Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Methods Mol Biol ; 1159: 109-33, 2014.
Article in English | MEDLINE | ID: mdl-24788264

ABSTRACT

The field of biomarker research has experienced a major boost in recent years, and the number of publications on biomarker studies evaluating given, but also proposing novel biomarker candidates is increasing rapidly for numerous clinically relevant disease areas. However, individual markers often lack sensitivity and specificity in the clinical context, resting essentially on the intra-individual phenotype variability hampering sensitivity, or on assessing more general processes downstream of the causative molecular events characterizing a disease term, in consequence impairing disease specificity. The trend to circumvent these shortcomings goes towards utilizing multimarker panels, thus combining the strength of individual markers to further enhance performance regarding both sensitivity and specificity. A way of identifying the optimal composition of individual markers in a panel approach is to pick each marker as representative for a specific pathophysiological (mechanistic) process relevant for the disease under investigation, hence resulting in a multimarker panel for covering the set of pathophysiological processes underlying the frequently multifactorial composition of a clinical phenotype.Here we outline a procedure of identifying such sets of disease-specific pathophysiological processes (units) delineated on the basis of disease-associated molecular feature lists derived from literature mining as well as aggregated, publicly available Omics profiling experiments. With such molecular units in hand, providing an improved reflection of a specific clinical phenotype, biomarker candidates can then be assigned to or novel candidates are to be selected from these units, subsequently resulting in a multimarker panel promising improved accuracy in disease diagnosis as well as prognosis.


Subject(s)
Biological Ontologies , Biomarkers/metabolism , Data Mining/methods , Knowledge Discovery/methods , Animals , Humans
2.
Electrophoresis ; 34(11): 1649-56, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23494759

ABSTRACT

Molecular profiling techniques have provided extensive sets of molecular features characterizing clinical phenotypes, but further extrapolation to mechanistic molecular models of disease pathophysiology faces major challenges. Here, we describe a computational procedure for delineating molecular disease models utilizing omics profiles, and exemplify the methodology on aspects of the cardiorenal syndrome describing the clinical association of declining kidney function and increased cardiovascular event rates. Individual molecular features as well as selected molecular processes were identified as linking cardiovascular and renal pathology as a combination of cross-organ mediators and common pathophysiology. The molecular characterization of the disease presents as a set of molecular processes together with their interactions, composing a molecular disease model of the cardiorenal syndrome. Integrating omics profiles describing aspects of cardiovascular disease and respective profiles for advanced chronic kidney disease on molecular interaction networks, computation of disease term-specific subgraphs, and complemented by subgraph segmentation allowed delineation of disease term-specific molecular models, at their intersection providing contributors to cardiorenal pathology. Building such molecular disease models allows in a generic way to integrate multi-omics sources for generating comprehensive sets of molecular processes, on such basis providing rationale for biomarker panel selection for further characterizing clinical phenotypes.


Subject(s)
Cardio-Renal Syndrome/physiopathology , Computational Biology/methods , Heart/physiopathology , Kidney/physiopathology , Cardio-Renal Syndrome/genetics , Cardio-Renal Syndrome/metabolism , Humans , Kidney/metabolism , Models, Molecular , Myocardium/metabolism , Myocardium/pathology
3.
Mol Biosyst ; 8(12): 3197-207, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23014771

ABSTRACT

Systematic study of the effect of mycophenolate mofetil (MMF) on the molecular level in the context of other drugs and molecular disease profiles became possible due to the availability of large scale molecular profiles on both disease characterization and drug mode of action. Such analysis is of particular value in elucidating alternative drug use for addressing clinically unmet needs, and the concept of synthetic lethality provides an alternative tool for such repositioning strategies. Resting on consolidation of transcriptomics data and literature mining, a MMF molecular footprint became available including a set of 170 genes specifically affected by the drug. Analysis of this profile on a molecular pathway level reveals a set of 14 pathways as affected. Next to assignment of molecular pathways and associated diseases synergistic drug combinations are proposed by utilizing the synthetic lethal interaction network. Of particular interest is the combination of MMF with adenosine deaminase inhibitors, sulfasalazine, and other selected drugs interfering with calcium-based regulatory pathways and metabolism. Indeed analysis of drugs in clinical trials positively identifies combinations with MMF in the context of synthetic lethality and affected pathways, particularly in diseases such as multiple sclerosis, vasculitis, GVHD and lupus nephritis. Importantly, the synthetic lethal interaction of the drug mode of action is an interesting basis for rational repositioning strategies by suggesting combinations which exhibit a synergistic rather than a mere additive effect, as for example is evident for the combination of tacrolimus and MMF. Inherent is also the assessment of possible adverse effects of drug combinations.


Subject(s)
Drug Interactions , Drug Therapy, Combination , Mycophenolic Acid/analogs & derivatives , Adenosine Deaminase Inhibitors/pharmacology , Calcium/metabolism , Drug Combinations , Drug Synergism , Humans , Immunosuppressive Agents/pharmacology , Metabolic Networks and Pathways/drug effects , Mycophenolic Acid/adverse effects , Mycophenolic Acid/pharmacology , Sulfasalazine/pharmacology , Tacrolimus/pharmacology
4.
OMICS ; 16(3): 105-12, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22401656

ABSTRACT

The risk of developing cardiovascular diseases (CVD) is dramatically increased in patients with chronic kidney diseases (CKD). Mechanisms leading to this cardiorenal syndrome (CRS) are multifactorial, and combined analyses of both failing organs may provide routes toward developing strategies for early risk assessment, prognosis, and consequently effective therapy. In order to identify molecular mechanisms involved in the crosstalk between the diseased cardiovascular system and kidney, we analyzed tissue specific transcriptomics profiles on atherosclerosis and diabetic nephropathy together with gene sets associated with cardiovascular and chronic kidney diseases that derived from a literature mining approach. We focused on enriched molecular pathways and highlight molecular interactions found within as well as between affected pathways identified for the two organs. Analysis on the level of molecular pathways pointed out the role of PPAR signaling, coagulation, inflammation, and focal adhesion pathways in formation and progression of the CRS. The proteins apolipoprotein A1 (APOA1) and albumin (ALB) turned out to be of particular importance in the context of dyslipidemia, one of the major risk factors for the development of CVD. In summary, our analyses highlight mechanisms associated with dyslipidemia, hemodynamic regulation, and inflammation on the interface between the cardiovascular and the renal system.


Subject(s)
Cardio-Renal Syndrome/metabolism , Cardiovascular Diseases/metabolism , Albumins/metabolism , Apolipoprotein A-I/metabolism , Humans , Renal Insufficiency, Chronic/metabolism
5.
Proteomics Clin Appl ; 5(5-6): 354-66, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21491608

ABSTRACT

PURPOSE: For diseases with complex phenotype such as diabetic nephropathy (DN), integration of multiple Omics sources promises an improved description of the disease pathophysiology, being the basis for novel diagnostics and therapy, but equally important personalization aspects. EXPERIMENTAL DESIGN: Molecular features on DN were retrieved from public domain Omics studies and by mining scientific literature, patent text and clinical trial specifications. Molecular feature sets were consolidated on a human protein interaction network and interpreted on the level of molecular pathways in the light of the pathophysiology of the disease and its clinical context defined as associated biomarkers and drug targets. RESULTS: About 1000 gene symbols each could be assigned to the pathophysiological description of DN and to the clinical context. Direct feature comparison showed minor overlap, whereas on the level of molecular pathways, the complement and coagulation cascade, PPAR signaling, and the renin-angiotensin system linked the disease descriptor space with biomarkers and targets. CONCLUSION AND CLINICAL RELEVANCE: Only the combined molecular feature landscapes closely reflect the clinical implications of DN in the context of hypertension and diabetes. Omics data integration on the level of interaction networks furthermore provides a platform for identification of pathway-specific biomarkers and therapy options.


Subject(s)
Computational Biology/methods , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Drug Delivery Systems , Biomarkers/metabolism , Case-Control Studies , Data Mining , Diabetic Nephropathies/diagnosis , Humans , Prognosis , Protein Interaction Mapping , Proteomics
6.
Methods Mol Biol ; 719: 379-97, 2011.
Article in English | MEDLINE | ID: mdl-21370093

ABSTRACT

Progress in experimental procedures has led to rapid availability of Omics profiles. Various open-access as well as commercial tools have been developed for storage, analysis, and interpretation of transcriptomics, proteomics, and metabolomics data. Generally, major analysis steps include data storage, retrieval, preprocessing, and normalization, followed by identification of differentially expressed features, functional annotation on the level of biological processes and molecular pathways, as well as interpretation of gene lists in the context of protein-protein interaction networks. In this chapter, we discuss a sequential transcriptomics data analysis workflow utilizing open-source tools, specifically exemplified on a gene expression dataset on familial hypercholesterolemia.


Subject(s)
Computational Biology/methods , Data Interpretation, Statistical , Databases, Genetic , Gene Expression Profiling , Humans , Hyperlipoproteinemia Type II/genetics , Internet , Molecular Sequence Annotation , Monocytes/metabolism
7.
Mol Biosyst ; 7(1): 200-14, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21031175

ABSTRACT

Chemotherapy of cancer experiences a number of shortcomings including development of drug resistance. This fact also holds true for neuroblastoma utilizing chemotherapeutics as vincristine. We performed a comparative analysis of molecular and cellular mechanisms associated with vincristine resistance utilizing cell line as well as human tissue data. Differential gene expression analysis revealed molecular features, processes and pathways afflicted with drug resistance mechanisms in general, and specifically with vincristine significantly involving actin associated features. However, specific mode of resistance as well as underlying genotype of parental, vincristine sensitive cells apparently exhibited significant heterogeneity. No consensus profile for vincristine resistance could be derived, but resistance-associated changes on the level of individual neuroblastoma cell lines as well as individual patient profiles became clearly evident. Based on these prerequisites we utilized the concept of synthetic lethality aimed at identifying hub proteins which when inhibited promise to induce cell death due to a synthetic lethal interaction with down-regulated, chemoresistance associated features. Our screening procedure identified synthetic lethal hub proteins afflicted with actin associated processes holding synthetic lethal interactions to down-regulated features individually found in all chemoresistant cell lines tested, therefore promising an improved therapeutic window. Verification of such synthetic lethal hub candidates in human neuroblastoma tissue expression profiles indicated the feasibility of this screening approach for addressing vincristine resistance in neuroblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/physiology , Neoplasm Proteins/metabolism , Neuroblastoma/metabolism , Vincristine/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Vitro Techniques , Nerve Tissue Proteins/metabolism , Polypyrimidine Tract-Binding Protein/metabolism
8.
Immunome Res ; 6 Suppl 2: S7, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-21067549

ABSTRACT

BACKGROUND: The last years have seen a renaissance of the vaccine area, driven by clinical needs in infectious diseases but also chronic diseases such as cancer and autoimmune disorders. Equally important are technological improvements involving nano-scale delivery platforms as well as third generation adjuvants. In parallel immunoinformatics routines have reached essential maturity for supporting central aspects in vaccinology going beyond prediction of antigenic determinants. On this basis computational vaccinology has emerged as a discipline aimed at ab-initio rational vaccine design.Here we present a computational workflow for implementing computational vaccinology covering aspects from vaccine target identification to functional characterization and epitope selection supported by a Systems Biology assessment of central aspects in host-pathogen interaction. We exemplify the procedures for Epstein Barr Virus (EBV), a clinically relevant pathogen causing chronic infection and suspected of triggering malignancies and autoimmune disorders. RESULTS: We introduce pBone/pView as a computational workflow supporting design and execution of immunoinformatics workflow modules, additionally involving aspects of results visualization, knowledge sharing and re-use. Specific elements of the workflow involve identification of vaccine targets in the realm of a Systems Biology assessment of host-pathogen interaction for identifying functionally relevant targets, as well as various methodologies for delineating B- and T-cell epitopes with particular emphasis on broad coverage of viral isolates as well as MHC alleles.Applying the workflow on EBV specifically proposes sequences from the viral proteins LMP2, EBNA2 and BALF4 as vaccine targets holding specific B- and T-cell epitopes promising broad strain and allele coverage. CONCLUSION: Based on advancements in the experimental assessment of genomes, transcriptomes and proteomes for both, pathogen and (human) host, the fundaments for rational design of vaccines have been laid out. In parallel, immunoinformatics modules have been designed and successfully applied for supporting specific aspects in vaccine design. Joining these advancements, further complemented by novel vaccine formulation and delivery aspects, have paved the way for implementing computational vaccinology for rational vaccine design tackling presently unmet vaccine challenges.

9.
Transplantation ; 88(3 Suppl): S14-9, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19667956

ABSTRACT

Ischemia reperfusion injury (IRI) is a choreographed process leading to delayed graft function (DGF) and reduced long-term patency of the transplanted organ. Early identification of recipients of grafts at risk would allow modification of the posttransplant management, and thereby potentially improve short- and long-term outcomes. The recently emerged "omics" technologies together with bioinformatics workup have allowed the integration and analysis of IRI-associated molecular profiles in the context of DGF. Such a systems biological approach promises qualitative information about interdependencies of complex processes such as IRI regulation, rather than offering descriptive tables of differentially regulated features on a transcriptome, proteome, or metabolome level leaking the functional, biological framework. In deceased-donor kidney transplantation as the primary causative factor resulting in IRI and DGF, a distinct signature and choreography of molecular events in the graft before harvesting seems to be associated with subsequent DGF. A systems biological assessment of these molecular changes suggests that processes along inflammation are of pivotal importance for the early stage of IRI. The causal proof of this association has been tested by a double-blinded, randomized, controlled trial of steroid or placebo infusion into deceased donors before the organs were harvested. Thorough systems biological analysis revealed a panel of biomarkers with excellent discrimination. In summary, integrated analysis of omics data has brought forward biomarker candidates and candidate panels that promise early assessment of IRI. However, the clinical utility of these markers still needs to be established in prospective trials in independent patient populations.


Subject(s)
Kidney Transplantation/immunology , Kidney Transplantation/pathology , Reperfusion Injury/immunology , Biomarkers/analysis , Cadaver , DNA, Complementary/genetics , Delayed Graft Function/genetics , Delayed Graft Function/immunology , Delayed Graft Function/pathology , Double-Blind Method , Gene Expression Profiling , Genome-Wide Association Study , Humans , Inflammation/immunology , Placebos , Proteome , Randomized Controlled Trials as Topic , Reperfusion Injury/genetics , Tissue Donors
10.
Mol Biosyst ; 5(12): 1720-31, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19585005

ABSTRACT

A central aim of differential gene expression profile analysis is to provide an interpretation of given data at the level of biological processes and pathways. However, traversing descriptive data into context is not straightforward. We present a gene-centric dependency graph approach supporting an interpretation of omics profiles at the level of affected networks. The core of our dependency graph comprises data objects encoding the functional categorization of a particular gene, its tissue-specific reference gene expression, as well as known interactions and subcellular location of assigned proteins. On the basis of these genome, transcriptome, and proteome data we compute pair-wise object (gene) dependencies and interpret them as weighted edges in a dependency graph. Mapping of omics profiles on this graph can be used to identify connectors linking features of the omics list, in turn providing the basis for identification of subgraphs and motifs characterizing the cellular state under analysis. We exemplify this approach by analyzing differential gene expression data characterizing B-cell lymphoma and demonstrate the identification of B-cell lymphoma associated subgraphs.


Subject(s)
Gene Expression Profiling/methods , Genomics/methods , Protein Interaction Mapping/methods , Databases, Genetic , Gene Expression Regulation, Neoplastic , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Metabolic Networks and Pathways/genetics , ROC Curve , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...