1,830 research outputs found

    Modeling cancer metabolism on a genome scale

    Get PDF
    Cancer cells have fundamentally altered cellular metabolism that is associated with their tumorigenicity and malignancy. In addition to the widely studied Warburg effect, several new key metabolic alterations in cancer have been established over the last decade, leading to the recognition that altered tumor metabolism is one of the hallmarks of cancer. Deciphering the full scope and functional implications of the dysregulated metabolism in cancer requires both the advancement of a variety of omics measurements and the advancement of computational approaches for the analysis and contextualization of the accumulated data. Encouragingly, while the metabolic network is highly interconnected and complex, it is at the same time probably the best characterized cellular network. Following, this review discusses the challenges that genome‐scale modeling of cancer metabolism has been facing. We survey several recent studies demonstrating the first strides that have been done, testifying to the value of this approach in portraying a network‐level view of the cancer metabolism and in identifying novel drug targets and biomarkers. Finally, we outline a few new steps that may further advance this field

    The role of network science in glioblastoma

    Get PDF
    Network science has long been recognized as a well-established discipline across many biological domains. In the particular case of cancer genomics, network discovery is challenged by the multitude of available high-dimensional heterogeneous views of data. Glioblastoma (GBM) is an example of such a complex and heterogeneous disease that can be tackled by network science. Identifying the architecture of molecular GBM networks is essential to understanding the information flow and better informing drug development and pre-clinical studies. Here, we review network-based strategies that have been used in the study of GBM, along with the available software implementations for reproducibility and further testing on newly coming datasets. Promising results have been obtained from both bulk and single-cell GBM data, placing network discovery at the forefront of developing a molecularly-informed-based personalized medicine.This work was partially supported by national funds through Fundação para a CiĂȘncia e a Tecnologia (FCT) with references CEECINST/00102/2018, CEECIND/00072/2018 and PD/BDE/143154/2019, UIDB/04516/2020, UIDB/00297/2020, UIDB/50021/2020, UIDB/50022/2020, UIDB/50026/2020, UIDP/50026/2020, NORTE-01-0145-FEDER-000013, and NORTE-01-0145-FEDER000023 and projects PTDC/CCI-BIO/4180/2020 and DSAIPA/DS/0026/2019. This project has received funding from the European Union’s Horizon 2020 research and innovation program under Grant Agreement No. 951970 (OLISSIPO project)

    INTEGRATIVE ANALYSIS OF OMICS DATA IN ADULT GLIOMA AND OTHER TCGA CANCERS TO GUIDE PRECISION MEDICINE

    Get PDF
    Transcriptomic profiling and gene expression signatures have been widely applied as effective approaches for enhancing the molecular classification, diagnosis, prognosis or prediction of therapeutic response towards personalized therapy for cancer patients. Thanks to modern genome-wide profiling technology, scientists are able to build engines leveraging massive genomic variations and integrating with clinical data to identify “at risk” individuals for the sake of prevention, diagnosis and therapeutic interventions. In my graduate work for my Ph.D. thesis, I have investigated genomic sequencing data mining to comprehensively characterise molecular classifications and aberrant genomic events associated with clinical prognosis and treatment response, through applying high-dimensional omics genomic data to promote the understanding of gene signatures and somatic molecular alterations contributing to cancer progression and clinical outcomes. Following this motivation, my dissertation has been focused on the following three topics in translational genomics. 1) Characterization of transcriptomic plasticity and its association with the tumor microenvironment in glioblastoma (GBM). I have integrated transcriptomic, genomic, protein and clinical data to increase the accuracy of GBM classification, and identify the association between the GBM mesenchymal subtype and reduced tumorpurity, accompanied with increased presence of tumor-associated microglia. Then I have tackled the sole source of microglial as intrinsic tumor bulk but not their corresponding neurosphere cells through both transcriptional and protein level analysis using a panel of sphere-forming glioma cultures and their parent GBM samples.FurthermoreI have demonstrated my hypothesis through longitudinal analysis of paired primary and recurrent GBM samples that the phenotypic alterations of GBM subtypes are not due to intrinsic proneural-to-mesenchymal transition in tumor cells, rather it is intertwined with increased level of microglia upon disease recurrence. Collectively I have elucidated the critical role of tumor microenvironment (Microglia and macrophages from central nervous system) contributing to the intra-tumor heterogeneity and accurate classification of GBM patients based on transcriptomic profiling, which will not only significantly impact on clinical perspective but also pave the way for preclinical cancer research. 2) Identification of prognostic gene signatures that stratify adult diffuse glioma patientsharboring1p/19q co-deletions. I have compared multiple statistical methods and derived a gene signature significantly associated with survival by applying a machine learning algorithm. Then I have identified inflammatory response and acetylation activity that associated with malignant progression of 1p/19q co-deleted glioma. In addition, I showed this signature translates to other types of adult diffuse glioma, suggesting its universality in the pathobiology of other subset gliomas. My efforts on integrative data analysis of this highly curated data set usingoptimizedstatistical models will reflect the pending update to WHO classification system oftumorsin the central nervous system (CNS). 3) Comprehensive characterization of somatic fusion transcripts in Pan-Cancers. I have identified a panel of novel fusion transcripts across all of TCGA cancer types through transcriptomic profiling. Then I have predicted fusion proteins with kinase activity and hub function of pathway network based on the annotation of genetically mobile domains and functional domain architectures. I have evaluated a panel of in -frame gene fusions as potential driver mutations based on network fusion centrality hypothesis. I have also characterised the emerging complexity of genetic architecture in fusion transcripts through integrating genomic structure and somatic variants and delineating the distinct genomic patterns of fusion events across different cancer types. Overall my exploration of the pathogenetic impact and clinical relevance of candidate gene fusions have provided fundamental insights into the management of a subset of cancer patients by predicting the oncogenic signalling and specific drug targets encoded by these fusion genes. Taken together, the translational genomic research I have conducted during my Ph.D. study will shed new light on precision medicine and contribute to the cancer research community. The novel classification concept, gene signature and fusion transcripts I have identified will address several hotly debated issues in translational genomics, such as complex interactions between tumor bulks and their adjacent microenvironments, prognostic markers for clinical diagnostics and personalized therapy, distinct patterns of genomic structure alterations and oncogenic events in different cancer types, therefore facilitating our understanding of genomic alterations and moving us towards the development of precision medicine

    Biologically Interpretable, Integrative Deep Learning for Cancer Survival Analysis

    Get PDF
    Identifying complex biological processes associated to patients\u27 survival time at the cellular and molecular level is critical not only for developing new treatments for patients but also for accurate survival prediction. However, highly nonlinear and high-dimension, low-sample size (HDLSS) data cause computational challenges in survival analysis. We developed a novel family of pathway-based, sparse deep neural networks (PASNet) for cancer survival analysis. PASNet family is a biologically interpretable neural network model where nodes in the network correspond to specific genes and pathways, while capturing nonlinear and hierarchical effects of biological pathways associated with certain clinical outcomes. Furthermore, integration of heterogeneous types of biological data from biospecimen holds promise of improving survival prediction and personalized therapies in cancer. Specifically, the integration of genomic data and histopathological images enhances survival predictions and personalized treatments in cancer study, while providing an in-depth understanding of genetic mechanisms and phenotypic patterns of cancer. Two proposed models will be introduced for integrating multi-omics data and pathological images, respectively. Each model in PASNet family was evaluated by comparing the performance of current cutting-edge models with The Cancer Genome Atlas (TCGA) cancer data. In the extensive experiments, PASNet family outperformed the benchmarking methods, and the outstanding performance was statistically assessed. More importantly, PASNet family showed the capability to interpret a multi-layered biological system. A number of biological literature in GBM supported the biological interpretation of the proposed models. The open-source software of PASNet family in PyTorch is publicly available at https://github.com/DataX-JieHao

    New hints towards a precision medicine strategy for IDH wild-type glioblastoma.

    Get PDF
    Glioblastoma represents the most common primary malignancy of the central nervous system in adults and remains a largely incurable disease. The elucidation of disease subtypes based on mutational profiling, gene expression and DNA methylation has so far failed to translate into improved clinical outcomes. However, new knowledge emerging from the subtyping effort in the IDH-wild-type setting may provide directions for future precision therapies. Here, we review recent learnings in the field, and further consider how tumour microenvironment differences across subtypes may reveal novel contexts of vulnerability. We discuss recent treatment approaches and ongoing trials in the IDH-wild-type glioblastoma setting, and propose an integrated discovery stratagem incorporating multi-omics, single-cell technologies and computational approaches

    eXamine: a Cytoscape app for exploring annotated modules in networks

    Get PDF
    Background. Biological networks have growing importance for the interpretation of high-throughput "omics" data. Statistical and combinatorial methods allow to obtain mechanistic insights through the extraction of smaller subnetwork modules. Further enrichment analyses provide set-based annotations of these modules. Results. We present eXamine, a set-oriented visual analysis approach for annotated modules that displays set membership as contours on top of a node-link layout. Our approach extends upon Self Organizing Maps to simultaneously lay out nodes, links, and set contours. Conclusions. We implemented eXamine as a freely available Cytoscape app. Using eXamine we study a module that is activated by the virally-encoded G-protein coupled receptor US28 and formulate a novel hypothesis about its functioning

    Genome-scale Precision Proteomics Identifies Cancer Signaling Networks and Therapeutic Vulnerabilities

    Get PDF
    Mass spectrometry (MS) based-proteomics technology has been emerging as an indispensable tool for biomedical research. But the highly diverse physical and chemical properties of the protein building blocks and the dramatic human proteome complexity largely limited proteomic profiling depth. Moreover, there was a lack of high-throughput quantitative strategies that were both precise and parallel to in-depth proteomic techniques. To solve these grand challenges, a high resolution liquid chromatography (LC) system that coupled with an advanced mass spectrometer was developed to allow genome-scale human proteome identification. Using the combination of pre-MS peptide fractionation, MS2-based interference detection and post-MS computational interference correction, we enabled precise proteome quantification with isobaric labeling. We then applied these advanced proteomics tools for cancer proteome analyses on high grade gliomas (HGG) and rhabdomyosarcomas (RMS). Using systems biology approaches, we demonstrated that these newly developed proteomic analysis pipelines are able to (i) define human proteotypes that link oncogenotypes to cancer phenotypes in HGG and to (ii) identify therapeutic vulnerabilities in RMS. Development of high resolution liquid chromatography is essential for improving the sensitivity and throughput of mass spectrometry-based proteomics to genome-scale. Here we present systematic optimization of a long gradient LC-MS/MS platform to enhance protein identification from a complex mixture. The platform employed an in-house fabricated, reverse phase long column (100 ”m x 150 cm, 5 ”m C18 beads) coupled with Q Exactive MS. The column was capable of achieving a peak capacity of approximately 700 in a 720 min gradient of 10-45% acetonitrile. The optimal loading amount was about 6 micrograms of peptides, although the column allowed loading as many as 20 micrograms. Gas phase fractionation of peptide ions further increased the number of peptides identified by ~10%. Moreover, the combination of basic pH LC pre-fractionation with the long gradient LC-MS/MS platform enabled the identification of 96,127 peptides and 10,544 proteins at 1% protein false discovery rate in a postmortem brain sample of Alzheimer’s disease. As deep RNA sequencing of the same specimen suggested that ~16,000 genes were expressed, current analysis covered more than 60% of the expressed proteome. Isobaric labeling quantification by mass spectrometry has emerged as a powerful technology for multiplexed large-scale protein profiling, but measurement accuracy in complex mixtures is confounded by the interference from co-isolated ions, resulting in ratio compression. Here we report that the ratio compression can be essentially resolved by the combination of pre-MS peptide fractionation, MS2-based interference detection and post-MS computational interference correction. To recapitulate the complexity of biological samples, we pooled tandem mass tag (TMT) labeled E. coli peptides at 1 : 3 : 10 ratios, and added in ~20-fold more rat peptides as background, followed by the analysis of two dimensional liquid chromatography-MS/MS. Systematic investigation indicated that the quantitative interference was impacted by LC fractionation depth, MS isolation window and peptide loading amount. Exhaustive fractionation (320 x 4 h) can nearly eliminate the interference and achieve results comparable to the MS3-based method. Importantly, the interference in MS2 scans can be estimated by the intensity of contaminated y1 product ions, and we thus developed an algorithm to correct reporter ion ratios of tryptic peptides. Our data indicated that intermediate fractionation (40 x 2 h) and y1 ion-based correction allowed accurate and deep TMT protein profiling, which represents a straightforward and affordable strategy in isobaric labeling proteomics High throughput omics approaches provide an unprecedented opportunity for dissecting molecular mechanisms in cancer biology. Here we present deep profiling of whole proteome, phosphoproteome and transcriptome in two high-grade glioma mouse models driven by mutated receptor tyrosine kinase (RTK) oncogenes, platelet-derived growth factor receptor alpha (PDGFRA) and neurotrophic receptor tyrosine kinase 1 (NTRK1), analyzing 13,860 proteins (11,941 genes) and 30,431 phosphosites by mass spectrometry. Systems biology approaches identified numerous functional modules and master regulators, including 41 kinases and 26 transcription factors. Pathway activity computation and mouse survival curves indicate the NTRK1 mutation induces a higher activation of AKT targets, drives a positive feedback loop to up-regulate multiple other RTKs, and shows higher oncogenic potency than the PDGFRA mutation. Further integration of the mouse data with human HGG transcriptome data determines shared regulators of invasion and stemness. Thus, multi-omics integrative profiling is a powerful avenue to characterize oncogenic activity. There is growing emphasis on personalizing cancer therapy based on somatic mutations identified in patient’s tumors. Among pediatric solid tumors, RAS pathway mutations in rhabdomyosarcoma are the most common potentially actionable lesions. Recent success targeting CDK4/6 and MEK in RAS mutant adult cancers led our collaborator Dr. Dyer’s group to test this approach for rhabdomyosarcoma. They achieved synergistic killing of RAS mutant rhabdomyosarcoma tumor cells by combining MEK and CDK4/6 inhibitors in culture but failed to achieve efficacy in vivo using orthotopic patient derived xenografts (O-PDXs). To determine how rhabdomyosarcomas evade targeting of CDK4/6 and MEK, we collaborated to perform large-scale deep proteomic, phosphoproteomic, and epigenomic profiling of RMS tumors. Integrative analysis of these omics data detected that RMS tumor cells rapidly compensate and overcome CDK4/6 and MEK combination therapy through 6 myogenic signal transduction pathways including WNT, HH, BMP, Adenyl Cyclase, P38/MAPK and PI3K. While it is not feasible to target each of these signal transduction pathways simultaneously in RMS, we discovered that they require the HSP90 chaperone to sustain the complex developmental signal transduction milieu. We achieved specific and synergistic killing of RMS cells using sub-therapeutic concentrations of an HSP90 inhibitor (ganetespib) in combination with conventional chemotherapy used for recurrent RMS. These effects were seen in the most aggressive recurrent RMS orthotopic patient derived xenografts irrespective of RAS pathway perturbations, histologic or molecular classification. Thus, multi-omics integrative cancer profiling using our newly developed tools is powerful to identify core signaling transduction networks, tumor vulnerability (master regulators) for novel cancer therapy

    Linking Proteomic and Transcriptional Data through the Interactome and Epigenome Reveals a Map of Oncogene-induced Signaling

    Get PDF
    Cellular signal transduction generally involves cascades of post-translational protein modifications that rapidly catalyze changes in protein-DNA interactions and gene expression. High-throughput measurements are improving our ability to study each of these stages individually, but do not capture the connections between them. Here we present an approach for building a network of physical links among these data that can be used to prioritize targets for pharmacological intervention. Our method recovers the critical missing links between proteomic and transcriptional data by relating changes in chromatin accessibility to changes in expression and then uses these links to connect proteomic and transcriptome data. We applied our approach to integrate epigenomic, phosphoproteomic and transcriptome changes induced by the variant III mutation of the epidermal growth factor receptor (EGFRvIII) in a cell line model of glioblastoma multiforme (GBM). To test the relevance of the network, we used small molecules to target highly connected nodes implicated by the network model that were not detected by the experimental data in isolation and we found that a large fraction of these agents alter cell viability. Among these are two compounds, ICG-001, targeting CREB binding protein (CREBBP), and PKF118–310, targeting ÎČ-catenin (CTNNB1), which have not been tested previously for effectiveness against GBM. At the level of transcriptional regulation, we used chromatin immunoprecipitation sequencing (ChIP-Seq) to experimentally determine the genome-wide binding locations of p300, a transcriptional co-regulator highly connected in the network. Analysis of p300 target genes suggested its role in tumorigenesis. We propose that this general method, in which experimental measurements are used as constraints for building regulatory networks from the interactome while taking into account noise and missing data, should be applicable to a wide range of high-throughput datasets.National Science Foundation (U.S.) (DB1-0821391)National Institutes of Health (U.S.) (Grant U54-CA112967)National Institutes of Health (U.S.) (Grant R01-GM089903)National Institutes of Health (U.S.) (P30-ES002109

    Developing methods for the context-specific reconstruction of metabolic models of cancer cells

    Get PDF
    Dissertação de mestrado em BioinformĂĄticaThe recent advances in genome sequencing technologies and other high-throughput methodologies allowed the identification and quantification of individual cell components. These efforts led to the development of genome-scale metabolic models (GSMMs), not only for humans but also for several other organisms. These models have been used to predict cellular metabolic phenotypes under a variety of physiological conditions and contexts, proving to be useful in tasks such as drug discovery, biomarker identification and interactions between hosts and pathogens. Therefore, these models provide a useful tool for targeting diseases such as cancer, Alzheimer or tuberculosis. However, the usefulness of GSSMs is highly dependent on their capabilities to predict phenotypes in the array of different cell types that compose the human body, making the development of tissue/context-specific models mandatory. To address this issue, several methods have been proposed to integrate omics data, such as transcriptomics or proteomics, to improve the phenotype prediction abilities of GSSMs. Despite these efforts, these methods still have some limitations. In most cases, their usage is locked behind commercially licensed software platforms, or not available in a user-friendly fashion, thus restricting their use to users with programming or command-line knowledge. In this work, an open-source tool was developed for the reconstruction of tissue/context-specific models based on a generic template GSMM and the integration of omics data. The Tissue-Specific Model Reconstruction (TSM-Rec) tool was developed under the Python programming language and features the FASTCORE algorithm for the reconstruction of tissue/context-specific metabolic models. Its functionalities include the loading of omics data from a variety of omics databases, a set of filtering and transformation methods to adjust the data for integration with a template metabolic model, and finally the reconstruction of tissue/context-specific metabolic models. To evaluate the functionality of the developed tool, a cancer related case-study was carried. Using omics data from 314 glioma patients, the TSM-Rec tool was used to reconstruct metabolic models of different grade gliomas. A total of three models were generated, corresponding to grade II, III and IV gliomas. These models were analysed regarding their differences and similarities in reactions and pathways. This comparison highlighted biological processes common to all glioma grades, and pathways that are more prominent in each glioma model. The results show that the tool developed during this work can be useful for the reconstruction of cancer metabolic models, in a search for insights into cancer metabolism and possible approaches towards drug-target discovery.Os avanços recentes nas tecnologias de sequenciação de genomas e noutras metodologias experimentais de alto rendimento permitiram a identificação e quantificação dos diversos componentes celulares. Estes esforços levaram ao desenvolvimento de Modelos MetabĂłlicos Ă  Escala GenĂłmica (MMEG) nĂŁo sĂł de humanos, mas tambĂ©m de diversos organismos. Estes modelos tĂȘm sido utilizados para a previsĂŁo de fenĂłtipos metabĂłlicos sob uma variedade de contextos e condiçÔes fisiolĂłgicas, mostrando a sua utilidade em ĂĄreas como a descoberta de fĂĄrmacos, a identificação de biomarcadores ou interaçÔes entre hĂłspede e patĂłgeno. Desta forma, estes modelos revelam-se ferramentas Ășteis para o estudo de doenças como o cancro, Alzheimer ou a tuberculose. Contudo, a utilidade dos MMEG estĂĄ altamente dependente das suas capacidades de previsĂŁo de fenĂłtipos nos diversostipos celulares que compĂ”em o corpo humano, tornando o desenvolvimento de modelos especĂ­ficos de tecidos uma tarefa obrigatĂłria. Para resolver este problema, vĂĄrios mĂ©todos tĂȘm proposto a integração de dados Ăłmicos como os de transcriptĂłmica ou proteĂłmica para melhorar as capacidades preditivas dos MMEG. Apesar disso, estes mĂ©todos ainda sofrem de algumas limitaçÔes. Na maioria dos casos o seu uso estĂĄ confinado a plataformas ou softwares com licenças comerciais, ou nĂŁo estĂĄ disponĂ­vel numa ferramenta de fĂĄcil uso, limitando a sua utilização a utilizadores com conhecimentos de programação ou de linha de comandos. Neste trabalho, foi desenvolvida uma ferramenta de acesso livre para a reconstrução de modelos metabĂłlicos especĂ­ficos para tecidos tendo por base um MMEG genĂ©rico e a integração de dados Ăłmicos. A ferramenta TSM-Rec (Tissue-Specific Model Reconstruction), foi desenvolvida na linguagem de programação Python e recorre ao algoritmo FASTCORE para efetuar a reconstrução de modelos metabĂłlicos especĂ­ficos. As suas funcionalidades permitem a leitura de dados Ăłmicos de diversas bases de dados Ăłmicas, a filtragem e transformação dos mesmos para permitir a sua integração com um modelo metabĂłlico genĂ©rico e por fim, a reconstrução de modelos metabĂłlicos especĂ­ficos. De forma a avaliar o funcionamento da ferramenta desenvolvida, esta foi aplicada num caso de estudo de cancro. Recorrendo a dados Ăłmicos de 314 pacientes com glioma, usou-se a ferramenta TSM-Rec para a reconstrução de modelos metabĂłlicos de gliomas de diferentes graus. No total, foram desenvolvidos trĂȘs modelos correspondentes a gliomas de grau II, grau III e grau IV. Estes modelos foram analisados no sentido de perceber as diferenças e as similaridades entre as reaçÔes e as vias metabĂłlicas envolvidas em cada um dos modelos. Esta comparação permitiu isolar processos biolĂłgicos comuns a todos os graus de glioma, assim como vias metabĂłlicas que se destacam em cada um dos graus. Os resultados obtidos demonstram que a ferramenta desenvolvida pode ser Ăștil para a reconstrução de modelos metabĂłlicos de cancro, na procura de um melhor conhecimento do metabolismo do cancro e possĂ­veis abordagens para a descoberta de fĂĄrmacos
    • 

    corecore