• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 13
  • 1
  • Tagged with
  • 16
  • 16
  • 4
  • 4
  • 4
  • 4
  • 3
  • 3
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

T cell clonality in coeliac disease and enteropathy associated T cell lymphoma

Murray, Anna January 1994 (has links)
No description available.
2

The role luteinizing hormone in Alzheimer Disease

Webber, Kate M. January 2007 (has links)
Thesis (Ph. D.)--Case Western Reserve University, 2006. / [School of Medicine] Department of Pathology. Includes bibliographical references. Available online via OhioLINK's ETD Center.
3

Differential expression of an endogenous retrovirus in MAIDS susceptible (C57BL/6) versus resistant (BALB/C) mice /

Socha,Amanda L. January 2006 (has links) (PDF)
Undergraduate honors paper--Mount Holyoke College, 2006. Dept. of Biological Sciences. / Includes bibliographical references (leaves 108-111).
4

CD8⁺ T-cell response potential, as determined by expression of the high affinity interleukin-2 receptor, in murine AIDS /

Krauchunas, Amber R. M. January 2006 (has links) (PDF)
Undergraduate honors paper--Mount Holyoke College, 2006. Dept. of Biological Sciences. / Includes bibliographical references (leaves 66-68).
5

Network Based Prioritization of Disease Genes

Erten, Mehmet Sinan January 2009 (has links)
No description available.
6

Comparative and integrative genomic approach toward disease gene identification: application to Bardet-Biedle Syndrome

Chiang, Annie Pei-Fen 01 January 2006 (has links)
The identification of disease genes (genes that when mutated cause human diseases) is an important and challenging problem. Proper diagnosis, prevention, as well as care for patients require an understanding of disease pathophysiology, which is best understood when the underlying causative gene(s) or genetic element(s) are identified. While the availability of the sequenced human genome helped to lead to the discovery of more than 1,900 disease genes, the rate of disease gene discovery is still occurring at a slow pace. The use of genetic linkage methods have successfully led to the identification of numerous disease genes. However, linkage studies are ultimately restricted by available meioses (clinical samples) which result in numerous candidate disease genes. This thesis addresses candidate gene prioritizations in disease gene discovery as applied toward a genetically heterogeneous disease known as Bardet-Biedl Syndrome (BBS). Specifically, the integration of various functional information and the development of a novel comparative genomic approach (Computational Orthologous Prioritization - COP) that led to the identification of BBS3 and BBS11. Functional data integration and application of the COP method may be helpful toward the identification of other disease genes.
7

Identifying protein complexes and disease genes from biomolecular networks

2014 November 1900 (has links)
With advances in high-throughput measurement techniques, large-scale biological data, such as protein-protein interaction (PPI) data, gene expression data, gene-disease association data, cellular pathway data, and so on, have been and will continue to be produced. Those data contain insightful information for understanding the mechanisms of biological systems and have been proved useful for developing new methods in disease diagnosis, disease treatment and drug design. This study focuses on two main research topics: (1) identifying protein complexes and (2) identifying disease genes from biomolecular networks. Firstly, protein complexes are groups of proteins that interact with each other at the same time and place within living cells. They are molecular entities that carry out cellular processes. The identification of protein complexes plays a primary role for understanding the organization of proteins and the mechanisms of biological systems. Many previous algorithms are designed based on the assumption that protein complexes are densely connected sub-graphs in PPI networks. In this research, a dense sub-graph detection algorithm is first developed following this assumption by using clique seeds and graph entropy. Although the proposed algorithm generates a large number of reasonable predictions and its f-score is better than many previous algorithms, it still cannot identify many known protein complexes. After that, we analyze characteristics of known yeast protein complexes and find that not all of the complexes exhibit dense structures in PPI networks. Many of them have a star-like structure, which is a very special case of the core-attachment structure and it cannot be identified by many previous core-attachment-structure-based algorithms. To increase the prediction accuracy of protein complex identification, a multiple-topological-structure-based algorithm is proposed to identify protein complexes from PPI networks. Four single-topological-structure-based algorithms are first employed to detect raw predictions with clique, dense, core-attachment and star-like structures, respectively. A merging and trimming step is then adopted to generate final predictions based on topological information or GO annotations of predictions. A comprehensive review about the identification of protein complexes from static PPI networks to dynamic PPI networks is also given in this study. Secondly, genetic diseases often involve the dysfunction of multiple genes. Various types of evidence have shown that similar disease genes tend to lie close to one another in various biomolecular networks. The identification of disease genes via multiple data integration is indispensable towards the understanding of the genetic mechanisms of many genetic diseases. However, the number of known disease genes related to similar genetic diseases is often small. It is not easy to capture the intricate gene-disease associations from such a small number of known samples. Moreover, different kinds of biological data are heterogeneous and no widely acceptable criterion is available to standardize them to the same scale. In this study, a flexible and reliable multiple data integration algorithm is first proposed to identify disease genes based on the theory of Markov random fields (MRF) and the method of Bayesian analysis. A novel global-characteristic-based parameter estimation method and an improved Gibbs sampling strategy are introduced, such that the proposed algorithm has the capability to tune parameters of different data sources automatically. However, the Markovianity characteristic of the proposed algorithm means it only considers information of direct neighbors to formulate the relationship among genes, ignoring the contribution of indirect neighbors in biomolecular networks. To overcome this drawback, a kernel-based MRF algorithm is further proposed to take advantage of the global characteristics of biological data via graph kernels. The kernel-based MRF algorithm generates predictions better than many previous disease gene identification algorithms in terms of the area under the receiver operating characteristic curve (AUC score). However, it is very time-consuming, since the Gibbs sampling process of the algorithm has to maintain a long Markov chain for every single gene. Finally, to reduce the computational time of the MRF-based algorithm, a fast and high performance logistic-regression-based algorithm is developed for identifying disease genes from biomolecular networks. Numerical experiments show that the proposed algorithm outperforms many existing methods in terms of the AUC score and running time. To summarize, this study has developed several computational algorithms for identifying protein complexes and disease genes from biomolecular networks, respectively. These proposed algorithms are better than many other existing algorithms in the literature.
8

Association Based Prioritization of Genes

January 2011 (has links)
abstract: Genes have widely different pertinences to the etiology and pathology of diseases. Thus, they can be ranked according to their disease-significance on a genomic scale, which is the subject of gene prioritization. Given a set of genes known to be related to a disease, it is reasonable to use them as a basis to determine the significance of other candidate genes, which will then be ranked based on the association they exhibit with respect to the given set of known genes. Experimental and computational data of various kinds have different reliability and relevance to a disease under study. This work presents a gene prioritization method based on integrated biological networks that incorporates and models the various levels of relevance and reliability of diverse sources. The method is shown to achieve significantly higher performance as compared to two well-known gene prioritization algorithms. Essentially, no bias in the performance was seen as it was applied to diseases of diverse ethnology, e.g., monogenic, polygenic and cancer. The method was highly stable and robust against significant levels of noise in the data. Biological networks are often sparse, which can impede the operation of associationbased gene prioritization algorithms such as the one presented here from a computational perspective. As a potential approach to overcome this limitation, we explore the value that transcription factor binding sites can have in elucidating suitable targets. Transcription factors are needed for the expression of most genes, especially in higher organisms and hence genes can be associated via their genetic regulatory properties. While each transcription factor recognizes specific DNA sequence patterns, such patterns are mostly unknown for many transcription factors. Even those that are known are inconsistently reported in the literature, implying a potentially high level of inaccuracy. We developed computational methods for prediction and improvement of transcription factor binding patterns. Tests performed on the improvement method by employing synthetic patterns under various conditions showed that the method is very robust and the patterns produced invariably converge to nearly identical series of patterns. Preliminary tests were conducted to incorporate knowledge from transcription factor binding sites into our networkbased model for prioritization, with encouraging results. Genes have widely different pertinences to the etiology and pathology of diseases. Thus, they can be ranked according to their disease-significance on a genomic scale, which is the subject of gene prioritization. Given a set of genes known to be related to a disease, it is reasonable to use them as a basis to determine the significance of other candidate genes, which will then be ranked based on the association they exhibit with respect to the given set of known genes. Experimental and computational data of various kinds have different reliability and relevance to a disease under study. This work presents a gene prioritization method based on integrated biological networks that incorporates and models the various levels of relevance and reliability of diverse sources. The method is shown to achieve significantly higher performance as compared to two well-known gene prioritization algorithms. Essentially, no bias in the performance was seen as it was applied to diseases of diverse ethnology, e.g., monogenic, polygenic and cancer. The method was highly stable and robust against significant levels of noise in the data. Biological networks are often sparse, which can impede the operation of associationbased gene prioritization algorithms such as the one presented here from a computational perspective. As a potential approach to overcome this limitation, we explore the value that transcription factor binding sites can have in elucidating suitable targets. Transcription factors are needed for the expression of most genes, especially in higher organisms and hence genes can be associated via their genetic regulatory properties. While each transcription factor recognizes specific DNA sequence patterns, such patterns are mostly unknown for many transcription factors. Even those that are known are inconsistently reported in the literature, implying a potentially high level of inaccuracy. We developed computational methods for prediction and improvement of transcription factor binding patterns. Tests performed on the improvement method by employing synthetic patterns under various conditions showed that the method is very robust and the patterns produced invariably converge to nearly identical series of patterns. Preliminary tests were conducted to incorporate knowledge from transcription factor binding sites into our networkbased model for prioritization, with encouraging results. To validate these approaches in a disease-specific context, we built a schizophreniaspecific network based on the inferred associations and performed a comprehensive prioritization of human genes with respect to the disease. These results are expected to be validated empirically, but computational validation using known targets are very positive. / Dissertation/Thesis / Ph.D. Computer Science 2011
9

Neural Inductive Matrix Completion for Predicting Disease-Gene Associations

Hou, Siqing 21 May 2018 (has links)
In silico prioritization of undiscovered associations can help find causal genes of newly discovered diseases. Some existing methods are based on known associations, and side information of diseases and genes. We exploit the possibility of using a neural network model, Neural inductive matrix completion (NIMC), in disease-gene prediction. Comparing to the state-of-the-art inductive matrix completion method, using neural networks allows us to learn latent features from non-linear functions of input features. Previous methods use disease features only from mining text. Comparing to text mining, disease ontology is a more informative way of discovering correlation of dis- eases, from which we can calculate the similarities between diseases and help increase the performance of predicting disease-gene associations. We compare the proposed method with other state-of-the-art methods for pre- dicting associated genes for diseases from the Online Mendelian Inheritance in Man (OMIM) database. Results show that both new features and the proposed NIMC model can improve the chance of recovering an unknown associated gene in the top 100 predicted genes. Best results are obtained by using both the new features and the new model. Results also show the proposed method does better in predicting associated genes for newly discovered diseases.
10

A Multimodal Graph Convolutional Approach to Predict Genes Associated with Rare Genetic Diseases

Sahasrabudhe, Dhruva Shrikrishna 11 September 2020 (has links)
There exist a large number of rare genetic diseases in humans. Our knowledge of the specific gene variants whose presence in the genome of a person predisposes them towards developing a disease, called gene associations, is incomplete. Computational tools which can predict genes which may be associated with a rare disease have great utility in healthcare. However, a majority of existing prediction algorithms require a set of already known "seed genes'' to further discover novel associations for a disease. This drawback becomes more serious for rare genetic diseases, since a large proportion do not have any known gene associations. In this work, we develop an approach for disease-gene association prediction that overcomes the reliance on seed genes. Our approach uses the similarity of the observable biological characteristics of diseases (i.e., phenotypes) along with a global map of direct and indirect human protein interactions, to transfer associations from diseases whose gene associations have been discovered to diseases with no known gene associations. We formulate disease-gene association prediction over a multimodal network of diseases and genes, and develop an approach based on graph convolutional networks. We show how our model design considerations impact prediction performance. We demonstrate that our approach outperforms simpler graph machine learning and traditional machine learning approaches, as well as a competitive network propagation based approach for the task of predicting disease-gene associations. / Master of Science / There exist a large number of rare genetic diseases in humans. Our knowledge of the specific gene variants whose presence in the genome of a person predisposes them towards developing a disease, called gene associations, is incomplete. Computational tools which can predict genes which may be associated with a rare disease have great utility in healthcare. However, a majority of existing prediction algorithms require a set of already known "seed genes'' to further discover novel associations for a disease. This drawback becomes more serious for rare genetic diseases, since a large proportion do not have any known gene associations. In this work, we develop an approach for disease-gene association prediction that overcomes the reliance on seed genes. Our approach uses the similarity of the observable biological characteristics of diseases (i.e. disease phenotypes) along with a global map of direct and indirect human protein interactions, to transfer gene associations from diseases whose gene associations have been discovered, to diseases with no known associations. We implement an approach based on the field of graph machine learning, namely graph convolutional networks, to predict the genes associated with rare genetic diseases. We show how our predictor performs, compared to other approaches, and analyze some of the choices made in the design of the predictor, along with some properties of the outputs of our predictor.

Page generated in 0.2419 seconds