Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Nature ; 632(8027): 995-1008, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38862027

ABSTRACT

The recent acceleration of commercial, private and multi-national spaceflight has created an unprecedented level of activity in low Earth orbit, concomitant with the largest-ever number of crewed missions entering space and preparations for exploration-class (lasting longer than one year) missions. Such rapid advancement into space from many new companies, countries and space-related entities has enabled a 'second space age'. This era is also poised to leverage, for the first time, modern tools and methods of molecular biology and precision medicine, thus enabling precision aerospace medicine for the crews. The applications of these biomedical technologies and algorithms are diverse, and encompass multi-omic, single-cell and spatial biology tools to investigate human and microbial responses to spaceflight. Additionally, they extend to the development of new imaging techniques, real-time cognitive assessments, physiological monitoring and personalized risk profiles tailored for astronauts. Furthermore, these technologies enable advancements in pharmacogenomics, as well as the identification of novel spaceflight biomarkers and the development of corresponding countermeasures. In this Perspective, we highlight some of the recent biomedical research from the National Aeronautics and Space Administration, Japan Aerospace Exploration Agency, European Space Agency and other space agencies, and detail the entrance of the commercial spaceflight sector (including SpaceX, Blue Origin, Axiom and Sierra Space) into aerospace medicine and space biology, the first aerospace medicine biobank, and various upcoming missions that will utilize these tools to ensure a permanent human presence beyond low Earth orbit, venturing out to other planets and moons.


Subject(s)
Aerospace Medicine , Astronauts , Multiomics , Space Flight , Humans , Aerospace Medicine/methods , Aerospace Medicine/trends , Biological Specimen Banks , Biomarkers/metabolism , Biomarkers/analysis , Cognition , Internationality , Monitoring, Physiologic/methods , Monitoring, Physiologic/trends , Multiomics/methods , Multiomics/trends , Pharmacogenetics/methods , Pharmacogenetics/trends , Precision Medicine/methods , Precision Medicine/trends , Space Flight/methods , Space Flight/trends
2.
Nucleic Acids Res ; 52(D1): D67-D71, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37971299

ABSTRACT

The Bioinformation and DNA Data Bank of Japan (DDBJ) Center (https://www.ddbj.nig.ac.jp) provides database archives that cover a wide range of fields in life sciences. As a founding member of the International Nucleotide Sequence Database Collaboration (INSDC), DDBJ accepts and distributes nucleotide sequence data as well as their study and sample information along with the National Center for Biotechnology Information in the United States and the European Bioinformatics Institute (EBI). Besides INSDC databases, the DDBJ Center provides databases for functional genomics (GEA: Genomic Expression Archive), metabolomics (MetaboBank) and human genetic and phenotypic data (JGA: Japanese Genotype-phenotype Archive). These database systems have been built on the National Institute of Genetics (NIG) supercomputer, which is also open for domestic life science researchers to analyze large-scale sequence data. This paper reports recent updates on the archival databases and the services of the DDBJ Center, highlighting the newly redesigned MetaboBank. MetaboBank uses BioProject and BioSample in its metadata description making it suitable for multi-omics large studies. Its collaboration with MetaboLights at EBI brings synergy in locating and reusing public data.


Subject(s)
Databases, Nucleic Acid , Metabolomics , Metadata , Humans , Computational Biology , Genomics , Internet , Japan , Multiomics/methods
4.
BMC Bioinformatics ; 25(1): 257, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107690

ABSTRACT

The recent advances in high-throughput single-cell sequencing have created an urgent demand for computational models which can address the high complexity of single-cell multiomics data. Meticulous single-cell multiomics integration models are required to avoid biases towards a specific modality and overcome sparsity. Batch effects obfuscating biological signals must also be taken into account. Here, we introduce a new single-cell multiomics integration model, Single-cell Multiomics Autoencoder Integration (scMaui) based on variational product-of-experts autoencoders and adversarial learning. scMaui calculates a joint representation of multiple marginal distributions based on a product-of-experts approach which is especially effective for missing values in the modalities. Furthermore, it overcomes limitations seen in previous VAE-based integration methods with regard to batch effect correction and restricted applicable assays. It handles multiple batch effects independently accepting both discrete and continuous values, as well as provides varied reconstruction loss functions to cover all possible assays and preprocessing pipelines. We demonstrate that scMaui achieves superior performance in many tasks compared to other methods. Further downstream analyses also demonstrate its potential in identifying relations between assays and discovering hidden subpopulations.


Subject(s)
Deep Learning , Single-Cell Analysis , Humans , Multiomics/methods , Single-Cell Analysis/methods
5.
J Proteome Res ; 23(8): 3149-3160, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-38415376

ABSTRACT

Human induced pluripotent stem cells (iPSCs) can be differentiated into neurons, providing living human neurons to model brain diseases. However, it is unclear how different types of molecules work together to regulate stem cell and neuron biology in healthy and disease states. In this study, we conducted integrated proteomics, lipidomics, and metabolomics analyses with confident identification, accurate quantification, and reproducible measurements to compare the molecular profiles of human iPSCs and iPSC-derived neurons. Proteins, lipids, and metabolites related to mitosis, DNA replication, pluripotency, glycosphingolipids, and energy metabolism were highly enriched in iPSCs, whereas synaptic proteins, neurotransmitters, polyunsaturated fatty acids, cardiolipins, and axon guidance pathways were highly enriched in neurons. Mutations in the GRN gene lead to the deficiency of the progranulin (PGRN) protein, which has been associated with various neurodegenerative diseases. Using this multiomics platform, we evaluated the impact of PGRN deficiency on iPSCs and neurons at the whole-cell level. Proteomics, lipidomics, and metabolomics analyses implicated PGRN's roles in neuroinflammation, purine metabolism, and neurite outgrowth, revealing commonly altered pathways related to neuron projection, synaptic dysfunction, and brain metabolism. Multiomics data sets also pointed toward the same hypothesis that neurons seem to be more susceptible to PGRN loss compared to iPSCs, consistent with the neurological symptoms and cognitive impairment from patients carrying inherited GRN mutations.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Multiomics , Neurons , Progranulins , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lipidomics/methods , Metabolomics/methods , Multiomics/methods , Neurons/metabolism , Progranulins/genetics , Progranulins/metabolism , Proteomics/methods
6.
J Proteome Res ; 23(8): 3332-3341, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-38967328

ABSTRACT

The prevalence of different metabolic syndromes has grown globally, and the farnesoid X receptor (FXR), a metabolic homeostat for glucose, lipid, and bile acid metabolisms, may serve an important role in the progression of metabolic disorders. Glucose intolerance by FXR deficiency was previously reported and observed in our study, but the underlying biology remained unclear. To investigate the ambiguity, we collected the nontargeted profiles of the fecal metaproteome, serum metabolome, and liver proteome in Fxr-null (Fxr-/-) and wild-type (WT) mice with LC-HRMS. FXR deficiency showed a global impact on the different molecular levels we monitored, suggesting its serious disruption in the gut microbiota, hepatic metabolism, and circulating biomolecules. The network and enrichment analyses of the dysregulated metabolites and proteins suggested the perturbation of carbohydrate and lipid metabolism by FXR deficiency. Fxr-/- mice presented lower levels of hepatic proteins involved in glycogenesis. The impairment of glycogenesis by an FXR deficiency may leave glucose to accumulate in the circulation, which may deteriorate glucose tolerance. Lipid metabolism was dysregulated by FXR deficiency in a structural-dependent manner. Fatty acid ß-oxidations were alleviated, but cholesterol metabolism was promoted by an FXR deficiency. Together, we explored the molecular events associated with glucose intolerance by impaired FXR with integrated novel multiomic data.


Subject(s)
Glucose Intolerance , Lipid Metabolism , Liver , Mice, Knockout , Multiomics , Receptors, Cytoplasmic and Nuclear , Animals , Male , Mice , Feces/chemistry , Gastrointestinal Microbiome , Glucose/metabolism , Glucose Intolerance/metabolism , Glucose Intolerance/blood , Glucose Intolerance/genetics , Lipid Metabolism/genetics , Liver/metabolism , Metabolome , Multiomics/methods , Proteome/metabolism , Proteomics/methods , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/deficiency
7.
Mol Cancer ; 23(1): 173, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39175001

ABSTRACT

BACKGROUND: Early detection of colorectal cancer (CRC) significantly enhances patient outcomes. Conventional CRC screening tools, like endoscopy and stool-based tests, have constraints due to their invasiveness or suboptimal patient adherence. Recently, liquid biopsy employing plasma cell-free DNA (cfDNA) has emerged as a potential noninvasive screening technique for various malignancies. METHODS: In this research, we harnessed the Mutation Capsule Plus (MCP) technology to profile an array of genomic characteristics from cfDNA procured from a single blood draw. This profiling encompassed DNA methylation, the 5' end motif, copy number variation (CNV), and genetic mutations. An integrated model built upon selected multiomics biomarkers was trained using a cohort of 93 CRC patients and 96 healthy controls. RESULTS: This model was subsequently validated in another cohort comprising 89 CRC patients and 95 healthy controls. Remarkably, the model achieved an area under the curve (AUC) of 0.981 (95% confidence interval (CI), 0.965-0.998) in the validation set, boasting a sensitivity of 92.1% (95% CI, 84.5%-96.8%) and a specificity of 94.7% (95% CI, 88.1%-98.3%). These numbers surpassed the performance of any single genomic feature. Importantly, the sensitivities reached 80% for stage I, 89.2% for stage II, and were 100% for stages III and IV. CONCLUSION: Our findings underscore the clinical potential of our multiomics liquid biopsy test, indicating its prospective role as a noninvasive method for early-stage CRC detection. This multiomics approach holds promise for further refinement and broader clinical application.


Subject(s)
Biomarkers, Tumor , Colorectal Neoplasms , DNA Methylation , Early Detection of Cancer , Multiomics , Adult , Aged , Female , Humans , Male , Middle Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/blood , Case-Control Studies , Cell-Free Nucleic Acids/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/blood , DNA Copy Number Variations , Early Detection of Cancer/methods , Genomics/methods , Liquid Biopsy/methods , Multiomics/methods , Mutation
8.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 252-259, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39097872

ABSTRACT

One of the main causes of cancer-related mortality for women worldwide is breast cancer (BC). The XRCC2 gene, essential for DNA repair, has been implicated in cancer susceptibility. This study aims to evaluate the association between XRCC2 and BC risk. The study was conducted at Zheen International Hospital in Erbil, Iraq, between 2021 and 2024 with a total of 88 samples, including 44 paired normal and cancer tissue samples. Mutation analysis was performed using Next-Generation Sequencing, coupled with in silico tools for variant impact prediction. Expression levels were assessed through RT-PCR, and methylation status was determined using methylation-sensitive restriction enzyme digestion PCR. The study identified seven inherited germline variants in the XRCC2 gene, with five of these mutations being Uncertain Significance, one being Likely Pathogenic, and one being Likely benign. RNA purity was found high with mean A260/280 ratios of 1.986 ± 0.097 in normal (N) and 1.963 ± 0.092 in tumor (T) samples. Tumor samples exhibited a higher RNA concentration (78.56 ± 40.87 ng/µL) than normal samples (71.44 ± 40.79 ng/µL). XRCC2 gene expression was significantly upregulated in tumor tissue, with marked increases in patients aged 40-55 and >56 years and in higher cancer grades (II and III) and invasive ductal carcinoma (p-values ranging from <0.0001 to 0.0392). DNA methylation rates in tumor tissues were low (7%), suggesting limited regulation by methylation. The study suggests that XRCC2 can be classified as an oncogene and that its structural investigation by targeted NGS and expression evaluation can be used as a potential biomarker in BC.


Subject(s)
Breast Neoplasms , DNA Methylation , DNA-Binding Proteins , Multiomics , Adult , Female , Humans , Middle Aged , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA Methylation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epigenomics/methods , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genomics/methods , Multiomics/methods , Transcriptome/genetics
9.
Nephrology (Carlton) ; 29(9): 565-578, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38637907

ABSTRACT

AIM: Saliva can reflect an individual's physiological status or susceptibility to systemic disease. However, little attention has been given to salivary analysis in children with idiopathic nephrotic syndrome (INS). We aimed to perform a comprehensive analysis of saliva from INS children. METHODS: A total of 18 children (9 children with INS and 9 normal controls) were recruited. Saliva was collected from each INS patient in the acute and remission phases. 16S rRNA gene sequencing, widely targeted metabolomics, and 4D-DIA proteomics were performed. RESULTS: Actinobacteria and Firmicutes were significantly enriched in the pretreatment group compared with the normal control group, while Bacteroidota and Proteobacteria were significantly decreased. A total of 146 metabolites were identified as significantly different between INS children before treatment and normal controls, which covers 17 of 23 categories. KEGG enrichment analysis revealed three significantly enriched pathways, including ascorbate and aldarate metabolism, pentose and glucuronate interconversions, and terpenoid backbone biosynthesis (P < 0.05). A total of 389 differentially expressed proteins were selected between INS children before treatment and normal controls. According to the KEGG and GO enrichment analyses of the KOGs, abnormal ribosome structure and function and humoral immune disorders were the most prominent differences between INS patients and normal controls in the proteomic analysis. CONCLUSION: Oral microbiota dysbiosis may modulate the metabolic profile of saliva in children with INS. It is hypothesized that children with INS might have "abnormal ribosome structure and function" and "humoral immune disorders".


Subject(s)
Dysbiosis , Multiomics , Nephrotic Syndrome , Saliva , Child , Female , Humans , Male , Case-Control Studies , Dysbiosis/diagnosis , Dysbiosis/metabolism , Dysbiosis/microbiology , Metabolomics/methods , Multiomics/methods , Nephrotic Syndrome/microbiology , Nephrotic Syndrome/metabolism , Proteomics/methods , RNA, Ribosomal, 16S/genetics , Saliva/microbiology , Saliva/metabolism
10.
Sci Rep ; 14(1): 17996, 2024 08 03.
Article in English | MEDLINE | ID: mdl-39097651

ABSTRACT

Detection of important genes affecting lung adenocarcinoma (LUAD) is critical to finding effective therapeutic targets for this highly lethal cancer. However, many existing approaches have focused on single outcomes or phenotypic associations, which may not be as thorough as investigating molecular transcript levels within cells. In this article, we apply a novel multivariate rank-distance correlation-based gene selection procedure (MrDcGene) to LUAD multi-omics data downloaded from The Cancer Genome Atlas (TCGA). MrDcGene provides additional opportunities for detecting novel susceptibility genes as it leverages information from multiple platforms, while efficiently handling challenges such as high dimensionality, low signal-to-noise ratio, unknown distributions, and non-linear structures, etc. Notably, the MrDcGene method is able to detect two different scenarios, i.e., strong association strength with a few gene expressions and weak association strength with several gene expressions. After thoroughly exploring the association between gene expression (GE) and multiple other platforms, including reverse phase protein array (RPPA), miRNA, copy number variation (CNV) and DNA methylation (ME), we detect several novel genes that may play an important role in LUAD (ZNF133, CCDC159, YWHAZ, HNRNPR. ITPR2, PTHLH, and WIPI2). In addition, we quantitatively validate several other susceptibility genes that were reported in the literature using different methods and studies. The accuracy of the MrDcGene approach is theoretically assured and empirically demonstrated by the simulation studies.


Subject(s)
Adenocarcinoma of Lung , DNA Copy Number Variations , Lung Neoplasms , Humans , Adenocarcinoma of Lung/genetics , DNA Methylation , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genomics/methods , Lung Neoplasms/genetics , Multiomics/methods
11.
OMICS ; 28(7): 319-323, 2024 07.
Article in English | MEDLINE | ID: mdl-38841897

ABSTRACT

Systems biology and multiomics research expand the prospects of planetary health innovations. In this context, this mini-review unpacks the twin scholarships of glycomedicine and precision medicine in the current era of single-cell multiomics. A significant growth in glycan research has been observed over the past decade, unveiling and establishing co- and post-translational modifications as dynamic indicators of both pathological and physiological conditions. Systems biology technologies have enabled large-scale and high-throughput glycoprofiling and access to data-intensive biological repositories for global research. These advancements have established glycans as a pivotal third code of life, alongside nucleic acids and amino acids. However, challenges persist, particularly in the simultaneous analysis of the glycome and transcriptome in single cells owing to technical limitations. In addition, holistic views of the complex molecular interactions between glycomics and other omics types remain elusive. We underscore and call for a paradigm shift toward the exploration of integrative glycan platforms and analysis methods for single-cell multiomics research and precision medicine biomarker discovery. The integration of multiple datasets from various single-cell omics levels represents a crucial application of systems biology in understanding complex cellular processes and is essential for advancing the twin scholarships of glycomedicine and precision medicine.


Subject(s)
Glycomics , Multiomics , Precision Medicine , Single-Cell Analysis , Humans , Biomarkers/metabolism , Glycomics/methods , Multiomics/methods , Polysaccharides/metabolism , Precision Medicine/methods , Single-Cell Analysis/methods
12.
Sci Rep ; 14(1): 17477, 2024 07 30.
Article in English | MEDLINE | ID: mdl-39080329

ABSTRACT

The combination of multi-omic techniques, such as genomics, transcriptomics, proteomics, metabolomics and epigenomics, has revolutionised studies in medical research. These techniques are employed to support biomarker discovery, better understand molecular pathways and identify novel drug targets. Despite concerted efforts in integrating omic datasets, there is an absence of protocols that integrate all four biomolecules in a single extraction process. Here, we demonstrate for the first time a minimally destructive integrated protocol for the simultaneous extraction of artificially degraded DNA, proteins, lipids and metabolites from pig brain samples. We used an MTBE-based approach to separate lipids and metabolites, followed by subsequent isolation of DNA and proteins. We have validated this protocol against standalone extraction protocols and show comparable or higher yields of all four biomolecules. This integrated protocol is key to facilitating the preservation of irreplaceable samples while promoting downstream analyses and successful data integration by removing bias from univariate dataset noise and varied distribution characteristics.


Subject(s)
Multiomics , Animals , Brain/metabolism , DNA/isolation & purification , Genomics/methods , Lipids/analysis , Metabolomics/methods , Multiomics/methods , Proteins/isolation & purification , Proteins/metabolism , Proteomics/methods , Swine
13.
Nat Commun ; 15(1): 6856, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127735

ABSTRACT

The formation of complex traits is the consequence of genotype and activities at multiple molecular levels. However, connecting genotypes and these activities to complex traits remains challenging. Here, we investigate whether integrating genomic, transcriptomic, and methylomic data can improve prediction for six Arabidopsis traits. We find that transcriptome- and methylome-based models have performances comparable to those of genome-based models. However, models built for flowering time using different omics data identify different benchmark genes. Nine additional genes identified as important for flowering time from our models are experimentally validated as regulating flowering. Gene contributions to flowering time prediction are accession-dependent and distinct genes contribute to trait prediction in different genotypes. Models integrating multi-omics data perform best and reveal known and additional gene interactions, extending knowledge about existing regulatory networks underlying flowering time determination. These results demonstrate the feasibility of revealing molecular mechanisms underlying complex traits through multi-omics data integration.


Subject(s)
Arabidopsis , Flowers , Multiomics , Transcriptome , Arabidopsis/genetics , Arabidopsis/metabolism , DNA Methylation , Flowers/genetics , Flowers/growth & development , Gene Expression Regulation, Plant , Gene Regulatory Networks , Genome, Plant , Genomics/methods , Genotype , Models, Genetic , Multiomics/methods , Phenotype , Quantitative Trait Loci/genetics
14.
Comput Biol Med ; 163: 107117, 2023 09.
Article in English | MEDLINE | ID: mdl-37329617

ABSTRACT

The advance of sequencing technologies has enabled a thorough molecular characterization of the genome in human cancers. To improve patient prognosis predictions and subsequent treatment strategies, it is imperative to develop advanced computational methods to analyze large-scale, high-dimensional genomic data. However, traditional machine learning methods face a challenge in handling the high-dimensional, low-sample size problem that is shown in most genomic data sets. To address this, our group has developed geometric network analysis techniques on multi-omics data in connection with prior biological knowledge derived from protein-protein interactions (PPIs) or pathways. Geometric features obtained from the genomic network, such as Ollivier-Ricci curvature and the invariant measure of the associated Markov chain, have been shown to be predictive of survival outcomes in various cancers. In this study, we propose a novel supervised deep learning method called geometric graph neural network (GGNN) that incorporates such geometric features into deep learning for enhanced predictive power and interpretability. More specifically, we utilize a state-of-the-art graph neural network with sparse connections between the hidden layers based on known biology of the PPI network and pathway information. Geometric features along with multi-omics data are then incorporated into the corresponding layers. The proposed approach utilizes a local-global principle in such a manner that highly predictive features are selected at the front layers and fed directly to the last layer for multivariable Cox proportional-hazards regression modeling. The method was applied to multi-omics data from the CoMMpass study of multiple myeloma and ten major cancers in The Cancer Genome Atlas (TCGA). In most experiments, our method showed superior predictive performance compared to other alternative methods.


Subject(s)
Deep Learning , Multiomics , Neoplasms , Humans , Genomics , Neoplasms/mortality , Prognosis , Survival , Multiomics/methods
15.
Sci Data ; 10(1): 455, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37443183

ABSTRACT

The development of high-throughput omics technology has greatly promoted the development of biomedicine. However, the poor reproducibility of omics techniques limits their application. It is necessary to use standard reference materials of complex RNAs or proteins to test and calibrate the accuracy and reproducibility of omics workflows. The transcriptome and proteome of most cell lines shift during culturing, which limits their applicability as standard samples. In this study, we demonstrated that the human hepatocellular cell line MHCC97H has a very stable transcriptome (r = 0.983~0.997) and proteome (r = 0.966~0.988 for data-dependent acquisition, r = 0.970~0.994 for data-independent acquisition) after 9 subculturing generations, which allows this steady standard sample to be consistently produced on an industrial scale in long term. Moreover, this stability was maintained across labs and platforms. In sum, our study provides omics standard reference material and reference datasets for transcriptomic and proteomics research. This helps to further standardize the workflow and data quality of omics techniques and thus promotes the application of omics technology in precision medicine.


Subject(s)
Multiomics , Proteome , Transcriptome , Humans , Multiomics/methods , Proteome/genetics , Proteomics/methods , Reproducibility of Results
16.
PLoS One ; 18(3): e0278272, 2023.
Article in English | MEDLINE | ID: mdl-36928437

ABSTRACT

Pathways are composed of proteins forming a network to represent specific biological mechanisms and are often used to measure enrichment scores based on a list of genes in means to measure their biological activity. The pathway analysis is a de facto standard downstream analysis procedure in most genomic and transcriptomic studies. Here, we present MOPA (Multi-Omics Pathway Analysis), which is a multi-omics integrative method that scores individual pathways in a sample wise manner in terms of enriched multi-omics regulatory activity, which we refer to mES (multi-omics Enrichment Score). The mES score reflects the strength of regulatory relations between multi-omics in units of pathways. In addition, MOPA is able to measure how much each omics contribute to mES that may be used to observe what kind of omics are active in a pathway within a sample group (e.g., subtype, gender), which we refer to OCR (Omics Contribution Rate). Using nine different cancer types, 93 clinical features and three types of omics (i.e., gene expression, miRNA and methylation), MOPA was used to search for clinical features that were explainable in context of multi-omics. By evaluating the performance of MOPA, we showed that it yielded higher or at least equal performance compared to previous single and multi-omics pathway analysis tools. We find that the advantage of MOPA is the ability to explain pathways in terms of omics relation using mES and OCR. As one of the results, the TGF-beta signaling pathway was captured as an important pathway that showed distinct mES and OCR values specific to the CMS4 subtype in colon adenocarcinoma. The mES and OCR metrics suggested that the mRNA and miRNA expressions were significantly different from the other subtypes, which was concordant with previous studies. The MOPA software is available at https://github.com/jaeminjj/MOPA.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Multiomics , Humans , Colonic Neoplasms/genetics , MicroRNAs/genetics , Multiomics/methods
17.
Comput Biol Med ; 163: 107220, 2023 09.
Article in English | MEDLINE | ID: mdl-37406589

ABSTRACT

Cancer drug response prediction based on genomic information plays a crucial role in modern pharmacogenomics, enabling individualized therapy. Given the expensive and complexity of biological experiments, computational methods serve as effective tools in predicting cancer drug sensitivity. In this study, we proposed a novel method called Multi-Omics Integrated Collective Variational Autoencoders (MOICVAE), which leverages integrated omics knowledge, including genomic and transcriptomic data, to fill in missing cancer-drug associations and enhance drug sensitivity prediction. Our method employs an encoder-decoder network to learn latent feature representations from cell lines. These learned feature vectors are then fed into a collective variational autoencoder network to train an association matrix. We evaluated MOICVAE on the GDSC and CCLE benchmark datasets using 10-fold cross-validation and achieved impressive AUCs of 0.856 and 0.808, respectively, outperforming state-of-the-art methods. Furthermore, on the TCGA dataset, consisting of 25 drugs across 7 cancer types, MOICVAE exhibited an average AUC of 0.91 in predicting drug sensitivity. Additionally, significant differences were observed in survival, tumor inflammatory assessment, and tumor microenvironment between the predicted drug-sensitive and drug-resistant groups. These results are consistent with predictions made on the METABRIC dataset. Moreover, we discovered that fusing omics data based on mRNA and CNV (copy number variations) yielded superior results in drug sensitivity prediction. MOICVAE not only achieved higher accuracy in drug sensitivity prediction but also provided additional value for combining immunotherapy with chemotherapy, offering patients with more precise treatment options. The code and dataset for MOICVAE are freely available at https://github.com/wanggnoc/MOICVAE.


Subject(s)
Antineoplastic Agents , Deep Learning , Multiomics , Neoplasms , Neoplasms/drug therapy , Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Humans , Cell Line, Tumor , Gene Expression Profiling , Genomics , Multiomics/methods
SELECTION OF CITATIONS
SEARCH DETAIL