661 research outputs found

    Identification of lung cancer gene markers through kernel maximum mean discrepancy and information entropy

    Full text link
    © 2019 The Author(s). Background: The early diagnosis of lung cancer has been a critical problem in clinical practice for a long time and identifying differentially expressed gene as disease marker is a promising solution. However, the most existing gene differential expression analysis (DEA) methods have two main drawbacks: First, these methods are based on fixed statistical hypotheses and not always effective; Second, these methods can not identify a certain expression level boundary when there is no obvious expression level gap between control and experiment groups. Methods: This paper proposed a novel approach to identify marker genes and gene expression level boundary for lung cancer. By calculating a kernel maximum mean discrepancy, our method can evaluate the expression differences between normal, normal adjacent to tumor (NAT) and tumor samples. For the potential marker genes, the expression level boundaries among different groups are defined with the information entropy method. Results: Compared with two conventional methods t-test and fold change, the top average ranked genes selected by our method can achieve better performance under all metrics in the 10-fold cross-validation. Then GO and KEGG enrichment analysis are conducted to explore the biological function of the top 100 ranked genes. At last, we choose the top 10 average ranked genes as lung cancer markers and their expression boundaries are calculated and reported. Conclusion: The proposed approach is effective to identify gene markers for lung cancer diagnosis. It is not only more accurate than conventional DEA methods but also provides a reliable method to identify the gene expression level boundaries

    Phenotype-driven identification of epithelial signalling clusters

    Get PDF
    In metazoans, epithelial architecture provides a context that dynamically modulates most if not all epithelial cell responses to intrinsic and extrinsic signals, including growth or survival signalling and transforming oncogene action. Three-dimensional ( 3D) epithelial culture systems provide tractable models to interrogate the function of human genetic determinants in establishment of context-dependency. We performed an arrayed genetic shRNA screen in mammary epithelial 3D cultures to identify new determinants of epithelial architecture, finding that the key phenotype impacting shRNAs altered not only the data population average but even more noticeably the population distribution. The broad distributions were attributable to sporadic gene silencing actions by shRNA in unselected populations. We employed Maximum Mean Discrepancy concept to capture similar population distribution patterns and demonstrate here the feasibility of the test in identifying an impact of shRNA in populations of 3D structures. Integration of the clustered morphometric data with protein-protein interactions data enabled hypothesis generation of novel biological pathways underlying similar 3D phenotype alterations. The results present a new strategy for 3D phenotype-driven pathway analysis, which is expected to accelerate discovery of context-dependent gene functions in epithelial biology and tumorigenesis.Peer reviewe

    Bayesian kernel two-sample testing

    Get PDF
    In modern data analysis, nonparametric measures of discrepancies between random variables are particularly important. The subject is well-studied in the frequentist literature, while the development in the Bayesian setting is limited where applications are often restricted to univariate cases. Here, we propose a Bayesian kernel two-sample testing procedure based on modelling the difference between kernel mean embeddings in the reproducing kernel Hilbert space utilising the framework established by Flaxman et al (2016). The use of kernel methods enables its application to random variables in generic domains beyond the multivariate Euclidean spaces. The proposed procedure results in a posterior inference scheme that allows an automatic selection of the kernel parameters relevant to the problem at hand. In a series of synthetic experiments and two real data experiments (i.e. testing network heterogeneity from high-dimensional data and six-membered monocyclic ring conformation comparison), we illustrate the advantages of our approach

    Feature selection and modelling methods for microarray data from acute coronary syndrome

    Get PDF
    Acute coronary syndrome (ACS) represents a leading cause of mortality and morbidity worldwide. Providing better diagnostic solutions and developing therapeutic strategies customized to the individual patient represent societal and economical urgencies. Progressive improvement in diagnosis and treatment procedures require a thorough understanding of the underlying genetic mechanisms of the disease. Recent advances in microarray technologies together with the decreasing costs of the specialized equipment enabled affordable harvesting of time-course gene expression data. The high-dimensional data generated demands for computational tools able to extract the underlying biological knowledge. This thesis is concerned with developing new methods for analysing time-course gene expression data, focused on identifying differentially expressed genes, deconvolving heterogeneous gene expression measurements and inferring dynamic gene regulatory interactions. The main contributions include: a novel multi-stage feature selection method, a new deconvolution approach for estimating cell-type specific signatures and quantifying the contribution of each cell type to the variance of the gene expression patters, a novel approach to identify the cellular sources of differential gene expression, a new approach to model gene expression dynamics using sums of exponentials and a novel method to estimate stable linear dynamical systems from noisy and unequally spaced time series data. The performance of the proposed methods was demonstrated on a time-course dataset consisting of microarray gene expression levels collected from the blood samples of patients with ACS and associated blood count measurements. The results of the feature selection study are of significant biological relevance. For the first time is was reported high diagnostic performance of the ACS subtypes up to three months after hospital admission. The deconvolution study exposed features of within and between groups variation in expression measurements and identified potential cell type markers and cellular sources of differential gene expression. It was shown that the dynamics of post-admission gene expression data can be accurately modelled using sums of exponentials, suggesting that gene expression levels undergo a transient response to the ACS events before returning to equilibrium. The linear dynamical models capturing the gene regulatory interactions exhibit high predictive performance and can serve as platforms for system-level analysis, numerical simulations and intervention studies

    RNA 상호작용 및 DNA 서열의 정보해독을 위한 기계학습 기법

    Get PDF
    학위논문(박사)--서울대학교 대학원 :공과대학 컴퓨터공학부,2020. 2. 김선.생물체 간 표현형의 차이는 각 개체의 유전적 정보 차이로부터 기인한다. 유전적 정보의 변화에 따라서, 각 생물체는 서로 다른 종으로 진화하기도 하고, 같은 병에 걸린 환자라도 서로 다른 예후를 보이기도 한다. 이처럼 중요한 생물학적 정보는 대용량 시퀀싱 분석 기법 등을 통해 다양한 오믹스 데이터로 측정된다. 그러나, 오믹스 데이터는 고차원 특징 및 소규모 표본 데이터이기 때문에, 오믹스 데이터로부터 생물학적 정보를 해석하는 것은 매우 어려운 문제이다. 일반적으로, 데이터 특징의 개수가 샘플의 개수보다 많을 때, 오믹스 데이터의 해석을 가장 난해한 기계학습 문제들 중 하나로 만듭니다. 본 박사학위 논문은 기계학습 기법을 활용하여 고차원적인 생물학적 데이터로부터 생물학적 정보를 추출하기 위한 새로운 생물정보학 방법들을 고안하는 것을 목표로 한다. 첫 번째 연구는 DNA 서열을 활용하여 종 간 비교와 동시에 DNA 서열상에 있는 다양한 지역에 담긴 생물학적 정보를 유전적 관점에서 해석해보고자 하였다. 이를 위해, 순위 기반 k 단어 문자열 비교방법, RKSS 커널을 개발하여 다양한 게놈 상의 지역에서 여러 종 간 비교 실험을 수행하였다. RKSS 커널은 기존의 k 단어 문자열 커널을 확장한 것으로, k 길이 단어의 순위 정보와 종 간 공통점을 표현하는 비교기준점 개념을 활용하였다. k 단어 문자열 커널은 k의 길이에 따라 단어 수가 급증하지만, 비교기준점은 극소수의 단어로 이루어져 있으므로 서열 간 유사도를 계산하는 데 필요한 계산량을 효율적으로 줄일 수 있다. 게놈 상의 세 지역에 대해서 실험을 진행한 결과, RKSS 커널은 기존의 커널에 비해 종 간 유사도 및 차이를 효율적으로 계산할 수 있었다. 또한, RKSS 커널은 실험에 사용된 생물학적 지역에 포함된 생물학적 정보량 차이를 생물학적 지식과 부합되는 순서로 비교할 수 있었다. 두 번째 연구는 생물학적 네트워크를 통해 복잡하게 얽힌 유전자 상호작용 간 정보를 해석하여, 더 나아가 생물학적 기능 해석을 통해 암의 아형을 분류하고자 하였다. 이를 위해, 그래프 컨볼루션 네트워크와 어텐션 메커니즘을 활용하여 패스웨이 기반 해석 가능한 암 아형 분류 모델(GCN+MAE)을 고안하였다. 그래프 컨볼루션 네트워크를 통해서 생물학적 사전 지식인 패스웨이 정보를 학습하여 복잡한 유전자 상호작용 정보를 효율적으로 다루었다. 또한, 여러 패스웨이 정보를 어텐션 메커니즘을 통해 해석 가능한 수준으로 병합하였다. 마지막으로, 학습한 패스웨이 레벨 정보를 보다 복잡하고 다양한 유전자 레벨로 효율적으로 전달하기 위해서 네트워크 전파 알고리즘을 활용하였다. 다섯 개의 암 데이터에 대해 GCN+MAE 모델을 적용한 결과, 기존의 암 아형 분류 모델들보다 나은 성능을 보였으며 암 아형 특이적인 패스웨이 및 생물학적 기능을 발굴할 수 있었다. 세 번째 연구는 패스웨이로부터 서브 패스웨이/네트워크를 찾기 위한 연구다. 패스웨이나 생물학적 네트워크에 단일 생물학적 기능이 아니라 다양한 생물학적 기능이 포함되어 있음에 주목하였다. 단일 기능을 지닌 유전자 조합을 찾기 위해서 생물학적 네트워크상에서 조건 특이적인 유전자 모듈을 찾고자 하였으며 MIDAS라는 도구를 개발하였다. 패스웨이로부터 유전자 상호작용 간 활성도를 유전자 발현량과 네트워크 구조를 통해 계산하였다. 계산된 활성도들을 활용하여 다중 클래스에서 서로 다르게 활성화된 서브 패스들을 통계적 기법에 기반하여 발굴하였다. 또한, 어텐션 메커니즘과 그래프 컨볼루션 네트워크를 통해서 해당 연구를 패스웨이보다 더 큰 생물학적 네트워크에 확장하려고 시도하였다. 유방암 데이터에 대해 실험을 진행한 결과, MIDAS와 딥러닝 모델을 다중 클래스에서 차이가 나는 유전자 모듈을 효과적으로 추출할 수 있었다. 결론적으로, 본 박사학위 논문은 DNA 서열에 담긴 진화적 정보량 비교, 패스웨이 기반 암 아형 분류, 조건 특이적인 유전자 모듈 발굴을 위한 새로운 기계학습 기법을 제안하였다.Phenotypic differences among organisms are mainly due to the difference in genetic information. As a result of genetic information modification, an organism may evolve into a different species and patients with the same disease may have different prognosis. This important biological information can be observed in the form of various omics data using high throughput instrument technologies such as sequencing instruments. However, interpretation of such omics data is challenging since omics data is with very high dimensions but with relatively small number of samples. Typically, the number of dimensions is higher than the number of samples, which makes the interpretation of omics data one of the most challenging machine learning problems. My doctoral study aims to develop new bioinformatics methods for decoding information in these high dimensional data by utilizing machine learning algorithms. The first study is to analyze the difference in the amount of information between different regions of the DNA sequence. To achieve the goal, a ranked-based k-spectrum string kernel, RKSS kernel, is developed for comparative and evolutionary comparison of various genomic region sequences among multiple species. RKSS kernel extends the existing k-spectrum string kernel by utilizing rank information of k-mers and landmarks of k-mers that represents a species. By using a landmark as a reference point for comparison, the number of k-mers needed to calculating sequence similarities is dramatically reduced. In the experiments on three different genomic regions, RKSS kernel captured more reliable distances between species according to genetic information contents of the target region. Also, RKSS kernel was able to rearrange each region to match a biological common insight. The second study aims to efficiently decode complex genetic interactions using biological networks and, then, to classify cancer subtypes by interpreting biological functions. To achieve the goal, a pathway-based deep learning model using graph convolutional network and multi-attention based ensemble (GCN+MAE) for cancer subtype classification is developed. In order to efficiently reduce the relationships between genes using pathway information, GCN+MAE is designed as an explainable deep learning structure using graph convolutional network and attention mechanism. Extracted pathway-level information of cancer subtypes is transported into gene-level again by network propagation. In the experiments of five cancer data sets, GCN+MAE showed better cancer subtype classification performances and captured subtype-specific pathways and their biological functions. The third study is to identify sub-networks of a biological pathway. The goal is to dissect a biological pathway into multiple sub-networks, each of which is to be of a single functional unit. To achieve the goal, a condition-specific sub-module detection method in a biological network, MIDAS (MIning Differentially Activated Subpaths) is developed. From the pathway, edge activities are measured by explicit gene expression and network topology. Using the activities, differentially activated subpaths are explored by a statistical approach. Also, by extending this idea on graph convolutional network, different sub-networks are highlighted by attention mechanisms. In the experiment with breast cancer data, MIDAS and the deep learning model successfully decomposed gene-level features into sub-modules of single functions. In summary, my doctoral study proposes new computational methods to compare genomic DNA sequences as information contents, to model pathway-based cancer subtype classifications and regulations, and to identify condition-specific sub-modules among multiple cancer subtypes.Chapter 1 Introduction 1 1.1 Biological questions with genetic information 2 1.1.1 Biological Sequences 2 1.1.2 Gene expression 2 1.2 Formulating computational problems for the biological questions 3 1.2.1 Decoding biological sequences by k-mer vectors 3 1.2.2 Interpretation of complex relationships between genes 7 1.3 Three computational problems for the biological questions 9 1.4 Outline of the thesis 14 Chapter 2 Ranked k-spectrum kernel for comparative and evolutionary comparison of DNA sequences 15 2.1 Motivation 16 2.1.1 String kernel for sequence comparison 17 2.1.2 Approach: RKSS kernel 19 2.2 Methods 21 2.2.1 Mapping biological sequences to k-mer space: the k-spectrum string kernel 23 2.2.2 The ranked k-spectrum string kernel with a landmark 24 2.2.3 Single landmark-based reconstruction of phylogenetic tree 27 2.2.4 Multiple landmark-based distance comparison of exons, introns, CpG islands 29 2.2.5 Sequence Data for analysis 30 2.3 Results 31 2.3.1 Reconstruction of phylogenetic tree on the exons, introns, and CpG islands 31 2.3.2 Landmark space captures the characteristics of three genomic regions 38 2.3.3 Cross-evaluation of the landmark-based feature space 45 Chapter 3 Pathway-based cancer subtype classification and interpretation by attention mechanism and network propagation 46 3.1 Motivation 47 3.2 Methods 52 3.2.1 Encoding biological prior knowledge using Graph Convolutional Network 52 3.2.2 Re-producing comprehensive biological process by Multi-Attention based Ensemble 53 3.2.3 Linking pathways and transcription factors by network propagation with permutation-based normalization 55 3.3 Results 58 3.3.1 Pathway database and cancer data set 58 3.3.2 Evaluation of individual GCN pathway models 60 3.3.3 Performance of ensemble of GCN pathway models with multi-attention 60 3.3.4 Identification of TFs as regulator of pathways and GO term analysis of TF target genes 67 Chapter 4 Detecting sub-modules in biological networks with gene expression by statistical approach and graph convolutional network 70 4.1 Motivation 70 4.1.1 Pathway based analysis of transcriptome data 71 4.1.2 Challenges and Summary of Approach 74 4.2 Methods 78 4.2.1 Convert single KEGG pathway to directed graph 79 4.2.2 Calculate edge activity for each sample 79 4.2.3 Mining differentially activated subpath among classes 80 4.2.4 Prioritizing subpaths by the permutation test 82 4.2.5 Extension: graph convolutional network and class activation map 83 4.3 Results 84 4.3.1 Identifying 36 subtype specific subpaths in breast cancer 86 4.3.2 Subpath activities have a good discrimination power for cancer subtype classification 88 4.3.3 Subpath activities have a good prognostic power for survival outcomes 90 4.3.4 Comparison with an existing tool, PATHOME 91 4.3.5 Extension: detection of subnetwork on PPI network 98 Chapter 5 Conclusions 101 국문초록 127Docto

    Inferential stability in systems biology

    Get PDF
    The modern biological sciences are fraught with statistical difficulties. Biomolecular stochasticity, experimental noise, and the “large p, small n” problem all contribute to the challenge of data analysis. Nevertheless, we routinely seek to draw robust, meaningful conclusions from observations. In this thesis, we explore methods for assessing the effects of data variability upon downstream inference, in an attempt to quantify and promote the stability of the inferences we make. We start with a review of existing methods for addressing this problem, focusing upon the bootstrap and similar methods. The key requirement for all such approaches is a statistical model that approximates the data generating process. We move on to consider biomarker discovery problems. We present a novel algorithm for proposing putative biomarkers on the strength of both their predictive ability and the stability with which they are selected. In a simulation study, we find our approach to perform favourably in comparison to strategies that select on the basis of predictive performance alone. We then consider the real problem of identifying protein peak biomarkers for HAM/TSP, an inflammatory condition of the central nervous system caused by HTLV-1 infection. We apply our algorithm to a set of SELDI mass spectral data, and identify a number of putative biomarkers. Additional experimental work, together with known results from the literature, provides corroborating evidence for the validity of these putative biomarkers. Having focused on static observations, we then make the natural progression to time course data sets. We propose a (Bayesian) bootstrap approach for such data, and then apply our method in the context of gene network inference and the estimation of parameters in ordinary differential equation models. We find that the inferred gene networks are relatively unstable, and demonstrate the importance of finding distributions of ODE parameter estimates, rather than single point estimates

    Modular Algorithms for Biomolecular Network Alignment

    Get PDF
    Comparative analysis of biomolecular networks constructed using measurements from different conditions, tissues, and organisms offer a powerful approach to understanding the structure, function, dynamics, and evolution of complex biological systems. The rapidly advancing field of systems biology aims to understand the structure, function, dynamics, and evolution of complex biological systems in terms of the underlying networks of interactions among the large number of molecular participants involved including genes, proteins, and metabolites. In particular, the comparative analysis of network models representing biomolecular interactions in different species or tissues offers an important tool for identifying conserved modules, predicting functions of specific genes or proteins and studying the evolution of biological processes, among other applications. The primary focus of this dissertation is on the biomolecular network alignment problem: Given two or more network models, the problem is to optimally match the nodes and links in one network with the nodes and links of the other. The Biomolecular Network Alignment (BiNA) Toolkit developed as part of this dissertation provides a set of efficient (in terms of the running time complexity) and accurate (in terms of various evaluation criteria discussed in the literature) network alignment algorithms for biomolecular networks. BiNA is scalable, user-friendly, modular, and extensible for performing alignments on diverse types of biomolecular networks. The algorithm is applicable to (1) undirected graphs in their weighted and unweighted variations (2) undirected graphs in their labeled and unlabeled variations (3) and has been applied to align multiple networks from hundreds of nodes with a few thousand edges to networks with tens of thousands of nodes with millions of edges. The dissertation provides various applications of network comparison tools including how results from such alignments have been utilized to (1) construct phylogenetic trees based on protein-protein interaction networks, and (2) find biochemical pathways involved in ligand recognition in B cells

    Cell type identification, differential expression analysis and trajectory inference in single-cell transcriptomics

    Get PDF
    Single-cell RNA-sequencing (scRNA-seq) is a cutting-edge technology that enables to quantify the transcriptome, the set of expressed RNA transcripts, of a group of cells at the single-cell level. It represents a significant upgrade from bulk RNA-seq, which measures the combined signal of thousands of cells. Measuring gene expression by bulk RNA-seq is an invaluable tool for biomedical researchers who want to understand how cells alter their gene expression due to an illness, differentiation, ternal stimulus, or other events. Similarly, scRNA-seq has become an essential method for biomedical researchers, and it has brought several new applications previously unavailable with bulk RNA-seq. scRNA-seq has the same applications as bulk RNA-seq. However, the single-cell resolution also enables cell annotation based on gene markers of clusters, that is, cell populations that have been identified based on machine learning to be, on average, dissimilar at the transcriptomic level. Researchers can use the cell clusters to detect cell-type-specific gene expression changes between conditions such as case and control groups. Clustering can sometimes even discover entirely new cell types. Besides the cluster-level representation, the single-cell resolution also enables to model cells as a trajectory, representing how the cells are related at the cell level and what is the dynamic differentiation process that the cells undergo in a tissue. This thesis introduces new computational methods for cell type identification and trajectory inference from scRNA-seq data. A new cell type identification method (ILoReg) was proposed, which enables high-resolution clustering of cells into populations with subtle transcriptomic differences. In addition, two new trajectory inference methods were developed: scShaper, which is an accurate and robust method for inferring linear trajectories; and Totem, which is a user-friendly and flexible method for inferring tree-shaped trajectories. In addition, one of the works benchmarked methods for detecting cell-type-specific differential states from scRNA-seq data with multiple subjects per comparison group, requiring tailored methods to confront false discoveries. KEYWORDS: Single-cell RNA sequencing, transcriptome, cell type identification, trajectory inference, differential expressionYksisoluinen RNA-sekvensointi on huipputeknologia, joka mahdollistaa transkriptomin eli ilmentyneiden RNA-transkriptien laskennallisen määrittämisen joukolle soluja yhden solun tarkkuudella, ja sen kehittäminen oli merkittävä askel eteenpäin perinteisestä bulkki-RNA-sekvensoinnista, joka mittaa tuhansien solujen yhteistä signaalia. Bulkki-RNA-sekvensointi on tärkeä työväline biolääketieteen tutkijoille, jotka haluavat ymmärtää miten solut muuttavat geenien ilmentymistä sairauden, erilaistumisen, ulkoisen ärsykkeen tai muun tapahtuman seurauksena. Yksisoluisesta RNA-sekvensoinnista on vastaavasti kehittynyt tärkeä työväline tutkijoille, ja se on tuonut useita uusia sovelluksia. Yksisoluisella RNA-sekvensoinnilla on samat sovellukset kuin bulkki-RNA-sekvensoinnilla, mutta sen lisäksi se mahdollistaa solujen tunnistamisen geenimarkkerien perusteella. Geenimarkkerit etsitään tilastollisin menetelmin solupopulaatioille, joiden on tunnistettu koneoppimisen menetelmin muodostavan transkriptomitasolla keskenään erilaisia joukkoja eli klustereita. Tutkijat voivat hyödyntää soluklustereita tutkimaan geeniekspressioeroja solutyyppien sisällä esimerkiksi sairaiden ja terveiden välillä, ja joskus klusterointi voi jopa tunnistaa uusia solutyyppejä. Yksisolutason mittaukset mahdollistavat myös solujen mallintamisen trajektorina, joka esittää kuinka solut kehittyvät dynaamisesti toisistaan geenien ilmentymistä vaativien prosessien aikana. Tämä väitöskirja esittelee uusia laskennallisia menetelmiä solutyyppien ja trajektorien tunnistamiseen yksisoluisesta RNA-sekvensointidatasta. Väitöskirja esittelee uuden solutyyppitunnistusmenetelmän (ILoReg), joka mahdollistaa hienovaraisia geeniekspressioeroja sisältävien solutyyppien tunnistamisen. Sen lisäksi väitöskirjassa kehitettiin kaksi uutta trajektorin tunnistusmenetelmää: scShaper, joka on tarkka ja robusti menetelmä lineaaristen trajektorien tunnistamiseen, sekä Totem, joka on käyttäjäystävällinen ja joustava menetelmä puumallisten trajektorien tunnistamiseen. Lopuksi väitöskirjassa vertailtiin menetelmiä solutyyppien sisäisten geeniekspressioerojen tunnistamiseen ryhmien välillä, joissa on useita koehenkilöitä tai muita biologisia replikaatteja, mikä vaatii erityisiä menetelmiä väärien positiivisten löydösten vähentämiseen. ASIASANAT: yksisoluinen RNA-sekvensointi, klusterointi, trajektorin tunnistus, geeniekspressi

    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
    corecore