1
|
Singh M, Sachdeva M, Kumar N. Assessment of the Anti-adipogenic Effect of Crateva religiosa Bark Extract for Molecular Regulation of Adipogenesis: In Silico and In Vitro Approaches for Management of Hyperlipidemia Through the 3T3-L1 Cell Line. Curr Pharm Biotechnol 2025; 26:778-794. [PMID: 39206484 DOI: 10.2174/0113892010314594240816050240] [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: 03/05/2024] [Revised: 06/16/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024]
Abstract
AIMS This study aimed to determine the phytoconstituents of Crateva religiosa bark (CRB) and evaluate the hypolipidemic effect of bioactive CRB extract by preventing adipocyte differentiation and lipogenesis. BACKGROUND After performing the preliminary phytochemicals screening, the antioxidant activity of CRB extracts was determined through a DPPH (2, 2-diphenyl-1-picrylhydrazyl) assay. Ethyl acetate extract (CREAE) and ethanol extract (CRETE) of CRB were selected for chromatographic evaluation. METHODS The antihyperlipidemic potential was analyzed by molecular docking through the PKCMS software platform. Further, a 3T3-L1 cell line study via in vitro sulforhodamine B assay and western blotting was performed to confirm the prevention of adipocyte differentiation and lipogenesis. RESULTS The total phenolic contents in CREAE and CRETE were estimated as 29.47 and 81.19 μg/mg equivalent to gallic acid, respectively. The total flavonoid content was found to be 8.78 and 49.08 μg/mg, equivalent to quercetin in CREAE and CRETE, respectively. CRETE exhibited greater scavenging activity with the IC50 value of 61.05 μg/ mL. GC-MS analysis confirmed the presence of three bioactive molecules, stigmasterol, gamma sitosterol, and lupeol, in CRETE. Molecular docking studies predicted that the bioactive molecules interact with HMG-CoA reductase, PPARγ, and CCAAT/EBP, which are responsible for lipid metabolism. In vitro, Sulforhodamine B assays revealed that CRETE dose-dependently reduced cell differentiation and viability. Cellular staining using 'Oil Red O' revealed a decreased lipid content in the CRETE-treated cell lines. CRETE significantly inhibited the induction of PPARγ and CCAAT/EBP expression, as determined through protein expression via western blotting. CONCLUSION The influence of CRETE on lipid metabolism in 3T3-L1 cells is potentially suggesting a new approach to managing hyperlipidemia.
Collapse
Affiliation(s)
- Monika Singh
- Department of Pharmacology, I.T.S. College of Pharmacy, Ghaziabad, U.P., Affiliated with Dr. A.P.J. Abdul Kalam Technical University, Lucknow, India
| | - Monika Sachdeva
- Department of Pharmacy, Raj Kumar Goel Institute of Technology, Ghaziabad U.P., Affiliated with Dr. A.P.J. Abdul Kalam Technical University, Lucknow, India
| | - Nitin Kumar
- Department of Pharmacy, Meerut Institute of Technology, Meerut, Affiliated with Dr. A.P.J. Abdul Kalam Technical University, Lucknow, India
| |
Collapse
|
2
|
Cogno N, Axenie C, Bauer R, Vavourakis V. Agent-based modeling in cancer biomedicine: applications and tools for calibration and validation. Cancer Biol Ther 2024; 25:2344600. [PMID: 38678381 PMCID: PMC11057625 DOI: 10.1080/15384047.2024.2344600] [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: 10/30/2023] [Accepted: 04/15/2024] [Indexed: 04/29/2024] Open
Abstract
Computational models are not just appealing because they can simulate and predict the development of biological phenomena across multiple spatial and temporal scales, but also because they can integrate information from well-established in vitro and in vivo models and test new hypotheses in cancer biomedicine. Agent-based models and simulations are especially interesting candidates among computational modeling procedures in cancer research due to the capability to, for instance, recapitulate the dynamics of neoplasia and tumor - host interactions. Yet, the absence of methods to validate the consistency of the results across scales can hinder adoption by turning fine-tuned models into black boxes. This review compiles relevant literature that explores strategies to leverage high-fidelity simulations of multi-scale, or multi-level, cancer models with a focus on verification approached as simulation calibration. We consolidate our review with an outline of modern approaches for agent-based models' validation and provide an ambitious outlook toward rigorous and reliable calibration.
Collapse
Affiliation(s)
- Nicolò Cogno
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Institute for Condensed Matter Physics, Technische Universit¨at Darmstadt, Darmstadt, Germany
| | - Cristian Axenie
- Computer Science Department and Center for Artificial Intelligence, Technische Hochschule Nürnberg Georg Simon Ohm, Nuremberg, Germany
| | - Roman Bauer
- Nature Inspired Computing and Engineering Research Group, Computer Science Research Centre, University of Surrey, Guildford, UK
| | - Vasileios Vavourakis
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
- Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
3
|
Tuna R, Yi W, Crespo Cruz E, Romero JP, Ren Y, Guan J, Li Y, Deng Y, Bluestein D, Liu ZL, Sheriff J. Platelet Biorheology and Mechanobiology in Thrombosis and Hemostasis: Perspectives from Multiscale Computation. Int J Mol Sci 2024; 25:4800. [PMID: 38732019 PMCID: PMC11083691 DOI: 10.3390/ijms25094800] [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: 02/11/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Thrombosis is the pathological clot formation under abnormal hemodynamic conditions, which can result in vascular obstruction, causing ischemic strokes and myocardial infarction. Thrombus growth under moderate to low shear (<1000 s-1) relies on platelet activation and coagulation. Thrombosis at elevated high shear rates (>10,000 s-1) is predominantly driven by unactivated platelet binding and aggregating mediated by von Willebrand factor (VWF), while platelet activation and coagulation are secondary in supporting and reinforcing the thrombus. Given the molecular and cellular level information it can access, multiscale computational modeling informed by biology can provide new pathophysiological mechanisms that are otherwise not accessible experimentally, holding promise for novel first-principle-based therapeutics. In this review, we summarize the key aspects of platelet biorheology and mechanobiology, focusing on the molecular and cellular scale events and how they build up to thrombosis through platelet adhesion and aggregation in the presence or absence of platelet activation. In particular, we highlight recent advancements in multiscale modeling of platelet biorheology and mechanobiology and how they can lead to the better prediction and quantification of thrombus formation, exemplifying the exciting paradigm of digital medicine.
Collapse
Affiliation(s)
- Rukiye Tuna
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
| | - Wenjuan Yi
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
| | - Esmeralda Crespo Cruz
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
| | - JP Romero
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
| | - Yi Ren
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32304, USA
| | - Jingjiao Guan
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
- Institute for Successful Longevity, Florida State University, Tallahassee, FL 32304, USA
| | - Yan Li
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
- Institute for Successful Longevity, Florida State University, Tallahassee, FL 32304, USA
| | - Yuefan Deng
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Danny Bluestein
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Zixiang Leonardo Liu
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA; (R.T.); (E.C.C.); (Z.L.L.)
- Institute for Successful Longevity, Florida State University, Tallahassee, FL 32304, USA
| | - Jawaad Sheriff
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA;
| |
Collapse
|
4
|
Gazo Hanna E, Younes K, Roufayel R, Khazaal M, Fajloun Z. Engineering innovations in medicine and biology: Revolutionizing patient care through mechanical solutions. Heliyon 2024; 10:e26154. [PMID: 38390063 PMCID: PMC10882044 DOI: 10.1016/j.heliyon.2024.e26154] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 01/24/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
The overlap between mechanical engineering and medicine is expanding more and more over the years. Engineers are now using their expertise to design and create functional biomaterials and are continually collaborating with physicians to improve patient health. In this review, we explore the state of scientific knowledge in the areas of biomaterials, biomechanics, nanomechanics, and computational fluid dynamics (CFD) in relation to the pharmaceutical and medical industry. Focusing on current research and breakthroughs, we provide an overview of how these fields are being used to create new technologies for medical treatments of human patients. Barriers and constraints in these fields, as well as ways to overcome them, are also described in this review. Finally, the potential for future advances in biomaterials to fundamentally change the current approach to medicine and biology is also discussed.
Collapse
Affiliation(s)
- Eddie Gazo Hanna
- College of Engineering and Technology, American University of the Middle East, Egaila, 54200, Kuwait
| | - Khaled Younes
- College of Engineering and Technology, American University of the Middle East, Egaila, 54200, Kuwait
| | - Rabih Roufayel
- College of Engineering and Technology, American University of the Middle East, Egaila, 54200, Kuwait
| | - Mickael Khazaal
- École Supérieure des Techniques Aéronautiques et de Construction Automobile, ISAE-ESTACA, France
| | - Ziad Fajloun
- Faculty of Sciences 3, Department of Biology, Lebanese University, Campus Michel Slayman Ras Maska, 1352, Tripoli, Lebanon
- Laboratory of Applied Biotechnology (LBA3B), Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, 1300, Tripoli, Lebanon
| |
Collapse
|
5
|
Tindall MJ, Cucurull-Sanchez L, Mistry H, Yates JWT. Quantitative Systems Pharmacology and Machine Learning: A Match Made in Heaven or Hell? J Pharmacol Exp Ther 2023; 387:92-99. [PMID: 37652709 DOI: 10.1124/jpet.122.001551] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 09/02/2023] Open
Abstract
As pharmaceutical development moves from early-stage in vitro experimentation to later in vivo and subsequent clinical trials, data and knowledge are acquired across multiple time and length scales, from the subcellular to whole patient cohort scale. Realizing the potential of this data for informing decision making in pharmaceutical development requires the individual and combined application of machine learning (ML) and mechanistic multiscale mathematical modeling approaches. Here we outline how these two approaches, both individually and in tandem, can be applied at different stages of the drug discovery and development pipeline to inform decision making compound development. The importance of discerning between knowledge and data are highlighted in informing the initial use of ML or mechanistic quantitative systems pharmacology (QSP) models. We discuss the application of sensitivity and structural identifiability analyses of QSP models in informing future experimental studies to which ML may be applied, as well as how ML approaches can be used to inform mechanistic model development. Relevant literature studies are highlighted and we close by discussing caveats regarding the application of each approach in an age of constant data acquisition. SIGNIFICANCE STATEMENT: We consider when best to apply machine learning (ML) and mechanistic quantitative systems pharmacology (QSP) approaches in the context of the drug discovery and development pipeline. We discuss the importance of prior knowledge and data available for the system of interest and how this informs the individual and combined application of ML and QSP approaches at each stage of the pipeline.
Collapse
Affiliation(s)
- Marcus John Tindall
- Department of Mathematics and Statistics and Institute of Cardiovascular and Metabolic Research, University of Reading, Whiteknights, Reading, United Kingdom (M.J.T.); GSK Medicines Research Centre, Stevenage, United Kingdom (L.C.-S., J.W.T.Y.); and Pharmacy, Division of Pharmacy and Optometry, University of Manchester, Oxford Road, Manchester, United Kingdom (H.M.)
| | - Lourdes Cucurull-Sanchez
- Department of Mathematics and Statistics and Institute of Cardiovascular and Metabolic Research, University of Reading, Whiteknights, Reading, United Kingdom (M.J.T.); GSK Medicines Research Centre, Stevenage, United Kingdom (L.C.-S., J.W.T.Y.); and Pharmacy, Division of Pharmacy and Optometry, University of Manchester, Oxford Road, Manchester, United Kingdom (H.M.)
| | - Hitesh Mistry
- Department of Mathematics and Statistics and Institute of Cardiovascular and Metabolic Research, University of Reading, Whiteknights, Reading, United Kingdom (M.J.T.); GSK Medicines Research Centre, Stevenage, United Kingdom (L.C.-S., J.W.T.Y.); and Pharmacy, Division of Pharmacy and Optometry, University of Manchester, Oxford Road, Manchester, United Kingdom (H.M.)
| | - James W T Yates
- Department of Mathematics and Statistics and Institute of Cardiovascular and Metabolic Research, University of Reading, Whiteknights, Reading, United Kingdom (M.J.T.); GSK Medicines Research Centre, Stevenage, United Kingdom (L.C.-S., J.W.T.Y.); and Pharmacy, Division of Pharmacy and Optometry, University of Manchester, Oxford Road, Manchester, United Kingdom (H.M.)
| |
Collapse
|
6
|
Luo S, Gong J, Zhao S, Li M, Li R. Deubiquitinase BAP1 regulates stability of BRCA1 protein and inactivates the NF-κB signaling to protect mice from sepsis-induced acute kidney injury. Chem Biol Interact 2023; 382:110621. [PMID: 37414201 DOI: 10.1016/j.cbi.2023.110621] [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: 03/21/2023] [Revised: 06/21/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Sepsis and its associated organ dysfunction syndrome is a leading cause of death in critically ill patients. Breast cancer susceptibility protein 1 (BRCA1)-associated protein 1 (BAP1) is a potential regulator in immune regulation and inflammatory responses. This study aims to investigate the function of BAP1 in sepsis-induced acute kidney injury (AKI). A mouse model with sepsis-induced AKI was induced by cecal ligation and puncture, and renal tubular epithelial cells (RTECs) were treated with lipopolysaccharide (LPS) to mimic an AKI condition in vitro. BAP1 was significantly poorly expressed in the kidney tissues of model mice and the LPS-treated RTECs. Artificial upregulation of BAP1 ameliorated the pathological changes, tissue injury and inflammatory responses in kidney tissues of the mice, and it reduced the LPS-induced injury and apoptosis of the RTECs. BAP1 was found to interact with BRCA1 and enhance stability of BRCA1 protein through deubiquitination modification. Further downregulation of BRCA1 activated the nuclear factor-kappa B (NF-κB) signaling pathway and blocked the protective roles of BAP1 in sepsis-induced AKI. In conclusion, this study demonstrates that BAP1 protects mice from sepsis-induced AKI through enhancing stability of BRCA1 protein and inactivating the NF-κB signaling.
Collapse
Affiliation(s)
- Shu Luo
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, PR China.
| | - Junzuo Gong
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, PR China
| | - Shiqiao Zhao
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, PR China
| | - Menqin Li
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, PR China
| | - Ruixiu Li
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, PR China
| |
Collapse
|
7
|
Ghosh K, Vernuccio S, Dowling AW. Nonlinear Reactor Design Optimization With Embedded Microkinetic Model Information. FRONTIERS IN CHEMICAL ENGINEERING 2022. [DOI: 10.3389/fceng.2022.898685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Despite the success of multiscale modeling in science and engineering, embedding molecular-level information into nonlinear reactor design and control optimization problems remains challenging. In this work, we propose a computationally tractable scale-bridging approach that incorporates information from multi-product microkinetic (MK) models with thousands of rates and chemical species into nonlinear reactor design optimization problems. We demonstrate reduced-order kinetic (ROK) modeling approaches for catalytic oligomerization in shale gas processing. We assemble a library of six candidate ROK models based on literature and MK model structure. We find that three metrics—quality of fit (e.g., mean squared logarithmic error), thermodynamic consistency (e.g., low conversion of exothermic reactions at high temperatures), and model identifiability—are all necessary to train and select ROK models. The ROK models that closely mimic the structure of the MK model offer the best compromise to emulate the product distribution. Using the four best ROK models, we optimize the temperature profiles in staged reactors to maximize conversions to heavier oligomerization products. The optimal temperature starts at 630–900K and monotonically decreases to approximately 560 K in the final stage, depending on the choice of ROK model. For all models, staging increases heavier olefin production by 2.5% and there is minimal benefit to more than four stages. The choice of ROK model, i.e., model-form uncertainty, results in a 22% difference in the objective function, which is twice the impact of parametric uncertainty; we demonstrate sequential eigendecomposition of the Fisher information matrix to identify and fix sloppy model parameters, which allows for more reliable estimation of the covariance of the identifiable calibrated model parameters. First-order uncertainty propagation determines this parametric uncertainty induces less than a 10% variability in the reactor optimization objective function. This result highlights the importance of quantifying model-form uncertainty, in addition to parametric uncertainty, in multi-scale reactor and process design and optimization. Moreover, the fast dynamic optimization solution times suggest the ROK strategy is suitable for incorporating molecular information in sequential modular or equation-oriented process simulation and optimization frameworks.
Collapse
|
8
|
Dałek P, Drabik D, Wołczańska H, Foryś A, Jagas M, Jędruchniewicz N, Przybyło M, Witkiewicz W, Langner M. Bioavailability by design — Vitamin D3 liposomal delivery vehicles. NANOMEDICINE: NANOTECHNOLOGY, BIOLOGY AND MEDICINE 2022; 43:102552. [PMID: 35346834 PMCID: PMC8957331 DOI: 10.1016/j.nano.2022.102552] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/01/2022] [Accepted: 03/15/2022] [Indexed: 11/29/2022]
Abstract
Vitamin D3 deficiency has serious health consequences, as demonstrated by its effect on severity and recovery after COVID-19 infection. Because of high hydrophobicity, its absorption and subsequent redistribution throughout the body are inherently dependent on the accompanying lipids and/or proteins. The effective oral vitamin D3 formulation should ensure penetration of the mucus layer followed by internalization by competent cells. Isothermal titration calorimetry and computer simulations show that vitamin D3 molecules cannot leave the hydrophobic environment, indicating that their absorption is predominantly driven by the digestion of the delivery vehicle. In the clinical experiment, liposomal vitamin D3 was compared to the oily formulation. The results obtained show that liposomal vitamin D3 causes a rapid increase in the plasma concentration of calcidiol. No such effect was observed when the oily formulation was used. The effect was especially pronounced for people with severe vitamin D3 deficiency.
Collapse
Affiliation(s)
- Paulina Dałek
- Laboratory for Biophysics of Macromolecular Aggregates, Department of Biomedical Engineering, Wrocław University of Science and Technology, Wrocław, Poland; Lipid Systems sp. z o.o., Wrocław, Poland.
| | - Dominik Drabik
- Laboratory for Biophysics of Macromolecular Aggregates, Department of Biomedical Engineering, Wrocław University of Science and Technology, Wrocław, Poland
| | | | - Aleksander Foryś
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | | | | | - Magdalena Przybyło
- Laboratory for Biophysics of Macromolecular Aggregates, Department of Biomedical Engineering, Wrocław University of Science and Technology, Wrocław, Poland; Lipid Systems sp. z o.o., Wrocław, Poland
| | - Wojciech Witkiewicz
- Research and Development Centre, Specialized Hospital in Wrocław, Wrocław, Poland
| | - Marek Langner
- Laboratory for Biophysics of Macromolecular Aggregates, Department of Biomedical Engineering, Wrocław University of Science and Technology, Wrocław, Poland; Lipid Systems sp. z o.o., Wrocław, Poland
| |
Collapse
|
9
|
Schmitt JM, Baumann JM, Morgen MM. Predicting Spray Dried Dispersion Particle Size Via Machine Learning Regression Methods. Pharm Res 2022; 39:3223-3239. [PMID: 35986124 PMCID: PMC9780133 DOI: 10.1007/s11095-022-03370-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/14/2022] [Indexed: 12/27/2022]
Abstract
Spray dried dispersion particle size is a critical quality attribute that impacts bioavailability and manufacturability of the spray drying process and final dosage form. Substantial experimentation has been required to relate formulation and process parameters to particle size with the results limited to a single active pharmaceutical ingredient (API). This is the first study that demonstrates prediction of particle size independent of API for a wide range of formulation and process parameters at pilot and commercial scale. Additionally we developed a strategy with formulation and target particle size as inputs to define a set of "first to try" process parameters. An ensemble machine learning model was created to predict dried particle size across pilot and production scale spray dryers, with prediction errors between -7.7% and 18.6% (25th/75th percentiles) for a hold-out evaluation set. Shapley additive explanations identified how changes in formulation and process parameters drove variations in model predictions of dried particle size and were found to be consistent with mechanistic understanding of the particle formation process. Additionally, an optimization strategy used the predictive model to determine initial estimates for process parameter values that best achieve a target particle size for a provided formulation. The optimization strategy was employed to estimate process parameters in the hold-out evaluation set and to illustrate selection of process parameters during scale-up. The results of this study illustrate how trained regression models can reduce the experimental effort required to create an in-silico design space for new molecules during early-stage process development and subsequent scale-up.
Collapse
Affiliation(s)
- John M. Schmitt
- Computational Science, Lonza, 1201 NW Wall St, Bend, OR 97703 USA
| | | | | |
Collapse
|
10
|
Lagoutte-Renosi J, Allemand F, Ramseyer C, Yesylevskyy S, Davani S. Molecular modeling in cardiovascular pharmacology: Current state of the art and perspectives. Drug Discov Today 2021; 27:985-1007. [PMID: 34863931 DOI: 10.1016/j.drudis.2021.11.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/02/2021] [Accepted: 11/25/2021] [Indexed: 01/10/2023]
Abstract
Molecular modeling in pharmacology is a promising emerging tool for exploring drug interactions with cellular components. Recent advances in molecular simulations, big data analysis, and artificial intelligence (AI) have opened new opportunities for rationalizing drug interactions with their pharmacological targets. Despite the obvious utility and increasing impact of computational approaches, their development is not progressing at the same speed in different fields of pharmacology. Here, we review current in silico techniques used in cardiovascular diseases (CVDs), cardiological drug discovery, and assessment of cardiotoxicity. In silico techniques are paving the way to a new era in cardiovascular medicine, but their use somewhat lags behind that in other fields.
Collapse
Affiliation(s)
- Jennifer Lagoutte-Renosi
- EA 3920 Université Bourgogne Franche-Comté, 25000 Besançon, France; Laboratoire de Pharmacologie Clinique et Toxicologie-CHU de Besançon, 25000 Besançon, France
| | - Florentin Allemand
- EA 3920 Université Bourgogne Franche-Comté, 25000 Besançon, France; Laboratoire Chrono Environnement UMR CNRS 6249, Université de Bourgogne Franche-Comté, 16 route de Gray, 25000 Besançon, France
| | - Christophe Ramseyer
- Laboratoire Chrono Environnement UMR CNRS 6249, Université de Bourgogne Franche-Comté, 16 route de Gray, 25000 Besançon, France
| | - Semen Yesylevskyy
- Laboratoire Chrono Environnement UMR CNRS 6249, Université de Bourgogne Franche-Comté, 16 route de Gray, 25000 Besançon, France; Department of Physics of Biological Systems, Institute of Physics of The National Academy of Sciences of Ukraine, Nauky Sve. 46, Kyiv, Ukraine; Receptor.ai inc, 16192 Coastal Highway, Lewes, DE, USA
| | - Siamak Davani
- EA 3920 Université Bourgogne Franche-Comté, 25000 Besançon, France; Laboratoire de Pharmacologie Clinique et Toxicologie-CHU de Besançon, 25000 Besançon, France.
| |
Collapse
|
11
|
Lohitha N, Vijayakumar V. Imidazole Appended Novel Phenoxyquinolines as New Inhibitors of α-Amylase and α-Glucosidase Evidenced with Molecular Docking Studies. Polycycl Aromat Compd 2021. [DOI: 10.1080/10406638.2021.1939069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- N. Lohitha
- Centre for Organic and Medicinal Chemistry, VIT University, Vellore, India
| | - V. Vijayakumar
- Centre for Organic and Medicinal Chemistry, VIT University, Vellore, India
| |
Collapse
|
12
|
DeMarco KR, Yang PC, Singh V, Furutani K, Dawson JRD, Jeng MT, Fettinger JC, Bekker S, Ngo VA, Noskov SY, Yarov-Yarovoy V, Sack JT, Wulff H, Clancy CE, Vorobyov I. Molecular determinants of pro-arrhythmia proclivity of d- and l-sotalol via a multi-scale modeling pipeline. J Mol Cell Cardiol 2021; 158:163-177. [PMID: 34062207 PMCID: PMC8906354 DOI: 10.1016/j.yjmcc.2021.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/03/2021] [Accepted: 05/24/2021] [Indexed: 11/20/2022]
Abstract
Drug isomers may differ in their proarrhythmia risk. An interesting example is the drug sotalol, an antiarrhythmic drug comprising d- and l- enantiomers that both block the hERG cardiac potassium channel and confer differing degrees of proarrhythmic risk. We developed a multi-scale in silico pipeline focusing on hERG channel – drug interactions and used it to probe and predict the mechanisms of pro-arrhythmia risks of the two enantiomers of sotalol. Molecular dynamics (MD) simulations predicted comparable hERG channel binding affinities for d- and l-sotalol, which were validated with electrophysiology experiments. MD derived thermodynamic and kinetic parameters were used to build multi-scale functional computational models of cardiac electrophysiology at the cell and tissue scales. Functional models were used to predict inactivated state binding affinities to recapitulate electrocardiogram (ECG) QT interval prolongation observed in clinical data. Our study demonstrates how modeling and simulation can be applied to predict drug effects from the atom to the rhythm for dl-sotalol and also increased proarrhythmia proclivity of d- vs. l-sotalol when accounting for stereospecific beta-adrenergic receptor blocking.
Collapse
Affiliation(s)
- Kevin R DeMarco
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - Pei-Chi Yang
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - Vikrant Singh
- Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Kazuharu Furutani
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Tokushima 770-8514, Japan
| | - John R D Dawson
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Biophysics Graduate Group, University of California Davis, Davis, CA 95616, USA
| | - Mao-Tsuen Jeng
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - James C Fettinger
- Department of Chemistry, University of California Davis, Davis, CA 95616, USA
| | - Slava Bekker
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Science and Engineering, American River College, Sacramento, CA 95841, USA
| | - Van A Ngo
- Centre for Molecular Simulation and Biochemistry Research Cluster, Department of Biological Sciences, University of Calgary, Calgary, AB T2N1N4, Canada
| | - Sergei Y Noskov
- Centre for Molecular Simulation and Biochemistry Research Cluster, Department of Biological Sciences, University of Calgary, Calgary, AB T2N1N4, Canada
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, CA 95616, USA
| | - Jon T Sack
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, CA 95616, USA
| | - Heike Wulff
- Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Colleen E Clancy
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA; Department of Pharmacology, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
13
|
Yang PC, Giles WR, Belardinelli L, Clancy CE. Mechanisms of flecainide induced negative inotropy: An in silico study. J Mol Cell Cardiol 2021; 158:26-37. [PMID: 34004185 PMCID: PMC8772296 DOI: 10.1016/j.yjmcc.2021.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/27/2022]
Abstract
It is imperative to develop better approaches to predict how antiarrhythmic drugs with multiple interactions and targets may alter the overall electrical and/or mechanical function of the heart. Safety Pharmacology studies have provided new insights into the multi-target effects of many different classes of drugs and have been aided by the addition of robust new in vitro and in silico technology. The primary focus of Safety Pharmacology studies has been to determine the risk profile of drugs and drug candidates by assessing their effects on repolarization of the cardiac action potential. However, for decades experimental and clinical studies have described substantial and potentially detrimental effects of Na+ channel blockers in addition to their well-known conduction slowing effects. One such side effect, associated with administration of some Na+ channel blocking drugs is negative inotropy. This reduces the pumping function of the heart, thereby resulting in hypotension. Flecainide is a well-known example of a Na+ channel blocking drug, that exhibits strong rate-dependent block of INa and may cause negative cardiac inotropy. While the phenomenon of Na+ channel suppression and resulting negative inotropy is well described, the mechanism(s) underlying this effect are not. Here, we set out to use a modeling and simulation approach to reveal plausible mechanisms that could explain the negative inotropic effect of flecainide. We utilized the Grandi-Bers model [1] of the cardiac ventricular myocyte because of its robust descriptions of ion homeostasis in order to characterize and resolve the relative effects of QRS widening, flecainide off-target effects and changes in intracellular Ca2+ and Na+ homeostasis. The results of our investigations and predictions reconcile multiple data sets and illustrate how multiple mechanisms may play a contributing role in the flecainide induced negative cardiac inotropic effect.
Collapse
Affiliation(s)
- Pei-Chi Yang
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, United States of America
| | - Wayne R Giles
- Department of Physiology & Pharmacology, University of Calgary, Canada
| | | | - Colleen E Clancy
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, United States of America.
| |
Collapse
|
14
|
Atomistic Basis of Microtubule Dynamic Instability Assessed Via Multiscale Modeling. Ann Biomed Eng 2021; 49:1716-1734. [PMID: 33537926 PMCID: PMC8302526 DOI: 10.1007/s10439-020-02715-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023]
Abstract
Microtubule “dynamic instability,” the abrupt switching from assembly to disassembly caused by the hydrolysis of GTP to GDP within the β subunit of the αβ-tubulin heterodimer, is necessary for vital cellular processes such as mitosis and migration. Despite existing high-resolution structural data, the key mechanochemical differences between the GTP and GDP states that mediate dynamic instability behavior remain unclear. Starting with a published atomic-level structure as an input, we used multiscale modeling to find that GTP hydrolysis results in both longitudinal bond weakening (~ 4 kBT) and an outward bending preference (~ 1.5 kBT) to both drive dynamic instability and give rise to the microtubule tip structures previously observed by light and electron microscopy. More generally, our study provides an example where atomic level structural information is used as the sole input to predict cellular level dynamics without parameter adjustment.
Collapse
|
15
|
Abstract
In the past several decades, the establishment of in vitro models of pluripotency has ushered in a golden era for developmental and stem cell biology. Research in this arena has led to profound insights into the regulatory features that shape early embryonic development. Nevertheless, an integrative theory of the epigenetic principles that govern the pluripotent nucleus remains elusive. Here, we summarize the epigenetic characteristics that define the pluripotent state. We cover what is currently known about the epigenome of pluripotent stem cells and reflect on the use of embryonic stem cells as an experimental system. In addition, we highlight insights from super-resolution microscopy, which have advanced our understanding of the form and function of chromatin, particularly its role in establishing the characteristically "open chromatin" of pluripotent nuclei. Further, we discuss the rapid improvements in 3C-based methods, which have given us a means to investigate the 3D spatial organization of the pluripotent genome. This has aided the adaptation of prior notions of a "pluripotent molecular circuitry" into a more holistic model, where hotspots of co-interacting domains correspond with the accumulation of pluripotency-associated factors. Finally, we relate these earlier hypotheses to an emerging model of phase separation, which posits that a biophysical mechanism may presuppose the formation of a pluripotent-state-defining transcriptional program.
Collapse
Affiliation(s)
| | - Eran Meshorer
- Department of Genetics, the Institute of Life Sciences
- Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University of Jerusalem, Jerusalem, Israel 9190400
| |
Collapse
|
16
|
Koolman PM, Bukshtynov V. A multiscale optimization framework for reconstructing binary images using multilevel PCA-based control space reduction. Biomed Phys Eng Express 2021; 7:025005. [PMID: 33522496 DOI: 10.1088/2057-1976/abd4be] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
An efficient computational approach for optimal reconstructing parameters of binary-type physical properties for models in biomedical applications is developed and validated. The methodology includes gradient-based multiscale optimization with multilevel control space reduction by using principal component analysis (PCA) coupled with dynamical control space upscaling. The reduced dimensional controls are used interchangeably at fine and coarse scales to accumulate the optimization progress and mitigate side effects at both scales. Flexibility is achieved through the proposed procedure for calibrating certain parameters to enhance the performance of the optimization algorithm. Reduced size of control spaces supplied with adjoint-based gradients obtained at both scales facilitate the application of this algorithm to models of higher complexity and also to a broad range of problems in biomedical sciences. This technique is shown to outperform regular gradient-based methods applied to fine scale only in terms of both qualities of binary images and computing time. Performance of the complete computational framework is tested in applications to 2D inverse problems of cancer detection by the electrical impedance tomography (EIT). The results demonstrate the efficient performance of the new method and its high potential for minimizing possibilities for false positive screening and improving the overall quality of the EIT-based procedures.
Collapse
Affiliation(s)
- Priscilla M Koolman
- College of Engineering & Science, Florida Institute of Technology, Melbourne, FL 32901, United States of America
| | | |
Collapse
|
17
|
Benzekry S, Sentis C, Coze C, Tessonnier L, André N. Development and Validation of a Prediction Model of Overall Survival in High-Risk Neuroblastoma Using Mechanistic Modeling of Metastasis. JCO Clin Cancer Inform 2021; 5:81-90. [PMID: 33439729 DOI: 10.1200/cci.20.00092] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Prognosis of high-risk neuroblastoma (HRNB) remains poor despite multimodal therapies. Better prediction of survival could help to refine patient stratification and better tailor treatments. We established a mechanistic model of metastasis in HRNB relying on two processes: growth and dissemination relying on two patient-specific parameters: the dissemination rate μ and the minimal visible lesion size Svis. This model was calibrated using diagnosis values of primary tumor size, lactate dehydrogenase circulating levels, and the meta-iodobenzylguanidine International Society for Paediatric Oncology European (SIOPEN) score from nuclear imaging, using data from 49 metastatic patients. It was able to describe the data of total tumor mass (lactate dehydrogenase, R2 > 0.99) and number of visible metastases (SIOPEN, R2 = 0.96). A prediction model of overall survival (OS) was then developed using Cox regression. Clinical variables alone were not able to generate a model with sufficient OS prognosis ability (P = .507). The parameter μ was found to be independent of the clinical variables and positively associated with OS (P = .0739 in multivariable analysis). Critically, addition of this computational biomarker significantly improved prediction of OS with a concordance index increasing from 0.675 (95% CI, 0.663 to 0.688) to 0.733 (95% CI, 0.722 to 0.744, P < .0001), resulting in significant OS prognosis ability (P = .0422).
Collapse
Affiliation(s)
- Sébastien Benzekry
- MONC Team, Inria Bordeaux Sud-Ouest and Institut de Mathématiques de Bordeaux, CNRS, University of Bordeaux, Bordeaux, France
| | - Coline Sentis
- Paediatric Hematology and Oncology Department, Hôpital pour enfant de La Timone, AP-HM, Marseille, France
| | - Carole Coze
- Paediatric Hematology and Oncology Department, Hôpital pour enfant de La Timone, AP-HM, Marseille, France.,Aix Marseille University, Marseille, France
| | - Laëtitia Tessonnier
- Department of Nuclear Medicine, Hôpital de La Timone, AP-HM, Marseille, France
| | - Nicolas André
- Paediatric Hematology and Oncology Department, Hôpital pour enfant de La Timone, AP-HM, Marseille, France.,SMARTc Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068, Aix Marseille University, Marseille, France
| |
Collapse
|
18
|
Sahai N, Gogoi M, Ahmad N. Mathematical Modeling and Simulations for Developing Nanoparticle-Based Cancer Drug Delivery Systems: A Review. CURRENT PATHOBIOLOGY REPORTS 2021. [DOI: 10.1007/s40139-020-00219-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
19
|
Combination of "generalized Trotter operator splitting" and "quadratic adaptive algorithm" method for tradeoff among speedup, stability, and accuracy in the Markov chain model of sodium ion channels in the ventricular cell model. Med Biol Eng Comput 2020; 58:2131-2141. [PMID: 32676840 DOI: 10.1007/s11517-020-02220-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 06/25/2020] [Indexed: 10/23/2022]
Abstract
The fast hybrid operator splitting (HOS) and stable uniformization (UNI) methods have been proposed to save computation cost and enhance stability for Markov chain model in cardiac cell simulations. Moreover, Chen-Chen-Luo's quadratic adaptive algorithm (CCL) combined with HOS or UNI was used to improve the tradeoff between speedup and stability, but without considering accuracy. To compromise among stability, acceleration, and accuracy, we propose a generalized Trotter operator splitting (GTOS) method combined with CCL independent of the asymptotic property of a particular ion-channel model. Due to the accuracy underestimation of the mixed root mean square error (MRMSE) method, threshold root mean square error (TRMSE) is proposed to evaluate computation accuracy. With the fixed time-step RK4 as a reference, the second-order GTOS combined with CCL (30.8-fold speedup) for the wild-type Markov chain model with nine states (WT-9 model) or (7.4-fold) for the wild-type Markov chain model with eight states (WT-8 model) is faster than UNI combined with CCL (15.6-fold) for WT-9 model or (1.2-fold) for WT-8 model, separately. Besides, the second-order GTOS combined with CCL has 3.81% TRMSE for WT-9 model or 4.32% TRMSE for WT-8 model more accurate than 72.43% TRMSE for WT-9 model or 136.17% TRMSE for WT-8 model of HOS combined with CCL. To compromise speedup and accuracy, low-order GTOS combined with CCL is suggested to have the advantages of high precision and low computation cost. For high-accuracy requirements, high-order GTOS combined with CCL is recommended. Graphical abstract.
Collapse
|
20
|
Liu AC, Patel K, Vunikili RD, Johnson KW, Abdu F, Belman SK, Glicksberg BS, Tandale P, Fontanez R, Mathew OK, Kasarskis A, Mukherjee P, Subramanian L, Dudley JT, Shameer K. Sepsis in the era of data-driven medicine: personalizing risks, diagnoses, treatments and prognoses. Brief Bioinform 2020; 21:1182-1195. [PMID: 31190075 PMCID: PMC8179509 DOI: 10.1093/bib/bbz059] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 04/04/2019] [Accepted: 04/18/2019] [Indexed: 12/26/2022] Open
Abstract
Sepsis is a series of clinical syndromes caused by the immunological response to infection. The clinical evidence for sepsis could typically attribute to bacterial infection or bacterial endotoxins, but infections due to viruses, fungi or parasites could also lead to sepsis. Regardless of the etiology, rapid clinical deterioration, prolonged stay in intensive care units and high risk for mortality correlate with the incidence of sepsis. Despite its prevalence and morbidity, improvement in sepsis outcomes has remained limited. In this comprehensive review, we summarize the current landscape of risk estimation, diagnosis, treatment and prognosis strategies in the setting of sepsis and discuss future challenges. We argue that the advent of modern technologies such as in-depth molecular profiling, biomedical big data and machine intelligence methods will augment the treatment and prevention of sepsis. The volume, variety, veracity and velocity of heterogeneous data generated as part of healthcare delivery and recent advances in biotechnology-driven therapeutics and companion diagnostics may provide a new wave of approaches to identify the most at-risk sepsis patients and reduce the symptom burden in patients within shorter turnaround times. Developing novel therapies by leveraging modern drug discovery strategies including computational drug repositioning, cell and gene-therapy, clustered regularly interspaced short palindromic repeats -based genetic editing systems, immunotherapy, microbiome restoration, nanomaterial-based therapy and phage therapy may help to develop treatments to target sepsis. We also provide empirical evidence for potential new sepsis targets including FER and STARD3NL. Implementing data-driven methods that use real-time collection and analysis of clinical variables to trace, track and treat sepsis-related adverse outcomes will be key. Understanding the root and route of sepsis and its comorbid conditions that complicate treatment outcomes and lead to organ dysfunction may help to facilitate identification of most at-risk patients and prevent further deterioration. To conclude, leveraging the advances in precision medicine, biomedical data science and translational bioinformatics approaches may help to develop better strategies to diagnose and treat sepsis in the next decade.
Collapse
Affiliation(s)
- Andrew C Liu
- Department of Information Services, Northwell Health, New Hyde Park, NY, USA
- Donald and Barbara School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY, USA
| | - Krishna Patel
- Department of Information Services, Northwell Health, New Hyde Park, NY, USA
- Donald and Barbara School of Medicine at Hofstra/Northwell, Northwell Health, Hempstead, NY, USA
| | - Ramya Dhatri Vunikili
- Center for Research Informatics and Innovation, Northwell Health, New Hyde Park, NY, USA
- Courant Institute of Mathematical Sciences, New York University, New York, NY, USA
| | - Kipp W Johnson
- Department of Genetics and Genomic Sciences, Mount Sinai Health System, New York, NY, USA
- Institute for Next Generation Healthcare, Mount Sinai Health System, New York, NY, USA
| | - Fahad Abdu
- Center for Research Informatics and Innovation, Northwell Health, New Hyde Park, NY, USA
- Stonybrook University, 100 Nicolls Rd, Stony Brook, NY, USA
| | - Shivani Kamath Belman
- Center for Research Informatics and Innovation, Northwell Health, New Hyde Park, NY, USA
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Benjamin S Glicksberg
- Department of Genetics and Genomic Sciences, Mount Sinai Health System, New York, NY, USA
- Institute for Next Generation Healthcare, Mount Sinai Health System, New York, NY, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Pratyush Tandale
- Center for Research Informatics and Innovation, Northwell Health, New Hyde Park, NY, USA
- School of Biotechnology and Bioinformatics, D Y Patil University, Navi Mumbai, India
| | - Roberto Fontanez
- Department of Information Services, Northwell Health, New Hyde Park, NY, USA
- Center for Research Informatics and Innovation, Northwell Health, New Hyde Park, NY, USA
| | | | - Andrew Kasarskis
- Department of Genetics and Genomic Sciences, Mount Sinai Health System, New York, NY, USA
| | | | | | - Joel T Dudley
- Department of Genetics and Genomic Sciences, Mount Sinai Health System, New York, NY, USA
- Institute for Next Generation Healthcare, Mount Sinai Health System, New York, NY, USA
| | - Khader Shameer
- Department of Information Services, Northwell Health, New Hyde Park, NY, USA
- Center for Research Informatics and Innovation, Northwell Health, New Hyde Park, NY, USA
- Institute for Next Generation Healthcare, Mount Sinai Health System, New York, NY, USA
| |
Collapse
|
21
|
Pruett WA, Clemmer JS, Hester RL. Physiological Modeling and Simulation-Validation, Credibility, and Application. Annu Rev Biomed Eng 2020; 22:185-206. [PMID: 32501771 DOI: 10.1146/annurev-bioeng-082219-051740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In this review, we discuss the science of model validation as it applies to physiological modeling. There is widespread disagreement and ambiguity about what constitutes model validity. In areas in which models affect real-world decision-making, including within the clinic, in regulatory science, or in the design and engineering of novel therapeutics, this question is of critical importance. Without an answer, it impairs the usefulness of models and casts a shadow over model credibility in all domains. To address this question, we examine the use of nonmathematical models in physiological research, in medical practice, and in engineering to see how models in other domains are used and accepted. We reflect on historic physiological models and how they have been presented to the scientific community. Finally, we look at various validation frameworks that have been proposed as potential solutions during the past decade.
Collapse
Affiliation(s)
- W Andrew Pruett
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA; , ,
| | - John S Clemmer
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA; , ,
| | - Robert L Hester
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA; , , .,John D. Bower School of Population Health, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA
| |
Collapse
|
22
|
Ali I, Mukhtar SD, Ali HS, Scotti MT, Scotti L. Advances in Nanoparticles as Anticancer Drug Delivery Vector: Need of this Century. Curr Pharm Des 2020; 26:1637-1649. [DOI: 10.2174/1381612826666200203124330] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/02/2019] [Indexed: 12/17/2022]
Abstract
Background:
Nanotechnology has contributed a great deal to the field of medical science. Smart drugdelivery
vectors, combined with stimuli-based characteristics, are becoming increasingly important. The use of
external and internal stimulating factors can have enormous benefits and increase the targeting efficiency of
nanotechnology platforms. The pH values of tumor vascular tissues are acidic in nature, allowing the improved
targeting of anticancer drug payloads using drug-delivery vectors. Nanopolymers are smart drug-delivery vectors
that have recently been developed and recommended for use by scientists because of their potential targeting
capabilities, non-toxicity and biocompatibility, and make them ideal nanocarriers for personalized drug delivery.
Method:
The present review article provides an overview of current advances in the use of nanoparticles (NPs) as
anticancer drug-delivery vectors.
Results:
This article reviews the molecular basis for the use of NPs in medicine, including personalized medicine,
personalized therapy, emerging vistas in anticancer therapy, nanopolymer targeting, passive and active targeting
transports, pH-responsive drug carriers, biological barriers, computer-aided drug design, future challenges and
perspectives, biodegradability and safety.
Conclusions:
This article will benefit academia, researchers, clinicians, and government authorities by providing a
basis for further research advancements.
Collapse
Affiliation(s)
- Imran Ali
- Department of Chemistry, College of Sciences, Taibah University, Al-Medina Al-Munawara – 41477, Saudi Arabia
| | - Sofi D. Mukhtar
- Department of Chemistry, Jamia Millia Islamia (Central University) New Delhi-110025, India
| | - Heyam S. Ali
- Department of Pharmaceutics, University of Khartoum, Khartoum, Sudan
| | - Marcus T. Scotti
- Cheminformatics Laboratory- Postgraduate Program in Natural Products and Synthetic Bioactive, Federal University of Paraíba-Campus I 58051-970, João Pessoa, PB, Brazil
| | - Luciana Scotti
- Teaching and Research Management - University Hospital, Cheminformatics Laboratory- Postgraduate Program in Natural Products and Synthetic Bioactive, Federal University of Paraíba-Campus I, 58051-970, João Pessoa, PB, Brazil
| |
Collapse
|
23
|
Dogra P, Butner JD, Nizzero S, Ruiz Ramírez J, Noureddine A, Peláez MJ, Elganainy D, Yang Z, Le AD, Goel S, Leong HS, Koay EJ, Brinker CJ, Cristini V, Wang Z. Image-guided mathematical modeling for pharmacological evaluation of nanomaterials and monoclonal antibodies. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1628. [PMID: 32314552 PMCID: PMC7507140 DOI: 10.1002/wnan.1628] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/06/2020] [Accepted: 02/15/2020] [Indexed: 12/13/2022]
Abstract
While plasma concentration kinetics has traditionally been the predictor of drug pharmacological effects, it can occasionally fail to represent kinetics at the site of action, particularly for solid tumors. This is especially true in the case of delivery of therapeutic macromolecules (drug-loaded nanomaterials or monoclonal antibodies), which can experience challenges to effective delivery due to particle size-dependent diffusion barriers at the target site. As a result, disparity between therapeutic plasma kinetics and kinetics at the site of action may exist, highlighting the importance of target site concentration kinetics in determining the pharmacodynamic effects of macromolecular therapeutic agents. Assessment of concentration kinetics at the target site has been facilitated by non-invasive in vivo imaging modalities. This allows for visualization and quantification of the whole-body disposition behavior of therapeutics that is essential for a comprehensive understanding of their pharmacokinetics and pharmacodynamics. Quantitative non-invasive imaging can also help guide the development and parameterization of mathematical models for descriptive and predictive purposes. Here, we present a review of the application of state-of-the-art imaging modalities for quantitative pharmacological evaluation of therapeutic nanoparticles and monoclonal antibodies, with a focus on their integration with mathematical models, and identify challenges and opportunities. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Joseph D Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Sara Nizzero
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Achraf Noureddine
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - María J Peláez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA.,Applied Physics Graduate Program, Rice University, Houston, Texas, USA
| | - Dalia Elganainy
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhen Yang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Anh-Dung Le
- Nanoscience and Microsystems Engineering, University of New Mexico, Albuquerque, New Mexico, USA
| | - Shreya Goel
- Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hon S Leong
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Eugene J Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - C Jeffrey Brinker
- Department of Chemical and Biological Engineering and UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
24
|
Herland A, Maoz BM, Das D, Somayaji MR, Prantil-Baun R, Novak R, Cronce M, Huffstater T, Jeanty SSF, Ingram M, Chalkiadaki A, Benson Chou D, Marquez S, Delahanty A, Jalili-Firoozinezhad S, Milton Y, Sontheimer-Phelps A, Swenor B, Levy O, Parker KK, Przekwas A, Ingber DE. Quantitative prediction of human pharmacokinetic responses to drugs via fluidically coupled vascularized organ chips. Nat Biomed Eng 2020; 4:421-436. [PMID: 31988459 PMCID: PMC8011576 DOI: 10.1038/s41551-019-0498-9] [Citation(s) in RCA: 284] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 11/25/2019] [Indexed: 01/15/2023]
Abstract
Analyses of drug pharmacokinetics (PKs) and pharmacodynamics (PDs) performed in animals are often not predictive of drug PKs and PDs in humans, and in vitro PK and PD modelling does not provide quantitative PK parameters. Here, we show that physiological PK modelling of first-pass drug absorption, metabolism and excretion in humans-using computationally scaled data from multiple fluidically linked two-channel organ chips-predicts PK parameters for orally administered nicotine (using gut, liver and kidney chips) and for intravenously injected cisplatin (using coupled bone marrow, liver and kidney chips). The chips are linked through sequential robotic liquid transfers of a common blood substitute by their endothelium-lined channels (as reported by Novak et al. in an associated Article) and share an arteriovenous fluid-mixing reservoir. We also show that predictions of cisplatin PDs match previously reported patient data. The quantitative in-vitro-to-in-vivo translation of PK and PD parameters and the prediction of drug absorption, distribution, metabolism, excretion and toxicity through fluidically coupled organ chips may improve the design of drug-administration regimens for phase-I clinical trials.
Collapse
Affiliation(s)
- Anna Herland
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- AIMES, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Ben M Maoz
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Debarun Das
- CFD Research Corporation, Huntsville, AL, USA
| | | | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Richard Novak
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Michael Cronce
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Tessa Huffstater
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sauveur S F Jeanty
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Miles Ingram
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Angeliki Chalkiadaki
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - David Benson Chou
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Susan Marquez
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Aaron Delahanty
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sasan Jalili-Firoozinezhad
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Portugal Graduate Program, Universidade de Lisboa, Lisbon, Portugal
| | - Yuka Milton
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Alexandra Sontheimer-Phelps
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ben Swenor
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Oren Levy
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Kevin K Parker
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | | | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
25
|
Lin X, Li X, Lin X. A Review on Applications of Computational Methods in Drug Screening and Design. Molecules 2020; 25:E1375. [PMID: 32197324 PMCID: PMC7144386 DOI: 10.3390/molecules25061375] [Citation(s) in RCA: 278] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/16/2020] [Accepted: 03/16/2020] [Indexed: 12/27/2022] Open
Abstract
Drug development is one of the most significant processes in the pharmaceutical industry. Various computational methods have dramatically reduced the time and cost of drug discovery. In this review, we firstly discussed roles of multiscale biomolecular simulations in identifying drug binding sites on the target macromolecule and elucidating drug action mechanisms. Then, virtual screening methods (e.g., molecular docking, pharmacophore modeling, and QSAR) as well as structure- and ligand-based classical/de novo drug design were introduced and discussed. Last, we explored the development of machine learning methods and their applications in aforementioned computational methods to speed up the drug discovery process. Also, several application examples of combining various methods was discussed. A combination of different methods to jointly solve the tough problem at different scales and dimensions will be an inevitable trend in drug screening and design.
Collapse
Affiliation(s)
- Xiaoqian Lin
- Institute of Single Cell Engineering, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China;
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xiu Li
- School of Chemistry and Material Science, Shanxi Normal University, Linfen 041004, China;
| | - Xubo Lin
- Institute of Single Cell Engineering, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China;
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
26
|
Facile Fabrication of Composite Scaffolds for Long-Term Controlled Dual Drug Release. ADVANCES IN POLYMER TECHNOLOGY 2020. [DOI: 10.1155/2020/3927860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone tuberculosis (TB) caused by mycobacterium tuberculosis continues to present a formidable challenge to humans. To effectively cure serious bone TB, a novel kind of composite scaffolds with long-term dual drug release behaviours were prepared to satisfy the needs of both bone regeneration and antituberculosis drug therapy. In virtue of an improved O/W emulsion technique, water-soluble isoniazid (INH)-loaded gelatin microparticles were obtained by tailoring the content of β-tricalcium phosphate (β-TCP), which played significant roles in INH entrapment efficiency and drug release behaviours. By mixing with the poly(ε-caprolactone)-block-poly (lactic-co-glycolic acid) (b-PLGC) solution containing oil-soluble rifampicin (RFP) via the particle leaching combined with phase separation technique, the dual drugs-loaded composite scaffolds were fabricated, which possessed interconnected porous structures and achieved the steady release of INH and RFP drugs for three months. Moreover, this dual drugs-loaded system could basically achieve their expectant roles of respective drugs without obvious influences with each other. This strategy on preparation of intelligent composite scaffolds with the multi-drugs loading capacity and controlled long-term release behaviour will be potential and promising substrates in clinical treatment of bone tuberculosis.
Collapse
|
27
|
Shamsi M, Mohammadi A, Manshadi MK, Sanati-Nezhad A. Mathematical and computational modeling of nano-engineered drug delivery systems. J Control Release 2019; 307:150-165. [DOI: 10.1016/j.jconrel.2019.06.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 12/20/2022]
|
28
|
Kudaibergenova M, Perissinotti LL, Noskov SY. Lipid roles in hERG function and interactions with drugs. Neurosci Lett 2019; 700:70-77. [DOI: 10.1016/j.neulet.2018.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/08/2018] [Accepted: 05/11/2018] [Indexed: 01/29/2023]
|
29
|
Sherman TD, Kagohara LT, Cao R, Cheng R, Satriano M, Considine M, Krigsfeld G, Ranaweera R, Tang Y, Jablonski SA, Stein-O'Brien G, Gaykalova DA, Weiner LM, Chung CH, Fertig EJ. CancerInSilico: An R/Bioconductor package for combining mathematical and statistical modeling to simulate time course bulk and single cell gene expression data in cancer. PLoS Comput Biol 2019; 14:e1006935. [PMID: 31002670 PMCID: PMC6504085 DOI: 10.1371/journal.pcbi.1006935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 05/07/2019] [Accepted: 03/11/2019] [Indexed: 11/18/2022] Open
Abstract
Bioinformatics techniques to analyze time course bulk and single cell omics data
are advancing. The absence of a known ground truth of the dynamics of molecular
changes challenges benchmarking their performance on real data. Realistic
simulated time-course datasets are essential to assess the performance of time
course bioinformatics algorithms. We develop an R/Bioconductor package,
CancerInSilico, to simulate bulk and single cell
transcriptional data from a known ground truth obtained from mathematical models
of cellular systems. This package contains a general R infrastructure for
running cell-based models and simulating gene expression data based on the model
states. We show how to use this package to simulate a gene expression data set
and consequently benchmark analysis methods on this data set with a known ground
truth. The package is freely available via Bioconductor: http://bioconductor.org/packages/CancerInSilico/
Collapse
Affiliation(s)
- Thomas D. Sherman
- Department of Oncology, Division of Biostatistics and Bioinformatics,
Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore,
MD United States of America
- * E-mail:
(TDS); (EJF)
| | - Luciane T. Kagohara
- Department of Oncology, Division of Biostatistics and Bioinformatics,
Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore,
MD United States of America
| | - Raymon Cao
- Department of Oncology, Division of Biostatistics and Bioinformatics,
Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore,
MD United States of America
| | - Raymond Cheng
- Science, Math and Computer Science Magnet Program, Poolesville High
School, Poolesville, MD United States of America
| | - Matthew Satriano
- Department of Mathematics, University of Waterloo, Waterloo, Ontario,
Canada
| | - Michael Considine
- Department of Oncology, Division of Biostatistics and Bioinformatics,
Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore,
MD United States of America
| | - Gabriel Krigsfeld
- Department of Oncology, Division of Biostatistics and Bioinformatics,
Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore,
MD United States of America
| | | | - Yong Tang
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington,
DC United States of America
| | - Sandra A. Jablonski
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington,
DC United States of America
| | - Genevieve Stein-O'Brien
- Department of Oncology, Division of Biostatistics and Bioinformatics,
Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore,
MD United States of America
- Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD
United States of America
| | - Daria A. Gaykalova
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins
University School of Medicine, Baltimore, MD United States of
America
| | - Louis M. Weiner
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington,
DC United States of America
| | | | - Elana J. Fertig
- Department of Oncology, Division of Biostatistics and Bioinformatics,
Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore,
MD United States of America
- Department of Applied Mathematics and Statistics, Johns Hopkins
University, Baltimore, MD United States of America
- Department of Biomedical Engineering, Johns Hopkins University,
Baltimore, MD United States of America
- * E-mail:
(TDS); (EJF)
| |
Collapse
|
30
|
DeMarco KR, Bekker S, Vorobyov I. Challenges and advances in atomistic simulations of potassium and sodium ion channel gating and permeation. J Physiol 2018; 597:679-698. [PMID: 30471114 DOI: 10.1113/jp277088] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 10/15/2018] [Indexed: 12/19/2022] Open
Abstract
Ion channels are implicated in many essential physiological events such as electrical signal propagation and cellular communication. The advent of K+ and Na+ ion channel structure determination has facilitated numerous investigations of molecular determinants of their behaviour. At the same time, rapid development of computer hardware and molecular simulation methodologies has made computational studies of large biological molecules in all-atom representation tractable. The concurrent evolution of experimental structural biology with biomolecular computer modelling has yielded mechanistic details of fundamental processes unavailable through experiments alone, such as ion conduction and ion channel gating. This review is a short survey of the atomistic computational investigations of K+ and Na+ ion channels, focusing on KcsA and several voltage-gated channels from the KV and NaV families, which have garnered many successes and engendered several long-standing controversies regarding the nature of their structure-function relationship. We review the latest advancements and challenges facing the field of molecular modelling and simulation regarding the structural and energetic determinants of ion channel function and their agreement with experimental observations.
Collapse
Affiliation(s)
- Kevin R DeMarco
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA.,Department of Pharmacology, School of Medicine, University of California, Davis, CA, USA
| | - Slava Bekker
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA.,Chemistry Department, American River College, Sacramento, CA, USA
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA.,Department of Pharmacology, School of Medicine, University of California, Davis, CA, USA
| |
Collapse
|
31
|
Vagos M, van Herck IGM, Sundnes J, Arevalo HJ, Edwards AG, Koivumäki JT. Computational Modeling of Electrophysiology and Pharmacotherapy of Atrial Fibrillation: Recent Advances and Future Challenges. Front Physiol 2018; 9:1221. [PMID: 30233399 PMCID: PMC6131668 DOI: 10.3389/fphys.2018.01221] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022] Open
Abstract
The pathophysiology of atrial fibrillation (AF) is broad, with components related to the unique and diverse cellular electrophysiology of atrial myocytes, structural complexity, and heterogeneity of atrial tissue, and pronounced disease-associated remodeling of both cells and tissue. A major challenge for rational design of AF therapy, particularly pharmacotherapy, is integrating these multiscale characteristics to identify approaches that are both efficacious and independent of ventricular contraindications. Computational modeling has long been touted as a basis for achieving such integration in a rapid, economical, and scalable manner. However, computational pipelines for AF-specific drug screening are in their infancy, and while the field is progressing quite rapidly, major challenges remain before computational approaches can fill the role of workhorse in rational design of AF pharmacotherapies. In this review, we briefly detail the unique aspects of AF pathophysiology that determine requirements for compounds targeting AF rhythm control, with emphasis on delimiting mechanisms that promote AF triggers from those providing substrate or supporting reentry. We then describe modeling approaches that have been used to assess the outcomes of drugs acting on established AF targets, as well as on novel promising targets including the ultra-rapidly activating delayed rectifier potassium current, the acetylcholine-activated potassium current and the small conductance calcium-activated potassium channel. Finally, we describe how heterogeneity and variability are being incorporated into AF-specific models, and how these approaches are yielding novel insights into the basic physiology of disease, as well as aiding identification of the important molecular players in the complex AF etiology.
Collapse
Affiliation(s)
- Márcia Vagos
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Ilsbeth G. M. van Herck
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Joakim Sundnes
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Center for Cardiological Innovation, Oslo, Norway
| | - Hermenegild J. Arevalo
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Center for Cardiological Innovation, Oslo, Norway
| | - Andrew G. Edwards
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Center for Cardiological Innovation, Oslo, Norway
| | - Jussi T. Koivumäki
- BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
32
|
Oduola WO, Li X. Multiscale Tumor Modeling With Drug Pharmacokinetic and Pharmacodynamic Profile Using Stochastic Hybrid System. Cancer Inform 2018; 17:1176935118790262. [PMID: 30083052 PMCID: PMC6073835 DOI: 10.1177/1176935118790262] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/16/2018] [Indexed: 12/16/2022] Open
Abstract
Effective cancer treatment strategy requires an understanding of cancer behavior and development across multiple temporal and spatial scales. This has resulted into a growing interest in developing multiscale mathematical models that can simulate cancer growth, development, and response to drug treatments. This study thus investigates multiscale tumor modeling that integrates drug pharmacokinetic and pharmacodynamic (PK/PD) information using stochastic hybrid system modeling framework. Specifically, (1) pathways modeled by differential equations are adopted for gene regulations at the molecular level; (2) cellular automata (CA) model is proposed for the cellular and multicellular scales. Markov chains are used to model the cell behaviors by taking into account the gene expression levels, cell cycle, and the microenvironment. The proposed model enables the prediction of tumor growth under given molecular properties, microenvironment conditions, and drug PK/PD profile. Simulation results demonstrate the effectiveness of the proposed approach and the results agree with observed tumor behaviors.
Collapse
Affiliation(s)
- Wasiu Opeyemi Oduola
- Department of Electrical and Computer Engineering (ECE), Prairie View A&M University, Prairie View, TX, USA
| | - Xiangfang Li
- Department of Electrical and Computer Engineering (ECE), Prairie View A&M University, Prairie View, TX, USA
| |
Collapse
|
33
|
Ceresa M, Olivares AL, Noailly J, González Ballester MA. Coupled Immunological and Biomechanical Model of Emphysema Progression. Front Physiol 2018; 9:388. [PMID: 29725304 PMCID: PMC5917021 DOI: 10.3389/fphys.2018.00388] [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: 12/15/2017] [Accepted: 03/28/2018] [Indexed: 12/16/2022] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a disabling respiratory pathology, with a high prevalence and a significant economic and social cost. It is characterized by different clinical phenotypes with different risk profiles. Detecting the correct phenotype, especially for the emphysema subtype, and predicting the risk of major exacerbations are key elements in order to deliver more effective treatments. However, emphysema onset and progression are influenced by a complex interaction between the immune system and the mechanical properties of biological tissue. The former causes chronic inflammation and tissue remodeling. The latter influences the effective resistance or appropriate mechanical response of the lung tissue to repeated breathing cycles. In this work we present a multi-scale model of both aspects, coupling Finite Element (FE) and Agent Based (AB) techniques that we would like to use to predict the onset and progression of emphysema in patients. The AB part is based on existing biological models of inflammation and immunological response as a set of coupled non-linear differential equations. The FE part simulates the biomechanical effects of repeated strain on the biological tissue. We devise a strategy to couple the discrete biological model at the molecular /cellular level and the biomechanical finite element simulations at the tissue level. We tested our implementation on a public emphysema image database and found that it can indeed simulate the evolution of clinical image biomarkers during disease progression.
Collapse
Affiliation(s)
- Mario Ceresa
- BCN-Medtech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Andy L Olivares
- BCN-Medtech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Jérôme Noailly
- BCN-Medtech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Miguel A González Ballester
- BCN-Medtech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain.,ICREA, Barcelona, Spain
| |
Collapse
|
34
|
DeMarco KR, Bekker S, Clancy CE, Noskov SY, Vorobyov I. Digging into Lipid Membrane Permeation for Cardiac Ion Channel Blocker d-Sotalol with All-Atom Simulations. Front Pharmacol 2018; 9:26. [PMID: 29449809 PMCID: PMC5799612 DOI: 10.3389/fphar.2018.00026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/10/2018] [Indexed: 12/31/2022] Open
Abstract
Interactions of drug molecules with lipid membranes play crucial role in their accessibility of cellular targets and can be an important predictor of their therapeutic and safety profiles. Very little is known about spatial localization of various drugs in the lipid bilayers, their active form (ionization state) or translocation rates and therefore potency to bind to different sites in membrane proteins. All-atom molecular simulations may help to map drug partitioning kinetics and thermodynamics, thus providing in-depth assessment of drug lipophilicity. As a proof of principle, we evaluated extensively lipid membrane partitioning of d-sotalol, well-known blocker of a cardiac potassium channel Kv11.1 encoded by the hERG gene, with reported substantial proclivity for arrhythmogenesis. We developed the positively charged (cationic) and neutral d-sotalol models, compatible with the biomolecular CHARMM force field, and subjected them to all-atom molecular dynamics (MD) simulations of drug partitioning through hydrated lipid membranes, aiming to elucidate thermodynamics and kinetics of their translocation and thus putative propensities for hydrophobic and aqueous hERG access. We found that only a neutral form of d-sotalol accumulates in the membrane interior and can move across the bilayer within millisecond time scale, and can be relevant to a lipophilic channel access. The computed water-membrane partitioning coefficient for this form is in good agreement with experiment. There is a large energetic barrier for a cationic form of the drug, dominant in water, to cross the membrane, resulting in slow membrane translocation kinetics. However, this form of the drug can be important for an aqueous access pathway through the intracellular gate of hERG. This route will likely occur after a neutral form of a drug crosses the membrane and subsequently re-protonates. Our study serves to demonstrate a first step toward a framework for multi-scale in silico safety pharmacology, and identifies some of the challenges that lie therein.
Collapse
Affiliation(s)
- Kevin R DeMarco
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States.,Department of Pharmacology, University of California, Davis, Davis, CA, United States.,Biophysics Graduate Group, University of California, Davis, Davis, CA, United States
| | - Slava Bekker
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States.,Hartnell College, Salinas, CA, United States
| | - Colleen E Clancy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States.,Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Sergei Y Noskov
- Centre for Molecular Simulations, Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States.,Department of Pharmacology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
35
|
Computational analysis of the mesenchymal signature landscape in gliomas. BMC Med Genomics 2017; 10:13. [PMID: 28279210 PMCID: PMC5345226 DOI: 10.1186/s12920-017-0252-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/03/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Epithelial to mesenchymal transition, and mimicking processes, contribute to cancer invasion and metastasis, and are known to be responsible for resistance to various therapeutic agents in many cancers. While a number of studies have proposed molecular signatures that characterize the spectrum of such transition, more work is needed to understand how the mesenchymal signature (MS) is regulated in non-epithelial cancers like gliomas, to identify markers with the most prognostic significance, and potential for therapeutic targeting. RESULTS Computational analysis of 275 glioma samples from "The Cancer Genome Atlas" was used to identify the regulatory changes between low grade gliomas with little expression of MS, and high grade glioblastomas with high expression of MS. TF (transcription factor)-gene regulatory networks were constructed for each of the cohorts, and 5 major pathways and 118 transcription factors were identified as involved in the differential regulation of the networks. The most significant pathway - Extracellular matrix organization - was further analyzed for prognostic relevance. A 20-gene signature was identified as having prognostic significance (HR (hazard ratio) 3.2, 95% CI (confidence interval) = 1.53-8.33), after controlling for known prognostic factors (age, and glioma grade). The signature's significance was validated in an independent data set. The putative stem cell marker CD44 was biologically validated in glioma cell lines and brain tissue samples. CONCLUSIONS Our results suggest that the differences between low grade gliomas and high grade glioblastoma are associated with differential expression of the signature genes, raising the possibility that targeting these genes might prolong survival in glioma patients.
Collapse
|
36
|
Affiliation(s)
- Zhihui Wang
- Center for Precision Biomedicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, (UTHealth) McGovern Medical School, Houston, TX 77030, USA
| | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK
| |
Collapse
|
37
|
Chiamvimonvat N, Chen-Izu Y, Clancy CE, Deschenes I, Dobrev D, Heijman J, Izu L, Qu Z, Ripplinger CM, Vandenberg JI, Weiss JN, Koren G, Banyasz T, Grandi E, Sanguinetti MC, Bers DM, Nerbonne JM. Potassium currents in the heart: functional roles in repolarization, arrhythmia and therapeutics. J Physiol 2017; 595:2229-2252. [PMID: 27808412 DOI: 10.1113/jp272883] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/11/2016] [Indexed: 12/19/2022] Open
Abstract
This is the second of the two White Papers from the fourth UC Davis Cardiovascular Symposium Systems Approach to Understanding Cardiac Excitation-Contraction Coupling and Arrhythmias (3-4 March 2016), a biennial event that brings together leading experts in different fields of cardiovascular research. The theme of the 2016 symposium was 'K+ channels and regulation', and the objectives of the conference were severalfold: (1) to identify current knowledge gaps; (2) to understand what may go wrong in the diseased heart and why; (3) to identify possible novel therapeutic targets; and (4) to further the development of systems biology approaches to decipher the molecular mechanisms and treatment of cardiac arrhythmias. The sessions of the Symposium focusing on the functional roles of the cardiac K+ channel in health and disease, as well as K+ channels as therapeutic targets, were contributed by Ye Chen-Izu, Gideon Koren, James Weiss, David Paterson, David Christini, Dobromir Dobrev, Jordi Heijman, Thomas O'Hara, Crystal Ripplinger, Zhilin Qu, Jamie Vandenberg, Colleen Clancy, Isabelle Deschenes, Leighton Izu, Tamas Banyasz, Andras Varro, Heike Wulff, Eleonora Grandi, Michael Sanguinetti, Donald Bers, Jeanne Nerbonne and Nipavan Chiamvimonvat as speakers and panel discussants. This article summarizes state-of-the-art knowledge and controversies on the functional roles of cardiac K+ channels in normal and diseased heart. We endeavour to integrate current knowledge at multiple scales, from the single cell to the whole organ levels, and from both experimental and computational studies.
Collapse
Affiliation(s)
- Nipavan Chiamvimonvat
- Department of Internal Medicine, University of California, Davis, Genome and Biomedical Science Facility, Rm 6315, Davis, CA, 95616, USA.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, 95655, USA
| | - Ye Chen-Izu
- Department of Internal Medicine, University of California, Davis, Genome and Biomedical Science Facility, Rm 6315, Davis, CA, 95616, USA.,Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA.,Department of Biomedical Engineering, University of California, Davis, Genome and Biomedical Science Facility, Rm 2303, Davis, CA, 95616, USA
| | - Colleen E Clancy
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Isabelle Deschenes
- Department of Physiology and Biophysics, and Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44109, USA.,Heart and Vascular Research Center, MetroHealth Medical Center, Cleveland, OH, 44109, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Leighton Izu
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Zhilin Qu
- Division of Cardiology, Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, 3645 MRL, Los Angeles, CA, 90095, USA
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Jamie I Vandenberg
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW, 2010, Australia
| | - James N Weiss
- Division of Cardiology, Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, 3645 MRL, Los Angeles, CA, 90095, USA
| | - Gideon Koren
- Cardiovascular Research Center, Rhode Island Hospital and the Cardiovascular Institute, The Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Tamas Banyasz
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eleonora Grandi
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Michael C Sanguinetti
- Department of Internal Medicine, University of Utah, Nora Eccles Harrison Cardiovascular Research & Training Institute, Salt Lake City, UT, 84112, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Jeanne M Nerbonne
- Departments of Developmental Biology and Internal Medicine, Cardiovascular Division, Washington University Medical School, St Louis, MO, 63110, USA
| |
Collapse
|
38
|
Cheng M, Bhujwalla ZM, Glunde K. Targeting Phospholipid Metabolism in Cancer. Front Oncol 2016; 6:266. [PMID: 28083512 PMCID: PMC5187387 DOI: 10.3389/fonc.2016.00266] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 12/14/2016] [Indexed: 12/14/2022] Open
Abstract
All cancers tested so far display abnormal choline and ethanolamine phospholipid metabolism, which has been detected with numerous magnetic resonance spectroscopy (MRS) approaches in cells, animal models of cancer, as well as the tumors of cancer patients. Since the discovery of this metabolic hallmark of cancer, many studies have been performed to elucidate the molecular origins of deregulated choline metabolism, to identify targets for cancer treatment, and to develop MRS approaches that detect choline and ethanolamine compounds for clinical use in diagnosis and treatment monitoring. Several enzymes in choline, and recently also ethanolamine, phospholipid metabolism have been identified, and their evaluation has shown that they are involved in carcinogenesis and tumor progression. Several already established enzymes as well as a number of emerging enzymes in phospholipid metabolism can be used as treatment targets for anticancer therapy, either alone or in combination with other chemotherapeutic approaches. This review summarizes the current knowledge of established and relatively novel targets in phospholipid metabolism of cancer, covering choline kinase α, phosphatidylcholine-specific phospholipase D1, phosphatidylcholine-specific phospholipase C, sphingomyelinases, choline transporters, glycerophosphodiesterases, phosphatidylethanolamine N-methyltransferase, and ethanolamine kinase. These enzymes are discussed in terms of their roles in oncogenic transformation, tumor progression, and crucial cancer cell properties such as fast proliferation, migration, and invasion. Their potential as treatment targets are evaluated based on the current literature.
Collapse
Affiliation(s)
- Menglin Cheng
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine Glunde
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
39
|
Nagaraja S, Chen L, Zhou J, Zhao Y, Fine D, DiPietro LA, Reifman J, Mitrophanov AY. Predictive Analysis of Mechanistic Triggers and Mitigation Strategies for Pathological Scarring in Skin Wounds. THE JOURNAL OF IMMUNOLOGY 2016; 198:832-841. [DOI: 10.4049/jimmunol.1601273] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/15/2016] [Indexed: 12/17/2022]
|
40
|
Tebani A, Afonso C, Marret S, Bekri S. Omics-Based Strategies in Precision Medicine: Toward a Paradigm Shift in Inborn Errors of Metabolism Investigations. Int J Mol Sci 2016; 17:ijms17091555. [PMID: 27649151 PMCID: PMC5037827 DOI: 10.3390/ijms17091555] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/06/2016] [Accepted: 09/07/2016] [Indexed: 12/20/2022] Open
Abstract
The rise of technologies that simultaneously measure thousands of data points represents the heart of systems biology. These technologies have had a huge impact on the discovery of next-generation diagnostics, biomarkers, and drugs in the precision medicine era. Systems biology aims to achieve systemic exploration of complex interactions in biological systems. Driven by high-throughput omics technologies and the computational surge, it enables multi-scale and insightful overviews of cells, organisms, and populations. Precision medicine capitalizes on these conceptual and technological advancements and stands on two main pillars: data generation and data modeling. High-throughput omics technologies allow the retrieval of comprehensive and holistic biological information, whereas computational capabilities enable high-dimensional data modeling and, therefore, accessible and user-friendly visualization. Furthermore, bioinformatics has enabled comprehensive multi-omics and clinical data integration for insightful interpretation. Despite their promise, the translation of these technologies into clinically actionable tools has been slow. In this review, we present state-of-the-art multi-omics data analysis strategies in a clinical context. The challenges of omics-based biomarker translation are discussed. Perspectives regarding the use of multi-omics approaches for inborn errors of metabolism (IEM) are presented by introducing a new paradigm shift in addressing IEM investigations in the post-genomic era.
Collapse
Affiliation(s)
- Abdellah Tebani
- Department of Metabolic Biochemistry, Rouen University Hospital, 76031 Rouen, France.
- Normandie University, UNIROUEN, INSERM, CHU Rouen, Laboratoire NeoVasc ERI28, 76000 Rouen, France.
- Normandie University, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France.
| | - Carlos Afonso
- Normandie University, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France.
| | - Stéphane Marret
- Normandie University, UNIROUEN, INSERM, CHU Rouen, Laboratoire NeoVasc ERI28, 76000 Rouen, France.
- Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, 76031 Rouen, France.
| | - Soumeya Bekri
- Department of Metabolic Biochemistry, Rouen University Hospital, 76031 Rouen, France.
- Normandie University, UNIROUEN, INSERM, CHU Rouen, Laboratoire NeoVasc ERI28, 76000 Rouen, France.
| |
Collapse
|
41
|
Gemmell P, Burrage K, Rodríguez B, Quinn TA. Rabbit-specific computational modelling of ventricular cell electrophysiology: Using populations of models to explore variability in the response to ischemia. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 121:169-84. [PMID: 27320382 PMCID: PMC5405055 DOI: 10.1016/j.pbiomolbio.2016.06.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/13/2016] [Indexed: 11/04/2022]
Abstract
Computational modelling, combined with experimental investigations, is a powerful method for investigating complex cardiac electrophysiological behaviour. The use of rabbit-specific models, due to the similarities of cardiac electrophysiology in this species with human, is especially prevalent. In this paper, we first briefly review rabbit-specific computational modelling of ventricular cell electrophysiology, multi-cellular simulations including cellular heterogeneity, and acute ischemia. This mini-review is followed by an original computational investigation of variability in the electrophysiological response of two experimentally-calibrated populations of rabbit-specific ventricular myocyte action potential models to acute ischemia. We performed a systematic exploration of the response of the model populations to varying degrees of ischemia and individual ischemic parameters, to investigate their individual and combined effects on action potential duration and refractoriness. This revealed complex interactions between model population variability and ischemic factors, which combined to enhance variability during ischemia. This represents an important step towards an improved understanding of the role that physiological variability may play in electrophysiological alterations during acute ischemia.
Collapse
Affiliation(s)
- Philip Gemmell
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Kevin Burrage
- Department of Computer Science, University of Oxford, Oxford, UK; School of Mathematical Sciences and ARC Centre of Excellence, ACEMS, Queensland University of Technology, Brisbane, Australia
| | - Blanca Rodríguez
- Department of Computer Science, University of Oxford, Oxford, UK
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, 5850 College St, Lab 3F, Halifax, NS B3H 4R2, Canada; School of Biomedical Engineering, Dalhousie University, 5850 College St, Lab 3F, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|