scispace - formally typeset
Search or ask a question
Author

Karthik Raman

Other affiliations: University of Zurich, Swiss Institute of Bioinformatics, Bosch  ...read more
Bio: Karthik Raman is an academic researcher from Indian Institute of Technology Madras. The author has contributed to research in topics: Flux balance analysis & Computer science. The author has an hindex of 20, co-authored 84 publications receiving 1822 citations. Previous affiliations of Karthik Raman include University of Zurich & Swiss Institute of Bioinformatics.


Papers
More filters
Journal ArticleDOI
TL;DR: The usefulness of FBA as a tool for gaining biological insights is reviewed, advances in methodology enabling integration of regulatory information and thermodynamic constraints are discussed, and the challenges that lie ahead are addressed.
Abstract: Systems level modelling and simulations of biological processes are proving to be invaluable in obtaining a quantitative and dynamic perspective of various aspects of cellular function. In particular, constraint-based analyses of metabolic networks have gained considerable popularity for simulating cellular metabolism, of which flux balance analysis (FBA), is most widely used. Unlike mechanistic simulations that depend on accurate kinetic data, which are scarcely available, FBA is based on the principle of conservation of mass in a network, which utilizes the stoichiometric matrix and a biologically relevant objective function to identify optimal reaction flux distributions. FBA has been used to analyse genome-scale reconstructions of several organisms; it has also been used to analyse the effect of perturbations, such as gene deletions or drug inhibitions in silico. This article reviews the usefulness of FBA as a tool for gaining biological insights, advances in methodology enabling integration of regulatory information and thermodynamic constraints, and finally addresses the challenges that lie ahead. Various use scenarios and biological insights obtained from FBA, and applications in fields such metabolic engineering and drug target identification, are also discussed. Genome-scale constraint-based models have an immense potential for building and testing hypotheses, as well as to guide experimentation.

381 citations

Journal ArticleDOI
TL;DR: A comprehensive in silico target identification pipeline, targetTB, for Mycobacterium tuberculosis, which provides rational schema for drug target identification that are likely to have high rates of success, which is expected to save enormous amounts of money, resources and time in the drug discovery process.
Abstract: Background: Tuberculosis still remains one of the largest killer infectious diseases, warranting the identification of newer targets and drugs. Identification and validation of appropriate targets for designing drugs are critical steps in drug discovery, which are at present major bottle-necks. A majority of drugs in current clinical use for many diseases have been designed without the knowledge of the targets, perhaps because standard methodologies to identify such targets in a high-throughput fashion do not really exist. With different kinds of 'omics' data that are now available, computational approaches can be powerful means of obtaining short-lists of possible targets for further experimental validation. Results: We report a comprehensive in silico target identification pipeline, targetTB, for Mycobacterium tuberculosis. The pipeline incorporates a network analysis of the protein-protein interactome, a flux balance analysis of the reactome, experimentally derived phenotype essentiality data, sequence analyses and a structural assessment of targetability, using novel algorithms recently developed by us. Using flux balance analysis and network analysis, proteins critical for survival of M. tuberculosis are first identified, followed by comparative genomics with the host, finally incorporating a novel structural analysis of the binding sites to assess the feasibility of a protein as a target. Further analyses include correlation with expression data and non-similarity to gut flora proteins as well as 'anti-targets' in the host, leading to the identification of 451 high-confidence targets. Through phylogenetic profiling against 228 pathogen genomes, shortlisted targets have been further explored to identify broad-spectrum antibiotic targets, while also identifying those specific to tuberculosis. Targets that address mycobacterial persistence and drug resistance mechanisms are also analysed. Conclusion: The pipeline developed provides rational schema for drug target identification that are likely to have high rates of success, which is expected to save enormous amounts of money, resources and time in the drug discovery process. A thorough comparison with previously suggested targets in the literature demonstrates the usefulness of the integrated approach used in our study, highlighting the importance of systems-level analyses in particular. The method has the potential to be used as a general strategy for target identification and validation and hence significantly impact most drug discovery programmes.

243 citations

Journal ArticleDOI
Sarah M. Keating1, Sarah M. Keating2, Dagmar Waltemath3, Matthias König4, Fengkai Zhang5, Andreas Dräger6, Claudine Chaouiya7, Claudine Chaouiya8, Frank Bergmann2, Andrew Finney9, Colin S. Gillespie10, Tomáš Helikar11, Stefan Hoops12, Rahuman S Malik-Sheriff, Stuart L. Moodie, Ion I. Moraru13, Chris J. Myers14, Aurélien Naldi15, Brett G. Olivier1, Brett G. Olivier2, Brett G. Olivier16, Sven Sahle2, James C. Schaff, Lucian P. Smith17, Lucian P. Smith1, Maciej J. Swat, Denis Thieffry15, Leandro Watanabe14, Darren J. Wilkinson10, Darren J. Wilkinson18, Michael L. Blinov13, Kimberly Begley1, James R. Faeder19, Harold F. Gómez20, Thomas M. Hamm6, Yuichiro Inagaki, Wolfram Liebermeister21, Allyson L. Lister22, Daniel Lucio23, Eric Mjolsness24, Carole J. Proctor10, Karthik Raman25, Nicolas Rodriguez26, Clifford A. Shaffer27, Bruce E. Shapiro28, Joerg Stelling20, Neil Swainston29, Naoki Tanimura, John Wagner30, Martin Meier-Schellersheim5, Herbert M. Sauro17, Bernhard O. Palsson31, Hamid Bolouri32, Hiroaki Kitano33, Akira Funahashi34, Henning Hermjakob, John Doyle1, Michael Hucka1, Richard R. Adams, Nicholas Alexander Allen35, Bastian R. Angermann5, Marco Antoniotti36, Gary D. Bader37, Jan Červený38, Mélanie Courtot, Christopher Cox39, Piero Dalle Pezze26, Emek Demir40, William S. Denney, Harish Dharuri41, Julien Dorier, Dirk Drasdo, Ali Ebrahim31, Johannes Eichner, Johan Elf42, Lukas Endler, Chris T. Evelo43, Christoph Flamm44, Ronan M. T. Fleming45, Martina Fröhlich, Mihai Glont, Emanuel Gonçalves46, Martin Golebiewski47, Hovakim Grabski48, Alex Gutteridge, Damon Hachmeister, Leonard A. Harris, Benjamin D. Heavner, Ron Henkel, William S. Hlavacek1, Bin Hu49, Daniel R. Hyduke50, Hidde de Jong, Nick Juty46, Peter D. Karp, Jonathan R. Karr51, Douglas B. Kell52, Roland Keller6, Ilya Kiselev53, Steffen Klamt54, Edda Klipp54, Christian Knüpfer55, Fedor A. Kolpakov, Falko Krause4, Martina Kutmon, Camille Laibe46, Conor Lawless7, Lu Li56, Leslie M. Loew10, Rainer Machné27, Yukiko Matsuoka, Pedro Mendes, Huaiyu Mi57, Florian Mittag2, Pedro T. Monteiro7, Kedar Nath Natarajan, Poul M. F. Nielsen17, Tramy Nguyen, Alida Palmisano58, Jean-Baptiste Pettit14, Thomas Pfau10, Robert Phair13, Tomas Radivoyevitch1, Johann M. Rohwer59, Oliver A. Ruebenacker60, Julio Saez-Rodriguez6, Martin Scharm61, Henning Schmidt47, Falk Schreiber48, Michael Schubert, Roman Schulte24, Stuart C. Sealfon10, Kieran Smallbone, Sylvain Soliman, Melanie I. Stefan1, Devin P. Sullivan28, Koichi Takahashi50, Bas Teusink, David Tolnay1, Ibrahim Vazirabad30, Axel von Kamp54, Ulrike Wittig52, Clemens Wrzodek6, Finja Wrzodek6, Ioannis Xenarios, Anna Zhukova, Jeremy Zucker62 
California Institute of Technology1, Heidelberg University2, University of Greifswald3, Humboldt University of Berlin4, National Institutes of Health5, University of Tübingen6, Instituto Gulbenkian de Ciência7, Aix-Marseille University8, Ansys9, Newcastle University10, University of Nebraska–Lincoln11, University of Virginia12, University of Connecticut13, University of Utah14, PSL Research University15, VU University Amsterdam16, University of Washington17, The Turing Institute18, University of Pittsburgh19, ETH Zurich20, Université Paris-Saclay21, University of Oxford22, North Carolina State University23, University of California, Irvine24, Indian Institute of Technology Madras25, Babraham Institute26, Virginia Tech27, California State University, Northridge28, University of Liverpool29, IBM30, University of California, San Diego31, Virginia Mason Medical Center32, Okinawa Institute of Science and Technology33, Keio University34, Amazon.com35, University of Milan36, University of Toronto37, Masaryk University38, University of Tennessee39, Oregon Health & Science University40, Illumina41, Uppsala University42, Maastricht University43, Alpen-Adria-Universität Klagenfurt44, Medical University of Vienna45, European Bioinformatics Institute46, University of Rostock47, Leibniz Association48, Lorentz Institute49, Shinshu University50, Icahn School of Medicine at Mount Sinai51, Heidelberg Institute for Theoretical Studies52, Greifswald University Hospital53, Max Planck Society54, University of Jena55, École Polytechnique56, University of Southern California57, École Normale Supérieure58, Stellenbosch University59, École Polytechnique Fédérale de Lausanne60, Mizuho Information & Research Institute61, Pacific Northwest National Laboratory62
TL;DR: The latest edition of the Systems Biology Markup Language (SBML) is reviewed, a format designed for this purpose that leverages two decades of SBML and a rich software ecosystem that transformed how systems biologists build and interact with models.
Abstract: Systems biology has experienced dramatic growth in the number, size, and complexity of computational models. To reproduce simulation results and reuse models, researchers must exchange unambiguous model descriptions. We review the latest edition of the Systems Biology Markup Language (SBML), a format designed for this purpose. A community of modelers and software authors developed SBML Level 3 over the past decade. Its modular form consists of a core suited to representing reaction-based models and packages that extend the core with features suited to other model types including constraint-based models, reaction-diffusion models, logical network models, and rule-based models. The format leverages two decades of SBML and a rich software ecosystem that transformed how systems biologists build and interact with models. More recently, the rise of multiscale models of whole cells and organs, and new data sources such as single-cell measurements and live imaging, has precipitated new ways of integrating data with models. We provide our perspectives on the challenges presented by these developments and how SBML Level 3 provides the foundation needed to support this evolution.

176 citations

Journal ArticleDOI
TL;DR: A comprehensive model of mycolic acid synthesis in the pathogen M. tuberculosis involving 197 metabolites participating in 219 reactions catalysed by 28 proteins is presented, demonstrating the application of FBA for rational identification of potential anti-tubercular drug targets, which can indeed be a general strategy in drug design.
Abstract: Mycobacterium tuberculosis is the focus of several investigations for design of newer drugs, as tuberculosis remains a major epidemic despite the availability of several drugs and a vaccine. Mycobacteria owe many of their unique qualities to mycolic acids, which are known to be important for their growth, survival, and pathogenicity. Mycolic acid biosynthesis has therefore been the focus of a number of biochemical and genetic studies. It also turns out to be the pathway inhibited by front-line anti-tubercular drugs such as isoniazid and ethionamide. Recent years have seen the emergence of systems-based methodologies that can be used to study microbial metabolism. Here, we seek to apply insights from flux balance analyses of the mycolic acid pathway (MAP) for the identification of anti-tubercular drug targets. We present a comprehensive model of mycolic acid synthesis in the pathogen M. tuberculosis involving 197 metabolites participating in 219 reactions catalysed by 28 proteins. Flux balance analysis (FBA) has been performed on the MAP model, which has provided insights into the metabolic capabilities of the pathway. In silico systematic gene deletions and inhibition of InhA by isoniazid, studied here, provide clues about proteins essential for the pathway and hence lead to a rational identification of possible drug targets. Feasibility studies using sequence analysis of the M. tuberculosis H37Rv and human proteomes indicate that, apart from the known InhA, potential targets for anti-tubercular drug design are AccD3, Fas, FabH, Pks13, DesA1/2, and DesA3. Proteins identified as essential by FBA correlate well with those previously identified experimentally through transposon site hybridisation mutagenesis. This study demonstrates the application of FBA for rational identification of potential anti-tubercular drug targets, which can indeed be a general strategy in drug design. The targets, chosen based on the critical points in the pathway, form a ready shortlist for experimental testing.

151 citations

Journal ArticleDOI
TL;DR: An introduction to network theory is presented, followed by a discussion of the parameters commonly used in analysing networks, important network topologies, as well as methods to identify important network components, based on perturbations.
Abstract: Protein–protein interactions form the basis for a vast majority of cellular events, including signal transduction and transcriptional regulation. It is now understood that the study of interactions between cellular macromolecules is fundamental to the understanding of biological systems. Interactions between proteins have been studied through a number of high-throughput experiments and have also been predicted through an array of computational methods that leverage the vast amount of sequence data generated in the last decade. In this review, I discuss some of the important computational methods for the prediction of functional linkages between proteins. I then give a brief overview of some of the databases and tools that are useful for a study of protein–protein interactions. I also present an introduction to network theory, followed by a discussion of the parameters commonly used in analysing networks, important network topologies, as well as methods to identify important network components, based on perturbations.

144 citations


Cited by
More filters
Journal ArticleDOI
TL;DR: The constraint-based reconstruction and analysis toolbox as discussed by the authors is a software package running in the Matlab environment, which allows for quantitative prediction of cellular behavior using a constraintbased approach and allows predictive computations of both steady-state and dynamic optimal growth behavior, the effects of gene deletions, comprehensive robustness analyses, sampling the range of possible cellular metabolic states and the determination of network modules.
Abstract: The manner in which microorganisms utilize their metabolic processes can be predicted using constraint-based analysis of genome-scale metabolic networks. Herein, we present the constraint-based reconstruction and analysis toolbox, a software package running in the Matlab environment, which allows for quantitative prediction of cellular behavior using a constraint-based approach. Specifically, this software allows predictive computations of both steady-state and dynamic optimal growth behavior, the effects of gene deletions, comprehensive robustness analyses, sampling the range of possible cellular metabolic states and the determination of network modules. Functions enabling these calculations are included in the toolbox, allowing a user to input a genome-scale metabolic model distributed in Systems Biology Markup Language format and perform these calculations with just a few lines of code. The results are predictions of cellular behavior that have been verified as accurate in a growing body of research. After software installation, calculation time is minimal, allowing the user to focus on the interpretation of the computational results.

1,827 citations

Journal Article
TL;DR: Why interactome networks are important to consider in biology, how they can be mapped and integrated with each other, what global properties are starting to emerge from interactome network models, and how these properties may relate to human disease are detailed.
Abstract: Complex biological systems and cellular networks may underlie most genotype to phenotype relationships. Here, we review basic concepts in network biology, discussing different types of interactome networks and the insights that can come from analyzing them. We elaborate on why interactome networks are important to consider in biology, how they can be mapped and integrated with each other, what global properties are starting to emerge from interactome network models, and how these properties may relate to human disease.

1,323 citations

Journal ArticleDOI
TL;DR: The biology of latent tuberculosis is discussed as part of a broad range of responses that occur following infection with Mycobacterium tuberculosis, which result in the formation of physiologically distinct granulomatous lesions that provide microenvironments with differential ability to support or suppress the persistence of viable bacteria.
Abstract: Immunological tests provide evidence of latent tuberculosis in one third of the global population, which corresponds to more than two billion individuals. Latent tuberculosis is defined by the absence of clinical symptoms but carries a risk of subsequent progression to clinical disease, particularly in the context of co-infection with HIV. In this Review we discuss the biology of latent tuberculosis as part of a broad range of responses that occur following infection with Mycobacterium tuberculosis, which result in the formation of physiologically distinct granulomatous lesions that provide microenvironments with differential ability to support or suppress the persistence of viable bacteria. We then show how this model can be used to develop a rational programme to discover effective drugs for the eradication of M. tuberculosis infection.

1,254 citations

Journal ArticleDOI
TL;DR: This review examines the many uses and future directions of genome‐scale metabolic reconstructions, and highlights trends and opportunities in the field that will make the greatest impact on many fields of biology.
Abstract: The availability and utility of genome-scale metabolic reconstructions have exploded since the first genome-scale reconstruction was published a decade ago. Reconstructions have now been built for a wide variety of organisms, and have been used toward five major ends: (1) contextualization of high-throughput data, (2) guidance of metabolic engineering, (3) directing hypothesis-driven discovery, (4) interrogation of multi-species relationships, and (5) network property discovery. In this review, we examine the many uses and future directions of genome-scale metabolic reconstructions, and we highlight trends and opportunities in the field that will make the greatest impact on many fields of biology.

839 citations

Journal ArticleDOI
TL;DR: It is shown how network techniques can help in the identification of single-target, edgetic, multi-target and allo-network drug target candidates and an optimized protocol of network-aided drug development is suggested, and a list of systems-level hallmarks of drug quality is provided.

806 citations