Щоб переглянути інші типи публікацій з цієї теми, перейдіть за посиланням: Multi-omics Integration.

Статті в журналах з теми "Multi-omics Integration"

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся з топ-50 статей у журналах для дослідження на тему "Multi-omics Integration".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Переглядайте статті в журналах для різних дисциплін та оформлюйте правильно вашу бібліографію.

1

Sathyanarayanan, Anita, Rohit Gupta, Erik W. Thompson, Dale R. Nyholt, Denis C. Bauer, and Shivashankar H. Nagaraj. "A comparative study of multi-omics integration tools for cancer driver gene identification and tumour subtyping." Briefings in Bioinformatics 21, no. 6 (November 27, 2019): 1920–36. http://dx.doi.org/10.1093/bib/bbz121.

Повний текст джерела
Анотація:
Abstract Oncogenesis and cancer can arise as a consequence of a wide range of genomic aberrations including mutations, copy number alterations, expression changes and epigenetic modifications encompassing multiple omics layers. Integrating genomic, transcriptomic, proteomic and epigenomic datasets via multi-omics analysis provides the opportunity to derive a deeper and holistic understanding of the development and progression of cancer. There are two primary approaches to integrating multi-omics data: multi-staged (focused on identifying genes driving cancer) and meta-dimensional (focused on establishing clinically relevant tumour or sample classifications). A number of ready-to-use bioinformatics tools are available to perform both multi-staged and meta-dimensional integration of multi-omics data. In this study, we compared nine different integration tools using real and simulated cancer datasets. The performance of the multi-staged integration tools were assessed at the gene, function and pathway levels, while meta-dimensional integration tools were assessed based on the sample classification performance. Additionally, we discuss the influence of factors such as data representation, sample size, signal and noise on multi-omics data integration. Our results provide current and much needed guidance regarding selection and use of the most appropriate and best performing multi-omics integration tools.
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Li, Chuan-Xing, Craig E. Wheelock, C. Magnus Sköld, and Åsa M. Wheelock. "Integration of multi-omics datasets enables molecular classification of COPD." European Respiratory Journal 51, no. 5 (March 15, 2018): 1701930. http://dx.doi.org/10.1183/13993003.01930-2017.

Повний текст джерела
Анотація:
Chronic obstructive pulmonary disease (COPD) is an umbrella diagnosis caused by a multitude of underlying mechanisms, and molecular sub-phenotyping is needed to develop molecular diagnostic/prognostic tools and efficacious treatments.The objective of these studies was to investigate whether multi-omics integration improves the accuracy of molecular classification of COPD in small cohorts.Nine omics data blocks (comprising mRNA, micro RNA, proteomes and metabolomes) collected from several anatomical locations from 52 female subjects were integrated by similarity network fusion (SNF). Multi-omics integration significantly improved the accuracy of group classification of COPD patients from healthy never-smokers and from smokers with normal spirometry, reducing required group sizes from n=30 to n=6 at 95% power. Seven different combinations of four to seven omics platforms achieved >95% accuracy.For the first time, a quantitative relationship between multi-omics data integration and accuracy of data-driven classification power has been demonstrated across nine omics data blocks. Integrating five to seven omics data blocks enabled 100% correct classification of COPD diagnosis with groups as small as n=6 individuals, despite strong confounding effects of current smoking. These results can serve as guidelines for the design of future systems-based multi-omics investigations, with indications that integrating five to six data blocks from several molecular levels and anatomical locations suffices to facilitate unsupervised molecular classification in small cohorts.
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Wu, Cen, Fei Zhou, Jie Ren, Xiaoxi Li, Yu Jiang, and Shuangge Ma. "A Selective Review of Multi-Level Omics Data Integration Using Variable Selection." High-Throughput 8, no. 1 (January 18, 2019): 4. http://dx.doi.org/10.3390/ht8010004.

Повний текст джерела
Анотація:
High-throughput technologies have been used to generate a large amount of omics data. In the past, single-level analysis has been extensively conducted where the omics measurements at different levels, including mRNA, microRNA, CNV and DNA methylation, are analyzed separately. As the molecular complexity of disease etiology exists at all different levels, integrative analysis offers an effective way to borrow strength across multi-level omics data and can be more powerful than single level analysis. In this article, we focus on reviewing existing multi-omics integration studies by paying special attention to variable selection methods. We first summarize published reviews on integrating multi-level omics data. Next, after a brief overview on variable selection methods, we review existing supervised, semi-supervised and unsupervised integrative analyses within parallel and hierarchical integration studies, respectively. The strength and limitations of the methods are discussed in detail. No existing integration method can dominate the rest. The computation aspects are also investigated. The review concludes with possible limitations and future directions for multi-level omics data integration.
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Bodein, Antoine, Marie-Pier Scott-Boyer, Olivier Perin, Kim-Anh Lê Cao, and Arnaud Droit. "Interpretation of network-based integration from multi-omics longitudinal data." Nucleic Acids Research 50, no. 5 (December 9, 2021): e27-e27. http://dx.doi.org/10.1093/nar/gkab1200.

Повний текст джерела
Анотація:
Abstract Multi-omics integration is key to fully understand complex biological processes in an holistic manner. Furthermore, multi-omics combined with new longitudinal experimental design can unreveal dynamic relationships between omics layers and identify key players or interactions in system development or complex phenotypes. However, integration methods have to address various experimental designs and do not guarantee interpretable biological results. The new challenge of multi-omics integration is to solve interpretation and unlock the hidden knowledge within the multi-omics data. In this paper, we go beyond integration and propose a generic approach to face the interpretation problem. From multi-omics longitudinal data, this approach builds and explores hybrid multi-omics networks composed of both inferred and known relationships within and between omics layers. With smart node labelling and propagation analysis, this approach predicts regulation mechanisms and multi-omics functional modules. We applied the method on 3 case studies with various multi-omics designs and identified new multi-layer interactions involved in key biological functions that could not be revealed with single omics analysis. Moreover, we highlighted interplay in the kinetics that could help identify novel biological mechanisms. This method is available as an R package netOmics to readily suit any application.
Стилі APA, Harvard, Vancouver, ISO та ін.
5

Wieder, Cecilia, Juliette Cooke, Clement Frainay, Nathalie Poupin, Russell Bowler, Fabien Jourdan, Katerina J. Kechris, Rachel PJ Lai, and Timothy Ebbels. "PathIntegrate: Multivariate modelling approaches for pathway-based multi-omics data integration." PLOS Computational Biology 20, no. 3 (March 25, 2024): e1011814. http://dx.doi.org/10.1371/journal.pcbi.1011814.

Повний текст джерела
Анотація:
As terabytes of multi-omics data are being generated, there is an ever-increasing need for methods facilitating the integration and interpretation of such data. Current multi-omics integration methods typically output lists, clusters, or subnetworks of molecules related to an outcome. Even with expert domain knowledge, discerning the biological processes involved is a time-consuming activity. Here we propose PathIntegrate, a method for integrating multi-omics datasets based on pathways, designed to exploit knowledge of biological systems and thus provide interpretable models for such studies. PathIntegrate employs single-sample pathway analysis to transform multi-omics datasets from the molecular to the pathway-level, and applies a predictive single-view or multi-view model to integrate the data. Model outputs include multi-omics pathways ranked by their contribution to the outcome prediction, the contribution of each omics layer, and the importance of each molecule in a pathway. Using semi-synthetic data we demonstrate the benefit of grouping molecules into pathways to detect signals in low signal-to-noise scenarios, as well as the ability of PathIntegrate to precisely identify important pathways at low effect sizes. Finally, using COPD and COVID-19 data we showcase how PathIntegrate enables convenient integration and interpretation of complex high-dimensional multi-omics datasets. PathIntegrate is available as an open-source Python package.
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Colomé-Tatché, M., and F. J. Theis. "Statistical single cell multi-omics integration." Current Opinion in Systems Biology 7 (February 2018): 54–59. http://dx.doi.org/10.1016/j.coisb.2018.01.003.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
7

Subramanian, Indhupriya, Srikant Verma, Shiva Kumar, Abhay Jere, and Krishanpal Anamika. "Multi-omics Data Integration, Interpretation, and Its Application." Bioinformatics and Biology Insights 14 (January 2020): 117793221989905. http://dx.doi.org/10.1177/1177932219899051.

Повний текст джерела
Анотація:
To study complex biological processes holistically, it is imperative to take an integrative approach that combines multi-omics data to highlight the interrelationships of the involved biomolecules and their functions. With the advent of high-throughput techniques and availability of multi-omics data generated from a large set of samples, several promising tools and methods have been developed for data integration and interpretation. In this review, we collected the tools and methods that adopt integrative approach to analyze multiple omics data and summarized their ability to address applications such as disease subtyping, biomarker prediction, and deriving insights into the data. We provide the methodology, use-cases, and limitations of these tools; brief account of multi-omics data repositories and visualization portals; and challenges associated with multi-omics data integration.
Стилі APA, Harvard, Vancouver, ISO та ін.
8

Haidar, Siwar, Julia Hooker, Simon Lackey, Mohamad Elian, Nathalie Puchacz, Krzysztof Szczyglowski, Frédéric Marsolais, Ashkan Golshani, Elroy R. Cober, and Bahram Samanfar. "Harnessing Multi-Omics Strategies and Bioinformatics Innovations for Advancing Soybean Improvement: A Comprehensive Review." Plants 13, no. 19 (September 28, 2024): 2714. http://dx.doi.org/10.3390/plants13192714.

Повний текст джерела
Анотація:
Soybean improvement has entered a new era with the advent of multi-omics strategies and bioinformatics innovations, enabling more precise and efficient breeding practices. This comprehensive review examines the application of multi-omics approaches in soybean—encompassing genomics, transcriptomics, proteomics, metabolomics, epigenomics, and phenomics. We first explore pre-breeding and genomic selection as tools that have laid the groundwork for advanced trait improvement. Subsequently, we dig into the specific contributions of each -omics field, highlighting how bioinformatics tools and resources have facilitated the generation and integration of multifaceted data. The review emphasizes the power of integrating multi-omics datasets to elucidate complex traits and drive the development of superior soybean cultivars. Emerging trends, including novel computational techniques and high-throughput technologies, are discussed in the context of their potential to revolutionize soybean breeding. Finally, we address the challenges associated with multi-omics integration and propose future directions to overcome these hurdles, aiming to accelerate the pace of soybean improvement. This review serves as a crucial resource for researchers and breeders seeking to leverage multi-omics strategies for enhanced soybean productivity and resilience.
Стилі APA, Harvard, Vancouver, ISO та ін.
9

Fiocchi, Claudio. "Omics and Multi-Omics in IBD: No Integration, No Breakthroughs." International Journal of Molecular Sciences 24, no. 19 (October 5, 2023): 14912. http://dx.doi.org/10.3390/ijms241914912.

Повний текст джерела
Анотація:
The recent advent of sophisticated technologies like sequencing and mass spectroscopy platforms combined with artificial intelligence-powered analytic tools has initiated a new era of “big data” research in various complex diseases of still-undetermined cause and mechanisms. The investigation of these diseases was, until recently, limited to traditional in vitro and in vivo biological experimentation, but a clear switch to in silico methodologies is now under way. This review tries to provide a comprehensive assessment of state-of-the-art knowledge on omes, omics and multi-omics in inflammatory bowel disease (IBD). The notion and importance of omes, omics and multi-omics in both health and complex diseases like IBD is introduced, followed by a discussion of the various omics believed to be relevant to IBD pathogenesis, and how multi-omics “big data” can generate new insights translatable into useful clinical tools in IBD such as biomarker identification, prediction of remission and relapse, response to therapy, and precision medicine. The pitfalls and limitations of current IBD multi-omics studies are critically analyzed, revealing that, regardless of the types of omes being analyzed, the majority of current reports are still based on simple associations of descriptive retrospective data from cross-sectional patient cohorts rather than more powerful longitudinally collected prospective datasets. Given this limitation, some suggestions are provided on how IBD multi-omics data may be optimized for greater clinical and therapeutic benefit. The review concludes by forecasting the upcoming incorporation of multi-omics analyses in the routine management of IBD.
Стилі APA, Harvard, Vancouver, ISO та ін.
10

Pinu, Farhana R., David J. Beale, Amy M. Paten, Konstantinos Kouremenos, Sanjay Swarup, Horst J. Schirra, and David Wishart. "Systems Biology and Multi-Omics Integration: Viewpoints from the Metabolomics Research Community." Metabolites 9, no. 4 (April 18, 2019): 76. http://dx.doi.org/10.3390/metabo9040076.

Повний текст джерела
Анотація:
The use of multiple omics techniques (i.e., genomics, transcriptomics, proteomics, and metabolomics) is becoming increasingly popular in all facets of life science. Omics techniques provide a more holistic molecular perspective of studied biological systems compared to traditional approaches. However, due to their inherent data differences, integrating multiple omics platforms remains an ongoing challenge for many researchers. As metabolites represent the downstream products of multiple interactions between genes, transcripts, and proteins, metabolomics, the tools and approaches routinely used in this field could assist with the integration of these complex multi-omics data sets. The question is, how? Here we provide some answers (in terms of methods, software tools and databases) along with a variety of recommendations and a list of continuing challenges as identified during a peer session on multi-omics integration that was held at the recent ‘Australian and New Zealand Metabolomics Conference’ (ANZMET 2018) in Auckland, New Zealand (Sept. 2018). We envisage that this document will serve as a guide to metabolomics researchers and other members of the community wishing to perform multi-omics studies. We also believe that these ideas may allow the full promise of integrated multi-omics research and, ultimately, of systems biology to be realized.
Стилі APA, Harvard, Vancouver, ISO та ін.
11

Luo, Haoran, Hong Liang, Hongwei Liu, Zhoujie Fan, Yanhui Wei, Xiaohui Yao, and Shan Cong. "TEMINET: A Co-Informative and Trustworthy Multi-Omics Integration Network for Diagnostic Prediction." International Journal of Molecular Sciences 25, no. 3 (January 29, 2024): 1655. http://dx.doi.org/10.3390/ijms25031655.

Повний текст джерела
Анотація:
Advancing the domain of biomedical investigation, integrated multi-omics data have shown exceptional performance in elucidating complex human diseases. However, as the variety of omics information expands, precisely perceiving the informativeness of intra- and inter-omics becomes challenging due to the intricate interrelations, thus presenting significant challenges in the integration of multi-omics data. To address this, we introduce a novel multi-omics integration approach, referred to as TEMINET. This approach enhances diagnostic prediction by leveraging an intra-omics co-informative representation module and a trustworthy learning strategy used to address inter-omics fusion. Considering the multifactorial nature of complex diseases, TEMINET utilizes intra-omics features to construct disease-specific networks; then, it applies graph attention networks and a multi-level framework to capture more collective informativeness than pairwise relations. To perceive the contribution of co-informative representations within intra-omics, we designed a trustworthy learning strategy to identify the reliability of each omics in integration. To integrate inter-omics information, a combined-beliefs fusion approach is deployed to harmonize the trustworthy representations of different omics types effectively. Our experiments across four different diseases using mRNA, methylation, and miRNA data demonstrate that TEMINET achieves advanced performance and robustness in classification tasks.
Стилі APA, Harvard, Vancouver, ISO та ін.
12

Rappoport, Nimrod, and Ron Shamir. "NEMO: cancer subtyping by integration of partial multi-omic data." Bioinformatics 35, no. 18 (January 30, 2019): 3348–56. http://dx.doi.org/10.1093/bioinformatics/btz058.

Повний текст джерела
Анотація:
Abstract Motivation Cancer subtypes were usually defined based on molecular characterization of single omic data. Increasingly, measurements of multiple omic profiles for the same cohort are available. Defining cancer subtypes using multi-omic data may improve our understanding of cancer, and suggest more precise treatment for patients. Results We present NEMO (NEighborhood based Multi-Omics clustering), a novel algorithm for multi-omics clustering. Importantly, NEMO can be applied to partial datasets in which some patients have data for only a subset of the omics, without performing data imputation. In extensive testing on ten cancer datasets spanning 3168 patients, NEMO achieved results comparable to the best of nine state-of-the-art multi-omics clustering algorithms on full data and showed an improvement on partial data. On some of the partial data tests, PVC, a multi-view algorithm, performed better, but it is limited to two omics and to positive partial data. Finally, we demonstrate the advantage of NEMO in detailed analysis of partial data of AML patients. NEMO is fast and much simpler than existing multi-omics clustering algorithms, and avoids iterative optimization. Availability and implementation Code for NEMO and for reproducing all NEMO results in this paper is in github: https://github.com/Shamir-Lab/NEMO. Supplementary information Supplementary data are available at Bioinformatics online.
Стилі APA, Harvard, Vancouver, ISO та ін.
13

Choi, Joung Min, Chaelin Park, and Heejoon Chae. "moSCminer: a cell subtype classification framework based on the attention neural network integrating the single-cell multi-omics dataset on the cloud." PeerJ 12 (February 26, 2024): e17006. http://dx.doi.org/10.7717/peerj.17006.

Повний текст джерела
Анотація:
Single-cell omics sequencing has rapidly advanced, enabling the quantification of diverse omics profiles at a single-cell resolution. To facilitate comprehensive biological insights, such as cellular differentiation trajectories, precise annotation of cell subtypes is essential. Conventional methods involve clustering cells and manually assigning subtypes based on canonical markers, a labor-intensive and expert-dependent process. Hence, an automated computational prediction framework is crucial. While several classification frameworks for predicting cell subtypes from single-cell RNA sequencing datasets exist, these methods solely rely on single-omics data, offering insights at a single molecular level. They often miss inter-omic correlations and a holistic understanding of cellular processes. To address this, the integration of multi-omics datasets from individual cells is essential for accurate subtype annotation. This article introduces moSCminer, a novel framework for classifying cell subtypes that harnesses the power of single-cell multi-omics sequencing datasets through an attention-based neural network operating at the omics level. By integrating three distinct omics datasets—gene expression, DNA methylation, and DNA accessibility—while accounting for their biological relationships, moSCminer excels at learning the relative significance of each omics feature. It then transforms this knowledge into a novel representation for cell subtype classification. Comparative evaluations against standard machine learning-based classifiers demonstrate moSCminer’s superior performance, consistently achieving the highest average performance on real datasets. The efficacy of multi-omics integration is further corroborated through an in-depth analysis of the omics-level attention module, which identifies potential markers for cell subtype annotation. To enhance accessibility and scalability, moSCminer is accessible as a user-friendly web-based platform seamlessly connected to a cloud system, publicly accessible at http://203.252.206.118:5568. Notably, this study marks the pioneering integration of three single-cell multi-omics datasets for cell subtype identification.
Стилі APA, Harvard, Vancouver, ISO та ін.
14

Cembrowska-Lech, Danuta, Adrianna Krzemińska, Tymoteusz Miller, Anna Nowakowska, Cezary Adamski, Martyna Radaczyńska, Grzegorz Mikiciuk, and Małgorzata Mikiciuk. "An Integrated Multi-Omics and Artificial Intelligence Framework for Advance Plant Phenotyping in Horticulture." Biology 12, no. 10 (September 30, 2023): 1298. http://dx.doi.org/10.3390/biology12101298.

Повний текст джерела
Анотація:
This review discusses the transformative potential of integrating multi-omics data and artificial intelligence (AI) in advancing horticultural research, specifically plant phenotyping. The traditional methods of plant phenotyping, while valuable, are limited in their ability to capture the complexity of plant biology. The advent of (meta-)genomics, (meta-)transcriptomics, proteomics, and metabolomics has provided an opportunity for a more comprehensive analysis. AI and machine learning (ML) techniques can effectively handle the complexity and volume of multi-omics data, providing meaningful interpretations and predictions. Reflecting the multidisciplinary nature of this area of research, in this review, readers will find a collection of state-of-the-art solutions that are key to the integration of multi-omics data and AI for phenotyping experiments in horticulture, including experimental design considerations with several technical and non-technical challenges, which are discussed along with potential solutions. The future prospects of this integration include precision horticulture, predictive breeding, improved disease and stress response management, sustainable crop management, and exploration of plant biodiversity. The integration of multi-omics and AI holds immense promise for revolutionizing horticultural research and applications, heralding a new era in plant phenotyping.
Стилі APA, Harvard, Vancouver, ISO та ін.
15

Zhu, Yingkun, Dengpan Bu, and Lu Ma. "Integration of Multiplied Omics, a Step Forward in Systematic Dairy Research." Metabolites 12, no. 3 (March 4, 2022): 225. http://dx.doi.org/10.3390/metabo12030225.

Повний текст джерела
Анотація:
Due to their unique multi-gastric digestion system highly adapted for rumination, dairy livestock has complicated physiology different from monogastric animals. However, the microbiome-based mechanism of the digestion system is congenial for biology approaches. Different omics and their integration have been widely applied in the dairy sciences since the previous decade for investigating their physiology, pathology, and the development of feed and management protocols. The rumen microbiome can digest dietary components into utilizable sugars, proteins, and volatile fatty acids, contributing to the energy intake and feed efficiency of dairy animals, which has become one target of the basis for omics applications in dairy science. Rumen, liver, and mammary gland are also frequently targeted in omics because of their crucial impact on dairy animals’ energy metabolism, production performance, and health status. The application of omics has made outstanding contributions to a more profound understanding of the physiology, etiology, and optimizing the management strategy of dairy animals, while the multi-omics method could draw information of different levels and organs together, providing an unprecedented broad scope on traits of dairy animals. This article reviewed recent omics and multi-omics researches on physiology, feeding, and pathology on dairy animals and also performed the potential of multi-omics on systematic dairy research.
Стилі APA, Harvard, Vancouver, ISO та ін.
16

Hachem, Sana, Amani Yehya, Jad El Masri, Nicole Mavingire, Jabril R. Johnson, Abdulrahman M. Dwead, Naim Kattour, et al. "Contemporary Update on Clinical and Experimental Prostate Cancer Biomarkers: A Multi-Omics-Focused Approach to Detection and Risk Stratification." Biology 13, no. 10 (September 25, 2024): 762. http://dx.doi.org/10.3390/biology13100762.

Повний текст джерела
Анотація:
Prostate cancer remains a significant health challenge, being the most prevalent non-cutaneous cancer in men worldwide. This review discusses the critical advancements in biomarker discovery using single-omics and multi-omics approaches. Multi-omics, integrating genomic, transcriptomic, proteomic, metabolomic, and epigenomic data, offers a comprehensive understanding of the molecular heterogeneity of prostate cancer, leading to the identification of novel biomarkers and therapeutic targets. This holistic approach not only enhances the specificity and sensitivity of prostate cancer detection but also supports the development of personalized treatment strategies. Key studies highlighted include the identification of novel genes, genetic mutations, peptides, metabolites, and potential biomarkers through multi-omics analyses, which have shown promise in improving prostate cancer management. The integration of multi-omics in clinical practice can potentially revolutionize prostate cancer prognosis and treatment, paving the way for precision medicine. This review underscores the importance of continued research and the application of multi-omics to overcome current challenges in prostate cancer diagnosis and therapy.
Стилі APA, Harvard, Vancouver, ISO та ін.
17

Khorraminezhad, Leila, Mickael Leclercq, Arnaud Droit, Jean-François Bilodeau, and Iwona Rudkowska. "Statistical and Machine-Learning Analyses in Nutritional Genomics Studies." Nutrients 12, no. 10 (October 14, 2020): 3140. http://dx.doi.org/10.3390/nu12103140.

Повний текст джерела
Анотація:
Nutritional compounds may have an influence on different OMICs levels, including genomics, epigenomics, transcriptomics, proteomics, metabolomics, and metagenomics. The integration of OMICs data is challenging but may provide new knowledge to explain the mechanisms involved in the metabolism of nutrients and diseases. Traditional statistical analyses play an important role in description and data association; however, these statistical procedures are not sufficiently enough powered to interpret the large integrated multiple OMICs (multi-OMICS) datasets. Machine learning (ML) approaches can play a major role in the interpretation of multi-OMICS in nutrition research. Specifically, ML can be used for data mining, sample clustering, and classification to produce predictive models and algorithms for integration of multi-OMICs in response to dietary intake. The objective of this review was to investigate the strategies used for the analysis of multi-OMICs data in nutrition studies. Sixteen recent studies aimed to understand the association between dietary intake and multi-OMICs data are summarized. Multivariate analysis in multi-OMICs nutrition studies is used more commonly for analyses. Overall, as nutrition research incorporated multi-OMICs data, the use of novel approaches of analysis such as ML needs to complement the traditional statistical analyses to fully explain the impact of nutrition on health and disease.
Стилі APA, Harvard, Vancouver, ISO та ін.
18

Cao, Kai, Xiangqi Bai, Yiguang Hong, and Lin Wan. "Unsupervised topological alignment for single-cell multi-omics integration." Bioinformatics 36, Supplement_1 (July 1, 2020): i48—i56. http://dx.doi.org/10.1093/bioinformatics/btaa443.

Повний текст джерела
Анотація:
Abstract Motivation Single-cell multi-omics data provide a comprehensive molecular view of cells. However, single-cell multi-omics datasets consist of unpaired cells measured with distinct unmatched features across modalities, making data integration challenging. Results In this study, we present a novel algorithm, termed UnionCom, for the unsupervised topological alignment of single-cell multi-omics integration. UnionCom does not require any correspondence information, either among cells or among features. It first embeds the intrinsic low-dimensional structure of each single-cell dataset into a distance matrix of cells within the same dataset and then aligns the cells across single-cell multi-omics datasets by matching the distance matrices via a matrix optimization method. Finally, it projects the distinct unmatched features across single-cell datasets into a common embedding space for feature comparability of the aligned cells. To match the complex non-linear geometrical distorted low-dimensional structures across datasets, UnionCom proposes and adjusts a global scaling parameter on distance matrices for aligning similar topological structures. It does not require one-to-one correspondence among cells across datasets, and it can accommodate samples with dataset-specific cell types. UnionCom outperforms state-of-the-art methods on both simulated and real single-cell multi-omics datasets. UnionCom is robust to parameter choices, as well as subsampling of features. Availability and implementation UnionCom software is available at https://github.com/caokai1073/UnionCom. Supplementary information Supplementary data are available at Bioinformatics online.
Стилі APA, Harvard, Vancouver, ISO та ін.
19

Eicher, Tara, Garrett Kinnebrew, Andrew Patt, Kyle Spencer, Kevin Ying, Qin Ma, Raghu Machiraju, and Ewy A. Mathé. "Metabolomics and Multi-Omics Integration: A Survey of Computational Methods and Resources." Metabolites 10, no. 5 (May 15, 2020): 202. http://dx.doi.org/10.3390/metabo10050202.

Повний текст джерела
Анотація:
As researchers are increasingly able to collect data on a large scale from multiple clinical and omics modalities, multi-omics integration is becoming a critical component of metabolomics research. This introduces a need for increased understanding by the metabolomics researcher of computational and statistical analysis methods relevant to multi-omics studies. In this review, we discuss common types of analyses performed in multi-omics studies and the computational and statistical methods that can be used for each type of analysis. We pinpoint the caveats and considerations for analysis methods, including required parameters, sample size and data distribution requirements, sources of a priori knowledge, and techniques for the evaluation of model accuracy. Finally, for the types of analyses discussed, we provide examples of the applications of corresponding methods to clinical and basic research. We intend that our review may be used as a guide for metabolomics researchers to choose effective techniques for multi-omics analyses relevant to their field of study.
Стилі APA, Harvard, Vancouver, ISO та ін.
20

Et al., Gonesh Chandra Saha. "Integrative Analysis of Multi-Omics Data with Deep Learning: Challenges and Opportunities in Bioinformatics." Tuijin Jishu/Journal of Propulsion Technology 44, no. 3 (September 11, 2023): 1384–92. http://dx.doi.org/10.52783/tjjpt.v44.i3.488.

Повний текст джерела
Анотація:
The advent of high-throughput technologies has ushered in an era of unprecedented data generation in the field of bioinformatics. Omics data, including genomics, transcriptomics, proteomics, and metabolomics, provide comprehensive insights into biological systems, but their integration poses significant challenges. Integrative analysis of multi-omics data holds the promise of unraveling complex biological phenomena and enabling personalized medicine. [1] Deep learning, a subset of machine learning, has gained prominence in bioinformatics due to its ability to automatically extract intricate patterns from large-scale multi-omics datasets. This paper presents an overview of the challenges and opportunities associated with the integrative analysis of multi-omics data using deep learning techniques in bioinformatics.The challenges in multi-omics integration primarily stem from data heterogeneity, dimensionality, and noise. One of the key opportunities presented by deep learning is its ability to capture complex, non-linear relationships in multi-omics data. The paper emphasizes the importance of interpretability and explainability in deep learning models applied to bioinformatics, as they play a crucial role in gaining biological insights and facilitating clinical decision-making. The integration of domain knowledge and biological context is highlighted as a critical aspect of model development. The paper showcases real-world applications of deep learning in multi-omics data integration, such as disease subtype classification, biomarker discovery, and drug response prediction. As the field continues to evolve, addressing these challenges and harnessing the potential of deep learning approaches will pave the way for transformative advancements in our understanding of complex biological systems and the development of precision medicine strategies.
Стилі APA, Harvard, Vancouver, ISO та ін.
21

ElKarami, Bashier, Abedalrhman Alkhateeb, Hazem Qattous, Lujain Alshomali, and Behnam Shahrrava. "Multi-omics Data Integration Model Based on UMAP Embedding and Convolutional Neural Network." Cancer Informatics 21 (January 2022): 117693512211242. http://dx.doi.org/10.1177/11769351221124205.

Повний текст джерела
Анотація:
Introduction: Multi-omics data integration facilitates collecting richer understanding and perceptions than separate omics data. Various promising integrative approaches have been utilized to analyze multi-omics data for biomedical applications, including disease prediction and disease subtypes, biomarker prediction, and others. Methods: In this paper, we introduce a multi-omics data integration method that is constructed using the combination of gene similarity network (GSN) based on uniform manifold approximation and projection (UMAP) and convolutional neural networks (CNNs). The method utilizes UMAP to embed gene expression, DNA methylation, and copy number alteration (CNA) to a lower dimension creating two-dimensional RGB images. Gene expression is used as a reference to construct the GSN and then integrate other omics data with the gene expression for better prediction. We used CNNs to predict the Gleason score levels of prostate cancer patients and the tumor stage in breast cancer patients. Results: The model proposed near perfection with accuracy above 99% with all other performance measurements at the same level. The proposed model outperformed the state-of-art iSOM-GSN model that constructs the GSN map based on the self-organizing map. Conclusion: The results show that UMAP as an embedding technique can better integrate multi-omics maps into the prediction model than SOM. The proposed model can also be applied to build a multi-omics prediction model for other types of cancer.
Стилі APA, Harvard, Vancouver, ISO та ін.
22

Sharifi-Noghabi, Hossein, Olga Zolotareva, Colin C. Collins, and Martin Ester. "MOLI: multi-omics late integration with deep neural networks for drug response prediction." Bioinformatics 35, no. 14 (July 2019): i501—i509. http://dx.doi.org/10.1093/bioinformatics/btz318.

Повний текст джерела
Анотація:
Abstract Motivation Historically, gene expression has been shown to be the most informative data for drug response prediction. Recent evidence suggests that integrating additional omics can improve the prediction accuracy which raises the question of how to integrate the additional omics. Regardless of the integration strategy, clinical utility and translatability are crucial. Thus, we reasoned a multi-omics approach combined with clinical datasets would improve drug response prediction and clinical relevance. Results We propose MOLI, a multi-omics late integration method based on deep neural networks. MOLI takes somatic mutation, copy number aberration and gene expression data as input, and integrates them for drug response prediction. MOLI uses type-specific encoding sub-networks to learn features for each omics type, concatenates them into one representation and optimizes this representation via a combined cost function consisting of a triplet loss and a binary cross-entropy loss. The former makes the representations of responder samples more similar to each other and different from the non-responders, and the latter makes this representation predictive of the response values. We validate MOLI on in vitro and in vivo datasets for five chemotherapy agents and two targeted therapeutics. Compared to state-of-the-art single-omics and early integration multi-omics methods, MOLI achieves higher prediction accuracy in external validations. Moreover, a significant improvement in MOLI’s performance is observed for targeted drugs when training on a pan-drug input, i.e. using all the drugs with the same target compared to training only on drug-specific inputs. MOLI’s high predictive power suggests it may have utility in precision oncology. Availability and implementation https://github.com/hosseinshn/MOLI. Supplementary information Supplementary data are available at Bioinformatics online.
Стилі APA, Harvard, Vancouver, ISO та ін.
23

Molla, Getnet, and Molalegne Bitew. "Revolutionizing Personalized Medicine: Synergy with Multi-Omics Data Generation, Main Hurdles, and Future Perspectives." Biomedicines 12, no. 12 (November 30, 2024): 2750. https://doi.org/10.3390/biomedicines12122750.

Повний текст джерела
Анотація:
The field of personalized medicine is undergoing a transformative shift through the integration of multi-omics data, which mainly encompasses genomics, transcriptomics, proteomics, and metabolomics. This synergy allows for a comprehensive understanding of individual health by analyzing genetic, molecular, and biochemical profiles. The generation and integration of multi-omics data enable more precise and tailored therapeutic strategies, improving the efficacy of treatments and reducing adverse effects. However, several challenges hinder the full realization of personalized medicine. Key hurdles include the complexity of data integration across different omics layers, the need for advanced computational tools, and the high cost of comprehensive data generation. Additionally, issues related to data privacy, standardization, and the need for robust validation in diverse populations remain significant obstacles. Looking ahead, the future of personalized medicine promises advancements in technology and methodologies that will address these challenges. Emerging innovations in data analytics, machine learning, and high-throughput sequencing are expected to enhance the integration of multi-omics data, making personalized medicine more accessible and effective. Collaborative efforts among researchers, clinicians, and industry stakeholders are crucial to overcoming these hurdles and fully harnessing the potential of multi-omics for individualized healthcare.
Стилі APA, Harvard, Vancouver, ISO та ін.
24

Moon, Sehwan, and Hyunju Lee. "JDSNMF: Joint Deep Semi-Non-Negative Matrix Factorization for Learning Integrative Representation of Molecular Signals in Alzheimer’s Disease." Journal of Personalized Medicine 11, no. 8 (July 21, 2021): 686. http://dx.doi.org/10.3390/jpm11080686.

Повний текст джерела
Анотація:
High dimensional multi-omics data integration can enhance our understanding of the complex biological interactions in human diseases. However, most studies involving unsupervised integration of multi-omics data focus on linear integration methods. In this study, we propose a joint deep semi-non-negative matrix factorization (JDSNMF) model, which uses a hierarchical non-linear feature extraction approach that can capture shared latent features from the complex multi-omics data. The extracted latent features obtained from JDSNMF enabled a variety of downstream tasks, including prediction of disease and module analysis. The proposed model is applicable not only to sample-matched multiple data (e.g., multi-omics data from one cohort) but also to feature-matched multiple data (e.g., omics data from multiple cohorts), and therefore it can be flexibly applied to various cases. We demonstrate the capabilities of JDSNMF using sample-matched simulated data and feature-matched multi-omics data from Alzheimer’s disease cohorts, evaluating the feature extraction performance in the context of classification. In a test application, we identify AD- and age-related modules from the latent matrices using an explainable artificial intelligence and regression model. These results show that the JDSNMF model is effective in identifying latent features having a complex interplay of potential biological signatures.
Стилі APA, Harvard, Vancouver, ISO та ін.
25

Zarayeneh, Neda, Euiseong Ko, Jung Hun Oh, Sang Suh, Chunyu Liu, Jean Gao, Donghyun Kim, and Mingon Kang. "Integration of multi-omics data for integrative gene regulatory network inference." International Journal of Data Mining and Bioinformatics 18, no. 3 (2017): 223. http://dx.doi.org/10.1504/ijdmb.2017.087178.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
26

Kang, Mingon, Donghyun Kim, Jean Gao, Chunyu Liu, Sang Suh, Jung Hun Oh, Neda Zarayeneh, and Euiseong Ko. "Integration of multi-omics data for integrative gene regulatory network inference." International Journal of Data Mining and Bioinformatics 18, no. 3 (2017): 223. http://dx.doi.org/10.1504/ijdmb.2017.10008266.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
27

Milner, Danny A., and Jochen K. Lennerz. "Technology and Future of Multi-Cancer Early Detection." Life 14, no. 7 (June 29, 2024): 833. http://dx.doi.org/10.3390/life14070833.

Повний текст джерела
Анотація:
Cancer remains a significant global health challenge due to its high morbidity and mortality rates. Early detection is essential for improving patient outcomes, yet current diagnostic methods lack the sensitivity and specificity needed for identifying early-stage cancers. Here, we explore the potential of multi-omics approaches, which integrate genomic, transcriptomic, proteomic, and metabolomic data, to enhance early cancer detection. We highlight the challenges and benefits of data integration from these diverse sources and discuss successful examples of multi-omics applications in other fields. By leveraging these advanced technologies, multi-omics can significantly improve the sensitivity and specificity of early cancer diagnostics, leading to better patient outcomes and more personalized cancer care. We underscore the transformative potential of multi-omics approaches in revolutionizing early cancer detection and the need for continued research and clinical integration.
Стилі APA, Harvard, Vancouver, ISO та ін.
28

Bakare, Olayemi Stephen. "AI-Driven Multi-Omics Integration for Precision Medicine in Complex Disease Diagnosis and Treatment." International Journal of Research Publication and Reviews 6, no. 6 (January 2025): 5070–84. https://doi.org/10.55248/gengpi.6.0125.0650.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
29

Ivanisevic, Tonci, and Raj N. Sewduth. "Multi-Omics Integration for the Design of Novel Therapies and the Identification of Novel Biomarkers." Proteomes 11, no. 4 (October 20, 2023): 34. http://dx.doi.org/10.3390/proteomes11040034.

Повний текст джерела
Анотація:
Multi-omics is a cutting-edge approach that combines data from different biomolecular levels, such as DNA, RNA, proteins, metabolites, and epigenetic marks, to obtain a holistic view of how living systems work and interact. Multi-omics has been used for various purposes in biomedical research, such as identifying new diseases, discovering new drugs, personalizing treatments, and optimizing therapies. This review summarizes the latest progress and challenges of multi-omics for designing new treatments for human diseases, focusing on how to integrate and analyze multiple proteome data and examples of how to use multi-proteomics data to identify new drug targets. We also discussed the future directions and opportunities of multi-omics for developing innovative and effective therapies by deciphering proteome complexity.
Стилі APA, Harvard, Vancouver, ISO та ін.
30

Adossa, Nigatu, Sofia Khan, Kalle T. Rytkönen, and Laura L. Elo. "Computational strategies for single-cell multi-omics integration." Computational and Structural Biotechnology Journal 19 (2021): 2588–96. http://dx.doi.org/10.1016/j.csbj.2021.04.060.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
31

Charmpi, Konstantina, Manopriya Chokkalingam, Ronja Johnen, and Andreas Beyer. "Optimizing network propagation for multi-omics data integration." PLOS Computational Biology 17, no. 11 (November 11, 2021): e1009161. http://dx.doi.org/10.1371/journal.pcbi.1009161.

Повний текст джерела
Анотація:
Network propagation refers to a class of algorithms that integrate information from input data across connected nodes in a given network. These algorithms have wide applications in systems biology, protein function prediction, inferring condition-specifically altered sub-networks, and prioritizing disease genes. Despite the popularity of network propagation, there is a lack of comparative analyses of different algorithms on real data and little guidance on how to select and parameterize the various algorithms. Here, we address this problem by analyzing different combinations of network normalization and propagation methods and by demonstrating schemes for the identification of optimal parameter settings on real proteome and transcriptome data. Our work highlights the risk of a ‘topology bias’ caused by the incorrect use of network normalization approaches. Capitalizing on the fact that network propagation is a regularization approach, we show that minimizing the bias-variance tradeoff can be utilized for selecting optimal parameters. The application to real multi-omics data demonstrated that optimal parameters could also be obtained by either maximizing the agreement between different omics layers (e.g. proteome and transcriptome) or by maximizing the consistency between biological replicates. Furthermore, we exemplified the utility and robustness of network propagation on multi-omics datasets for identifying ageing-associated genes in brain and liver tissues of rats and for elucidating molecular mechanisms underlying prostate cancer progression. Overall, this work compares different network propagation approaches and it presents strategies for how to use network propagation algorithms to optimally address a specific research question at hand.
Стилі APA, Harvard, Vancouver, ISO та ін.
32

Mohr, Alex E., Carmen P. Ortega-Santos, Corrie M. Whisner, Judith Klein-Seetharaman, and Paniz Jasbi. "Navigating Challenges and Opportunities in Multi-Omics Integration for Personalized Healthcare." Biomedicines 12, no. 7 (July 5, 2024): 1496. http://dx.doi.org/10.3390/biomedicines12071496.

Повний текст джерела
Анотація:
The field of multi-omics has witnessed unprecedented growth, converging multiple scientific disciplines and technological advances. This surge is evidenced by a more than doubling in multi-omics scientific publications within just two years (2022–2023) since its first referenced mention in 2002, as indexed by the National Library of Medicine. This emerging field has demonstrated its capability to provide comprehensive insights into complex biological systems, representing a transformative force in health diagnostics and therapeutic strategies. However, several challenges are evident when merging varied omics data sets and methodologies, interpreting vast data dimensions, streamlining longitudinal sampling and analysis, and addressing the ethical implications of managing sensitive health information. This review evaluates these challenges while spotlighting pivotal milestones: the development of targeted sampling methods, the use of artificial intelligence in formulating health indices, the integration of sophisticated n-of-1 statistical models such as digital twins, and the incorporation of blockchain technology for heightened data security. For multi-omics to truly revolutionize healthcare, it demands rigorous validation, tangible real-world applications, and smooth integration into existing healthcare infrastructures. It is imperative to address ethical dilemmas, paving the way for the realization of a future steered by omics-informed personalized medicine.
Стилі APA, Harvard, Vancouver, ISO та ін.
33

Andrew, Zhou, Zhang Charlie, and Eminaga Okyaz. "Advances in deep learning-based cancer outcome prediction using multi-omics data." Annals of Proteomics and Bioinformatics 7, no. 1 (May 1, 2023): 010–13. http://dx.doi.org/10.29328/journal.apb.1001020.

Повний текст джерела
Анотація:
Cancer prognosis reflects a complex biological process measured by multiple types of omics data. Deep learning frameworks have been proposed to integrate multi-omics data and predict patient outcomes in different cancer types, potentially revolutionizing cancer prognosis with superior performance. This minireview summarizes the advances in the strategies for multi-omics data integration and the performance of different deep learning models in prognosis prediction of diverse cancer types using multi-omics data published in the past 18 months. The challenges and limitations of deep learning models for predicting cancer outcomes based on multi-omics data are discussed.
Стилі APA, Harvard, Vancouver, ISO та ін.
34

Li, Peng, and Bo Sun. "Integration of Multi-Omics Data to Identify Cancer Biomarkers." Journal of Information Technology Research 15, no. 1 (January 2022): 1–15. http://dx.doi.org/10.4018/jitr.2022010105.

Повний текст джерела
Анотація:
A novel method for integrating multi-omics data, including gene expression, copy number variation, DNA methylation, and miRNA data, is proposed to identify biomarkers of cancer prognosis. First, survival analysis was performed for these four types of omics data to obtain survival-related genes. Next, survival-related genes detected in at least two types of omics data were selected as candidate genes. The four types of omics data only composed of candidate genes were subjected to dimension reduction using an autoencoder to obtain a one-dimensional data representation. The mRMR algorithm was used to screen for key genes. This method was applied to lung squamous cell carcinoma and 20 cancer-related genes were identified. Gene function analysis revealed that the genes were related to cancer. Using survival analysis, the genes were verified to distinguish between high- and low-risk groups. These results indicate that the genes can be used as biomarkers for cancer.
Стилі APA, Harvard, Vancouver, ISO та ін.
35

Chen, Tyrone, Al J. Abadi, Kim-Anh Lê Cao, and Sonika Tyagi. "multiomics: A user-friendly multi-omics data harmonisation R pipeline." F1000Research 10 (July 6, 2021): 538. http://dx.doi.org/10.12688/f1000research.53453.1.

Повний текст джерела
Анотація:
Data from multiple omics layers of a biological system is growing in quantity, heterogeneity and dimensionality. Simultaneous multi-omics data integration is a growing field of research as it has strong potential to unlock information on previously hidden biological relationships leading to early diagnosis, prognosis and expedited treatments. Many tools for multi-omics data integration are being developed. However, these tools are often restricted to highly specific experimental designs, and types of omics data. While some general methods do exist, they require specific data formats and experimental conditions. A major limitation in the field is a lack of a single or multi-omics pipeline which can accept data in an unrefined, information-rich form pre-integration and subsequently generate output for further investigation. There is an increasing demand for a generic multi-omics pipeline to facilitate general-purpose data exploration and analysis of heterogeneous data. Therefore, we present our R multiomics pipeline as an easy to use and flexible pipeline that takes unrefined multi-omics data as input, sample information and user-specified parameters to generate a list of output plots and data tables for quality control and downstream analysis. We have demonstrated application of the pipeline on two separate COVID-19 case studies. We enabled limited checkpointing where intermediate output is staged to allow continuation after errors or interruptions in the pipeline and generate a script for reproducing the analysis to improve reproducibility. A seamless integration with the mixOmics R package is achieved, as the R data object can be loaded and manipulated with mixOmics functions. Our pipeline can be installed as an R package or from the git repository, and is accompanied by detailed documentation with walkthroughs on two case studies. The pipeline is also available as Docker and Singularity containers.
Стилі APA, Harvard, Vancouver, ISO та ін.
36

Chen, Tyrone, Al J. Abadi, Kim-Anh Lê Cao, and Sonika Tyagi. "multiomics: A user-friendly multi-omics data harmonisation R pipeline." F1000Research 10 (August 2, 2023): 538. http://dx.doi.org/10.12688/f1000research.53453.2.

Повний текст джерела
Анотація:
Data from multiple omics layers of a biological system is growing in quantity, heterogeneity and dimensionality. Simultaneous multi-omics data integration is of immense interest to researchers as it has potential to unlock previously hidden biomolecular relationships leading to early diagnosis, prognosis, and expedited treatments. Many tools for multi-omics data integration are developed. However, these tools are often restricted to highly specific experimental designs, types of omics data, and specific data formats. A major limitation of the field is the lack of a pipeline that can accept data in unrefined form to preserve maximum biology in an individual dataset prior to integration. We fill this gap by developing a flexible, generic multi-omics pipeline called multiomics, to facilitate general-purpose data exploration and analysis of heterogeneous data. The pipeline takes unrefined multi-omics data as input, sample information and user-specified parameters to generate a list of output plots and data tables for quality control and downstream analysis. We have demonstrated its application on a sepsis case study. We enabled limited checkpointing functionality where intermediate output is staged to allow continuation after errors or interruptions in the pipeline and generate a script for reproducing the analysis to improve reproducibility. Our pipeline can be installed as an R package or manually from the git repository, and is accompanied by detailed documentation with walkthroughs on three case studies.
Стилі APA, Harvard, Vancouver, ISO та ін.
37

Duan, Ran, Lin Gao, Yong Gao, Yuxuan Hu, Han Xu, Mingfeng Huang, Kuo Song, et al. "Evaluation and comparison of multi-omics data integration methods for cancer subtyping." PLOS Computational Biology 17, no. 8 (August 12, 2021): e1009224. http://dx.doi.org/10.1371/journal.pcbi.1009224.

Повний текст джерела
Анотація:
Computational integrative analysis has become a significant approach in the data-driven exploration of biological problems. Many integration methods for cancer subtyping have been proposed, but evaluating these methods has become a complicated problem due to the lack of gold standards. Moreover, questions of practical importance remain to be addressed regarding the impact of selecting appropriate data types and combinations on the performance of integrative studies. Here, we constructed three classes of benchmarking datasets of nine cancers in TCGA by considering all the eleven combinations of four multi-omics data types. Using these datasets, we conducted a comprehensive evaluation of ten representative integration methods for cancer subtyping in terms of accuracy measured by combining both clustering accuracy and clinical significance, robustness, and computational efficiency. We subsequently investigated the influence of different omics data on cancer subtyping and the effectiveness of their combinations. Refuting the widely held intuition that incorporating more types of omics data always produces better results, our analyses showed that there are situations where integrating more omics data negatively impacts the performance of integration methods. Our analyses also suggested several effective combinations for most cancers under our studies, which may be of particular interest to researchers in omics data analysis.
Стилі APA, Harvard, Vancouver, ISO та ін.
38

Sun, Wenli, and Mohamad Hesam Shahrajabian. "Survey on Multi-omics, and Multi-omics Data Analysis, Integration and Application." Current Pharmaceutical Analysis 19 (April 6, 2023). http://dx.doi.org/10.2174/1573412919666230406100948.

Повний текст джерела
Анотація:
Abstract: Multi-omics approaches have developed as a profitable technique for plant systems, a popular method in medical and biological sciences underlining the necessity to outline new integrative technology and functions to facilitate the multi-scale depiction of biological systems. Understanding a biological system through various omics layers reveals supplementary sources of variability and probably inferring the sequence of cases leading to a definitive process. Manuscripts and reviews were searched on PubMed with the keywords of multi-omics, data analysis, omics, data analysis, data integration, deep learning multi-omics, and multi-omics integration. Articles that were published after 2010 were prioritized. The authors focused mainly on popular publications developing new approaches. Omics reveal interesting tools to produce behavioral and interactions data in microbial communities, and integrating omics details into microbial risk assessment will have an impact on food safety, and also on relevant spoilage control procedures. Omics datasets, comprehensively characterizing biological cases at a molecular level, are continually increasing in both dimensionality and complexity. Multi-omics data analysis is appropriate for treatment optimization, molecular testing and disease prognosis, and to achieve mechanistic understandings of diseases. New effective solutions for multi-omics data analysis together with well-designed components are recommended for many trials. The goal of this mini-review article is to introduce multi-omics technologies considering different multi-omics analyses.
Стилі APA, Harvard, Vancouver, ISO та ін.
39

Li, Yunfan, Dan Zhang, Mouxing Yang, Dezhong Peng, Jun Yu, Yu Liu, Jiancheng Lv, Lu Chen, and Xi Peng. "scBridge embraces cell heterogeneity in single-cell RNA-seq and ATAC-seq data integration." Nature Communications 14, no. 1 (September 28, 2023). http://dx.doi.org/10.1038/s41467-023-41795-5.

Повний текст джерела
Анотація:
AbstractSingle-cell multi-omics data integration aims to reduce the omics difference while keeping the cell type difference. However, it is daunting to model and distinguish the two differences due to cell heterogeneity. Namely, even cells of the same omics and type would have various features, making the two differences less significant. In this work, we reveal that instead of being an interference, cell heterogeneity could be exploited to improve data integration. Specifically, we observe that the omics difference varies in cells, and cells with smaller omics differences are easier to be integrated. Hence, unlike most existing works that homogeneously treat and integrate all cells, we propose a multi-omics data integration method (dubbed scBridge) that integrates cells in a heterogeneous manner. In brief, scBridge iterates between i) identifying reliable scATAC-seq cells that have smaller omics differences, and ii) integrating reliable scATAC-seq cells with scRNA-seq data to narrow the omics gap, thus benefiting the integration for the rest cells. Extensive experiments on seven multi-omics datasets demonstrate the superiority of scBridge compared with six representative baselines.
Стилі APA, Harvard, Vancouver, ISO та ін.
40

Liu, Zhe, and Taesung Park. "DMOIT: denoised multi-omics integration approach based on transformer multi-head self-attention mechanism." Frontiers in Genetics 15 (December 10, 2024). https://doi.org/10.3389/fgene.2024.1488683.

Повний текст джерела
Анотація:
Multi-omics data integration has become increasingly crucial for a deeper understanding of the complexity of biological systems. However, effectively integrating and analyzing multi-omics data remains challenging due to their heterogeneity and high dimensionality. Existing methods often struggle with noise, redundant features, and the complex interactions between different omics layers, leading to suboptimal performance. Additionally, they face difficulties in adequately capturing intra-omics interactions due to simplistic concatenation techiniques, and they risk losing critical inter-omics interaction information when using hierarchical attention layers. To address these challenges, we propose a novel Denoised Multi-Omics Integration approach that leverages the Transformer multi-head self-attention mechanism (DMOIT). DMOIT consists of three key modules: a generative adversarial imputation network for handling missing values, a sampling-based robust feature selection module to reduce noise and redundant features, and a multi-head self-attention (MHSA) based feature extractor with a noval architecture that enchance the intra-omics interaction capture. We validated model porformance using cancer datasets from the Cancer Genome Atlas (TCGA), conducting two tasks: survival time classification across different cancer types and estrogen receptor status classification for breast cancer. Our results show that DMOIT outperforms traditional machine learning methods and the state-of-the-art integration method MoGCN in terms of accuracy and weighted F1 score. Furthermore, we compared DMOIT with various alternative MHSA-based architectures to further validate our approach. Our results show that DMOIT consistently outperforms these models across various cancer types and different omics combinations. The strong performance and robustness of DMOIT demonstrate its potential as a valuable tool for integrating multi-omics data across various applications.
Стилі APA, Harvard, Vancouver, ISO та ін.
41

Wang, Haohua, Kai Lin, Qiang Zhang, Jinlong Shi, Xinyu Song, Jue Wu, Chenghui Zhao, and Kunlun He. "HyperTMO: a trusted multi-omics integration framework based on hypergraph convolutional network for patient classification." Bioinformatics, March 26, 2024. http://dx.doi.org/10.1093/bioinformatics/btae159.

Повний текст джерела
Анотація:
Abstract Motivation The rapid development of high-throughput biomedical technologies can provide researchers with detailed multi-omics data. The multi-omics integrated analysis approach based on machine learning contributes a more comprehensive perspective to human disease research. However, there are still significant challenges in representing single-omics data and integrating multi-omics information. Results This paper presents HyperTMO, a Trusted Multi-Omics integration framework based on Hypergraph convolutional network for patient classification. HyperTMO constructs hypergraph structures to represent the association between samples in single-omics data, then evidence extraction is performed by hypergraph convolutional network, and multi-omics information is integrated at an evidence level. Lastly, we experimentally demonstrate that HyperTMO outperforms other state-of-the-art methods in breast cancer subtype classification and Alzheimer’s disease classification tasks using multi-omics data from TCGA (BRCA) and ROSMAP datasets. Importantly, HyperTMO is the first attempt to integrate hypergraph structure, evidence theory, and multi-omics integration for patient classification. Its accurate and robust properties bring great potential for applications in clinical diagnosis. Availability HyperTMO and datasets are publicly available at https://github.com/ippousyuga/HyperTMO. Supplementary Information Supplementary data are available at Bioinformatics online.
Стилі APA, Harvard, Vancouver, ISO та ін.
42

Kwoji, Iliya Dauda, Olayinka Ayobami Aiyegoro, Moses Okpeku, and Matthew Adekunle Adeleke. "‘Multi-omics’ data integration: applications in probiotics studies." npj Science of Food 7, no. 1 (June 5, 2023). http://dx.doi.org/10.1038/s41538-023-00199-x.

Повний текст джерела
Анотація:
AbstractThe concept of probiotics is witnessing increasing attention due to its benefits in influencing the host microbiome and the modulation of host immunity through the strengthening of the gut barrier and stimulation of antibodies. These benefits, combined with the need for improved nutraceuticals, have resulted in the extensive characterization of probiotics leading to an outburst of data generated using several ‘omics’ technologies. The recent development in system biology approaches to microbial science is paving the way for integrating data generated from different omics techniques for understanding the flow of molecular information from one ‘omics’ level to the other with clear information on regulatory features and phenotypes. The limitations and tendencies of a ‘single omics’ application to ignore the influence of other molecular processes justify the need for ‘multi-omics’ application in probiotics selections and understanding its action on the host. Different omics techniques, including genomics, transcriptomics, proteomics, metabolomics and lipidomics, used for studying probiotics and their influence on the host and the microbiome are discussed in this review. Furthermore, the rationale for ‘multi-omics’ and multi-omics data integration platforms supporting probiotics and microbiome analyses was also elucidated. This review showed that multi-omics application is useful in selecting probiotics and understanding their functions on the host microbiome. Hence, recommend a multi-omics approach for holistically understanding probiotics and the microbiome.
Стилі APA, Harvard, Vancouver, ISO та ін.
43

Tiwari, Prateek, Raghvendra Pandey, and Sonia Chadha. "Integrative Multi-Omics Approaches for Personalized Medicine and Health." Current Bioinformatics 20 (February 10, 2025). https://doi.org/10.2174/0115748936360644250127095005.

Повний текст джерела
Анотація:
Introduction: Multi-omics data integration has transformed personalized medicine, providing a comprehensive understanding of disease mechanisms and informed precision therapeutic options. Multi-omics data generated for the same samples/patients can help in getting insights into the flow of biological information at several levels, thereby providing in-depth information regarding the molecular mechanisms underlying pathological conditions. Multi-omics integration plays a pivotal role in personalized medicine by providing comprehensive insights into the complex biological systems of individual patients. This review provides a comprehensive account of the current and future progress brought into multi-omics methodologies, promising to refine diagnostics and therapeutic strategy by integrating genomic, transcriptomic analyses, proteomics approaches and metabolome screens. Methods: A literature search was performed in PubMed using keywords like genomics, proteomics, transcriptomics, metabolomics, multi-omics, and precision medicine to identify published research articles. A thorough review of all results was then conducted, and their results and conclusions were compiled and summarized. Result: By analyzing various omics layers, such as genomics, transcriptomics, proteomics, and metabolomics, multi-omics approaches enable the identification of patient-specific molecular traits and the discovery of new clinical therapeutics for diseases. Integration of various data types augments diagnostics, optimizes therapeutic regimens and supports personalized medicine according to an individual patient profile. Conclusion: Integration of multi-omics data and its applications in various fields, such as cancer research, helps in optimizing patient-specific treatment and improvement of patient health. With time, as these technologies reach more people, they stand to democratize precision medicine and hopefully bridge health disparities. In conclusion, the present review highlights multiomics data integration as a transformative step towards personalized medicine and ultimately changing patient care from empirical-based to precision or individualized.
Стилі APA, Harvard, Vancouver, ISO та ін.
44

Kang, Mingon, Euiseong Ko, and Tesfaye B. Mersha. "A roadmap for multi-omics data integration using deep learning." Briefings in Bioinformatics 23, no. 1 (November 12, 2021). http://dx.doi.org/10.1093/bib/bbab454.

Повний текст джерела
Анотація:
Abstract High-throughput next-generation sequencing now makes it possible to generate a vast amount of multi-omics data for various applications. These data have revolutionized biomedical research by providing a more comprehensive understanding of the biological systems and molecular mechanisms of disease development. Recently, deep learning (DL) algorithms have become one of the most promising methods in multi-omics data analysis, due to their predictive performance and capability of capturing nonlinear and hierarchical features. While integrating and translating multi-omics data into useful functional insights remain the biggest bottleneck, there is a clear trend towards incorporating multi-omics analysis in biomedical research to help explain the complex relationships between molecular layers. Multi-omics data have a role to improve prevention, early detection and prediction; monitor progression; interpret patterns and endotyping; and design personalized treatments. In this review, we outline a roadmap of multi-omics integration using DL and offer a practical perspective into the advantages, challenges and barriers to the implementation of DL in multi-omics data.
Стилі APA, Harvard, Vancouver, ISO та ін.
45

Selvarajoo, Kumar, and Sebastian Maurer-Stroh. "Towards multi-omics synthetic data integration." Briefings in Bioinformatics 25, no. 3 (March 27, 2024). http://dx.doi.org/10.1093/bib/bbae213.

Повний текст джерела
Анотація:
Abstract Across many scientific disciplines, the development of computational models and algorithms for generating artificial or synthetic data is gaining momentum. In biology, there is a great opportunity to explore this further as more and more big data at multi-omics level are generated recently. In this opinion, we discuss the latest trends in biological applications based on process-driven and data-driven aspects. Moving ahead, we believe these methodologies can help shape novel multi-omics-scale cellular inferences.
Стилі APA, Harvard, Vancouver, ISO та ін.
46

Pang, Jiali, Bilin Liang, Ruifeng Ding, Qiujuan Yan, Ruiyao Chen, and Jie Xu. "A denoised multi-omics integration framework for cancer subtype classification and survival prediction." Briefings in Bioinformatics, August 18, 2023. http://dx.doi.org/10.1093/bib/bbad304.

Повний текст джерела
Анотація:
Abstract The availability of high-throughput sequencing data creates opportunities to comprehensively understand human diseases as well as challenges to train machine learning models using such high dimensions of data. Here, we propose a denoised multi-omics integration framework, which contains a distribution-based feature denoising algorithm, Feature Selection with Distribution (FSD), for dimension reduction and a multi-omics integration framework, Attention Multi-Omics Integration (AttentionMOI) to predict cancer prognosis and identify cancer subtypes. We demonstrated that FSD improved model performance either using single omic data or multi-omics data in 15 The Cancer Genome Atlas Program (TCGA) cancers for survival prediction and kidney cancer subtype identification. And our integration framework AttentionMOI outperformed machine learning models and current multi-omics integration algorithms with high dimensions of features. Furthermore, FSD identified features that were associated to cancer prognosis and could be considered as biomarkers.
Стилі APA, Harvard, Vancouver, ISO та ін.
47

Tsagiopoulou, Maria, Nikolaos Pechlivanis, Maria Christina Maniou, and Fotis Psomopoulos. "InterTADs: integration of multi-omics data on topologically associated domains, application to chronic lymphocytic leukemia." NAR Genomics and Bioinformatics 4, no. 1 (January 14, 2022). http://dx.doi.org/10.1093/nargab/lqab121.

Повний текст джерела
Анотація:
ABSTRACT The integration of multi-omics data can greatly facilitate the advancement of research in Life Sciences by highlighting new interactions. However, there is currently no widespread procedure for meaningful multi-omics data integration. Here, we present a robust framework, called InterTADs, for integrating multi-omics data derived from the same sample, and considering the chromatin configuration of the genome, i.e. the topologically associating domains (TADs). Following the integration process, statistical analysis highlights the differences between the groups of interest (normal versus cancer cells) relating to (i) independent and (ii) integrated events through TADs. Finally, enrichment analysis using KEGG database, Gene Ontology and transcription factor binding sites and visualization approaches are available. We applied InterTADs to multi-omics datasets from 135 patients with chronic lymphocytic leukemia (CLL) and found that the integration through TADs resulted in a dramatic reduction of heterogeneity compared to individual events. Significant differences for individual events and on TADs level were identified between patients differing in the somatic hypermutation status of the clonotypic immunoglobulin genes, the core biological stratifier in CLL, attesting to the biomedical relevance of InterTADs. In conclusion, our approach suggests a new perspective towards analyzing multi-omics data, by offering reasonable execution time, biological benchmarking and potentially contributing to pattern discovery through TADs.
Стилі APA, Harvard, Vancouver, ISO та ін.
48

Maitra, Chayan, Dibyendu B. Seal, Vivek Das, and Rajat K. De. "Unsupervised neural network for single cell Multi-omics INTegration (UMINT): an application to health and disease." Frontiers in Molecular Biosciences 10 (May 24, 2023). http://dx.doi.org/10.3389/fmolb.2023.1184748.

Повний текст джерела
Анотація:
Multi-omics studies have enabled us to understand the mechanistic drivers behind complex disease states and progressions, thereby providing novel and actionable biological insights into health status. However, integrating data from multiple modalities is challenging due to high dimensionality and diverse nature of data, and noise associated with each platform. Sparsity in data, non-overlapping features and technical batch effects make the task of learning more complicated. Conventional machine learning (ML) tools are not quite effective against such data integration hazards due to their simplistic nature with less capacity. In addition, existing methods for single cell multi-omics integration are computationally expensive. Therefore, in this work, we have introduced a novel Unsupervised neural network for single cell Multi-omics INTegration (UMINT). UMINT serves as a promising model for integrating variable number of single cell omics layers with high dimensions. It has a light-weight architecture with substantially reduced number of parameters. The proposed model is capable of learning a latent low-dimensional embedding that can extract useful features from the data facilitating further downstream analyses. UMINT has been applied to integrate healthy and disease CITE-seq (paired RNA and surface proteins) datasets including a rare disease Mucosa-Associated Lymphoid Tissue (MALT) tumor. It has been benchmarked against existing state-of-the-art methods for single cell multi-omics integration. Furthermore, UMINT is capable of integrating paired single cell gene expression and ATAC-seq (Transposase-Accessible Chromatin) assays as well.
Стилі APA, Harvard, Vancouver, ISO та ін.
49

Canzler, Sebastian, Kristin Schubert, Ulrike E. Rolle-Kampczyk, Zhipeng Wang, Stephan Schreiber, Hervé Seitz, Sophie Mockly, et al. "Evaluating the performance of multi-omics integration: a thyroid toxicity case study." Archives of Toxicology, October 23, 2024. http://dx.doi.org/10.1007/s00204-024-03876-2.

Повний текст джерела
Анотація:
AbstractMulti-omics data integration has been repeatedly discussed as the way forward to more comprehensively cover the molecular responses of cells or organisms to chemical exposure in systems toxicology and regulatory risk assessment. In Canzler et al. (Arch Toxicol 94(2):371–388. https://doi.org/10.1007/s00204-020-02656-y), we reviewed the state of the art in applying multi-omics approaches in toxicological research and chemical risk assessment. We developed best practices for the experimental design of multi-omics studies, omics data acquisition, and subsequent omics data integration. We found that multi-omics data sets for toxicological research questions were generally rare, with no data sets comprising more than two omics layers adhering to these best practices. Due to these limitations, we could not fully assess the benefits of different data integration approaches or quantitatively evaluate the contribution of various omics layers for toxicological research questions. Here, we report on a multi-omics study on thyroid toxicity that we conducted in compliance with these best practices. We induced direct and indirect thyroid toxicity through Propylthiouracil (PTU) and Phenytoin, respectively, in a 28-day plus 14-day recovery oral rat toxicity study. We collected clinical and histopathological data and six omics layers, including the long and short transcriptome, proteome, phosphoproteome, and metabolome from plasma, thyroid, and liver. We demonstrate that the multi-omics approach is superior to single-omics in detecting responses at the regulatory pathway level. We also show how combining omics data with clinical and histopathological parameters facilitates the interpretation of the data. Furthermore, we illustrate how multi-omics integration can hint at the involvement of non-coding RNAs in post-transcriptional regulation. Also, we show that multi-omics facilitates grouping, and we assess how much information individual and combinations of omics layers contribute to this approach.
Стилі APA, Harvard, Vancouver, ISO та ін.
50

Xu, Jing, Peng Wu, Yuehui Chen, Qingfang Meng, Hussain Dawood, and Hassan Dawood. "A hierarchical integration deep flexible neural forest framework for cancer subtype classification by integrating multi-omics data." BMC Bioinformatics 20, no. 1 (October 28, 2019). http://dx.doi.org/10.1186/s12859-019-3116-7.

Повний текст джерела
Анотація:
Abstract Background Cancer subtype classification attains the great importance for accurate diagnosis and personalized treatment of cancer. Latest developments in high-throughput sequencing technologies have rapidly produced multi-omics data of the same cancer sample. Many computational methods have been proposed to classify cancer subtypes, however most of them generate the model by only employing gene expression data. It has been shown that integration of multi-omics data contributes to cancer subtype classification. Results A new hierarchical integration deep flexible neural forest framework is proposed to integrate multi-omics data for cancer subtype classification named as HI-DFNForest. Stacked autoencoder (SAE) is used to learn high-level representations in each omics data, then the complex representations are learned by integrating all learned representations into a layer of autoencoder. Final learned data representations (from the stacked autoencoder) are used to classify patients into different cancer subtypes using deep flexible neural forest (DFNForest) model.Cancer subtype classification is verified on BRCA, GBM and OV data sets from TCGA by integrating gene expression, miRNA expression and DNA methylation data. These results demonstrated that integrating multiple omics data improves the accuracy of cancer subtype classification than only using gene expression data and the proposed framework has achieved better performance compared with other conventional methods. Conclusion The new hierarchical integration deep flexible neural forest framework(HI-DFNForest) is an effective method to integrate multi-omics data to classify cancer subtypes.
Стилі APA, Harvard, Vancouver, ISO та ін.
Ми пропонуємо знижки на всі преміум-плани для авторів, чиї праці увійшли до тематичних добірок літератури. Зв'яжіться з нами, щоб отримати унікальний промокод!

До бібліографії