1
|
Qiao L, Khalilimeybodi A, Linden-Santangeli NJ, Rangamani P. The Evolution of Systems Biology and Systems Medicine: From Mechanistic Models to Uncertainty Quantification. Annu Rev Biomed Eng 2025; 27:425-447. [PMID: 39971380 DOI: 10.1146/annurev-bioeng-102723-065309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Understanding interaction mechanisms within cells, tissues, and organisms is crucial for driving developments across biology and medicine. Mathematical modeling is an essential tool for simulating such biological systems. Building on experiments, mechanistic models are widely used to describe small-scale intracellular networks. The development of sequencing techniques and computational tools has recently enabled multiscale models. Combining such larger scale network modeling with mechanistic modeling provides us with an opportunity to reveal previously unknown disease mechanisms and pharmacological interventions. Here, we review systems biology models from mechanistic models to multiscale models that integrate multiple layers of cellular networks and discuss how they can be used to shed light on disease states and even wellness-related states. Additionally, we introduce several methods that increase the certainty and accuracy of model predictions. Thus, combining mechanistic models with emerging mathematical and computational techniques can provide us with increasingly powerful tools to understand disease states and inspire drug discoveries.
Collapse
Affiliation(s)
- Lingxia Qiao
- Department of Pharmacology, University of California San Diego, La Jolla, California, USA;
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA
| | - Ali Khalilimeybodi
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA
| | | | - Padmini Rangamani
- Department of Pharmacology, University of California San Diego, La Jolla, California, USA;
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
2
|
Świerczek A, Batko D, Wyska E. The Role of Pharmacometrics in Advancing the Therapies for Autoimmune Diseases. Pharmaceutics 2024; 16:1559. [PMID: 39771538 PMCID: PMC11676367 DOI: 10.3390/pharmaceutics16121559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/14/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Autoimmune diseases (AIDs) are a group of disorders in which the immune system attacks the body's own tissues, leading to chronic inflammation and organ damage. These diseases are difficult to treat due to variability in drug PK among individuals, patient responses to treatment, and the side effects of long-term immunosuppressive therapies. In recent years, pharmacometrics has emerged as a critical tool in drug discovery and development (DDD) and precision medicine. The aim of this review is to explore the diverse roles that pharmacometrics has played in addressing the challenges associated with DDD and personalized therapies in the treatment of AIDs. Methods: This review synthesizes research from the past two decades on pharmacometric methodologies, including Physiologically Based Pharmacokinetic (PBPK) modeling, Pharmacokinetic/Pharmacodynamic (PK/PD) modeling, disease progression (DisP) modeling, population modeling, model-based meta-analysis (MBMA), and Quantitative Systems Pharmacology (QSP). The incorporation of artificial intelligence (AI) and machine learning (ML) into pharmacometrics is also discussed. Results: Pharmacometrics has demonstrated significant potential in optimizing dosing regimens, improving drug safety, and predicting patient-specific responses in AIDs. PBPK and PK/PD models have been instrumental in personalizing treatments, while DisP and QSP models provide insights into disease evolution and pathophysiological mechanisms in AIDs. AI/ML implementation has further enhanced the precision of these models. Conclusions: Pharmacometrics plays a crucial role in bridging pre-clinical findings and clinical applications, driving more personalized and effective treatments for AIDs. Its integration into DDD and translational science, in combination with AI and ML algorithms, holds promise for advancing therapeutic strategies and improving autoimmune patients' outcomes.
Collapse
Affiliation(s)
- Artur Świerczek
- Department of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; (D.B.); (E.W.)
| | | | | |
Collapse
|
3
|
Tie Y, Liu J, Wu Y, Qiang Y, Cai’Li G, Xu P, Xue M, Xu L, Li X, Zhou X. A Dataset for Constructing the Network Pharmacology of Overactive Bladder and Its Application to Reveal the Potential Therapeutic Targets of Rhynchophylline. Pharmaceuticals (Basel) 2024; 17:1253. [PMID: 39458894 PMCID: PMC11510256 DOI: 10.3390/ph17101253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/06/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Objectives: Network pharmacology is essential for understanding the multi-target and multi-pathway therapeutic mechanisms of traditional Chinese medicine. This study aims to evaluate the influence of database quality on target identification and to explore the therapeutic potential of rhynchophylline (Rhy) in treating overactive bladder (OAB). Methods: An OAB dataset was constructed through extensive literature screening. Using this dataset, we applied network pharmacology to predict potential targets for Rhy, which is known for its therapeutic effects but lacks a well-defined target profile. Predicted targets were validated through in vitro experiments, including DARTS and CETSA. Results: Our analysis identified Rhy as a potential modulator of the M3 receptor and TRPM8 channel in the treatment of OAB. Validation experiments confirmed the interaction between Rhy and these targets. Additionally, the GeneCards database predicted other targets that are not directly linked to OAB, corroborated by the literature. Conclusions: We established a more accurate and comprehensive dataset of OAB targets, enhancing the reliability of target identification for drug treatments. This study underscores the importance of database quality in network pharmacology and contributes to the potential therapeutic strategies for OAB.
Collapse
Affiliation(s)
- Yan Tie
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (Y.T.); (J.L.); (Y.W.); (Y.Q.); (G.C.); (P.X.); (M.X.)
- School of Chinese Medicine, Capital Medical University, Beijing 100069, China;
| | - Jihan Liu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (Y.T.); (J.L.); (Y.W.); (Y.Q.); (G.C.); (P.X.); (M.X.)
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yushan Wu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (Y.T.); (J.L.); (Y.W.); (Y.Q.); (G.C.); (P.X.); (M.X.)
| | - Yining Qiang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (Y.T.); (J.L.); (Y.W.); (Y.Q.); (G.C.); (P.X.); (M.X.)
| | - Ge’Er Cai’Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (Y.T.); (J.L.); (Y.W.); (Y.Q.); (G.C.); (P.X.); (M.X.)
| | - Pingxiang Xu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (Y.T.); (J.L.); (Y.W.); (Y.Q.); (G.C.); (P.X.); (M.X.)
| | - Ming Xue
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (Y.T.); (J.L.); (Y.W.); (Y.Q.); (G.C.); (P.X.); (M.X.)
| | - Liping Xu
- School of Chinese Medicine, Capital Medical University, Beijing 100069, China;
| | - Xiaorong Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (Y.T.); (J.L.); (Y.W.); (Y.Q.); (G.C.); (P.X.); (M.X.)
| | - Xuelin Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (Y.T.); (J.L.); (Y.W.); (Y.Q.); (G.C.); (P.X.); (M.X.)
| |
Collapse
|
4
|
Hsiao YC, Dutta A. Network Modeling and Control of Dynamic Disease Pathways, Review and Perspectives. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:1211-1230. [PMID: 38498762 DOI: 10.1109/tcbb.2024.3378155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Dynamic disease pathways are a combination of complex dynamical processes among bio-molecules in a cell that leads to diseases. Network modeling of disease pathways considers disease-related bio-molecules (e.g. DNA, RNA, transcription factors, enzymes, proteins, and metabolites) and their interaction (e.g. DNA methylation, histone modification, alternative splicing, and protein modification) to study disease progression and predict therapeutic responses. These bio-molecules and their interactions are the basic elements in the study of the misregulation in the disease-related gene expression that lead to abnormal cellular responses. Gene regulatory networks, cell signaling networks, and metabolic networks are the three major types of intracellular networks for the study of the cellular responses elicited from extracellular signals. The disease-related cellular responses can be prevented or regulated by designing control strategies to manipulate these extracellular or other intracellular signals. The paper reviews the regulatory mechanisms, the dynamic models, and the control strategies for each intracellular network. The applications, limitations and the prospective for modeling and control are also discussed.
Collapse
|
5
|
Cosme M, Thomas C, Gaucherel C. On the History of Ecosystem Dynamical Modeling: The Rise and Promises of Qualitative Models. ENTROPY (BASEL, SWITZERLAND) 2023; 25:1526. [PMID: 37998218 PMCID: PMC10670156 DOI: 10.3390/e25111526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/25/2023]
Abstract
Ecosystem modeling is a complex and multidisciplinary modeling problem which emerged in the 1950s. It takes advantage of the computational turn in sciences to better understand anthropogenic impacts and improve ecosystem management. For that purpose, ecosystem simulation models based on difference or differential equations were built. These models were relevant for studying dynamical phenomena and still are. However, they face important limitations in data-poor situations. As a response, several formal and non-formal qualitative dynamical modeling approaches were independently developed to overcome some limitations of the existing methods. Qualitative approaches allow studying qualitative dynamics as relevant abstractions of those provided by quantitative models (e.g., response to press perturbations). Each modeling framework can be viewed as a different assemblage of properties (e.g., determinism, stochasticity or synchronous update of variable values) designed to satisfy some scientific objectives. Based on four stated objectives commonly found in complex environmental sciences ((1) grasping qualitative dynamics, (2) making as few assumptions as possible about parameter values, (3) being explanatory and (4) being predictive), our objectives were guided by the wish to model complex and multidisciplinary issues commonly found in ecosystem modeling. We then discussed the relevance of existing modeling approaches and proposed the ecological discrete-event networks (EDEN) modeling framework for this purpose. The EDEN models propose a qualitative, discrete-event, partially synchronous and possibilistic view of ecosystem dynamics. We discussed each of these properties through ecological examples and existing analysis techniques for such models and showed how relevant they are for environmental science studies.
Collapse
Affiliation(s)
- Maximilien Cosme
- UMR AMAP, INRAE, University of Montpellier (Faculté des Sciences), IRD, CIRAD, CNRS, 34398 Montpellier, France
- UMR DECOD, Institut Agro Rennes-Angers (Campus Rennes), 65 rue de Saint-Brieuc, 35042 Rennes, France
| | - Colin Thomas
- IBISC, University of Evry, 91025 Evry, France (C.G.)
| | | |
Collapse
|
6
|
Ballesta A, Gallo JM. Quantitative Systems Pharmacology: A Foundation To Establish Precision Medicine-Editorial. J Pharmacol Exp Ther 2023; 387:27-30. [PMID: 37714689 DOI: 10.1124/jpet.123.001842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 09/17/2023] Open
Affiliation(s)
- Annabelle Ballesta
- INSERM U900, Institut Curie, Mines ParisTech CBIO, Université PSL, Paris, France (A.B.) and Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York (J.M.G.)
| | - James M Gallo
- INSERM U900, Institut Curie, Mines ParisTech CBIO, Université PSL, Paris, France (A.B.) and Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York (J.M.G.)
| |
Collapse
|
7
|
Hemedan AA, Schneider R, Ostaszewski M. Applications of Boolean modeling to study the dynamics of a complex disease and therapeutics responses. FRONTIERS IN BIOINFORMATICS 2023; 3:1189723. [PMID: 37325771 PMCID: PMC10267406 DOI: 10.3389/fbinf.2023.1189723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/18/2023] [Indexed: 06/17/2023] Open
Abstract
Computational modeling has emerged as a critical tool in investigating the complex molecular processes involved in biological systems and diseases. In this study, we apply Boolean modeling to uncover the molecular mechanisms underlying Parkinson's disease (PD), one of the most prevalent neurodegenerative disorders. Our approach is based on the PD-map, a comprehensive molecular interaction diagram that captures the key mechanisms involved in the initiation and progression of PD. Using Boolean modeling, we aim to gain a deeper understanding of the disease dynamics, identify potential drug targets, and simulate the response to treatments. Our analysis demonstrates the effectiveness of this approach in uncovering the intricacies of PD. Our results confirm existing knowledge about the disease and provide valuable insights into the underlying mechanisms, ultimately suggesting potential targets for therapeutic intervention. Moreover, our approach allows us to parametrize the models based on omics data for further disease stratification. Our study highlights the value of computational modeling in advancing our understanding of complex biological systems and diseases, emphasizing the importance of continued research in this field. Furthermore, our findings have potential implications for the development of novel therapies for PD, which is a pressing public health concern. Overall, this study represents a significant step forward in the application of computational modeling to the investigation of neurodegenerative diseases, and underscores the power of interdisciplinary approaches in tackling challenging biomedical problems.
Collapse
|
8
|
Langston JC, Yang Q, Kiani MF, Kilpatrick LE. LEUKOCYTE PHENOTYPING IN SEPSIS USING OMICS, FUNCTIONAL ANALYSIS, AND IN SILICO MODELING. Shock 2023; 59:224-231. [PMID: 36377365 PMCID: PMC9957940 DOI: 10.1097/shk.0000000000002047] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
ABSTRACT Sepsis is a major health issue and a leading cause of death in hospitals globally. The treatment of sepsis is largely supportive, and there are no therapeutics available that target the underlying pathophysiology of the disease. The development of therapeutics for the treatment of sepsis is hindered by the heterogeneous nature of the disease. The presence of multiple, distinct immune phenotypes ranging from hyperimmune to immunosuppressed can significantly impact the host response to infection. Recently, omics, biomarkers, cell surface protein expression, and immune cell profiles have been used to classify immune status of sepsis patients. However, there has been limited studies of immune cell function during sepsis and even fewer correlating omics and biomarker alterations to functional consequences. In this review, we will discuss how the heterogeneity of sepsis and associated immune cell phenotypes result from changes in the omic makeup of cells and its correlation with leukocyte dysfunction. We will also discuss how emerging techniques such as in silico modeling and machine learning can help in phenotyping sepsis patients leading to precision medicine.
Collapse
Affiliation(s)
- Jordan C. Langston
- Department of Bioengineering, Temple University, Philadelphia, PA, 19122
| | - Qingliang Yang
- Department of Mechanical Engineering, Temple University, Philadelphia, PA, 19122
| | - Mohammad F. Kiani
- Department of Bioengineering, Temple University, Philadelphia, PA, 19122
- Department of Mechanical Engineering, Temple University, Philadelphia, PA, 19122
| | - Laurie E. Kilpatrick
- Center for Inflammation and Lung Research, Department of Microbiology, Immunology and Inflammation, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
| |
Collapse
|
9
|
A system pharmacology Boolean network model for the TLR4-mediated inflammatory response in early sepsis. J Pharmacokinet Pharmacodyn 2022; 49:645-655. [PMID: 36261775 DOI: 10.1007/s10928-022-09828-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/06/2022] [Indexed: 10/24/2022]
Abstract
Sepsis is a life-threatening condition driven by the dysregulation of the host immune response to an infection. The complex and interacting mechanisms underlying sepsis remain not fully understood. By integrating prior knowledge from literature using mathematical modelling techniques, we aimed to obtain a deeper mechanistic insight into sepsis pathogenesis and to evaluate promising novel therapeutic targets, with a focus on Toll-like receptor 4 (TLR4)-mediated pathways. A Boolean network of regulatory relationships was developed for key immune components associated with sepsis pathogenesis after TLR4 activation. Perturbation analyses were conducted to identify therapeutic targets associated with organ dysfunction or antibacterial activity. The developed model consisted of 42 nodes and 183 interactions. Perturbation analyses suggest that over-expression of tumour necrosis factor alpha (TNF-α) or inhibition of soluble receptor sTNF-R, tissue factor, and inflammatory cytokines (IFN-γ, IL-12) may lead to a reduced activation of organ dysfunction related endpoints. Over-expression of complement factor C3b and C5b led to an increase in the bacterial clearance related endpoint. We identified that combinatory blockade of IFN-γ and IL-10 may reduce the risk of organ dysfunction. Finally, we found that combining antibiotic treatment with IL-1β targeted therapy may have the potential to decrease thrombosis. In summary, we demonstrate how existing biological knowledge can be effectively integrated using Boolean network analysis for hypothesis generation of potential treatment strategies and characterization of biomarker responses associated with the early inflammatory response in sepsis.
Collapse
|
10
|
Hemedan AA, Niarakis A, Schneider R, Ostaszewski M. Boolean modelling as a logic-based dynamic approach in systems medicine. Comput Struct Biotechnol J 2022; 20:3161-3172. [PMID: 35782730 PMCID: PMC9234349 DOI: 10.1016/j.csbj.2022.06.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Molecular mechanisms of health and disease are often represented as systems biology diagrams, and the coverage of such representation constantly increases. These static diagrams can be transformed into dynamic models, allowing for in silico simulations and predictions. Boolean modelling is an approach based on an abstract representation of the system. It emphasises the qualitative modelling of biological systems in which each biomolecule can take two possible values: zero for absent or inactive, one for present or active. Because of this approximation, Boolean modelling is applicable to large diagrams, allowing to capture their dynamic properties. We review Boolean models of disease mechanisms and compare a range of methods and tools used for analysis processes. We explain the methodology of Boolean analysis focusing on its application in disease modelling. Finally, we discuss its practical application in analysing signal transduction and gene regulatory pathways in health and disease.
Collapse
Affiliation(s)
- Ahmed Abdelmonem Hemedan
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Anna Niarakis
- Université Paris-Saclay, Laboratoire Européen de Recherche pour la Polyarthrite rhumatoïde – Genhotel, Univ Evry, Evry, France
- Lifeware Group, Inria, Saclay-île de France, 91120 Palaiseau, France
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
11
|
In silico Methods for Identification of Potential Therapeutic Targets. Interdiscip Sci 2022; 14:285-310. [PMID: 34826045 PMCID: PMC8616973 DOI: 10.1007/s12539-021-00491-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 10/19/2021] [Accepted: 11/01/2021] [Indexed: 11/01/2022]
Abstract
AbstractAt the initial stage of drug discovery, identifying novel targets with maximal efficacy and minimal side effects can improve the success rate and portfolio value of drug discovery projects while simultaneously reducing cycle time and cost. However, harnessing the full potential of big data to narrow the range of plausible targets through existing computational methods remains a key issue in this field. This paper reviews two categories of in silico methods—comparative genomics and network-based methods—for finding potential therapeutic targets among cellular functions based on understanding their related biological processes. In addition to describing the principles, databases, software, and applications, we discuss some recent studies and prospects of the methods. While comparative genomics is mostly applied to infectious diseases, network-based methods can be applied to infectious and non-infectious diseases. Nonetheless, the methods often complement each other in their advantages and disadvantages. The information reported here guides toward improving the application of big data-driven computational methods for therapeutic target discovery.
Graphical abstract
Collapse
|
12
|
Sonawane AR, Aikawa E, Aikawa M. Connections for Matters of the Heart: Network Medicine in Cardiovascular Diseases. Front Cardiovasc Med 2022; 9:873582. [PMID: 35665246 PMCID: PMC9160390 DOI: 10.3389/fcvm.2022.873582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/19/2022] [Indexed: 01/18/2023] Open
Abstract
Cardiovascular diseases (CVD) are diverse disorders affecting the heart and vasculature in millions of people worldwide. Like other fields, CVD research has benefitted from the deluge of multiomics biomedical data. Current CVD research focuses on disease etiologies and mechanisms, identifying disease biomarkers, developing appropriate therapies and drugs, and stratifying patients into correct disease endotypes. Systems biology offers an alternative to traditional reductionist approaches and provides impetus for a comprehensive outlook toward diseases. As a focus area, network medicine specifically aids the translational aspect of in silico research. This review discusses the approach of network medicine and its application to CVD research.
Collapse
Affiliation(s)
- Abhijeet Rajendra Sonawane
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
13
|
Van Giang T, Hiraishi K. On Attractor Detection and Optimal Control of Deterministic Generalized Asynchronous Random Boolean Networks. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:1794-1806. [PMID: 33301407 DOI: 10.1109/tcbb.2020.3043785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Deterministic asynchronous Boolean networks play a crucial role in modeling and analysis of gene regulatory networks. In this paper, we focus on a typical type of deterministic asynchronous Boolean networks called deterministic generalized asynchronous random Boolean networks (DGARBNs). We first formulate the extended state transition graph, which captures the whole dynamics of a DGARBN and paves potential ways to analyze this DGARBN. We then propose two SMT-based methods for attractor detection and optimal control of DGARBNs. These methods are implemented in a JAVA tool called DABoolNet. Two experiments are designed to highlight the scalability of the proposed methods. We also formally state and prove several relations between DGARBNs and other models including deterministic asynchronous models, block-sequential Boolean networks, generalized asynchronous random Boolean networks, and mixed-context random Boolean networks. Several case studies are presented to show the applications of our methods.
Collapse
|
14
|
Jafari Nivlouei S, Soltani M, Shirani E, Salimpour MR, Travasso R, Carvalho J. A multiscale cell-based model of tumor growth for chemotherapy assessment and tumor-targeted therapy through a 3D computational approach. Cell Prolif 2022; 55:e13187. [PMID: 35132721 PMCID: PMC8891571 DOI: 10.1111/cpr.13187] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/09/2021] [Accepted: 01/03/2022] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES Computational modeling of biological systems is a powerful tool to clarify diverse processes contributing to cancer. The aim is to clarify the complex biochemical and mechanical interactions between cells, the relevance of intracellular signaling pathways in tumor progression and related events to the cancer treatments, which are largely ignored in previous studies. MATERIALS AND METHODS A three-dimensional multiscale cell-based model is developed, covering multiple time and spatial scales, including intracellular, cellular, and extracellular processes. The model generates a realistic representation of the processes involved from an implementation of the signaling transduction network. RESULTS Considering a benign tumor development, results are in good agreement with the experimental ones, which identify three different phases in tumor growth. Simulating tumor vascular growth, results predict a highly vascularized tumor morphology in a lobulated form, a consequence of cells' motile behavior. A novel systematic study of chemotherapy intervention, in combination with targeted therapy, is presented to address the capability of the model to evaluate typical clinical protocols. The model also performs a dose comparison study in order to optimize treatment efficacy and surveys the effect of chemotherapy initiation delays and different regimens. CONCLUSIONS Results not only provide detailed insights into tumor progression, but also support suggestions for clinical implementation. This is a major step toward the goal of predicting the effects of not only traditional chemotherapy but also tumor-targeted therapies.
Collapse
Affiliation(s)
- Sahar Jafari Nivlouei
- Department of Mechanical Engineering, Isfahan University of Technology, Isafahan, Iran.,Department of Physics, CFisUC, University of Coimbra, Coimbra, Portugal
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.,Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada.,Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, Canada.,Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran.,Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Ebrahim Shirani
- Department of Mechanical Engineering, Isfahan University of Technology, Isafahan, Iran.,Department of Mechanical Engineering, Foolad Institute of Technology, Fooladshahr, Iran
| | | | - Rui Travasso
- Department of Physics, CFisUC, University of Coimbra, Coimbra, Portugal
| | - João Carvalho
- Department of Physics, CFisUC, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
15
|
Putnins M, Campagne O, Mager DE, Androulakis IP. From data to QSP models: a pipeline for using Boolean networks for hypothesis inference and dynamic model building. J Pharmacokinet Pharmacodyn 2022; 49:101-115. [PMID: 34988912 PMCID: PMC9876619 DOI: 10.1007/s10928-021-09797-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/27/2021] [Indexed: 01/27/2023]
Abstract
Quantitative Systems Pharmacology (QSP) models capture the physiological underpinnings driving the response to a drug and express those in a semi-mechanistic way, often involving ordinary differential equations (ODEs). The process of developing a QSP model generally starts with the definition of a set of reasonable hypotheses that would support a mechanistic interpretation of the expected response which are used to form a network of interacting elements. This is a hypothesis-driven and knowledge-driven approach, relying on prior information about the structure of the network. However, with recent advances in our ability to generate large datasets rapidly, often in a hypothesis-neutral manner, the opportunity emerges to explore data-driven approaches to establish the network topologies and models in a robust, repeatable manner. In this paper, we explore the possibility of developing complex network representations of physiological responses to pharmaceuticals using a logic-based analysis of available data and then convert the logic relations to dynamic ODE-based models. We discuss an integrated pipeline for converting data to QSP models. This pipeline includes using k-means clustering to binarize continuous data, inferring likely network relationships using a Best-Fit Extension method to create a Boolean network, and finally converting the Boolean network to a continuous ODE model. We utilized an existing QSP model for the dual-affinity re-targeting antibody flotetuzumab to demonstrate the robustness of the process. Key output variables from the QSP model were used to generate a continuous data set for use in the pipeline. This dataset was used to reconstruct a possible model. This reconstruction had no false-positive relationships, and the output of each of the species was similar to that of the original QSP model. This demonstrates the ability to accurately infer relationships in a hypothesis-neutral manner without prior knowledge of a system using this pipeline.
Collapse
Affiliation(s)
- M. Putnins
- Biomedical Engineering Department, Rutgers University, Piscataway, USA
| | - O. Campagne
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, USA
| | - D. E. Mager
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, USA
| | - I. P. Androulakis
- Biomedical Engineering Department, Rutgers University, Piscataway, USA,Chemical & Biochemical Engineering Department, Rutgers University, Piscataway, USA
| |
Collapse
|
16
|
Langston JC, Rossi MT, Yang Q, Ohley W, Perez E, Kilpatrick LE, Prabhakarpandian B, Kiani MF. Omics of endothelial cell dysfunction in sepsis. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2022; 4:R15-R34. [PMID: 35515704 PMCID: PMC9066943 DOI: 10.1530/vb-22-0003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/07/2022] [Indexed: 12/19/2022]
Abstract
During sepsis, defined as life-threatening organ dysfunction due to dysregulated host response to infection, systemic inflammation activates endothelial cells and initiates a multifaceted cascade of pro-inflammatory signaling events, resulting in increased permeability and excessive recruitment of leukocytes. Vascular endothelial cells share many common properties but have organ-specific phenotypes with unique structure and function. Thus, therapies directed against endothelial cell phenotypes are needed to address organ-specific endothelial cell dysfunction. Omics allow for the study of expressed genes, proteins and/or metabolites in biological systems and provide insight on temporal and spatial evolution of signals during normal and diseased conditions. Proteomics quantifies protein expression, identifies protein-protein interactions and can reveal mechanistic changes in endothelial cells that would not be possible to study via reductionist methods alone. In this review, we provide an overview of how sepsis pathophysiology impacts omics with a focus on proteomic analysis of mouse endothelial cells during sepsis/inflammation and its relationship with the more clinically relevant omics of human endothelial cells. We discuss how omics has been used to define septic endotype signatures in different populations with a focus on proteomic analysis in organ-specific microvascular endothelial cells during sepsis or septic-like inflammation. We believe that studies defining septic endotypes based on proteomic expression in endothelial cell phenotypes are urgently needed to complement omic profiling of whole blood and better define sepsis subphenotypes. Lastly, we provide a discussion of how in silico modeling can be used to leverage the large volume of omics data to map response pathways in sepsis.
Collapse
Affiliation(s)
- Jordan C Langston
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania, USA
| | | | - Qingliang Yang
- Department of Mechanical Engineering, Temple University, Philadelphia, Pennsylvania, USA
| | - William Ohley
- Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Edwin Perez
- Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Laurie E Kilpatrick
- Center for Inflammation and Lung Research, Department of Microbiology, Immunology and Inflammation, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Balabhaskar Prabhakarpandian
- Center for Inflammation and Lung Research, Department of Microbiology, Immunology and Inflammation, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Mohammad F Kiani
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania, USA
- Department of Mechanical Engineering, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
17
|
Bloomingdale P, Meregalli C, Pollard K, Canta A, Chiorazzi A, Fumagalli G, Monza L, Pozzi E, Alberti P, Ballarini E, Oggioni N, Carlson L, Liu W, Ghandili M, Ignatowski TA, Lee KP, Moore MJ, Cavaletti G, Mager DE. Systems Pharmacology Modeling Identifies a Novel Treatment Strategy for Bortezomib-Induced Neuropathic Pain. Front Pharmacol 2022; 12:817236. [PMID: 35126148 PMCID: PMC8809372 DOI: 10.3389/fphar.2021.817236] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/16/2021] [Indexed: 11/17/2022] Open
Abstract
Chemotherapy-induced peripheral neurotoxicity is a common dose-limiting side effect of several cancer chemotherapeutic agents, and no effective therapies exist. Here we constructed a systems pharmacology model of intracellular signaling in peripheral neurons to identify novel drug targets for preventing peripheral neuropathy associated with proteasome inhibitors. Model predictions suggested the combinatorial inhibition of TNFα, NMDA receptors, and reactive oxygen species should prevent proteasome inhibitor-induced neuronal apoptosis. Dexanabinol, an inhibitor of all three targets, partially restored bortezomib-induced reduction of proximal action potential amplitude and distal nerve conduction velocity in vitro and prevented bortezomib-induced mechanical allodynia and thermal hyperalgesia in rats, including a partial recovery of intraepidermal nerve fiber density. Dexanabinol failed to restore bortezomib-induced decreases in electrophysiological endpoints in rats, and it did not compromise bortezomib anti-cancer effects in U266 multiple myeloma cells and a murine xenograft model. Owing to its favorable safety profile in humans and preclinical efficacy, dexanabinol might represent a treatment option for bortezomib-induced neuropathic pain.
Collapse
Affiliation(s)
- Peter Bloomingdale
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Cristina Meregalli
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Kevin Pollard
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, United States
| | - Annalisa Canta
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Alessia Chiorazzi
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Giulia Fumagalli
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Laura Monza
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Eleonora Pozzi
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Paola Alberti
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Elisa Ballarini
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Norberto Oggioni
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Louise Carlson
- Department of Immunology, Roswell Park Comprehensive Cancer Center, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Wensheng Liu
- Department of Immunology, Roswell Park Comprehensive Cancer Center, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Mehrnoosh Ghandili
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Tracey A. Ignatowski
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Kelvin P. Lee
- Department of Immunology, Roswell Park Comprehensive Cancer Center, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Michael J. Moore
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, United States
- AxoSim, Inc., New Orleans, LA, United States
| | - Guido Cavaletti
- Experimental Neurology Unit and Milan Center for Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- *Correspondence: Guido Cavaletti, ; Donald E. Mager,
| | - Donald E. Mager
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
- Enhanced Pharmacodynamics, LLC, Buffalo, NY, United States
- *Correspondence: Guido Cavaletti, ; Donald E. Mager,
| |
Collapse
|
18
|
Bonate PL. Editor's note on the themed issue: integration of machine learning and quantitative systems pharmacology. J Pharmacokinet Pharmacodyn 2022; 49:1-3. [PMID: 35041146 DOI: 10.1007/s10928-022-09803-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
19
|
Maudsley S, Leysen H, van Gastel J, Martin B. Systems Pharmacology: Enabling Multidimensional Therapeutics. COMPREHENSIVE PHARMACOLOGY 2022:725-769. [DOI: 10.1016/b978-0-12-820472-6.00017-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
20
|
Kaushik AC, Sahi S, Wei DQ. Computational Methods for Structure-Based Drug Design Through System Biology. Methods Mol Biol 2022; 2385:161-174. [PMID: 34888721 DOI: 10.1007/978-1-0716-1767-0_9] [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] [Indexed: 06/13/2023]
Abstract
The advances in computational chemistry and biology, computer science, structural biology, and molecular biology go in parallel with the rapid progress in target-based systems. This technique has become a powerful tool in medicinal chemistry for the identification of hit molecules. The recent developments in target-based systems have played a major role in the creation of libraries of compounds, and it has also been widely applied for the design of molecular docking methods. The main advantage of this method is that it hits the fragment that has the strongest binding, has relatively small size, and leads to better compounds in terms of pharmacokinetic properties when compared with virtual screening (VS) and high-throughput screening (HTS) hits. De novo design is an essential aspect of target-based systems and requires the synthesis of chemical to allow the design of promising compound.
Collapse
Affiliation(s)
| | - Shakti Sahi
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Research Laboratory of Metabolic & Developmental Sciences and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
- Peng Cheng Laboratory, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
21
|
Aghamiri SS, Amin R, Helikar T. Recent applications of quantitative systems pharmacology and machine learning models across diseases. J Pharmacokinet Pharmacodyn 2021; 49:19-37. [PMID: 34671863 PMCID: PMC8528185 DOI: 10.1007/s10928-021-09790-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/07/2021] [Indexed: 12/29/2022]
Abstract
Quantitative systems pharmacology (QSP) is a quantitative and mechanistic platform describing the phenotypic interaction between drugs, biological networks, and disease conditions to predict optimal therapeutic response. In this meta-analysis study, we review the utility of the QSP platform in drug development and therapeutic strategies based on recent publications (2019-2021). We gathered recent original QSP models and described the diversity of their applications based on therapeutic areas, methodologies, software platforms, and functionalities. The collection and investigation of these publications can assist in providing a repository of recent QSP studies to facilitate the discovery and further reusability of QSP models. Our review shows that the largest number of QSP efforts in recent years is in Immuno-Oncology. We also addressed the benefits of integrative approaches in this field by presenting the applications of Machine Learning methods for drug discovery and QSP models. Based on this meta-analysis, we discuss the advantages and limitations of QSP models and propose fields where the QSP approach constitutes a valuable interface for more investigations to tackle complex diseases and improve drug development.
Collapse
Affiliation(s)
- Sara Sadat Aghamiri
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Rada Amin
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA.
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
22
|
Niu J, Nguyen VA, Ghasemi M, Chen T, Mager DE. Cluster Gauss-Newton and CellNOpt Parameter Estimation in a Small Protein Signaling Network of Vorinostat and Bortezomib Pharmacodynamics. AAPS JOURNAL 2021; 23:110. [PMID: 34622346 DOI: 10.1208/s12248-021-00640-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 08/19/2021] [Indexed: 11/30/2022]
Abstract
Ordinary differential equation (ODE)-based models of signal transduction pathways often contain parameters that are unidentifiable or unmeasurable by experimental data, and calibrating such models to data remains challenging. Here, two efficient parameter estimation methods, cluster Gauss-Newton (CGN) and CellNOpt (CNO), were applied to fit a signaling network model of U266 multiple myeloma cells to the activity dynamics of key proteins in response to vorinostat and/or bortezomib. A logic-based network model was constructed and transformed to 17 ODEs with 79 parameters estimated within broad ranges of biologically plausible values. The top 10% best-fit parameters by both methods had high uncertainties with CV > 50% for the majority of parameters. The root mean square and prediction errors were comparable without statistically significant differences between the two methods. Despite uncertain parameter estimation, protein dynamics after the sequential combination of bortezomib and vorinostat was predicted with reasonable accuracy and precision. Global sensitivity analyses of partial rank correlation coefficients and Sobol sensitivity demonstrated that apoptosis induction was most sensitive to parameters governing the activity of the proteasome-JNK-caspase-8 axis. Simulations revealed that the greatest magnitude of pharmacodynamic drug interactions between bortezomib and vorinostat occurred at caspase-9, AKT, and Bcl-2. Two sequential combinations were explored in silico, and the outcome matched qualitatively with an empirical evaluation of the pharmacodynamic interaction based on cell viability. Overall, the CGN and CNO algorithms performed similarly for this ODE-based network model calibration, and the calibrated model provided meaningful insights into cellular signaling mechanisms in response to pharmacological perturbations.
Collapse
Affiliation(s)
- Jin Niu
- Department of Pharmaceutical Sciences, University At Buffalo, State University of New York, 431 Pharmacy Building, Buffalo, NY, 14214, USA
| | - Van Anh Nguyen
- Department of Pharmaceutical Sciences, University At Buffalo, State University of New York, 431 Pharmacy Building, Buffalo, NY, 14214, USA
| | - Mohammad Ghasemi
- Department of Pharmaceutical Sciences, University At Buffalo, State University of New York, 431 Pharmacy Building, Buffalo, NY, 14214, USA
| | - Ting Chen
- Department of Pharmaceutical Sciences, University At Buffalo, State University of New York, 431 Pharmacy Building, Buffalo, NY, 14214, USA
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University At Buffalo, State University of New York, 431 Pharmacy Building, Buffalo, NY, 14214, USA.
| |
Collapse
|
23
|
Qi Y, Zhou N, Jiang Q, Wang Z, Zhang Y, Li B, Xu W, Liu J, Wang Z, Zhu L. Dose-Dependent Variation of Synchronous Metabolites and Modules in a Yin/Yang Transformation Model of Appointed Ischemia Metabolic Networks. Front Neurosci 2021; 15:645185. [PMID: 34531713 PMCID: PMC8439200 DOI: 10.3389/fnins.2021.645185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/22/2021] [Indexed: 01/26/2023] Open
Abstract
Aim Chinese medicine Danhong injection (DHI) is an effective pharmaceutical preparation for treating cerebral infarction. Our previous study shows that DHI can remarkably reduce the ischemic stroke-induced infarct volume in a dose-dependent manner, but the pharmacological mechanism of the DHI dose-dependent relationship is not clear. Therefore, the dose-dependent efficacy of DHI on cerebral ischemia and the underlying mechanisms were further investigated in this study. Methods A middle cerebral artery occlusion (MCAO) model was established and the rats were randomly divided into six groups: sham, vehicle, DHI dose-1, DHI dose-2, DHI dose-3, and DHI dose-4. Forty-one metabolites in serum were selected as candidate biomarkers of efficacy phenotypes by the Agilent 1290 rapid-resolution liquid chromatography system coupled with the Agilent 6550 Q-TOF MS system. Then, the metabolic networks in each group were constructed using the Weighted Correlation Network analysis (WGCNA). Moreover, the Yang and Yin transformation of six patterns (which are defined by up- and downregulation of metabolites) and synchronous modules divided from a synchronous network were used to dynamically analyze the mechanism of the drug’s effectiveness. Results The neuroprotective effect of DHI has shown a dose-dependent manner, and the high-dose group (DH3 and DH4) effect is better. The entropy of the metabolic network and the Yin/Yang index both showed a consistent dose–response relationship. Seven dose-sensitive metabolites maintained constant inverse upregulation or downregulation in the four dose groups. Three synchronous modules for the DH1–DH4 full-course network were identified. Glycine, N-acetyl-L-glutamate, and tetrahydrofolate as a new emerging module appeared in DH2/DH3 and enriched in glutamine and glutamate metabolism-related pathways. Conclusion This study takes the DHI metabolic network as an example to provide a new method for the discovery of multiple targets related to pharmacological effects. Our results show that the three conservative allosteric module nodes, taurine, L-tyrosine, and L-leucine, may be one of the basic mechanisms of DHI in the treatment of cerebral infarction, and the other three new emerging module nodes glyoxylate, L-glutamate, and L-valine may participate in the glutamine and glutamate metabolism pathway to improve the efficacy of DHI.
Collapse
Affiliation(s)
- Yifei Qi
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,Xiyuan Hospital, Institute of Geriatrics, China Academy of Chinese Medical Sciences, Beijing, China
| | - Niwen Zhou
- Center for Statistics and Data Science, Beijing Normal University at Zhuhai, Zhuhai, China
| | - Qing Jiang
- Center for Statistics and Data Science, Beijing Normal University at Zhuhai, Zhuhai, China
| | - Zhi Wang
- Global Business Services, International Business Machines Corporation, Shanghai, China
| | - Yingying Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Bing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenjuan Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lixing Zhu
- Center for Statistics and Data Science, Beijing Normal University at Zhuhai, Zhuhai, China.,Department of Mathematics, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
24
|
Nanavati C, Mager DE. Network-Based Systems Analysis Explains Sequence-Dependent Synergism of Bortezomib and Vorinostat in Multiple Myeloma. AAPS JOURNAL 2021; 23:101. [PMID: 34403034 DOI: 10.1208/s12248-021-00622-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/05/2021] [Indexed: 12/22/2022]
Abstract
Bortezomib and vorinostat exhibit synergistic effects in multiple myeloma (MM) cells when given in sequence, and the purpose of this study was to evaluate the molecular determinants of the interaction using a systems pharmacology approach. A Boolean network model consisting of 79 proteins and 225 connections was developed using literature information characterizing mechanisms of drug action and intracellular signal transduction. Network visualization and structural analysis were conducted, and model simulations were compared with experimental data. Critical biomarkers, such as pNFκB, p53, cellular stress, and p21, were identified using measures of network centrality and model reduction. U266 cells were then exposed to bortezomib (3 nM) and vorinostat (2 μM) as single agents or in simultaneous and sequential (bortezomib for first 24 h, followed by addition of vorinostat for another 24 h) combinations. Temporal changes for nine of the critical proteins in the reduced Boolean model were measured over 48 h, and cellular proliferation was measured over 96 h. A mechanism-based systems model was developed that captured the biological basis of a bortezomib and vorinostat sequence-dependent pharmacodynamic interaction. The model was further extended in vivo by linking in vitro parameter values and dynamics of p21, caspase-3, and pAKT biomarkers to tumor growth in xenograft mice reported in the literature. Network-based methodologies and pharmacodynamic principles were integrated successfully to evaluate bortezomib and vorinostat interactions in a mechanistic and quantitative manner. The model can be potentially applied to evaluate their combination regimens and explore in vivo dosing regimens.
Collapse
Affiliation(s)
- Charvi Nanavati
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, 431 Pharmacy Building Buffalo, New York, 14214, USA
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, 431 Pharmacy Building Buffalo, New York, 14214, USA.
| |
Collapse
|
25
|
Sun Z, Shen K, Xie Y, Hu B, He P, Lu Y, Xue H. Shiquan Yuzhen Decoction inhibits angiogenesis and tumor apoptosis caused by non-small cell lung cancer and promotes immune response. Am J Transl Res 2021; 13:7492-7507. [PMID: 34377231 PMCID: PMC8340251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/23/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND TCM treatment for lung carcinoma has been reported by many researches. Shiquan Yuzhen Decoction can be used in the clinical treatment of lung carcinoma, but its specific mechanism is still under exploration at present. METHODS The active ingredients and mechanism of Shiquan Yuzhen Decoction on non-small cell lung carcinoma were discussed by network pharmacology. The main active ingredients, targets and disease genes of non-small cell lung carcinoma of Shiquan Yuzhen Decoction were screened through relevant databases. Lewis lung carcinoma bearing mice model was established by inoculating Lewis lung carcinoma cells to C57BL/6 mice under the right armpit. Different doses of Shiquan Yuzhen Decoction were used to observe the apoptosis and angiogenesis changes of tumor tissues in mice. RESULTS A total of 26 key active compounds meeting the evaluation of generic properties and 182 main targets were screened out. The multi-level network model shows that Shiquan Yuzhen Decoction can regulate the target gene network of non-small cell lung carcinoma. And it can inhibit tumor growth in tumor-bearing mice, induce apoptosis of tumor cells, and evidently increase the activities of Caspase-3, 8 and 9. The dose of 17.4 g/kg can evidently inhibit the formation of microvessels in transplanted tumor tissues, improve the sensitivity of mice's diet and activities, increase the spleen index of tumor-bearing mice, and inhibit inflammatory factors. CONCLUSION Shiquan Yuzhen Decoction can evidently improve the quality of life of Lewis lung carcinoma-bearing mice and inhibit tumor growth in mice, which is a potential clinical treatment plan.
Collapse
Affiliation(s)
- Zhengda Sun
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Keping Shen
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Yage Xie
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Bing Hu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Ping He
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Yanlin Lu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Haiyan Xue
- Department of Acupuncture and Moxibustion, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| |
Collapse
|
26
|
Azer K, Kaddi CD, Barrett JS, Bai JPF, McQuade ST, Merrill NJ, Piccoli B, Neves-Zaph S, Marchetti L, Lombardo R, Parolo S, Immanuel SRC, Baliga NS. History and Future Perspectives on the Discipline of Quantitative Systems Pharmacology Modeling and Its Applications. Front Physiol 2021; 12:637999. [PMID: 33841175 PMCID: PMC8027332 DOI: 10.3389/fphys.2021.637999] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/25/2021] [Indexed: 12/24/2022] Open
Abstract
Mathematical biology and pharmacology models have a long and rich history in the fields of medicine and physiology, impacting our understanding of disease mechanisms and the development of novel therapeutics. With an increased focus on the pharmacology application of system models and the advances in data science spanning mechanistic and empirical approaches, there is a significant opportunity and promise to leverage these advancements to enhance the development and application of the systems pharmacology field. In this paper, we will review milestones in the evolution of mathematical biology and pharmacology models, highlight some of the gaps and challenges in developing and applying systems pharmacology models, and provide a vision for an integrated strategy that leverages advances in adjacent fields to overcome these challenges.
Collapse
Affiliation(s)
- Karim Azer
- Quantitative Sciences, Bill and Melinda Gates Medical Research Institute, Cambridge, MA, United States
| | - Chanchala D. Kaddi
- Quantitative Sciences, Bill and Melinda Gates Medical Research Institute, Cambridge, MA, United States
| | | | - Jane P. F. Bai
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Sean T. McQuade
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, United States
| | - Nathaniel J. Merrill
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, United States
| | - Benedetto Piccoli
- Department of Mathematical Sciences and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, United States
| | - Susana Neves-Zaph
- Translational Disease Modeling, Data and Data Science, Sanofi, Bridgewater, NJ, United States
| | - Luca Marchetti
- Fondazione the Microsoft Research – University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, Italy
| | - Rosario Lombardo
- Fondazione the Microsoft Research – University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, Italy
| | - Silvia Parolo
- Fondazione the Microsoft Research – University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, Italy
| | | | | |
Collapse
|
27
|
Bloomingdale P, Karelina T, Cirit M, Muldoon SF, Baker J, McCarty WJ, Geerts H, Macha S. Quantitative systems pharmacology in neuroscience: Novel methodologies and technologies. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:412-419. [PMID: 33719204 PMCID: PMC8129713 DOI: 10.1002/psp4.12607] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/02/2020] [Accepted: 02/03/2021] [Indexed: 11/12/2022]
Abstract
The development and application of quantitative systems pharmacology models in neuroscience have been modest relative to other fields, such as oncology and immunology, which may reflect the complexity of the brain. Technological and methodological advancements have enhanced the quantitative understanding of brain physiology and pathophysiology and the effects of pharmacological interventions. To maximize the knowledge gained from these novel data types, pharmacometrics modelers may need to expand their toolbox to include additional mathematical and statistical frameworks. A session was held at the 10th annual American Conference on Pharmacometrics (ACoP10) to highlight several recent advancements in quantitative and systems neuroscience. In this mini‐review, we provide a brief overview of technological and methodological advancements in the neuroscience therapeutic area that were discussed during the session and how these can be leveraged with quantitative systems pharmacology modeling to enhance our understanding of neurological diseases. Microphysiological systems using human induced pluripotent stem cells (IPSCs), digital biomarkers, and large‐scale imaging offer more clinically relevant experimental datasets, enhanced granularity, and a plethora of data to potentially improve the preclinical‐to‐clinical translation of therapeutics. Network neuroscience methodologies combined with quantitative systems models of neurodegenerative disease could help bridge the gap between cellular and molecular alterations and clinical end points through the integration of information on neural connectomics. Additional topics, such as the neuroimmune system, microbiome, single‐cell transcriptomic technologies, and digital device biomarkers, are discussed in brief.
Collapse
Affiliation(s)
- Peter Bloomingdale
- Quantitative Pharmacology and Pharmacometrics, Merck & Co. Inc, Kenilworth, New Jersey, USA
| | | | - Murat Cirit
- Javelin Biotech, Inc, Woburn, Massachusetts, USA
| | - Sarah F Muldoon
- Mathematics Department, CDSE Program, Neuroscience Program, University at Buffalo, SUNY, Buffalo, New York, USA
| | - Justin Baker
- Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Sreeraj Macha
- Quantitative Pharmacology, Sanofi, Bridgewater, New Jersey, USA
| |
Collapse
|
28
|
McComb M, Bies R, Ramanathan M. Machine learning in pharmacometrics: Opportunities and challenges. Br J Clin Pharmacol 2021; 88:1482-1499. [PMID: 33634893 DOI: 10.1111/bcp.14801] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
The explosive growth in medical devices, imaging and diagnostics, computing, and communication and information technologies in drug development and healthcare has created an ever-expanding data landscape that the pharmacometrics (PMX) research community must now traverse. The tools of machine learning (ML) have emerged as a powerful computational approach in other data-rich disciplines but its effective utilization in the pharmaceutical sciences and PMX modelling is in its infancy. ML-based methods can complement PMX modelling by enabling the information in diverse sources of big data, e.g. population-based public databases and disease-specific clinical registries, to be harnessed because they are capable of efficiently identifying salient variables associated with outcomes and delineating their interdependencies. ML algorithms are computationally efficient, have strong predictive capabilities and can enable learning in the big data setting. ML algorithms can be viewed as providing a computational bridge from big data to complement PMX modelling. This review provides an overview of the strengths and weaknesses of ML approaches vis-à-vis population methods, assesses current research into ML applications in the pharmaceutical sciences and provides perspective for potential opportunities and strategies for the successful integration and utilization of ML in PMX.
Collapse
Affiliation(s)
- Mason McComb
- Department of Pharmaceutical Sciences, University at Buffalo, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Robert Bies
- Department of Pharmaceutical Sciences, University at Buffalo, University at Buffalo, State University of New York, Buffalo, NY, USA.,Institute for Computational Data Science, University at Buffalo, NY, USA
| | - Murali Ramanathan
- Department of Pharmaceutical Sciences, University at Buffalo, University at Buffalo, State University of New York, Buffalo, NY, USA.,Department of Neurology, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
29
|
Sinisi S, Alimguzhin V, Mancini T, Tronci E, Leeners B. Complete populations of virtual patients for in silico clinical trials. Bioinformatics 2020; 36:5465-5472. [PMID: 33325489 DOI: 10.1093/bioinformatics/btaa1026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/30/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
MOTIVATION Model-based approaches to safety and efficacy assessment of pharmacological drugs, treatment strategies, or medical devices (In Silico Clinical Trial, ISCT) aim to decrease time and cost for the needed experimentations, reduce animal and human testing, and enable precision medicine. Unfortunately, in presence of non-identifiable models (e.g., reaction networks), parameter estimation is not enough to generate complete populations of Virtual Patient (VPs), i.e., populations guaranteed to show the entire spectrum of model behaviours (phenotypes), thus ensuring representativeness of the trial. RESULTS We present methods and software based on global search driven by statistical model checking that, starting from a (non-identifiable) quantitative model of the human physiology (plus drugs PK/PD) and suitable biological and medical knowledge elicited from experts, compute a population of VPs whose behaviours are representative of the whole spectrum of phenotypes entailed by the model (completeness) and pairwise distinguishable according to user-provided criteria. This enables full granularity control on the size of the population to employ in an ISCT, guaranteeing representativeness while avoiding over-representation of behaviours.We proved the effectiveness of our algorithm on a non-identifiable ODE-based model of the female Hypothalamic-Pituitary-Gonadal axis, by generating a population of 4 830 264 VPs stratified into 7 levels (at different granularity of behaviours), and assessed its representativeness against 86 retrospective health records from Pfizer, Hannover Medical School and University Hospital of Lausanne. The datasets are respectively covered by our VPs within Average Normalised Mean Absolute Error of 15%, 20%, and 35% (90% of the latter dataset is covered within 20% error).
Collapse
Affiliation(s)
- S Sinisi
- Computer Science Department, Sapienza University of Rome, Italy
| | - V Alimguzhin
- Computer Science Department, Sapienza University of Rome, Italy
| | - T Mancini
- Computer Science Department, Sapienza University of Rome, Italy
| | - E Tronci
- Computer Science Department, Sapienza University of Rome, Italy
| | - B Leeners
- Division of Reproductive Endocrinology, University Hospital Zurich, Switzerland
| |
Collapse
|
30
|
Marku M, Verstraete N, Raynal F, Madrid-Mencía M, Domagala M, Fournié JJ, Ysebaert L, Poupot M, Pancaldi V. Insights on TAM Formation from a Boolean Model of Macrophage Polarization Based on In Vitro Studies. Cancers (Basel) 2020; 12:cancers12123664. [PMID: 33297362 PMCID: PMC7762229 DOI: 10.3390/cancers12123664] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary The recent success of immunotherapy treatments against cancer relies on helping our own body’s defenses in the fight against tumours, namely reinvigorating the cancer killing action of T cells. Unfortunately, in a large proportion of patients these therapies are ineffective, in part due to the presence of other immune cells, macrophages, which are mis-educated by the cancer cells into promoting tumour growth. Here we start from an existing model of macrophage polarization and extend it to the specific conditions encountered inside a tumour by adding signals, receptors, transcription factors and cytokines that are known to be the key components in establishing the cancer cell-macrophage interaction. Then we use a mathematical Boolean model applied to a gene regulatory network of this biological process to simulate its temporal behaviour and explore scenarios that have not been experimentally tested so far. Additionally, the KO and overexpression simulations successfully reproduce the known experimental results while predicting the potential role of regulators (such as STAT1 and EGF) in preventing the formation of pro-tumoural macrophages, which can be tested experimentally. Abstract The tumour microenvironment is the surrounding of a tumour, including blood vessels, fibroblasts, signaling molecules, the extracellular matrix and immune cells, especially neutrophils and monocyte-derived macrophages. In a tumour setting, macrophages encompass a spectrum between a tumour-suppressive (M1) or tumour-promoting (M2) state. The biology of macrophages found in tumours (Tumour Associated Macrophages) remains unclear, but understanding their impact on tumour progression is highly important. In this paper, we perform a comprehensive analysis of a macrophage polarization network, following two lines of enquiry: (i) we reconstruct the macrophage polarization network based on literature, extending it to include important stimuli in a tumour setting, and (ii) we build a dynamical model able to reproduce macrophage polarization in the presence of different stimuli, including the contact with cancer cells. Our simulations recapitulate the documented macrophage phenotypes and their dependencies on specific receptors and transcription factors, while also unravelling the formation of a special type of tumour associated macrophages in an in vitro model of chronic lymphocytic leukaemia. This model constitutes the first step towards elucidating the cross-talk between immune and cancer cells inside tumours, with the ultimate goal of identifying new therapeutic targets that could control the formation of tumour associated macrophages in patients.
Collapse
Affiliation(s)
- Malvina Marku
- INSERM, Centre de Recherches en Cancérologie de Toulouse, 2 Avenue Hubert Curien, 31037 Toulouse, France; (N.V.); (F.R.); (M.M.-M.); (M.D.); (J.-J.F.); (L.Y.); (M.P.)
- Université III Toulouse Paul Sabatier, Route de Narbonne, 31330 Toulouse, France
- Correspondence: (M.M.); (V.P.); Tel.: +33-5-82-74-17-74 (M.M.)
| | - Nina Verstraete
- INSERM, Centre de Recherches en Cancérologie de Toulouse, 2 Avenue Hubert Curien, 31037 Toulouse, France; (N.V.); (F.R.); (M.M.-M.); (M.D.); (J.-J.F.); (L.Y.); (M.P.)
- Université III Toulouse Paul Sabatier, Route de Narbonne, 31330 Toulouse, France
| | - Flavien Raynal
- INSERM, Centre de Recherches en Cancérologie de Toulouse, 2 Avenue Hubert Curien, 31037 Toulouse, France; (N.V.); (F.R.); (M.M.-M.); (M.D.); (J.-J.F.); (L.Y.); (M.P.)
- Université III Toulouse Paul Sabatier, Route de Narbonne, 31330 Toulouse, France
| | - Miguel Madrid-Mencía
- INSERM, Centre de Recherches en Cancérologie de Toulouse, 2 Avenue Hubert Curien, 31037 Toulouse, France; (N.V.); (F.R.); (M.M.-M.); (M.D.); (J.-J.F.); (L.Y.); (M.P.)
- Université III Toulouse Paul Sabatier, Route de Narbonne, 31330 Toulouse, France
| | - Marcin Domagala
- INSERM, Centre de Recherches en Cancérologie de Toulouse, 2 Avenue Hubert Curien, 31037 Toulouse, France; (N.V.); (F.R.); (M.M.-M.); (M.D.); (J.-J.F.); (L.Y.); (M.P.)
- Université III Toulouse Paul Sabatier, Route de Narbonne, 31330 Toulouse, France
| | - Jean-Jacques Fournié
- INSERM, Centre de Recherches en Cancérologie de Toulouse, 2 Avenue Hubert Curien, 31037 Toulouse, France; (N.V.); (F.R.); (M.M.-M.); (M.D.); (J.-J.F.); (L.Y.); (M.P.)
- Université III Toulouse Paul Sabatier, Route de Narbonne, 31330 Toulouse, France
| | - Loïc Ysebaert
- INSERM, Centre de Recherches en Cancérologie de Toulouse, 2 Avenue Hubert Curien, 31037 Toulouse, France; (N.V.); (F.R.); (M.M.-M.); (M.D.); (J.-J.F.); (L.Y.); (M.P.)
- Université III Toulouse Paul Sabatier, Route de Narbonne, 31330 Toulouse, France
- Service d’Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, 31330 Toulouse, France
| | - Mary Poupot
- INSERM, Centre de Recherches en Cancérologie de Toulouse, 2 Avenue Hubert Curien, 31037 Toulouse, France; (N.V.); (F.R.); (M.M.-M.); (M.D.); (J.-J.F.); (L.Y.); (M.P.)
- Université III Toulouse Paul Sabatier, Route de Narbonne, 31330 Toulouse, France
| | - Vera Pancaldi
- INSERM, Centre de Recherches en Cancérologie de Toulouse, 2 Avenue Hubert Curien, 31037 Toulouse, France; (N.V.); (F.R.); (M.M.-M.); (M.D.); (J.-J.F.); (L.Y.); (M.P.)
- Université III Toulouse Paul Sabatier, Route de Narbonne, 31330 Toulouse, France
- Barcelona Supercomputing Center, Carrer de Jordi Girona, 29, 31, 08034 Barcelona, Spain
- Correspondence: (M.M.); (V.P.); Tel.: +33-5-82-74-17-74 (M.M.)
| |
Collapse
|
31
|
Abstract
Network theory provides one of the most potent analysis tools for the study of complex systems. In this paper, we illustrate the network-based perspective in drug research and how it is coherent with the new paradigm of drug discovery. We first present data sources from which networks are built, then show some examples of how the networks can be used to investigate drug-related systems. A section is devoted to network-based inference applications, i.e., prediction methods based on interactomes, that can be used to identify putative drug-target interactions without resorting to 3D modeling. Finally, we present some aspects of Boolean networks dynamics, anticipating that it might become a very potent modeling framework to develop in silico screening protocols able to simulate phenotypic screening experiments. We conclude that network applications integrated with machine learning and 3D modeling methods will become an indispensable tool for computational drug discovery in the next years.
Collapse
Affiliation(s)
- Maurizio Recanatini
- Department of Pharmacy and
Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Chiara Cabrelle
- Department of Pharmacy and
Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| |
Collapse
|
32
|
Rajapakse VN, Herrada S, Lavi O. Phenotype stability under dynamic brain-tumor environment stimuli maps glioblastoma progression in patients. SCIENCE ADVANCES 2020; 6:eaaz4125. [PMID: 32832595 PMCID: PMC7439317 DOI: 10.1126/sciadv.aaz4125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 03/20/2020] [Indexed: 06/11/2023]
Abstract
Although tumor invasiveness is known to drive glioblastoma (GBM) recurrence, current approaches to treatment assume a fairly simple GBM phenotype transition map. We provide new analyses to estimate the likelihood of reaching or remaining in a phenotype under dynamic, physiologically likely perturbations of stimuli ("phenotype stability"). We show that higher stability values of the motile phenotype (Go) are associated with reduced patient survival. Moreover, induced motile states are capable of driving GBM recurrence. We found that the Dormancy and Go phenotypes are equally represented in advanced GBM samples, with natural transitioning between the two. Furthermore, Go and Grow phenotype transitions are mostly driven by tumor-brain stimuli. These are difficult to regulate directly, but could be modulated by reprogramming tumor-associated cell types. Our framework provides a foundation for designing targeted perturbations of the tumor-brain environment, by assessing their impact on GBM phenotypic plasticity, and is corroborated by analyses of patient data.
Collapse
Affiliation(s)
- Vinodh N. Rajapakse
- Integrative Cancer Dynamics Unit, Laboratory of Cell Biology, CCR, NCI, NIH, Bethesda, MD, USA
| | - Sylvia Herrada
- Integrative Cancer Dynamics Unit, Laboratory of Cell Biology, CCR, NCI, NIH, Bethesda, MD, USA
| | - Orit Lavi
- Integrative Cancer Dynamics Unit, Laboratory of Cell Biology, CCR, NCI, NIH, Bethesda, MD, USA
| |
Collapse
|
33
|
Deritei D, Rozum J, Ravasz Regan E, Albert R. A feedback loop of conditionally stable circuits drives the cell cycle from checkpoint to checkpoint. Sci Rep 2019; 9:16430. [PMID: 31712566 PMCID: PMC6848090 DOI: 10.1038/s41598-019-52725-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022] Open
Abstract
We perform logic-based network analysis on a model of the mammalian cell cycle. This model is composed of a Restriction Switch driving cell cycle commitment and a Phase Switch driving mitotic entry and exit. By generalizing the concept of stable motif, i.e., a self-sustaining positive feedback loop that maintains an associated state, we introduce the concept of a conditionally stable motif, the stability of which is contingent on external conditions. We show that the stable motifs of the Phase Switch are contingent on the state of three nodes through which it receives input from the rest of the network. Biologically, these conditions correspond to cell cycle checkpoints. Holding these nodes locked (akin to a checkpoint-free cell) transforms the Phase Switch into an autonomous oscillator that robustly toggles through the cell cycle phases G1, G2 and mitosis. The conditionally stable motifs of the Phase Switch Oscillator are organized into an ordered sequence, such that they serially stabilize each other but also cause their own destabilization. Along the way they channel the dynamics of the module onto a narrow path in state space, lending robustness to the oscillation. Self-destabilizing conditionally stable motifs suggest a general negative feedback mechanism leading to sustained oscillations.
Collapse
Affiliation(s)
- Dávid Deritei
- Department of Physics, Pennsylvania State University, University Park, PA, United States of America
- Department of Network and Data Science, Central European University, Budapest, Hungary
| | - Jordan Rozum
- Department of Physics, Pennsylvania State University, University Park, PA, United States of America
| | - Erzsébet Ravasz Regan
- Biochemistry and Molecular Biology, The College of Wooster, Wooster, OH, United States of America
| | - Réka Albert
- Department of Physics, Pennsylvania State University, University Park, PA, United States of America.
| |
Collapse
|
34
|
Rabajante JF, Del Rosario RCH. Modeling Long ncRNA-Mediated Regulation in the Mammalian Cell Cycle. Methods Mol Biol 2019; 1912:427-445. [PMID: 30635904 DOI: 10.1007/978-1-4939-8982-9_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Long noncoding RNAs (lncRNAs) are transcripts longer than 200 nucleotides that are not translated into proteins. They have recently gained widespread attention due to the finding that tens of thousands of lncRNAs reside in the human genome, and due to an increasing number of lncRNAs that are found to be associated with disease. Some lncRNAs, including disease-associated ones, play different roles in regulating the cell cycle. Mathematical models of the cell cycle have been useful in better understanding this biological system, such as how it could be robust to some perturbations and how the cell cycle checkpoints could act as a switch. Here, we discuss mathematical modeling techniques for studying lncRNA regulation of the mammalian cell cycle. We present examples on how modeling via network analysis and differential equations can provide novel predictions toward understanding cell cycle regulation in response to perturbations such as DNA damage.
Collapse
Affiliation(s)
- Jomar F Rabajante
- Institute of Mathematical Sciences and Physics, University of the Philippines Los Baños, Laguna, Philippines.
| | - Ricardo C H Del Rosario
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
35
|
Niu J, Straubinger RM, Mager DE. Pharmacodynamic Drug-Drug Interactions. Clin Pharmacol Ther 2019; 105:1395-1406. [PMID: 30912119 PMCID: PMC6529235 DOI: 10.1002/cpt.1434] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/13/2019] [Indexed: 01/01/2023]
Abstract
Pharmacodynamic drug-drug interactions (DDIs) occur when the pharmacological effect of one drug is altered by that of another drug in a combination regimen. DDIs often are classified as synergistic, additive, or antagonistic in nature, albeit these terms are frequently misused. Within a complex pathophysiological system, the mechanism of interaction may occur at the same target or through alternate pathways. Quantitative evaluation of pharmacodynamic DDIs by employing modeling and simulation approaches is needed to identify and optimize safe and effective combination therapy regimens. This review investigates the opportunities and challenges in pharmacodynamic DDI studies and highlights examples of quantitative methods for evaluating pharmacodynamic DDIs, with a particular emphasis on the use of mechanism-based modeling and simulation in DDI studies. Advancements in both experimental and computational techniques will enable the application of better, model-informed assessments of pharmacodynamic DDIs in drug discovery, development, and therapeutics.
Collapse
Affiliation(s)
- Jin Niu
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Robert M. Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Donald E. Mager
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| |
Collapse
|
36
|
Campbell C, Albert R. Edgetic perturbations to eliminate fixed-point attractors in Boolean regulatory networks. CHAOS (WOODBURY, N.Y.) 2019; 29:023130. [PMID: 30823730 DOI: 10.1063/1.5083060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 01/29/2019] [Indexed: 06/09/2023]
Abstract
The dynamics of complex biological networks may be modeled in a Boolean framework, where the state of each system component is either abundant (ON) or scarce/absent (OFF), and each component's dynamic trajectory is determined by a logical update rule involving the state(s) of its regulator(s). It is possible to encode the update rules in the topology of the so-called expanded graph, analysis of which reveals the long-term behavior, or attractors, of the network. Here, we develop an algorithm to perturb the expanded graph (or, equivalently, the logical update rules) to eliminate stable motifs: subgraphs that cause a subset of components to stabilize to one state. Depending on the topology of the expanded graph, these perturbations lead to the modification or loss of the corresponding attractor. While most perturbations of biological regulatory networks in the literature involve the knockout (fixing to OFF) or constitutive activation (fixing to ON) of one or more nodes, we here consider edgetic perturbations, where a node's update rule is modified such that one or more of its regulators is viewed as ON or OFF regardless of its actual state. We apply the methodology to two biological networks. In a network representing T-LGL leukemia, we identify edgetic perturbations that eliminate the cancerous attractor, leaving only the healthy attractor representing cell death. In a network representing drought-induced closure of plant stomata, we identify edgetic perturbations that modify the single attractor such that stomata, instead of being fixed in the closed state, oscillates between the open and closed states.
Collapse
Affiliation(s)
- Colin Campbell
- Department of Physics, Washington College, Chestertown, Maryland 21620, USA
| | - Réka Albert
- Department of Physics, Pennsylvania State University, University Park, Pennsylvania 16802, USA
| |
Collapse
|
37
|
|
38
|
Hou W, Ruan P, Ching WK, Akutsu T. On the number of driver nodes for controlling a Boolean network when the targets are restricted to attractors. J Theor Biol 2018; 463:1-11. [PMID: 30543810 DOI: 10.1016/j.jtbi.2018.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/01/2018] [Accepted: 12/10/2018] [Indexed: 01/28/2023]
Abstract
It is known that many driver nodes are required to control complex biological networks. Previous studies imply that O(N) driver nodes are required in both linear complex network and Boolean network models with N nodes if an arbitrary state is specified as the target. In order to cope with this intrinsic difficulty, we consider a special case of the control problem in which the targets are restricted to attractors. For this special case, we mathematically prove under the uniform distribution of states in basins that the expected number of driver nodes is only O(log2N+log2M) for controlling Boolean networks, where M is the number of attractors. Since it is expected that M is not very large in many practical networks, the new model requires a much smaller number of driver nodes. This result is based on discovery of novel relationships between control problems on Boolean networks and the coupon collector's problem, a well-known concept in combinatorics. We also provide lower bounds of the number of driver nodes as well as simulation results using artificial and realistic network data, which support our theoretical findings.
Collapse
Affiliation(s)
- Wenpin Hou
- Department of Computer Science, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218-2608, USA; Advanced Modeling and Applied Computing Laboratory, Department of Mathematics, The University of Hong Kong, Pokfulam Road, Hong Kong.
| | - Peiying Ruan
- Deep Learning Solution Architect, NVIDIA, Tokyo, Japan.
| | - Wai-Ki Ching
- Advanced Modeling and Applied Computing Laboratory, Department of Mathematics, The University of Hong Kong, Pokfulam Road, Hong Kong; Hughes Hall, Wollaston Road, Cambridge, UK; School of Economics and Management, Beijing University of Chemical Technology, North Third Ring Road, Beijing, China.
| | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Kyoto, Japan.
| |
Collapse
|
39
|
van Hasselt JGC, Iyengar R. Systems Pharmacology: Defining the Interactions of Drug Combinations. Annu Rev Pharmacol Toxicol 2018; 59:21-40. [PMID: 30260737 DOI: 10.1146/annurev-pharmtox-010818-021511] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The majority of diseases are associated with alterations in multiple molecular pathways and complex interactions at the cellular and organ levels. Single-target monotherapies therefore have intrinsic limitations with respect to their maximum therapeutic benefits. The potential of combination drug therapies has received interest for the treatment of many diseases and is well established in some areas, such as oncology. Combination drug treatments may allow us to identify synergistic drug effects, reduce adverse drug reactions, and address variability in disease characteristics between patients. Identification of combination therapies remains challenging. We discuss current state-of-the-art systems pharmacology approaches to enable rational identification of combination therapies. These approaches, which include characterization of mechanisms of disease and drug action at a systems level, can enable understanding of drug interactions at the molecular, cellular, physiological, and organismal levels. Such multiscale understanding can enable precision medicine by promoting the rational development of combination therapy at the level of individual patients for many diseases.
Collapse
Affiliation(s)
- J G Coen van Hasselt
- Department of Pharmacological Sciences, Systems Biology Center, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; .,Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug Research, Leiden University, 2333 Leiden, Netherlands;
| | - Ravi Iyengar
- Department of Pharmacological Sciences, Systems Biology Center, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| |
Collapse
|
40
|
Piñero J, Furlong LI, Sanz F. In silico models in drug development: where we are. Curr Opin Pharmacol 2018; 42:111-121. [PMID: 30205360 DOI: 10.1016/j.coph.2018.08.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/30/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023]
Abstract
The use and utility of computational models in drug development has significantly grown in the last decades, fostered by the availability of high throughput datasets and new data analysis strategies. These in silico approaches are demonstrating their ability to generate reliable predictions as well as new knowledge on the mode of action of drugs and the mechanisms underlying their side effects, altogether helping to reduce the costs of drug development. The aim of this review is to provide a panorama of developments in the field in the last two years.
Collapse
Affiliation(s)
- Janet Piñero
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Laura I Furlong
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Ferran Sanz
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain.
| |
Collapse
|
41
|
G T Zañudo J, Steinway SN, Albert R. Discrete dynamic network modeling of oncogenic signaling: Mechanistic insights for personalized treatment of cancer. ACTA ACUST UNITED AC 2018; 9:1-10. [PMID: 32954058 PMCID: PMC7487767 DOI: 10.1016/j.coisb.2018.02.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Targeted drugs disrupting proteins that are dysregulated in cancer have emerged as promising treatments because of their specificity to cancer cell aberrations and thus their improved side effect profile. However, their success remains limited, largely due to existing or emergent therapy resistance. We suggest that this is due to limited understanding of the entire relevant cellular landscape. A class of mathematical models called discrete dynamic network models can be used to understand the integrated effect of an individual tumor's aberrations. We review the recent literature on discrete dynamic models of cancer and highlight their predicted therapeutic strategies. We believe dynamic network modeling can be used to drive treatment decision-making in a personalized manner to direct improved treatments in cancer. Cancer is rooted in incorrect cellular decisions caused by genetic alterations. Dynamic models of signaling networks can map the relevant repertoire of alterations. Discrete dynamic network models can predict therapeutic interventions. Progress in personalized medicine needs integration of multiple data and model types.
Collapse
Affiliation(s)
- Jorge G T Zañudo
- Department of Physics, The Pennsylvania State University, University Park, PA 16802, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute and Broad Institute of Harvard and MIT, Boston MA, USA
| | - Steven N Steinway
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Réka Albert
- Department of Physics, The Pennsylvania State University, University Park, PA 16802, USA.,Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
42
|
Yang G, Gómez Tejeda Zañudo J, Albert R. Target Control in Logical Models Using the Domain of Influence of Nodes. Front Physiol 2018; 9:454. [PMID: 29867523 PMCID: PMC5951947 DOI: 10.3389/fphys.2018.00454] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/13/2018] [Indexed: 11/13/2022] Open
Abstract
Dynamical models of biomolecular networks are successfully used to understand the mechanisms underlying complex diseases and to design therapeutic strategies. Network control and its special case of target control, is a promising avenue toward developing disease therapies. In target control it is assumed that a small subset of nodes is most relevant to the system's state and the goal is to drive the target nodes into their desired states. An example of target control would be driving a cell to commit to apoptosis (programmed cell death). From the experimental perspective, gene knockout, pharmacological inhibition of proteins, and providing sustained external signals are among practical intervention techniques. We identify methodologies to use the stabilizing effect of sustained interventions for target control in Boolean network models of biomolecular networks. Specifically, we define the domain of influence (DOI) of a node (in a certain state) to be the nodes (and their corresponding states) that will be ultimately stabilized by the sustained state of this node regardless of the initial state of the system. We also define the related concept of the logical domain of influence (LDOI) of a node, and develop an algorithm for its identification using an auxiliary network that incorporates the regulatory logic. This way a solution to the target control problem is a set of nodes whose DOI can cover the desired target node states. We perform greedy randomized adaptive search in node state space to find such solutions. We apply our strategy to in silico biological network models of real systems to demonstrate its effectiveness.
Collapse
Affiliation(s)
- Gang Yang
- Department of Physics, Pennsylvania State University, University Park, PA, United States
| | - Jorge Gómez Tejeda Zañudo
- Department of Physics, Pennsylvania State University, University Park, PA, United States.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Réka Albert
- Department of Physics, Pennsylvania State University, University Park, PA, United States.,Department of Biology, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
43
|
Chen YL, Zhang YL, Dai YC, Tang ZP. Systems pharmacology approach reveals the antiinflammatory effects of Ampelopsis grossedentata on dextran sodium sulfate-induced colitis. World J Gastroenterol 2018; 24:1398-1409. [PMID: 29632421 PMCID: PMC5889820 DOI: 10.3748/wjg.v24.i13.1398] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 02/12/2018] [Accepted: 03/03/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the protective effects of Ampelopsis grossedentata (AMP) on dextran sulfate sodium (DSS)-induced colitis in mice based on systems pharmacology approach.
METHODS Systems pharmacology approach was used to predict the active ingredients, candidate targets and the efficacy of AMP on ulcerative colitis (UC) using a holistic process of active compound screening, target fishing, network construction and analysis. A DSS-induced colitis model in C57BL/6 mice (n = 10/group) was constructed and treated with 5-aminosalicylic acid (100 mg/kg/d) and AMP (400 mg/kg/d) to confirm the underlying mechanisms and effects of AMP on UC with western blot analyses, polymerase chain reaction, histological staining and immunohistochemistry.
RESULTS The therapeutic effects of AMP against DSS-induced colitis were determined in the beginning, and the results showed that AMP significantly improved the disease in general observations and histopathology analysis. Subsequent systems pharmacology predicted 89 corresponding targets for the four candidate compounds of AMP, as well as 123 candidate targets of UC, and protein-protein interaction networks were constructed for the interaction of putative targets of AMP against UC. Enrichment analyses on TNF-α and RANKL/RANK, a receptor activator of NF-κB signaling pathways, were then carried out. Experimental validation revealed that inflammation-related signaling pathways were activated in the DSS group, and AMP significantly suppressed DSS-induced high expression of IRAK1, TRAF6, IκB and NF-κB, and inhibited the elevated expression levels of TNF-α, IL-1β, IL-6 and IL-8.
CONCLUSION AMP could exert protective effects on UC via suppressing the IRAK1/TRAF6/NF-κB-mediated inflammatory signaling pathways.
Collapse
Affiliation(s)
- You-Lan Chen
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Department of Gastroenterology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ya-Li Zhang
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yan-Cheng Dai
- Department of Gastroenterology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Zhi-Peng Tang
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Department of Gastroenterology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|