Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Rui Kuang is active.

Publication


Featured researches published by Rui Kuang.


PLOS Computational Biology | 2013

Network-based Survival Analysis Reveals Subnetwork Signatures for Predicting Outcomes of Ovarian Cancer Treatment

Wei Zhang; Takayo Ota; Viji Shridhar; Jeremy Chien; Baolin Wu; Rui Kuang

Cox regression is commonly used to predict the outcome by the time to an event of interest and in addition, identify relevant features for survival analysis in cancer genomics. Due to the high-dimensionality of high-throughput genomic data, existing Cox models trained on any particular dataset usually generalize poorly to other independent datasets. In this paper, we propose a network-based Cox regression model called Net-Cox and applied Net-Cox for a large-scale survival analysis across multiple ovarian cancer datasets. Net-Cox integrates gene network information into the Coxs proportional hazard model to explore the co-expression or functional relation among high-dimensional gene expression features in the gene network. Net-Cox was applied to analyze three independent gene expression datasets including the TCGA ovarian cancer dataset and two other public ovarian cancer datasets. Net-Cox with the network information from gene co-expression or functional relations identified highly consistent signature genes across the three datasets, and because of the better generalization across the datasets, Net-Cox also consistently improved the accuracy of survival prediction over the Cox models regularized by or . This study focused on analyzing the death and recurrence outcomes in the treatment of ovarian carcinoma to identify signature genes that can more reliably predict the events. The signature genes comprise dense protein-protein interaction subnetworks, enriched by extracellular matrix receptors and modulators or by nuclear signaling components downstream of extracellular signal-regulated kinases. In the laboratory validation of the signature genes, a tumor array experiment by protein staining on an independent patient cohort from Mayo Clinic showed that the protein expression of the signature gene FBN1 is a biomarker significantly associated with the early recurrence after 12 months of the treatment in the ovarian cancer patients who are initially sensitive to chemotherapy. Net-Cox toolbox is available at http://compbio.cs.umn.edu/Net-Cox/.


Bioinformatics | 2004

Protein backbone angle prediction with machine learning approaches

Rui Kuang; Christina S. Leslie; An-Suei Yang

MOTIVATION Protein backbone torsion angle prediction provides useful local structural information that goes beyond conventional three-state (alpha, beta and coil) secondary structure predictions. Accurate prediction of protein backbone torsion angles will substantially improve modeling procedures for local structures of protein sequence segments, especially in modeling loop conformations that do not form regular structures as in alpha-helices or beta-strands. RESULTS We have devised two novel automated methods in protein backbone conformational state prediction: one method is based on support vector machines (SVMs); the other method combines a standard feed-forward back-propagation artificial neural network (NN) with a local structure-based sequence profile database (LSBSP1). Extensive benchmark experiments demonstrate that both methods have improved the prediction accuracy rate over the previously published methods for conformation state prediction when using an alphabet of three or four states. AVAILABILITY LSBSP1 and the NN algorithm have been implemented in PrISM.1, which is available from www.columbia.edu/~ay1/. SUPPLEMENTARY INFORMATION Supplementary data for the SVM method can be downloaded from the Website www.cs.columbia.edu/compbio/backbone.


Bioinformatics | 2009

A hypergraph-based learning algorithm for classifying gene expression and arrayCGH data with prior knowledge

Ze Tian; Tae Hyun Hwang; Rui Kuang

MOTIVATION Incorporating biological prior knowledge into predictive models is a challenging data integration problem in analyzing high-dimensional genomic data. We introduce a hypergraph-based semi-supervised learning algorithm called HyperPrior to classify gene expression and array-based comparative genomic hybridization (arrayCGH) data using biological knowledge as constraints on graph-based learning. HyperPrior is a robust two-step iterative method that alternatively finds the optimal labeling of the samples and the optimal weighting of the features, guided by constraints encoding prior knowledge. The prior knowledge for analyzing gene expression data is that cancer-related genes tend to interact with each other in a protein-protein interaction network. Similarly, the prior knowledge for analyzing arrayCGH data is that probes that are spatially nearby in their layout along the chromosomes tend to be involved in the same amplification or deletion event. Based on the prior knowledge, HyperPrior imposes a consistent weighting of the correlated genomic features in graph-based learning. RESULTS We applied HyperPrior to test two arrayCGH datasets and two gene expression datasets for both cancer classification and biomarker identification. On all the datasets, HyperPrior achieved competitive classification performance, compared with SVMs and the other baselines utilizing the same prior knowledge. HyperPrior also identified several discriminative regions on chromosomes and discriminative subnetworks in the PPI, both of which contain cancer-related genomic elements. Our results suggest that HyperPrior is promising in utilizing biological prior knowledge to achieve better classification performance and more biologically interpretable findings in gene expression and arrayCGH data. AVAILABILITY http://compbio.cs.umn.edu/HyperPrior CONTACT [email protected] SUPPLEMENTARY INFORMATION Supplementary data are available at bioinformatics online.


conference on learning theory | 2003

Fast Kernels for Inexact String Matching

Christina S. Leslie; Rui Kuang

We introduce several new families of string kernels designed in particular for use with support vector machines (SVMs) for classification of protein sequence data. These kernels – restricted gappy kernels, substitution kernels, and wildcard kernels – are based on feature spaces indexed by k-length subsequences from the string alphabet Σ (or the alphabet augmented by a wildcard character), and hence they are related to the recently presented (k,m)-mismatch kernel and string kernels used in text classification. However, for all kernels we define here, the kernel value K(x,y) can be computed in O(c K (|x| + |y|)) time, where the constant c K depends on the parameters of the kernel but is independent of the size |Σ| of the alphabet. Thus the computation of these kernels is linear in the length of the sequences, like the mismatch kernel, but we improve upon the parameter-dependent constant \(c_K = k^{m+1} |\Sigma|^m\) of the mismatch kernel. We compute the kernels efficiently using a recursive function based on a trie data structure and relate our new kernels to the recently described transducer formalism. Finally, we report protein classification experiments on a benchmark SCOP dataset, where we show that our new faster kernels achieve SVM classification performance comparable to the mismatch kernel and the Fisher kernel derived from profile hidden Markov models.


Nucleic Acids Research | 2012

Co-clustering phenome–genome for phenotype classification and disease gene discovery

Tae Hyun Hwang; Gowtham Atluri; Maoqiang Xie; Sanjoy Dey; Changjin Hong; Vipin Kumar; Rui Kuang

Understanding the categorization of human diseases is critical for reliably identifying disease causal genes. Recently, genome-wide studies of abnormal chromosomal locations related to diseases have mapped >2000 phenotype–gene relations, which provide valuable information for classifying diseases and identifying candidate genes as drug targets. In this article, a regularized non-negative matrix tri-factorization (R-NMTF) algorithm is introduced to co-cluster phenotypes and genes, and simultaneously detect associations between the detected phenotype clusters and gene clusters. The R-NMTF algorithm factorizes the phenotype–gene association matrix under the prior knowledge from phenotype similarity network and protein–protein interaction network, supervised by the label information from known disease classes and biological pathways. In the experiments on disease phenotype–gene associations in OMIM and KEGG disease pathways, R-NMTF significantly improved the classification of disease phenotypes and disease pathway genes compared with support vector machines and Label Propagation in cross-validation on the annotated phenotypes and genes. The newly predicted phenotypes in each disease class are highly consistent with human phenotype ontology annotations. The roles of the new member genes in the disease pathways are examined and validated in the protein–protein interaction subnetworks. Extensive literature review also confirmed many new members of the disease classes and pathways as well as the predicted associations between disease phenotype classes and pathways.


BMC Bioinformatics | 2006

Protein Ranking by Semi-Supervised Network Propagation

Jason Weston; Rui Kuang; Christina S. Leslie; William Stafford Noble

BackgroundBiologists regularly search DNA or protein databases for sequences that share an evolutionary or functional relationship with a given query sequence. Traditional search methods, such as BLAST and PSI-BLAST, focus on detecting statistically significant pairwise sequence alignments and often miss more subtle sequence similarity. Recent work in the machine learning community has shown that exploiting the global structure of the network defined by these pairwise similarities can help detect more remote relationships than a purely local measure.MethodsWe review RankProp, a ranking algorithm that exploits the global network structure of similarity relationships among proteins in a database by performing a diffusion operation on a protein similarity network with weighted edges. The original RankProp algorithm is unsupervised. Here, we describe a semi-supervised version of the algorithm that uses labeled examples. Three possible ways of incorporating label information are considered: (i) as a validation set for model selection, (ii) to learn a new network, by choosing which transfer function to use for a given query, and (iii) to estimate edge weights, which measure the probability of inferring structural similarity.ResultsBenchmarked on a human-curated database of protein structures, the original RankProp algorithm provides significant improvement over local network search algorithms such as PSI-BLAST. Furthermore, we show here that labeled data can be used to learn a network without any need for estimating parameters of the transfer function, and that diffusion on this learned network produces better results than the original RankProp algorithm with a fixed network.ConclusionIn order to gain maximal information from a network, labeled and unlabeled data should be used to extract both local and global structure.


Frontiers in Oncology | 2013

Platinum-Sensitive Recurrence in Ovarian Cancer: The Role of Tumor Microenvironment

Jeremy Chien; Rui Kuang; Charles N. Landen; Viji Shridhar

Despite several advances in the understanding of ovarian cancer pathobiology, in terms of driver genetic alterations in high-grade serous cancer, histologic heterogeneity of epithelial ovarian cancer, cell-of-origin for ovarian cancer, the survival rate from ovarian cancer is disappointingly low when compared to that of breast or prostate cancer. One of the factors contributing to the poor survival rate from ovarian cancer is the development of chemotherapy resistance following several rounds of chemotherapy. Although unicellular drug resistance mechanisms contribute to chemotherapy resistance, tumor microenvironment and the extracellular matrix (ECM), in particular, is emerging as a significant determinant of a tumor’s response to chemotherapy. In this review, we discuss the potential role of the tumor microenvironment in ovarian cancer recurrence and resistance to chemotherapy. Finally, we propose an alternative view of platinum-sensitive recurrence to describe a potential role of the ECM in the process.


Bioinformatics | 2011

Inferring disease and gene set associations with rank coherence in networks

Tae Hyun Hwang; Wei Zhang; Maoqiang Xie; Jinfeng Liu; Rui Kuang

MOTIVATION To validate the candidate disease genes identified from high-throughput genomic studies, a necessary step is to elucidate the associations between the set of candidate genes and disease phenotypes. The conventional gene set enrichment analysis often fails to reveal associations between disease phenotypes and the gene sets with a short list of poorly annotated genes, because the existing annotations of disease-causative genes are incomplete. This article introduces a network-based computational approach called rcNet to discover the associations between gene sets and disease phenotypes. A learning framework is proposed to maximize the coherence between the predicted phenotype-gene set relations and the known disease phenotype-gene associations. An efficient algorithm coupling ridge regression with label propagation and two variants are designed to find the optimal solution to the objective functions of the learning framework. RESULTS We evaluated the rcNet algorithms with leave-one-out cross-validation on Online Mendelian Inheritance in Man (OMIM) data and an independent test set of recently discovered disease-gene associations. In the experiments, the rcNet algorithms achieved best overall rankings compared with the baselines. To further validate the reproducibility of the performance, we applied the algorithms to identify the target diseases of novel candidate disease genes obtained from recent studies of Genome-Wide Association Study (GWAS), DNA copy number variation analysis and gene expression profiling. The algorithms ranked the target disease of the candidate genes at the top of the rank list in many cases across all the three case studies. AVAILABILITY http://compbio.cs.umn.edu/dgsa_rcNet CONTACT [email protected].


Bioinformatics | 2008

Robust and efficient identification of biomarkers by classifying features on graphs

Tae Hyun Hwang; Hugues Sicotte; Ze Tian; Baolin Wu; Jean-Pierre A. Kocher; Dennis A. Wigle; Vipin Kumar; Rui Kuang

MOTIVATION A central problem in biomarker discovery from large-scale gene expression or single nucleotide polymorphism (SNP) data is the computational challenge of taking into account the dependence among all the features. Methods that ignore the dependence usually identify non-reproducible biomarkers across independent datasets. We introduce a new graph-based semi-supervised feature classification algorithm to identify discriminative disease markers by learning on bipartite graphs. Our algorithm directly classifies the feature nodes in a bipartite graph as positive, negative or neutral with network propagation to capture the dependence among both samples and features (clinical and genetic variables) by exploring bi-cluster structures in a graph. Two features of our algorithm are: (1) our algorithm can find a global optimal labeling to capture the dependence among all the features and thus, generates highly reproducible results across independent microarray or other high-thoughput datasets, (2) our algorithm is capable of handling hundreds of thousands of features and thus, is particularly useful for biomarker identification from high-throughput gene expression and SNP data. In addition, although designed for classifying features, our algorithm can also simultaneously classify test samples for disease prognosis/diagnosis. RESULTS We applied the network propagation algorithm to study three large-scale breast cancer datasets. Our algorithm achieved competitive classification performance compared with SVMs and other baseline methods, and identified several markers with clinical or biological relevance with the disease. More importantly, our algorithm also identified highly reproducible marker genes and enriched functions from the independent datasets. AVAILABILITY Supplementary results and source code are available at http://compbio.cs.umn.edu/Feature_Class. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.


international conference on data mining | 2008

Learning on Weighted Hypergraphs to Integrate Protein Interactions and Gene Expressions for Cancer Outcome Prediction

Tae Hyun Hwang; Ze Tian; Rui Kuang; Jean-Pierre A. Kocher

Building reliable predictive models from multiple complementary genomic data for cancer study is a crucial step towards successful cancer treatment and a full understanding of the underlying biological principles. To tackle this challenging data integration problem, we propose a hypergraph-based learning algorithm called HyperGene to integrate microarray gene expressions and protein-protein interactions for cancer outcome prediction and biomarker identification. HyperGene is a robust two-step iterative method that alternatively finds the optimal outcome prediction and the optimal weighting of the marker genes guided by a protein-protein interaction network. Under the hypothesis that cancer-related genes tend to interact with each other, the HyperGene algorithm uses a protein-protein interaction network as prior knowledge by imposing a consistent weighting of interacting genes. Our experimental results on two large-scale breast cancer gene expression datasets show that HyperGene utilizing a curated protein-protein interaction network achieves significantly improved cancer outcome prediction. Moreover, HyperGene can also retrieve many known cancer genes as highly weighted marker genes.

Collaboration


Dive into the Rui Kuang's collaboration.

Top Co-Authors

Avatar

Tae Hyun Hwang

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Wei Zhang

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Ze Tian

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Huanan Zhang

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Jianying Gu

City University of New York

View shared research outputs
Top Co-Authors

Avatar

Vipin Kumar

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Yufeng Wang

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Baolin Wu

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Hong Cai

University of Texas at San Antonio

View shared research outputs
Researchain Logo
Decentralizing Knowledge