1
|
Hassan J, Saeed SM, Deka L, Uddin MJ, Das DB. Applications of Machine Learning (ML) and Mathematical Modeling (MM) in Healthcare with Special Focus on Cancer Prognosis and Anticancer Therapy: Current Status and Challenges. Pharmaceutics 2024; 16:260. [PMID: 38399314 PMCID: PMC10892549 DOI: 10.3390/pharmaceutics16020260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
The use of data-driven high-throughput analytical techniques, which has given rise to computational oncology, is undisputed. The widespread use of machine learning (ML) and mathematical modeling (MM)-based techniques is widely acknowledged. These two approaches have fueled the advancement in cancer research and eventually led to the uptake of telemedicine in cancer care. For diagnostic, prognostic, and treatment purposes concerning different types of cancer research, vast databases of varied information with manifold dimensions are required, and indeed, all this information can only be managed by an automated system developed utilizing ML and MM. In addition, MM is being used to probe the relationship between the pharmacokinetics and pharmacodynamics (PK/PD interactions) of anti-cancer substances to improve cancer treatment, and also to refine the quality of existing treatment models by being incorporated at all steps of research and development related to cancer and in routine patient care. This review will serve as a consolidation of the advancement and benefits of ML and MM techniques with a special focus on the area of cancer prognosis and anticancer therapy, leading to the identification of challenges (data quantity, ethical consideration, and data privacy) which are yet to be fully addressed in current studies.
Collapse
Affiliation(s)
- Jasmin Hassan
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (S.M.S.)
| | | | - Lipika Deka
- Faculty of Computing, Engineering and Media, De Montfort University, Leicester LE1 9BH, UK;
| | - Md Jasim Uddin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Diganta B. Das
- Department of Chemical Engineering, Loughborough University, Loughborough LE11 3TU, UK
| |
Collapse
|
2
|
Konsman JP. Expanding the notion of mechanism to further understanding of biopsychosocial disorders? Depression and medically-unexplained pain as cases in point. STUDIES IN HISTORY AND PHILOSOPHY OF SCIENCE 2024; 103:123-136. [PMID: 38157672 DOI: 10.1016/j.shpsa.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/24/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024]
Abstract
Evidence-Based Medicine has little consideration for mechanisms and philosophers of science and medicine have recently made pleas to increase the place of mechanisms in the medical evidence hierarchy. However, in this debate the notions of mechanisms seem to be limited to 'mechanistic processes' and 'complex-systems mechanisms,' understood as 'componential causal systems'. I believe that this will not do full justice to how mechanisms are used in biological, psychological and social sciences and, consequently, in a more biopsychosocial approach to medicine. Here, I propose, following (Kuorikoski, 2009), to pay more attention to 'abstract forms of interaction' mechanisms. The present work scrutinized review articles on depression and medically unexplained pain, which are considered to be of multifactorial pathogenesis, for their use of mechanisms. In review articles on these disorders there seemed to be a range of uses between more 'abstract forms of interaction' and 'componential causal system' mechanisms. I therefore propose to expand the notions of mechanisms considered in medicine to include that of more 'abstract forms of interaction' to better explain and manage biopsychosocial disorders.
Collapse
Affiliation(s)
- Jan Pieter Konsman
- ImmunoConcEpT, CNRS UMR 5164, University of Bordeaux, 33076, Bordeaux, France.
| |
Collapse
|
3
|
Roberts JA, Varma VR, Candia J, Tanaka T, Ferrucci L, Bennett DA, Thambisetty M. Unbiased proteomics and multivariable regularized regression techniques identify SMOC1, NOG, APCS, and NTN1 in an Alzheimer's disease brain proteomic signature. NPJ AGING 2023; 9:18. [PMID: 37414805 PMCID: PMC10326005 DOI: 10.1038/s41514-023-00112-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 05/18/2023] [Indexed: 07/08/2023]
Abstract
Advancements in omics methodologies have generated a wealth of high-dimensional Alzheimer's disease (AD) datasets, creating significant opportunities and challenges for data interpretation. In this study, we utilized multivariable regularized regression techniques to identify a reduced set of proteins that could discriminate between AD and cognitively normal (CN) brain samples. Utilizing eNetXplorer, an R package that tests the accuracy and significance of a family of elastic net generalized linear models, we identified 4 proteins (SMOC1, NOG, APCS, NTN1) that accurately discriminated between AD (n = 31) and CN (n = 22) middle frontal gyrus (MFG) tissue samples from Religious Orders Study participants with 83 percent accuracy. We then validated this signature in MFG samples from Baltimore Longitudinal Study of Aging participants using leave-one-out logistic regression cross-validation, finding that the signature again accurately discriminated AD (n = 31) and CN (n = 19) participants with a receiver operating characteristic curve area under the curve of 0.863. These proteins were strongly correlated with the burden of neurofibrillary tangle and amyloid pathology in both study cohorts. We additionally tested whether these proteins differed between AD and CN inferior temporal gyrus (ITG) samples and blood serum samples at the time of AD diagnosis in ROS and BLSA, finding that the proteins differed between AD and CN ITG samples but not in blood serum samples. The identified proteins may provide mechanistic insights into the pathophysiology of AD, and the methods utilized in this study may serve as the basis for further work with additional high-dimensional datasets in AD.
Collapse
Affiliation(s)
- Jackson A Roberts
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA.
| | - Vijay R Varma
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Julián Candia
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Toshiko Tanaka
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
4
|
Araujo R, Brumley D, Cursons J, Day K, Faria M, Flegg JA, Germano D, Hunt H, Hunter P, Jenner A, Johnston S, McCaw JM, Maini P, Miller C, Muskovic W, Osborne J, Pan M, Rajagopal V, Shahidi N, Siekmann I, Stumpf M, Zanca A. Frontiers of Mathematical Biology: A workshop honouring Professor Edmund Crampin. Math Biosci 2023; 359:109007. [PMID: 37062447 DOI: 10.1016/j.mbs.2023.109007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/28/2023] [Accepted: 04/02/2023] [Indexed: 04/18/2023]
Affiliation(s)
- Robyn Araujo
- School of Mathematical Sciences, Queensland University of Technology, Australia
| | - Douglas Brumley
- School of Mathematics and Statistics, The University of Melbourne, Australia
| | | | - Karen Day
- Bio21 Institute, The University of Melbourne, Australia
| | - Matthew Faria
- Department of Biomedical Engineering, The University of Melbourne, Australia
| | - Jennifer A Flegg
- School of Mathematics and Statistics, The University of Melbourne, Australia
| | - Domenic Germano
- School of Mathematics and Statistics, The University of Melbourne, Australia
| | - Hilary Hunt
- Department of Biology, University of Oxford, United Kingdom
| | - Peter Hunter
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - Adrianne Jenner
- School of Mathematical Sciences, Queensland University of Technology, Australia
| | - Stuart Johnston
- School of Mathematics and Statistics, The University of Melbourne, Australia
| | - James M McCaw
- School of Mathematics and Statistics, The University of Melbourne, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Australia.
| | - Philip Maini
- Mathematical Institute, University of Oxford, United Kingdom
| | - Claire Miller
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | | | - James Osborne
- School of Mathematics and Statistics, The University of Melbourne, Australia
| | - Michael Pan
- School of Mathematics and Statistics, The University of Melbourne, Australia
| | - Vijay Rajagopal
- Department of Biomedical Engineering, The University of Melbourne, Australia
| | - Niloofar Shahidi
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - Ivo Siekmann
- School of Computer Science and Mathematics, Liverpool John Moores University, United Kingdom
| | - Michael Stumpf
- School of Mathematics and Statistics, The University of Melbourne, Australia; Melbourne Integrative Genomics, The University of Melbourne, Australia
| | - Adriana Zanca
- School of Mathematics and Statistics, The University of Melbourne, Australia
| |
Collapse
|
5
|
Jeynes-Smith C, Araujo RP. Protein-protein complexes can undermine ultrasensitivity-dependent biological adaptation. J R Soc Interface 2023; 20:20220553. [PMID: 36596458 PMCID: PMC9810431 DOI: 10.1098/rsif.2022.0553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 12/09/2022] [Indexed: 01/05/2023] Open
Abstract
Robust perfect adaptation (RPA) is a ubiquitously observed signalling response across all scales of biological organization. A major class of network architectures that drive RPA in complex networks is the Opposer module-a feedback-regulated network into which specialized integral-computing 'opposer node(s)' are embedded. Although ultrasensitivity-generating chemical reactions have long been considered a possible mechanism for such adaptation-conferring opposer nodes, this hypothesis has relied on simplified Michaelian models, which neglect the presence of protein-protein complexes. Here we develop complex-complete models of interlinked covalent-modification cycles with embedded ultrasensitivity, explicitly capturing all molecular interactions and protein complexes. Strikingly, we demonstrate that the presence of protein-protein complexes thwarts the network's capacity for RPA in any 'free' active protein form, conferring RPA capacity instead on the concentration of a larger protein pool consisting of two distinct forms of a single protein. We further show that the presence of enzyme-substrate complexes, even at comparatively low concentrations, play a crucial and previously unrecognized role in controlling the RPA response-significantly reducing the range of network inputs for which RPA can obtain, and imposing greater parametric requirements on the RPA response. These surprising results raise fundamental new questions as to the biochemical requirements for adaptation-conferring Opposer modules within complex cellular networks.
Collapse
Affiliation(s)
- C. Jeynes-Smith
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia
| | - R. P. Araujo
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
6
|
Araujo RP, Liotta LA. Design Principles Underlying Robust Adaptation of Complex Biochemical Networks. Methods Mol Biol 2023; 2634:3-32. [PMID: 37074572 DOI: 10.1007/978-1-0716-3008-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
Biochemical networks are often characterized by tremendous complexity-both in terms of the sheer number of interacting molecules ("nodes") and in terms of the varied and incompletely understood interactions among these molecules ("interconnections" or "edges"). Strikingly, the vast and intricate networks of interacting proteins that exist within each living cell have the capacity to perform remarkably robustly, and reproducibly, despite significant variations in concentrations of the interacting components from one cell to the next and despite mutability over time of biochemical parameters. Here we consider the ubiquitously observed and fundamentally important signalling response known as robust perfect adaptation (RPA). We have recently shown that all RPA-capable networks, even the most complex ones, must satisfy an extremely rigid set of design principles, and are modular, being decomposable into just two types of network building-blocks-opposer modules and balancer modules. Here we present an overview of the design principles that characterize all RPA-capable network topologies through a detailed examination of a collection of simple examples. We also introduce a diagrammatic method for studying the potential of a network to exhibit RPA, which may be applied without a detailed knowledge of the complex mathematical principles governing RPA.
Collapse
Affiliation(s)
- Robyn P Araujo
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.
- Institute of Health and Biomedical Innovation (IHBI), Kelvin Grove, QLD, Australia.
| | - Lance A Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| |
Collapse
|
7
|
Xue Z, Zeng J, Li Y, Meng B, Gong X, Zhao Y, Dai X. Proteomics reveals that cell density could affect the efficacy of drug treatment. Biochem Biophys Rep 2022; 33:101403. [PMID: 36561432 PMCID: PMC9763681 DOI: 10.1016/j.bbrep.2022.101403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
In vitro cell biology study plays a fundamental role in biological and drug development research, but the repeatability and accuracy of cell studies remain to be low. Various uncertainties during the cell culture process could introduce bias into drug research. In this study, we evaluate the potential effects and underlying mechanisms induced by cell number differences in the cell seeding process. Normally, drug experiments are initiated 24 h after cell seeding, and the difference in the cell number at the time of inoculation leads to the difference in cell confluence (cell density) when drug research is conducted. While cell confluence is closely related to intercellular communication, surface protein interaction, cell autocrine as well as paracrine protein expression of cells, it might have a potential impact on the effect of biological studies such as drug treatment. This study used proteomics technology to comprehensively explore the different protein expression patterns between cells with different confluences. Due to the high sensitivity and high throughput of liquid chromatography-mass spectrometry (LC-MS/MS) detection, it was hired to evaluate the protein expression differences of Hep3B cells with 3 different confluences (30%, 50%, and 70%). The differential expressed proteins were analyzed by the Reactome pathway and the Gene Ontology (GO) pathway. Significant differences were identified across three confluences in terms of the number of proteins identified, the protein expression pattern, and the expression level of certain KEGG pathways. We found that those proteins involved in the cell cycle pathway were differently expressed: the higher the cell confluence, the higher these proteins expressed. A cell cycle inhibitor palbociclib was selected to further verify this observation. Palbociclib in the same dose was applied to cells with different confluence, the results indicated that the growth inhibition effect of palbociclib increases along with the increasing trend of cell cycle protein expression. The result indicated that cell density did influence the effect of drug treatment. Furthermore, three other drugs, cisplatin, paclitaxel, and imatinib, were used to treat the three liver cancer cell lines Hep3B, SUN387, and MHCC97, and a similar observation was obtained that drug effect would be different when the cell confluences were different. Therefore, selecting an appropriate number of cells for plating is vitally important at the beginning of a drug study.
Collapse
Affiliation(s)
- Zhichao Xue
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, PR China
| | - Jiaming Zeng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, PR China,Shenyang University of Chemical Technology, College of Chemical Engineering, Shenyang, 110142, PR China
| | - Yongshu Li
- Shenzhen Institute for Technology Innovation, National Institute of Metrology, Shenzhen, 518055, PR China
| | - Bo Meng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, PR China
| | - Xiaoyun Gong
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, PR China
| | - Yang Zhao
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, PR China
| | - Xinhua Dai
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, PR China,Corresponding author.
| |
Collapse
|
8
|
Stožer A, Šterk M, Paradiž Leitgeb E, Markovič R, Skelin Klemen M, Ellis CE, Križančić Bombek L, Dolenšek J, MacDonald PE, Gosak M. From Isles of Königsberg to Islets of Langerhans: Examining the Function of the Endocrine Pancreas Through Network Science. Front Endocrinol (Lausanne) 2022; 13:922640. [PMID: 35784543 PMCID: PMC9240343 DOI: 10.3389/fendo.2022.922640] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
Islets of Langerhans are multicellular microorgans located in the pancreas that play a central role in whole-body energy homeostasis. Through secretion of insulin and other hormones they regulate postprandial storage and interprandial usage of energy-rich nutrients. In these clusters of hormone-secreting endocrine cells, intricate cell-cell communication is essential for proper function. Electrical coupling between the insulin-secreting beta cells through gap junctions composed of connexin36 is particularly important, as it provides the required, most important, basis for coordinated responses of the beta cell population. The increasing evidence that gap-junctional communication and its modulation are vital to well-regulated secretion of insulin has stimulated immense interest in how subpopulations of heterogeneous beta cells are functionally arranged throughout the islets and how they mediate intercellular signals. In the last decade, several novel techniques have been proposed to assess cooperation between cells in islets, including the prosperous combination of multicellular imaging and network science. In the present contribution, we review recent advances related to the application of complex network approaches to uncover the functional connectivity patterns among cells within the islets. We first provide an accessible introduction to the basic principles of network theory, enumerating the measures characterizing the intercellular interactions and quantifying the functional integration and segregation of a multicellular system. Then we describe methodological approaches to construct functional beta cell networks, point out possible pitfalls, and specify the functional implications of beta cell network examinations. We continue by highlighting the recent findings obtained through advanced multicellular imaging techniques supported by network-based analyses, giving special emphasis to the current developments in both mouse and human islets, as well as outlining challenges offered by the multilayer network formalism in exploring the collective activity of islet cell populations. Finally, we emphasize that the combination of these imaging techniques and network-based analyses does not only represent an innovative concept that can be used to describe and interpret the physiology of islets, but also provides fertile ground for delineating normal from pathological function and for quantifying the changes in islet communication networks associated with the development of diabetes mellitus.
Collapse
Affiliation(s)
- Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Marko Šterk
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Eva Paradiž Leitgeb
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Rene Markovič
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Institute of Mathematics and Physics, Faculty of Electrical Engineering and Computer Science, University of Maribor, Maribor, Slovenia
| | - Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Cara E. Ellis
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | | | - Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Patrick E. MacDonald
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Marko Gosak
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| |
Collapse
|
9
|
Estrada AC, Irons L, Rego BV, Li G, Tellides G, Humphrey JD. Roles of mTOR in thoracic aortopathy understood by complex intracellular signaling interactions. PLoS Comput Biol 2021; 17:e1009683. [PMID: 34898595 PMCID: PMC8700007 DOI: 10.1371/journal.pcbi.1009683] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/23/2021] [Accepted: 11/26/2021] [Indexed: 02/01/2023] Open
Abstract
Thoracic aortopathy–aneurysm, dissection, and rupture–is increasingly responsible for significant morbidity and mortality. Advances in medical genetics and imaging have improved diagnosis and thus enabled earlier prophylactic surgical intervention in many cases. There remains a pressing need, however, to understand better the underlying molecular and cellular mechanisms with the hope of finding robust pharmacotherapies. Diverse studies in patients and mouse models of aortopathy have revealed critical changes in multiple smooth muscle cell signaling pathways that associate with disease, yet integrating information across studies and models has remained challenging. We present a new quantitative network model that includes many of the key smooth muscle cell signaling pathways and validate the model using a detailed data set that focuses on hyperactivation of the mechanistic target of rapamycin (mTOR) pathway and its inhibition using rapamycin. We show that the model can be parameterized to capture the primary experimental findings both qualitatively and quantitatively. We further show that simulating a population of cells by varying receptor reaction weights leads to distinct proteomic clusters within the population, and that these clusters emerge due to a bistable switch driven by positive feedback in the PI3K/AKT/mTOR signaling pathway. Cell signaling drives changes across scales, from altered transcription at the single-cell level to tissue-level growth and remodeling. Studying complex interactions within cell signaling pathways can lead to a better understanding of the progression of disease. In particular, we are interested in how vascular cells can change their phenotype in a way that exacerbates aortopathy, namely, the development of aneurysms, dissections, and rupture. In this study we built a novel cell signaling network model of a vascular smooth muscle cell using archival data and used it to capture the effects of a genetic knock-out and subsequent pharmacologic rescue. We then used the model to simulate populations of smooth muscle cells and found that small perturbations to the strength of signaling can lead to distinct clusters of cells. With further analysis of the network substructures, we found that a positive feedback loop within the network was responsible for the distinct phenotypes we saw in our clusters of simulated cells. We believe that this work not only helps us to understand changes in smooth muscle cell phenotype but also opens the possibility to study other signaling perturbations associated with aortopathy.
Collapse
Affiliation(s)
- Ana C. Estrada
- Department of Biomedical Engineering, Yale University; New Haven, Connecticut, United States of America
| | - Linda Irons
- Department of Biomedical Engineering, Yale University; New Haven, Connecticut, United States of America
| | - Bruno V. Rego
- Department of Biomedical Engineering, Yale University; New Haven, Connecticut, United States of America
| | - Guangxin Li
- Department of Surgery, Yale School of Medicine; New Haven, Connecticut, United States of America
| | - George Tellides
- Department of Surgery, Yale School of Medicine; New Haven, Connecticut, United States of America
- Vascular Biology and Therapeutics Program, Yale School of Medicine; New Haven, Connecticut, United States of America
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University; New Haven, Connecticut, United States of America
- Vascular Biology and Therapeutics Program, Yale School of Medicine; New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
10
|
Jeynes-Smith C, Araujo RP. Ultrasensitivity and bistability in covalent-modification cycles with positive autoregulation. Proc Math Phys Eng Sci 2021; 477:20210069. [PMID: 35153570 PMCID: PMC8331239 DOI: 10.1098/rspa.2021.0069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 07/02/2021] [Indexed: 12/17/2022] Open
Abstract
Switch-like behaviours in biochemical networks are of fundamental significance in biological signal processing, and exist as two distinct types: ultra-sensitivity and bistability. Here we propose two new models of a reversible covalent-modification cycle with positive autoregulation (PAR), a motif structure that is thought to be capable of both ultrasensitivity and bistability in different parameter regimes. These new models appeal to a modelling framework that we call complex-complete, which accounts fully for the molecular complexities of the underlying signalling mechanisms. Each of the two new models encodes a specific molecular mechanism for PAR. We demonstrate that the modelling simplifications for PAR models that have been used in previous work, which rely on Michaelian approximations, are unable to accurately recapitulate the qualitative signalling responses supported by our detailed models. Strikingly, we show that complex-complete PAR models are capable of new qualitative responses such as one-way switches and a 'prozone' effect, depending on the specific PAR-encoding mechanism, which are not supported by Michaelian simplifications. Our results highlight the critical importance of accurately representing the molecular details of biochemical signalling mechanisms, and strongly suggest that the Michaelian approximation is inadequate for predictive models of enzyme-mediated chemical reactions with added regulations such as PAR.
Collapse
Affiliation(s)
- Cailan Jeynes-Smith
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia
- Institute of Health and Biomedical Innovation (IHBI), Brisbane, Australia
| | - Robyn P. Araujo
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia
- Institute of Health and Biomedical Innovation (IHBI), Brisbane, Australia
| |
Collapse
|
11
|
Sadria M, Layton AT. Interactions among mTORC, AMPK and SIRT: a computational model for cell energy balance and metabolism. Cell Commun Signal 2021; 19:57. [PMID: 34016143 PMCID: PMC8135154 DOI: 10.1186/s12964-021-00706-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/11/2021] [Indexed: 12/26/2022] Open
Abstract
Background Cells adapt their metabolism and activities in response to signals from their surroundings, and this ability is essential for their survival in the face of perturbations. In tissues a deficit of these mechanisms is commonly associated with cellular aging and diseases, such as cardiovascular disease, cancer, immune system decline, and neurological pathologies. Several proteins have been identified as being able to respond directly to energy, nutrient, and growth factor levels and stress stimuli in order to mediate adaptations in the cell. In particular, mTOR, AMPK, and sirtuins are known to play an essential role in the management of metabolic stress and energy balance in mammals. Methods To understand the complex interactions of these signalling pathways and environmental signals, and how those interactions may impact lifespan and health-span, we have developed a computational model of metabolic signalling pathways. Specifically, the model includes (i) the insulin/IGF-1 pathway, which couples energy and nutrient abundance to the execution of cell growth and division, (ii) mTORC1 and the amino acid sensors such as sestrin, (iii) the Preiss-Handler and salvage pathways, which regulate the metabolism of NAD+ and the NAD+ -consuming factor SIRT1, (iv) the energy sensor AMPK, and (v) transcription factors FOXO and PGC-1α. Results The model simulates the interactions among key regulators such as AKT, mTORC1, AMPK, NAD+ , and SIRT, and predicts their dynamics. Key findings include the clinically important role of PRAS40 and diet in mTORC1 inhibition, and a potential link between SIRT1-activating compounds and premature autophagy. Moreover, the model captures the exquisite interactions of leucine, sestrin2, and arginine, and the resulting signal to the mTORC1 pathway. These results can be leveraged in the development of novel treatment of cancers and other diseases. Conclusions This study presents a state-of-the-art computational model for investigating the interactions among signaling pathways and environmental stimuli in growth, ageing, metabolism, and diseases. The model can be used as an essential component to simulate gene manipulation, therapies (e.g., rapamycin and wortmannin), calorie restrictions, and chronic stress, and assess their functional implications on longevity and ageing‐related diseases. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-021-00706-1.
Collapse
Affiliation(s)
- Mehrshad Sadria
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON, Canada.
| | - Anita T Layton
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON, Canada.,Department of Biology, Cheriton School of Computer Science, and School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
12
|
Maqbool T, Hadi F, Razzaq S, Naz S, Aftab S, Khurshid S, Awan SJ, Nawaz A, Abid F, Malik A. Synergistic Effect of Barbadensis miller and Marsdenia Condurango Extracts Induces Apoptosis Promotes Oxidative Stress by Limiting Proliferation of Cervical Cancer and Liver Cancer Cells. Asian Pac J Cancer Prev 2021; 22:843-852. [PMID: 33773549 PMCID: PMC8286677 DOI: 10.31557/apjcp.2021.22.3.843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Indexed: 12/04/2022] Open
Abstract
Background: Drug synergy is the combine effect of drug efficacy. Synergistic combinations of active ingredients have proven to be highly effective and more useful in therapeutics. In contrast, the individual effect of drug is usually undesirable and mostly used for selecting drug-resistant mutations. Purpose of this study was to check synergistic effects of both plants (Barbadensis miller and Marsdenia condurango) against liver and cervical cancer. Methodology: Culturing of HeLa (cervical cancer cell line) and HepG2 (liver cancer cell line) cells, IC50 evaluation, viability assays (trypan blue, crystal violet), p53 ELISA and immunocytochemistry, MUSE analysis (count and viability), antioxidants (GSH, SOD, CAT), at the end RT-PCR was performed. Results: IC50 evaluation was done of each plant individually and with combination for synergistic effects, IC50 with plants combination (synergism) was applied on further viability assays (trypan blue, crystal violet, MUSE analysis via count and viability kit) p53 ELISA and immunocytochemistry for evaluation of cellular apoptosis, antioxidants assays (GSH, SOD, CAT), and RT-PCR with proliferative and apoptotic markers along with internal control. Conclusion: According to current study it was observed that synergistic effect of these plants has more anticancer properties with minimum effective dose. It was also observed that extracts possess the ability to induce apoptosis, restrict proliferation and enhanced oxidative stress.
Collapse
Affiliation(s)
- Tahir Maqbool
- Faculty of Pharmacy, University of Lahore, Defence Road, Lahore, Pakistan
| | - Faheem Hadi
- Faculty of Pharmacy, University of Lahore, Defence Road, Lahore, Pakistan
| | - Sehrish Razzaq
- Faculty of Pharmacy, University of Lahore, Defence Road, Lahore, Pakistan
| | - Sadia Naz
- Faculty of Pharmacy, University of Lahore, Defence Road, Lahore, Pakistan
| | - Saira Aftab
- Faculty of Pharmacy, University of Lahore, Defence Road, Lahore, Pakistan
| | - Sameera Khurshid
- Faculty of Pharmacy, University of Lahore, Defence Road, Lahore, Pakistan
| | - Sana Javaid Awan
- Faculty of Pharmacy, University of Lahore, Defence Road, Lahore, Pakistan
| | - Aisha Nawaz
- Faculty of Pharmacy, University of Lahore, Defence Road, Lahore, Pakistan
| | - Farah Abid
- Faculty of Pharmacy, University of Lahore, Defence Road, Lahore, Pakistan
| | - Arif Malik
- Faculty of Pharmacy, University of Lahore, Defence Road, Lahore, Pakistan
| |
Collapse
|
13
|
Singh P, Kumar A. Deciphering the function of unknown Leishmania donovani cytosolic proteins using hyperparameter-tuned random forest. NETWORK MODELING ANALYSIS IN HEALTH INFORMATICS AND BIOINFORMATICS 2020; 9:2. [DOI: 10.1007/s13721-019-0208-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/02/2019] [Accepted: 11/22/2019] [Indexed: 08/30/2023]
|
14
|
Wang Y, Jia R, Tan W. The molecular mechanism behind protein kinase B natural mutant E17K affecting the allosteric inhibitor sensitivity: a molecular dynamics simulation study. J Biomol Struct Dyn 2020; 39:3158-3171. [PMID: 32452271 DOI: 10.1080/07391102.2020.1769731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glu17Lys (E17K) is one of the natural variants of Akt1, which is associated with multiple human cancers. This mutation is also indicated to affect the sensitivity of certain allosteric inhibitors. In order to explain the molecular mechanism that E17K mutation of Akt1 affects the sensitivity of allosteric inhibitors, we performed molecular dynamics simulations on Akt1 to its allosteric inhibitors for both wild type and E17K. We analyzed the simulated data in terms of structural stability, hydrogen bond formation, π-π interactions, binding free energy etc. We found that E17K substitution will affect the interaction of K297 residues with allosteric inhibitors, which was a key residue in allosteric inhibitors binding. This will eventually lead to allosteric inhibitors leaving the binding site in the E17K system. Our results can provide a theoretical basis for the design of novel allosteric inhibitors targeting E17K mutants in the future.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, People's Republic of China
| | - Ran Jia
- Institute of Theoretical Chemistry, Jilin University, Changchun, People's Republic of China
| | - Wen Tan
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, People's Republic of China
| |
Collapse
|
15
|
Abstract
Background For treating a complex disease such as cancer, some effective means are needed to control biological networks that underlies the disease. The one-target one-drug paradigm has been the dominating drug discovery approach in the past decades. Compared to single target-based drugs, combination drug targets may overcome many limitations of single drug target and achieve a more effective and safer control of the disease. Most of existing combination drug targets are developed based on clinical experience or text-and-trial strategy, which cannot provide theoretical guidelines for designing and screening effective drug combinations. Therefore, systematic identification of multiple drug targets and optimal intervention strategy needs to be developed. Results We developed a strategy to screen the synergistic combinations of two drug targets in disease networks based on the classification of single drug targets. The method tried to identify the sensitivity of single intervention and then the combination of multiple interventions that can restore the disease network to a desired normal state. In our strategy of screening drug target combinations, we first classified all drug targets into sensitive and insensitive single drug targets. Then, we identified the synergistic and antagonistic of drug target combinations, including the combinations of sensitive drug targets, the combinations of insensitive drug target and the combination of sensitive and insensitive targets. Finally, we applied our strategy to Arachidonic Acid (AA) metabolic network and found 18 pairs of synergistic drug target combinations, five of which have been proven to be viable by biological or medical experiments. Conclusions Different from traditional methods for judging drug synergy and antagonism, we propose the framework of how to enhance the efficiency by perturbing two sensitive targets in a combinatorial way, how to decrease the drug dose and therefore its side effect and cost by perturbing combinatorially a main sensitive target and an auxiliary insensitive target, and how to perturb two insensitive targets to realize the transition from a disease state to a healthy one which cannot be realized by perturbing each insensitive target alone. Although the idea is mainly applied to an AA metabolic network, the strategy holds for more general molecular networks such as combinatorial regulation in gene regulatory networks. Electronic supplementary material The online version of this article (10.1186/s12859-019-2730-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Min Luo
- Department of Mathematics, Shanghai University, No.99, Shangda Road, Shanghai, China
| | - Jianfeng Jiao
- Department of Mathematics, Shanghai University, No.99, Shangda Road, Shanghai, China
| | - Ruiqi Wang
- Department of Mathematics, Shanghai University, No.99, Shangda Road, Shanghai, China.
| |
Collapse
|
16
|
Sriram K. Bifurcation analysis of insulin regulated mTOR signalling pathway in cancer cells. IET Syst Biol 2018; 12:205-212. [PMID: 30259865 PMCID: PMC8687200 DOI: 10.1049/iet-syb.2018.0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/01/2018] [Accepted: 04/12/2018] [Indexed: 11/19/2022] Open
Abstract
Insulin induced mTOR signalling pathway is a complex network implicated in many types of cancers. The molecular mechanism of this pathway is highly complex and the dynamics is tightly regulated by intricate positive and negative feedback loops. In breast cancer cell lines, metformin has been shown to induce phosphorylation at specific serine sites in insulin regulated substrate of mTOR pathway that results in apoptosis over cell proliferation. The author models and performs bifurcation analysis to simulate cell proliferation and apoptosis in mTOR signalling pathway to capture the dynamics both in the presence and absence of metformin in cancer cells. Metformin is shown to negatively regulate PI3K through AMPK induced IRS1 phosphorylation and this brings about a reversal of AKT bistablity in codimension-1 bifurcation diagram from S-shaped, related to cell proliferation in the absence of drug metformin, to Z-shaped, related to apoptosis in the presence of drug metformin. The author hypothesises and explains how this negative regulation acts a circuit breaker, as a result of which mTOR network favours apoptosis of cancer cells over its proliferation. The implication of reversing the shape of bistable dynamics from S to Z or vice-versa in biological networks in general is discussed.
Collapse
Affiliation(s)
- Krishnamachari Sriram
- Centre for Computational Biology, Indraprastha Institute of Information Technology-Delhi, Okhla Phase-III, New Delhi, India.
| |
Collapse
|
17
|
Jacquier M, Kuriakose S, Bhardwaj A, Zhang Y, Shrivastav A, Portet S, Varma Shrivastav S. Investigation of Novel Regulation of N-myristoyltransferase by Mammalian Target of Rapamycin in Breast Cancer Cells. Sci Rep 2018; 8:12969. [PMID: 30154572 PMCID: PMC6113272 DOI: 10.1038/s41598-018-30447-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 07/16/2018] [Indexed: 01/02/2023] Open
Abstract
Breast cancer is the most common cancer in women worldwide. Hormone receptor breast cancers are the most common ones and, about 2 out of every 3 cases of breast cancer are estrogen receptor (ER) positive. Selective ER modulators, such as tamoxifen, are the first line of endocrine treatment of breast cancer. Despite the expression of hormone receptors some patients develop tamoxifen resistance and 50% present de novo tamoxifen resistance. Recently, we have demonstrated that activated mammalian target of rapamycin (mTOR) is positively associated with overall survival and recurrence free survival in ER positive breast cancer patients who were later treated with tamoxifen. Since altered expression of protein kinase B (PKB)/Akt in breast cancer cells affect N-myristoyltransferase 1 (NMT1) expression and activity, we investigated whether mTOR, a downstream target of PKB/Akt, regulates NMT1 in ER positive breast cancer cells (MCF7 cells). We inhibited mTOR by treating MCF7 cells with rapamycin and observed that the expression of NMT1 increased with rapamycin treatment over the period of time with a concomitant decrease in mTOR phosphorylation. We further employed mathematical modelling to investigate hitherto not known relationship of mTOR with NMT1. We report here for the first time a collection of models and data validating regulation of NMT1 by mTOR.
Collapse
Affiliation(s)
- Marine Jacquier
- Department of Mathematics, University of Manitoba, Winnipeg, Canada
| | - Shiby Kuriakose
- Department of Biology, University of Winnipeg, Winnipeg, Manitoba, Canada
| | - Apurva Bhardwaj
- Department of Biology, University of Winnipeg, Winnipeg, Manitoba, Canada
| | - Yang Zhang
- Department of Mathematics, University of Manitoba, Winnipeg, Canada
| | - Anuraag Shrivastav
- Department of Biology, University of Winnipeg, Winnipeg, Manitoba, Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Canada.,Research Institute of Hematology and Oncology, CancerCare Manitoba, Winnipeg, Manitoba, Canada
| | - Stéphanie Portet
- Department of Mathematics, University of Manitoba, Winnipeg, Canada
| | | |
Collapse
|
18
|
Araujo RP, Liotta LA. The topological requirements for robust perfect adaptation in networks of any size. Nat Commun 2018; 9:1757. [PMID: 29717141 PMCID: PMC5931626 DOI: 10.1038/s41467-018-04151-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/03/2018] [Indexed: 12/13/2022] Open
Abstract
Robustness, and the ability to function and thrive amid changing and unfavorable environments, is a fundamental requirement for living systems. Until now it has been an open question how large and complex biological networks can exhibit robust behaviors, such as perfect adaptation to a variable stimulus, since complexity is generally associated with fragility. Here we report that all networks that exhibit robust perfect adaptation (RPA) to a persistent change in stimulus are decomposable into well-defined modules, of which there exist two distinct classes. These two modular classes represent a topological basis for all RPA-capable networks, and generate the full set of topological realizations of the internal model principle for RPA in complex, self-organizing, evolvable bionetworks. This unexpected result supports the notion that evolutionary processes are empowered by simple and scalable modular design principles that promote robust performance no matter how large or complex the underlying networks become. Robust perfect adaptation (RPA), the ability of a system to return to its pre-stimulus state in the presence of a new signal, enables organisms to respond to further changes in stimuli. Here, the authors identify the modular structure of the full set of network topologies that can confer RPA on complex networks.
Collapse
Affiliation(s)
- Robyn P Araujo
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, QLD, 4000, Australia. .,Institute of Health and Biomedical Innovation (IHBI), 60 Musk Avenue, Kelvin Grove, Brisbane, QLD, 4059, Australia.
| | - Lance A Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, Manassas, Virginia, 20110, USA
| |
Collapse
|
19
|
Molecular mechanisms of detection and discrimination of dynamic signals. Sci Rep 2018; 8:2480. [PMID: 29410522 PMCID: PMC5802782 DOI: 10.1038/s41598-018-20842-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/24/2018] [Indexed: 12/25/2022] Open
Abstract
Many molecules decode not only the concentration of cellular signals, but also their temporal dynamics. However, little is known about the mechanisms that underlie the detection and discrimination of dynamic signals. We used computational modelling of the interaction of a ligand with multiple targets to investigate how kinetic and thermodynamic parameters regulate their capabilities to respond to dynamic signals. Our results demonstrated that the detection and discrimination of temporal features of signal inputs occur for reactions proceeding outside mass-action equilibrium. For these reactions, thermodynamic parameters such as affinity do not predict their outcomes. Additionally, we showed that, at non-equilibrium, the association rate constants determine the amount of product formed in reversible reactions. In contrast, the dissociation rate constants regulate the time interval required for reversible reactions to achieve equilibrium and, consequently, control their ability to detect and discriminate dynamic features of cellular signals.
Collapse
|
20
|
Varusai TM, Nguyen LK. Dynamic modelling of the mTOR signalling network reveals complex emergent behaviours conferred by DEPTOR. Sci Rep 2018; 8:643. [PMID: 29330362 PMCID: PMC5766521 DOI: 10.1038/s41598-017-18400-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/01/2017] [Indexed: 01/10/2023] Open
Abstract
The mechanistic Target of Rapamycin (mTOR) signalling network is an evolutionarily conserved network that controls key cellular processes, including cell growth and metabolism. Consisting of the major kinase complexes mTOR Complex 1 and 2 (mTORC1/2), the mTOR network harbours complex interactions and feedback loops. The DEP domain-containing mTOR-interacting protein (DEPTOR) was recently identified as an endogenous inhibitor of both mTORC1 and 2 through direct interactions, and is in turn degraded by mTORC1/2, adding an extra layer of complexity to the mTOR network. Yet, the dynamic properties of the DEPTOR-mTOR network and the roles of DEPTOR in coordinating mTORC1/2 activation dynamics have not been characterised. Using computational modelling, systems analysis and dynamic simulations we show that DEPTOR confers remarkably rich and complex dynamic behaviours to mTOR signalling, including abrupt, bistable switches, oscillations and co-existing bistable/oscillatory responses. Transitions between these distinct modes of behaviour are enabled by modulating DEPTOR expression alone. We characterise the governing conditions for the observed dynamics by elucidating the network in its vast multi-dimensional parameter space, and develop strategies to identify core network design motifs underlying these dynamics. Our findings provide new systems-level insights into the complexity of mTOR signalling contributed by DEPTOR.
Collapse
Affiliation(s)
- Thawfeek M Varusai
- European Bioinformatics Institute, EMBL-EBI, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SD, UK.,Systems Biology Ireland, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Lan K Nguyen
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, 3800, Australia. .,Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, 3800, Australia. .,Systems Biology Ireland, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
21
|
Nayak C, Chandra I, Singh P, Singh SK. Omics-Based Nanomedicine. Synth Biol (Oxf) 2018. [DOI: 10.1007/978-981-10-8693-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
22
|
Qiao Z, Shiozawa K, Kondo T. Proteomic approach toward determining the molecular background of pazopanib resistance in synovial sarcoma. Oncotarget 2017; 8:109587-109595. [PMID: 29312631 PMCID: PMC5752544 DOI: 10.18632/oncotarget.22730] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/28/2017] [Indexed: 12/13/2022] Open
Abstract
Pazopanib, a multitarget tyrosine kinase (TK) inhibitor, has been approved for treatment of soft tissue sarcoma. Elucidation of the molecular background of pazopanib resistance should lead to improved clinical outcomes in sarcomas; accordingly, we investigated this in synovial sarcoma using a proteomic approach. Pazopanib sensitivity was examined in four synovial sarcoma cell lines: SYO-1, HS-SYII, 1273/99, and YaFuSS. The 1273/99 cell line showed significantly higher IC50 values than the others for pazopanib. Expression levels of 90 TKs in the cell lines were examined by western blotting. Among these, the levels of PDGFRB, DDR1, AXL, MET, and PYK2 were higher, and those of FGFR1 and VEGFR3 were lower in the 1273/99 cell line than the other cell lines. Gene silencing analysis of the TKs upregulated in 1273/99 cells showed differing effects on cell growth: PDGFRB, MET, and PYK2 knockdown induced cell growth inhibition, whereas DDR1 and AXL knockdown did not influence cell growth. Using the PamChip peptide microarray, we found that 18 peptide substrates were highly phosphorylated in the 1273/99 cell line compared with other cell lines. Using the PhosphoNet database, we found that kinases FGFR3, RET, VEGFR1, EPHA2, EPHA4, TRKA, and SRC phosphorylated these 18 peptide substrates. Moreover, the results for overexpressed and aberrantly activated TKs in pazopanib-resistant cells showed no overlap. Taken together, our study indicates that identification of comprehensive TK profiles represents an essential approach to determining the molecular background of pazopanib resistance in synovial sarcoma.
Collapse
Affiliation(s)
- Zhiwei Qiao
- Division of Rare Cancer Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| | - Kumiko Shiozawa
- Division of Rare Cancer Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
23
|
Wang J, Guo Z, Fu Y, Wu Z, Huang C, Zheng C, Shar PA, Wang Z, Xiao W, Wang Y. Weak-binding molecules are not drugs?-toward a systematic strategy for finding effective weak-binding drugs. Brief Bioinform 2017; 18:321-332. [PMID: 26962012 DOI: 10.1093/bib/bbw018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Indexed: 12/16/2022] Open
Abstract
Designing maximally selective ligands that act on individual drug targets with high binding affinity has been the central dogma of drug discovery and development for the past two decades. However, many low-affinity drugs that aim for several targets at the same time are found more effective than the high-affinity binders when faced with complex disease conditions, such as cancers, Alzheimer's disease and cardiovascular diseases. The aim of this study was to appreciate the importance and reveal the features of weak-binding drugs and propose an integrated strategy for discovering them. Weak-binding drugs can be characterized by their high dissociation rates and transient interactions with their targets. In addition, network topologies and dynamics parameters involved in the targets of weak-binding drugs also influence the effects of the drugs. Here, we first performed a dynamics analysis for 33 elementary subgraphs to determine the desirable topology and dynamics parameters among targets. Then, by applying the elementary subgraphs to the mitogen-activated protein kinase (MAPK) pathway, several optimal target combinations were obtained. Combining drug-target interaction prediction with molecular dynamics simulation, we got two potential weak-binding drug candidates, luteolin and tanshinone IIA, acting on these targets. Further, the binding affinity of these two compounds to their targets and the anti-inflammatory effects of them were validated through in vitro experiments. In conclusion, weak-binding drugs have real opportunities for maximum efficiency and may show reduced adverse reactions, which can offer a bright and promising future for new drug discovery.
Collapse
Affiliation(s)
- Jinan Wang
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Zihu Guo
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Yingxue Fu
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Ziyin Wu
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Chao Huang
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Chunli Zheng
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Piar Ali Shar
- College of Life Science, Northwest A & F University, Yangling, Shaanxi, 712100, China; Center of Bioinformatics, Northwest A & F University, Yangling, Shaanxi, China
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, PR China
| | - Wei Xiao
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutical Process, Lianyungang, Jiangsu, China
| | - Yonghua Wang
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| |
Collapse
|
24
|
Tarca AL, Fitzgerald W, Chaemsaithong P, Xu Z, Hassan SS, Grivel J, Gomez‐Lopez N, Panaitescu B, Pacora P, Maymon E, Erez O, Margolis L, Romero R. The cytokine network in women with an asymptomatic short cervix and the risk of preterm delivery. Am J Reprod Immunol 2017; 78:e12686. [PMID: 28585708 PMCID: PMC5575567 DOI: 10.1111/aji.12686] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 03/20/2017] [Indexed: 01/06/2023] Open
Abstract
PROBLEM To characterize the amniotic fluid (AF) inflammatory-related protein (IRP) network in patients with a sonographic short cervix (SCx) and to determine its relation to early preterm delivery (ePTD). METHOD OF STUDY A retrospective cohort study included women with a SCx (≤25 mm; n=223) who had amniocentesis and were classified according to gestational age (GA) at diagnosis and delivery (ePTD <32 weeks of gestation). RESULTS (i) In women with a SCx ≤ 22 1/7 weeks, the concentration of most IRPs increased as the cervix shortened; those with ePTD had a higher rate of increase in MIP-1α, MCP-1, and IL-6 concentrations than those delivering later; and (ii) the concentration of most IRPs and the correlation between several IRP pairs were higher in the ePTD group than for those delivering later. CONCLUSION Women with a SCx at 16-22 1/7 weeks have a unique AF cytokine network that correlates with cervical length at diagnosis and GA at delivery. This network may aid in predicting ePTD.
Collapse
Affiliation(s)
- Adi L. Tarca
- Perinatology Research BranchProgram for Perinatal Research and ObstetricsDivision of Intramural ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMD, and Detroit, MIUSA
- Department of Obstetrics and GynecologyWayne State University School of MedicineDetroitMIUSA
| | - Wendy Fitzgerald
- Section on Intercellular InteractionsProgram on Physical BiologyEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMDUSA
| | - Piya Chaemsaithong
- Perinatology Research BranchProgram for Perinatal Research and ObstetricsDivision of Intramural ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMD, and Detroit, MIUSA
- Department of Obstetrics and GynecologyWayne State University School of MedicineDetroitMIUSA
| | - Zhonghui Xu
- Perinatology Research BranchProgram for Perinatal Research and ObstetricsDivision of Intramural ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMD, and Detroit, MIUSA
| | - Sonia S. Hassan
- Perinatology Research BranchProgram for Perinatal Research and ObstetricsDivision of Intramural ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMD, and Detroit, MIUSA
- Department of Obstetrics and GynecologyWayne State University School of MedicineDetroitMIUSA
| | - Jean‐Charles Grivel
- Division of Translational MedicineSidra Medical and Research CenterDohaQatar
| | - Nardhy Gomez‐Lopez
- Perinatology Research BranchProgram for Perinatal Research and ObstetricsDivision of Intramural ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMD, and Detroit, MIUSA
- Department of Obstetrics and GynecologyWayne State University School of MedicineDetroitMIUSA
- Department of ImmunologyMicrobiology and BiochemistryWayne State University School of MedicineDetroitMIUSA
| | - Bogdan Panaitescu
- Perinatology Research BranchProgram for Perinatal Research and ObstetricsDivision of Intramural ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMD, and Detroit, MIUSA
- Department of Obstetrics and GynecologyWayne State University School of MedicineDetroitMIUSA
| | - Percy Pacora
- Perinatology Research BranchProgram for Perinatal Research and ObstetricsDivision of Intramural ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMD, and Detroit, MIUSA
- Department of Obstetrics and GynecologyWayne State University School of MedicineDetroitMIUSA
| | - Eli Maymon
- Perinatology Research BranchProgram for Perinatal Research and ObstetricsDivision of Intramural ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMD, and Detroit, MIUSA
- Department of Obstetrics and GynecologyWayne State University School of MedicineDetroitMIUSA
| | - Offer Erez
- Perinatology Research BranchProgram for Perinatal Research and ObstetricsDivision of Intramural ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMD, and Detroit, MIUSA
- Department of Obstetrics and GynecologyWayne State University School of MedicineDetroitMIUSA
| | - Leonid Margolis
- Section on Intercellular InteractionsProgram on Physical BiologyEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMDUSA
| | - Roberto Romero
- Perinatology Research BranchProgram for Perinatal Research and ObstetricsDivision of Intramural ResearchEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMD, and Detroit, MIUSA
- Department of Obstetrics and GynecologyUniversity of MichiganAnn ArborMIUSA
- Department of Epidemiology and BiostatisticsMichigan State UniversityEast LansingMIUSA
- Center for Molecular Medicine and GeneticsWayne State UniversityDetroitMIUSA
| |
Collapse
|
25
|
Sulaimanov N, Klose M, Busch H, Boerries M. Understanding the mTOR signaling pathway via mathematical modeling. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2017; 9:e1379. [PMID: 28186392 PMCID: PMC5573916 DOI: 10.1002/wsbm.1379] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/09/2016] [Accepted: 12/07/2016] [Indexed: 12/12/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is a central regulatory pathway that integrates a variety of environmental cues to control cellular growth and homeostasis by intricate molecular feedbacks. In spite of extensive knowledge about its components, the molecular understanding of how these function together in space and time remains poor and there is a need for Systems Biology approaches to perform systematic analyses. In this work, we review the recent progress how the combined efforts of mathematical models and quantitative experiments shed new light on our understanding of the mTOR signaling pathway. In particular, we discuss the modeling concepts applied in mTOR signaling, the role of multiple feedbacks and the crosstalk mechanisms of mTOR with other signaling pathways. We also discuss the contribution of principles from information and network theory that have been successfully applied in dissecting design principles of the mTOR signaling network. We finally propose to classify the mTOR models in terms of the time scale and network complexity, and outline the importance of the classification toward the development of highly comprehensive and predictive models. WIREs Syst Biol Med 2017, 9:e1379. doi: 10.1002/wsbm.1379 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Nurgazy Sulaimanov
- Department of Electrical Engineering and Information TechnologyTechnische Universität DarmstadtDarmstadtGermany
- Department of BiologyTechnische Universitat DarmstadtDarmstadtGermany
| | - Martin Klose
- Systems Biology of the Cellular Microenvironment at the DKFZ Partner Site Freiburg ‐ Member of the German Cancer Consortium, Institute of Molecular Medicine and Cell ResearchAlbert‐Ludwigs‐University FreiburgFreiburgGermany and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hauke Busch
- Systems Biology of the Cellular Microenvironment at the DKFZ Partner Site Freiburg ‐ Member of the German Cancer Consortium, Institute of Molecular Medicine and Cell ResearchAlbert‐Ludwigs‐University FreiburgFreiburgGermany and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melanie Boerries
- Systems Biology of the Cellular Microenvironment at the DKFZ Partner Site Freiburg ‐ Member of the German Cancer Consortium, Institute of Molecular Medicine and Cell ResearchAlbert‐Ludwigs‐University FreiburgFreiburgGermany and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
26
|
Mukherjee S, Karolak A, Debant M, Buscaglia P, Renaudineau Y, Mignen O, Guida WC, Brooks WH. Molecular Dynamics Simulations of Membrane-Bound STIM1 to Investigate Conformational Changes during STIM1 Activation upon Calcium Release. J Chem Inf Model 2017; 57:335-344. [PMID: 28151650 DOI: 10.1021/acs.jcim.6b00475] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Calcium is involved in important intracellular processes, such as intracellular signaling from cell membrane receptors to the nucleus. Typically, calcium levels are kept at less than 100 nM in the nucleus and cytosol, but some calcium is stored in the endoplasmic reticulum (ER) lumen for rapid release to activate intracellular calcium-dependent functions. Stromal interacting molecule 1 (STIM1) plays a critical role in early sensing of changes in the ER's calcium level, especially when there is a sudden release of stored calcium from the ER. Inactive STIM1, which has a bound calcium ion, is activated upon ion release. Following activation of STIM1, there is STIM1-assisted initiation of extracellular calcium entry through channels in the cell membrane. This extracellular calcium entering the cell then amplifies intracellular calcium-dependent actions. At the end of the process, ER levels of stored calcium are reestablished. The main focus of this work was to study the conformational changes accompanying homo- or heterodimerization of STIM1. For this purpose, the ER luminal portion of STIM1 (residues 58-236), which includes the sterile alpha motif (SAM) domain plus the calcium-binding EF-hand domains 1 and 2 attached to the STIM1 transmembrane region (TM), was modeled and embedded in a virtual membrane. Next, molecular dynamics simulations were performed to study the conformational changes that take place during STIM1 activation and subsequent protein-protein interactions. Indeed, the simulations revealed exposure of residues in the EF-hand domains, which may be important for dimerization steps. Altogether, understanding conformational changes in STIM1 can help in drug discovery when targeting this key protein in intracellular calcium functions.
Collapse
Affiliation(s)
- Sreya Mukherjee
- Department of Chemistry, University of South Florida , Tampa, Florida 33620, United States
| | - Aleksandra Karolak
- Department of Chemistry, University of South Florida , Tampa, Florida 33620, United States
| | - Marjolaine Debant
- INSERM ESPRI, ERI29/EA2216 Laboratory of Immunotherapy and B Cell Pathologies, Laboratory of Immunology and Immunotherapy, CHRU Morvan, European University of Brittany , F29609 Brest, France.,Network "Ion channels and cancer-Cancéropole Grand Ouest (IC-CGO)" , F29609 Brest, France.,INSERM U1078, Brest University Medical School , F29609 Brest, France
| | - Paul Buscaglia
- Network "Ion channels and cancer-Cancéropole Grand Ouest (IC-CGO)" , F29609 Brest, France.,INSERM U1078, Brest University Medical School , F29609 Brest, France
| | - Yves Renaudineau
- INSERM ESPRI, ERI29/EA2216 Laboratory of Immunotherapy and B Cell Pathologies, Laboratory of Immunology and Immunotherapy, CHRU Morvan, European University of Brittany , F29609 Brest, France.,Network "Ion channels and cancer-Cancéropole Grand Ouest (IC-CGO)" , F29609 Brest, France
| | - Olivier Mignen
- Network "Ion channels and cancer-Cancéropole Grand Ouest (IC-CGO)" , F29609 Brest, France.,INSERM U1078, Brest University Medical School , F29609 Brest, France
| | - Wayne C Guida
- Department of Chemistry, University of South Florida , Tampa, Florida 33620, United States
| | - Wesley H Brooks
- Department of Chemistry, University of South Florida , Tampa, Florida 33620, United States
| |
Collapse
|
27
|
Jin S, Wu FX, Zou X. Domain control of nonlinear networked systems and applications to complex disease networks. ACTA ACUST UNITED AC 2017. [DOI: 10.3934/dcdsb.2017091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
28
|
Sinha AK, Singh P, Prakash A, Pal D, Dube A, Kumar A. Putative Drug and Vaccine Target Identification in Leishmania donovani Membrane Proteins Using Naïve Bayes Probabilistic Classifier. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2017; 14:204-211. [PMID: 28182549 DOI: 10.1109/tcbb.2016.2570217] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Predicting the role of protein is one of the most challenging problems. There are few approaches available for the prediction of role of unknown protein in terms of drug target or vaccine candidate. We propose here Naïve Bayes probabilistic classifier, a promising method for reliable predictions. This method is tested on the proteins identified in our mass spectrometry based membrane protemics study of Leishmania donovani parasite that causes a fatal disease (Visceral Leishmaniasis) in humans all around the world. Most of the vaccine/drug targets belonging to membrane proteins are represented as key players in the pathogenesis of Leishmania infection. Analyses of our previous results, using Naïve Bayes probabilistic classifier, indicate that this method predicts the role of unknown/hypothetical protein (as drug target/vaccine candidate) significantly with higher precision. We have employed this method in order to provide probabilistic predictions of unknown/hypothetical proteins as targets. This study reports the unknown/hypothetical proteins of Leishmania membrane fraction as a potential drug targets and vaccine candidate which is vital information for this parasite. Future molecular studies and characterization of these potent targets may produce a recombinant therapeutic/prophylactic tool against Visceral Leishmaniasis. These unknown/hypothetical proteins may open a vast research field to be exploited for novel treatment strategies.
Collapse
|
29
|
Wang M, Kussrow AK, Ocana MF, Chabot JR, Lepsy CS, Bornhop DJ, O'Hara DM. Physiologically relevant binding affinity quantification of monoclonal antibody PF-00547659 to mucosal addressin cell adhesion molecule for in vitro in vivo correlation. Br J Pharmacol 2016; 174:70-81. [PMID: 27760281 PMCID: PMC5221447 DOI: 10.1111/bph.13654] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/30/2016] [Accepted: 10/06/2016] [Indexed: 12/29/2022] Open
Abstract
Background and Purpose A monoclonal antibody (PF‐00547659) against mucosal addressin cell adhesion molecule (MAdCAM), expressed as both soluble (sMAdCAM) and trans‐membrane (mMAdCAM) target forms, showed over 30‐fold difference in antibody‐target KD between in vitro (Biacore) and clinically derived (KD,in‐vivo) values. Back‐scattering interferometry (BSI) was applied to acquire physiologically relevant KD values which were used to establish in vitro and in vivo correlation (IVIVC). Experimental Approach BSI was applied to obtain KD values between PF‐00547659 and recombinant human MAdCAM in buffer or CHO cells and endogenous MAdCAM in human serum or colon tissue. CHO cells and tissue were minimally processed to yield homogenate containing membrane vesicles and soluble proteins. A series of binding affinities in serum with various dilution factors was used to estimate both KD,in‐vivo and target concentrations; MAdCAM concentrations were also measured using LC–MS/MS. Key Results BSI measurements revealed low KD values (higher affinity) for sMAdCAM in buffer and serum, yet a 20‐fold higher KD value (lower affinity) for mMAdCAM in CHO, mMAdCAM and sMAdCAM in tissue. BSI predicted KD,in‐vivo in serum was similar to clinically derived KD,in‐vivo, and the BSI‐estimated serum sMAdCAM concentration also matched the measured concentration by LC–MS/MS. Conclusions and Implications Our results successfully demonstrated that BSI measurements of physiologically relevant KD values can be used to establish IVIVC, for PF‐00547659 to MAdCAM despite the lack of correlation when using Biacore measured KD and accurately estimates endogenous target concentrations. The application of BSI would greatly enhance successful basic pharmacological research and drug development.
Collapse
Affiliation(s)
- Mengmeng Wang
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc, Andover, MA, USA
| | - Amanda K Kussrow
- Department of Chemistry, Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | | | - Jeffrey R Chabot
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc, Andover, MA, USA
| | | | - Darryl J Bornhop
- Department of Chemistry, Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - Denise M O'Hara
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc, Andover, MA, USA
| |
Collapse
|
30
|
Chemoinformatics in the New Era: From Molecular Dynamics to Systems Dynamics. Molecules 2016; 21:71. [PMID: 26950111 PMCID: PMC6273631 DOI: 10.3390/molecules21030071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/22/2015] [Accepted: 01/05/2016] [Indexed: 11/16/2022] Open
Abstract
Chemoinformatics, due to its power in gathering information at the molecular level, has a wide array of important applications to biology, including fundamental biochemical studies and drug discovery and optimization. As modern "omics" based profiling and network based modeling and simulation techniques grow in sophistication, chemoinformatics now faces a great opportunity to include systems-level control mechanisms as one of its pillar components to extend and refine its various applications. This viewpoint article, through the example of computer aided targeting of the PI3K/Akt/mTOR pathway, outlines major steps of integrating systems dynamics simulations into molecular dynamics simulations to facilitate a higher level of chemoinformatics that would revolutionize drug lead optimization, personalized therapy, and possibly other applications.
Collapse
|
31
|
Wang G, Zhang M. Tunable ultrasensitivity: functional decoupling and biological insights. Sci Rep 2016; 6:20345. [PMID: 26847155 PMCID: PMC4742884 DOI: 10.1038/srep20345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/30/2015] [Indexed: 01/21/2023] Open
Abstract
Sensitivity has become a basic concept in biology, but much less is known about its tuning, probably because allosteric cooperativity, the best known mechanism of sensitivity, is determined by rigid conformations of interacting molecules and is thus difficult to tune. Reversible covalent modification (RCM), owing to its systems-level ingenuity, can generate concentration based, tunable sensitivity. Using a mathematical model of regulated RCM, we find sensitivity tuning can be decomposed into two orthogonal modes, which provide great insights into vital biological processes such as tissue development and cell cycle progression. We find that decoupling of the two modes of sensitivity tuning is critical to fidelity of cell fate decision; the decoupling is thus important in development. The decomposition also allows us to solve the ‘wasteful degradation conundrum’ in budding yeast cell cycle checkpoint, which further leads to discovery of a subtle but essential difference between positive feedback and double negative feedback. The latter guarantees revocability of stress-induced cell cycle arrest; while the former does not. By studying concentration conditions in the system, we extend applicability of ultrasensitivity and explain the ubiquity of reversible covalent modification.
Collapse
Affiliation(s)
- Guanyu Wang
- Department of Biology, South University of Science and Technology of China, Shenzhen, Guangdong 518055, China
| | - Mengshi Zhang
- Department of Biology, South University of Science and Technology of China, Shenzhen, Guangdong 518055, China
| |
Collapse
|
32
|
Romero R, Grivel JC, Tarca AL, Chaemsaithong P, Xu Z, Fitzgerald W, Hassan SS, Chaiworapongsa T, Margolis L. Evidence of perturbations of the cytokine network in preterm labor. Am J Obstet Gynecol 2015; 213:836.e1-836.e18. [PMID: 26232508 DOI: 10.1016/j.ajog.2015.07.037] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/26/2015] [Accepted: 07/21/2015] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Intraamniotic inflammation/infection is the only mechanism of disease with persuasive evidence of causality for spontaneous preterm labor/delivery. Previous studies about the behavior of cytokines in preterm labor have been largely based on the analysis of the behavior of each protein independently. Emerging evidence indicates that the study of biologic networks can provide insight into the pathobiology of disease and improve biomarker discovery. The goal of this study was to characterize the inflammatory-related protein network in the amniotic fluid of patients with preterm labor. STUDY DESIGN A retrospective cohort study was conducted that included women with singleton pregnancies who had spontaneous preterm labor and intact membranes (n = 135). These patients were classified according to the results of amniotic fluid culture, broad-range polymerase chain reaction coupled with electrospray ionization mass spectrometry, and amniotic fluid concentration of interleukin (IL)-6 into the following groups: (1) those without intraamniotic inflammation (n = 85), (2) those with microbial-associated intraamniotic inflammation (n = 15), and (3) those with intraamniotic inflammation without detectable bacteria (n = 35). Amniotic fluid concentrations of 33 inflammatory-related proteins were determined with the use of a multiplex bead array assay. RESULTS Patients with preterm labor and intact membranes who had microbial-associated intraamniotic inflammation had a higher amniotic fluid inflammatory-related protein concentration correlation than those without intraamniotic inflammation (113 perturbed correlations). IL-1β, IL-6, macrophage inflammatory protein (MIP)-1α, and IL-1α were the most connected nodes (highest degree) in this differential correlation network (degrees of 20, 16, 12, and 12, respectively). Patients with sterile intraamniotic inflammation had correlation patterns of inflammatory-related proteins, both increased and decreased, when compared to those without intraamniotic inflammation (50 perturbed correlations). IL-1α, MIP-1α, and IL-1β were the most connected nodes in this differential correlation network (degrees of 12, 10, and 7, respectively). There were more coordinated inflammatory-related protein concentrations in the amniotic fluid of women with microbial-associated intraamniotic inflammation than in those with sterile intraamniotic inflammation (60 perturbed correlations), with IL-4 and IL-33 having the largest number of perturbed correlations (degrees of 15 and 13, respectively). CONCLUSIONS We report for the first time an analysis of the inflammatory-related protein network in spontaneous preterm labor. Patients with preterm labor and microbial-associated intraamniotic inflammation had more coordinated amniotic fluid inflammatory-related proteins than either those with sterile intraamniotic inflammation or those without intraamniotic inflammation. The correlations were also stronger in patients with sterile intraamniotic inflammation than in those without intraamniotic inflammation. The findings herein could be of value in the development of biomarkers of preterm labor.
Collapse
|
33
|
Zahari MS, Wu X, Pinto SM, Nirujogi RS, Kim MS, Fetics B, Philip M, Barnes SR, Godfrey B, Gabrielson E, Nevo E, Pandey A. Phosphoproteomic profiling of tumor tissues identifies HSP27 Ser82 phosphorylation as a robust marker of early ischemia. Sci Rep 2015; 5:13660. [PMID: 26329039 PMCID: PMC4557083 DOI: 10.1038/srep13660] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/27/2015] [Indexed: 01/06/2023] Open
Abstract
Delays between tissue collection and tissue fixation result in ischemia and ischemia-associated changes in protein phosphorylation levels, which can misguide the examination of signaling pathway status. To identify a biomarker that serves as a reliable indicator of ischemic changes that tumor tissues undergo, we subjected harvested xenograft tumors to room temperature for 0, 2, 10 and 30 minutes before freezing in liquid nitrogen. Multiplex TMT-labeling was conducted to achieve precise quantitation, followed by TiO2 phosphopeptide enrichment and high resolution mass spectrometry profiling. LC-MS/MS analyses revealed phosphorylation level changes of a number of phosphosites in the ischemic samples. The phosphorylation of one of these sites, S82 of the heat shock protein 27 kDa (HSP27), was especially abundant and consistently upregulated in tissues with delays in freezing as short as 2 minutes. In order to eliminate effects of ischemia, we employed a novel cryogenic biopsy device which begins freezing tissues in situ before they are excised. Using this device, we showed that the upregulation of phosphorylation of S82 on HSP27 was abrogated. We thus demonstrate that our cryogenic biopsy device can eliminate ischemia-induced phosphoproteome alterations, and measurements of S82 on HSP27 can be used as a robust marker of ischemia in tissues.
Collapse
Affiliation(s)
- Muhammad Saddiq Zahari
- McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Xinyan Wu
- McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Sneha M Pinto
- Institute of Bioinformatics, International Tech Park, Bangalore, 560066 India
| | | | - Min-Sik Kim
- McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Barry Fetics
- Robin Medical, Inc., P.O. Box 2414, Baltimore, MD 21203, USA
| | - Mathew Philip
- Robin Medical, Inc., P.O. Box 2414, Baltimore, MD 21203, USA
| | - Sheri R Barnes
- Charles River Discovery Research Services, 3300 Gateway Centre Boulevard, Morrisville NC 27560
| | - Beverly Godfrey
- Charles River Discovery Research Services, 3300 Gateway Centre Boulevard, Morrisville NC 27560
| | - Edward Gabrielson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | - Erez Nevo
- Robin Medical, Inc., P.O. Box 2414, Baltimore, MD 21203, USA
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA.,Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130, USA
| |
Collapse
|
34
|
Liu J, Wang Z. Diverse array-designed modes of combination therapies in Fangjiomics. Acta Pharmacol Sin 2015; 36:680-8. [PMID: 25864646 PMCID: PMC4594182 DOI: 10.1038/aps.2014.125] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 10/30/2014] [Indexed: 12/11/2022] Open
Abstract
In line with the complexity of disease networks, diverse combination therapies have been demonstrated potential in the treatment of different patients with complex diseases in a personal combination profile. However, the identification of rational, compatible and effective drug combinations remains an ongoing challenge. Based on a holistic theory integrated with reductionism, Fangjiomics systematically develops multiple modes of array-designed combination therapies. We define diverse "magic shotgun" vertical, horizontal, focusing, siege and dynamic arrays according to different spatiotemporal distributions of hits on targets, pathways and networks. Through these multiple adaptive modes for treating complex diseases, Fangjiomics may help to identify rational drug combinations with synergistic or additive efficacy but reduced adverse side effects that reverse complex diseases by reconstructing or rewiring multiple targets, pathways and networks. Such a novel paradigm for combination therapies may allow us to achieve more precise treatments by developing phenotype-driven quantitative multi-scale modeling for rational drug combinations.
Collapse
Affiliation(s)
- Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
35
|
Vassilakopoulou M, Parisi F, Siddiqui S, England AM, Zarella ER, Anagnostou V, Kluger Y, Hicks DG, Rimm DL, Neumeister VM. Preanalytical variables and phosphoepitope expression in FFPE tissue: quantitative epitope assessment after variable cold ischemic time. J Transl Med 2015; 95:334-41. [PMID: 25418580 DOI: 10.1038/labinvest.2014.139] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 10/01/2014] [Accepted: 10/02/2014] [Indexed: 11/09/2022] Open
Abstract
Individualized targeted therapies for cancer patients require accurate and reproducible assessment of biomarkers to be able to plan treatment accordingly. Recent studies have shown highly variable effects of preanalytical variables on gene expression profiling and protein levels of different tissue types. Several publications have described protein degradation of tissue samples as a direct result of delay of formalin fixation of the tissue. Phosphorylated proteins are more labile and epitope degradation can happen within 30 min of cold ischemic time. To address this issue, we evaluated the change in antigenicity of a series of phosphoproteins in paraffin-embedded samples from breast tumors as a function of time to formalin fixation. A tissue microarray consisting of 93 breast cancer specimens with documented time-to-fixation was used to evaluate changes in antigenicity of 12 phosphoepitopes frequently used in research settings as a function of cold ischemic time. Analysis was performed in a quantitative manner using the AQUA technology for quantitative immunofluorescence. For each marker, least squares univariate linear regression was performed and confidence intervals were computed using bootstrapping. The majority of the epitopes tested revealed changes in expression levels with increasing time to formalin fixation. Some phosphorylated proteins, such as phospho-HSP27 and phospho-S6 RP, involved in post-translational modification and stress response pathways increased in expression or phosphorylation levels. Others (like phospho-AKT, phosphor-ERK1/2, phospho-Tyrosine, phospho-MET, and others) are quite labile and loss of antigenicity can be reported within 1-2 h of cold ischemic time. Therefore specimen collection should be closely monitored and subjected to quality control measures to ensure accurate measurement of these epitopes. However, a few phosphoepitopes (like phospho-JAK2 and phospho-ER) are sufficiently robust for routine usage in companion diagnostic testing.
Collapse
Affiliation(s)
| | - Fabio Parisi
- Department of Pathology, School of Medicine, Yale University, New Haven, CT, USA
| | - Summar Siddiqui
- Department of Pathology, School of Medicine, Yale University, New Haven, CT, USA
| | - Allison M England
- Department of Pathology, School of Medicine, Yale University, New Haven, CT, USA
| | - Elizabeth R Zarella
- Department of Pathology, School of Medicine, Yale University, New Haven, CT, USA
| | - Valsamo Anagnostou
- Department of Pathology, School of Medicine, Yale University, New Haven, CT, USA
| | - Yuval Kluger
- Department of Pathology, School of Medicine, Yale University, New Haven, CT, USA
| | - David G Hicks
- Department of Pathology, School of Medicine, University of Rochester, Rochester, NY, USA
| | - David L Rimm
- Department of Pathology, School of Medicine, Yale University, New Haven, CT, USA
| | | |
Collapse
|
36
|
Firempong CK, Cao X, Tong S, Yu J, Xu X. Prospects for multitarget lipid-raft-coated silica beads: a remarkable online biomaterial for discovering multitarget antitumor lead compounds. RSC Adv 2015. [DOI: 10.1039/c5ra08322b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Application of lipid raft biomaterial with multiple cancer-related receptors for screening novel multitarget antitumour lead compounds.
Collapse
Affiliation(s)
- Caleb Kesse Firempong
- Department of Pharmaceutics
- School of Pharmacy
- Centre for Nano Drug/Gene Delivery and Tissue Engineering
- Jiangsu University
- Zhenjiang
| | - Xia Cao
- Department of Pharmaceutics
- School of Pharmacy
- Centre for Nano Drug/Gene Delivery and Tissue Engineering
- Jiangsu University
- Zhenjiang
| | - Shanshan Tong
- Department of Pharmaceutics
- School of Pharmacy
- Centre for Nano Drug/Gene Delivery and Tissue Engineering
- Jiangsu University
- Zhenjiang
| | - Jiangnan Yu
- Department of Pharmaceutics
- School of Pharmacy
- Centre for Nano Drug/Gene Delivery and Tissue Engineering
- Jiangsu University
- Zhenjiang
| | - Ximing Xu
- Department of Pharmaceutics
- School of Pharmacy
- Centre for Nano Drug/Gene Delivery and Tissue Engineering
- Jiangsu University
- Zhenjiang
| |
Collapse
|
37
|
Nizard P, Ezan F, Bonnier D, Le Meur N, Langouët S, Baffet G, Arlot-Bonnemains Y, Théret N. Integrative analysis of high-throughput RNAi screen data identifies the FER and CRKL tyrosine kinases as new regulators of the mitogenic ERK-dependent pathways in transformed cells. BMC Genomics 2014; 15:1169. [PMID: 25540073 PMCID: PMC4367906 DOI: 10.1186/1471-2164-15-1169] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 12/12/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cell proliferation is a hallmark of cancer and depends on complex signaling networks that are chiefly supported by protein kinase activities. Therapeutic strategies have been used to target specific kinases but new methods are required to identify combined targets and improve treatment. Here, we propose a small interfering RNA genetic screen and an integrative approach to identify kinase networks involved in the proliferation of cancer cells. RESULTS The functional siRNA screen of 714 kinases in HeLa cells identified 91 kinases implicated in the regulation of cell growth, most of them never being reported in previous whole-genome siRNA screens. Based on gene ontology annotations, we have further discriminated between two classes of kinases that, when suppressed, result in alterations of the mitotic index and provoke cell-cycle arrest. Extinguished kinases that lead to a low mitotic index mostly include kinases implicated in cytosolic signaling. In contrast, extinguished kinases that result in a high mitotic index mostly include kinases implicated in cell division. By mapping hit kinases in the PhosphPOINT phosphoprotein database, we generated scale-free networks consisting of 449 and 661 protein-protein interactions for kinases from low MI and high MI groups, respectively. Further analyses of the kinase interactomes revealed specific modules such as FER- and CRKL-containing modules that connect three members of the epidermal growth factor receptor (EGFR) family, suggesting a tight control of the mitogenic EGF-dependent pathway. Based on experimental studies, we confirm the involvement of these two kinases in the regulation of tumor cell growth. CONCLUSION Based on a combined approach of large kinome-wide siRNA screens and ontology annotations, our study identifies for the first time two kinase groups differentially implicated in the control of cell proliferation. We further demonstrate that integrative analysis of the kinase interactome provides key information which can be used to facilitate or optimize target design for new therapeutic strategies. The complete list of protein-protein interactions from the two functional kinase groups will provide a useful database for future investigations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Nathalie Théret
- INSERM UMR1085, Institut de Recherche sur la Santé l'Environnement et le Travail IRSET, Université Rennes 1, SFR Biosit, Rennes, France.
| |
Collapse
|
38
|
Guo NL, Wan YW. Network-based identification of biomarkers coexpressed with multiple pathways. Cancer Inform 2014; 13:37-47. [PMID: 25392692 PMCID: PMC4218687 DOI: 10.4137/cin.s14054] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/25/2014] [Accepted: 06/29/2014] [Indexed: 02/07/2023] Open
Abstract
Unraveling complex molecular interactions and networks and incorporating clinical information in modeling will present a paradigm shift in molecular medicine. Embedding biological relevance via modeling molecular networks and pathways has become increasingly important for biomarker identification in cancer susceptibility and metastasis studies. Here, we give a comprehensive overview of computational methods used for biomarker identification, and provide a performance comparison of several network models used in studies of cancer susceptibility, disease progression, and prognostication. Specifically, we evaluated implication networks, Boolean networks, Bayesian networks, and Pearson’s correlation networks in constructing gene coexpression networks for identifying lung cancer diagnostic and prognostic biomarkers. The results show that implication networks, implemented in Genet package, identified sets of biomarkers that generated an accurate prediction of lung cancer risk and metastases; meanwhile, implication networks revealed more biologically relevant molecular interactions than Boolean networks, Bayesian networks, and Pearson’s correlation networks when evaluated with MSigDB database.
Collapse
Affiliation(s)
- Nancy Lan Guo
- Mary Babb Randolph Cancer Center/School of Public Health, West Virginia University, Morgantown, WV, USA
| | - Ying-Wooi Wan
- Mary Babb Randolph Cancer Center/School of Public Health, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
39
|
Geppert T, Koeppen H. Biological Networks and Drug Discovery-Where Do We Stand? Drug Dev Res 2014; 75:271-82. [DOI: 10.1002/ddr.21207] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Tim Geppert
- Lead Identification and Optimization Support; Boehringer Ingelheim Pharma GmbH & Co. KG; Biberach/Riss 88397 Germany
| | - Herbert Koeppen
- Lead Identification and Optimization Support; Boehringer Ingelheim Pharma GmbH & Co. KG; Biberach/Riss 88397 Germany
| |
Collapse
|
40
|
Pei J, Yin N, Ma X, Lai L. Systems Biology Brings New Dimensions for Structure-Based Drug Design. J Am Chem Soc 2014; 136:11556-65. [DOI: 10.1021/ja504810z] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jianfeng Pei
- Center
for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Ning Yin
- Center
for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Xiaomin Ma
- Center
for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Luhua Lai
- Center
for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Beijing
National Laboratory for Molecular Science, State Key Laboratory for
Structural Chemistry of Unstable and Stable Species, College of Chemistry
and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua
Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
41
|
Xie L, Ge X, Tan H, Xie L, Zhang Y, Hart T, Yang X, Bourne PE. Towards structural systems pharmacology to study complex diseases and personalized medicine. PLoS Comput Biol 2014; 10:e1003554. [PMID: 24830652 PMCID: PMC4022462 DOI: 10.1371/journal.pcbi.1003554] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Genome-Wide Association Studies (GWAS), whole genome sequencing, and high-throughput omics techniques have generated vast amounts of genotypic and molecular phenotypic data. However, these data have not yet been fully explored to improve the effectiveness and efficiency of drug discovery, which continues along a one-drug-one-target-one-disease paradigm. As a partial consequence, both the cost to launch a new drug and the attrition rate are increasing. Systems pharmacology and pharmacogenomics are emerging to exploit the available data and potentially reverse this trend, but, as we argue here, more is needed. To understand the impact of genetic, epigenetic, and environmental factors on drug action, we must study the structural energetics and dynamics of molecular interactions in the context of the whole human genome and interactome. Such an approach requires an integrative modeling framework for drug action that leverages advances in data-driven statistical modeling and mechanism-based multiscale modeling and transforms heterogeneous data from GWAS, high-throughput sequencing, structural genomics, functional genomics, and chemical genomics into unified knowledge. This is not a small task, but, as reviewed here, progress is being made towards the final goal of personalized medicines for the treatment of complex diseases.
Collapse
Affiliation(s)
- Lei Xie
- Department of Computer Science, Hunter College, The City University of New York, New York, New York, United States of America
- Ph.D. Program in Computer Science, Biology, and Biochemistry, The Graduate Center, The City University of New York, New York, New York, United States of America
- * E-mail:
| | - Xiaoxia Ge
- Department of Computer Science, Hunter College, The City University of New York, New York, New York, United States of America
| | - Hepan Tan
- Department of Computer Science, Hunter College, The City University of New York, New York, New York, United States of America
| | - Li Xie
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Yinliang Zhang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Thomas Hart
- Department of Biological Sciences, Hunter College, The City University of New York, New York, New York, United States of America
| | - Xiaowei Yang
- School of Public Health, Hunter College, The City University of New York, New York, New York, United States of America
| | - Philip E. Bourne
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
42
|
Akbani R, Becker KF, Carragher N, Goldstein T, de Koning L, Korf U, Liotta L, Mills GB, Nishizuka SS, Pawlak M, Petricoin EF, Pollard HB, Serrels B, Zhu J. Realizing the promise of reverse phase protein arrays for clinical, translational, and basic research: a workshop report: the RPPA (Reverse Phase Protein Array) society. Mol Cell Proteomics 2014; 13:1625-43. [PMID: 24777629 DOI: 10.1074/mcp.o113.034918] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Reverse phase protein array (RPPA) technology introduced a miniaturized "antigen-down" or "dot-blot" immunoassay suitable for quantifying the relative, semi-quantitative or quantitative (if a well-accepted reference standard exists) abundance of total protein levels and post-translational modifications across a variety of biological samples including cultured cells, tissues, and body fluids. The recent evolution of RPPA combined with more sophisticated sample handling, optical detection, quality control, and better quality affinity reagents provides exquisite sensitivity and high sample throughput at a reasonable cost per sample. This facilitates large-scale multiplex analysis of multiple post-translational markers across samples from in vitro, preclinical, or clinical samples. The technical power of RPPA is stimulating the application and widespread adoption of RPPA methods within academic, clinical, and industrial research laboratories. Advances in RPPA technology now offer scientists the opportunity to quantify protein analytes with high precision, sensitivity, throughput, and robustness. As a result, adopters of RPPA technology have recognized critical success factors for useful and maximum exploitation of RPPA technologies, including the following: preservation and optimization of pre-analytical sample quality, application of validated high-affinity and specific antibody (or other protein affinity) detection reagents, dedicated informatics solutions to ensure accurate and robust quantification of protein analytes, and quality-assured procedures and data analysis workflows compatible with application within regulated clinical environments. In 2011, 2012, and 2013, the first three Global RPPA workshops were held in the United States, Europe, and Japan, respectively. These workshops provided an opportunity for RPPA laboratories, vendors, and users to share and discuss results, the latest technology platforms, best practices, and future challenges and opportunities. The outcomes of the workshops included a number of key opportunities to advance the RPPA field and provide added benefit to existing and future participants in the RPPA research community. The purpose of this report is to share and disseminate, as a community, current knowledge and future directions of the RPPA technology.
Collapse
Affiliation(s)
- Rehan Akbani
- From the *University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | | | - Neil Carragher
- §Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Ted Goldstein
- ¶Center for Biomolecular Science and Engineering, University of California, Santa Cruz, California
| | | | - Ulrike Korf
- **German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Gordon B Mills
- From the *University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | | | - Michael Pawlak
- §§§The Natural and Medical Sciences Institute, Reutlingen, Germany
| | | | - Harvey B Pollard
- ¶¶Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Bryan Serrels
- §Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Jingchun Zhu
- ¶Center for Biomolecular Science and Engineering, University of California, Santa Cruz, California
| |
Collapse
|
43
|
Yin N, Ma W, Pei J, Ouyang Q, Tang C, Lai L. Synergistic and antagonistic drug combinations depend on network topology. PLoS One 2014; 9:e93960. [PMID: 24713621 PMCID: PMC3979733 DOI: 10.1371/journal.pone.0093960] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/10/2014] [Indexed: 11/18/2022] Open
Abstract
Drug combinations may exhibit synergistic or antagonistic effects. Rational design of synergistic drug combinations remains a challenge despite active experimental and computational efforts. Because drugs manifest their action via their targets, the effects of drug combinations should depend on the interaction of their targets in a network manner. We therefore modeled the effects of drug combinations along with their targets interacting in a network, trying to elucidate the relationships between the network topology involving drug targets and drug combination effects. We used three-node enzymatic networks with various topologies and parameters to study two-drug combinations. These networks can be simplifications of more complex networks involving drug targets, or closely connected target networks themselves. We found that the effects of most of the combinations were not sensitive to parameter variation, indicating that drug combinational effects largely depend on network topology. We then identified and analyzed consistent synergistic or antagonistic drug combination motifs. Synergistic motifs encompass a diverse range of patterns, including both serial and parallel combinations, while antagonistic combinations are relatively less common and homogenous, mostly composed of a positive feedback loop and a downstream link. Overall our study indicated that designing novel synergistic drug combinations based on network topology could be promising, and the motifs we identified could be a useful catalog for rational drug combination design in enzymatic systems.
Collapse
Affiliation(s)
- Ning Yin
- Center for Quantitative Biology, Peking University, Beijing, China
| | - Wenzhe Ma
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jianfeng Pei
- Center for Quantitative Biology, Peking University, Beijing, China
| | - Qi Ouyang
- Center for Quantitative Biology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- School of Physics, Peking University, Beijing, China
| | - Chao Tang
- Center for Quantitative Biology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- School of Physics, Peking University, Beijing, China
| | - Luhua Lai
- Center for Quantitative Biology, Peking University, Beijing, China
- BNLMS, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- * E-mail:
| |
Collapse
|
44
|
González-Díaz H, Herrera-Ibatá DM, Duardo-Sánchez A, Munteanu CR, Orbegozo-Medina RA, Pazos A. ANN Multiscale Model of Anti-HIV Drugs Activity vs AIDS Prevalence in the US at County Level Based on Information Indices of Molecular Graphs and Social Networks. J Chem Inf Model 2014; 54:744-55. [DOI: 10.1021/ci400716y] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Humberto González-Díaz
- Department
of Organic Chemistry II, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940, Leioa, Vizcaya, Spain
- IKERBASQUE, Basque
Foundation for Science, 48011, Bilbao, Vizcaya, Spain
| | - Diana María Herrera-Ibatá
- Department of Information and Communication Technologies, University of A Coruña UDC, 15071, A Coruña, A Coruña, Spain
| | - Aliuska Duardo-Sánchez
- Department of Information and Communication Technologies, University of A Coruña UDC, 15071, A Coruña, A Coruña, Spain
| | - Cristian R. Munteanu
- Department of Information and Communication Technologies, University of A Coruña UDC, 15071, A Coruña, A Coruña, Spain
| | - Ricardo Alfredo Orbegozo-Medina
- Department
of Microbiology and Parasitology, University of Santiago de Compostela (USC), 15782, Santiago de Compostela, A Coruña, Spain
| | - Alejandro Pazos
- Department of Information and Communication Technologies, University of A Coruña UDC, 15071, A Coruña, A Coruña, Spain
| |
Collapse
|
45
|
HER. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
46
|
Vandamme D, Minke BA, Fitzmaurice W, Kholodenko BN, Kolch W. Systems biology-embedded target validation: improving efficacy in drug discovery. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2013; 6:1-11. [PMID: 24214316 DOI: 10.1002/wsbm.1253] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/24/2013] [Accepted: 10/11/2013] [Indexed: 12/31/2022]
Abstract
The pharmaceutical industry is faced with a range of challenges with the ever-escalating costs of drug development and a drying out of drug pipelines. By harnessing advances in -omics technologies and moving away from the standard, reductionist model of drug discovery, there is significant potential to reduce costs and improve efficacy. Embedding systems biology approaches in drug discovery, which seek to investigate underlying molecular mechanisms of potential drug targets in a network context, will reduce attrition rates by earlier target validation and the introduction of novel targets into the currently stagnant market. Systems biology approaches also have the potential to assist in the design of multidrug treatments and repositioning of existing drugs, while stratifying patients to give a greater personalization of medical treatment.
Collapse
Affiliation(s)
- Drieke Vandamme
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | | | | | | | | |
Collapse
|
47
|
Janes KA, Lauffenburger DA. Models of signalling networks - what cell biologists can gain from them and give to them. J Cell Sci 2013; 126:1913-21. [PMID: 23720376 DOI: 10.1242/jcs.112045] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Computational models of cell signalling are perceived by many biologists to be prohibitively complicated. Why do math when you can simply do another experiment? Here, we explain how conceptual models, which have been formulated mathematically, have provided insights that directly advance experimental cell biology. In the past several years, models have influenced the way we talk about signalling networks, how we monitor them, and what we conclude when we perturb them. These insights required wet-lab experiments but would not have arisen without explicit computational modelling and quantitative analysis. Today, the best modellers are cross-trained investigators in experimental biology who work closely with collaborators but also undertake experimental work in their own laboratories. Biologists would benefit by becoming conversant in core principles of modelling in order to identify when a computational model could be a useful complement to their experiments. Although the mathematical foundations of a model are useful to appreciate its strengths and weaknesses, they are not required to test or generate a worthwhile biological hypothesis computationally.
Collapse
Affiliation(s)
- Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | | |
Collapse
|
48
|
Falcetta F, Lupi M, Colombo V, Ubezio P. Dynamic rendering of the heterogeneous cell response to anticancer treatments. PLoS Comput Biol 2013; 9:e1003293. [PMID: 24146610 PMCID: PMC3798276 DOI: 10.1371/journal.pcbi.1003293] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 09/05/2013] [Indexed: 11/29/2022] Open
Abstract
The antiproliferative response to anticancer treatment is the result of concurrent responses in all cell cycle phases, extending over several cell generations, whose complexity is not captured by current methods. In the proposed experimental/computational approach, the contemporary use of time-lapse live cell microscopy and flow cytometric data supported the computer rendering of the proliferative process through the cell cycle and subsequent generations during/after treatment. The effects of treatments were modelled with modules describing the functional activity of the main pathways causing arrest, repair and cell death in each phase. A framework modelling environment was created, enabling us to apply different types of modules in each phase and test models at the complexity level justified by the available data. We challenged the method with time-course measures taken in parallel with flow cytometry and time-lapse live cell microscopy in X-ray-treated human ovarian cancer cells, spanning a wide range of doses. The most suitable model of the treatment, including the dose-response of each effect, was progressively built, combining modules with a rational strategy and fitting simultaneously all data of different doses and platforms. The final model gave for the first time the complete rendering in silico of the cycling process following X-ray exposure, providing separate and quantitative measures of the dose-dependence of G1, S and G2M checkpoint activities in subsequent generations, reconciling known effects of ionizing radiations and new insights in a unique scenario. The antiproliferative response to anticancer treatment is the result of concurrent effects in all cell cycle phases, where molecular control pathways (checkpoints) are activated and cells may be arrested to repair DNA damage or killed if not able to succeed in the repair process. The complexity and inter-cell variability of these phenomena are not captured by the available methods, and the origin of the dose-dependence of the response remains elusive. In this work, we present an experimental-computational method that discloses and measures the individual responses of cell cycle controls in each phase and generation. We demonstrate that the method, exploiting jointly data sets obtained by flow cytometry and time-lapse in vivo imaging with a suitable experimental design, is able to achieve a full reconstruction in silico of the actual movement of cell cohorts following X-ray exposure, providing separate and quantitative measures of the dose-dependence of G1, S and G2M checkpoint activities in subsequent generations. Best fit parameters values are actual measures of the probability of activation of the specific pathways of arrest, repair or death within the cell population, linking the molecular scale to the “macroscopic” response, with full appreciation of its dynamics and inter-cell heterogeneity.
Collapse
Affiliation(s)
- Francesca Falcetta
- Biophysics Unit, Laboratory of Anticancer Pharmacology, Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche “Mario Negri,” Milano, Italy
| | - Monica Lupi
- Biophysics Unit, Laboratory of Anticancer Pharmacology, Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche “Mario Negri,” Milano, Italy
| | - Valentina Colombo
- Biophysics Unit, Laboratory of Anticancer Pharmacology, Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche “Mario Negri,” Milano, Italy
| | - Paolo Ubezio
- Biophysics Unit, Laboratory of Anticancer Pharmacology, Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche “Mario Negri,” Milano, Italy
- * E-mail:
| |
Collapse
|
49
|
Advanced systems biology methods in drug discovery and translational biomedicine. BIOMED RESEARCH INTERNATIONAL 2013; 2013:742835. [PMID: 24171171 PMCID: PMC3792523 DOI: 10.1155/2013/742835] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 08/26/2013] [Indexed: 02/08/2023]
Abstract
Systems biology is in an exponential development stage in recent years and has been widely utilized in biomedicine to better understand the molecular basis of human disease and the mechanism of drug action. Here, we discuss the fundamental concept of systems biology and its two computational methods that have been commonly used, that is, network analysis and dynamical modeling. The applications of systems biology in elucidating human disease are highlighted, consisting of human disease networks, treatment response prediction, investigation of disease mechanisms, and disease-associated gene prediction. In addition, important advances in drug discovery, to which systems biology makes significant contributions, are discussed, including drug-target networks, prediction of drug-target interactions, investigation of drug adverse effects, drug repositioning, and drug combination prediction. The systems biology methods and applications covered in this review provide a framework for addressing disease mechanism and approaching drug discovery, which will facilitate the translation of research findings into clinical benefits such as novel biomarkers and promising therapies.
Collapse
|
50
|
Bhattacharya S, Shoda LKM, Zhang Q, Woods CG, Howell BA, Siler SQ, Woodhead JL, Yang Y, McMullen P, Watkins PB, Andersen ME. Modeling drug- and chemical-induced hepatotoxicity with systems biology approaches. Front Physiol 2012; 3:462. [PMID: 23248599 PMCID: PMC3522076 DOI: 10.3389/fphys.2012.00462] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 11/21/2012] [Indexed: 12/22/2022] Open
Abstract
We provide an overview of computational systems biology approaches as applied to the study of chemical- and drug-induced toxicity. The concept of “toxicity pathways” is described in the context of the 2007 US National Academies of Science report, “Toxicity testing in the 21st Century: A Vision and A Strategy.” Pathway mapping and modeling based on network biology concepts are a key component of the vision laid out in this report for a more biologically based analysis of dose-response behavior and the safety of chemicals and drugs. We focus on toxicity of the liver (hepatotoxicity) – a complex phenotypic response with contributions from a number of different cell types and biological processes. We describe three case studies of complementary multi-scale computational modeling approaches to understand perturbation of toxicity pathways in the human liver as a result of exposure to environmental contaminants and specific drugs. One approach involves development of a spatial, multicellular “virtual tissue” model of the liver lobule that combines molecular circuits in individual hepatocytes with cell–cell interactions and blood-mediated transport of toxicants through hepatic sinusoids, to enable quantitative, mechanistic prediction of hepatic dose-response for activation of the aryl hydrocarbon receptor toxicity pathway. Simultaneously, methods are being developing to extract quantitative maps of intracellular signaling and transcriptional regulatory networks perturbed by environmental contaminants, using a combination of gene expression and genome-wide protein-DNA interaction data. A predictive physiological model (DILIsym™) to understand drug-induced liver injury (DILI), the most common adverse event leading to termination of clinical development programs and regulatory actions on drugs, is also described. The model initially focuses on reactive metabolite-induced DILI in response to administration of acetaminophen, and spans multiple biological scales.
Collapse
Affiliation(s)
- Sudin Bhattacharya
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences Research Triangle Park, NC, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|