Engineering Center (CBMEG), University of Campinas (UNICAMP), Campinas, Brazil
- São Francisco University (USF), Itatiba, Brazil
| | - Anete Pereira de Souza
- Molecular Biology and Genetic Engineering Center (CBMEG), University of Campinas (UNICAMP), Campinas, Brazil
- Department of Plant Biology, Biology Institute, University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
36
|
Minami Y, Yuan Y, Ueda HR. Towards organism-level systems biology by next-generation genetics and whole-organ cell profiling. Biophys Rev 2021; 13:1113-1126. [PMID: 35059031 PMCID: PMC8724464 DOI: 10.1007/s12551-021-00859-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023] Open
Abstract
The system-level identification and analysis of molecular and cellular networks in mammals can be accelerated by "next-generation" genetics, which is defined as genetics that can achieve desired genetic makeup in a single generation without any animal crossing. We recently established a highly efficient procedure for producing knock-out (KO) mice using the "Triple-CRISPR" method, which targets a single gene by triple gRNAs in the CRISPR/Cas9 system. This procedure achieved an almost perfect KO efficiency (96-100%). We also established a highly efficient procedure, the "ES-mouse" method, for producing knock-in (KI) mice within a single generation. In this method, ES cells were treated with three inhibitors to keep their potency and then injected into 8-cell-stage embryos. These procedures dramatically shortened the time required to produce KO or KI mice from years down to about 3 months. The produced KO and KI mice can also be systematically profiled at a single-cell resolution by the "whole-organ cell profiling," which was realized by tissue-clearing methods, such as CUBIC, and an advanced light-sheet microscopy. The review describes the establishment and application of these technologies above in analyzing the three states (NREM sleep, REM sleep, and awake) of mammalian brains. It also discusses the role of calcium and muscarinic receptors in these states as well as the current challenges and future opportunities in the next-generation mammalian genetics and whole-organ cell profiling for organism-level systems biology.
Collapse
Affiliation(s)
- Yoichi Minami
- Department of Systems Pharmacology, Graduate School of Medicine, the University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Yufei Yuan
- Department of Systems Pharmacology, Graduate School of Medicine, the University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Hiroki R. Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, the University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033 Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, 1-3 Yamadaoka, Suita, Osaka 565-0871 Japan
| |
Collapse
|
37
|
|
38
|
Castro LHE, Sant'Anna CMR. Molecular Modeling Techniques Applied to the Design of Multitarget Drugs: Methods and Applications. Curr Top Med Chem 2021; 22:333-346. [PMID: 34844540 DOI: 10.2174/1568026621666211129140958] [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: 07/01/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022]
Abstract
Multifactorial diseases, such as cancer and diabetes present a challenge for the traditional "one-target, one disease" paradigm due to their complex pathogenic mechanisms. Although a combination of drugs can be used, a multitarget drug may be a better choice face of its efficacy, lower adverse effects and lower chance of resistance development. The computer-based design of these multitarget drugs can explore the same techniques used for single-target drug design, but the difficulties associated to the obtention of drugs that are capable of modulating two or more targets with similar efficacy impose new challenges, whose solutions involve the adaptation of known techniques and also to the development of new ones, including machine-learning approaches. In this review, some SBDD and LBDD techniques for the multitarget drug design are discussed, together with some cases where the application of such techniques led to effective multitarget ligands.
Collapse
Affiliation(s)
| | - Carlos Mauricio R Sant'Anna
- Programa de Pós-Graduação em Química, Instituto de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica. Brazil
| |
Collapse
|
39
|
Wu MX, Zou Y, Yu YH, Chen BX, Zheng QW, Ye ZW, Wei T, Ye SQ, Guo LQ, Lin JF. Comparative transcriptome and proteome provide new insights into the regulatory mechanisms of the postharvest deterioration of Pleurotus tuoliensis fruitbodies during storage. Food Res Int 2021; 147:110540. [PMID: 34399517 DOI: 10.1016/j.foodres.2021.110540] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/05/2021] [Accepted: 06/14/2021] [Indexed: 10/21/2022]
Abstract
The Pleurotus tuoliensis (Pt), a precious edible mushroom with high economic value, is widely popular for its rich nutrition and meaty texture. However, rapid postharvest deterioration depreciates the commercial value of Pt and severely restricts its marketing. By RNA-Seq transcriptomic and TMT-MS MS proteomic, we study the regulatory mechanisms of the postharvest storage of Pt fruitbodies at 25 ℃ for 0, 38, and 76 h (these three-time points recorded as groups A, B, and C, respectively). 2,008 DEGs (Differentially expressed genes) were identified, and all DEGs shared 265 factors with all DEPs (Differentially expressed proteins). Jointly, the DEGs and DEPs of two-omics showed that the category of the metabolic process contained the most DEGs and DEPs in the biological process by GO (Gene Ontology) classification. The top 17 KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways with the highest sum of DEG and DEP numbers in groups B/A (38 h vs. 0 h) and C/A (76 h vs. 0 h) and pathways closely related to energy metabolism were selected for analysis and discussion. Actively expression of CAZymes (Carbohydrate active enzymes), represented by laccase, chitinase, and β-glucanase, directly leads to the softening of fruitbodies. The transcription factor Rlm1 of 1,3-β-glucan synthase attracted attention with a significant down-regulation of gene levels in the C/A group. Laccase also contributes, together with phenylalanine ammonia-lyase (PAL), to the discoloration reaction in the first 76 h of the fruitbodies. Significant expression of several crucial enzymes for EMP (Glycolysis), Fatty acid degradation, and Valine, leucine and isoleucine degradation at the gene or protein level supply substantial amounts of acetyl-CoA to the TCA cycle. Citrate synthase (CS), isocitrate dehydrogenase (ICDH), and three mitochondrial respiratory complexes intensify respiration and produce high levels of ROS (Reactive oxygen species) by significant up-regulation. In the ROS scavenging system, only Mn-SOD was significantly up-regulated at the gene level and was probably interacted with Hsp60 (Heat shock protein 60), which was significantly up-regulated at the protein level, to play a dominant role in antioxidation. Three types of stresses - cell wall stress, starvation, and oxidative stress - were suffered by Pt fruitbodies postharvest, resulting in cell cycle arrest and gene expression disorder.
Collapse
Affiliation(s)
- Mu-Xiu Wu
- College of Food Science, South China Agricultural University, Guangzhou 510642, China; Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510642, China
| | - Yuan Zou
- College of Food Science, South China Agricultural University, Guangzhou 510642, China; Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510642, China
| | - Ying-Hao Yu
- College of Food Science, South China Agricultural University, Guangzhou 510642, China; Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510642, China
| | - Bai-Xiong Chen
- College of Food Science, South China Agricultural University, Guangzhou 510642, China; Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510642, China
| | - Qian-Wang Zheng
- College of Food Science, South China Agricultural University, Guangzhou 510642, China; Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510642, China
| | - Zhi-Wei Ye
- College of Food Science, South China Agricultural University, Guangzhou 510642, China; Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510642, China
| | - Tao Wei
- College of Food Science, South China Agricultural University, Guangzhou 510642, China; Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510642, China
| | - Si-Qiang Ye
- College of Food Science, South China Agricultural University, Guangzhou 510642, China; Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510642, China
| | - Li-Qiong Guo
- College of Food Science, South China Agricultural University, Guangzhou 510642, China; Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510642, China.
| | - Jun-Fang Lin
- College of Food Science, South China Agricultural University, Guangzhou 510642, China; Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510642, China.
| |
Collapse
|
40
|
Integrated Inference of Asymmetric Protein Interaction Networks Using Dynamic Model and Individual Patient Proteomics Data. Symmetry (Basel) 2021. [DOI: 10.3390/sym13061097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Recent advances in experimental biology studies have produced large amount of molecular activity data. In particular, individual patient data provide non-time series information for the molecular activities in disease conditions. The challenge is how to design effective algorithms to infer regulatory networks using the individual patient datasets and consequently address the issue of network symmetry. This work is aimed at developing an efficient pipeline to reverse-engineer regulatory networks based on the individual patient proteomic data. The first step uses the SCOUT algorithm to infer the pseudo-time trajectory of individual patients. Then the path-consistent method with part mutual information is used to construct a static network that contains the potential protein interactions. To address the issue of network symmetry in terms of undirected symmetric network, a dynamic model of ordinary differential equations is used to further remove false interactions to derive asymmetric networks. In this work a dataset from triple-negative breast cancer patients is used to develop a protein-protein interaction network with 15 proteins.
Collapse
|
41
|
Wang Y, Zhou M, Zou Q, Xu L. Machine learning for phytopathology: from the molecular scale towards the network scale. Brief Bioinform 2021; 22:6204793. [PMID: 33787847 DOI: 10.1093/bib/bbab037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/09/2021] [Accepted: 01/26/2021] [Indexed: 01/16/2023] Open
Abstract
With the increasing volume of high-throughput sequencing data from a variety of omics techniques in the field of plant-pathogen interactions, sorting, retrieving, processing and visualizing biological information have become a great challenge. Within the explosion of data, machine learning offers powerful tools to process these complex omics data by various algorithms, such as Bayesian reasoning, support vector machine and random forest. Here, we introduce the basic frameworks of machine learning in dissecting plant-pathogen interactions and discuss the applications and advances of machine learning in plant-pathogen interactions from molecular to network biology, including the prediction of pathogen effectors, plant disease resistance protein monitoring and the discovery of protein-protein networks. The aim of this review is to provide a summary of advances in plant defense and pathogen infection and to indicate the important developments of machine learning in phytopathology.
Collapse
Affiliation(s)
- Yansu Wang
- Postdoctoral Innovation Practice Base, Shenzhen Polytechnic, China
| | | | - Quan Zou
- University of Electronic Science and Technology of China
| | - Lei Xu
- Shenzhen Polytechnic, China
| |
Collapse
|
42
|
Xu L, Pierroz G, Wipf HML, Gao C, Taylor JW, Lemaux PG, Coleman-Derr D. Holo-omics for deciphering plant-microbiome interactions. MICROBIOME 2021; 9:69. [PMID: 33762001 PMCID: PMC7988928 DOI: 10.1186/s40168-021-01014-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/02/2021] [Indexed: 05/02/2023]
Abstract
Host-microbiome interactions are recognized for their importance to host health. An improved understanding of the molecular underpinnings of host-microbiome relationships will advance our capacity to accurately predict host fitness and manipulate interaction outcomes. Within the plant microbiome research field, unlocking the functional relationships between plants and their microbial partners is the next step to effectively using the microbiome to improve plant fitness. We propose that strategies that pair host and microbial datasets-referred to here as holo-omics-provide a powerful approach for hypothesis development and advancement in this area. We discuss several experimental design considerations and present a case study to highlight the potential for holo-omics to generate a more holistic perspective of molecular networks within the plant microbiome system. In addition, we discuss the biggest challenges for conducting holo-omics studies; specifically, the lack of vetted analytical frameworks, publicly available tools, and required technical expertise to process and integrate heterogeneous data. Finally, we conclude with a perspective on appropriate use-cases for holo-omics studies, the need for downstream validation, and new experimental techniques that hold promise for the plant microbiome research field. We argue that utilizing a holo-omics approach to characterize host-microbiome interactions can provide important opportunities for broadening system-level understandings and significantly inform microbial approaches to improving host health and fitness. Video abstract.
Collapse
Affiliation(s)
- Ling Xu
- Department of Plant and Microbial Biology, University of California, Berkeley, CA USA
| | - Grady Pierroz
- Department of Plant and Microbial Biology, University of California, Berkeley, CA USA
| | - Heidi M.-L. Wipf
- Department of Plant and Microbial Biology, University of California, Berkeley, CA USA
| | - Cheng Gao
- Department of Plant and Microbial Biology, University of California, Berkeley, CA USA
| | - John W. Taylor
- Department of Plant and Microbial Biology, University of California, Berkeley, CA USA
| | - Peggy G. Lemaux
- Department of Plant and Microbial Biology, University of California, Berkeley, CA USA
| | - Devin Coleman-Derr
- Department of Plant and Microbial Biology, University of California, Berkeley, CA USA
- Plant Gene Expression Center, USDA-ARS, Albany, CA USA
| |
Collapse
|
43
|
Zhou M, Varol A, Efferth T. Multi-omics approaches to improve malaria therapy. Pharmacol Res 2021; 167:105570. [PMID: 33766628 DOI: 10.1016/j.phrs.2021.105570] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/02/2021] [Accepted: 03/16/2021] [Indexed: 01/07/2023]
Abstract
Malaria contributes to the most widespread infectious diseases worldwide. Even though current drugs are commercially available, the ever-increasing drug resistance problem by malaria parasites poses new challenges in malaria therapy. Hence, searching for efficient therapeutic strategies is of high priority in malaria control. In recent years, multi-omics technologies have been extensively applied to provide a more holistic view of functional principles and dynamics of biological mechanisms. We briefly review multi-omics technologies and focus on recent malaria progress conducted with the help of various omics methods. Then, we present up-to-date advances for multi-omics approaches in malaria. Next, we describe resistance phenomena to established antimalarial drugs and underlying mechanisms. Finally, we provide insight into novel multi-omics approaches, new drugs and vaccine developments and analyze current gaps in multi-omics research. Although multi-omics approaches have been successfully used in malaria studies, they are still limited. Many gaps need to be filled to bridge the gap between basic research and treatment of malaria patients. Multi-omics approaches will foster a better understanding of the molecular mechanisms of Plasmodium that are essential for the development of novel drugs and vaccines to fight this disastrous disease.
Collapse
Affiliation(s)
- Min Zhou
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Ayşegül Varol
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany.
| |
Collapse
|
44
|
Jendoubi T. Approaches to Integrating Metabolomics and Multi-Omics Data: A Primer. Metabolites 2021; 11:184. [PMID: 33801081 PMCID: PMC8003953 DOI: 10.3390/metabo11030184] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolomics deals with multiple and complex chemical reactions within living organisms and how these are influenced by external or internal perturbations. It lies at the heart of omics profiling technologies not only as the underlying biochemical layer that reflects information expressed by the genome, the transcriptome and the proteome, but also as the closest layer to the phenome. The combination of metabolomics data with the information available from genomics, transcriptomics, and proteomics offers unprecedented possibilities to enhance current understanding of biological functions, elucidate their underlying mechanisms and uncover hidden associations between omics variables. As a result, a vast array of computational tools have been developed to assist with integrative analysis of metabolomics data with different omics. Here, we review and propose five criteria-hypothesis, data types, strategies, study design and study focus- to classify statistical multi-omics data integration approaches into state-of-the-art classes under which all existing statistical methods fall. The purpose of this review is to look at various aspects that lead the choice of the statistical integrative analysis pipeline in terms of the different classes. We will draw particular attention to metabolomics and genomics data to assist those new to this field in the choice of the integrative analysis pipeline.
Collapse
Affiliation(s)
- Takoua Jendoubi
- Department of Statistical Science, University College London, London WC1E 6BT, UK
| |
Collapse
|
45
|
Compendium of Methods to Uncover RNA-Protein Interactions In Vivo. Methods Protoc 2021; 4:mps4010022. [PMID: 33808611 PMCID: PMC8006020 DOI: 10.3390/mps4010022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 01/01/2023] Open
Abstract
Control of gene expression is critical in shaping the pro-and eukaryotic organisms’ genotype and phenotype. The gene expression regulatory pathways solely rely on protein–protein and protein–nucleic acid interactions, which determine the fate of the nucleic acids. RNA–protein interactions play a significant role in co- and post-transcriptional regulation to control gene expression. RNA-binding proteins (RBPs) are a diverse group of macromolecules that bind to RNA and play an essential role in RNA biology by regulating pre-mRNA processing, maturation, nuclear transport, stability, and translation. Hence, the studies aimed at investigating RNA–protein interactions are essential to advance our knowledge in gene expression patterns associated with health and disease. Here we discuss the long-established and current technologies that are widely used to study RNA–protein interactions in vivo. We also present the advantages and disadvantages of each method discussed in the review.
Collapse
|
46
|
Planell N, Lagani V, Sebastian-Leon P, van der Kloet F, Ewing E, Karathanasis N, Urdangarin A, Arozarena I, Jagodic M, Tsamardinos I, Tarazona S, Conesa A, Tegner J, Gomez-Cabrero D. STATegra: Multi-Omics Data Integration - A Conceptual Scheme With a Bioinformatics Pipeline. Front Genet 2021; 12:620453. [PMID: 33747045 PMCID: PMC7970106 DOI: 10.3389/fgene.2021.620453] [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: 10/22/2020] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Technologies for profiling samples using different omics platforms have been at the forefront since the human genome project. Large-scale multi-omics data hold the promise of deciphering different regulatory layers. Yet, while there is a myriad of bioinformatics tools, each multi-omics analysis appears to start from scratch with an arbitrary decision over which tools to use and how to combine them. Therefore, it is an unmet need to conceptualize how to integrate such data and implement and validate pipelines in different cases. We have designed a conceptual framework (STATegra), aiming it to be as generic as possible for multi-omics analysis, combining available multi-omic anlaysis tools (machine learning component analysis, non-parametric data combination, and a multi-omics exploratory analysis) in a step-wise manner. While in several studies, we have previously combined those integrative tools, here, we provide a systematic description of the STATegra framework and its validation using two The Cancer Genome Atlas (TCGA) case studies. For both, the Glioblastoma and the Skin Cutaneous Melanoma (SKCM) cases, we demonstrate an enhanced capacity of the framework (and beyond the individual tools) to identify features and pathways compared to single-omics analysis. Such an integrative multi-omics analysis framework for identifying features and components facilitates the discovery of new biology. Finally, we provide several options for applying the STATegra framework when parametric assumptions are fulfilled and for the case when not all the samples are profiled for all omics. The STATegra framework is built using several tools, which are being integrated step-by-step as OpenSource in the STATegRa Bioconductor package.
Collapse
Affiliation(s)
- Nuria Planell
- Translational Bioinformatics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Vincenzo Lagani
- Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
- Gnosis Data Analysis P.C., Heraklion, Greece
| | - Patricia Sebastian-Leon
- Department of Genomic and Systems Reproductive Medicine, IVI-RMA (Instituto Valenciano de Infertilidad – Reproductive Medicine Associates) IVI Foundation, Valencia, Spain
| | - Frans van der Kloet
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Ewoud Ewing
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Nestoras Karathanasis
- Institute of Computer Science, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Computational Medicine Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Arantxa Urdangarin
- Translational Bioinformatics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Imanol Arozarena
- Cancer Signalling Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Health Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ioannis Tsamardinos
- Gnosis Data Analysis P.C., Heraklion, Greece
- Computer Science Department, University of Crete, Heraklion, Greece
| | - Sonia Tarazona
- Department of Applied Statistics, Operations Research and Quality, Universitat Politècnica de València, València, Spain
| | - Ana Conesa
- Microbiology and Cell Science, Institute for Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
- Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Jesper Tegner
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Unit of Computational Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Science for Life Laboratory, Solna, Sweden
| | - David Gomez-Cabrero
- Translational Bioinformatics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Unit of Computational Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Mucosal & Salivary Biology DivisionKing’s College London Dental Institute, London, United Kingdom
| |
Collapse
|
47
|
Reyes-López FE, Ibarz A, Ordóñez-Grande B, Vallejos-Vidal E, Andree KB, Balasch JC, Fernández-Alacid L, Sanahuja I, Sánchez-Nuño S, Firmino JP, Pavez L, Polo J, Tort L, Gisbert E. Skin Multi-Omics-Based Interactome Analysis: Integrating the Tissue and Mucus Exuded Layer for a Comprehensive Understanding of the Teleost Mucosa Functionality as Model of Study. Front Immunol 2021; 11:613824. [PMID: 33613538 PMCID: PMC7890662 DOI: 10.3389/fimmu.2020.613824] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/24/2020] [Indexed: 01/13/2023] Open
Abstract
From a general structural perspective, a mucosal tissue is constituted by two main matrices: the tissue and the secreted mucus. Jointly, they fulfill a wide range of functions including the protection of the epithelial layer. In this study, we simultaneously analyzed the epithelial tissue and the secreted mucus response using a holistic interactome-based multi-omics approach. The effect of the gilthead sea bream (Sparus aurata) skin mucosa to a dietary inclusion of spray-dried porcine plasma (SDPP) was evaluated. The epithelial skin microarrays-based transcriptome data showed 194 differentially expressed genes, meanwhile the exuded mucus proteome analysis 35 differentially synthesized proteins. Separately, the skin transcripteractome revealed an expression profile that favored biological mechanisms associated to gene expression, biogenesis, vesicle function, protein transport and localization to the membrane. Mucus proteome showed an enhanced protective role with putatively higher antioxidant and antimicrobial properties. The integrated skin mucosa multi-interactome analysis evidenced the interrelationship and synergy between the metabolism and the exuded mucus functions improving specifically the tissue development, innate defenses, and environment recognition. Histologically, the skin increased in thickness and in number of mucous cells. A positive impact on animal performance, growth and feed efficiency was also registered. Collectively, the results suggest an intimate crosstalk between skin tissue and its exuded mucus in response to the nutritional stimulus (SDPP supplementation) that favors the stimulation of cell protein turnover and the activation of the exudation machinery in the skin mucosa. Thus, the multi-omics-based interactome analysis provides a comprehensive understanding of the biological context of response that takes place in a mucosal tissue. In perspective, this strategy is applicable for evaluating the effect of any experimental variable on any mucosal tissue functionality, including the benefits this assessment may provide on the study of the mammalian mucosa.
Collapse
Affiliation(s)
- Felipe E Reyes-López
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat de Autònoma de Barcelona (UAB), Bellatera, Spain.,Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Providencia, Chile.,Consorcio Tecnológico de Sanidad Acuícola, Ictio Biotechnologies S.A., Santiago, Chile
| | - Antoni Ibarz
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat de Barcelona (UB), Barcelona, Spain
| | - Borja Ordóñez-Grande
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat de Barcelona (UB), Barcelona, Spain
| | - Eva Vallejos-Vidal
- Centro de Biotecnología Acuícola, Facultad de Química y Biología, Universidad de Santiago de Chile, Edificio de Investigación Eduardo Morales, Santiago, Chile
| | - Karl B Andree
- IRTA-SCR, Aquaculture Program, Sant Carles de la Rápita, Spain
| | - Joan Carles Balasch
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat de Autònoma de Barcelona (UAB), Bellatera, Spain
| | - Laura Fernández-Alacid
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat de Barcelona (UB), Barcelona, Spain
| | - Ignasi Sanahuja
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat de Barcelona (UB), Barcelona, Spain
| | - Sergio Sánchez-Nuño
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat de Barcelona (UB), Barcelona, Spain
| | - Joana P Firmino
- IRTA-SCR, Aquaculture Program, Sant Carles de la Rápita, Spain.,PhD Program in Aquaculture, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Leonardo Pavez
- Instituto de Ciencias Naturales, Universidad de las Américas, Santiago, Chile
| | | | - Lluis Tort
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat de Autònoma de Barcelona (UAB), Bellatera, Spain
| | - Enric Gisbert
- IRTA-SCR, Aquaculture Program, Sant Carles de la Rápita, Spain
| |
Collapse
|
48
|
Riley LA, Guss AM. Approaches to genetic tool development for rapid domestication of non-model microorganisms. BIOTECHNOLOGY FOR BIOFUELS 2021; 14:30. [PMID: 33494801 PMCID: PMC7830746 DOI: 10.1186/s13068-020-01872-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/30/2020] [Indexed: 05/04/2023]
Abstract
Non-model microorganisms often possess complex phenotypes that could be important for the future of biofuel and chemical production. They have received significant interest the last several years, but advancement is still slow due to the lack of a robust genetic toolbox in most organisms. Typically, "domestication" of a new non-model microorganism has been done on an ad hoc basis, and historically, it can take years to develop transformation and basic genetic tools. Here, we review the barriers and solutions to rapid development of genetic transformation tools in new hosts, with a major focus on Restriction-Modification systems, which are a well-known and significant barrier to efficient transformation. We further explore the tools and approaches used for efficient gene deletion, DNA insertion, and heterologous gene expression. Finally, more advanced and high-throughput tools are now being developed in diverse non-model microbes, paving the way for rapid and multiplexed genome engineering for biotechnology.
Collapse
Affiliation(s)
- Lauren A Riley
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA
- Bredesen Center, University of Tennessee, Knoxville, TN, 37996, USA
| | - Adam M Guss
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, USA.
- Bredesen Center, University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
49
|
Irshad O, Ghani Khan MU. Formalization and Semantic Integration of Heterogeneous Omics Annotations for Exploratory Searches. Curr Bioinform 2021. [DOI: 10.2174/1574893615666200127122818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aim:
To facilitate researchers and practitioners for unveiling the mysterious functional aspects of human cellular system through performing exploratory searching on semantically integrated heterogeneous and geographically dispersed omics annotations.
Background:
Improving health standards of life is one of the motives which continuously instigates researchers and practitioners to strive for uncovering the mysterious aspects of human cellular system. Inferring new knowledge from known facts always requires reasonably large amount of data in well-structured, integrated and unified form. Due to the advent of especially high throughput and sensor technologies, biological data is growing heterogeneously and geographically at astronomical rate. Several data integration systems have been deployed to cope with the issues of data heterogeneity and global dispersion. Systems based on semantic data integration models are more flexible and expandable than syntax-based ones but still lack aspect-based data integration, persistence and querying. Furthermore, these systems do not fully support to warehouse biological entities in the form of semantic associations as naturally possessed by the human cell.
Objective:
To develop aspect-oriented formal data integration model for semantically integrating heterogeneous and geographically dispersed omics annotations for providing exploratory querying on integrated data.
Method:
We propose an aspect-oriented formal data integration model which uses web semantics standards to formally specify its each construct. Proposed model supports aspect-oriented representation of biological entities while addressing the issues of data heterogeneity and global dispersion. It associates and warehouses biological entities in the way they relate with
Result:
To show the significance of proposed model, we developed a data warehouse and information retrieval system based on proposed model compliant multi-layered and multi-modular software architecture. Results show that our model supports well for gathering, associating, integrating, persisting and querying each entity with respect to its all possible aspects within or across the various associated omics layers.
Conclusion:
Formal specifications better facilitate for addressing data integration issues by providing formal means for understanding omics data based on meaning instead of syntax
Collapse
Affiliation(s)
- Omer Irshad
- Department of Computer Science & Engineering, Faculty of Electrical Engineering, The University of Engineering and Technology, Lahore,Pakistan
| | - Muhammad Usman Ghani Khan
- Department of Computer Science & Engineering, Faculty of Electrical Engineering, The University of Engineering and Technology, Lahore,Pakistan
| |
Collapse
|
50
|
Wright NR, Rønnest NP, Sonnenschein N. Single-Cell Technologies to Understand the Mechanisms of Cellular Adaptation in Chemostats. Front Bioeng Biotechnol 2020; 8:579841. [PMID: 33392163 PMCID: PMC7775484 DOI: 10.3389/fbioe.2020.579841] [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: 07/03/2020] [Accepted: 11/30/2020] [Indexed: 11/13/2022] Open
Abstract
There is a growing interest in continuous manufacturing within the bioprocessing community. In this context, the chemostat process is an important unit operation. The current application of chemostat processes in industry is limited although many high yielding processes are reported in literature. In order to reach the full potential of the chemostat in continuous manufacture, the output should be constant. However, adaptation is often observed resulting in changed productivities over time. The observed adaptation can be coupled to the selective pressure of the nutrient-limited environment in the chemostat. We argue that population heterogeneity should be taken into account when studying adaptation in the chemostat. We propose to investigate adaptation at the single-cell level and discuss the potential of different single-cell technologies, which could be used to increase the understanding of the phenomena. Currently, none of the discussed single-cell technologies fulfill all our criteria but in combination they may reveal important information, which can be used to understand and potentially control the adaptation.
Collapse
Affiliation(s)
- Naia Risager Wright
- Novo Nordisk A/S, Bagsvaerd, Denmark
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Nikolaus Sonnenschein
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|