1
|
Hurtado M, Suarez-Álvarez S, Castander-Olarieta A, Montalbán IA, Goicoechea PG, López de Heredia U, Marino D, Moncaleán P. Physiological and molecular response to drought in somatic plants from Pinus radiata embryonal masses induced at high temperatures. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2025; 224:109886. [PMID: 40262399 DOI: 10.1016/j.plaphy.2025.109886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/04/2025] [Indexed: 04/24/2025]
Abstract
Drought and heat are among the major abiotic stresses in forest trees and are directly related with the consequences of climatic change. Many responses to abiotic stresses in plants have been associated with plant memory but mechanisms underlying this phenomenon remain unclear. Somatic embryogenesis, which is considered one of the most important methods for large-scale vegetative propagation of plants, is also used for stress induction and study the mechanisms involved in adaptation to abiotic stress. Specifically, heat stress during initiation stage of somatic embryogenesis has shown to have an impact in differential expression of stress related genes in pines. Modifications caused by a previous stress could eventually influence the stress tolerance of somatic plants years later. In this study we analysed the response to drought in 2-year-old radiata pine somatic plants, derived from embryonal masses initiated at 60 °C, at physiological, transcriptomic and amino acid accumulation level. Our results showed a more pronounce response to drought in plants coming from 60 °C treatment, which presented lower values in several physiological parameters as well as higher proline and tyrosine levels. Additionally, the transcriptomic response to drought was stronger in heat primed plants compared to control plants, suggesting a memory acquired two years before.
Collapse
Affiliation(s)
- Mikel Hurtado
- Department Forestry Sciences, NEIKER-BRTA, Instituto Vasco de Investigación y Desarrollo Agrario, Campus Agroalimentario de Arkaute, Ctra N-104 km 355, Arkaute, Álava, 01192, Spain; Department of Plant Biology and Ecology, Facultad de Ciencia y Tecnología, Universidad del País Vasco-Euskal Herriko Unibertsitatea (UPV/EHU), Barrio Sarriena s/n, Leioa, Bizkaia, 48940, Spain
| | - Sonia Suarez-Álvarez
- Department Plant Production. NEIKER-BRTA, Instituto Vasco de Investigación y Desarrollo Agrario, Campus Agroalimentario de Arkaute, Ctra N-104 km 355, Arkaute, Álava, 01192, Spain
| | - Ander Castander-Olarieta
- Department Forestry Sciences, NEIKER-BRTA, Instituto Vasco de Investigación y Desarrollo Agrario, Campus Agroalimentario de Arkaute, Ctra N-104 km 355, Arkaute, Álava, 01192, Spain
| | - Itziar A Montalbán
- Department Forestry Sciences, NEIKER-BRTA, Instituto Vasco de Investigación y Desarrollo Agrario, Campus Agroalimentario de Arkaute, Ctra N-104 km 355, Arkaute, Álava, 01192, Spain
| | - Pablo G Goicoechea
- Department Forestry Sciences, NEIKER-BRTA, Instituto Vasco de Investigación y Desarrollo Agrario, Campus Agroalimentario de Arkaute, Ctra N-104 km 355, Arkaute, Álava, 01192, Spain
| | - Unai López de Heredia
- GI en Desarrollo de Especies y Comunidades Leñosas (WooSP), Dpto. Sistemas y Recursos Naturales, ETSI Montes, Forestal y del Medio Natural, Universidad Politécnica de Madrid, Ciudad Universitaria s/n, 28040, Madrid, Spain
| | - Daniel Marino
- Department of Plant Biology and Ecology, Facultad de Ciencia y Tecnología, Universidad del País Vasco-Euskal Herriko Unibertsitatea (UPV/EHU), Barrio Sarriena s/n, Leioa, Bizkaia, 48940, Spain
| | - Paloma Moncaleán
- Department Forestry Sciences, NEIKER-BRTA, Instituto Vasco de Investigación y Desarrollo Agrario, Campus Agroalimentario de Arkaute, Ctra N-104 km 355, Arkaute, Álava, 01192, Spain.
| |
Collapse
|
2
|
Bertagna M, Bright L, Ye F, Jiang YY, Sarkar D, Pradhan A, Kumar S, Gao S, Turkewitz A, Tsypin LZ. Inferring gene-pathway associations from consolidated transcriptome datasets: an interactive gene network explorer for Tetrahymena thermophila. NAR Genom Bioinform 2025; 7:lqaf067. [PMID: 40432793 PMCID: PMC12107436 DOI: 10.1093/nargab/lqaf067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/11/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
Although an established model organism, Tetrahymena thermophila remains comparatively inaccessible to high throughput screens, and alternative bioinformatic approaches still rely on unconnected datasets and outdated algorithms. Here, we report a new approach to consolidating RNA-seq and microarray data based on a systematic exploration of parameters and computational controls, enabling us to infer functional gene associations from their co-expression patterns. To illustrate the power of this approach, we took advantage of new data regarding a previously studied pathway, the biogenesis of a secretory organelle called the mucocyst. Our untargeted clustering approach recovered over 80% of the genes that were previously verified to play a role in mucocyst biogenesis. Furthermore, we tested four new genes that we predicted to be mucocyst-associated based on their co-expression and found that knocking out each of them results in mucocyst secretion defects. We also found that our approach succeeds in clustering genes associated with several other cellular pathways that we evaluated based on prior literature. We present the Tetrahymena Gene Network Explorer (TGNE) as an interactive tool for genetic hypothesis generation and functional annotation in this organism and as a framework for building similar tools for other systems.
Collapse
Affiliation(s)
- Michael A Bertagna
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, 60637, United States
| | - Lydia J Bright
- Department of Biology, State University of New York at New Paltz, New Paltz, NY, 12561, United States
| | - Fei Ye
- MOE Key Laboratory of Evolution & Marine Biodiversity and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Yu-Yang Jiang
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, 60637, United States
| | - Debolina Sarkar
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune, Maharashtra State, 411007, India
| | - Ajay Pradhan
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune, Maharashtra State, 411007, India
| | - Santosh Kumar
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune, Maharashtra State, 411007, India
| | - Shan Gao
- MOE Key Laboratory of Evolution & Marine Biodiversity and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Aaron P Turkewitz
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, 60637, United States
| | - Lev M Z Tsypin
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA, 94305, United States
| |
Collapse
|
3
|
Gandham K, Thomas J, Riaz A, Balakrishnan D, Pereira A, Kariyat R. Rice master regulator 'HYR' enhances growth and defense mechanisms with consequences for fall armyworm growth and host selection. PLANT & CELL PHYSIOLOGY 2025; 66:687-704. [PMID: 40045601 DOI: 10.1093/pcp/pcaf025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 06/01/2025]
Abstract
Rice (Oryza sativa L.), the staple food for half of the world's population, suffers heavy damage by insect herbivores, especially the emerging fall armyworm (FAW), Spodoptera frugiperda. HIGHER YIELD RICE (HYR), a master regulator of multiple biological pathways with an established gene regulatory network, has been found to improve rice yield to ∼29% and tolerance to environmental stress. However, its impact on defense has not been explored. We hypothesized that, FAW would target HYR plants because of its vigorous growth and lead to trade-offs for defense. Through a series of experiments with HYR and its wild type (WT), we show that HYR plants have enhanced below-ground growth, physiological traits, and direct and indirect defense traits including leaf trichomes, wax, and volatile organic compounds. To test possible phytohormone-mediated defense signaling, we focussed on jasmonic acid and salicylic acid gene expression panel and found that most of these genes are highly expressed in HYR when compared to its WT counterpart. Bioassays examining developmental milestones also revealed that HYR plants effectively deter FAW, and when force-fed, caused negative effects. Collectively, our findings suggest that the master regulator HYR (Higher Yield Rice expressing) plants enhance growth and physiological traits, as well as physical and chemical defense mechanisms through co-ordinated defense gene expression, which deter herbivore feeding, growth, development, and host selection.
Collapse
Affiliation(s)
- Krishnarao Gandham
- Department of Entomology and Plant Pathology, University of Arkansas, Fayetteville, AR 72701, United States of America
| | - Julie Thomas
- Department of Crop, Soil and Environmental Sciences, University of Arkansas, Fayetteville, AR 72701, United States of America
| | - Awais Riaz
- Department of Crop, Soil and Environmental Sciences, University of Arkansas, Fayetteville, AR 72701, United States of America
| | - Devi Balakrishnan
- Department of Entomology and Plant Pathology, University of Arkansas, Fayetteville, AR 72701, United States of America
| | - Andy Pereira
- Department of Crop, Soil and Environmental Sciences, University of Arkansas, Fayetteville, AR 72701, United States of America
| | - Rupesh Kariyat
- Department of Entomology and Plant Pathology, University of Arkansas, Fayetteville, AR 72701, United States of America
| |
Collapse
|
4
|
Qiao L, Khalilimeybodi A, Linden-Santangeli NJ, Rangamani P. The Evolution of Systems Biology and Systems Medicine: From Mechanistic Models to Uncertainty Quantification. Annu Rev Biomed Eng 2025; 27:425-447. [PMID: 39971380 DOI: 10.1146/annurev-bioeng-102723-065309] [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] [Indexed: 02/21/2025]
Abstract
Understanding interaction mechanisms within cells, tissues, and organisms is crucial for driving developments across biology and medicine. Mathematical modeling is an essential tool for simulating such biological systems. Building on experiments, mechanistic models are widely used to describe small-scale intracellular networks. The development of sequencing techniques and computational tools has recently enabled multiscale models. Combining such larger scale network modeling with mechanistic modeling provides us with an opportunity to reveal previously unknown disease mechanisms and pharmacological interventions. Here, we review systems biology models from mechanistic models to multiscale models that integrate multiple layers of cellular networks and discuss how they can be used to shed light on disease states and even wellness-related states. Additionally, we introduce several methods that increase the certainty and accuracy of model predictions. Thus, combining mechanistic models with emerging mathematical and computational techniques can provide us with increasingly powerful tools to understand disease states and inspire drug discoveries.
Collapse
Affiliation(s)
- Lingxia Qiao
- Department of Pharmacology, University of California San Diego, La Jolla, California, USA;
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA
| | - Ali Khalilimeybodi
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA
| | | | - Padmini Rangamani
- Department of Pharmacology, University of California San Diego, La Jolla, California, USA;
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
5
|
de Silva K, Coelho C, Gao J, Brooks MD. Shining light on Arabidopsis regulatory networks integrating nitrogen use and photosynthesis. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2025; 122:e70211. [PMID: 40358388 PMCID: PMC12071342 DOI: 10.1111/tpj.70211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 03/06/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025]
Abstract
Nitrogen and light availability are well-known to influence photosynthesis, having both individual and synergistic effects. However, the regulatory interactions between these signaling pathways, especially the transcription factors (TFs) that perceive and integrate these cues, remain to be elucidated. Arabidopsis grown in a matrix of nitrogen and light treatments exhibited distinct physiological and transcriptomic responses. Notably, the effect of nitrogen dose on biomass, nitrogen use efficiency, carbon-to-nitrogen ratio, and gene expression was highly dependent on light intensity. Genes differentially expressed across the treatments were enriched for photosynthetic processes, including the pentose-phosphate cycle, light-harvesting, and chlorophyll biosynthesis. TFs coordinating photosynthesis, carbon-to-nitrogen balance, and nitrogen uptake were identified based on motif enrichment, validated binding data, and gene regulatory network analysis. Dynamic light-by-nitrogen responses were found for TFs previously linked to either nitrogen or light signaling, which now emerge as regulatory hubs that integrate these signals. Among these TFs, we identified bZIP and MYB-related family transcription factors as pivotal players in harmonizing photosynthesis, nitrogen assimilation, and light responses. The transcription factors unveiled in this study have the potential to unlock new strategies for optimizing photosynthetic activity and nutrient-use efficiency in plants.
Collapse
Affiliation(s)
- Kithmee de Silva
- Department of Plant BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinois61801USA
| | - Camila Coelho
- Department of BiologyNew York UniversityNew YorkNew York10003USA
| | - Jenny Gao
- Department of BiologyNew York UniversityNew YorkNew York10003USA
| | - Matthew D. Brooks
- Global Change and Photosynthesis Research UnitUSDA ARSUrbanaIllinois61801USA
| |
Collapse
|
6
|
Sebastian S, Roy S, Kalita J. Network-based analysis of Alzheimer's Disease genes using multi-omics network integration with graph diffusion. J Biomed Inform 2025; 164:104797. [PMID: 39993589 DOI: 10.1016/j.jbi.2025.104797] [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: 10/23/2024] [Revised: 01/16/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
Alzheimer's Disease (AD) is a complex neurodegenerative disorder affecting millions worldwide. Despite extensive research, the mechanisms behind AD remain elusive. Many studies suggest that disease-responsible genes often act as hub genes in biological networks. However, this assumption requires further investigation in the context of AD. To examine the network characteristics of known AD genes, it is crucial to construct a highly confident network, which is challenging to achieve using a single data source. This work integrates multi-omics networks inferred from microarray, single-cell RNA sequencing, and single-nuclei RNA sequencing expression data, weighted with protein interaction and gene ontology information. We generate a high-quality integrated network by utilizing various inference methods and combining them through a graph diffusion-based integration approach. This network is then analyzed to investigate the properties of known AD-specific genes. Our findings reveal that AD genes are not always high-degree or central hub nodes in the network. Instead, these genes are distributed across different quartiles of degree centrality while maintaining significant interconnections for effective regulation. Furthermore, our study highlights that peripheral genes, often overlooked, also play crucial roles by connecting to relevant disease nodes and hub genes. These findings challenge the conventional understanding that AD-responsible genes are primarily the hub genes in the network, offering new insights into the complex regulatory mechanisms of AD and suggesting novel directions for future research.
Collapse
Affiliation(s)
- Softya Sebastian
- Network Reconstruction and Analysis (NetRA) Lab, Department of Computer Applications, Sikkim (Central) University, India; School of Computer Science and Engineering, Vellore Institute of Technology, Chennai, India
| | - Swarup Roy
- Network Reconstruction and Analysis (NetRA) Lab, Department of Computer Applications, Sikkim (Central) University, India.
| | - Jugal Kalita
- Department of Computer Science, University of Colorado at Colorado Springs, USA
| |
Collapse
|
7
|
Borg JP, Colinge J, Ravel P. Testing and overcoming the limitations of modular response analysis. Brief Bioinform 2025; 26:bbaf098. [PMID: 40062619 PMCID: PMC11891662 DOI: 10.1093/bib/bbaf098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/04/2025] [Accepted: 02/25/2025] [Indexed: 05/13/2025] Open
Abstract
Modular response analysis (MRA) is an effective method to infer biological networks from perturbation data. However, it has several limitations such as strong sensitivity to noise, need of performing independent perturbations that hit a single node at a time, and linear approximation of dependencies within the network. Previously, we addressed the sensitivity of MRA to noise by reinterpreting MRA as a multilinear regression problem. We demonstrated the advantages of this approach over the conventional MRA and other known inference methods, particularly in handling noise measurements and nonlinear networks. Here, we provide new contributions to complement this theory. First, we overcome the need of perturbations to be independent, thereby augmenting MRA applicability. Second, using analysis of variance and lack-of-fit tests, we can now assess MRA compatibility with the data and identify the primary source of errors. In cases where nonlinearity prevails, we propose extending the model to a second-order polynomial. Third, we demonstrate how to effectively use prior knowledge about a network. We validated these results using 4 networks with known dynamics (3, 4, and 6 nodes) and 40 simulated networks, ranging from 10 to 200 nodes. Finally, we incorporated these innovations into our R software package MRARegress to offer a comprehensive, extended theory for MRA and to facilitate its use by the community. Mathematical aspects, tests details, and scripts are provided as Supplementary Information (see 'Data Availability Statement').
Collapse
Affiliation(s)
- Jean-Pierre Borg
- Université de Montpellier, 5 Bd Henri IV, 34000 Montpellier, France
- Institut régional du Cancer Montpellier (ICM), 208 Av. des Apothicaires, 34090 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, 208 Av. des Apothicaires, 34090 Montpellier, France
| | - Jacques Colinge
- Université de Montpellier, 5 Bd Henri IV, 34000 Montpellier, France
- Institut régional du Cancer Montpellier (ICM), 208 Av. des Apothicaires, 34090 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, 208 Av. des Apothicaires, 34090 Montpellier, France
| | - Patrice Ravel
- Université de Montpellier, 5 Bd Henri IV, 34000 Montpellier, France
- Institut régional du Cancer Montpellier (ICM), 208 Av. des Apothicaires, 34090 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, 208 Av. des Apothicaires, 34090 Montpellier, France
| |
Collapse
|
8
|
Majeed A, Seth R, Sharma B, Devi A, Sharma S, Masand M, Rahim MS, Verma N, Kumar D, Sharma RK. Deep transcriptome and metabolome analysis to dissect untapped spatial dynamics of specialized metabolism in Saussurea costus (Falc.) Lipsch. Funct Integr Genomics 2025; 25:46. [PMID: 40019562 DOI: 10.1007/s10142-025-01549-6] [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: 10/03/2024] [Revised: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Saussurea costus (Falc.) is an endangered medicinal plant possessing diverse phytochemical compounds with clinical significance and used to treat numerous human ailments. Despite the source of enriched phytochemicals, molecular insights into spatialized metabolism are poorly understood in S. costus. This study investigated the dynamics of organ-specific secondary metabolite biosynthesis using deep transcriptome sequencing and high-throughput UHPLC-QTOF based untargeted metabolomic profiling. A de novo assembly from quality reads fetched 59,725 transcripts with structural (53.02%) and functional (66.13%) annotations of non-redundant transcripts. Of the 7,683 predicted gene families, 3,211 were categorized as 'single gene families'. Interestingly, out of the 4,664 core gene families within the Asterids, 4,560 families were captured in S. costus. Organ-specific differential gene expression analysis revealed significant variations between leaves vs. stems (23,102 transcripts), leaves vs. roots (30,590 transcripts), and roots vs. stems (21,759 transcripts). Like-wise, putative metabolites (PMs) were recorded with significant differences in leaves vs. roots (250 PMs), leaves vs. stem (350 PMs), and roots vs. stem (107 PMs). The integrative transcriptomic and metabolomic analysis identified organ-specific differences in the accumulation of important metabolites, including secologanin, menthofuran, taraxerol, lupeol, acetyleugenol, scopoletin, costunolide, and dehydrocostus lactone. Furthermore, a global gene co-expression network (GCN) identified putative regulators controlling the expression of key target genes of secondary metabolite pathways including terpenoid, phenylpropanoid, and flavonoid. The comprehensive functionally relevant genomic resource created here provides beneficial insights for upscaling targeted metabolite biosynthesis through genetic engineering, and for expediting association mapping efforts to elucidate the casual genetic elements controlling specific bioactive metabolites.
Collapse
Affiliation(s)
- Aasim Majeed
- Molecular Genetic and Genomics Laboratory, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, India
| | - Romit Seth
- Molecular Genetic and Genomics Laboratory, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, India
| | - Balraj Sharma
- Molecular Genetic and Genomics Laboratory, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Amna Devi
- Molecular Genetic and Genomics Laboratory, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shikha Sharma
- Molecular Genetic and Genomics Laboratory, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, India
| | - Mamta Masand
- Molecular Genetic and Genomics Laboratory, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Mohammed Saba Rahim
- Molecular Genetic and Genomics Laboratory, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, India
| | - Naveen Verma
- Molecular Genetic and Genomics Laboratory, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, India
| | - Dinesh Kumar
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ram Kumar Sharma
- Molecular Genetic and Genomics Laboratory, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
9
|
Xu J, Lu C, Jin S, Meng Y, Fu X, Zeng X, Nussinov R, Cheng F. Deep learning-based cell-specific gene regulatory networks inferred from single-cell multiome data. Nucleic Acids Res 2025; 53:gkaf138. [PMID: 40037709 PMCID: PMC11879466 DOI: 10.1093/nar/gkaf138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/03/2025] [Accepted: 02/13/2025] [Indexed: 03/06/2025] Open
Abstract
Gene regulatory networks (GRNs) provide a global representation of how genetic/genomic information is transferred in living systems and are a key component in understanding genome regulation. Single-cell multiome data provide unprecedented opportunities to reconstruct GRNs at fine-grained resolution. However, the inference of GRNs is hindered by insufficient single omic profiles due to the characteristic high loss rate of single-cell sequencing data. In this study, we developed scMultiomeGRN, a deep learning framework to infer transcription factor (TF) regulatory networks via unique integration of single-cell genomic (single-cell RNA sequencing) and epigenomic (single-cell ATAC sequencing) data. We create scMultiomeGRN to elucidate these networks by conceptualizing TF network graph structures. Specifically, we build modality-specific neighbor aggregators and cross-modal attention modules to learn latent representations of TFs from single-cell multi-omics. We demonstrate that scMultiomeGRN outperforms state-of-the-art models on multiple benchmark datasets involved in diseases and health. Via scMultiomeGRN, we identified Alzheimer's disease-relevant regulatory network of SPI1 and RUNX1 for microglia. In summary, scMultiomeGRN offers a deep learning framework to identify cell type-specific gene regulatory network from single-cell multiome data.
Collapse
Affiliation(s)
- Junlin Xu
- School of Computer Science and Technology, Wuhan University of Science and Technology, Wuhan, Hubei 430065, China
| | - Changcheng Lu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Shuting Jin
- School of Computer Science and Technology, Wuhan University of Science and Technology, Wuhan, Hubei 430065, China
| | - Yajie Meng
- School of Computer Science and Artificial Intelligence, Wuhan Textile University, Wuhan, Hubei 430200, China
| | - Xiangzheng Fu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Xiangxiang Zeng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Ruth Nussinov
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, United States
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, United States
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, United States
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, United States
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| |
Collapse
|
10
|
Wang C, Liu ZP. Diffusion-based generation of gene regulatory networks from scRNA-seq data with DigNet. Genome Res 2025; 35:340-354. [PMID: 39694856 PMCID: PMC11874984 DOI: 10.1101/gr.279551.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
A gene regulatory network (GRN) intricately encodes the interconnectedness of identities and functionalities of genes within cells, ultimately shaping cellular specificity. Despite decades of endeavors, reverse engineering of GRNs from gene expression profiling data remains a profound challenge, particularly when it comes to reconstructing cell-specific GRNs that are tailored to precise cellular and genetic contexts. Here, we propose a discrete diffusion generation model, called DigNet, capable of generating corresponding GRNs from high-throughput single-cell RNA sequencing (scRNA-seq) data. DigNet embeds the network generation process into a multistep recovery procedure with Markov properties. Each intermediate step has a specific model to recover a portion of the gene regulatory architectures. It thus can ensure compatibility between global network structures and regulatory modules through the unique multistep diffusion procedure. Furthermore, through iMetacell integration and non-Euclidean discrete space modeling, DigNet is robust to the presence of noise in scRNA-seq data and the sparsity of GRNs. Benchmark evaluation results against more than a dozen state-of-the-art network inference methods demonstrate that DigNet achieves superior performance across various single-cell GRN reconstruction experiments. Furthermore, DigNet provides unique insights into the immune response in breast cancer, derived from differential gene regulation identified in T cells. As an open-source software, DigNet offers a powerful and effective tool for generating cell-specific GRNs from scRNA-seq data.
Collapse
Affiliation(s)
- Chuanyuan Wang
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Zhi-Ping Liu
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| |
Collapse
|
11
|
Liu F, Li N, Yan ZY, Chen X. Time-series transcriptome analysis reveals the cascade mechanism of biological processes following the perturbation of the MVA pathway in Salvia miltiorrhiza. PLANT MOLECULAR BIOLOGY 2025; 115:20. [PMID: 39821838 PMCID: PMC11742292 DOI: 10.1007/s11103-024-01547-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 12/13/2024] [Indexed: 01/19/2025]
Abstract
Various biological processes are interconnected in plants. Transcription factors (TFs) often act as regulatory hubs to regulate plant growth and responses to stress by integrating various biological pathways. Despite extensive studies on TFs functions in various plant species, our understanding of the details of TFs regulation remains limited. In this study, clonal seedlings of Salvia miltiorrhiza were exposed to specific inhibitors for 12 h. Time-series transcriptome data, sampled hourly, were used to construct co-expression networks and gene regulatory networks (GRNs). Transcriptome dynamic analysis was utilized to capture the gene expression dynamics of various biological processes and decipher the potential molecular mechanisms that regulate these processes. The perturbation results showed the growth and development processes of S.miltiorrhiza were primarily affected at the early stage, whereas stress response-related biological processes were mainly influenced at the later stage. And there was a correlation between the series of key differentially expressed genes in terpenoid biosynthesis pathways and the topological distribution of these pathways. Furthermore, the GRNs based on TFs indicate that TFs play a crucial role in connecting various biological processes. In the cytoplasmic lysate gene regulatory module, SmWRKY48-SmTCP4-SmWRKY28 constituted a regulation hub regulating S.miltiorrhiza responses to perturbation of the MVA pathway. The regulation hub mediated various pathways, including pyruvate metabolism, glycolysis/gluconeogenesis, amino acid metabolism, and ubiquinone and other terpenoid-quinone biosynthesis.Our findings suggest that perturbation of a key biological pathway in S.miltiorrhiza has time-dependent effects on other biological processes. And SmWRKY48-SmTCP4-SmWRKY28 constitutes the regulatory hub in S.miltiorrhiza responses to perturbation of MVA pathway.
Collapse
Affiliation(s)
- Fang Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Nan Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhu-Yun Yan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xin Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
12
|
Jing R, Falchetti M, Han T, Najia M, Hensch LT, Meader E, Lummertz da Rocha E, Kononov M, Wang S, Bingham T, Li Z, Zhao Y, Frenis K, Kubaczka C, Yang S, Jha D, Rodrigues-Luiz GF, Rowe RG, Schlaeger TM, Maus MV, North TE, Zon LI, Daley GQ. Maturation and persistence of CAR T cells derived from human pluripotent stem cells via chemical inhibition of G9a/GLP. Cell Stem Cell 2025; 32:71-85.e5. [PMID: 39504968 PMCID: PMC11698653 DOI: 10.1016/j.stem.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/27/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024]
Abstract
Elucidating mechanisms of T cell development can guide in vitro T cell differentiation from induced pluripotent stem cells (iPSCs) and facilitate off-the-shelf T cell-based immunotherapies. Using a stroma-free human iPSC-T cell differentiation platform, we screened for epigenetic modulators that influence T cell specification and identified the H3K9-directed histone methyltransferases G9a/GLP as repressors of T cell fate. We show that G9a/GLP inhibition during specific time windows of differentiation of hematopoietic stem and progenitor cells (HSPCs) skews cell fates toward lymphoid lineages. Inhibition of G9a/GLP promotes the production of lymphoid cells during zebrafish embryonic hematopoiesis, demonstrating the evolutionary conservation of G9a/GLP function. Importantly, chemical inhibition of G9a/GLP facilitates the generation of mature iPSC-T cells that bear transcriptional similarity to peripheral blood αβ T cells. When engineered to express chimeric antigen receptors, the epigenetically engineered iPSC-T cells exhibit enhanced effector functions in vitro and durable, persistent antitumor activity in a xenograft tumor-rechallenge model.
Collapse
Affiliation(s)
- Ran Jing
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Marcelo Falchetti
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Tianxiao Han
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Mohamad Najia
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Luca T Hensch
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Eleanor Meader
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Edroaldo Lummertz da Rocha
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | - Martin Kononov
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Stephanie Wang
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Trevor Bingham
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Zhiheng Li
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Yunliang Zhao
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Katie Frenis
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Caroline Kubaczka
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Song Yang
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Deepak Jha
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Gabriela F Rodrigues-Luiz
- Graduate Program of Pharmacology, Center for Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil
| | - R Grant Rowe
- Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA 02115, USA
| | | | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Charlestown, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Trista E North
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
| | - Leonard I Zon
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - George Q Daley
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Bertagna MA, Bright LJ, Ye F, Jiang YY, Sarkar D, Pradhan A, Kumar S, Gao S, Turkewitz AP, Tsypin LMZ. Inferring gene-pathway associations from consolidated transcriptome datasets: an interactive gene network explorer for Tetrahymena thermophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.627356. [PMID: 39713406 PMCID: PMC11661410 DOI: 10.1101/2024.12.12.627356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Although an established model organism, Tetrahymena thermophila remains comparatively inaccessible to high throughput screens, and alternative bioinformatic approaches still rely on unconnected datasets and outdated algorithms. Here, we report a new approach to consolidating RNA-seq and microarray data based on a systematic exploration of parameters and computational controls, enabling us to infer functional gene associations from their co-expression patterns. To illustrate the power of this approach, we took advantage of new data regarding a previously studied pathway, the biogenesis of a secretory organelle called the mucocyst. Our untargeted clustering approach recovered over 80% of the genes that were previously verified to play a role in mucocyst biogenesis. Furthermore, we tested four new genes that we predicted to be mucocyst-associated based on their co-expression and found that knocking out each of them results in mucocyst secretion defects. We also found that our approach succeeds in clustering genes associated with several other cellular pathways that we evaluated based on prior literature. We present the Tetrahymena Gene Network Explorer (TGNE) as an interactive tool for genetic hypothesis generation and functional annotation in this organism and as a framework for building similar tools for other systems.
Collapse
Affiliation(s)
- Michael A. Bertagna
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, USA
| | - Lydia J. Bright
- Department of Biology, State University of New York at New Paltz, New Paltz, NY, USA
| | - Fei Ye
- MOE Key Laboratory of Evolution & Marine Biodiversity and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Yu-Yang Jiang
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, USA
| | - Debolina Sarkar
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune, 411007 Maharashtra State, India
| | - Ajay Pradhan
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune, 411007 Maharashtra State, India
| | - Santosh Kumar
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune, 411007 Maharashtra State, India
| | - Shan Gao
- MOE Key Laboratory of Evolution & Marine Biodiversity and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Aaron P. Turkewitz
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, USA
| | - Lev M. Z. Tsypin
- Department of Pathology, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
14
|
Gillenwater LA, Galbraith MD, Rachubinski AL, Eduthan NP, Sullivan KD, Espinosa JM, Costello JC. Integrated analysis of immunometabolic interactions in Down syndrome. SCIENCE ADVANCES 2024; 10:eadq3073. [PMID: 39671500 PMCID: PMC11641111 DOI: 10.1126/sciadv.adq3073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/05/2024] [Indexed: 12/15/2024]
Abstract
Down syndrome (DS), caused by trisomy 21 (T21), results in immune and metabolic dysregulation. People with DS experience co-occurring conditions at higher rates than the euploid population. However, the interplay between immune and metabolic alterations and the clinical manifestations of DS are poorly understood. Here, we report an integrated analysis of immunometabolic pathways in DS. Using multi-omics data, we infered cytokine-metabolite relationships mediated by specific transcriptional programs. We observed increased mediation of immunometabolic interactions in those with DS compared to euploid controls by genes in interferon response, heme metabolism, and oxidative phosphorylation. Unsupervised clustering of immunometabolic relationships in people with DS revealed subgroups with different frequencies of co-occurring conditions. Across the subgroups, we observed distinct mediation by DNA repair, Hedgehog signaling, and angiogenesis. The molecular stratification associates with the clinical heterogeneity observed in DS, suggesting that integrating multiple omic profiles reveals axes of coordinated dysregulation specific to DS co-occurring conditions.
Collapse
Affiliation(s)
- Lucas A. Gillenwater
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Matthew D. Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angela L. Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Neetha Paul Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kelly D. Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joaquin M. Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - James C. Costello
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
15
|
Yang B, Li J, Li X, Liu S. Gene regulatory network inference based on novel ensemble method. Brief Funct Genomics 2024; 23:866-878. [PMID: 39324652 DOI: 10.1093/bfgp/elae036] [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: 03/11/2024] [Revised: 08/09/2024] [Accepted: 09/06/2024] [Indexed: 09/27/2024] Open
Abstract
Gene regulatory networks (GRNs) contribute toward understanding the function of genes and the development of cancer or the impact of key genes on diseases. Hence, this study proposes an ensemble method based on 13 basic classification methods and a flexible neural tree (FNT) to improve GRN identification accuracy. The primary classification methods contain ridge classification, stochastic gradient descent, Gaussian process classification, Bernoulli Naive Bayes, adaptive boosting, gradient boosting decision tree, hist gradient boosting classification, eXtreme gradient boosting (XGBoost), multilayer perceptron, light gradient boosting machine, random forest, support vector machine, and k-nearest neighbor algorithm, which are regarded as the input variable set of FNT model. Additionally, a hybrid evolutionary algorithm based on a gene programming variant and particle swarm optimization is developed to search for the optimal FNT model. Experiments on three simulation datasets and three real single-cell RNA-seq datasets demonstrate that the proposed ensemble feature outperforms 13 supervised algorithms, seven unsupervised algorithms (ARACNE, CLR, GENIE3, MRNET, PCACMI, GENECI, and EPCACMI) and four single cell-specific methods (SCODE, BiRGRN, LEAP, and BiGBoost) based on the area under the receiver operating characteristic curve, area under the precision-recall curve, and F1 metrics.
Collapse
Affiliation(s)
- Bin Yang
- School of Information Science and Engineering, Zaozhuang University, No. 1 Beian Road, Zaozhuang 277160, China
| | - Jing Li
- School of Information Science and Engineering, Zaozhuang University, No. 1 Beian Road, Zaozhuang 277160, China
| | - Xiang Li
- Information Department, Qingdao Eighth People's Hospital, No. 84 Fengshan Road, Qingdao 266121, China
| | - Sanrong Liu
- School of Information Science and Engineering, Zaozhuang University, No. 1 Beian Road, Zaozhuang 277160, China
| |
Collapse
|
16
|
Jin H, Kim W, Yuan M, Li X, Yang H, Li M, Shi M, Turkez H, Uhlen M, Zhang C, Mardinoglu A. Identification of SPP1 + macrophages as an immune suppressor in hepatocellular carcinoma using single-cell and bulk transcriptomics. Front Immunol 2024; 15:1446453. [PMID: 39691723 PMCID: PMC11649653 DOI: 10.3389/fimmu.2024.1446453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Introduction Macrophages and T cells play crucial roles in liver physiology, but their functional diversity in hepatocellular carcinoma (HCC) remains largely unknown. Methods Two bulk RNA-sequencing (RNA-seq) cohorts for HCC were analyzed using gene co-expression network analysis. Key gene modules and networks were mapped to single-cell RNA-sequencing (scRNA-seq) data of HCC. Cell type fraction of bulk RNA-seq data was estimated by deconvolution approach using single-cell RNA-sequencing data as a reference. Survival analysis was carried out to estimate the prognosis of different immune cell types in bulk RNA-seq cohorts. Cell-cell interaction analysis was performed to identify potential links between immune cell types in HCC. Results In this study, we analyzed RNA-seq data from two large-scale HCC cohorts, revealing a major and consensus gene co-expression cluster with significant implications for immunosuppression. Notably, these genes exhibited higher enrichment in liver macrophages than T cells, as confirmed by scRNA-seq data from HCC patients. Integrative analysis of bulk and single-cell RNA-seq data pinpointed SPP1 + macrophages as an unfavorable cell type, while VCAN + macrophages, C1QA + macrophages, and CD8 + T cells were associated with a more favorable prognosis for HCC patients. Subsequent scRNA-seq investigations and in vitro experiments elucidated that SPP1, predominantly secreted by SPP1 + macrophages, inhibits CD8 + T cell proliferation. Finally, targeting SPP1 in tumor-associated macrophages through inhibition led to a shift towards a favorable phenotype. Discussion This study underpins the potential of SPP1 as a translational target in immunotherapy for HCC.
Collapse
Affiliation(s)
- Han Jin
- Central Laboratory, Tianjin Medical University General Hospital, Tianjin, China
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Woonghee Kim
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Meng Yuan
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Xiangyu Li
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Hong Yang
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Mengzhen Li
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Mengnan Shi
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Türkiye
| | - Mathias Uhlen
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH – Royal Institute of Technology, Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
17
|
Blassick CM, Lugagne JB, Dunlop MJ. Dynamic heterogeneity in an E. coli stress response regulon mediates gene activation and antimicrobial peptide tolerance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625634. [PMID: 39677761 PMCID: PMC11642793 DOI: 10.1101/2024.11.27.625634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The bacterial stress response is an intricately regulated system that plays a critical role in cellular resistance to drug treatment. The complexity of this response is further complicated by cell-to-cell heterogeneity in the expression of bacterial stress response genes. These genes are often organized into networks comprising one or more transcriptional regulators that control expression of a suite of downstream genes. While the expression heterogeneity of many of these upstream regulators has been characterized, the way in which this variability affects the larger downstream stress response remains hard to predict, prompting two key questions. First, how does heterogeneity and expression noise in stress response regulators propagate to the diverse downstream genes in their regulons. Second, when expression levels vary, how do multiple downstream genes act together to protect cells from stress. To address these questions, we focus on the transcription factor PhoP, a critical virulence regulator which coordinates pathogenicity in several gram-negative species. We use optogenetic stimulation to precisely control PhoP expression levels and examine how variations in PhoP affect the downstream activation of genes in the PhoP regulon. We find that these downstream genes exhibit differences both in mean expression level and sensitivity to increasing levels of PhoP. These response functions can also vary between individual cells, increasing heterogeneity in the population. We tie these variations to cell survival when bacteria are exposed to a clinically-relevant antimicrobial peptide, showing that high expression of the PhoP-regulon gene pmrD provides a protective effect against Polymyxin B. Overall, we demonstrate that even subtle heterogeneity in expression of a stress response regulator can have clear consequences for enabling bacteria to survive stress.
Collapse
|
18
|
Bonev B, Castelo-Branco G, Chen F, Codeluppi S, Corces MR, Fan J, Heiman M, Harris K, Inoue F, Kellis M, Levine A, Lotfollahi M, Luo C, Maynard KR, Nitzan M, Ramani V, Satijia R, Schirmer L, Shen Y, Sun N, Green GS, Theis F, Wang X, Welch JD, Gokce O, Konopka G, Liddelow S, Macosko E, Ali Bayraktar O, Habib N, Nowakowski TJ. Opportunities and challenges of single-cell and spatially resolved genomics methods for neuroscience discovery. Nat Neurosci 2024; 27:2292-2309. [PMID: 39627587 PMCID: PMC11999325 DOI: 10.1038/s41593-024-01806-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 09/23/2024] [Indexed: 12/13/2024]
Abstract
Over the past decade, single-cell genomics technologies have allowed scalable profiling of cell-type-specific features, which has substantially increased our ability to study cellular diversity and transcriptional programs in heterogeneous tissues. Yet our understanding of mechanisms of gene regulation or the rules that govern interactions between cell types is still limited. The advent of new computational pipelines and technologies, such as single-cell epigenomics and spatially resolved transcriptomics, has created opportunities to explore two new axes of biological variation: cell-intrinsic regulation of cell states and expression programs and interactions between cells. Here, we summarize the most promising and robust technologies in these areas, discuss their strengths and limitations and discuss key computational approaches for analysis of these complex datasets. We highlight how data sharing and integration, documentation, visualization and benchmarking of results contribute to transparency, reproducibility, collaboration and democratization in neuroscience, and discuss needs and opportunities for future technology development and analysis.
Collapse
Affiliation(s)
- Boyan Bonev
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Fei Chen
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - M Ryan Corces
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Jean Fan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Myriam Heiman
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- The Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
| | - Kenneth Harris
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Fumitaka Inoue
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Manolis Kellis
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ariel Levine
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Mo Lotfollahi
- Institute of Computational Biology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Chongyuan Luo
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mor Nitzan
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Racah Institute of Physics, The Hebrew University of Jerusalem, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Vijay Ramani
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, San Francisco, CA, USA
| | - Rahul Satijia
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Lucas Schirmer
- Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yin Shen
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Na Sun
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gilad S Green
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Fabian Theis
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xiao Wang
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joshua D Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Ozgun Gokce
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA.
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Shane Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, NYU Grossman School of Medicine, New York, NY, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
| | - Evan Macosko
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
| | | | - Naomi Habib
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Tomasz J Nowakowski
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
19
|
Dong H, Ma B, Meng Y, Wu Y, Liu Y, Zeng T, Huang J. GRNMOPT: Inference of gene regulatory networks based on a multi-objective optimization approach. Comput Biol Chem 2024; 113:108223. [PMID: 39340962 DOI: 10.1016/j.compbiolchem.2024.108223] [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: 06/17/2024] [Revised: 08/21/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND AND OBJECTIVE The reconstruction of gene regulatory networks (GRNs) stands as a vital approach in deciphering complex biological processes. The application of nonlinear ordinary differential equations (ODEs) models has demonstrated considerable efficacy in predicting GRNs. Notably, the decay rate and time delay are pivotal in authentic gene regulation, yet their systematic determination in ODEs models remains underexplored. The development of a comprehensive optimization framework for the effective estimation of these key parameters is essential for accurate GRN inference. METHOD This study introduces GRNMOPT, an innovative methodology for inferring GRNs from time-series and steady-state data. GRNMOPT employs a combined use of decay rate and time delay in constructing ODEs models to authentically represent gene regulatory processes. It incorporates a multi-objective optimization approach, optimizing decay rate and time delay concurrently to derive Pareto optimal sets for these factors, thereby maximizing accuracy metrics such as AUROC (Area Under the Receiver Operating Characteristic curve) and AUPR (Area Under the Precision-Recall curve). Additionally, the use of XGBoost for calculating feature importance aids in identifying potential regulatory gene links. RESULTS Comprehensive experimental evaluations on two simulated datasets from DREAM4 and three real gene expression datasets (Yeast, In vivo Reverse-engineering and Modeling Assessment [IRMA], and Escherichia coli [E. coli]) reveal that GRNMOPT performs commendably across varying network scales. Furthermore, cross-validation experiments substantiate the robustness of GRNMOPT. CONCLUSION We propose a novel approach called GRNMOPT to infer GRNs based on a multi-objective optimization framework, which effectively improves inference accuracy and provides a powerful tool for GRNs inference.
Collapse
Affiliation(s)
- Heng Dong
- School of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Baoshan Ma
- School of Information Science and Technology, Dalian Maritime University, Dalian 116026, China.
| | - Yangyang Meng
- School of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Yiming Wu
- School of Information Science and Technology, Dalian Maritime University, Dalian 116026, China
| | - Yongjing Liu
- Biomedical big data center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou 310003, China; Zhejiang University Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Tao Zeng
- Biomedical big data center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou 310003, China; Zhejiang University Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Jinyan Huang
- Biomedical big data center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou 310003, China; Zhejiang University Cancer Center, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
20
|
Barman S, Farid FA, Gope HL, Hafiz MFB, Khan NA, Ahmad S, Mansor S. LBF-MI: Limited Boolean Functions and Mutual Information to Infer a Gene Regulatory Network from Time-Series Gene Expression Data. Genes (Basel) 2024; 15:1530. [PMID: 39766797 PMCID: PMC11675687 DOI: 10.3390/genes15121530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND In the realm of system biology, it is a challenging endeavor to infer a gene regulatory network from time-series gene expression data. Numerous Boolean network inference techniques have emerged for reconstructing a gene regulatory network from a time-series gene expression dataset. However, most of these techniques pose scalability concerns given their capability to consider only two to three regulatory genes over a specific target gene. METHODS To overcome this limitation, a novel inference method, LBF-MI, has been proposed in this research. This two-phase method utilizes limited Boolean functions and multivariate mutual information to reconstruct a Boolean gene regulatory network from time-series gene expression data. Initially, Boolean functions are applied to determine the optimum solutions. In case of failure, multivariate mutual information is applied to obtain the optimum solutions. RESULTS This research conducted a performance-comparison experiment between LBF-MI and three other methods: mutual information-based Boolean network inference, context likelihood relatedness, and relevance network. When examined on artificial as well as real-time-series gene expression data, the outcomes exhibited that the proposed LBF-MI method outperformed mutual information-based Boolean network inference, context likelihood relatedness, and relevance network on artificial datasets, and two real Escherichia coli datasets (E. coli gene regulatory network, and SOS response of E. coli regulatory network). CONCLUSIONS LBF-MI's superior performance in gene regulatory network inference enables researchers to uncover the regulatory mechanisms and cellular behaviors of various organisms.
Collapse
Affiliation(s)
- Shohag Barman
- Department of Computer Science and Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Pirojpur 8500, Bangladesh
| | - Fahmid Al Farid
- Faculty of Engineering, Multimedia University, Cyberjaya 63000, Selangor, Malaysia;
| | - Hira Lal Gope
- Faculty of Agricultural Engineering and Technology, Sylhet Agricultural University, Sylhet 3100, Bangladesh;
| | - Md. Ferdous Bin Hafiz
- Department of Computer Science and Engineering, Southeast University, Dhaka 1208, Bangladesh;
| | - Niaz Ashraf Khan
- Department of Computer Science and Engineering, University of Liberal Arts Bangladesh, Dhaka 1207, Bangladesh;
| | - Sabbir Ahmad
- Department of Computer Science and Engineering, University of Chittagong, Chittagong 4331, Bangladesh;
| | - Sarina Mansor
- Faculty of Engineering, Multimedia University, Cyberjaya 63000, Selangor, Malaysia;
| |
Collapse
|
21
|
Wajid MA, Sharma P, Majeed A, Bhat S, Angmo T, Fayaz M, Pal K, Andotra S, Bhat WW, Misra P. Transcriptome-wide investigation and functional characterization reveal a terpene synthase involved in γ-terpinene biosynthesis in Monarda citriodora. Funct Integr Genomics 2024; 24:222. [PMID: 39589550 DOI: 10.1007/s10142-024-01491-z] [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: 08/30/2024] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 11/27/2024]
Abstract
Monarda citriodora Cerv. ex Lag. is a rich source of industrially important compounds like γ-terpinene, carvacrol, thymol and thymoquinone. Understanding the regulation of γ-terpinene biosynthesis, a precursor for other monoterpenes, could facilitate upscaling of these metabolites in M. citriodora. Therefore, the present study aimed to unravel and characterize the terpene synthase (TPS) involved in γ-terpinene biosynthesis. Homology searches revealed 33 TPS members in the transcriptome assembly of M. citriodora. Based on the correlation of expression patterns and phytochemical profile, McTPS22 emerged as the putative TPS for γ-terpinene biosynthesis. Molecular docking suggested geranyl diphosphate (GPP) as a potential substrate for McTPS22. Heterologous expression in Escherichia coli and Nicotiana benthamiana confirmed the role of McTPS22 in γ-terpinene biosynthesis. Both in-silico prediction and confocal microscopy indicated plastidial localization of the McTPS22. Gene co-expression network analysis revealed 507 genes interacting with McTPS22, including 80 transcription factors (TFs). Of these, 46 TFs had binding sites in the McTPS22 promoter, and 36 showed significant correlations with γ-terpinene accumulation, suggesting they may be potential regulators. Promoter analysis indicated regulation by phytohormones and abiotic factors, confirmed by phytohormone elicitation and QRT-PCR. The histochemical GUS staining suggested that McTPS22 is primarily active in the glandular trichomes of M. citriodora. The present work provides insights into the molecular regulation of biosynthesis of γ-terpinene in M. citriodora.
Collapse
Affiliation(s)
- Mir Abdul Wajid
- Plant Sciences and Agrotechnology Division, CSIR-Indian Institute of Integrative Medicine Canal Road, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Priyanka Sharma
- Plant Sciences and Agrotechnology Division, CSIR-Indian Institute of Integrative Medicine Canal Road, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Aasim Majeed
- Plant Sciences and Agrotechnology Division, CSIR-Indian Institute of Integrative Medicine Canal Road, Jammu, 180001, India
| | - Sheetal Bhat
- Plant Sciences and Agrotechnology Division, CSIR-Indian Institute of Integrative Medicine Canal Road, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Tsering Angmo
- Plant Sciences and Agrotechnology Division, CSIR-Indian Institute of Integrative Medicine Canal Road, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Mohd Fayaz
- Plant Sciences and Agrotechnology Division, CSIR-Indian Institute of Integrative Medicine Canal Road, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Koushik Pal
- Plant Sciences and Agrotechnology Division, CSIR-Indian Institute of Integrative Medicine Canal Road, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sonali Andotra
- Plant Sciences and Agrotechnology Division, CSIR-Indian Institute of Integrative Medicine Canal Road, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Wajid Waheed Bhat
- Division of Basic Sciences and Humanities, SKUAST-Kashmir, Shalimar, 190025, Srinagar, India
| | - Prashant Misra
- Plant Sciences and Agrotechnology Division, CSIR-Indian Institute of Integrative Medicine Canal Road, Jammu, 180001, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
22
|
Karamveer, Uzun Y. Approaches for Benchmarking Single-Cell Gene Regulatory Network Methods. Bioinform Biol Insights 2024; 18:11779322241287120. [PMID: 39502448 PMCID: PMC11536393 DOI: 10.1177/11779322241287120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/10/2024] [Indexed: 11/08/2024] Open
Abstract
Gene regulatory networks are powerful tools for modeling genetic interactions that control the expression of genes driving cell differentiation, and single-cell sequencing offers a unique opportunity to build these networks with high-resolution genomic data. There are many proposed computational methods to build these networks using single-cell data, and different approaches are used to benchmark these methods. However, a comprehensive discussion specifically focusing on benchmarking approaches is missing. In this article, we lay the GRN terminology, present an overview of common gold-standard studies and data sets, and define the performance metrics for benchmarking network construction methodologies. We also point out the advantages and limitations of different benchmarking approaches, suggest alternative ground truth data sets that can be used for benchmarking, and specify additional considerations in this context.
Collapse
Affiliation(s)
- Karamveer
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Yasin Uzun
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
23
|
Dong J, Li J, Wang F. Deep Learning in Gene Regulatory Network Inference: A Survey. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:2089-2101. [PMID: 39137088 DOI: 10.1109/tcbb.2024.3442536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Understanding the intricate regulatory relationships among genes is crucial for comprehending the development, differentiation, and cellular response in living systems. Consequently, inferring gene regulatory networks (GRNs) based on observed data has gained significant attention as a fundamental goal in biological applications. The proliferation and diversification of available data present both opportunities and challenges in accurately inferring GRNs. Deep learning, a highly successful technique in various domains, holds promise in aiding GRN inference. Several GRN inference methods employing deep learning models have been proposed; however, the selection of an appropriate method remains a challenge for life scientists. In this survey, we provide a comprehensive analysis of 12 GRN inference methods that leverage deep learning models. We trace the evolution of these major methods and categorize them based on the types of applicable data. We delve into the core concepts and specific steps of each method, offering a detailed evaluation of their effectiveness and scalability across different scenarios. These insights enable us to make informed recommendations. Moreover, we explore the challenges faced by GRN inference methods utilizing deep learning and discuss future directions, providing valuable suggestions for the advancement of data scientists in this field.
Collapse
|
24
|
Zhu H, Slonim D. From Noise to Knowledge: Diffusion Probabilistic Model-Based Neural Inference of Gene Regulatory Networks. J Comput Biol 2024; 31:1087-1103. [PMID: 39387266 PMCID: PMC11698671 DOI: 10.1089/cmb.2024.0607] [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] [Indexed: 10/15/2024] Open
Abstract
Understanding gene regulatory networks (GRNs) is crucial for elucidating cellular mechanisms and advancing therapeutic interventions. Original methods for GRN inference from bulk expression data often struggled with the high dimensionality and inherent noise in the data. Here we introduce RegDiffusion, a new class of Denoising Diffusion Probabilistic Models focusing on the regulatory effects among feature variables. RegDiffusion introduces Gaussian noise to the input data following a diffusion schedule and uses a neural network with a parameterized adjacency matrix to predict the added noise. We show that using this process, GRNs can be learned effectively with a surprisingly simple model architecture. In our benchmark experiments, RegDiffusion shows superior performance compared to several baseline methods in multiple datasets. We also demonstrate that RegDiffusion can infer biologically meaningful regulatory networks from real-world single-cell data sets with over 15,000 genes in under 5 minutes. This work not only introduces a fresh perspective on GRN inference but also highlights the promising capacity of diffusion-based models in the area of single-cell analysis. The RegDiffusion software package and experiment data are available at https://github.com/TuftsBCB/RegDiffusion.
Collapse
Affiliation(s)
- Hao Zhu
- Department of Computer Science, Tufts University, Medford, Massachusetts, USA
| | - Donna Slonim
- Department of Computer Science, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
25
|
Cingiz MÖ. k- Strong Inference Algorithm: A Hybrid Information Theory Based Gene Network Inference Algorithm. Mol Biotechnol 2024; 66:3213-3225. [PMID: 37950851 DOI: 10.1007/s12033-023-00929-2] [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: 02/23/2023] [Accepted: 10/05/2023] [Indexed: 11/13/2023]
Abstract
Gene networks allow researchers to understand the underlying mechanisms between diseases and genes while reducing the need for wet lab experiments. Numerous gene network inference (GNI) algorithms have been presented in the literature to infer accurate gene networks. We proposed a hybrid GNI algorithm, k-Strong Inference Algorithm (ksia), to infer more reliable and robust gene networks from omics datasets. To increase reliability, ksia integrates Pearson correlation coefficient (PCC) and Spearman rank correlation coefficient (SCC) scores to determine mutual information scores between molecules to increase diversity of relation predictions. To infer a more robust gene network, ksia applies three different elimination steps to remove redundant and spurious relations between genes. The performance of ksia was evaluated on microbe microarrays database in the overlap analysis with other GNI algorithms, namely ARACNE, C3NET, CLR, and MRNET. Ksia inferred less number of relations due to its strict elimination steps. However, ksia generally performed better on Escherichia coli (E.coli) and Saccharomyces cerevisiae (yeast) gene expression datasets due to F- measure and precision values. The integration of association estimator scores and three elimination stages slightly increases the performance of ksia based gene networks. Users can access ksia R package and user manual of package via https://github.com/ozgurcingiz/ksia .
Collapse
Affiliation(s)
- Mustafa Özgür Cingiz
- Computer Engineering Department, Faculty of Engineering and Natural Sciences, Bursa Technical University, Mimar Sinan Campus, Yildirim, 16310, Bursa, Turkey.
| |
Collapse
|
26
|
Bustad E, Petry E, Gu O, Griebel BT, Rustad TR, Sherman DR, Yang JH, Ma S. Predicting bacterial fitness in Mycobacterium tuberculosis with transcriptional regulatory network-informed interpretable machine learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614645. [PMID: 39386570 PMCID: PMC11463588 DOI: 10.1101/2024.09.23.614645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis disease, the greatest source of global mortality by a bacterial pathogen. Mtb adapts and responds to diverse stresses such as antibiotics by inducing transcriptional stress-response regulatory programs. Understanding how and when these mycobacterial regulatory programs are activated could enable novel treatment strategies for potentiating the efficacy of new and existing drugs. Here we sought to define and analyze Mtb regulatory programs that modulate bacterial fitness. We assembled a large Mtb RNA expression compendium and applied these to infer a comprehensive Mtb transcriptional regulatory network and compute condition-specific transcription factor activity profiles. We utilized transcriptomic and functional genomics data to train an interpretable machine learning model that can predict Mtb fitness from transcription factor activity profiles. We demonstrated that this transcription factor activity-based model can successfully predict Mtb growth arrest and growth resumption under hypoxia and reaeration using only RNA-seq expression data as a starting point. These integrative network modeling and machine learning analyses thus enable the prediction of mycobacterial fitness under different environmental and genetic contexts. We envision these models can potentially inform the future design of prognostic assays and therapeutic intervention that can cripple Mtb growth and survival to cure tuberculosis disease.
Collapse
Affiliation(s)
- Ethan Bustad
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
| | - Edson Petry
- Center for Emerging and Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark NJ, USA
| | - Oliver Gu
- Center for Emerging and Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark NJ, USA
| | - Braden T. Griebel
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
- Department of Chemical Engineering, University of Washington, Seattle WA, USA
| | | | - David R. Sherman
- Department of Microbiology, University of Washington, Seattle WA, USA
| | - Jason H. Yang
- Center for Emerging and Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark NJ, USA
- Department of Microbiology, Biochemistry, & Molecular Genetics, Rutgers New Jersey Medical School, Newark NJ, USA
| | - Shuyi Ma
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
- Department of Chemical Engineering, University of Washington, Seattle WA, USA
- Department of Pediatrics, University of Washington, Seattle WA, USA
- Pathobiology Graduate Program, Department of Global Health, University of Washington, Seattle WA, USA
| |
Collapse
|
27
|
Deng Y, Mao J, Choi J, Lê Cao KA. StableMate: a statistical method to select stable predictors in omics data. NAR Genom Bioinform 2024; 6:lqae130. [PMID: 39345755 PMCID: PMC11437361 DOI: 10.1093/nargab/lqae130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/16/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Identifying statistical associations between biological variables is crucial to understanding molecular mechanisms. Most association studies are based on correlation or linear regression analyses, but the identified associations often lack reproducibility and interpretability due to the complexity and variability of omics datasets, making it difficult to translate associations into meaningful biological hypotheses. We developed StableMate, a regression framework, to address these challenges through a process of variable selection across heterogeneous datasets. Given datasets from different environments, such as experimental batches, StableMate selects environment-agnostic (stable) and environment-specific predictors in predicting the response of interest. Stable predictors represent robust functional dependencies with the response, and can be used to build regression models that make generalizable predictions in unseen environments. We applied StableMate to (i) RNA sequencing data of breast cancer to discover genes that consistently predict estrogen receptor expression across disease status; (ii) metagenomics data to identify microbial signatures that show persistent association with colon cancer across study cohorts; and (iii) single-cell RNA sequencing data of glioblastoma to discern signature genes associated with the development of pro-tumour microglia regardless of cell location. Our case studies demonstrate that StableMate is adaptable to regression and classification analyses and achieves comprehensive characterization of biological systems for different omics data types.
Collapse
Affiliation(s)
- Yidi Deng
- Melbourne Integrative Genomics, School of Mathematics and Statistics, The University of Melbourne, Royal Parade, Melbourne, 3052, Australia
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Grattan Street, Melbourne, 3010, Australia
| | - Jiadong Mao
- Melbourne Integrative Genomics, School of Mathematics and Statistics, The University of Melbourne, Royal Parade, Melbourne, 3052, Australia
| | - Jarny Choi
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Grattan Street, Melbourne, 3010, Australia
| | - Kim-Anh Lê Cao
- Melbourne Integrative Genomics, School of Mathematics and Statistics, The University of Melbourne, Royal Parade, Melbourne, 3052, Australia
| |
Collapse
|
28
|
Luomaranta M, Grones C, Choudhary S, Milhinhos A, Kalman TA, Nilsson O, Robinson KM, Street NR, Tuominen H. Systems genetic analysis of lignin biosynthesis in Populus tremula. THE NEW PHYTOLOGIST 2024; 243:2157-2174. [PMID: 39072753 DOI: 10.1111/nph.19993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024]
Abstract
The genetic control underlying natural variation in lignin content and composition in trees is not fully understood. We performed a systems genetic analysis to uncover the genetic regulation of lignin biosynthesis in a natural 'SwAsp' population of aspen (Populus tremula) trees. We analyzed gene expression by RNA sequencing (RNA-seq) in differentiating xylem tissues, and lignin content and composition using Pyrolysis-GC-MS in mature wood of 268 trees from 99 genotypes. Abundant variation was observed for lignin content and composition, and genome-wide association study identified proteins in the pentose phosphate pathway and arabinogalactan protein glycosylation among the top-ranked genes that are associated with these traits. Variation in gene expression and the associated genetic polymorphism was revealed through the identification of 312 705 local and 292 003 distant expression quantitative trait loci (eQTL). A co-expression network analysis suggested modularization of lignin biosynthesis and novel functions for the lignin-biosynthetic CINNAMYL ALCOHOL DEHYDROGENASE 2 and CAFFEOYL-CoA O-METHYLTRANSFERASE 3. PHENYLALANINE AMMONIA LYASE 3 was co-expressed with HOMEOBOX PROTEIN 5 (HB5), and the role of HB5 in stimulating lignification was demonstrated in transgenic trees. The systems genetic approach allowed linking natural variation in lignin biosynthesis to trees´ responses to external cues such as mechanical stimulus and nutrient availability.
Collapse
Affiliation(s)
- Mikko Luomaranta
- Umeå Plant Science Centre, Department of Plant Physiology, Umeå University, 90187, Umeå, Sweden
| | - Carolin Grones
- Umeå Plant Science Centre, Department of Plant Physiology, Umeå University, 90187, Umeå, Sweden
| | - Shruti Choudhary
- Department of Forest Genetics and Plant Physiology, Umeå Plant Science Centre, Swedish University of Agricultural Sciences, 90183, Umeå, Sweden
| | - Ana Milhinhos
- Umeå Plant Science Centre, Department of Plant Physiology, Umeå University, 90187, Umeå, Sweden
| | - Teitur Ahlgren Kalman
- Umeå Plant Science Centre, Department of Plant Physiology, Umeå University, 90187, Umeå, Sweden
| | - Ove Nilsson
- Department of Forest Genetics and Plant Physiology, Umeå Plant Science Centre, Swedish University of Agricultural Sciences, 90183, Umeå, Sweden
| | - Kathryn M Robinson
- Umeå Plant Science Centre, Department of Plant Physiology, Umeå University, 90187, Umeå, Sweden
| | - Nathaniel R Street
- Umeå Plant Science Centre, Department of Plant Physiology, Umeå University, 90187, Umeå, Sweden
- SciLifeLab, Umeå University, 90187, Umeå, Sweden
| | - Hannele Tuominen
- Department of Forest Genetics and Plant Physiology, Umeå Plant Science Centre, Swedish University of Agricultural Sciences, 90183, Umeå, Sweden
| |
Collapse
|
29
|
Canovi C, Stojkovič K, Benítez AA, Delhomme N, Egertsdotter U, Street NR. A resource of identified and annotated lincRNAs expressed during somatic embryogenesis development in Norway spruce. PHYSIOLOGIA PLANTARUM 2024; 176:e14537. [PMID: 39319989 DOI: 10.1111/ppl.14537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/26/2024]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as important regulators of many biological processes, although their regulatory roles remain poorly characterized in woody plants, especially in gymnosperms. A major challenge of working with lncRNAs is to assign functional annotations, since they have a low coding potential and low cross-species conservation. We utilised an existing RNA-Sequencing resource and performed short RNA sequencing of somatic embryogenesis developmental stages in Norway spruce (Picea abies L. Karst). We implemented a pipeline to identify lncRNAs located within the intergenic space (lincRNAs) and generated a co-expression network including protein coding, lincRNA and miRNA genes. To assign putative functional annotation, we employed a guilt-by-association approach using the co-expression network and integrated these results with annotation assigned using semantic similarity and co-expression. Moreover, we evaluated the relationship between lincRNAs and miRNAs, and identified which lincRNAs are conserved in other species. We identified lincRNAs with clear evidence of differential expression during somatic embryogenesis and used network connectivity to identify those with the greatest regulatory potential. This work provides the most comprehensive view of lincRNAs in Norway spruce and is the first study to perform global identification of lincRNAs during somatic embryogenesis in conifers. The data have been integrated into the expression visualisation tools at the PlantGenIE.org web resource to enable easy access to the community. This will facilitate the use of the data to address novel questions about the role of lincRNAs in the regulation of embryogenesis and facilitate future comparative genomics studies.
Collapse
Affiliation(s)
- Camilla Canovi
- Umeå Plant Science Centre, Department of Plant Physiology, Umeå University, Umeå, Sweden
| | - Katja Stojkovič
- Umeå Plant Science Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Aarón Ayllón Benítez
- Umeå Plant Science Centre, Department of Plant Physiology, Umeå University, Umeå, Sweden
| | - Nicolas Delhomme
- Umeå Plant Science Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Ulrika Egertsdotter
- Umeå Plant Science Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
- Renewable Bioproducts Institute, Georgia Institute of Technology Atlanta, USA
| | - Nathaniel R Street
- Umeå Plant Science Centre, Department of Plant Physiology, Umeå University, Umeå, Sweden
- SciLifeLab, Umeå University, Umeå, Sweden
| |
Collapse
|
30
|
Segura-Ortiz A, García-Nieto J, Aldana-Montes JF, Navas-Delgado I. Multi-objective context-guided consensus of a massive array of techniques for the inference of Gene Regulatory Networks. Comput Biol Med 2024; 179:108850. [PMID: 39013340 DOI: 10.1016/j.compbiomed.2024.108850] [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: 02/08/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND AND OBJECTIVE Gene Regulatory Network (GRN) inference is a fundamental task in biology and medicine, as it enables a deeper understanding of the intricate mechanisms of gene expression present in organisms. This bioinformatics problem has been addressed in the literature through multiple computational approaches. Techniques developed for inferring from expression data have employed Bayesian networks, ordinary differential equations (ODEs), machine learning, information theory measures and neural networks, among others. The diversity of implementations and their respective customization have led to the emergence of many tools and multiple specialized domains derived from them, understood as subsets of networks with specific characteristics that are challenging to detect a priori. This specialization has introduced significant uncertainty when choosing the most appropriate technique for a particular dataset. This proposal, named MO-GENECI, builds upon the basic idea of the previous proposal GENECI and optimizes consensus among different inference techniques, through a carefully refined multi-objective evolutionary algorithm guided by various objective functions, linked to the biological context at hand. METHODS MO-GENECI has been tested on an extensive and diverse academic benchmark of 106 gene regulatory networks from multiple sources and sizes. The evaluation of MO-GENECI compared its performance to individual techniques using key metrics (AUROC and AUPR) for gene regulatory network inference. Friedman's statistical ranking provided an ordered classification, followed by non-parametric Holm tests to determine statistical significance. RESULTS MO-GENECI's Pareto front approximation facilitates easy selection of an appropriate solution based on generic input data characteristics. The best solution consistently emerged as the winner in all statistical tests, and in many cases, the median precision solution showed no statistically significant difference compared to the winner. CONCLUSIONS MO-GENECI has not only demonstrated achieving more accurate results than individual techniques, but has also overcome the uncertainty associated with the initial choice due to its flexibility and adaptability. It is shown intelligently to select the most suitable techniques for each case. The source code is hosted in a public repository at GitHub under MIT license: https://github.com/AdrianSeguraOrtiz/MO-GENECI. Moreover, to facilitate its installation and use, the software associated with this implementation has been encapsulated in a Python package available at PyPI: https://pypi.org/project/geneci/.
Collapse
Affiliation(s)
- Adrián Segura-Ortiz
- Department de Lenguajes y Ciencias de la Computación, ITIS Software, Universidad de Málaga, Málaga, 29071, Spain.
| | - José García-Nieto
- Department de Lenguajes y Ciencias de la Computación, ITIS Software, Universidad de Málaga, Málaga, 29071, Spain; Biomedical Research Institute of Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
| | - José F Aldana-Montes
- Department de Lenguajes y Ciencias de la Computación, ITIS Software, Universidad de Málaga, Málaga, 29071, Spain; Biomedical Research Institute of Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
| | - Ismael Navas-Delgado
- Department de Lenguajes y Ciencias de la Computación, ITIS Software, Universidad de Málaga, Málaga, 29071, Spain; Biomedical Research Institute of Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
| |
Collapse
|
31
|
Kernfeld E, Keener R, Cahan P, Battle A. Transcriptome data are insufficient to control false discoveries in regulatory network inference. Cell Syst 2024; 15:709-724.e13. [PMID: 39173585 PMCID: PMC11642480 DOI: 10.1016/j.cels.2024.07.006] [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: 05/24/2023] [Revised: 05/31/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024]
Abstract
Inference of causal transcriptional regulatory networks (TRNs) from transcriptomic data suffers notoriously from false positives. Approaches to control the false discovery rate (FDR), for example, via permutation, bootstrapping, or multivariate Gaussian distributions, suffer from several complications: difficulty in distinguishing direct from indirect regulation, nonlinear effects, and causal structure inference requiring "causal sufficiency," meaning experiments that are free of any unmeasured, confounding variables. Here, we use a recently developed statistical framework, model-X knockoffs, to control the FDR while accounting for indirect effects, nonlinear dose-response, and user-provided covariates. We adjust the procedure to estimate the FDR correctly even when measured against incomplete gold standards. However, benchmarking against chromatin immunoprecipitation (ChIP) and other gold standards reveals higher observed than reported FDR. This indicates that unmeasured confounding is a major driver of FDR in TRN inference. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Eric Kernfeld
- Department of Biomedical Engineering, Johns Hopkins University, 3400 N. Charles Street, Wyman Park Building, Suite 400 West, Baltimore, MD 21218, USA
| | - Rebecca Keener
- Department of Biomedical Engineering, Johns Hopkins University, 3400 N. Charles Street, Wyman Park Building, Suite 400 West, Baltimore, MD 21218, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Johns Hopkins University, 3400 N. Charles Street, Wyman Park Building, Suite 400 West, Baltimore, MD 21218, USA; Institute for Cell Engineering, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD, USA.
| | - Alexis Battle
- Department of Biomedical Engineering, Johns Hopkins University, 3400 N. Charles Street, Wyman Park Building, Suite 400 West, Baltimore, MD 21218, USA; Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins Medicine, Baltimore, MD, USA; Malone Center for Engineering and Healthcare, Johns Hopkins University, Baltimore, MD, USA; Data Science and AI Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
32
|
Zitnik M, Li MM, Wells A, Glass K, Morselli Gysi D, Krishnan A, Murali TM, Radivojac P, Roy S, Baudot A, Bozdag S, Chen DZ, Cowen L, Devkota K, Gitter A, Gosline SJC, Gu P, Guzzi PH, Huang H, Jiang M, Kesimoglu ZN, Koyuturk M, Ma J, Pico AR, Pržulj N, Przytycka TM, Raphael BJ, Ritz A, Sharan R, Shen Y, Singh M, Slonim DK, Tong H, Yang XH, Yoon BJ, Yu H, Milenković T. Current and future directions in network biology. BIOINFORMATICS ADVANCES 2024; 4:vbae099. [PMID: 39143982 PMCID: PMC11321866 DOI: 10.1093/bioadv/vbae099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/31/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024]
Abstract
Summary Network biology is an interdisciplinary field bridging computational and biological sciences that has proved pivotal in advancing the understanding of cellular functions and diseases across biological systems and scales. Although the field has been around for two decades, it remains nascent. It has witnessed rapid evolution, accompanied by emerging challenges. These stem from various factors, notably the growing complexity and volume of data together with the increased diversity of data types describing different tiers of biological organization. We discuss prevailing research directions in network biology, focusing on molecular/cellular networks but also on other biological network types such as biomedical knowledge graphs, patient similarity networks, brain networks, and social/contact networks relevant to disease spread. In more detail, we highlight areas of inference and comparison of biological networks, multimodal data integration and heterogeneous networks, higher-order network analysis, machine learning on networks, and network-based personalized medicine. Following the overview of recent breakthroughs across these five areas, we offer a perspective on future directions of network biology. Additionally, we discuss scientific communities, educational initiatives, and the importance of fostering diversity within the field. This article establishes a roadmap for an immediate and long-term vision for network biology. Availability and implementation Not applicable.
Collapse
Affiliation(s)
- Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, United States
| | - Michelle M Li
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, United States
| | - Aydin Wells
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
- Lucy Family Institute for Data and Society, University of Notre Dame, Notre Dame, IN 46556, United States
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Deisy Morselli Gysi
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States
- Department of Statistics, Federal University of Paraná, Curitiba, Paraná 81530-015, Brazil
- Department of Physics, Northeastern University, Boston, MA 02115, United States
| | - Arjun Krishnan
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - T M Murali
- Department of Computer Science, Virginia Tech, Blacksburg, VA 24061, United States
| | - Predrag Radivojac
- Khoury College of Computer Sciences, Northeastern University, Boston, MA 02115, United States
| | - Sushmita Roy
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53715, United States
- Wisconsin Institute for Discovery, Madison, WI 53715, United States
| | - Anaïs Baudot
- Aix Marseille Université, INSERM, MMG, Marseille, France
| | - Serdar Bozdag
- Department of Computer Science and Engineering, University of North Texas, Denton, TX 76203, United States
- Department of Mathematics, University of North Texas, Denton, TX 76203, United States
| | - Danny Z Chen
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Lenore Cowen
- Department of Computer Science, Tufts University, Medford, MA 02155, United States
| | - Kapil Devkota
- Department of Computer Science, Tufts University, Medford, MA 02155, United States
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53715, United States
- Morgridge Institute for Research, Madison, WI 53715, United States
| | - Sara J C Gosline
- Biological Sciences Division, Pacific Northwest National Laboratory, Seattle, WA 98109, United States
| | - Pengfei Gu
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Pietro H Guzzi
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, 88100, Italy
| | - Heng Huang
- Department of Computer Science, University of Maryland College Park, College Park, MD 20742, United States
| | - Meng Jiang
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Ziynet Nesibe Kesimoglu
- Department of Computer Science and Engineering, University of North Texas, Denton, TX 76203, United States
- National Center of Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20814, United States
| | - Mehmet Koyuturk
- Department of Computer and Data Sciences, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Jian Ma
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, United States
| | - Alexander R Pico
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, United States
| | - Nataša Pržulj
- Department of Computer Science, University College London, London, WC1E 6BT, England
- ICREA, Catalan Institution for Research and Advanced Studies, Barcelona, 08010, Spain
- Barcelona Supercomputing Center (BSC), Barcelona, 08034, Spain
| | - Teresa M Przytycka
- National Center of Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20814, United States
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, Princeton, NJ 08544, United States
| | - Anna Ritz
- Department of Biology, Reed College, Portland, OR 97202, United States
| | - Roded Sharan
- School of Computer Science, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Yang Shen
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, United States
| | - Mona Singh
- Department of Computer Science, Princeton University, Princeton, NJ 08544, United States
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, United States
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, MA 02155, United States
| | - Hanghang Tong
- Department of Computer Science, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States
| | - Xinan Holly Yang
- Department of Pediatrics, University of Chicago, Chicago, IL 60637, United States
| | - Byung-Jun Yoon
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, United States
- Computational Science Initiative, Brookhaven National Laboratory, Upton, NY 11973, United States
| | - Haiyuan Yu
- Department of Computational Biology, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, United States
| | - Tijana Milenković
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
- Lucy Family Institute for Data and Society, University of Notre Dame, Notre Dame, IN 46556, United States
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, United States
| |
Collapse
|
33
|
Jiang H, Wang Y, Yin C, Pan H, Chen L, Feng K, Chang Y, Sun H. SLIVER: Unveiling large scale gene regulatory networks of single-cell transcriptomic data through causal structure learning and modules aggregation. Comput Biol Med 2024; 178:108690. [PMID: 38879931 DOI: 10.1016/j.compbiomed.2024.108690] [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: 04/04/2024] [Revised: 05/19/2024] [Accepted: 06/01/2024] [Indexed: 06/18/2024]
Abstract
Prevalent Gene Regulatory Network (GRN) construction methods rely on generalized correlation analysis. However, in biological systems, regulation is essentially a causal relationship that cannot be adequately captured solely through correlation. Therefore, it is more reasonable to infer GRNs from a causal perspective. Existing causal discovery algorithms typically rely on Directed Acyclic Graphs (DAGs) to model causal relationships, but it often requires traversing the entire network, which result in computational demands skyrocketing as the number of nodes grows and make causal discovery algorithms only suitable for small networks with one or two hundred nodes or fewer. In this study, we propose the SLIVER (cauSaL dIscovery Via dimEnsionality Reduction) algorithm which integrates causal structural equation model and graph decomposition. SLIVER introduces a set of factor nodes, serving as abstractions of different functional modules to integrate the regulatory relationships between genes based on their respective functions or pathways, thus reducing the GRN to the product of two low-dimensional matrices. Subsequently, we employ the structural causal model (SCM) to learn the GRN within the gene node space, enforce the DAG constraint in the low-dimensional space, and guide each factor to aggregate various functions through cosine similarity. We evaluate the performance of the SLIVER algorithm on 12 real single cell transcriptomic datasets, and demonstrate it outperforms other 12 widely used methods both in GRN inference performance and computational resource usage. The analysis of the gene information integrated by factor nodes also demonstrate the biological explanation of factor nodes in GRNs. We apply it to scRNA-seq of Type 2 diabetes mellitus to capture the transcriptional regulatory structural changes of β cells under high insulin demand.
Collapse
Affiliation(s)
- Hongyang Jiang
- School of Artificial Intelligence, Jilin University, Changchun, 130012, China
| | - Yuezhu Wang
- School of Artificial Intelligence, Jilin University, Changchun, 130012, China
| | - Chaoyi Yin
- School of Artificial Intelligence, Jilin University, Changchun, 130012, China
| | - Hao Pan
- College of Software, Jilin University, Changchun, 130012, China
| | - Liqun Chen
- School of Artificial Intelligence, Jilin University, Changchun, 130012, China
| | - Ke Feng
- School of Artificial Intelligence, Jilin University, Changchun, 130012, China
| | - Yi Chang
- School of Artificial Intelligence, Jilin University, Changchun, 130012, China; International Center of Future Science, Jilin University, Changchun, China; Engineering Research Center of Knowledge-Driven Human-Machine Intelligence, MOE, China
| | - Huiyan Sun
- School of Artificial Intelligence, Jilin University, Changchun, 130012, China; International Center of Future Science, Jilin University, Changchun, China; Engineering Research Center of Knowledge-Driven Human-Machine Intelligence, MOE, China.
| |
Collapse
|
34
|
Loers JU, Vermeirssen V. A single-cell multimodal view on gene regulatory network inference from transcriptomics and chromatin accessibility data. Brief Bioinform 2024; 25:bbae382. [PMID: 39207727 PMCID: PMC11359808 DOI: 10.1093/bib/bbae382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/27/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Eukaryotic gene regulation is a combinatorial, dynamic, and quantitative process that plays a vital role in development and disease and can be modeled at a systems level in gene regulatory networks (GRNs). The wealth of multi-omics data measured on the same samples and even on the same cells has lifted the field of GRN inference to the next stage. Combinations of (single-cell) transcriptomics and chromatin accessibility allow the prediction of fine-grained regulatory programs that go beyond mere correlation of transcription factor and target gene expression, with enhancer GRNs (eGRNs) modeling molecular interactions between transcription factors, regulatory elements, and target genes. In this review, we highlight the key components for successful (e)GRN inference from (sc)RNA-seq and (sc)ATAC-seq data exemplified by state-of-the-art methods as well as open challenges and future developments. Moreover, we address preprocessing strategies, metacell generation and computational omics pairing, transcription factor binding site detection, and linear and three-dimensional approaches to identify chromatin interactions as well as dynamic and causal eGRN inference. We believe that the integration of transcriptomics together with epigenomics data at a single-cell level is the new standard for mechanistic network inference, and that it can be further advanced with integrating additional omics layers and spatiotemporal data, as well as with shifting the focus towards more quantitative and causal modeling strategies.
Collapse
Affiliation(s)
- Jens Uwe Loers
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Zwijnaarde-Technologiepark 71, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Vanessa Vermeirssen
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Zwijnaarde-Technologiepark 71, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| |
Collapse
|
35
|
Cui W, Long Q, Xiao M, Wang X, Feng G, Li X, Wang P, Zhou Y. Refining computational inference of gene regulatory networks: integrating knockout data within a multi-task framework. Brief Bioinform 2024; 25:bbae361. [PMID: 39082651 PMCID: PMC11289685 DOI: 10.1093/bib/bbae361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/09/2024] [Accepted: 07/16/2024] [Indexed: 08/03/2024] Open
Abstract
Constructing accurate gene regulatory network s (GRNs), which reflect the dynamic governing process between genes, is critical to understanding the diverse cellular process and unveiling the complexities in biological systems. With the development of computer sciences, computational-based approaches have been applied to the GRNs inference task. However, current methodologies face challenges in effectively utilizing existing topological information and prior knowledge of gene regulatory relationships, hindering the comprehensive understanding and accurate reconstruction of GRNs. In response, we propose a novel graph neural network (GNN)-based Multi-Task Learning framework for GRN reconstruction, namely MTLGRN. Specifically, we first encode the gene promoter sequences and the gene biological features and concatenate the corresponding feature representations. Then, we construct a multi-task learning framework including GRN reconstruction, Gene knockout predict, and Gene expression matrix reconstruction. With joint training, MTLGRN can optimize the gene latent representations by integrating gene knockout information, promoter characteristics, and other biological attributes. Extensive experimental results demonstrate superior performance compared with state-of-the-art baselines on the GRN reconstruction task, efficiently leveraging biological knowledge and comprehensively understanding the gene regulatory relationships. MTLGRN also pioneered attempts to simulate gene knockouts on bulk data by incorporating gene knockout information.
Collapse
Affiliation(s)
- Wentao Cui
- Computer Network Information Center, Chinese Academy of Sciences, CAS Informatization Plaza No. 2 Dong Sheng Nan Lu, Haidian District, Beijing, 100083, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Qingqing Long
- Computer Network Information Center, Chinese Academy of Sciences, CAS Informatization Plaza No. 2 Dong Sheng Nan Lu, Haidian District, Beijing, 100083, China
| | - Meng Xiao
- Computer Network Information Center, Chinese Academy of Sciences, CAS Informatization Plaza No. 2 Dong Sheng Nan Lu, Haidian District, Beijing, 100083, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Xuezhi Wang
- Computer Network Information Center, Chinese Academy of Sciences, CAS Informatization Plaza No. 2 Dong Sheng Nan Lu, Haidian District, Beijing, 100083, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Guihai Feng
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Shijingshan District, Beijing, 100049, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Xin Li
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Shijingshan District, Beijing, 100049, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Pengfei Wang
- Computer Network Information Center, Chinese Academy of Sciences, CAS Informatization Plaza No. 2 Dong Sheng Nan Lu, Haidian District, Beijing, 100083, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Yuanchun Zhou
- Computer Network Information Center, Chinese Academy of Sciences, CAS Informatization Plaza No. 2 Dong Sheng Nan Lu, Haidian District, Beijing, 100083, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Shijingshan District, Beijing, 100049, China
| |
Collapse
|
36
|
Gómez-Pascual A, Rocamora-Pérez G, Ibanez L, Botía JA. Targeted co-expression networks for the study of traits. Sci Rep 2024; 14:16675. [PMID: 39030261 PMCID: PMC11271532 DOI: 10.1038/s41598-024-67329-7] [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: 12/28/2023] [Accepted: 07/10/2024] [Indexed: 07/21/2024] Open
Abstract
Weighted Gene Co-expression Network Analysis (WGCNA) is a widely used approach for the generation of gene co-expression networks. However, networks generated with this tool usually create large modules with a large set of functional annotations hard to decipher. We have developed TGCN, a new method to create Targeted Gene Co-expression Networks. This method identifies the transcripts that best predict the trait of interest based on gene expression using a refinement of the LASSO regression. Then, it builds the co-expression modules around those transcripts. Algorithm properties were characterized using the expression of 13 brain regions from the Genotype-Tissue Expression project. When comparing our method with WGCNA, TGCN networks lead to more precise modules that have more specific and yet rich biological meaning. Then, we illustrate its applicability by creating an APP-TGCN on The Religious Orders Study and Memory and Aging Project dataset, aiming to identify the molecular pathways specifically associated with APP role in Alzheimer's disease. Main biological findings were further validated in two independent cohorts. In conclusion, we provide a new framework that serves to create targeted networks that are smaller, biologically relevant and useful in high throughput hypothesis driven research. The TGCN R package is available on Github: https://github.com/aliciagp/TGCN .
Collapse
Affiliation(s)
- A Gómez-Pascual
- Communications Engineering and Information Department, University of Murcia, 30100, Murcia, Spain
| | - G Rocamora-Pérez
- Department of Genetics and Genomic Medicine Research and Teaching, UCL GOS Institute of Child Health, London, WC1N 1EH, UK
| | - L Ibanez
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - J A Botía
- Communications Engineering and Information Department, University of Murcia, 30100, Murcia, Spain.
| |
Collapse
|
37
|
Peng H, Xu J, Liu K, Liu F, Zhang A, Zhang X. EIEPCF: accurate inference of functional gene regulatory networks by eliminating indirect effects from confounding factors. Brief Funct Genomics 2024; 23:373-383. [PMID: 37642217 DOI: 10.1093/bfgp/elad040] [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] [Received: 05/05/2023] [Revised: 07/07/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023] Open
Abstract
Reconstructing functional gene regulatory networks (GRNs) is a primary prerequisite for understanding pathogenic mechanisms and curing diseases in animals, and it also provides an important foundation for cultivating vegetable and fruit varieties that are resistant to diseases and corrosion in plants. Many computational methods have been developed to infer GRNs, but most of the regulatory relationships between genes obtained by these methods are biased. Eliminating indirect effects in GRNs remains a significant challenge for researchers. In this work, we propose a novel approach for inferring functional GRNs, named EIEPCF (eliminating indirect effects produced by confounding factors), which eliminates indirect effects caused by confounding factors. This method eliminates the influence of confounding factors on regulatory factors and target genes by measuring the similarity between their residuals. The validation results of the EIEPCF method on simulation studies, the gold-standard networks provided by the DREAM3 Challenge and the real gene networks of Escherichia coli demonstrate that it achieves significantly higher accuracy compared to other popular computational methods for inferring GRNs. As a case study, we utilized the EIEPCF method to reconstruct the cold-resistant specific GRN from gene expression data of cold-resistant in Arabidopsis thaliana. The source code and data are available at https://github.com/zhanglab-wbgcas/EIEPCF.
Collapse
Affiliation(s)
- Huixiang Peng
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074 China
- University of Chinese Academy of Sciences, Beijing 100049 China
| | - Jing Xu
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074 China
- University of Chinese Academy of Sciences, Beijing 100049 China
| | - Kangchen Liu
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074 China
- University of Chinese Academy of Sciences, Beijing 100049 China
| | - Fang Liu
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074 China
| | - Aidi Zhang
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074 China
| | - Xiujun Zhang
- Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074 China
- Center of Economic Botany, Core Botanical Gardens, Chinese Academy of Sciences, Wuhan 430074, China
| |
Collapse
|
38
|
Chee FT, Harun S, Mohd Daud K, Sulaiman S, Nor Muhammad NA. Exploring gene regulation and biological processes in insects: Insights from omics data using gene regulatory network models. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 189:1-12. [PMID: 38604435 DOI: 10.1016/j.pbiomolbio.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/18/2023] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Gene regulatory network (GRN) comprises complicated yet intertwined gene-regulator relationships. Understanding the GRN dynamics will unravel the complexity behind the observed gene expressions. Insect gene regulation is often complicated due to their complex life cycles and diverse ecological adaptations. The main interest of this review is to have an update on the current mathematical modelling methods of GRNs to explain insect science. Several popular GRN architecture models are discussed, together with examples of applications in insect science. In the last part of this review, each model is compared from different aspects, including network scalability, computation complexity, robustness to noise and biological relevancy.
Collapse
Affiliation(s)
- Fong Ting Chee
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, 43600 UKM Bangi, Selangor, Malaysia
| | - Sarahani Harun
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, 43600 UKM Bangi, Selangor, Malaysia
| | - Kauthar Mohd Daud
- Faculty of Information Science and Technology, Universiti Kebangsaan Malaysia, 43600, UKM Bangi, Selangor, Malaysia
| | - Suhaila Sulaiman
- FGV R&D Sdn Bhd, FGV Innovation Center, PT23417 Lengkuk Teknologi, Bandar Baru Enstek, 71760 Nilai, Negeri Sembilan, Malaysia
| | - Nor Azlan Nor Muhammad
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, 43600 UKM Bangi, Selangor, Malaysia.
| |
Collapse
|
39
|
Yu J, Leng J, Yuan F, Sun D, Wu LY. Reverse network diffusion to remove indirect noise for better inference of gene regulatory networks. Bioinformatics 2024; 40:btae435. [PMID: 38963312 PMCID: PMC11236096 DOI: 10.1093/bioinformatics/btae435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/05/2024] Open
Abstract
MOTIVATION Gene regulatory networks (GRNs) are vital tools for delineating regulatory relationships between transcription factors and their target genes. The boom in computational biology and various biotechnologies has made inferring GRNs from multi-omics data a hot topic. However, when networks are constructed from gene expression data, they often suffer from false-positive problem due to the transitive effects of correlation. The presence of spurious noise edges obscures the real gene interactions, which makes downstream analyses, such as detecting gene function modules and predicting disease-related genes, difficult and inefficient. Therefore, there is an urgent and compelling need to develop network denoising methods to improve the accuracy of GRN inference. RESULTS In this study, we proposed a novel network denoising method named REverse Network Diffusion On Random walks (RENDOR). RENDOR is designed to enhance the accuracy of GRNs afflicted by indirect effects. RENDOR takes noisy networks as input, models higher-order indirect interactions between genes by transitive closure, eliminates false-positive effects using the inverse network diffusion method, and produces refined networks as output. We conducted a comparative assessment of GRN inference accuracy before and after denoising on simulated networks and real GRNs. Our results emphasized that the network derived from RENDOR more accurately and effectively captures gene interactions. This study demonstrates the significance of removing network indirect noise and highlights the effectiveness of the proposed method in enhancing the signal-to-noise ratio of noisy networks. AVAILABILITY AND IMPLEMENTATION The R package RENDOR is provided at https://github.com/Wu-Lab/RENDOR and other source code and data are available at https://github.com/Wu-Lab/RENDOR-reproduce.
Collapse
Affiliation(s)
- Jiating Yu
- School of Mathematics and Statistics, Nanjing University of Information Science & Technology, Nanjing 210044, China
- IAM, MADIS, NCMIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiacheng Leng
- IAM, MADIS, NCMIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Zhejiang Lab, Hangzhou 311121, China
| | - Fan Yuan
- IAM, MADIS, NCMIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Duanchen Sun
- School of Mathematics, Shandong University, Jinan 250100, China
| | - Ling-Yun Wu
- IAM, MADIS, NCMIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
40
|
Wang Y, Zhou F, Guan J. SFINN: inferring gene regulatory network from single-cell and spatial transcriptomic data with shared factor neighborhood and integrated neural network. Bioinformatics 2024; 40:btae433. [PMID: 38950180 PMCID: PMC11236097 DOI: 10.1093/bioinformatics/btae433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/03/2024] Open
Abstract
MOTIVATION The rise of single-cell RNA sequencing (scRNA-seq) technology presents new opportunities for constructing detailed cell type-specific gene regulatory networks (GRNs) to study cell heterogeneity. However, challenges caused by noises, technical errors, and dropout phenomena in scRNA-seq data pose significant obstacles to GRN inference, making the design of accurate GRN inference algorithms still essential. The recent growth of both single-cell and spatial transcriptomic sequencing data enables the development of supervised deep learning methods to infer GRNs on these diverse single-cell datasets. RESULTS In this study, we introduce a novel deep learning framework based on shared factor neighborhood and integrated neural network (SFINN) for inferring potential interactions and causalities between transcription factors and target genes from single-cell and spatial transcriptomic data. SFINN utilizes shared factor neighborhood to construct cellular neighborhood network based on gene expression data and additionally integrates cellular network generated from spatial location information. Subsequently, the cell adjacency matrix and gene pair expression are fed into an integrated neural network framework consisting of a graph convolutional neural network and a fully-connected neural network to determine whether the genes interact. Performance evaluation in the tasks of gene interaction and causality prediction against the existing GRN reconstruction algorithms demonstrates the usability and competitiveness of SFINN across different kinds of data. SFINN can be applied to infer GRNs from conventional single-cell sequencing data and spatial transcriptomic data. AVAILABILITY AND IMPLEMENTATION SFINN can be accessed at GitHub: https://github.com/JGuan-lab/SFINN.
Collapse
Affiliation(s)
- Yongjie Wang
- Department of Automation, Xiamen University, Xiamen, Fujian 361102, China
| | - Fengfan Zhou
- Department of Automation, Xiamen University, Xiamen, Fujian 361102, China
| | - Jinting Guan
- Department of Automation, Xiamen University, Xiamen, Fujian 361102, China
- Key Laboratory of System Control and Information Processing, Ministry of Education, Shanghai 200240, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
41
|
Moeckel C, Mouratidis I, Chantzi N, Uzun Y, Georgakopoulos-Soares I. Advances in computational and experimental approaches for deciphering transcriptional regulatory networks: Understanding the roles of cis-regulatory elements is essential, and recent research utilizing MPRAs, STARR-seq, CRISPR-Cas9, and machine learning has yielded valuable insights. Bioessays 2024; 46:e2300210. [PMID: 38715516 PMCID: PMC11444527 DOI: 10.1002/bies.202300210] [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: 10/31/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024]
Abstract
Understanding the influence of cis-regulatory elements on gene regulation poses numerous challenges given complexities stemming from variations in transcription factor (TF) binding, chromatin accessibility, structural constraints, and cell-type differences. This review discusses the role of gene regulatory networks in enhancing understanding of transcriptional regulation and covers construction methods ranging from expression-based approaches to supervised machine learning. Additionally, key experimental methods, including MPRAs and CRISPR-Cas9-based screening, which have significantly contributed to understanding TF binding preferences and cis-regulatory element functions, are explored. Lastly, the potential of machine learning and artificial intelligence to unravel cis-regulatory logic is analyzed. These computational advances have far-reaching implications for precision medicine, therapeutic target discovery, and the study of genetic variations in health and disease.
Collapse
Affiliation(s)
- Camille Moeckel
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Ioannis Mouratidis
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Nikol Chantzi
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Yasin Uzun
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
42
|
Jimenez-Cyrus D, Adusumilli VS, Stempel MH, Maday S, Ming GL, Song H, Bond AM. Molecular cascade reveals sequential milestones underlying hippocampal neural stem cell development into an adult state. Cell Rep 2024; 43:114339. [PMID: 38852158 PMCID: PMC11320877 DOI: 10.1016/j.celrep.2024.114339] [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: 05/27/2023] [Revised: 04/16/2024] [Accepted: 05/23/2024] [Indexed: 06/11/2024] Open
Abstract
Quiescent adult neural stem cells (NSCs) in the mammalian brain arise from proliferating NSCs during development. Beyond acquisition of quiescence, an adult NSC hallmark, little is known about the process, milestones, and mechanisms underlying the transition of developmental NSCs to an adult NSC state. Here, we performed targeted single-cell RNA-seq analysis to reveal the molecular cascade underlying NSC development in the early postnatal mouse dentate gyrus. We identified two sequential steps, first a transition to quiescence followed by further maturation, each of which involved distinct changes in metabolic gene expression. Direct metabolic analysis uncovered distinct milestones, including an autophagy burst before NSC quiescence acquisition and cellular reactive oxygen species level elevation along NSC maturation. Functionally, autophagy is important for the NSC transition to quiescence during early postnatal development. Together, our study reveals a multi-step process with defined milestones underlying establishment of the adult NSC pool in the mammalian brain.
Collapse
Affiliation(s)
- Dennisse Jimenez-Cyrus
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vijay S Adusumilli
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Max H Stempel
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sandra Maday
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; The Epigenetics Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Allison M Bond
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
43
|
Mansoor S, Tripathi P, Ghimire A, Hamid S, Abd El-Moniem D, Chung YS, Kim Y. Comparative transcriptomic analysis of the nodulation-competent zone and inference of transcription regulatory network in silicon applied Glycine max [L.]-Merr. Roots. PLANT CELL REPORTS 2024; 43:169. [PMID: 38864921 PMCID: PMC11169057 DOI: 10.1007/s00299-024-03250-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024]
Abstract
KEY MESSAGE The study unveils Si's regulatory influence by regulating DEGs, TFs, and TRs. Further bHLH subfamily and auxin transporter pathway elucidates the mechanisms enhancing root development and nodulation. Soybean is a globally important crop serving as a primary source of vegetable protein for millions of individuals. The roots of these plants harbour essential nitrogen fixing structures called nodules. This study investigates the multifaceted impact of silicon (Si) application on soybean, with a focus on root development, and nodulation employing comprehensive transcriptomic analyses and gene regulatory network. RNA sequence analysis was utilised to examine the change in gene expression and identify the noteworthy differentially expressed genes (DEGs) linked to the enhancement of soybean root nodulation and root development. A set of 316 genes involved in diverse biological and molecular pathways are identified, with emphasis on transcription factors (TFs) and transcriptional regulators (TRs). The study uncovers TF and TR genes, categorized into 68 distinct families, highlighting the intricate regulatory landscape influenced by Si in soybeans. Upregulated most important bHLH subfamily and the involvement of the auxin transporter pathway underscore the molecular mechanisms contributing to enhanced root development and nodulation. The study bridges insights from other research, reinforcing Si's impact on stress-response pathways and phenylpropanoid biosynthesis crucial for nodulation. The study reveals significant alterations in gene expression patterns associated with cellular component functions, root development, and nodulation in response to Si.
Collapse
Affiliation(s)
- Sheikh Mansoor
- Department of Plant Resources and Environment, Jeju National University, Jeju, 63243, Republic of Korea
| | - Pooja Tripathi
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
- Department of Horticulture and Crop Science, The Ohio State University, Columbus, OH, USA
| | - Amit Ghimire
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
- Department of Integrative Biology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Saira Hamid
- Watson Crick Centre for Molecular Medicine, Islamia University of Science and Technology, Awantipora, Pulwama, J&K, India
| | - Diaa Abd El-Moniem
- Department of Plant Production (Genetic Branch), Faculty of Environmental Agricultural Sciences, Arish University, El-Arish, 45511, Egypt
| | - Yong Suk Chung
- Department of Plant Resources and Environment, Jeju National University, Jeju, 63243, Republic of Korea.
| | - Yoonha Kim
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Department of Integrative Biology, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
44
|
Wu S, Jin K, Tang M, Xia Y, Gao W. Inference of Gene Regulatory Networks Based on Multi-view Hierarchical Hypergraphs. Interdiscip Sci 2024; 16:318-332. [PMID: 38342857 DOI: 10.1007/s12539-024-00604-3] [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: 08/16/2023] [Revised: 11/26/2023] [Accepted: 01/03/2024] [Indexed: 02/13/2024]
Abstract
Since gene regulation is a complex process in which multiple genes act simultaneously, accurately inferring gene regulatory networks (GRNs) is a long-standing challenge in systems biology. Although graph neural networks can formally describe intricate gene expression mechanisms, current GRN inference methods based on graph learning regard only transcription factor (TF)-target gene interactions as pairwise relationships, and cannot model the many-to-many high-order regulatory patterns that prevail among genes. Moreover, these methods often rely on limited prior regulatory knowledge, ignoring the structural information of GRNs in gene expression profiles. Therefore, we propose a multi-view hierarchical hypergraphs GRN (MHHGRN) inference model. Specifically, multiple heterogeneous biological information is integrated to construct multi-view hierarchical hypergraphs of TFs and target genes, using hypergraph convolution networks to model higher order complex regulatory relationships. Meanwhile, the coupled information diffusion mechanism and the cross-domain messaging mechanism facilitate the information sharing between genes to optimise gene embedding representations. Finally, a unique channel attention mechanism is used to adaptively learn feature representations from multiple views for GRN inference. Experimental results show that MHHGRN achieves better results than the baseline methods on the E. coli and S. cerevisiae benchmark datasets of the DREAM5 challenge, and it has excellent cross-species generalization, achieving comparable or better performance on scRNA-seq datasets from five mouse and two human cell lines.
Collapse
Affiliation(s)
- Songyang Wu
- School of Information Science and Technology, Yunnan Normal University, Kunming, 650500, China
| | - Kui Jin
- School of Information Science and Technology, Yunnan Normal University, Kunming, 650500, China
| | - Mingjing Tang
- School of Life Science, Yunnan Normal University, Kunming, 650500, China.
- Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Ministry of Education, Yunnan Normal University, Kunming, 650500, China.
| | - Yuelong Xia
- School of Information Science and Technology, Yunnan Normal University, Kunming, 650500, China
| | - Wei Gao
- School of Information Science and Technology, Yunnan Normal University, Kunming, 650500, China
| |
Collapse
|
45
|
Montalban E, Giralt A, Taing L, Nakamura Y, Pelosi A, Brown M, de Pins B, Valjent E, Martin M, Nairn AC, Greengard P, Flajolet M, Hervé D, Gambardella N, Roussarie JP, Girault JA. Operant Training for Highly Palatable Food Alters Translating Messenger RNA in Nucleus Accumbens D 2 Neurons and Reveals a Modulatory Role of Ncdn. Biol Psychiatry 2024; 95:926-937. [PMID: 37579933 PMCID: PMC11059129 DOI: 10.1016/j.biopsych.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Highly palatable food triggers behavioral responses including strong motivation. These effects involve the reward system and dopamine neurons, which modulate neurons in the nucleus accumbens (NAc). The molecular mechanisms underlying the long-lasting effects of highly palatable food on feeding behavior are poorly understood. METHODS We studied the effects of 2-week operant conditioning of mice with standard or isocaloric highly palatable food. We investigated the behavioral responses and dendritic spine modifications in the NAc. We compared the translating messenger RNA in NAc neurons identified by the type of dopamine receptors they express, depending on the kind of food and training. We tested the consequences of invalidation of an abundant downregulated gene, Ncdn. RESULTS Operant conditioning for highly palatable food increased motivation for food even in well-fed mice. In wild-type mice, free choice between regular and highly palatable food increased weight compared with access to regular food only. Highly palatable food increased spine density in the NAc. In animals trained for highly palatable food, translating messenger RNAs were modified in NAc neurons expressing dopamine D2 receptors, mostly corresponding to striatal projection neurons, but not in neurons expressing D1 receptors. Knockout of Ncdn, an abundant downregulated gene, opposed the conditioning-induced changes in satiety-sensitive feeding behavior and apparent motivation for highly palatable food, suggesting that downregulation may be a compensatory mechanism. CONCLUSIONS Our results emphasize the importance of messenger RNA alterations in D2 striatal projection neurons in the NAc in the behavioral consequences of highly palatable food conditioning and suggest a modulatory contribution of Ncdn downregulation.
Collapse
Affiliation(s)
- Enrica Montalban
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France.
| | - Albert Giralt
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Lieng Taing
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Yuki Nakamura
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Assunta Pelosi
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Mallory Brown
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Benoit de Pins
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Emmanuel Valjent
- Institut de Génomique Fonctionnelle, University of Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Miquel Martin
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, Spain; Instituto de investigaciones médicas Hospital del Mar, Barcelona, Spain
| | - Angus C Nairn
- Department of Psychiatry, Yale School of Medicine, Connecticut Mental Health Center, New Haven, Connecticut
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Marc Flajolet
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Denis Hervé
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | | | - Jean-Pierre Roussarie
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Jean-Antoine Girault
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France.
| |
Collapse
|
46
|
Kion-Crosby W, Barquist L. Network depth affects inference of gene sets from bacterial transcriptomes using denoising autoencoders. BIOINFORMATICS ADVANCES 2024; 4:vbae066. [PMID: 39027639 PMCID: PMC11256956 DOI: 10.1093/bioadv/vbae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/05/2024] [Accepted: 05/02/2024] [Indexed: 07/20/2024]
Abstract
Summary The increasing number of publicly available bacterial gene expression data sets provides an unprecedented resource for the study of gene regulation in diverse conditions, but emphasizes the need for self-supervised methods for the automated generation of new hypotheses. One approach for inferring coordinated regulation from bacterial expression data is through neural networks known as denoising autoencoders (DAEs) which encode large datasets in a reduced bottleneck layer. We have generalized this application of DAEs to include deep networks and explore the effects of network architecture on gene set inference using deep learning. We developed a DAE-based pipeline to extract gene sets from transcriptomic data in Escherichia coli, validate our method by comparing inferred gene sets with known pathways, and have used this pipeline to explore how the choice of network architecture impacts gene set recovery. We find that increasing network depth leads the DAEs to explain gene expression in terms of fewer, more concisely defined gene sets, and that adjusting the width results in a tradeoff between generalizability and biological inference. Finally, leveraging our understanding of the impact of DAE architecture, we apply our pipeline to an independent uropathogenic E.coli dataset to identify genes uniquely induced during human colonization. Availability and implementation https://github.com/BarquistLab/DAE_architecture_exploration.
Collapse
Affiliation(s)
- Willow Kion-Crosby
- Helmholtz Institute for RNA-based Infection Research (HIRI)/Helmholtz Centre for Infection Research (HZI), 97080 Würzburg, Germany
- Faculty of Medicine, University of Würzburg, 97080 Würzburg, Germany
| | - Lars Barquist
- Helmholtz Institute for RNA-based Infection Research (HIRI)/Helmholtz Centre for Infection Research (HZI), 97080 Würzburg, Germany
- Faculty of Medicine, University of Würzburg, 97080 Würzburg, Germany
- Department of Biology, University of Toronto, Mississauga, ON L5L 1C6, Canada
| |
Collapse
|
47
|
Chodkowski JL, Shade A. Bioactive exometabolites drive maintenance competition in simple bacterial communities. mSystems 2024; 9:e0006424. [PMID: 38470039 PMCID: PMC11019792 DOI: 10.1128/msystems.00064-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 02/19/2024] [Indexed: 03/13/2024] Open
Abstract
During prolonged resource limitation, bacterial cells can persist in metabolically active states of non-growth. These maintenance periods, such as those experienced in stationary phase, can include upregulation of secondary metabolism and release of exometabolites into the local environment. As resource limitation is common in many environmental microbial habitats, we hypothesized that neighboring bacterial populations employ exometabolites to compete or cooperate during maintenance and that these exometabolite-facilitated interactions can drive community outcomes. Here, we evaluated the consequences of exometabolite interactions over the stationary phase among three environmental strains: Burkholderia thailandensis E264, Chromobacterium subtsugae ATCC 31532, and Pseudomonas syringae pv. tomato DC3000. We assembled them into synthetic communities that only permitted chemical interactions. We compared the responses (transcripts) and outputs (exometabolites) of each member with and without neighbors. We found that transcriptional dynamics were changed with different neighbors and that some of these changes were coordinated between members. The dominant competitor B. thailandensis consistently upregulated biosynthetic gene clusters to produce bioactive exometabolites for both exploitative and interference competition. These results demonstrate that competition strategies during maintenance can contribute to community-level outcomes. It also suggests that the traditional concept of defining competitiveness by growth outcomes may be narrow and that maintenance competition could be an additional or alternative measure. IMPORTANCE Free-living microbial populations often persist and engage in environments that offer few or inconsistently available resources. Thus, it is important to investigate microbial interactions in this common and ecologically relevant condition of non-growth. This work investigates the consequences of resource limitation for community metabolic output and for population interactions in simple synthetic bacterial communities. Despite non-growth, we observed active, exometabolite-mediated competition among the bacterial populations. Many of these interactions and produced exometabolites were dependent on the community composition but we also observed that one dominant competitor consistently produced interfering exometabolites regardless. These results are important for predicting and understanding microbial interactions in resource-limited environments.
Collapse
Affiliation(s)
- John L. Chodkowski
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Ashley Shade
- Universite Claude Bernard Lyon 1, Laboratoire d'Ecologie Microbienne, UMR CNRS 5557, UMR INRAE 1418, VetAgro Sup, Villeurbanne, France
| |
Collapse
|
48
|
Mousavi R, Lobo D. Automatic design of gene regulatory mechanisms for spatial pattern formation. NPJ Syst Biol Appl 2024; 10:35. [PMID: 38565850 PMCID: PMC10987498 DOI: 10.1038/s41540-024-00361-5] [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: 11/21/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
Gene regulatory mechanisms (GRMs) control the formation of spatial and temporal expression patterns that can serve as regulatory signals for the development of complex shapes. Synthetic developmental biology aims to engineer such genetic circuits for understanding and producing desired multicellular spatial patterns. However, designing synthetic GRMs for complex, multi-dimensional spatial patterns is a current challenge due to the nonlinear interactions and feedback loops in genetic circuits. Here we present a methodology to automatically design GRMs that can produce any given two-dimensional spatial pattern. The proposed approach uses two orthogonal morphogen gradients acting as positional information signals in a multicellular tissue area or culture, which constitutes a continuous field of engineered cells implementing the same designed GRM. To efficiently design both the circuit network and the interaction mechanisms-including the number of genes necessary for the formation of the target spatial pattern-we developed an automated algorithm based on high-performance evolutionary computation. The tolerance of the algorithm can be configured to design GRMs that are either simple to produce approximate patterns or complex to produce precise patterns. We demonstrate the approach by automatically designing GRMs that can produce a diverse set of synthetic spatial expression patterns by interpreting just two orthogonal morphogen gradients. The proposed framework offers a versatile approach to systematically design and discover complex genetic circuits producing spatial patterns.
Collapse
Affiliation(s)
- Reza Mousavi
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, MD, USA
| | - Daniel Lobo
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, MD, USA.
- Greenebaum Comprehensive Cancer Center and Center for Stem Cell Biology & Regenerative Medicine, University of Maryland, Baltimore, Baltimore, MD, USA.
| |
Collapse
|
49
|
Lukauskas S, Tvardovskiy A, Nguyen NV, Stadler M, Faull P, Ravnsborg T, Özdemir Aygenli B, Dornauer S, Flynn H, Lindeboom RGH, Barth TK, Brockers K, Hauck SM, Vermeulen M, Snijders AP, Müller CL, DiMaggio PA, Jensen ON, Schneider R, Bartke T. Decoding chromatin states by proteomic profiling of nucleosome readers. Nature 2024; 627:671-679. [PMID: 38448585 PMCID: PMC10954555 DOI: 10.1038/s41586-024-07141-5] [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: 05/10/2021] [Accepted: 01/31/2024] [Indexed: 03/08/2024]
Abstract
DNA and histone modifications combine into characteristic patterns that demarcate functional regions of the genome1,2. While many 'readers' of individual modifications have been described3-5, how chromatin states comprising composite modification signatures, histone variants and internucleosomal linker DNA are interpreted is a major open question. Here we use a multidimensional proteomics strategy to systematically examine the interaction of around 2,000 nuclear proteins with over 80 modified dinucleosomes representing promoter, enhancer and heterochromatin states. By deconvoluting complex nucleosome-binding profiles into networks of co-regulated proteins and distinct nucleosomal features driving protein recruitment or exclusion, we show comprehensively how chromatin states are decoded by chromatin readers. We find highly distinctive binding responses to different features, many factors that recognize multiple features, and that nucleosomal modifications and linker DNA operate largely independently in regulating protein binding to chromatin. Our online resource, the Modification Atlas of Regulation by Chromatin States (MARCS), provides in-depth analysis tools to engage with our results and advance the discovery of fundamental principles of genome regulation by chromatin states.
Collapse
Affiliation(s)
- Saulius Lukauskas
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Andrey Tvardovskiy
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Nhuong V Nguyen
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Mara Stadler
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Statistics, Ludwig Maximilian University Munich, Munich, Germany
| | - Peter Faull
- MRC Laboratory of Medical Sciences (LMS), London, UK
- Proteomic Sciences Technology Platform, The Francis Crick Institute, London, UK
- Northwestern Proteomics Core Facility, Northwestern University, Chicago, IL, USA
| | - Tina Ravnsborg
- VILLUM Center for Bioanalytical Sciences and Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | | - Scarlett Dornauer
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Helen Flynn
- Proteomic Sciences Technology Platform, The Francis Crick Institute, London, UK
| | - Rik G H Lindeboom
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Teresa K Barth
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Munich, Germany
- Clinical Protein Analysis Unit (ClinZfP), Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University Munich, Martinsried, Germany
| | - Kevin Brockers
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Munich, Germany
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Christian L Müller
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Statistics, Ludwig Maximilian University Munich, Munich, Germany
- Center for Computational Mathematics, Flatiron Institute, New York, NY, USA
| | - Peter A DiMaggio
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Ole N Jensen
- VILLUM Center for Bioanalytical Sciences and Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Robert Schneider
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany
- Faculty of Biology, Ludwig Maximilian University Munich, Martinsried, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Till Bartke
- Institute of Functional Epigenetics, Helmholtz Zentrum München, Neuherberg, Germany.
- MRC Laboratory of Medical Sciences (LMS), London, UK.
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
50
|
Yao Q, Feng Y, Wang J, Zhang Y, Yi F, Li Z, Zhang M. Integrated Metabolome and Transcriptome Analysis of Gibberellins Mediated the Circadian Rhythm of Leaf Elongation by Regulating Lignin Synthesis in Maize. Int J Mol Sci 2024; 25:2705. [PMID: 38473951 DOI: 10.3390/ijms25052705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/08/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
Plant growth exhibits rhythmic characteristics, and gibberellins (GAs) are involved in regulating cell growth, but it is still unclear how GAs crosstalk with circadian rhythm to regulate cell elongation. The study analyzed growth characteristics of wild-type (WT), zmga3ox and zmga3ox with GA3 seedlings. We integrated metabolomes and transcriptomes to study the interaction between GAs and circadian rhythm in mediating leaf elongation. The rates of leaf growth were higher in WT than zmga3ox, and zmga3ox cell length was shorter when proliferated in darkness than light, and GA3 restored zmga3ox leaf growth. The differentially expressed genes (DEGs) between WT and zmga3ox were mainly enriched in hormone signaling and cell wall synthesis, while DEGs in zmga3ox were restored to WT by GA3. Moreover, the number of circadian DEGs that reached the peak expression in darkness was more than light, and the upregulated circadian DEGs were mainly enriched in cell wall synthesis. The differentially accumulated metabolites (DAMs) were mainly attributed to flavonoids and phenolic acid. Twenty-two DAMs showed rhythmic accumulation, especially enriched in lignin synthesis. The circadian DEGs ZmMYBr41/87 and ZmHB34/70 were identified as regulators of ZmHCT8 and ZmBM1, which were enzymes in lignin synthesis. Furthermore, GAs regulated ZmMYBr41/87 and ZmHB34/70 to modulate lignin biosynthesis for mediating leaf rhythmic growth.
Collapse
Affiliation(s)
- Qingqing Yao
- State Key Laboratory of Plant Environmental Resilience, Engineering Research Center of Plant Growth Regulator, Ministry of Education, College of Agronomy and Biotechnology, China Agricultural University, No 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Ying Feng
- State Key Laboratory of Plant Environmental Resilience, Engineering Research Center of Plant Growth Regulator, Ministry of Education, College of Agronomy and Biotechnology, China Agricultural University, No 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Jiajie Wang
- State Key Laboratory of Plant Environmental Resilience, Engineering Research Center of Plant Growth Regulator, Ministry of Education, College of Agronomy and Biotechnology, China Agricultural University, No 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Yushi Zhang
- State Key Laboratory of Plant Environmental Resilience, Engineering Research Center of Plant Growth Regulator, Ministry of Education, College of Agronomy and Biotechnology, China Agricultural University, No 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Fei Yi
- State Key Laboratory of Plant Environmental Resilience, Engineering Research Center of Plant Growth Regulator, Ministry of Education, College of Agronomy and Biotechnology, China Agricultural University, No 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Zhaohu Li
- State Key Laboratory of Plant Environmental Resilience, Engineering Research Center of Plant Growth Regulator, Ministry of Education, College of Agronomy and Biotechnology, China Agricultural University, No 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Mingcai Zhang
- State Key Laboratory of Plant Environmental Resilience, Engineering Research Center of Plant Growth Regulator, Ministry of Education, College of Agronomy and Biotechnology, China Agricultural University, No 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| |
Collapse
|