1
|
Su F, Su M, Wei W, Wu J, Chen L, Sun X, Liu M, Sun S, Mao R, Bourgonje AR, Hu S. Integrating multi-omics data to reveal the host-microbiota interactome in inflammatory bowel disease. Gut Microbes 2025; 17:2476570. [PMID: 40063366 PMCID: PMC11901428 DOI: 10.1080/19490976.2025.2476570] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Numerous studies have accelerated the knowledge expansion on the role of gut microbiota in inflammatory bowel disease (IBD). However, the precise mechanisms behind host-microbe cross-talk remain largely undefined, due to the complexity of the human intestinal ecosystem and multiple external factors. In this review, we introduce the interactome concept to systematically summarize how intestinal dysbiosis is involved in IBD pathogenesis in terms of microbial composition, functionality, genomic structure, transcriptional activity, and downstream proteins and metabolites. Meanwhile, this review also aims to present an updated overview of the relevant mechanisms, high-throughput multi-omics methodologies, different types of multi-omics cohort resources, and computational methods used to understand host-microbiota interactions in the context of IBD. Finally, we discuss the challenges pertaining to the integration of multi-omics data in order to reveal host-microbiota cross-talk and offer insights into relevant future research directions.
Collapse
Affiliation(s)
- Fengyuan Su
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Meng Su
- The First Clinical Medical School, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Wenting Wei
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jiayun Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Leyan Chen
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xiqiao Sun
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Moyan Liu
- Amsterdam UMC location Academic Medical Center, Department of Experimental Vascular Medicine, Amsterdam, The Netherlands
| | - Shiqiang Sun
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Arno R. Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shixian Hu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
2
|
Ye X, Shi T, Huang D, Sakurai T. Multi-Omics clustering by integrating clinical features from large language model. Methods 2025; 239:64-71. [PMID: 40180255 DOI: 10.1016/j.ymeth.2025.03.017] [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: 01/14/2025] [Revised: 03/16/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025] Open
Abstract
Multi-omics clustering has emerged as a powerful approach for understanding complex biological systems and enabling cancer subtyping by integrating diverse omics data. Existing methods primarily focus on the integration of different types of omics data, often overlooking the value of clinical context. In this study, we propose a novel framework that incorporates clinical features extracted from large language model (LLM) to enhance multi-omics clustering. Leveraging clinical data extracted from pathology reports using a BERT-based model, our framework converts unstructured medical text into structured clinical features. These features are integrated with omics data through an autoencoder, enriching the information content of each omics layer to improve feature extraction. The extracted features are then projected into a latent subspace using singular value decomposition (SVD), followed by spectral clustering to obtain the final clustering result. We evaluate the proposed framework on six cancer datasets on three omics levels, comparing it with several state-of-the-art methods. The experimental results demonstrate that the proposed framework outperforms existing methods in multi-omics clustering for cancer subtyping. Moreover, the results highlight the efficacy of integrating clinical features derived from LLM, significantly enhancing clustering performance. This work underscores the importance of clinical context in multi-omics analysis and showcases the transformative potential of LLM in advancing precision medicine.
Collapse
Affiliation(s)
- Xiucai Ye
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan.
| | - Tianyi Shi
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan
| | - Dong Huang
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan.
| | - Tetsuya Sakurai
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan
| |
Collapse
|
3
|
Creus-Martí I, Moya A, Santonja FJ. Methodology for microbiome data analysis: An overview. Comput Biol Med 2025; 192:110157. [PMID: 40279974 DOI: 10.1016/j.compbiomed.2025.110157] [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: 06/30/2024] [Revised: 03/07/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025]
Abstract
It is known that microbiome and health are related, in addition, recent research has found that microbiome has potential clinical uses. These facts highlight the importance of the microbiome in actual science. However, microbiome data has some characteristics that makes its statistical study challenging. In recent years, longitudinal and non-longitudinal methods have been designed to analyze the microbiota and knowing more about the bacterial behavior. In this article in the form of a review we summarize the characteristics of microbiome data and the statistical methods most widespread to analyze it. We have taken into account if the strategies are longitudinal or not. We also classify the methods based on their specific analytical objectives and based on their mathematical characteristics. The methods are structured according to their biological goals and mathematical features, ensuring that the insights provided are both relevant and accessible to professionals in biology and statistics. We present this review as a reference for the most widely used methods in microbiome data analysis and as a foundation for identifying potential areas for future research. We want to point out that this review can be particularly useful to remark the importance of the methodology designed in order to study microbiome longitudinal datasets.
Collapse
Affiliation(s)
- Irene Creus-Martí
- Department of Applied Mathematics, Universitat Politècnica de València, Valencia, Spain.
| | - Andrés Moya
- Institute for Integrative Systems Biology (I2Sysbio), Universitat de València and CSIC, València, Spain; The Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), Valencia, Spain; CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Francisco J Santonja
- Department of Statistics and Operation Research, Universitat de València, Valencia, Spain
| |
Collapse
|
4
|
Zhang S, Kong W, Wang S, Wei K, Liu K, Wen G, Yu Y. Effective Integration of Single-Cell Multi-Omics Data Using Improved Network-Based Integrative Clustering with Multigraph Regularization. J Comput Biol 2025. [PMID: 40401439 DOI: 10.1089/cmb.2023.0460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
The purpose of integrating different omics data is to study cellular heterogeneity at the level of transcriptional regulation from different gene levels, which can effectively identify cell types and reveal the pathogenesis of Alzheimer's disease (AD) from two perspectives. However, implementing such algorithms faces challenges such as high data noise levels, increased dimensionality, and computational complexity. In this study, multigraph regularization constraints were introduced in the network-based integrative clustering algorithm (MGR-NIC) to remove redundant features and keep the geometry structures underlying the data by fusing two types of data (snRNA-seq and snATAC-seq) of glial cells from AD samples. The effectiveness of the MGR-NIC algorithm was validated using both simulation datasets and real datasets derived from various tissues. The MGR-NIC algorithm can improve clustering accuracy by selecting features that better represent the dataset's structure. The clustering results obtained with the MGR-NIC algorithm show strong consistency with the clustering results inherent to the published DLPFC dataset, while the classification results generated using the NIC algorithm often lead to cluster overlap when applied to the DLPFC dataset. We will use the same state-of-the-art algorithms for a comprehensive evaluation with our proposed MGR-NIC algorithm, including NIC, scAI, Multi-Omics Factor Analysis v2, and JSNMF. MGR-NIC is the most stable and reliable method, implying its robustness across different datasets and its reliability in yielding consistent and accurate results.
Collapse
Affiliation(s)
- Shunqin Zhang
- College of Information Engineering, Shanghai Maritime University, Shanghai, P.R. China
| | - Wei Kong
- College of Information Engineering, Shanghai Maritime University, Shanghai, P.R. China
| | - Shuaiqun Wang
- College of Information Engineering, Shanghai Maritime University, Shanghai, P.R. China
| | - Kai Wei
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Kun Liu
- College of Information Engineering, Shanghai Maritime University, Shanghai, P.R. China
| | - Gen Wen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yaling Yu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
5
|
Chen M, Cheng R, He J, Chen J, Zhang J. SMOPCA: spatially aware dimension reduction integrating multi-omics improves the efficiency of spatial domain detection. Genome Biol 2025; 26:135. [PMID: 40399936 PMCID: PMC12096709 DOI: 10.1186/s13059-025-03576-9] [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: 02/14/2024] [Accepted: 04/12/2025] [Indexed: 05/23/2025] Open
Abstract
Technological advances have enabled us to profile multiple omics layers with spatial information, significantly enhancing spatial domain detection and advancing a variety of biomedical research fields. Despite these advancements, there is a notable lack of effective methods for modeling spatial multi-omics data. We introduce SMOPCA, a Spatial Multi-Omics Principal Component Analysis method designed to perform joint dimension reduction on multimodal data while preserving spatial dependencies. Extensive experiments reveal that SMOPCA outperforms existing single-modal and multimodal dimension reduction and clustering methods, across both single-cell and spatial multi-omics datasets derived from diverse technologies and tissue structures.
Collapse
Affiliation(s)
- Mo Chen
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, Jiangsu, China
- School of Artificial Intelligence, Nanjing University, Nanjing, Jiangsu, China
| | - Ruihua Cheng
- Big Data Statistics Research Center, Tianjin University of Finance and Economics, Tianjin, China
| | - Jianuo He
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, Jiangsu, China
- School of Artificial Intelligence, Nanjing University, Nanjing, Jiangsu, China
| | - Jun Chen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA.
| | - Jie Zhang
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing, Jiangsu, China.
- School of Artificial Intelligence, Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
6
|
Zhang R, Wang Y, Yang W, Wen J, Liu W, Zhi S, Li G, Chai N, Huang J, Xie Y, Xie X, Chen L, Gu M, Liu YG, Zhu Q. PlantGPT: An Arabidopsis-Based Intelligent Agent that Answers Questions about Plant Functional Genomics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e03926. [PMID: 40397417 DOI: 10.1002/advs.202503926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/15/2025] [Indexed: 05/22/2025]
Abstract
Research into plant gene function is crucial for developing strategies to increase crop yields. The recent introduction of large language models (LLMs) offers a means to aggregate large amounts of data into a queryable format, but the output can contain inaccurate or false claims known as hallucinations. To minimize such hallucinations and produce high-quality knowledge-based outputs, the abstracts of over 60 000 plant research articles are compiled into a Chroma database for retrieval-augmented generation (RAG). Then linguistic data are used from 13 993 Arabidopsis (Arabidopsis thaliana) phenotypes and 23 323 gene functions to fine-tune the LLM Llama3-8B, producing PlantGPT, a virtual expert in Arabidopsis phenotype-gene research. By evaluating answers to test questions, it is demonstrated that PlantGPT outperforms general LLMs in answering specialized questions. The findings provide a blueprint for functional genomics research in food crops and demonstrate the potential for developing LLMs for plant research modalities. To provide broader access and facilitate adoption, the online tool http://www.plantgpt.icu is developed, which will allow researchers to use PlantGPT in their scientific investigations.
Collapse
Affiliation(s)
- Ruixiang Zhang
- Guangdong Basic Research Center of Excellence for Precise Breeding of Future Crops, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Agriculture, College of Life Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yu Wang
- School of Life Sciences, Institute for Immunology, State Key Laboratory of Membrane Biology, China Ministry of Education Key Laboratory of Protein Sciences, Tsinghua University, Beijing, 100084, China
| | - Weiyang Yang
- Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Jun Wen
- Guangdong Basic Research Center of Excellence for Precise Breeding of Future Crops, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Agriculture, College of Life Science, South China Agricultural University, Guangzhou, 510642, China
| | - Weizhi Liu
- Guangdong Basic Research Center of Excellence for Precise Breeding of Future Crops, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Agriculture, College of Life Science, South China Agricultural University, Guangzhou, 510642, China
| | - Shipeng Zhi
- Department of Medicine, Tsinghua University, Beijing, 100084, China
| | - Guangzhou Li
- Guangdong Basic Research Center of Excellence for Precise Breeding of Future Crops, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Agriculture, College of Life Science, South China Agricultural University, Guangzhou, 510642, China
| | - Nan Chai
- Guangdong Basic Research Center of Excellence for Precise Breeding of Future Crops, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Agriculture, College of Life Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jiaqi Huang
- Engineering Research Center of Protection and Utilization of Plant Resources, College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, 110866, China
| | - Yongyao Xie
- Guangdong Basic Research Center of Excellence for Precise Breeding of Future Crops, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Agriculture, College of Life Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xianrong Xie
- Guangdong Basic Research Center of Excellence for Precise Breeding of Future Crops, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Agriculture, College of Life Science, South China Agricultural University, Guangzhou, 510642, China
| | - Letian Chen
- Guangdong Basic Research Center of Excellence for Precise Breeding of Future Crops, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Agriculture, College of Life Science, South China Agricultural University, Guangzhou, 510642, China
| | - Miao Gu
- Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Yao-Guang Liu
- Guangdong Basic Research Center of Excellence for Precise Breeding of Future Crops, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Agriculture, College of Life Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qinlong Zhu
- Guangdong Basic Research Center of Excellence for Precise Breeding of Future Crops, Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Agriculture, College of Life Science, South China Agricultural University, Guangzhou, 510642, China
| |
Collapse
|
7
|
Taha BA, Abdulrahm ZM, Addie AJ, Haider AJ, Alkawaz AN, Yaqoob IAM, Arsad N. Advancing optical nanosensors with artificial intelligence: A powerful tool to identify disease-specific biomarkers in multi-omics profiling. Talanta 2025; 287:127693. [PMID: 39919475 DOI: 10.1016/j.talanta.2025.127693] [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: 12/04/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/09/2025]
Abstract
Multi-omics profiling integrates genomic, epigenomic, transcriptomic, and proteomic data, essential for understanding complex health and disease pathways. This review highlights the transformative potential of combining optical nanosensors with artificial intelligence (AI). It is possible to identify disease-specific biomarkers using real-time and sensitive molecular interactions. These technologies are precious for genetic, epigenetic, and proteomic changes critical to disease progression and treatment response. AI improves multi-omics profiling by analyzing large, diverse data sets and common patterns traditional methods overlook. Machine learning tools Biomarkers Discovery is revolutionizing, drug resistance is being understood, and medicine is being personalized as the combination of AI and nanosensors has advanced the detection of DNA methylation and proteomic signatures and improved our understanding of cancer, cardiovascular disease and vascular disease. Despite these advances, challenges still exist. Difficulties in integrating data sets, retaining sensors, and building scalable computing tools are the biggest obstacles. It also examines various solutions with advanced AI algorithms and innovations, including fabrication in nanosensor design. Moreover, it highlights the potential of nanosensor-assisted, AI-driven multi-omics profiling to revolutionize disease diagnosis and treatment. As technology advances, these tools pave the way for faster diagnosis, more accurate treatment and improved patient outcomes, offering new hope for personalized medicine.
Collapse
Affiliation(s)
- Bakr Ahmed Taha
- Photonics Technology Lab, Department of Electrical, Electronic and Systems Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, UKM, Bangi, 43600, Malaysia; Alimam University College, Balad, Iraq.
| | | | - Ali J Addie
- Center of Industrial Applications and Materials Technology, Scientific Research Commission, Baghdad 10070, Iraq.
| | - Adawiya J Haider
- Applied Sciences Department/Laser Science and Technology Branch, University of Technology, Iraq.
| | - Ali Najem Alkawaz
- Department of Electrical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, 50603, Malaysia.
| | - Isam Ahmed M Yaqoob
- Faculty of Computer Sciences, Universiti Putra Malaysia, 43400, Selangor, Malaysia.
| | - Norhana Arsad
- Photonics Technology Lab, Department of Electrical, Electronic and Systems Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, UKM, Bangi, 43600, Malaysia.
| |
Collapse
|
8
|
Goldmann U, Wiedmer T, Garofoli A, Sedlyarov V, Bichler M, Haladik B, Wolf G, Christodoulaki E, Ingles-Prieto A, Ferrada E, Frommelt F, Teoh ST, Leippe P, Onea G, Pfeifer M, Kohlbrenner M, Chang L, Selzer P, Reinhardt J, Digles D, Ecker GF, Osthushenrich T, MacNamara A, Malarstig A, Hepworth D, Superti-Furga G. Data- and knowledge-derived functional landscape of human solute carriers. Mol Syst Biol 2025:10.1038/s44320-025-00108-2. [PMID: 40355757 DOI: 10.1038/s44320-025-00108-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/28/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
The human solute carrier (SLC) superfamily of ~460 membrane transporters remains the largest understudied protein family despite its therapeutic potential. To advance SLC research, we developed a comprehensive knowledgebase that integrates systematic multi-omics data sets with selected curated information from public sources. We annotated SLC substrates through literature curation, compiled SLC disease associations using data mining techniques, and determined the subcellular localization of SLCs by combining annotations from public databases with an immunofluorescence imaging approach. This SLC-centric knowledge is made accessible to the scientific community via a web portal featuring interactive dashboards and visualization tools. Utilizing this systematically collected and curated resource, we computationally derived an integrated functional landscape for the entire human SLC superfamily. We identified clusters with distinct properties and established functional distances between transporters. Based on all available data sets and their integration, we assigned biochemical/biological functions to each SLC, making this study one of the largest systematic annotations of human gene function and a potential blueprint for future research endeavors.
Collapse
Affiliation(s)
- Ulrich Goldmann
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Tabea Wiedmer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Andrea Garofoli
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Vitaly Sedlyarov
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Manuel Bichler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Ben Haladik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Gernot Wolf
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Eirini Christodoulaki
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Alvaro Ingles-Prieto
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Evandro Ferrada
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Fabian Frommelt
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Shao Thing Teoh
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Philipp Leippe
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Gabriel Onea
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | - Daniela Digles
- University of Vienna, Department of Pharmaceutical Sciences, Vienna, Austria
| | - Gerhard F Ecker
- University of Vienna, Department of Pharmaceutical Sciences, Vienna, Austria
| | | | | | | | | | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
- Fondazione Ri.MED, Palermo, Italy.
| |
Collapse
|
9
|
Reierson MM, Acharjee A. Unsupervised machine learning-based stratification and immune deconvolution of liver hepatocellular carcinoma. BMC Cancer 2025; 25:853. [PMID: 40349011 PMCID: PMC12066050 DOI: 10.1186/s12885-025-14242-5] [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: 12/06/2024] [Accepted: 04/29/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most prevalent type of liver cancer and a leading cause of cancer-related deaths globally. The tumour microenvironment (TME) influences treatment response and prognosis, yet its heterogeneity remains unclear. METHODS The unsupervised machine learning methods- agglomerative hierarchical clustering, Multi-Omics Factor Analysis with K-means++, and an autoencoder with K-means++ - stratified patients using microarray data from HCC samples. Immune deconvolution algorithms estimated the proportions of infiltrating immune cells across identified clusters. RESULTS Thirteen genes were found to influence HCC subtyping in both primary and validation datasets, with three genes-TOP2A, DCN, and MT1E-showing significant associations with survival and recurrence. DCN, a known tumour suppressor, was significant across datasets and associated with improved survival, potentially by modulating the TME and promoting an anti-tumour immune response. CONCLUSIONS The discovery of the 13 conserved genes is an important step toward understanding HCC heterogeneity and the TME, potentially leading to the identification of more reliable biomarkers and therapeutic targets. We have stratified and validated the liver cancer populations. The findings suggest further research is needed to explore additional factors influencing the TME beyond gene expression, such as tumour microbiome and stromal cell interactions.
Collapse
Affiliation(s)
- Mae Montserrat Reierson
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, B15 2TT, UK
| | - Animesh Acharjee
- Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, B15 2TT, UK.
- Institute of Translational Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TT, UK.
- MRC Health Data Research UK (HDR), Midlands Site, UK.
- Centre for Health Data Research, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
10
|
Tripathy RK, Frohock Z, Wang H, Cary GA, Keegan S, Carter GW, Li Y. Effective integration of multi-omics with prior knowledge to identify biomarkers via explainable graph neural networks. NPJ Syst Biol Appl 2025; 11:43. [PMID: 40341543 PMCID: PMC12062277 DOI: 10.1038/s41540-025-00519-9] [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: 10/06/2024] [Accepted: 04/11/2025] [Indexed: 05/10/2025] Open
Abstract
The rapid growth of multi-omics datasets and the wealth of biological knowledge necessitates the development of effective methods for their integration. Such methods are essential for building predictive models and identifying drug targets based on a limited number of samples. We propose a framework called GNNRAI for the supervised integration of multi-omics data with biological priors represented as knowledge graphs. Our framework leverages graph neural networks (GNNs) to model the correlation structures among features from high-dimensional 'omics data, which reduces the effective dimensions in data and enables us to analyze thousands of genes simultaneously using hundreds of samples. Furthermore, our framework incorporates explainability methods to elucidate informative biomarkers. We apply our framework to Alzheimer's disease (AD) multi-omics data, showing that the integration of transcriptomics and proteomics data with prior AD knowledge is effective, improving the prediction accuracy of AD status over single-omics analyses and highlighting both known and novel AD-predictive biomarkers.
Collapse
Affiliation(s)
- Rohit K Tripathy
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Zachary Frohock
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Hong Wang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | | | | | - Yi Li
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
| |
Collapse
|
11
|
Manurung MD, Heieis GA, König M, Azimi S, Ndao M, Veldhuizen T, Hoving D, Hoekstra PT, Kruize YCM, Wammes LJ, Menafra R, Cisse M, Mboup S, Dieye A, Kloet S, Tahapary DL, Supali T, Wuhrer M, Hokke CH, Everts B, Mahfouz A, Jochems SP, Yazdanbakhsh M, Mbow M. Systems analysis unravels a common rural-urban gradient in immunological profile, function, and metabolic dependencies. SCIENCE ADVANCES 2025; 11:eadu0419. [PMID: 40305616 PMCID: PMC12042899 DOI: 10.1126/sciadv.adu0419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025]
Abstract
Urbanization affects environmental exposures and lifestyle, shaping immune system variation and influencing disease susceptibility and vaccine responses. Here, we present systems analysis of immune profiles across the rural-urban gradient, comparing rural and urban Senegalese with urban Dutch individuals. By integrating single-cell phenotyping, metabolic profiling, and functional analysis, we reveal a trajectory of immune remodeling along the gradient. This includes enrichment of proinflammatory CD11c+ B cells associated with altered IgG Fc glycosylation, adaptive NK cells with reduced responsiveness to accessory cytokines, and CD161+CD4+T cells with enhanced cytokine production in rural settings. Metabolic perturbation studies demonstrated distinct dependencies on glycolysis, pentose phosphate pathway, and fatty acid synthesis for cellular cytokine responses across populations. We validate core rural-urban immune signatures in an independent Indonesian cohort, suggesting shared immunological adaptations to urbanization across ancestries and geographical areas. Our findings provide insights into rural-urban immune function in understudied populations.
Collapse
Affiliation(s)
- Mikhael D. Manurung
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Graham A. Heieis
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Marion König
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Shohreh Azimi
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Malick Ndao
- Department of Immunology, Faculty of Medicine, Pharmacy, and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal
| | - Tom Veldhuizen
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Dennis Hoving
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Pytsje T. Hoekstra
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Yvonne C. M. Kruize
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Linda J. Wammes
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Roberta Menafra
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, Netherlands
| | - Marouba Cisse
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
- Department of Immunology, Faculty of Medicine, Pharmacy, and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal
| | - Souleymane Mboup
- Institute of Health Research, Epidemiological Surveillance, and Training, Dakar, Senegal
| | - Alioune Dieye
- Department of Immunology, Faculty of Medicine, Pharmacy, and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal
| | - Susan Kloet
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, Netherlands
| | - Dicky L. Tahapary
- Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Taniawati Supali
- Department of Parasitology, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Cornelis H. Hokke
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Bart Everts
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Ahmed Mahfouz
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Simon P. Jochems
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Moustapha Mbow
- Department of Immunology, Faculty of Medicine, Pharmacy, and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal
| |
Collapse
|
12
|
Guo Y, Zou H, Alam MS, Luo S. Integrative Multi-Omics and Multivariate Longitudinal Data Analysis for Dynamic Risk Estimation in Alzheimer's Disease. Stat Med 2025; 44:e70105. [PMID: 40387018 DOI: 10.1002/sim.70105] [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: 02/27/2024] [Revised: 03/19/2025] [Accepted: 04/11/2025] [Indexed: 05/20/2025]
Abstract
Alzheimer's disease (AD) is a complex and progressive neurodegenerative disorder, characterized by diverse cognitive and functional impairments that manifest heterogeneously across individuals, domains, and time. The accurate assessment of AD's severity and progression requires integrating a variety of data modalities, including multivariate longitudinal neuropsychological tests and multi-omics datasets such as metabolomics and lipidomics. These data sources provide valuable insights into risk factors associated with dementia onset. However, effectively utilizing omics data in dynamic risk estimation for AD progression is challenging due to issues including high dimensionality, heterogeneity, and complex intercorrelations. To address these challenges, we develop a novel joint-modeling framework that effectively combines multi-omics factor analysis (MOFA) for dimension reduction and feature extraction with a multivariate functional mixed model (MFMM) for modeling longitudinal outcomes. This integrative joint modeling approach enables dynamic evaluation of dementia risk by leveraging both omics and longitudinal data. We validate the efficacy of our integrative model through extensive simulation studies and its practical application to the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset.
Collapse
Affiliation(s)
- Yuanyuan Guo
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Haotian Zou
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Mohammad Samsul Alam
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| |
Collapse
|
13
|
Yang T, Li WV. Generalized Probabilistic Canonical Correlation Analysis for Multi-modal Data Integration with Full or Partial Observations. ARXIV 2025:arXiv:2504.11610v1. [PMID: 40321951 PMCID: PMC12047925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Background The integration and analysis of multi-modal data are increasingly essential across various domains including bioinformatics. As the volume and complexity of such data grow, there is a pressing need for computational models that not only integrate diverse modalities but also leverage their complementary information to improve clustering accuracy and insights, especially when dealing with partial observations with missing data. Results We propose Generalized Probabilistic Canonical Correlation Analysis (GPCCA), an unsupervised method for the integration and joint dimensionality reduction of multi-modal data. GPCCA addresses key challenges in multi-modal data analysis by handling missing values within the model, enabling the integration of more than two modalities, and identifying informative features while accounting for correlations within individual modalities. The model demonstrates robustness to various missing data patterns and provides low-dimensional embeddings that facilitate downstream clustering and analysis. In a range of simulation settings, GPCCA outperforms existing methods in capturing essential patterns across modalities. Additionally, we demonstrate its applicability to multi-omics data from TCGA cancer datasets and a multi-view image dataset. Conclusion GPCCA offers a useful framework for multi-modal data integration, effectively handling missing data and providing informative low-dimensional embeddings. Its performance across cancer genomics and multi-view image data highlights its robustness and potential for broad application. To make the method accessible to the wider research community, we have released an R package, GPCCA, which is available at https://github.com/Kaversoniano/GPCCA.
Collapse
Affiliation(s)
- Tianjian Yang
- Department of Statistics, University of California, Riverside
| | - Wei Vivian Li
- Department of Statistics, University of California, Riverside
| |
Collapse
|
14
|
Yue C, Chen B, Pan F, Wang Z, Yu H, Liu G, Li W, Wang R, Tang Y. TCnet: A Novel Strategy to Predict Target Combination of Alzheimer's Disease via Network-Based Methods. J Chem Inf Model 2025; 65:3866-3878. [PMID: 40172120 DOI: 10.1021/acs.jcim.5c00172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder with an unclear pathogenesis; the traditional ″single gene-single target-single drug″ strategy is insufficient for effective treatment. This study explores a novel strategy for the multitarget therapy of AD by integrating multiomics data and employing network analysis. Different from conventional single-target methods, TCnet adopts a mechanism-driven strategy, utilizing multiomics data to decompose disease mechanisms, construct potential target combinations, and prioritize the optimal combinations using a scoring function. TCnet not only advances our understanding of disease mechanisms but also facilitates large-scale drug screening. This approach was further employed to screen active compounds from Huang-Lian-Jie-Du-Tang (HLJDT), identifying quercetin as a candidate targeting GSK3β and ADAM17. Subsequent in vitro experiments confirmed the neuroprotective and anti-inflammatory effects of quercetin. Overall, TCnet offers a promising approach for predicting target combinations and provides new insights and directions for drug discovery in AD.
Collapse
Affiliation(s)
- Chengyuan Yue
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Baiyu Chen
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Fei Pan
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Ze Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Hongbo Yu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Rui Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
15
|
Morabito A, De Simone G, Pastorelli R, Brunelli L, Ferrario M. Algorithms and tools for data-driven omics integration to achieve multilayer biological insights: a narrative review. J Transl Med 2025; 23:425. [PMID: 40211300 PMCID: PMC11987215 DOI: 10.1186/s12967-025-06446-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/30/2025] [Indexed: 04/13/2025] Open
Abstract
Systems biology is a holistic approach to biological sciences that combines experimental and computational strategies, aimed at integrating information from different scales of biological processes to unravel pathophysiological mechanisms and behaviours. In this scenario, high-throughput technologies have been playing a major role in providing huge amounts of omics data, whose integration would offer unprecedented possibilities in gaining insights on diseases and identifying potential biomarkers. In the present review, we focus on strategies that have been applied in literature to integrate genomics, transcriptomics, proteomics, and metabolomics in the year range 2018-2024. Integration approaches were divided into three main categories: statistical-based approaches, multivariate methods, and machine learning/artificial intelligence techniques. Among them, statistical approaches (mainly based on correlation) were the ones with a slightly higher prevalence, followed by multivariate approaches, and machine learning techniques. Integrating multiple biological layers has shown great potential in uncovering molecular mechanisms, identifying putative biomarkers, and aid classification, most of the time resulting in better performances when compared to single omics analyses. However, significant challenges remain. The high-throughput nature of omics platforms introduces issues such as variable data quality, missing values, collinearity, and dimensionality. These challenges further increase when combining multiple omics datasets, as the complexity and heterogeneity of the data increase with integration. We report different strategies that have been found in literature to cope with these challenges, but some open issues still remain and should be addressed to disclose the full potential of omics integration.
Collapse
Affiliation(s)
- Aurelia Morabito
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy.
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133, Milan, Italy.
| | - Giulia De Simone
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
- Department of Biotechnologies and Biosciences, Università degli Studi Milano Bicocca, 20126, Milan, Italy
| | - Roberta Pastorelli
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
| | - Laura Brunelli
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
| | - Manuela Ferrario
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133, Milan, Italy
| |
Collapse
|
16
|
Zillich L, Artioli A, Pohořalá V, Zillich E, Stertz L, Belschner H, Jabali A, Frank J, Streit F, Avetyan D, Völker MP, Müller S, Hansson AC, Meyer TD, Rietschel M, Ladewig J, Spanagel R, Oliveira AMM, Walss-Bass C, Bernardi RE, Koch P, Witt SH. Cell type-specific multi-omics analysis of cocaine use disorder in the human caudate nucleus. Nat Commun 2025; 16:3381. [PMID: 40204703 PMCID: PMC11982542 DOI: 10.1038/s41467-025-57339-y] [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: 08/20/2024] [Accepted: 02/11/2025] [Indexed: 04/11/2025] Open
Abstract
Structural and functional alterations in the brain's reward circuitry are present in cocaine use disorder (CocUD), but their molecular underpinnings remain unclear. To investigate these mechanisms, we performed single-nuclei multiome profiling on postmortem caudate nucleus tissue from six individuals with CocUD and eight controls. We profiled 30,030 nuclei, identifying 13 cell types including D1- and D2-medium spiny neurons (MSNs) and glial cells. We observed 1485 differentially regulated genes and 10,342 differentially accessible peaks, with alterations in MSNs and astrocytes related to neurotransmitter activity and synapse organization. Gene regulatory network analysis identified transcription factors including ZEB1 as exhibiting distinct CocUD-specific subclusters, activating downstream expression of ion- and calcium-channels in MSNs. Further, PDE10A emerged as a potential drug target, showing conserved effects in a rat model. This study highlights cell type-specific molecular alterations in CocUD and provides targets for further investigation, demonstrating the value of multi-omics approaches in addiction research.
Collapse
Affiliation(s)
- Lea Zillich
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Department of Translational Brain Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- HITBR Hector Institute for Translational Brain Research gGmbH, Mannheim, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Center for Mental Health (DZPG), partner site Mannheim/Heidelberg/Ulm, Mannheim, Germany.
| | - Annasara Artioli
- Department of Translational Brain Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Mannheim, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Veronika Pohořalá
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Eric Zillich
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Laura Stertz
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hanna Belschner
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ammar Jabali
- Department of Translational Brain Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Mannheim, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Fabian Streit
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Center for Mental Health (DZPG), partner site Mannheim/Heidelberg/Ulm, Mannheim, Germany
| | - Diana Avetyan
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Maja P Völker
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Svenja Müller
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Center for Mental Health (DZPG), partner site Mannheim/Heidelberg/Ulm, Mannheim, Germany
| | - Anita C Hansson
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas D Meyer
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Julia Ladewig
- Department of Translational Brain Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Mannheim, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rainer Spanagel
- German Center for Mental Health (DZPG), partner site Mannheim/Heidelberg/Ulm, Mannheim, Germany
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ana M M Oliveira
- German Center for Mental Health (DZPG), partner site Mannheim/Heidelberg/Ulm, Mannheim, Germany
- Department of Molecular and Cellular Cognition Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Consuelo Walss-Bass
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rick E Bernardi
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philipp Koch
- Department of Translational Brain Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Mannheim, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Center for Mental Health (DZPG), partner site Mannheim/Heidelberg/Ulm, Mannheim, Germany
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Center for Mental Health (DZPG), partner site Mannheim/Heidelberg/Ulm, Mannheim, Germany
- Center for Innovative Psychiatric and Psychotherapeutic Research, Biobank, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
17
|
Gao Y, Wen P, Liu Y, Sun Y, Qian H, Zhang X, Peng H, Gao Y, Li C, Gu Z, Zeng H, Hong Z, Wang W, Yan R, Hu Z, Fu H. Application of artificial intelligence in the diagnosis of malignant digestive tract tumors: focusing on opportunities and challenges in endoscopy and pathology. J Transl Med 2025; 23:412. [PMID: 40205603 PMCID: PMC11983949 DOI: 10.1186/s12967-025-06428-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: 11/20/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Malignant digestive tract tumors are highly prevalent and fatal tumor types globally, often diagnosed at advanced stages due to atypical early symptoms, causing patients to miss optimal treatment opportunities. Traditional endoscopic and pathological diagnostic processes are highly dependent on expert experience, facing problems such as high misdiagnosis rates and significant inter-observer variations. With the development of artificial intelligence (AI) technologies such as deep learning, real-time lesion detection with endoscopic assistance and automated pathological image analysis have shown potential in improving diagnostic accuracy and efficiency. However, relevant applications still face challenges including insufficient data standardization, inadequate interpretability, and weak clinical validation. OBJECTIVE This study aims to systematically review the current applications of artificial intelligence in diagnosing malignant digestive tract tumors, focusing on the progress and bottlenecks in two key areas: endoscopic examination and pathological diagnosis, and to provide feasible ideas and suggestions for subsequent research and clinical translation. METHODS A systematic literature search strategy was adopted to screen relevant studies published between 2017 and 2024 from databases including PubMed, Web of Science, Scopus, and IEEE Xplore, supplemented with searches of early classical literature. Inclusion criteria included studies on malignant digestive tract tumors such as esophageal cancer, gastric cancer, or colorectal cancer, involving the application of artificial intelligence technology in endoscopic diagnosis or pathological analysis. The effects and main limitations of AI diagnosis were summarized through comprehensive analysis of research design, algorithmic methods, and experimental results from relevant literature. RESULTS In the field of endoscopy, multiple deep learning models have significantly improved detection rates in real-time polyp detection, early gastric cancer, and esophageal cancer screening, with some commercialized systems successfully entering clinical trials. However, the scale and quality of data across different studies vary widely, and the generalizability of models to multi-center, multi-device environments remains to be verified. In pathological analysis, using convolutional neural networks, multimodal pre-training models, etc., automatic tissue segmentation, tumor grading, and assisted diagnosis can be achieved, showing good scalability in interactive question-answering. Nevertheless, clinical implementation still faces obstacles such as non-uniform data standards, lack of large-scale prospective validation, and insufficient model interpretability and continuous learning mechanisms. CONCLUSION Artificial intelligence provides new technological opportunities for endoscopic and pathological diagnosis of malignant digestive tract tumors, achieving positive results in early lesion identification and assisted decision-making. However, to achieve the transition from research to widespread clinical application, data standardization, model reliability, and interpretability still need to be improved through multi-center joint research, and a complete regulatory and ethical system needs to be established. In the future, artificial intelligence will play a more important role in the standardization and precision management of diagnosis and treatment of digestive tract tumors.
Collapse
Affiliation(s)
- Yinhu Gao
- Department of Gastroenterology, Shaanxi Province Rehabilitation Hospital, Xi'an, Shaanxi, China
| | - Peizhen Wen
- Department of General Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Yuan Liu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yahuang Sun
- Division of Colorectal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Hui Qian
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Xin Zhang
- Department of Gastrointestinal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Huan Peng
- Division of Colorectal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Yanli Gao
- Infection Control Office, Shaanxi Province Rehabilitation Hospital, Xi'an, Shaanxi, China
| | - Cuiyu Li
- Department of Radiology, The First Hospital of Nanchang, the Third Affiliated Hospital of Nanchang University, Nanchang, 330008, Jiangxi, China
| | - Zhangyuan Gu
- Tongji University School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China
| | - Huajin Zeng
- Department of General Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Zhijun Hong
- Tongji University School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China
| | - Weijun Wang
- Department of Gastrointestinal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Ronglin Yan
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Zunqi Hu
- Department of Gastrointestinal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Hongbing Fu
- Department of Gastrointestinal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
18
|
Milite S, Caravagna G, Sottoriva A. MIDAA: deep archetypal analysis for interpretable multi-omic data integration based on biological principles. Genome Biol 2025; 26:90. [PMID: 40200293 PMCID: PMC11980162 DOI: 10.1186/s13059-025-03530-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 03/06/2025] [Indexed: 04/10/2025] Open
Abstract
High-throughput multi-omic molecular profiling allows the probing of biological systems at unprecedented resolution. However, integrating and interpreting high-dimensional, sparse, and noisy multimodal datasets remains challenging. Deriving new biological insights with current methods is difficult because they are not rooted in biological principles but prioritise tasks like dimensionality reduction. Here, we introduce a framework that combines archetypal analysis, an approach grounded in biological principles, with deep learning. Using archetypes based on evolutionary trade-offs and Pareto optimality, MIDAA finds extreme data points that define the geometry of the latent space, preserving the complexity of biological interactions while retaining an interpretable output. We demonstrate that these extreme points represent cellular programmes reflecting the underlying biology. Moreover, we show that, compared to alternative methods, MIDAA can identify parsimonious, interpretable, and biologically relevant patterns from real and simulated multi-omics.
Collapse
Affiliation(s)
- Salvatore Milite
- Computational Biology Research Centre, Human Technopole, Milan, Italy.
| | - Giulio Caravagna
- Department of Mathematics, Informatics and Geosciences, University of Trieste, Trieste, Italy.
| | - Andrea Sottoriva
- Computational Biology Research Centre, Human Technopole, Milan, Italy.
| |
Collapse
|
19
|
Xu H, Li M, Yao H, Chen G, Chen J, Hou X, Yang H, Yu C, Lin Z, Zhu J, Wang R, Qiu S, Liu X, Wang Z, Tao X, Liu L. Multi-tissue metabolomics analysis reveals susceptible factors for chemotherapy-induced hepatotoxicity in colorectal cancer patients. Front Pharmacol 2025; 16:1517446. [PMID: 40255576 PMCID: PMC12006014 DOI: 10.3389/fphar.2025.1517446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/19/2025] [Indexed: 04/22/2025] Open
Abstract
Amis Chemotherapy-induced hepatotoxicity (CIH) is a significant concern in colorectal cancer (CRC) patients treated with the CAPEOX (capecitabine and oxaliplatin) regimen. Identifying predictive factors for CIH is crucial for clinical management. Patients and Methods This study analyzed colorectal tissue (CRT), plasma, and urine samples from CRC patients. Differentially expressed metabolites (DEMs) across these tissues were integrated for multi-omics analysis, and predictive models for CIH susceptibility were developed. An independent set of 75 plasma samples was used for validation. Results A total of 492 differentially expressed compounds were identified in samples from 63 CRC patients, including 105, 149, and 238 DEMs in CRT, plasma, and urine, respectively. Lipids and lipid-like molecules were predominant in all samples. Among these, urine samples exhibited the highest variability and provided the strongest predictive power for CIH susceptibility. Principal component analysis (PCA) effectively differentiated normal patients from those with CIH. The study revealed steatosis as the primary pathological feature of CIH, with disrupted lipid metabolism emerging as a key characteristic. Predictive models constructed from multi-tissue metabolites profile exhibited high accuracy, with the plasma model achieving an AUC of 0.933 in external validation set. Our study underscores the importance of individual metabolic variations in CIH susceptibility, reflecting the complex interplay of genetic, environmental, and lifestyle factors. Conclusion This study emphasizes the critical role of alterations in lipid, polyamine, and purine metabolism, as well as impaired tissue repair mechanisms, were identified as key endogenous factors underlying CIH susceptibility. The developed predictive models demonstrate potential for clinical application in assessing CIH risk in CRC patients undergoing CAPEOX chemotherapy.
Collapse
Affiliation(s)
- Huilin Xu
- Institutes of Biomedical Sciences and Intelligent Medicine Institute, Fudan University, Shanghai, China
| | - Mingming Li
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Houshan Yao
- Department of General Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Guoliang Chen
- Department of General Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jiani Chen
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xinyun Hou
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hong Yang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chenghang Yu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People’s Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, China
| | - Zeshuai Lin
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jiawei Zhu
- Traditional Chinese Medicine Resource and Technology Center, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Wang
- Traditional Chinese Medicine Resource and Technology Center, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shi Qiu
- Traditional Chinese Medicine Resource and Technology Center, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuan Liu
- Department of Traditional Chinese Medicine, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhipeng Wang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xia Tao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Lei Liu
- Institutes of Biomedical Sciences and Intelligent Medicine Institute, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Cominetti O, Dayon L. Unravelling disease complexity: integrative analysis of multi-omic data in clinical research. Expert Rev Proteomics 2025; 22:149-162. [PMID: 40207843 DOI: 10.1080/14789450.2025.2491357] [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: 01/24/2025] [Revised: 03/28/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
INTRODUCTION A holistic view on biological systems is today a reality with the application of multi-omic technologies. These technologies allow the profiling of genome, epigenome, transcriptome, proteome, metabolome as well as newly emerging 'omes.' While the multiple layers of data accumulate, their integration and reconciliation in a single system map is a cumbersome exercise that faces many challenges. Application to human health and disease requires large sample sizes, robust methodologies and high-quality standards. AREAS COVERED We review the different methods used to integrate multi-omics, as recent ones including artificial intelligence. With proteomics as an anchor technology, we then present selected applications of its data combination with other omics layers in clinical research, mainly covering literature from the last five years in the Scopus and/or PubMed databases. EXPERT OPINION Multi-omics is powerful to comprehensively type molecular layers and link them to phenotype. Yet, technologies and data are very diverse and still strategies and methodologies to properly integrate these modalities are needed.
Collapse
Affiliation(s)
- Ornella Cominetti
- Proteomics, Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, Lausanne, Switzerland
| | - Loïc Dayon
- Proteomics, Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, Lausanne, Switzerland
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
21
|
Giordano R, Arendt-Nielsen L, Gerra MC, Kappel A, Østergaard SE, Capriotti C, Dallabona C, Petersen KKS. Pain mechanistic networks: the development using supervised multivariate data analysis and implications for chronic pain. Pain 2025; 166:847-857. [PMID: 39297729 DOI: 10.1097/j.pain.0000000000003410] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/20/2024] [Indexed: 03/20/2025]
Abstract
ABSTRACT Chronic postoperative pain is present in approximately 20% of patients undergoing total knee arthroplasty. Studies indicate that pain mechanisms are associated with development and maintenance of chronic postoperative pain. The current study assessed pain sensitivity, inflammation, microRNAs, and psychological factors and combined these in a network to describe chronic postoperative pain. This study involved 75 patients with and without chronic postoperative pain after total knee arthroplasty. Clinical pain intensity, Oxford Knee Score, and pain catastrophizing were assessed as clinical parameters. Quantitative sensory testing was assessed to evaluate pain sensitivity and microRNAs, and inflammatory markers were likewise analyzed. Supervised multivariate data analysis with "Data Integration Analysis for Biomarker Discovery" using Latent cOmponents (DIABLO) was used to describe the chronic postoperative pain intensity. Two DIABLO models were constructed by dividing the patients into 3 groups or 2 defined by clinical pain intensities. Data Integration Analysis for Biomarker discovery using Latent cOmponents model explained chronic postoperative pain and identified factors involved in pain mechanistic networks among assessments included in the analysis. Developing models of 3 or 2 patient groups using the assessments and the networks could explain 81% and 69% of the variability in clinical postoperative pain intensity. The reduction of the number of parameters stabilized the models and reduced the explanatory value to 69% and 51%. This is the first study to use the DIABLO model for chronic postoperative pain and to demonstrate how different pain mechanisms form a pain mechanistic network. The complex model explained 81% of the variability of clinical pain intensity, whereas the less complex model explained 51% of the variability of clinical pain intensity.
Collapse
Affiliation(s)
- Rocco Giordano
- Department of Oral and Maxillofacial Surgery, Aalborg University Hospital, Aalborg, Denmark
- Center for Neuroplasticity and Pain (CNAP), SMI®, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Lars Arendt-Nielsen
- Center for Neuroplasticity and Pain (CNAP), SMI®, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Center for Mathematical Modeling of Knee Osteoarthritis (MathKOA), Department of Material and Production, Faculty of Engineering and Science, Aalborg University, Aalborg, Denmark
- Department of Gastroenterology & Hepatology, MechSense, Aalborg University Hospital, Aalborg, Denmark
- Steno Diabetes Center North Denmark, Clinical Institute, Aalborg University Hospital, Aalborg, Denmark
| | - Maria Carla Gerra
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Andreas Kappel
- Interdisciplinary Orthopedics, Department of Orthopedic Surgery, Aalborg University Hospital, Aalborg University Hospital, Aalborg, Denmark
| | - Svend Erik Østergaard
- Interdisciplinary Orthopedics, Department of Orthopedic Surgery, Aalborg University Hospital, Aalborg University Hospital, Aalborg, Denmark
| | - Camila Capriotti
- Center for Neuroplasticity and Pain (CNAP), SMI®, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Cristina Dallabona
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Kristian Kjær-Staal Petersen
- Center for Neuroplasticity and Pain (CNAP), SMI®, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Center for Mathematical Modeling of Knee Osteoarthritis (MathKOA), Department of Material and Production, Faculty of Engineering and Science, Aalborg University, Aalborg, Denmark
| |
Collapse
|
22
|
Llinas-Bertran A, Butjosa-Espín M, Barberi V, Seoane JA. Multimodal data integration in early-stage breast cancer. Breast 2025; 80:103892. [PMID: 39922065 PMCID: PMC11973824 DOI: 10.1016/j.breast.2025.103892] [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: 10/10/2024] [Revised: 12/13/2024] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
The use of biomarkers in breast cancer has significantly improved patient outcomes through targeted therapies, such as hormone therapy anti-Her2 therapy and CDK4/6 or PARP inhibitors. However, existing knowledge does not fully encompass the diverse nature of breast cancer, particularly in triple-negative tumors. The integration of multi-omics and multimodal data has the potential to provide new insights into biological processes, to improve breast cancer patient stratification, enhance prognosis and response prediction, and identify new biomarkers. This review presents a comprehensive overview of the state-of-the-art multimodal (including molecular and image) data integration algorithms developed and with applicability to breast cancer stratification, prognosis, or biomarker identification. We examined the primary challenges and opportunities of these multimodal data integration algorithms, including their advantages, limitations, and critical considerations for future research. We aimed to describe models that are not only academically and preclinically relevant, but also applicable to clinical settings.
Collapse
Affiliation(s)
- Arnau Llinas-Bertran
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Maria Butjosa-Espín
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Vittoria Barberi
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jose A Seoane
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
23
|
Pearce EM, Evans E, Mayday MY, Reyes G, Simon MR, Blum J, Kim H, Mu J, Shaw PJ, Rowan CM, Auletta JJ, Martin PL, Hurley C, Kreml EM, Qayed M, Abdel-Azim H, Keating AK, Cuvelier GDE, Hume JR, Killinger JS, Godder K, Hanna R, Duncan CN, Quigg TC, Castillo P, Lalefar NR, Fitzgerald JC, Mahadeo KM, Satwani P, Moore TB, Hanisch B, Abdel-Mageed A, Davis DB, Hudspeth MP, Yanik GA, Pulsipher MA, Dvorak CCJL, Zinter MS. Integrating Pulmonary and Systemic Transcriptomic Profiles to Characterize Lung Injury after Pediatric Hematopoietic Stem Cell Transplant. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.31.25324969. [PMID: 40236411 PMCID: PMC11998824 DOI: 10.1101/2025.03.31.25324969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Hematopoietic stem cell transplantation (HCT) is potentially curative for numerous malignant and non-malignant diseases but can lead to lung injury due to chemoradiation toxicity, infection, and immune dysregulation. Bronchoalveolar lavage (BAL) is the most commonly used procedure for diagnostic sampling of the lung but is invasive, cannot be performed in medically fragile patients, and is challenging to perform serially. We previously showed that BAL transcriptomes representing pulmonary inflammation and cellular injury can phenotype post-HCT lung injury and predict mortality outcomes. However, whether peripheral blood testing is a suitable minimally-invasive surrogate for pulmonary sampling in the HCT population remains unknown. To address this question, we compared 210 paired BAL and peripheral blood transcriptomes obtained from 166 pediatric HCT patients at 27 children's hospitals. BAL and blood mRNA abundance showed minimal overall correlation at the level of individual genes, gene set enrichment scores, imputed cell fractions, and T- and B-cell receptor clonotypes. Instead, we identified significant site-specific transcriptional programs. In BAL, expression of innate and adaptive immune pathways was tightly co-regulated with expression of epithelial mesenchymal transition and hypoxia pathways, and these signatures were associated with mortality. In contrast, in blood, expression of endothelial injury, DNA repair, and cellular metabolism pathways was associated with mortality. Integration of paired BAL and blood transcriptomes dichotomized patients into two groups, of which one group showed twice the rate of hypoxia and significantly worse outcomes within 7 days of enrollment. These findings reveal a compartmentalized injury response, where BAL and peripheral blood transcriptomes provide distinct but complementary insights into local and systemic mechanisms of post-HCT lung injury.
Collapse
Affiliation(s)
- Emma M Pearce
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Erica Evans
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Madeline Y Mayday
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Departments of Laboratory Medicine and Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Gustavo Reyes
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Miriam R Simon
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Jacob Blum
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Hanna Kim
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica Mu
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Peter J Shaw
- The Children`s Hospital at Westmead, Westmead, NSW, Australia
| | - Courtney M Rowan
- Indiana University, Department of Pediatrics, Division of Critical Care Medicine, Indianapolis, IN, USA
| | - Jeffrey J Auletta
- Hematology/Oncology/BMT and Infectious Diseases, Nationwide Children's Hospital, Columbus, OH, USA
- CIBMTR (Center for International Blood and Marrow Transplant Research), National Marrow Donor Program/Be The Match, Minneapolis, MN, USA
| | - Paul L Martin
- Division of Pediatric and Cellular Therapy, Duke University Medical Center, Durham, NC, USA
| | - Caitlin Hurley
- Division of Critical Care, Department of Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Erin M Kreml
- Department of Child Health, Division of Critical Care Medicine, University of Arizona, Phoenix, AZ, USA
| | - Muna Qayed
- Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, GA, USA
| | - Hisham Abdel-Azim
- Department of Pediatrics, Division of Hematology/Oncology and Transplant and Cell Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Loma Linda University School of Medicine, Cancer Center, Children Hospital and Medical Center, Loma Linda, CA, USA
| | - Amy K Keating
- Harvard Medical School, Boston, Massachusetts; Division of Pediatric Oncology, Department of Pediatrics, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado and University of Colorado, Aurora, CO, USA
| | - Geoffrey D E Cuvelier
- CancerCare Manitoba, Manitoba Blood and Marrow Transplant Program, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Janet R Hume
- University of Minnesota, Department of Pediatrics, Division of Critical Care Medicine, Minneapolis, MN, USA
| | - James S Killinger
- Division of Pediatric Critical Care, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Kamar Godder
- Cancer and Blood Disorders Center, Nicklaus Children's Hospital, Miami, FL, USA
| | - Rabi Hanna
- Department of Pediatric Hematology, Oncology and Blood and Marrow Transplantation, Pediatric Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Christine N Duncan
- Harvard Medical School, Boston, Massachusetts; Division of Pediatric Oncology, Department of Pediatrics, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, USA
| | - Troy C Quigg
- Pediatric Blood and Marrow Transplantation Program, Texas Transplant Institute, Methodist Children's Hospital, San Antonio, TX, USA
- Section of Pediatric BMT and Cellular Therapy, Helen DeVos Children's Hospital, Grand Rapids, MI, USA
| | - Paul Castillo
- University of Florida, Gainesville, UF Health Shands Children's Hospital, Gainesville, FL, USA
| | - Nahal R Lalefar
- Division of Pediatric Hematology/Oncology, UCSF Benioff Children's Hospital Oakland, University of California San Francisco, Oakland, CA, USA
| | - Julie C Fitzgerald
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Kris M Mahadeo
- Department of Pediatrics, Division of Hematology/Oncology, MD Anderson Cancer Center, Houston, TX, USA
- Division of Pediatric and Cellular Therapy, Duke University Medical Center, Durham, NC, USA
| | - Prakash Satwani
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, Columbia University, New York, NY, USA
| | - Theodore B Moore
- Department of Pediatric Hematology-Oncology, Mattel Children's Hospital, University of California, Los Angeles, CA, USA
| | - Benjamin Hanisch
- Children's National Hospital, Washington, District of Columbia, USA
| | - Aly Abdel-Mageed
- Section of Pediatric BMT and Cellular Therapy, Helen DeVos Children's Hospital, Grand Rapids, MI, USA
| | - Dereck B Davis
- Department of Pediatrics, Hematology/Oncology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michelle P Hudspeth
- Adult and Pediatric Blood & Marrow Transplantation, Pediatric Hematology/Oncology, Medical University of South Carolina Children's Hospital/Hollings Cancer Center, Charleston, SC, USA
| | - Greg A Yanik
- Pediatric Blood and Bone Marrow Transplantation, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Michael A Pulsipher
- Division of Pediatric Hematology and Oncology, Intermountain Primary Children's Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, UT, USA
| | - Christopher C Joseph L Dvorak
- Division of Allergy, Immunology, and Bone Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Matt S Zinter
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Division of Allergy, Immunology, and Bone Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
24
|
Wan Z, Sun X, Li Y, Chu T, Hao X, Cao Y, Zhang P. Applications of Artificial Intelligence in Drug Repurposing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411325. [PMID: 40047357 PMCID: PMC11984889 DOI: 10.1002/advs.202411325] [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: 09/15/2024] [Revised: 12/12/2024] [Indexed: 04/12/2025]
Abstract
Drug repurposing identifies new therapeutic uses for the existing drugs originally developed for different indications, aiming at capitalizing on the established safety and efficacy profiles of known drugs. Thus, it is beneficial to bypass of early stages of drug development, and to reduction of the time and cost associated with bringing new therapies to market. Traditional experimental methods are often time-consuming and expensive, making artificial intelligence (AI) a promising alternative due to its lower cost, computational advantages, and ability to uncover hidden patterns. This review focuses on the availability of AI algorithms in drug development, and their positive and specific roles in revealing repurposing of the existing drugs, especially being integrated with virtual screening. It is shown that the existing AI algorithms excel at analyzing large-scale datasets, identifying the complicated patterns of drug responses from these datasets, and making predictions for potential drug repurposing. Building on these insights, challenges remain in developing efficient AI algorithms and future research, including integrating drug-related data across databases for better repurposing, enhancing AI computational efficiency, and advancing personalized medicine.
Collapse
Affiliation(s)
- Zhaoman Wan
- State Key Laboratory of Common Mechanism Research for Major DiseasesSuzhou Institute of Systems MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeSuzhouJiangsu215123China
| | - Xinran Sun
- Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100193China
| | - Yi Li
- Hunan Agriculture University College of Plant ProtectionChangshaHunan410128China
| | - Tianyao Chu
- Beijing Key Laboratory for Genetics of Birth DefectsBeijing Pediatric Research InstituteMOE Key Laboratory of Major Diseases in ChildrenRare Disease CenterBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijing100045China
| | - Xueyu Hao
- Beijing Key Laboratory for Genetics of Birth DefectsBeijing Pediatric Research InstituteMOE Key Laboratory of Major Diseases in ChildrenRare Disease CenterBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijing100045China
| | - Yang Cao
- College of Life SciencesSichuan UniversityChengduSichuan610041China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth DefectsBeijing Pediatric Research InstituteMOE Key Laboratory of Major Diseases in ChildrenRare Disease CenterBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijing100045China
| |
Collapse
|
25
|
Guan H, Zhang P, Park RF, Ding Y. Genomics Research on the Road of Studying Biology and Virulence of Cereal Rust Fungi. MOLECULAR PLANT PATHOLOGY 2025; 26:e70082. [PMID: 40181494 PMCID: PMC11968332 DOI: 10.1111/mpp.70082] [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: 09/09/2024] [Revised: 03/06/2025] [Accepted: 03/23/2025] [Indexed: 04/05/2025]
Abstract
Rust fungi are highly destructive pathogens that pose a significant threat to crop production worldwide, especially cereals. Obligate biotrophy and, in many cases, complex life cycles make rust fungi particularly challenging to study. However, recent rapid advances in sequencing technologies and genomic analysis tools have revolutionised rust fungal research. It is anticipated that the increasing availability and ongoing substantial improvements in genome assemblies will propel the field of rust biology into the post-genomic era, instigating a cascade of research endeavours encompassing multi-omics and gene discoveries. This is especially the case for many cereal rust pathogens, for which continental-scale studies of virulence have been conducted over many years and historical collections of viable isolates have been sequenced and assembled. Genomic analysis plays a crucial role in uncovering the underlying causes of the high variability of virulence and the complexity of population dynamics in rust fungi. Here, we provide an overview of progress in rust genomics, discuss the strategies employed in genomic analysis, and elucidate the strides that will drive cereal rust biology into the post-genomic era.
Collapse
Affiliation(s)
- Haixia Guan
- School of Life and Environment SciencesPlant Breeding Institute, The University of SydneyCobbittyNew South WalesAustralia
| | - Peng Zhang
- School of Life and Environment SciencesPlant Breeding Institute, The University of SydneyCobbittyNew South WalesAustralia
| | - Robert F. Park
- School of Life and Environment SciencesPlant Breeding Institute, The University of SydneyCobbittyNew South WalesAustralia
| | - Yi Ding
- School of Life and Environment SciencesPlant Breeding Institute, The University of SydneyCobbittyNew South WalesAustralia
| |
Collapse
|
26
|
Johnson Z, Anderson D, Cheung MS, Bohutskyi P. Gene network centrality analysis identifies key regulators coordinating day-night metabolic transitions in Synechococcus elongatus PCC 7942 despite limited accuracy in predicting direct regulator-gene interactions. Front Microbiol 2025; 16:1569559. [PMID: 40207147 PMCID: PMC11979508 DOI: 10.3389/fmicb.2025.1569559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Synechococcus elongatus PCC 7942 is a model organism for studying circadian regulation and bioproduction, where precise temporal control of metabolism significantly impacts photosynthetic efficiency and CO2-to-bioproduct conversion. Despite extensive research on core clock components, our understanding of the broader regulatory network orchestrating genome-wide metabolic transitions remains incomplete. We address this gap by applying machine learning tools and network analysis to investigate the transcriptional architecture governing circadian-controlled gene expression. While our approach showed moderate accuracy in predicting individual transcription factor-gene interactions - a common challenge with real expression data - network-level topological analysis successfully revealed the organizational principles of circadian regulation. Our analysis identified distinct regulatory modules coordinating day-night metabolic transitions, with photosynthesis and carbon/nitrogen metabolism controlled by day-phase regulators, while nighttime modules orchestrate glycogen mobilization and redox metabolism. Through network centrality analysis, we identified potentially significant but previously understudied transcriptional regulators: HimA as a putative DNA architecture regulator, and TetR and SrrB as potential coordinators of nighttime metabolism, working alongside established global regulators RpaA and RpaB. This work demonstrates how network-level analysis can extract biologically meaningful insights despite limitations in predicting direct regulatory interactions. The regulatory principles uncovered here advance our understanding of how cyanobacteria coordinate complex metabolic transitions and may inform metabolic engineering strategies for enhanced photosynthetic bioproduction from CO2.
Collapse
Affiliation(s)
- Zachary Johnson
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, United States
- Department of Biological Systems Engineering, Washington State University, Pullman, WA, United States
| | - David Anderson
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Margaret S. Cheung
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, United States
- Department of Physics, University of Washington, Seattle, WA, United States
| | - Pavlo Bohutskyi
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, United States
- Department of Biological Systems Engineering, Washington State University, Pullman, WA, United States
| |
Collapse
|
27
|
Tillett BJ, Dwiyanto J, Secombe KR, George T, Zhang V, Anderson D, Duggan E, Giri R, Loo D, Stoll T, Morrison M, Begun J, Hill MM, Gurzov EN, Bell KJ, Saad S, Barlow CK, Creek DJ, Chong CW, Mariño E, Hamilton-Williams EE. SCFA biotherapy delays diabetes in humanized gnotobiotic mice by remodeling mucosal homeostasis and metabolome. Nat Commun 2025; 16:2893. [PMID: 40133336 PMCID: PMC11937418 DOI: 10.1038/s41467-025-58319-y] [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: 04/17/2024] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Type 1 diabetes (T1D) is linked to an altered gut microbiota characterized by reduced short-chain fatty acid (SCFA) production. Oral delivery of a SCFA-yielding biotherapy in adults with T1D was followed by increased SCFAs, altered gut microbiota and immunoregulation, as well as delaying diabetes in preclinical models. Here, we show that SCFA-biotherapy in humans is accompanied by remodeling of the gut proteome and mucosal immune homeostasis. Metabolomics showed arginine, glutamate, nucleotide and tryptophan metabolism were enriched following the SCFA-biotherapy, and found metabolites that correlated with glycemic control. Fecal microbiota transfer demonstrated that the microbiota of SCFA-responders delayed diabetes progression in humanized gnotobiotic mice. The protected mice increased similar metabolite pathways to the humans including producing aryl-hydrocarbon receptor ligands and reducing inflammatory mucosal immunity and increasing IgA production in the gut. These data demonstrate that a potent SCFA immunomodulator promotes multiple beneficial pathways and supports targeting the microbiota as an approach against T1D. Trial registration: Australia New Zealand Clinical Trials Registry ACTRN12618001391268.
Collapse
Affiliation(s)
- Bree J Tillett
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jacky Dwiyanto
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Kate R Secombe
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Thomas George
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Vivian Zhang
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Dovile Anderson
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
- Monash Proteomics and Metabolomics Platform, Monash University, MelbourneVIC, Australia
| | - Emily Duggan
- Translational Research Institute, Brisbane, QLD, Australia
| | - Rabina Giri
- Mater Research Institute-The University of Queensland, Brisbane, QLD, Australia
| | - Dorothy Loo
- Translational Research Institute, Brisbane, QLD, Australia
| | - Thomas Stoll
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Mark Morrison
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Jakob Begun
- Mater Research Institute-The University of Queensland, Brisbane, QLD, Australia
| | - Michelle M Hill
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Esteban N Gurzov
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, Brussels, Belgium
| | - Kirstine J Bell
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Sonia Saad
- Department of Medicine, Kolling Institute, University of Sydney, Sydney, NSW, Australia
| | - Christopher K Barlow
- Monash Proteomics and Metabolomics Platform, Monash University, MelbourneVIC, Australia
- Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Darren J Creek
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
- Monash Proteomics and Metabolomics Platform, Monash University, MelbourneVIC, Australia
| | - Chun Wie Chong
- Monash University Microbiome Research Centre, School of Pharmacy, Monash University Malaysia, Selangor, Malaysia
| | - Eliana Mariño
- Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.
- ImmunoBiota Therapeutics Pty Ltd, Melbourne, VIC, Australia.
| | | |
Collapse
|
28
|
Al-Mansour FSH, Almasoudi HH, Albarrati A. Mapping molecular landscapes in triple-negative breast cancer: insights from spatial transcriptomics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04057-3. [PMID: 40119898 DOI: 10.1007/s00210-025-04057-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
The tumor microenvironment (TME) of triple-negative breast cancer (TNBC) is a highly heterogeneous and very aggressive form of the disease that has few suitable treatment options; however, spatial transcriptomics (ST) is a powerful tool for elucidation of the TME in TNBC. Because of its spatial context preservation, ST has a unique capability to map tumor-stroma interactions, immune infiltration, and therapy resistance mechanisms (which are key to understanding TNBC progression), compared with conventional transcriptomics. This review shows the use of ST in TNBC, its utilization in spatial biomarker identification, intratumoral heterogeneity definition, molecular subtyping refinement, and prediction of immunotherapy responses. Recent insight from ST-driven insights has explained the key spatial patterns on immune evasion, chemotherapy resistance, racial disparities of TNBC, and aspects for patient stratification and therapeutic decision. With the integration of ST with the subjects of proteomics and imaging mass cytometry, this approach has been enlarged and is now applied in precision medicine and biomarker discovery. Recently, advancements in AI-based spatial analysis for tumor classification, identification of biomarkers, and creation of therapy prediction models have occurred. However, continued developments in ST technologies, computational tools, and partnerships amongst multiple centers to facilitate the integration of ST into clinical routine practice are needed to unlock novel therapeutic targets.
Collapse
Affiliation(s)
- Fares Saeed H Al-Mansour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Hassan H Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Ali Albarrati
- Rehabilitation Sciences Department, College of Applied Medical Sciences, King Saud University, 11451, Riyadh, Saudi Arabia.
| |
Collapse
|
29
|
Liao X, Li Y, Li S, Wen L, Li X, Yu B. Enhanced Integration of Single-Cell Multi-Omics Data Using Graph Attention Networks. ACS Synth Biol 2025; 14:931-942. [PMID: 39888834 DOI: 10.1021/acssynbio.4c00864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
Abstract
The continuous advancement of single-cell multimodal omics (scMulti-omics) technologies offers unprecedented opportunities to measure various modalities, including RNA expression, protein abundance, gene perturbation, DNA methylation, and chromatin accessibility at single-cell resolution. These advances hold significant potential for breakthroughs by integrating diverse omics modalities. However, the data generated from different omics layers often face challenges due to high dimensionality, heterogeneity, and sparsity, which can adversely impact the accuracy and efficiency of data integration analyses. To address these challenges, we propose a high-precision analysis method called scMGAT (single-cell multiomics data analysis based on multihead graph attention networks). This method effectively coordinates reliable information across multiomics data sets using a multihead attention mechanism, allowing for better management of the heterogeneous characteristics inherent in scMulti-omics data. We evaluated scMGAT's performance on eight sets of real scMulti-omics data, including samples from both human and mouse. The experimental results demonstrate that scMGAT significantly enhances the quality of multiomics data and improves the accuracy of cell-type annotation compared to state-of-the-art methods. scMGAT is now freely accessible at https://github.com/Xingyu-Liao/scMGAT.
Collapse
Affiliation(s)
- Xingyu Liao
- School of Computer Science, Northwestern Polytechnical University (NPU), Chang'an Campus, Xi'an, Shaanxi 710072, P.R. China
| | - Yanyan Li
- School of Computer Science, Northwestern Polytechnical University (NPU), Chang'an Campus, Xi'an, Shaanxi 710072, P.R. China
| | - Shuangyi Li
- School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, P.R. China
| | - Long Wen
- School of Computer Science, Northwestern Polytechnical University (NPU), Chang'an Campus, Xi'an, Shaanxi 710072, P.R. China
| | - Xingyi Li
- School of Computer Science, Northwestern Polytechnical University (NPU), Chang'an Campus, Xi'an, Shaanxi 710072, P.R. China
| | - Bin Yu
- School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, P.R. China
| |
Collapse
|
30
|
Mulvey JF, Meyer EL, Svenningsen MS, Lundby A. Integrating -Omic Technologies across Modality, Space, and Time to Decipher Remodeling in Cardiac Disease. Curr Cardiol Rep 2025; 27:74. [PMID: 40116972 PMCID: PMC11928419 DOI: 10.1007/s11886-025-02226-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 03/23/2025]
Abstract
PURPOSE OF REVIEW Despite significant efforts to understand pathophysiological processes underlying cardiac diseases, the molecular causes for the most part remain unresolved. Rapid advancements in -omics technologies, and their application in cardiac research, offer new insight into cardiac remodeling in disease states. This review aims to provide an accessible overview of recent advances in omics approaches for studying cardiac remodeling, catering to readers without extensive prior expertise. RECENT FINDINGS We provide a methodologically focused overview of current methods for performing transcriptomics and proteomics, including their extensions for single-cell and spatial measurements. We discuss approaches to integrate data across modalities, resolutions and time. Key recent applications within the cardiac field are highlighted. Each -omics modality can provide insight, yet each existing experimental method has technical or conceptual limitations. Integrating data across multiple modalities can leverage strengths and mitigate weaknesses, ultimately enhancing our understanding of cardiac pathophysiology.
Collapse
Affiliation(s)
- John F Mulvey
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emily L Meyer
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Skjoldan Svenningsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
31
|
Zhang S, Hu H, Wang X, Xiong C, Asmann YW, Ren Y. Single-cell multiomics reveals disrupted glial gene regulatory programs in Alzheimer's disease via interpretable machine learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643349. [PMID: 40166228 PMCID: PMC11957018 DOI: 10.1101/2025.03.14.643349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Recent development of single-cell technology across multiple omics platforms has provided new ways to obtain holistic views of cells to study disease pathobiology. Alzheimer's disease (AD) is the most common form of dementia worldwide, yet the detailed understanding of its cellular and molecular mechanisms remains limited. In this study, we analyzed paired single-cell transcriptomic (scRNA-seq) and chromatin accessibility (scATAC-seq) data from the Seattle Alzheimer's Disease Brain Cell Atlas (SEA-AD) Consortium to investigate the molecular mechanisms of AD at a cell-subpopulation-specific resolution focusing on glial cells. We benchmarked various multi-omics integration methods using diverse metrics and built an analytic workflow that enabled effective batch correction and cross-modality alignment, creating a unified cell state space. Through integrative analysis of 26 human brain samples, we uncovered AD-associated gene expression and pathway changes in glial subpopulations and highlighted important transcriptomic and epigenomic signatures via functional inference and interpretable machine learning paradigms, discovering the profound involvement of the Solute Carrier proteins (SLC) family genes in multiple glial cell types. We also identified glial cell-specific regulatory programs mediated by key transcription factors such as JUN and FOSL2 in astrocytes, the Zinc Finger (ZNF) family genes in microglia, and the SOX family of transcription factors in oligodendrocytes. Our study provides a comprehensive workflow and a high-resolution view of how glial regulatory programs are disrupted in AD. Our findings offer novel insights into disease-related changes in gene regulation and suggest potential targets for further research and therapy.
Collapse
|
32
|
Alakwaa F, Das V, Majumdar A, Nair V, Fermin D, Dey AB, Slidel T, Reilly DF, Myshkin E, Duffin KL, Chen Y, Bitzer M, Pennathur S, Brosius FC, Kretzler M, Ju W, Karihaloo A, Eddy S. Leveraging complementary multi-omics data integration methods for mechanistic insights in kidney diseases. JCI Insight 2025; 10:e186070. [PMID: 40059827 PMCID: PMC11949029 DOI: 10.1172/jci.insight.186070] [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: 08/23/2024] [Accepted: 01/22/2025] [Indexed: 03/29/2025] Open
Abstract
Chronic kidney diseases (CKDs) are a global health concern, necessitating a comprehensive understanding of their complex pathophysiology. This study explores the use of 2 complementary multidimensional -omics data integration methods to elucidate mechanisms of CKD progression as a proof of concept. Baseline biosamples from 37 participants with CKD in the Clinical Phenotyping and Resource Biobank Core (C-PROBE) cohort with prospective longitudinal outcome data ascertained over 5 years were used to generate molecular profiles. Tissue transcriptomic, urine and plasma proteomic, and targeted urine metabolomic profiling were integrated using 2 orthogonal multi-omics data integration approaches, one unsupervised and the other supervised. Both integration methods identified 8 urinary proteins significantly associated with long-term outcomes, which were replicated in an adjusted survival model using 94 samples from an independent validation group in the same cohort. The 2 methods also identified 3 shared enriched pathways: the complement and coagulation cascades, cytokine-cytokine receptor interaction pathway, and the JAK/STAT signaling pathway. Use of different multiscalar data integration strategies on the same data enabled identification and prioritization of disease mechanisms associated with CKD progression. Approaches like this will be invaluable with the expansion of high-dimension data in kidney diseases.
Collapse
Affiliation(s)
- Fadhl Alakwaa
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | - Viji Nair
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - Damian Fermin
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Timothy Slidel
- Biopharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | | | | | | | - Yu Chen
- Eli Lilly & Co., Indianapolis, Indiana, USA
| | - Markus Bitzer
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - Subramaniam Pennathur
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Matthias Kretzler
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - Wenjun Ju
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - Anil Karihaloo
- Novo Nordisk Research Center Seattle, Inc, Seattle, Washington, USA
| | - Sean Eddy
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
33
|
Guan A, Quek C. Single-Cell Multi-Omics: Insights into Therapeutic Innovations to Advance Treatment in Cancer. Int J Mol Sci 2025; 26:2447. [PMID: 40141092 PMCID: PMC11942442 DOI: 10.3390/ijms26062447] [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: 02/06/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Advances in single-cell multi-omics technologies have deepened our understanding of cancer biology by integrating genomic, transcriptomic, epigenomic, and proteomic data at single-cell resolution. These single-cell multi-omics technologies provide unprecedented insights into tumour heterogeneity, tumour microenvironment, and mechanisms of therapeutic resistance, enabling the development of precision medicine strategies. The emerging field of single-cell multi-omics in genomic medicine has improved patient outcomes. However, most clinical applications still depend on bulk genomic approaches, which fail to directly capture the genomic variations driving cellular heterogeneity. In this review, we explore the common single-cell multi-omics platforms and discuss key analytical steps for data integration. Furthermore, we highlight emerging knowledge in therapeutic resistance and immune evasion, and the potential of new therapeutic innovations informed by single-cell multi-omics. Finally, we discuss the future directions of the application of single-cell multi-omics technologies. By bridging the gap between technological advancements and clinical implementation, this review provides a roadmap for leveraging single-cell multi-omics to improve cancer treatment and patient outcomes.
Collapse
Affiliation(s)
- Angel Guan
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia;
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Camelia Quek
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia;
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
34
|
Patte C, Pommier RM, Ferrari A, Fei-Lei Chung F, Ouzounova M, Moullé P, Richaud M, Khoueiry R, Hervieu M, Breusa S, Allio M, Rama N, Gérard L, Hervieu V, Poncet G, Fenouil T, Cahais V, Sertier AS, Boland A, Bacq-Daian D, Ducarouge B, Marie JC, Deleuze JF, Viari A, Scoazec JY, Roche C, Mehlen P, Walter T, Gibert B. Comprehensive molecular portrait reveals genetic diversity and distinct molecular subtypes of small intestinal neuroendocrine tumors. Nat Commun 2025; 16:2197. [PMID: 40038310 PMCID: PMC11880452 DOI: 10.1038/s41467-025-57305-8] [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: 01/20/2024] [Accepted: 02/18/2025] [Indexed: 03/06/2025] Open
Abstract
Small intestinal neuroendocrine tumors (siNETs) are rare bowel tumors arising from malignant enteroendocrine cells, which normally regulate digestion throughout the intestine. Though infrequent, their incidence is rising through better diagnosis, fostering research into their origin and treatment. To date, siNETs are considered to be a single entity and are clinically treated as such. Here, by performing a multi-omics analysis of siNETs, we unveil four distinct molecular groups with strong clinical relevance and provide a resource to study their origin and clinical features. Transcriptomic, genetic and DNA methylation profiles identify two groups linked to distinct enteroendocrine differentiation patterns, another with a strong immune phenotype, and the last with mesenchymal properties. This latter subtype displays the worst prognosis and resistance to treatments in line with infiltration of cancer-associated fibroblasts. These data provide insights into the origin and diversity of these rare diseases, in the hope of improving clinical research into their management.
Collapse
Affiliation(s)
- Céline Patte
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
| | - Roxane M Pommier
- Plateforme Bioinformatique Gilles Thomas, Synergie Lyon Cancer, Centre Léon Bérard, Lyon, France
| | - Anthony Ferrari
- Plateforme Bioinformatique Gilles Thomas, Synergie Lyon Cancer, Centre Léon Bérard, Lyon, France
| | - Felicia Fei-Lei Chung
- Epigenetics Group, International Agency for Research on Cancer (IARC), Lyon, France
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Malaysia
| | - Maria Ouzounova
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
| | - Pauline Moullé
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
| | - Mathieu Richaud
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
| | - Rita Khoueiry
- Epigenetics Group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Maëva Hervieu
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
| | - Silvia Breusa
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
| | - Marion Allio
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
| | - Nicolas Rama
- Apoptosis, Cancer and Development (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
| | - Laura Gérard
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Gastroentérologie et d'Oncologie Digestive, Lyon, cedex 03, France
| | - Valérie Hervieu
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
- Hospices Civils de Lyon, Institut de Pathologie Multi-sites, Groupement Hospitalier Est, Bron, France
| | - Gilles Poncet
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Chirurgie Digestive, Lyon, France
| | - Tanguy Fenouil
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
- Hospices Civils de Lyon, Institut de Pathologie Multi-sites, Groupement Hospitalier Est, Bron, France
| | - Vincent Cahais
- Epigenetics Group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Anne-Sophie Sertier
- Plateforme Bioinformatique Gilles Thomas, Synergie Lyon Cancer, Centre Léon Bérard, Lyon, France
- Apoptosis, Cancer and Development (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
| | - Anne Boland
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France
| | - Delphine Bacq-Daian
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France
| | | | - Julien C Marie
- TGF-beta and Immune Response (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Equipe labellisée Ligue nationale contre le cancer, Cancer Research Center of Lyon, Lyon, France
| | - Jean-François Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France
| | - Alain Viari
- Plateforme Bioinformatique Gilles Thomas, Synergie Lyon Cancer, Centre Léon Bérard, Lyon, France
| | - Jean-Yves Scoazec
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - Colette Roche
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
| | - Patrick Mehlen
- Apoptosis, Cancer and Development (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France
| | - Thomas Walter
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France.
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Gastroentérologie et d'Oncologie Digestive, Lyon, cedex 03, France.
| | - Benjamin Gibert
- Gastroenterology and technologies for health (Université Claude Bernard Lyon 1, INSERM U1052, CNRS UMR5286, Centre Léon Bérard), Cancer Research Center of Lyon, Lyon, France.
| |
Collapse
|
35
|
Reinisch I, Ghosh A, Noé F, Sun W, Dong H, Leary P, Dietrich A, Hoffmann A, Blüher M, Wolfrum C. Unveiling adipose populations linked to metabolic health in obesity. Cell Metab 2025; 37:640-655.e4. [PMID: 39694039 DOI: 10.1016/j.cmet.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 08/06/2024] [Accepted: 11/10/2024] [Indexed: 12/20/2024]
Abstract
Precision medicine is still not considered as a standard of care in obesity treatment, despite a large heterogeneity in the metabolic phenotype of individuals with obesity. One of the strongest factors influencing the variability in metabolic disease risk is adipose tissue (AT) dysfunction; however, there is little understanding of the link between distinct cell populations, cell-type-specific transcriptional programs, and disease severity. Here, we generated a comprehensive cellular map of subcutaneous and visceral AT of individuals with metabolically healthy and unhealthy obesity. By combining single-nucleus RNA-sequencing data with bulk transcriptomics and clinical parameters, we identified that mesothelial cells, adipocytes, and adipocyte-progenitor cells exhibit the strongest correlation with metabolic disease. Furthermore, we uncovered cell-specific transcriptional programs, such as the transitioning of mesothelial cells to a mesenchymal phenotype, that are involved in uncoupling obesity from metabolic disease. Together, these findings provide valuable insights by revealing biological drivers of clinical endpoints.
Collapse
Affiliation(s)
- Isabel Reinisch
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Adhideb Ghosh
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland; Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Falko Noé
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland; Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Wenfei Sun
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Hua Dong
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland; Stem Cell Bio Regenerative Med Institute, Stanford University, Stanford, CA, USA
| | - Peter Leary
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Arne Dietrich
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany; Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
36
|
Keele GR, Dzieciatkowska M, Hay AM, Vincent M, O'Connor C, Stephenson D, Reisz JA, Nemkov T, Hansen KC, Page GP, Zimring JC, Churchill GA, D'Alessandro A. Genetic architecture of the red blood cell proteome in genetically diverse mice reveals central role of hemoglobin beta cysteine redox status in maintaining circulating glutathione pools. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640676. [PMID: 40093052 PMCID: PMC11908137 DOI: 10.1101/2025.02.27.640676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Red blood cells (RBCs) transport oxygen but accumulate oxidative damage over time, reducing function in vivo and during storage-critical for transfusions. To explore genetic influences on RBC resilience, we profiled proteins, metabolites, and lipids from fresh and stored RBCs obtained from 350 genetically diverse mice. Our analysis identified over 6,000 quantitative trait loci (QTL). Compared to other tissues, prevalence of trans genetic effects over cis reflects the absence of de novo protein synthesis in anucleated RBCs. QTL hotspots at Hbb, Hba, Mon1a, and storage-specific Steap3 linked ferroptosis to hemolysis. Proteasome components clustered at multiple loci, underscoring the importance of degrading oxidized proteins. Post-translational modifications (PTMs) mapped predominantly to hemoglobins, particularly cysteine residues. Loss of reactive C93 in humanized mice (HBB C93A) disrupted redox balance, affecting glutathione pools, protein glutathionylation, and redox PTMs. These findings highlight genetic regulation of RBC oxidation, with implications for transfusion biology and oxidative stress-dependent hemolytic disorders.
Collapse
|
37
|
Liu S, Yu T. Nonlinear embedding and integration of omics data: a fast and tuning-free approach. Brief Bioinform 2025; 26:bbaf184. [PMID: 40254834 PMCID: PMC12009717 DOI: 10.1093/bib/bbaf184] [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: 01/03/2025] [Revised: 02/26/2025] [Accepted: 03/16/2025] [Indexed: 04/22/2025] Open
Abstract
The rapid progress of single-cell technology has facilitated cost-effective acquisition of diverse omics data, allowing biologists to unravel the complexities of cell populations, disease states, and more. Additionally, single-cell multi-omics technologies have opened new avenues for studying biological interactions. However, the high dimensionality and sparsity of omics data present significant analytical challenges. Dimension reduction (DR) techniques are hence essential for analyzing such complex data, yet many existing methods have inherent limitations. Linear methods like principal component analysis (PCA) struggle to capture intricate associations within data. In response, nonlinear techniques have emerged, but they may face scalability issues, be restricted to single-omics data, or prioritize visualization over generating informative embeddings. Here, we introduce dissimilarity based on conditional ordered list (DCOL) correlation, a novel measure for quantifying nonlinear relationships between variables. Based on this measure, we propose DCOL-PCA and DCOL-Canonical Correlation Analysis for dimension reduction and integration of single- and multi-omics data. In simulations, our methods outperformed nine DR methods and four joint dimension reduction methods, demonstrating stable performance across various settings. We also validated these methods on real datasets, with our method demonstrating its ability to detect intricate signals within and between omics data and generate lower dimensional embeddings that preserve the essential information and latent structures.
Collapse
Affiliation(s)
- Shengjie Liu
- School of Data Science, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 2001 Longxiang Boulevard, Longgang District, Shenzhen 518172, Guangdong, P.R. China
| | - Tianwei Yu
- School of Data Science, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 2001 Longxiang Boulevard, Longgang District, Shenzhen 518172, Guangdong, P.R. China
| |
Collapse
|
38
|
Chuah J, Cordi CV, Hahn J, Hurley JM. Dual-approach co-expression analysis framework (D-CAF) enables identification of novel circadian co-regulation from multi-omic timeseries data. BMC Bioinformatics 2025; 26:72. [PMID: 40038581 PMCID: PMC11881278 DOI: 10.1186/s12859-025-06089-1] [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: 09/18/2024] [Accepted: 02/18/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND The circadian clock is a central driver of many biological and behavioral processes, regulating the levels of many genes and proteins, termed clock controlled genes and proteins (CCGs/CCPs), to impart biological timing at the molecular level. While transcriptomic and proteomic data has been analyzed to find potential CCGs and CCPs, multi-omic modeling of circadian data, which has the potential to enhance the understanding of circadian control of biological timing, remains relatively rare due to several methodological hurdles. To address this gap, a dual-approach co-expression analysis framework (D-CAF) was created to perform co-expression analysis that is robust to Gaussian noise perturbations on time-series measurements of both transcripts and proteins. RESULTS Applying this D-CAF framework to previously gathered transcriptomic and proteomic data from mouse macrophages gathered over circadian time, we identified small, highly significant clusters of oscillating transcripts and proteins in the unweighted similarity matrices and larger, less significant clusters of of oscillating transcripts and proteins using the weighted similarity network. Functional enrichment analysis of these clusters identified novel immunological response pathways that appear to be under circadian control. CONCLUSIONS Overall, our findings suggest that D-CAF is a tool that can be used by the circadian community to integrate multi-omic circadian data to improve our understanding of the mechanisms of circadian regulation of molecular processes.
Collapse
Affiliation(s)
- Joshua Chuah
- Department of Electrical, Computer, and Biomedical Engineering, Union College, 807 Union St, Schenectady, NY, 12308, USA.
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA.
| | - Carmalena V Cordi
- Department of Biological Sciences, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Juergen Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Jennifer M Hurley
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA.
- Department of Biological Sciences, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA.
| |
Collapse
|
39
|
Narayana JK, Mac Aogáin M, Hansbro PM, Chotirmall SH. The bronchiectasis microbiome: current understanding and treatment implications. Curr Opin Pulm Med 2025; 31:135-144. [PMID: 39492755 DOI: 10.1097/mcp.0000000000001131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
PURPOSE OF REVIEW Advances in DNA sequencing and analysis of the respiratory microbiome highlight its close association with bronchiectasis phenotypes, revealing fresh opportunities for diagnosis, stratification, and personalized clinical intervention. An under-recognized condition, bronchiectasis is increasingly the subject of recent large-scale, multicentre, and longitudinal clinical studies including detailed analysis of the microbiome. In this review, we summarize recent progress in our understanding of the bronchiectasis microbiome within the context of its potential use in treatment decisions. RECENT FINDINGS Diverse microbiome profiles exist in bronchiectasis, in line with the established disease heterogeneity including treatment response. Classical microbiology has established Pseudomonas aeruginosa and Haemophilus influenza as two microbial markers of disease, while holistic microbiome analysis has uncovered important associations with less common bacterial taxa including commensal an/or pathobiont species, including the emerging role of the fungal mycobiome, virome, and interactome. Integration of airway microbiomes with other high-dimensional biological and clinical datasets holds significant promise to determining treatable traits and mechanisms of disease related to the microbiome. SUMMARY The bronchiectasis microbiome is an emerging and key area of study with significant implications for understanding bronchiectasis, influencing treatment decisions and ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Jayanth Kumar Narayana
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Micheál Mac Aogáin
- Department of Biochemistry, St. James's Hospital
- School of Medicine, Trinity College, Dublin, Ireland
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, New South Wales, Australia
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| |
Collapse
|
40
|
Huang Y, Zhu S, Yao S, Zhai H, Liu C, Han JDJ. Unraveling aging from transcriptomics. Trends Genet 2025; 41:218-235. [PMID: 39424502 DOI: 10.1016/j.tig.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/21/2024]
Abstract
Research into aging constitutes a pivotal endeavor aimed at elucidating the underlying biological mechanisms governing aging and age-associated diseases, as well as promoting healthy longevity. Recent advances in transcriptomic technologies, such as bulk RNA sequencing (RNA-seq), single-cell transcriptomics, and spatial transcriptomics, have revolutionized our ability to study aging at unprecedented resolution and scale. These technologies present novel opportunities for the discovery of biomarkers, elucidation of molecular pathways, and development of targeted therapeutic strategies for age-related disorders. This review surveys recent breakthroughs in different types of transcripts on aging, such as mRNA, long noncoding (lnc)RNA, tRNA, and miRNA, highlighting key findings and discussing their potential implications for future studies in this field.
Collapse
Affiliation(s)
- Yuanfang Huang
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shouxuan Zhu
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shuai Yao
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Haotian Zhai
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Chenyang Liu
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, China.
| |
Collapse
|
41
|
Shinde P, Willemsen L, Anderson M, Aoki M, Basu S, Burel JG, Cheng P, Ghosh Dastidar S, Dunleavy A, Einav T, Forschmiedt J, Fourati S, Garcia J, Gibson W, Greenbaum JA, Guan L, Guan W, Gygi JP, Ha B, Hou J, Hsiao J, Huang Y, Jansen R, Kakoty B, Kang Z, Kobie JJ, Kojima M, Konstorum A, Lee J, Lewis SA, Li A, Lock EF, Mahita J, Mendes M, Meng H, Neher A, Nili S, Olsen LR, Orfield S, Overton JA, Pai N, Parker C, Qian B, Rasmussen M, Reyna J, Richardson E, Safo S, Sorenson J, Srinivasan A, Thrupp N, Tippalagama R, Trevizani R, Ventz S, Wang J, Wu CC, Ay F, Grant B, Kleinstein SH, Peters B. Putting computational models of immunity to the test-An invited challenge to predict B.pertussis vaccination responses. PLoS Comput Biol 2025; 21:e1012927. [PMID: 40163550 PMCID: PMC11978014 DOI: 10.1371/journal.pcbi.1012927] [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: 09/23/2024] [Revised: 04/08/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
Systems vaccinology studies have been used to build computational models that predict individual vaccine responses and identify the factors contributing to differences in outcome. Comparing such models is challenging due to variability in study designs. To address this, we established a community resource to compare models predicting B. pertussis booster responses and generate experimental data for the explicit purpose of model evaluation. We here describe our second computational prediction challenge using this resource, where we benchmarked 49 algorithms from 53 scientists. We found that the most successful models stood out in their handling of nonlinearities, reducing large feature sets to representative subsets, and advanced data preprocessing. In contrast, we found that models adopted from literature that were developed to predict vaccine antibody responses in other settings performed poorly, reinforcing the need for purpose-built models. Overall, this demonstrates the value of purpose-generated datasets for rigorous and open model evaluations to identify features that improve the reliability and applicability of computational models in vaccine response prediction.
Collapse
Affiliation(s)
- Pramod Shinde
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Lisa Willemsen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Michael Anderson
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Minori Aoki
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Saonli Basu
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Julie G. Burel
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Peng Cheng
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Souradipto Ghosh Dastidar
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Aidan Dunleavy
- School of Statistics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Tal Einav
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Department of Medicine, University of California San Diego, San Diego, California, United States of America
| | - Jamie Forschmiedt
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Slim Fourati
- Department of Medicine, Division of Allergy and Immunology, Feinberg School of Medicine and Center for Human Immunobiology, Northwestern University, Chicago, Illinois, United States of America
| | - Javier Garcia
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - William Gibson
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institute of Health, Bethesda, Maryland, United States of America
| | - Jason A. Greenbaum
- LJI Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Leying Guan
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Weikang Guan
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Jeremy P. Gygi
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
| | - Brendan Ha
- LJI Bioinformatics Core, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Joe Hou
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jason Hsiao
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Rick Jansen
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Bhargob Kakoty
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Zhiyu Kang
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - James J. Kobie
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Mari Kojima
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Anna Konstorum
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Laboratory for Systems Biology, University of Florida, Gainesville, Florida, United States of America
| | - Jiyeun Lee
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Sloan A. Lewis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Aixin Li
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Eric F. Lock
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jarjapu Mahita
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Marcus Mendes
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Hailong Meng
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Aidan Neher
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Somayeh Nili
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Lars Rønn Olsen
- Department of Immunology and Microbiology, LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Shelby Orfield
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | | | - Nidhi Pai
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Cokie Parker
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, Maryland, United States of America
| | - Brian Qian
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Mikkel Rasmussen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Joaquin Reyna
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, California, United States of America
| | - Eve Richardson
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Sandra Safo
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Josey Sorenson
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Aparna Srinivasan
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Nicola Thrupp
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Rashmi Tippalagama
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Raphael Trevizani
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Fundação Oswaldo Cruz, Fiocruz - Ceará, Brazil
| | - Steffen Ventz
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jiuzhou Wang
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Cheng-Chang Wu
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ferhat Ay
- Department of Medicine, University of California San Diego, San Diego, California, United States of America
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, California, United States of America
| | - Barry Grant
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Steven H. Kleinstein
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Bjoern Peters
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
- Department of Medicine, University of California San Diego, San Diego, California, United States of America
| |
Collapse
|
42
|
Patterson WB, Young ND, Holzhausen EA, Lurmann F, Liang D, Walker DI, Jones DP, Liao J, Chen Z, Conti DV, Chatzi L, Goodrich JA, Alderete TL. Oxidative gaseous air pollutant exposure interacts with PNPLA3-I148M genotype to influence liver fat fraction and multi-omics profiles in young adults. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125692. [PMID: 39864653 PMCID: PMC11859754 DOI: 10.1016/j.envpol.2025.125692] [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: 08/08/2024] [Revised: 12/10/2024] [Accepted: 01/13/2025] [Indexed: 01/28/2025]
Abstract
PNPLA3-I148M genotype is the strongest predictive single-nucleotide polymorphism for liver fat. We examine whether PNPLA3-I148M modifies associations between oxidative gaseous air pollutant exposure (Oxwt) with i) liver fat and ii) multi-omics profiles of miRNAs and metabolites linked to liver fat. Participants were 69 young adults (17-22 years) from the Meta-AIR cohort. Prior-month residential Oxwt exposure (redox-weighted oxidative capacity of nitrogen dioxide and ozone) was spatially interpolated from monitoring stations via inverse-distance-squared weighting. Liver fat fraction was assessed by MRI. Serum miRNAs and metabolites were assayed via NanoString nCounter and LC-HRMS, respectively. Multi-omics factor analysis (MOFA) was used to identify latent factors with shared variance across omics layers. Multivariable linear regression models adjusted for age, sex, body mass index, and genotype with liver fat or MOFA factors as an outcome and examined PNPLA3 (rs738409; CC/CG vs. GG) as a multiplicative interaction term. Overall, a standard deviation difference in Oxwt exposure was associated with 8.9% relative increase in liver fat (p = 0.04) and this relationship differed by PNPLA3 genotype (p-value for interaction term: pintx<0.001), whereby relative increases in liver fat for GG and CC/CG participants were 71.8% and 2.4%, respectively. There was no main effect of Oxwt on MOFA Factor 1 expression (p = 0.85), but there was an interaction with PNPLA3 genotype (pintx = 0.01), whereby marginal slopes were 0.211 and -0.017 for GG and CC/CG participants, respectively. MOFA Factor 1 in turn was associated with liver fat (p = 0.006). MOFA Factor 1 miRNAs targeted genes in Fatty Acid Biosynthesis and Metabolism and Lysine Degradation pathways. MOFA Factor 9 was also associated with liver fat and was comprised of branched-chain keto acid and amino acid metabolites. The effects of Oxwt exposure on liver fat is exacerbated in young adults with two PNPLA3 risk alleles, potentially through differential effects on miRNA and/or metabolite profiles.
Collapse
Affiliation(s)
- William B Patterson
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nathan D Young
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Elizabeth A Holzhausen
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Donghai Liang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Douglas I Walker
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Jiawen Liao
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Zhanghua Chen
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - David V Conti
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Lida Chatzi
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Jesse A Goodrich
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Tanya L Alderete
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
43
|
Suo Y, Song Y, Wang Y, Liu Q, Rodriguez H, Zhou H. Advancements in proteogenomics for preclinical targeted cancer therapy research. BIOPHYSICS REPORTS 2025; 11:56-76. [PMID: 40070661 PMCID: PMC11891078 DOI: 10.52601/bpr.2024.240053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 03/14/2025] Open
Abstract
Advancements in molecular characterization technologies have accelerated targeted cancer therapy research at unprecedented resolution and dimensionality. Integrating comprehensive multi-omic molecular profiling of a tumor, proteogenomics, marks a transformative milestone for preclinical cancer research. In this paper, we initially provided an overview of proteogenomics in cancer research, spanning genomics, transcriptomics, and proteomics. Subsequently, the applications were introduced and examined from different perspectives, including but not limited to genetic alterations, molecular quantifications, single-cell patterns, different post-translational modification levels, subtype signatures, and immune landscape. We also paid attention to the combined multi-omics data analysis and pan-cancer analysis. This paper highlights the crucial role of proteogenomics in preclinical targeted cancer therapy research, including but not limited to elucidating the mechanisms of tumorigenesis, discovering effective therapeutic targets and promising biomarkers, and developing subtype-specific therapies.
Collapse
Affiliation(s)
- Yuying Suo
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanli Song
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuqiu Wang
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Qian Liu
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Rockville, MD 20850, USA
| | - Hu Zhou
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
44
|
Gill J, Dasgupta A, Manry B, Markuzon N. Combining single-cell ATAC and RNA sequencing for supervised cell annotation. BMC Bioinformatics 2025; 26:67. [PMID: 40011801 PMCID: PMC11863512 DOI: 10.1186/s12859-025-06084-6] [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: 11/20/2024] [Accepted: 02/13/2025] [Indexed: 02/28/2025] Open
Abstract
MOTIVATION Single-cell analysis offers insights into cellular heterogeneity and individual cell function. Cell type annotation is the first and critical step for performing such an analysis. Current methods mostly utilize single-cell RNA sequencing data. Several studies demonstrated improved unsupervised annotation when combining RNA with single-cell ATAC sequencing, but improvements in supervised methods have not been explored. RESULTS Single-cell 10x genomics multiome datasets containing paired ATAC and RNA from human peripheral blood mononuclear cells (PBMC) and neuronal cells with Alzheimer's Disease were used for supervised annotation. Using linear and nonlinear dimensionality reduction methods and random forest, support vector machine and logistic regression classification models, we demonstrate the improvement in supervised annotation and prediction confidence in PBMC data when using a combination of RNA seq and ATAC-seq data. No such improvement was observed when annotating neuronal cells. Specifically, F1 scores were improved when using scVI embeddings to annotate PBMC sub-types. CD4 T effector memory cells showed the largest improvement in F1 score.
Collapse
Affiliation(s)
- Jaidip Gill
- School of Public Health, Imperial College London, London, England
| | | | - Brychan Manry
- Oncology Data Science, AstraZeneca, Gaithersburg, MD, USA
| | | |
Collapse
|
45
|
Zillich E, Artioli A, Rossetti AC, Avetyan D, Belschner H, Frank J, Stein F, Schwarz JJ, Mechawar N, Turecki G, Nöthen MM, Hansson AC, Witt CC, Rietschel M, Koch P, Spanagel R, Zillich L, Witt SH. A multi-omics and cell type-specific characterization of the ventral striatum in human cocaine use disorder. Cell Rep 2025; 44:115332. [PMID: 39954253 DOI: 10.1016/j.celrep.2025.115332] [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: 10/10/2024] [Revised: 12/11/2024] [Accepted: 01/28/2025] [Indexed: 02/17/2025] Open
Abstract
Epigenome, transcriptome, and proteome analyses of postmortem brains have revealed initial molecular insights into cocaine use disorder (CUD). However, the inter-relationship between these omics and the contribution of individual cell types remains largely unknown. We present an in-depth analysis of molecular changes in the ventral striatum in CUD at multi-omics and single-cell resolution. Integrative multi-omics analyses of microRNA sequencing (microRNA-seq), RNA sequencing (RNA-seq), and proteomics datasets in 41 individuals and single-nuclei RNA-seq in a subset of 16 individuals revealed conserved deregulation of metabolic pathways, oxidative phosphorylation, and glutamatergic signaling. Cell type-specific analyses identified inverse metabolic pathway deregulation patterns in glial and neuronal cells, notably in astrocytes and medium-spiny neurons (MSNs). Characterizing astrocyte-neuron crosstalk revealed altered glutamatergic and cell-cell adhesion signaling in CUD. By applying a comprehensive multi-omics analytical framework, our study provides novel insights into CUD-associated molecular changes in the ventral striatum highlighting the perturbation of astrocytes, MSNs, and their crosstalk in CUD.
Collapse
Affiliation(s)
- Eric Zillich
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Annasara Artioli
- Department of Translational Brain Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany; HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Andrea C Rossetti
- Department of Translational Brain Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany; HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Diana Avetyan
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Hanna Belschner
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Jennifer J Schwarz
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, 53127 Bonn, Germany
| | - Anita C Hansson
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Christian C Witt
- Department of Anesthesiology and Operative Intensive Care, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Philipp Koch
- Department of Translational Brain Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany; HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany; German Center for Mental Health (DZPG), partner site Mannheim/Heidelberg/Ulm, 68159 Mannheim, Germany
| | - Lea Zillich
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany; Department of Translational Brain Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany; HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany; German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; German Center for Mental Health (DZPG), partner site Mannheim/Heidelberg/Ulm, 68159 Mannheim, Germany.
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany; German Center for Mental Health (DZPG), partner site Mannheim/Heidelberg/Ulm, 68159 Mannheim, Germany; Center for Innovative Psychiatric and Psychotherapeutic Research, Biobank, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany.
| |
Collapse
|
46
|
Bailin SS, Ma S, Perry AS, Terry JG, Carr JJ, Nair S, Silver HJ, Shi M, Mashayekhi M, Kropski JA, Ferguson JF, Wanjalla CN, Das SR, Shah R, Koethe JR, Gabriel CL. The Primacy of Adipose Tissue Gene Expression and Plasma Lipidome in Cardiometabolic Disease in Persons With HIV. J Infect Dis 2025; 231:e407-e418. [PMID: 39657693 PMCID: PMC11841643 DOI: 10.1093/infdis/jiae532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Persons with HIV (PWH) on contemporary antiretroviral therapy (ART) are at elevated risk for developing age-related cardiometabolic diseases. We hypothesized that integrative analysis of cross-tissue, multimodal data from PWH could provide insight into molecular programming that defines cardiometabolic phenotypes in this high-risk group. METHODS We enrolled 93 PWH without diabetes who were virologically suppressed on contemporary ART and obtained measures of insulin resistance, glucose intolerance, and adiposity. We performed circulating lipidomics, proteomics, and metabolomics, as well as subcutaneous adipose tissue (SAT) bulk transcriptomics, and used multiomics factor analysis (MOFA) to perform integrative analyses of these datasets. RESULTS The median age was 43 years, median body mass index 30.8 kg/m2, 81% were male, and 56% were self-identified non-Hispanic White. We identified a specific MOFA factor associated with visceral adipose tissue volume (ρ = -0.43), homeostasis model assessment 2 insulin resistance score (ρ = -0.52), liver density (ρ = 0.43), and other cardiometabolic risk factors, which explained more variance in the SAT transcriptome and circulating lipidome compared with the circulating proteome and metabolome. Gene set enrichment analysis of this factor showed extracellular matrix and inflammatory pathways that primarily mapped to SAT myeloid cells and adipose progenitor cells using single-cell deconvolution. Lipidomic analysis showed that this factor was significantly enriched for triacylglycerol and diacylglycerol species. CONCLUSIONS Our multiomic analysis demonstrated coordinated, multitissue molecular reprogramming in virologically suppressed PWH with elevated cardiometabolic disease risk. Longitudinal studies of PWH with assessments of adipose tissue and lipid handling are necessary to understand mechanisms of cardiometabolic disease in PWH. Clinical Trials Registration. NCT04451980.
Collapse
Affiliation(s)
- Samuel S Bailin
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Siyuan Ma
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Andrew S Perry
- Department of Medicine, Division of Cardiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James G Terry
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John Jeffrey Carr
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sangeeta Nair
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Heidi J Silver
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Mingjian Shi
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mona Mashayekhi
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jonathan A Kropski
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jane F Ferguson
- Department of Medicine, Division of Cardiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Celestine N Wanjalla
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Suman R Das
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ravi Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Department of Medicine, Division of Cardiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John R Koethe
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Curtis L Gabriel
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
47
|
Zheng H, Sarkar H, Raphael BJ. Joint imputation and deconvolution of gene expression across spatial transcriptomics platforms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638195. [PMID: 40027720 PMCID: PMC11870578 DOI: 10.1101/2025.02.17.638195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Spatially resolved transcriptomics (SRT) technologies measure gene expression across thousands of spatial locations within a tissue slice. Multiple SRT technologies are currently available and others are in active development with each technology having varying spatial resolution (subcellular, single-cell, or multicellular regions), gene coverage (targeted vs. whole-transcriptome), and sequencing depth per location. For example, the widely used 10x Genomics Visium platform measures whole transcriptomes from multiple-cell-sized spots, while the 10x Genomics Xenium platform measures a few hundred genes at subcellular resolution. A number of studies apply multiple SRT technologies to slices that originate from the same biological tissue. Integration of data from different SRT technologies can overcome limitations of the individual technologies enabling the imputation of expression from unmeasured genes in targeted technologies and/or the deconvolution of ad-mixed expression from technologies with lower spatial resolution. We introduce Spatial Integration for Imputation and Deconvolution (SIID), an algorithm to reconstruct a latent spatial gene expression matrix from a pair of observations from different SRT technologies. SIID leverages a spatial alignment and uses a joint non-negative factorization model to accurately impute missing gene expression and infer gene expression signatures of cell types from ad-mixed SRT data. In simulations involving paired SRT datasets from different technologies (e.g., Xenium and Visium), SIID shows superior performance in reconstructing spot-to-cell-type assignments, recovering cell-type-specific gene expression, and imputing missing data compared to contemporary tools. When applied to real-world 10x Xenium-Visium pairs from human breast and colon cancer tissues, SIID achieves highest performance in imputing holdout gene expression. A PyTorch implementation of SIID is available at https://github.com/raphael-group/siid .
Collapse
Affiliation(s)
- Hongyu Zheng
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Hirak Sarkar
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Ludwig Cancer Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
| | | |
Collapse
|
48
|
Pateras J, Lodi M, Rana P, Ghosh P. Heterogeneous Clustering of Multiomics Data for Breast Cancer Subgroup Classification and Detection. Int J Mol Sci 2025; 26:1707. [PMID: 40004168 PMCID: PMC11855380 DOI: 10.3390/ijms26041707] [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: 01/09/2025] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
The rapid growth of diverse -omics datasets has made multiomics data integration crucial in cancer research. This study adapts the expectation-maximization routine for the joint latent variable modeling of multiomics patient profiles. By combining this approach with traditional biological feature selection methods, this study optimizes latent distribution, enabling efficient patient clustering from well-studied cancer types with reduced computational expense. The proposed optimization subroutines enhance survival analysis and improve runtime performance. This article presents a framework for distinguishing cancer subtypes and identifying potential biomarkers for breast cancer. Key insights into individual subtype expression and function were obtained through differentially expressed gene analysis and pathway enrichment for BRCA patients. The analysis compared 302 tumor samples to 113 normal samples across 60,660 genes. The highly upregulated gene COL10A1, promoting breast cancer progression and poor prognosis, and the consistently downregulated gene CDG300LG, linked to brain metastatic cancer, were identified. Pathway enrichment analysis revealed similarities in cellular matrix organization pathways across subtypes, with notable differences in functions like cell proliferation regulation and endocytosis by host cells. GO Semantic Similarity analysis quantified gene relationships in each subtype, identifying potential biomarkers like MATN2, similar to COL10A1. These insights suggest deeper relationships within clusters and highlight personalized treatment potential based on subtypes.
Collapse
Affiliation(s)
- Joseph Pateras
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Musaddiq Lodi
- Integrative Life Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Pratip Rana
- Department of Computer Science, Old Dominion University, Norfolk, VA 23529, USA
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA;
| |
Collapse
|
49
|
Sabit H, Arneth B, Altrawy A, Ghazy A, Abdelazeem RM, Adel A, Abdel-Ghany S, Alqosaibi AI, Deloukas P, Taghiyev ZT. Genetic and Epigenetic Intersections in COVID-19-Associated Cardiovascular Disease: Emerging Insights and Future Directions. Biomedicines 2025; 13:485. [PMID: 40002898 PMCID: PMC11852909 DOI: 10.3390/biomedicines13020485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/23/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
The intersection of COVID-19 and cardiovascular disease (CVD) has emerged as a significant area of research, particularly in understanding the impact of antiplatelet therapies like ticagrelor and clopidogrel. COVID-19 has been associated with acute cardiovascular complications, including myocardial infarction, thrombosis, and heart failure, exacerbated by the virus's ability to trigger widespread inflammation and endothelial dysfunction. MicroRNAs (miRNAs) play a critical role in regulating these processes by modulating the gene expressions involved in platelet function, inflammation, and vascular homeostasis. This study explores the potential of miRNAs such as miR-223 and miR-126 as biomarkers for predicting resistance or responsiveness to antiplatelet therapies in COVID-19 patients with cardiovascular disease. Identifying miRNA signatures linked to drug efficacy could optimize treatment strategies for patients at high risk of thrombotic events during COVID-19 infection. Moreover, understanding miRNA-mediated pathways offers new insights into how SARS-CoV-2 exacerbates CVD, particularly through mechanisms like cytokine storms and endothelial damage. The findings of this research could lead to personalized therapeutic approaches, improving patient outcomes and reducing mortality in COVID-19-associated cardiovascular events. With global implications, this study addresses the urgent need for effective management of CVD in the context of COVID-19, focusing on the integration of molecular biomarkers to enhance the precision of antiplatelet therapy.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Afaf Altrawy
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Aysha Ghazy
- Department of Agri-Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Rawan M. Abdelazeem
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Amro Adel
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Amany I. Alqosaibi
- Department of Biology, College of Science, Imam Abdulrahman bin Faisal University, Dammam 31441, Saudi Arabia
| | - Panos Deloukas
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK;
| | - Zulfugar T. Taghiyev
- Department of Cardiovascular Surgery, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
50
|
Gabernet G, Maciuch J, Gygi JP, Moore JF, Hoch A, Syphurs C, Chu T, Jayavelu ND, Corry DB, Kheradmand F, Baden LR, Sekaly RP, McComsey GA, Haddad EK, Cairns CB, Rouphael N, Fernandez-Sesma A, Simon V, Metcalf JP, Agudelo Higuita NI, Hough CL, Messer WB, Davis MM, Nadeau KC, Pulendran B, Kraft M, Bime C, Reed EF, Schaenman J, Erle DJ, Calfee CS, Atkinson MA, Brackenridge SC, Melamed E, Shaw AC, Hafler DA, Ozonoff A, Bosinger SE, Eckalbar W, Maecker HT, Kim-Schulze S, Steen H, Krammer F, Westendorf K, Network I, Peters B, Fourati S, Altman MC, Levy O, Smolen KK, Montgomery RR, Diray-Arce J, Kleinstein SH, Guan L, Ehrlich LIR. Identification of a multi-omics factor predictive of long COVID in the IMPACC study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637926. [PMID: 39990442 PMCID: PMC11844572 DOI: 10.1101/2025.02.12.637926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Following SARS-CoV-2 infection, ∼10-35% of COVID-19 patients experience long COVID (LC), in which often debilitating symptoms persist for at least three months. Elucidating the biologic underpinnings of LC could identify therapeutic opportunities. We utilized machine learning methods on biologic analytes and patient reported outcome surveys provided over 12 months after hospital discharge from >500 hospitalized COVID-19 patients in the IMPACC cohort to identify a multi-omics "recovery factor". IMPACC participants who experienced LC had lower recovery factor scores compared to participants without LC. Biologic characterization revealed increased levels of plasma proteins associated with inflammation, elevated transcriptional signatures of heme metabolism, and decreased androgenic steroids in LC patients. The recovery factor was also associated with altered circulating immune cell frequencies. Notably, recovery factor scores were predictive of LC occurrence in patients as early as hospital admission, irrespective of acute disease severity. Thus, the recovery factor identifies patients at risk of LC early after SARS-CoV-2 infection and reveals LC biomarkers and potential treatment targets.
Collapse
|