scispace - formally typeset
Search or ask a question
Author

Michael Snyder

Bio: Michael Snyder is an academic researcher from Stanford University. The author has contributed to research in topics: Genome & Exome. The author has an hindex of 5, co-authored 9 publications receiving 810 citations.

Papers
More filters
Journal ArticleDOI
28 Jul 2016-Cell
TL;DR: A view of how the somatic genome drives the cancer proteome and associations between protein and post-translational modification levels and clinical outcomes in HGSC is provided.

728 citations

Journal ArticleDOI
TL;DR: This work describes a multimodal strategy for isolating pancreatic endocrine and exocrine cells from children and adults to identify age-dependent gene expression and chromatin changes on a genomic scale and provides a unique resource to study human-specific regulators of islet cell maturation and function.

198 citations

01 Jun 2016
TL;DR: In this article, a detailed analysis of the molecular components and underlying mechanisms associated with ovarian cancer was provided, such as how different copy-number alterna-tions in the Proteome, the proteins associated with chromosomal instability, the sets of signalingpathways that diverse genome rearrangements converge on, and the ones associated with short overall survival.
Abstract: To provide a detailed analysis of the molecular com-ponents and underlying mechanisms associatedwith ovarian cancer, we performed a comprehensivemass-spectrometry-based proteomic characteriza-tion of 174 ovarian tumors previously analyzed byThe Cancer Genome Atlas (TCGA), of which 169were high-grade serous carcinomas (HGSCs). Inte-grating our proteomic measurements with thegenomic data yielded a number of insights into dis-ease, such as how different copy-number alterna-tionsinfluencetheproteome,theproteinsassociatedwith chromosomal instability, the sets of signalingpathways that diverse genome rearrangementsconverge on, and the ones most associated withshort overall survival. Specific protein acetylationsassociated with homologous recombination defi-ciency suggest a potential means for stratifying pa-tients for therapy. In addition to providing a valuableresource,thesefindingsprovideaviewofhowtheso-maticgenomedrivesthecancerproteomeandasso-ciations between protein and post-translationalmodification levels and clinical outcomes in HGSC.

160 citations

Journal ArticleDOI
TL;DR: iNetModels as mentioned in this paper is an interactive database and visualization platform of multi-Omics Biological Networks (MOBNs), which describes the associations between the clinical chemistry, anthropometric parameters, plasma proteomics, plasma metabolomics, as well as metagenomics for oral and gut microbiome obtained from the same individuals.
Abstract: It is essential to reveal the associations between various omics data for a comprehensive understanding of the altered biological process in human wellness and disease. To date, very few studies have focused on collecting and exhibiting multi-omics associations in a single database. Here, we present iNetModels, an interactive database and visualization platform of Multi-Omics Biological Networks (MOBNs). This platform describes the associations between the clinical chemistry, anthropometric parameters, plasma proteomics, plasma metabolomics, as well as metagenomics for oral and gut microbiome obtained from the same individuals. Moreover, iNetModels includes tissue- and cancer-specific Gene Co-expression Networks (GCNs) for exploring the connections between the specific genes. This platform allows the user to interactively explore a single feature's association with other omics data and customize its particular context (e.g. male/female specific). The users can also register their data for sharing and visualization of the MOBNs and GCNs. Moreover, iNetModels allows users who do not have a bioinformatics background to facilitate human wellness and disease research. iNetModels can be accessed freely at https://inetmodels.com without any limitation.

19 citations

Posted ContentDOI
14 Jan 2021-bioRxiv
TL;DR: iNetModels is presented, an interactive database and visualization platform of Multi-Omics Biological Networks (MOBNs), which describes the associations between the clinical chemistry, anthropometric parameters, plasma proteomics and metabolomics as well as metagenomics for oral and gut microbiome obtained from the same individuals.
Abstract: It is essential to reveal the associations between different omics data for a comprehensive understanding of the altered biological process in human wellness and disease. To date, very few studies have focused on collecting and exhibiting multi-omics associations in a single database. Here, we present iNetModels, an interactive database and visualization platform of Multi-Omics Biological Networks (MOBNs). This platform describes the associations between the clinical chemistry, anthropometric parameters, plasma proteomics and metabolomics as well as metagenomics for oral and gut microbiome obtained from the same individuals. Moreover, iNetModels includes tissue- and cancer-specific Gene Co-expression Networks (GCNs) for exploring the connections between the specific genes. This platform allows the user to interactively explore a single feature9s association with other omics data and customize its particular context (e.g. male/female specific). The users can also register their own data for sharing and visualization of the MOBNs and GCNs. Moreover, iNetModels allows users who do not have a bioinformatics background to facilitate human wellness and diseases research. iNetModels can be accessed freely at https://inetmodels.com without any limitation.

9 citations


Cited by
More filters
Journal ArticleDOI
TL;DR: It is demonstrated that LinkedOmics provides a unique platform for biologists and clinicians to access, analyze and compare cancer multi-omics data within and across tumor types.
Abstract: The LinkedOmics database contains multi-omics data and clinical data for 32 cancer types and a total of 11 158 patients from The Cancer Genome Atlas (TCGA) project. It is also the first multi-omics database that integrates mass spectrometry (MS)-based global proteomics data generated by the Clinical Proteomic Tumor Analysis Consortium (CPTAC) on selected TCGA tumor samples. In total, LinkedOmics has more than a billion data points. To allow comprehensive analysis of these data, we developed three analysis modules in the LinkedOmics web application. The LinkFinder module allows flexible exploration of associations between a molecular or clinical attribute of interest and all other attributes, providing the opportunity to analyze and visualize associations between billions of attribute pairs for each cancer cohort. The LinkCompare module enables easy comparison of the associations identified by LinkFinder, which is particularly useful in multi-omics and pan-cancer analyses. The LinkInterpreter module transforms identified associations into biological understanding through pathway and network analysis. Using five case studies, we demonstrate that LinkedOmics provides a unique platform for biologists and clinicians to access, analyze and compare cancer multi-omics data within and across tumor types. LinkedOmics is freely available at http://www.linkedomics.org.

1,256 citations

Journal ArticleDOI
TL;DR: The potential for combining diverse types of data and the utility of this approach in human health and disease is discussed and examples of data integration to understand, diagnose and inform treatment of diseases, including rare and common diseases as well as cancer and transplant biology.
Abstract: Advances in omics technologies - such as genomics, transcriptomics, proteomics and metabolomics - have begun to enable personalized medicine at an extraordinarily detailed molecular level. Individually, these technologies have contributed medical advances that have begun to enter clinical practice. However, each technology individually cannot capture the entire biological complexity of most human diseases. Integration of multiple technologies has emerged as an approach to provide a more comprehensive view of biology and disease. In this Review, we discuss the potential for combining diverse types of data and the utility of this approach in human health and disease. We provide examples of data integration to understand, diagnose and inform treatment of diseases, including rare and common diseases as well as cancer and transplant biology. Finally, we discuss technical and other challenges to clinical implementation of integrative omics.

589 citations

Journal ArticleDOI
TL;DR: This review 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.
Abstract: 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.

542 citations

Journal ArticleDOI
TL;DR: The authors' comprehensive analysis of alternative splicing across 32 The Cancer Genome Atlas cancer types from 8,705 patients detectsAlternative splicing events and tumor variants by reanalyzing RNA and whole-exome sequencing data.

529 citations

Journal ArticleDOI
11 Mar 2020-Nature
TL;DR: Microbial nucleic acids are detected in samples of tissues and blood from more than 10,000 patients with cancer, and machine learning is used to show that these can be used to discriminate between and among different types of cancer, suggesting a new microbiome-based diagnostic approach.
Abstract: Systematic characterization of the cancer microbiome provides the opportunity to develop techniques that exploit non-human, microorganism-derived molecules in the diagnosis of a major human disease. Following recent demonstrations that some types of cancer show substantial microbial contributions1–10, we re-examined whole-genome and whole-transcriptome sequencing studies in The Cancer Genome Atlas11 (TCGA) of 33 types of cancer from treatment-naive patients (a total of 18,116 samples) for microbial reads, and found unique microbial signatures in tissue and blood within and between most major types of cancer. These TCGA blood signatures remained predictive when applied to patients with stage Ia–IIc cancer and cancers lacking any genomic alterations currently measured on two commercial-grade cell-free tumour DNA platforms, despite the use of very stringent decontamination analyses that discarded up to 92.3% of total sequence data. In addition, we could discriminate among samples from healthy, cancer-free individuals (n = 69) and those from patients with multiple types of cancer (prostate, lung, and melanoma; 100 samples in total) solely using plasma-derived, cell-free microbial nucleic acids. This potential microbiome-based oncology diagnostic tool warrants further exploration. Microbial nucleic acids are detected in samples of tissues and blood from more than 10,000 patients with cancer, and machine learning is used to show that these can be used to discriminate between and among different types of cancer, suggesting a new microbiome-based diagnostic approach.

524 citations