Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Angiogenesis ; 16(1): 159-70, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23053781

ABSTRACT

Angiogenesis is central to many physiological and pathological processes. Here we show two potent bioinformatically-identified peptides, one derived from collagen IV and translationally optimized, and one from a somatotropin domain-containing protein, synergize in angiogenesis and lymphangiogenesis assays including cell adhesion, migration and in vivo Matrigel plugs. Peptide-peptide combination therapies have recently been applied to diseases such as human immunodeficiency virus (HIV), but remain uncommon thus far in cancer, age-related macular degeneration and other angiogenesis-dependent diseases. Previous work from our group has shown that the collagen IV-derived peptide primarily binds ß1 integrins, while the receptor for the somatotropin-derived peptide remains unknown. We investigate these peptides' mechanisms of action and find both peptides affect the vascular endothelial growth factor (VEGF) pathway as well as focal adhesion kinase (FAK) by changes in phosphorylation level and total protein content. Blocking of FAK both through binding of ß1 integrins and through inhibition of VEGFR2 accounts for the synergy we observe. Since resistance through activation of multiple signaling pathways is a central problem of anti-angiogenic therapies in diseases such as cancer, we suggest that peptide combinations such as these are an approach that should be considered as a means to sustain anti-angiogenic and anti-lymphangiogenic therapy and improve efficacy of treatment.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Collagen Type IV/chemistry , Growth Hormone/chemistry , Lymphangiogenesis/drug effects , Neovascularization, Physiologic/drug effects , Peptides/pharmacology , Amino Acid Sequence , Angiogenesis Inhibitors/chemistry , Cell Adhesion/drug effects , Cell Movement/drug effects , Drug Synergism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Activation/drug effects , Focal Adhesion Kinase 1/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Models, Biological , Molecular Sequence Data , Peptides/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/metabolism , Reproducibility of Results , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
2.
Physiol Genomics ; 44(19): 915-24, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22911453

ABSTRACT

Angiogenesis is the formation of new blood vessels from pre-existing microvessels. Excessive and insufficient angiogenesis have been associated with many diseases including cancer, age-related macular degeneration, ischemic heart, brain, and skeletal muscle diseases. A comprehensive understanding of angiogenesis regulatory processes is needed to improve treatment of these diseases. To identify proteins related to angiogenesis, we developed a novel integrative framework for diverse sources of high-throughput data. The system, called GeneHits, was used to expand on known angiogenesis pathways to construct the angiome, a protein-protein interaction network for angiogenesis. The network consists of 478 proteins and 1,488 interactions. The network was validated through cross validation and analysis of five gene expression datasets from in vitro angiogenesis assays. We calculated the topological properties of the angiome. We analyzed the functional enrichment of angiogenesis-annotated and associated proteins. We also constructed an extended angiome with 1,233 proteins and 5,726 interactions to derive a more complete map of protein-protein interactions in angiogenesis. Finally, the extended angiome was used to identify growth factor signaling networks that drive angiogenesis and antiangiogenic signaling networks. The results of this analysis can be used to identify genes and proteins in different disease conditions and putative targets for therapeutic interventions as high-ranked candidates for experimental validation.


Subject(s)
Computational Biology/methods , Databases, Genetic , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Protein Interaction Mapping/methods , Protein Interaction Maps/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Microarray Analysis , Search Engine/methods , Signal Transduction/genetics , Systems Biology/methods
3.
Transl Oncol ; 5(2): 92-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22496925

ABSTRACT

Angiogenesis is the formation of neovasculature from preexisting microvessels. Several endogenous proteins regulate the balance of vessel formation and regression in the body including pigment epithelium-derived factor (PEDF), which has been shown to be antiangiogenic and to suppress tumor growth. Using sequence homology and bioinformatics, we previously identified several peptide sequences homologous to an active region of PEDF existing in multiple proteins in the human proteome. These short 11-mer peptides are found in a DEAH box helicase protein, CKIP-1 and caspase 10, and show similar activity in altering endothelial cell adhesion, migration and inducing apoptosis.We tested the peptide derived from DEAH box helicase protein in a triple-negative MDA-MB-231 breast orthotopic xenograft model in severe combined immunodeficient mice and show significant tumor suppression.

4.
BMC Bioinformatics ; 13 Suppl 3: S9, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22536907

ABSTRACT

BACKGROUND: The normal functioning of a living cell is characterized by complex interaction networks involving many different types of molecules. Associations detected between diseases and perturbations in well-defined pathways within such interaction networks have the potential to illuminate the molecular mechanisms underlying disease progression and response to treatment. RESULTS: In this paper, we present a computational method that compares expression profiles of genes in cancer samples to samples from normal tissues in order to detect perturbations of pre-defined pathways in the cancer. In contrast to many previous methods, our scoring function approach explicitly takes into account the interactions between the gene products in a pathway. Moreover, we compute the sub-pathway that has the highest score, as opposed to merely computing the score for the entire pathway. We use a permutation test to assess the statistical significance of the most perturbed sub-pathway. We apply our method to 20 pathways in the Netpath database and to the Global Cancer Map of gene expression in 18 cancers. We demonstrate that our method yields more sensitive results than alternatives that do not consider interactions or measure the perturbation of a pathway as a whole. We perform a sensitivity analysis to show that our approach is robust to modest changes in the input data. Our method confirms numerous well-known connections between pathways and cancers. CONCLUSIONS: Our results indicate that integrating differential gene expression with the interaction structure in a pathway is a powerful approach for detecting links between a cancer and the pathways perturbed in it. Our results also suggest that even well-studied pathways may be perturbed only partially in any given cancer. Further analysis of cancer-specific sub-pathways may shed new light on the similarities and differences between cancers.


Subject(s)
Algorithms , Neoplasms/genetics , Neoplasms/metabolism , Protein Interaction Maps , Signal Transduction , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans
5.
PLoS One ; 6(9): e24887, 2011.
Article in English | MEDLINE | ID: mdl-21931866

ABSTRACT

Angiogenesis is important for many physiological processes, diseases, and also regenerative medicine. Therapies that inhibit the vascular endothelial growth factor (VEGF) pathway have been used in the clinic for cancer and macular degeneration. In cancer applications, these treatments suffer from a "tumor escape phenomenon" where alternative pathways are upregulated and angiogenesis continues. The redundancy of angiogenesis regulation indicates the need for additional studies and new drug targets. We aimed to (i) identify novel and missing angiogenesis annotations and (ii) verify their significance to angiogenesis. To achieve these goals, we integrated the human interactome with known angiogenesis-annotated proteins to identify a set of 202 angiogenesis-associated proteins. Across endothelial cell lines, we found that a significant fraction of these proteins had highly perturbed gene expression during angiogenesis. After treatment with VEGF-A, we found increasing expression of HIF-1α, APP, HIV-1 tat interactive protein 2, and MEF2C, while endoglin, liprin ß1 and HIF-2α had decreasing expression across three endothelial cell lines. The analysis showed differential regulation of HIF-1α and HIF-2α. The data also provided additional evidence for the role of endothelial cells in Alzheimer's disease.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Antigens, CD/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Endoglin , Gene Expression/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MADS Domain Proteins/metabolism , MEF2 Transcription Factors , Myogenic Regulatory Factors/metabolism , Receptors, Cell Surface/metabolism , Vascular Endothelial Growth Factor A/pharmacology
6.
J Med Chem ; 54(19): 6492-500, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21866962

ABSTRACT

Angiogenesis is the growth of new blood vessels from existing vasculature. Excessive vascularization is associated with a number of diseases including cancer. Antiangiogenic therapies have the potential to stunt cancer progression. Peptides derived from type IV collagen are potent inhibitors of angiogenesis. We wanted to gain a better understanding of collagen IV structure-activity relationships using a ligand-based approach. We developed novel peptide-specific QSAR models to study the activity of the peptides in endothelial cell proliferation, migration, and adhesion inhibition assays. We found that the models produced quantitatively accurate predictions of activity and provided insight into collagen IV derived peptide structure-activity relationships.


Subject(s)
Angiogenesis Inhibitors/chemistry , Collagen Type IV/chemistry , Peptides/chemistry , Quantitative Structure-Activity Relationship , Algorithms , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/pharmacology , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Human Umbilical Vein Endothelial Cells , Humans , Ligands , Models, Molecular , Neovascularization, Physiologic/drug effects , Peptides/chemical synthesis , Peptides/pharmacology
7.
Ann Biomed Eng ; 39(8): 2213-22, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21590489

ABSTRACT

Excessive vascularization is a hallmark of many diseases including cancer, rheumatoid arthritis, diabetic nephropathy, pathologic obesity, age-related macular degeneration, and asthma. Compounds that inhibit angiogenesis represent potential therapeutics for many diseases. Karagiannis and Popel [Proc. Natl. Acad. Sci. USA 105(37):13775-13780, 2008] used a bioinformatics approach to identify more than 100 peptides with sequence homology to known angiogenesis inhibitors. The peptides could be grouped into families by the conserved domain of the proteins they were derived from. The families included type IV collagen fibrils, CXC chemokine ligands, and type I thrombospondin domain-containing proteins. The relationships between these families have received relatively little attention. To investigate these relationships, we approached the problem by placing the families of proteins in the context of the human interactome including >120,000 physical interactions among proteins, genes, and transcripts. We built on a graph theoretic approach to identify proteins that may represent conduits of crosstalk between protein families. We validated these findings by statistical analysis and analysis of a time series gene expression data set taken during angiogenesis. We identified six proteins at the center of the angiogenesis-associated network including three syndecans, MMP9, CD44, and versican. These findings shed light on the complex signaling networks that govern angiogenesis phenomena.


Subject(s)
Chemokines, CXC/metabolism , Collagen Type IV/metabolism , Models, Cardiovascular , Neovascularization, Pathologic/metabolism , Signal Transduction , Thrombospondin 1/metabolism , Animals , Humans , Hyaluronan Receptors/metabolism , Matrix Metalloproteinase 9/metabolism , Neovascularization, Pathologic/pathology , Versicans/metabolism
8.
Curr Pharm Biotechnol ; 12(8): 1101-16, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21470139

ABSTRACT

Peptides have emerged as important therapeutics that are being rigorously tested in angiogenesis-dependent diseases due to their low toxicity and high specificity. Since the discovery of endogenous proteins and protein fragments that inhibit microvessel formation (thrombospondin, endostatin) several peptides have shown promise in pre-clinical and clinical studies for cancer. Peptides have been derived from thrombospondin, collagens, chemokines, coagulation cascade proteins, growth factors, and other classes of proteins and target different receptors. Here we survey recent developments for anti-angiogenic peptides with length not exceeding 50 amino acid residues that have shown activity in pre-clinical models of cancer or have been tested in clinical trials; some of the peptides have been modified and optimized, e.g., through L-to-D and non-natural amino acid substitutions. We highlight technological advances in peptide discovery and optimization including computational and bioinformatics tools and novel experimental techniques.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/drug therapy , Peptides/therapeutic use , Amino Acid Sequence , Computational Biology , Drug Discovery , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Peptides/chemistry
9.
PLoS One ; 5(8): e12089, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20711500

ABSTRACT

BACKGROUND: Bacillus anthracis, Francisella tularensis, and Yersinia pestis are bacterial pathogens that can cause anthrax, lethal acute pneumonic disease, and bubonic plague, respectively, and are listed as NIAID Category A priority pathogens for possible use as biological weapons. However, the interactions between human proteins and proteins in these bacteria remain poorly characterized leading to an incomplete understanding of their pathogenesis and mechanisms of immune evasion. METHODOLOGY: In this study, we used a high-throughput yeast two-hybrid assay to identify physical interactions between human proteins and proteins from each of these three pathogens. From more than 250,000 screens performed, we identified 3,073 human-B. anthracis, 1,383 human-F. tularensis, and 4,059 human-Y. pestis protein-protein interactions including interactions involving 304 B. anthracis, 52 F. tularensis, and 330 Y. pestis proteins that are uncharacterized. Computational analysis revealed that pathogen proteins preferentially interact with human proteins that are hubs and bottlenecks in the human PPI network. In addition, we computed modules of human-pathogen PPIs that are conserved amongst the three networks. Functionally, such conserved modules reveal commonalities between how the different pathogens interact with crucial host pathways involved in inflammation and immunity. SIGNIFICANCE: These data constitute the first extensive protein interaction networks constructed for bacterial pathogens and their human hosts. This study provides novel insights into host-pathogen interactions.


Subject(s)
Bacillus anthracis/metabolism , Bacterial Proteins/metabolism , Computational Biology , Francisella tularensis/metabolism , Host-Pathogen Interactions , Yersinia pestis/metabolism , Bacillus anthracis/physiology , Francisella tularensis/physiology , Humans , Protein Binding , Yersinia pestis/physiology
10.
BMC Bioinformatics ; 11 Suppl 1: S61, 2010 Jan 18.
Article in English | MEDLINE | ID: mdl-20122237

ABSTRACT

BACKGROUND: As the size of the known human interactome grows, biologists increasingly rely on computational tools to identify patterns that represent protein complexes and pathways. Previous studies have shown that densely connected network components frequently correspond to community structure and functionally related modules. In this work, we present a novel method to identify densely connected and bipartite network modules based on a log odds score for shared neighbours. RESULTS: To evaluate the performance of our method (NeMo), we compare it to other widely used tools for community detection including kMetis, MCODE, and spectral clustering. We test these methods on a collection of synthetically constructed networks and the set of MIPS human complexes. We apply our method to the CXC chemokine pathway and find a high scoring functional module of 12 disconnected phospholipase isoforms. CONCLUSION: We present a novel method that combines a unique neighbour-sharing score with hierarchical agglomerative clustering to identify diverse network communities. The approach is unique in that we identify both dense network and dense bipartite network structures in a single approach. Our results suggest that the performance of NeMo is better than or competitive with leading approaches on both real and synthetic datasets. We minimize model complexity and generalization error in the Bayesian spirit by integrating out nuisance parameters. An implementation of our method is freely available for download as a plugin to Cytoscape through our website and through Cytoscape itself.


Subject(s)
Proteomics/methods , Software , Algorithms , Cluster Analysis , Databases, Protein , Protein Interaction Mapping/methods
11.
J Comput Biol ; 15(7): 829-44, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18707557

ABSTRACT

Publicly available datasets provide detailed and large-scale information on multiple types of molecular interaction networks in a number of model organisms. The wiring diagrams composed of these interaction networks capture a static view of cellular state. An important challenge in systems biology is obtaining a dynamic perspective on these networks by integrating them with gene expression measurements taken under multiple conditions. We present a top-down computational approach to identify building blocks of molecular interaction networks by: (i) integrating gene expression measurements for a particular disease state (e.g., leukemia) or experimental condition (e.g., treatment with growth serum) with molecular interactions to reveal an active network, which is the network of interactions active in the cell in that disease state or condition; and (ii) systematically combining active networks computed for different experimental conditions using set-theoretic formulae to reveal network legos, which are modules of coherently interacting genes and gene products in the wiring diagram. We propose efficient methods to compute active networks, systematically mine candidate legos, assess the statistical significance of these candidates, arrange them in a directed acyclic graph (DAG), and exploit the structure of the DAG to identify true network legos. We describe methods to assess the stability of our computations to changes in the input and to recover active networks by composing network legos. We analyze two human datasets using our method. A comparison of three leukemias demonstrates how a biologist can use our system to identify specific differences between these diseases. A larger-scale analysis of 13 distinct stresses illustrates our ability to compute the building blocks of the interaction networks activated in response to these stresses. Source code implementing our algorithms is available under version 2 of the GNU General Public License at http://bioinformatics.cs.vt.edu/ murali/software/network-lego.


Subject(s)
Algorithms , Cells/metabolism , Gene Regulatory Networks , Models, Biological , Protein Interaction Mapping/methods , Systems Biology/methods , Computational Biology/methods , Computer Simulation , Gene Expression Profiling/methods , Humans , Leukemia/metabolism , Leukemia/physiopathology , Oligonucleotide Array Sequence Analysis/methods
12.
Nucleic Acids Res ; 34(Web Server issue): W340-4, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16845022

ABSTRACT

Dramatic advances in sequencing technology and sophisticated experimental assays that interrogate the cell, combined with the public availability of the resulting data, herald the era of systems biology. However, the biological functions of more than 40% of the genes in sequenced genomes are unknown, posing a fundamental barrier to progress in systems biology. The large scale and diversity of available data requires the development of techniques that can automatically utilize these datasets to make quantified and robust predictions of gene function that can be experimentally verified. We present a service called the VIRtual Gene Ontology (VIRGO) that (i) constructs a functional linkage network (FLN) from gene expression and molecular interaction data, (ii) labels genes in the FLN with their functional annotations in the Gene Ontology and (iii) systematically propagates these labels across the FLN in order to precisely predict the functions of unlabelled genes. VIRGO assigns confidence estimates to predicted functions so that a biologist can prioritize predictions for further experimental study. For each prediction, VIRGO also provides an informative 'propagation diagram' that traces the flow of information in the FLN that led to the prediction. VIRGO is available at http://whipple.cs.vt.edu:8080/virgo.


Subject(s)
Computational Biology/methods , Genes/physiology , Genomics/methods , Software , Algorithms , Gene Expression , Humans , Internet , Saccharomyces cerevisiae/genetics , User-Computer Interface , Vocabulary, Controlled
SELECTION OF CITATIONS
SEARCH DETAIL
...