1
|
Mendoza L, Vázquez-Ramírez R, Tzompantzi-de-Ita JM. The regulatory network that controls lymphopoiesis. Biosystems 2025; 248:105399. [PMID: 39828207 DOI: 10.1016/j.biosystems.2025.105399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Lymphopoiesis is the generation of the T, B and NK cell lineages from a common lymphoid-biased haematopoietic stem cell. The experimental study of this process has generated a large amount of cellular and molecular data. As a result, there is a considerable number of mathematical and computational models regarding different aspects of lymphopoiesis. We hereby present a regulatory network consisting of 95 nodes and 202 regulatory interactions among them. The network is studied as a qualitative dynamical system, which has as stationary states the molecular patterns reported for CLP, pre-B, B naive, PC, pNK, iNK, NK, DP, CD8 naive, CTL, CD4 naive, Th1, Th2, Th17 and Treg cells. Also, we show that the system is able to respond to specific stimuli to reproduce the ontogeny of the T, B and NK cell lineages.
Collapse
Affiliation(s)
- Luis Mendoza
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 3er Circuito Exterior s/n, Ciudad Universitaria, 04510, CdMx, México.
| | - Ricardo Vázquez-Ramírez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 3er Circuito Exterior s/n, Ciudad Universitaria, 04510, CdMx, México
| | - Juan Manuel Tzompantzi-de-Ita
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 3er Circuito Exterior s/n, Ciudad Universitaria, 04510, CdMx, México
| |
Collapse
|
2
|
Calzone L, Noël V, Barillot E, Kroemer G, Stoll G. Modeling signaling pathways in biology with MaBoSS: From one single cell to a dynamic population of heterogeneous interacting cells. Comput Struct Biotechnol J 2022; 20:5661-5671. [PMID: 36284705 PMCID: PMC9582792 DOI: 10.1016/j.csbj.2022.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/24/2022] Open
Abstract
As a result of the development of experimental technologies and the accumulation of data, biological and molecular processes can be described as complex networks of signaling pathways. These networks are often directed and signed, where nodes represent entities (genes/proteins) and arrows interactions. They are translated into mathematical models by adding a dynamic layer onto them. Such mathematical models help to understand and interpret non-intuitive experimental observations and to anticipate the response to external interventions such as drug effects on phenotypes. Several frameworks for modeling signaling pathways exist. The choice of the appropriate framework is often driven by the experimental context. In this review, we present MaBoSS, a tool based on Boolean modeling using a continuous time approach, which predicts time-dependent probabilities of entities in different biological contexts. MaBoSS was initially built to model the intracellular signaling in non-interacting homogeneous cell populations. MaBoSS was then adapted to model heterogeneous cell populations (EnsembleMaBoSS) by considering families of models rather than a unique model. To account for more complex questions, MaBoSS was extended to simulate dynamical interacting populations (UPMaBoSS), with a precise spatial distribution (PhysiBoSS). To illustrate all these levels of description, we show how each of these tools can be used with a running example of a simple model of cell fate decisions. Finally, we present practical applications to cancer biology and studies of the immune response.
Collapse
Affiliation(s)
- Laurence Calzone
- Institut Curie, PSL Research University, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, F-75006 Paris, France
| | - Vincent Noël
- Institut Curie, PSL Research University, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, F-75006 Paris, France
| | - Emmanuel Barillot
- Institut Curie, PSL Research University, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, F-75006 Paris, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisé par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Europén Georges Pompidou, AP-HP, Paris, France
| | - Gautier Stoll
- Centre de Recherche des Cordeliers, Equipe labellisé par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| |
Collapse
|
3
|
ROY PRITIKUMAR, ROY AMITKUMAR, KHAILOV EVGENIIN, AL BASIR FAHAD, GRIGORIEVA ELLINAV. A MODEL OF THE OPTIMAL IMMUNOTHERAPY OF PSORIASIS BY INTRODUCING IL-10 AND IL-22 INHIBITORS. J BIOL SYST 2020; 28:609-639. [DOI: 10.1142/s0218339020500084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Psoriasis is a chronic skin disease in which the process of hyper-proliferation (excessive division) of skin cells starts. Externally, psoriasis appears as red papules, on the surface of which there are scales of white–gray color. There is substantial evidence that T-helper cells take vital accountability for creating the hyper-proliferation of keratinocytes (skin cells), which causes itching of skin patches. In this paper, we propose a mathematical model describing the concentrations of T-helper and keratinocyte cell populations to predict cellular behaviors for psoriasis regulation under normal or anomalous immune circumstances. Local and global asymptotic stabilities of the model equilibria are investigated. Additionally, by introducing two scalar bounded controls into the model, the effect of combined immunotherapy using IL-10 and IL-22 inhibitors is analyzed. The optimal control problem of minimizing the cost of immune therapy and simultaneous optimizing the effect of this therapy on T-helper cells and keratinocytes proliferation is formulated and solved by applying the Pontryagin maximum principle. Within the restrictions of the proposed model, the obtained analytical and numerical outcomes suggest that the optimal strategy of injecting IL-10 and IL-22 inhibitors can be effective for psoriasis treatment.
Collapse
Affiliation(s)
- PRITI KUMAR ROY
- Centre for Mathematical Biology and Ecology, Department of Mathematics, Jadavpur University, Kolkata 700032, India
| | - AMIT KUMAR ROY
- Centre for Mathematical Biology and Ecology, Department of Mathematics, Jadavpur University, Kolkata 700032, India
| | - EVGENII N. KHAILOV
- Faculty of Computational Mathematics and Cybernetics, Lomonosov Moscow State University, Moscow 119992, Russia
| | - FAHAD AL BASIR
- Department of Mathematics, Asansol Girls College, Asansol-4, West Bengal 713304, India
| | - ELLINA V. GRIGORIEVA
- Department of Mathematics and Computer Sciences, Texas Woman’s University, Denton, TX 76204, USA
| |
Collapse
|
4
|
Stalidzans E, Zanin M, Tieri P, Castiglione F, Polster A, Scheiner S, Pahle J, Stres B, List M, Baumbach J, Lautizi M, Van Steen K, Schmidt HH. Mechanistic Modeling and Multiscale Applications for Precision Medicine: Theory and Practice. NETWORK AND SYSTEMS MEDICINE 2020. [DOI: 10.1089/nsm.2020.0002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Egils Stalidzans
- Computational Systems Biology Group, University of Latvia, Riga, Latvia
- Latvian Biomedical Reasearch and Study Centre, Riga, Latvia
| | - Massimiliano Zanin
- Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Pozuelo de Alarcón, Spain
| | - Paolo Tieri
- CNR National Research Council, IAC Institute for Applied Computing, Rome, Italy
| | - Filippo Castiglione
- CNR National Research Council, IAC Institute for Applied Computing, Rome, Italy
| | | | - Stefan Scheiner
- Institute for Mechanics of Materials and Structures, Vienna University of Technology, Vienna, Austria
| | - Jürgen Pahle
- BioQuant, Heidelberg University, Heidelberg, Germany
| | - Blaž Stres
- Department of Animal Science, University of Ljubljana, Ljubljana, Slovenia
- Faculty of Civil and Geodetic Engineering, University of Ljubljana, Ljubljana, Slovenia
- Department of Automation, Biocybernetics and Robotics, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Markus List
- Big Data in BioMedicine Research Group, Chair of Experimental Bioinformatics, TUM School of Weihenstephan, Technical University of Munich, Freising, Germany
| | - Jan Baumbach
- Chair of Experimental Bioinformatics, TUM School of Weihenstephan, Technical University of Munich, Freising, Germany
| | - Manuela Lautizi
- Computational Systems Medicine Research Group, Chair of Experimental Bioinformatics, TUM School of Weihenstephan, Technical University of Munich, Freising, Germany
| | - Kristel Van Steen
- BIO-Systems Genetics, GIGA-R, University of Liège, Liège, Belgium
- BIO3—Systems Medicine, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Harald H.H.W. Schmidt
- Department of Pharmacology and Personalised Medicine, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
5
|
Analyzing pharmacological intervention points: A method to calculate external stimuli to switch between steady states in regulatory networks. PLoS Comput Biol 2019; 15:e1007075. [PMID: 31310618 PMCID: PMC6660093 DOI: 10.1371/journal.pcbi.1007075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 07/26/2019] [Accepted: 05/07/2019] [Indexed: 11/21/2022] Open
Abstract
Once biological systems are modeled by regulatory networks, the next step is to include external stimuli, which model the experimental possibilities to affect the activity level of certain network’s nodes, in a mathematical framework. Then, this framework can be interpreted as a mathematical optimal control framework such that optimization algorithms can be used to determine external stimuli which cause a desired switch from an initial state of the network to another final state. These external stimuli are the intervention points for the corresponding biological experiment to obtain the desired outcome of the considered experiment. In this work, the model of regulatory networks is extended to controlled regulatory networks. For this purpose, external stimuli are considered which can affect the activity of the network’s nodes by activation or inhibition. A method is presented how to calculate a selection of external stimuli which causes a switch between two different steady states of a regulatory network. A software solution based on Jimena and Mathworks Matlab is provided. Furthermore, numerical examples are presented to demonstrate application and scope of the software on networks of 4 nodes, 11 nodes and 36 nodes. Moreover, we analyze the aggregation of platelets and the behavior of a basic T-helper cell protein-protein interaction network and its maturation towards Th0, Th1, Th2, Th17 and Treg cells in accordance with experimental data. Organisms can be seen as molecular networks being able to react on external stimuli. Experiments are performed to understand the underlying regulating mechanisms within the molecular network. A common purpose for these efforts is to reveal mechanisms with which the molecular networks can be affected to achieve a desired behavior. To cover the complexity of life these models of molecular networks often need to be quite huge and need to have many cross connections between the different agents of the network that regulate the behavior of the network. A useful tool to structure this complexity are mathematical methods. Once the model based on experiments is set up the experimental data can be further processed by mathematical methods. As experiments are cumbersome, the present work provides a framework that can be used to systematically figure out intervention points in molecular networks to cause a desired effect. In this way promising intervention strategies can be obtained. For instance the process of obtaining new drugs for pharmacological modulation can be shortened as in the best case the calculated intervention strategy just has to be validated with one experiment and the time consuming procedure of searching an intervention strategy with several experiments can be saved.
Collapse
|
6
|
Breitenbach T, Lorenz K, Dandekar T. How to Steer and Control ERK and the ERK Signaling Cascade Exemplified by Looking at Cardiac Insufficiency. Int J Mol Sci 2019; 20:E2179. [PMID: 31052520 PMCID: PMC6539830 DOI: 10.3390/ijms20092179] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/16/2019] [Accepted: 04/24/2019] [Indexed: 12/13/2022] Open
Abstract
Mathematical optimization framework allows the identification of certain nodes within a signaling network. In this work, we analyzed the complex extracellular-signal-regulated kinase 1 and 2 (ERK1/2) cascade in cardiomyocytes using the framework to find efficient adjustment screws for this cascade that is important for cardiomyocyte survival and maladaptive heart muscle growth. We modeled optimal pharmacological intervention points that are beneficial for the heart, but avoid the occurrence of a maladaptive ERK1/2 modification, the autophosphorylation of ERK at threonine 188 (ERK Thr 188 phosphorylation), which causes cardiac hypertrophy. For this purpose, a network of a cardiomyocyte that was fitted to experimental data was equipped with external stimuli that model the pharmacological intervention points. Specifically, two situations were considered. In the first one, the cardiomyocyte was driven to a desired expression level with different treatment strategies. These strategies were quantified with respect to beneficial effects and maleficent side effects and then which one is the best treatment strategy was evaluated. In the second situation, it was shown how to model constitutively activated pathways and how to identify drug targets to obtain a desired activity level that is associated with a healthy state and in contrast to the maleficent expression pattern caused by the constitutively activated pathway. An implementation of the algorithms used for the calculations is also presented in this paper, which simplifies the application of the presented framework for drug targeting, optimal drug combinations and the systematic and automatic search for pharmacological intervention points. The codes were designed such that they can be combined with any mathematical model given by ordinary differential equations.
Collapse
Affiliation(s)
- Tim Breitenbach
- Biozentrum, Universität Würzburg, Am Hubland, 97074 Würzburg, Germany.
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, Versbacher Straße 9, 97078 Würzburg, Germany.
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany.
| | - Thomas Dandekar
- Biozentrum, Universität Würzburg, Am Hubland, 97074 Würzburg, Germany.
| |
Collapse
|
7
|
Figueiredo AS, Schumacher A. The T helper type 17/regulatory T cell paradigm in pregnancy. Immunology 2016; 148:13-21. [PMID: 26855005 DOI: 10.1111/imm.12595] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/15/2016] [Accepted: 02/04/2016] [Indexed: 12/28/2022] Open
Abstract
T helper type 17 (Th17) and regulatory T (Treg) cells are active players in the establishment of tolerance and defence. These attributes of the immune system enmesh to guarantee the right level of protection. The healthy immune system, on the one hand, recognizes and eliminates dangerous non-self pathogens and, on the other hand, protects the healthy self. However, there are circumstances where this fine balance is disrupted. In fact, in situations such as in pregnancy, the foreign fetal antigens challenge the maternal immune system and Treg cells will dominate Th17 cells to guarantee fetal survival. In other situations such as autoimmunity, where the Th17 responses are often overwhelming, the immune system shifts towards an inflammatory profile and attacks the healthy tissue from the self. Interestingly, autoimmune patients have meliorating symptoms during pregnancy. This connects with the antagonist role of Th17 and Treg cells, and their specific profiles during these two immune challenging situations. In this review, we put into perspective the Th17/Treg ratio during pregnancy and autoimmunity, as well as in pregnant women with autoimmune conditions. We further review existing systems biology approaches that study specific mechanisms of these immune cells using mathematical modelling and we point out possible future directions of investigation. Understanding what maintains or disrupts the balance between these two opponent yet reciprocal cells in healthy physiological settings, sheds light into the development of innovative pharmacological approaches to fight pregnancy loss and autoimmunity.
Collapse
Affiliation(s)
- Ana Sofia Figueiredo
- Medical Faculty, Institute for Experimental Internal Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Schumacher
- Medical Faculty, Institute for Experimental Obstetrics and Gynecology, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
8
|
Méndez A, Mendoza L. A Network Model to Describe the Terminal Differentiation of B Cells. PLoS Comput Biol 2016; 12:e1004696. [PMID: 26751566 PMCID: PMC4720151 DOI: 10.1371/journal.pcbi.1004696] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 12/07/2015] [Indexed: 01/31/2023] Open
Abstract
Terminal differentiation of B cells is an essential process for the humoral immune response in vertebrates and is achieved by the concerted action of several transcription factors in response to antigen recognition and extracellular signals provided by T-helper cells. While there is a wealth of experimental data regarding the molecular and cellular signals involved in this process, there is no general consensus regarding the structure and dynamical properties of the underlying regulatory network controlling this process. We developed a dynamical model of the regulatory network controlling terminal differentiation of B cells. The structure of the network was inferred from experimental data available in the literature, and its dynamical behavior was analyzed by modeling the network both as a discrete and a continuous dynamical systems. The steady states of these models are consistent with the patterns of activation reported for the Naive, GC, Mem, and PC cell types. Moreover, the models are able to describe the patterns of differentiation from the precursor Naive to any of the GC, Mem, or PC cell types in response to a specific set of extracellular signals. We simulated all possible single loss- and gain-of-function mutants, corroborating the importance of Pax5, Bcl6, Bach2, Irf4, and Blimp1 as key regulators of B cell differentiation process. The model is able to represent the directional nature of terminal B cell differentiation and qualitatively describes key differentiation events from a precursor cell to terminally differentiated B cells. Generation of antibody-producing cells through terminal B cell differentiation represents a good model to study the formation of multiple effector cells from a progenitor cell type. This process is controlled by the action of several molecules that maintain cell type specific programs in response to cytokines, antigen recognition and the direct contact with T helper cells, forming a complex regulatory network. While there is a large body of experimental data regarding some of the key molecules involved in this process and there have been several efforts to reconstruct the underlying regulatory network, a general consensus about the structure and dynamical behavior of this network is lacking. Moreover, it is not well understood how this network controls the establishment of specific B cell expression patterns and how it responds to specific external signals. We present a model of the regulatory network controlling terminal B cell differentiation and analyze its dynamical behavior under normal and mutant conditions. The model recovers the patterns of differentiation of B cells and describes a large set of gain- and loss-of-function mutants. This model provides an unified framework to generate qualitative descriptions to interpret the role of intra- and extracellular regulators of B cell differentiation.
Collapse
Affiliation(s)
- Akram Méndez
- Programa de Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Ciudad de México, México
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Luis Mendoza
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
- C3, Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Ciudad de México, México
- * E-mail:
| |
Collapse
|
9
|
Intosalmi J, Ahlfors H, Rautio S, Mannerstöm H, Chen ZJ, Lahesmaa R, Stockinger B, Lähdesmäki H. Analyzing Th17 cell differentiation dynamics using a novel integrative modeling framework for time-course RNA sequencing data. BMC SYSTEMS BIOLOGY 2015; 9:81. [PMID: 26578352 PMCID: PMC4650136 DOI: 10.1186/s12918-015-0223-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/28/2015] [Indexed: 02/07/2023]
Abstract
Background The differentiation of naive CD 4+ helper T (Th) cells into effector Th17 cells is steered by extracellular cytokines that activate and control the lineage specific transcriptional program. While the inducing cytokine signals and core transcription factors driving the differentiation towards Th17 lineage are well known, detailed mechanistic interactions between the key components are poorly understood. Results We develop an integrative modeling framework which combines RNA sequencing data with mathematical modeling and enables us to construct a mechanistic model for the core Th17 regulatory network in a data-driven manner. Conclusions Our results show significant evidence, for instance, for inhibitory mechanisms between the transcription factors and reveal a previously unknown dependency between the dosage of the inducing cytokine TGF β and the expression of the master regulator of competing (induced) regulatory T cell lineage. Further, our experimental validation approves this dependency in Th17 polarizing conditions. Electronic supplementary material The online version of this article (doi:10.1186/s12918-015-0223-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jukka Intosalmi
- Department of Computer Science, Aalto University, Aalto, FI-00076, Finland.
| | - Helena Ahlfors
- The Francis Crick Institute, Mill Hill Laboratory, Mill HillLondon, UK. .,Current affiliation: Lymphocyte Signalling and Development, The Babraham Institute, Cambridge, UK.
| | - Sini Rautio
- Department of Computer Science, Aalto University, Aalto, FI-00076, Finland.
| | - Henrik Mannerstöm
- Department of Computer Science, Aalto University, Aalto, FI-00076, Finland.
| | - Zhi Jane Chen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi, Turku, Finland.
| | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi, Turku, Finland.
| | | | - Harri Lähdesmäki
- Department of Computer Science, Aalto University, Aalto, FI-00076, Finland. .,Turku Centre for Biotechnology, University of Turku and Åbo Akademi, Turku, Finland.
| |
Collapse
|
10
|
Mendoza L, Méndez A. A dynamical model of the regulatory network controlling lymphopoiesis. Biosystems 2015; 137:26-33. [PMID: 26408858 DOI: 10.1016/j.biosystems.2015.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 08/22/2015] [Accepted: 09/21/2015] [Indexed: 12/22/2022]
Abstract
Due to the large number of diseases associated to a malfunction of the hematopoietic system, there is an interest in knowing the molecular mechanisms controlling the differentiation of blood cell lineages. However, the structure and dynamical properties of the underlying regulatory network controlling this process is not well understood. This manuscript presents a regulatory network of 81 nodes, representing several types of molecules that regulate each other during the process of lymphopoiesis. The regulatory interactions were inferred mostly from published experimental data. However, 15 out of 159 regulatory interactions are predictions arising from the present study. The network is modelled as a continuous dynamical system, in the form of a set of differential equations. The dynamical behaviour of the model describes the differentiation process from the common lymphocyte precursor (CLP) to several mature B and T cell types; namely, plasma cell (PC), cytotoxic T lymphocyte (CTL), T helper 1 (Th1), Th2, Th17, and T regulatory (Treg) cells. The model qualitatively recapitulates key cellular differentiation events, being able to represent the directional and branched nature of lymphopoiesis, going from a multipotent progenitor to fully differentiated cell types.
Collapse
Affiliation(s)
- Luis Mendoza
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, Mexico.
| | - Akram Méndez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, Mexico; Programa de Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, México, Mexico
| |
Collapse
|
11
|
Martinez-Sanchez ME, Mendoza L, Villarreal C, Alvarez-Buylla ER. A Minimal Regulatory Network of Extrinsic and Intrinsic Factors Recovers Observed Patterns of CD4+ T Cell Differentiation and Plasticity. PLoS Comput Biol 2015; 11:e1004324. [PMID: 26090929 PMCID: PMC4475012 DOI: 10.1371/journal.pcbi.1004324] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 05/07/2015] [Indexed: 12/24/2022] Open
Abstract
CD4+ T cells orchestrate the adaptive immune response in vertebrates. While both experimental and modeling work has been conducted to understand the molecular genetic mechanisms involved in CD4+ T cell responses and fate attainment, the dynamic role of intrinsic (produced by CD4+ T lymphocytes) versus extrinsic (produced by other cells) components remains unclear, and the mechanistic and dynamic understanding of the plastic responses of these cells remains incomplete. In this work, we studied a regulatory network for the core transcription factors involved in CD4+ T cell-fate attainment. We first show that this core is not sufficient to recover common CD4+ T phenotypes. We thus postulate a minimal Boolean regulatory network model derived from a larger and more comprehensive network that is based on experimental data. The minimal network integrates transcriptional regulation, signaling pathways and the micro-environment. This network model recovers reported configurations of most of the characterized cell types (Th0, Th1, Th2, Th17, Tfh, Th9, iTreg, and Foxp3-independent T regulatory cells). This transcriptional-signaling regulatory network is robust and recovers mutant configurations that have been reported experimentally. Additionally, this model recovers many of the plasticity patterns documented for different T CD4+ cell types, as summarized in a cell-fate map. We tested the effects of various micro-environments and transient perturbations on such transitions among CD4+ T cell types. Interestingly, most cell-fate transitions were induced by transient activations, with the opposite behavior associated with transient inhibitions. Finally, we used a novel methodology was used to establish that T-bet, TGF-β and suppressors of cytokine signaling proteins are keys to recovering observed CD4+ T cell plastic responses. In conclusion, the observed CD4+ T cell-types and transition patterns emerge from the feedback between the intrinsic or intracellular regulatory core and the micro-environment. We discuss the broader use of this approach for other plastic systems and possible therapeutic interventions.
Collapse
Affiliation(s)
- Mariana Esther Martinez-Sanchez
- Departamento de Ecología Funcional, Instituto de Ecología, Universidad Nacional Autónoma de México, Coyoacán, México Distrito Federal, México
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Coyoacán, México Distrito Federal, México
| | - Luis Mendoza
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México Distrito Federal, México
| | - Carlos Villarreal
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Coyoacán, México Distrito Federal, México
- Departamento de Física Teórica, Instituto de Física, Universidad Nacional Autónoma de México, México Distrito Federal, México
| | - Elena R. Alvarez-Buylla
- Departamento de Ecología Funcional, Instituto de Ecología, Universidad Nacional Autónoma de México, Coyoacán, México Distrito Federal, México
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Coyoacán, México Distrito Federal, México
- * E-mail:
| |
Collapse
|
12
|
Karl S, Dandekar T. Convergence behaviour and Control in Non-Linear Biological Networks. Sci Rep 2015; 5:9746. [PMID: 26068060 PMCID: PMC4464179 DOI: 10.1038/srep09746] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 03/13/2015] [Indexed: 12/18/2022] Open
Abstract
Control of genetic regulatory networks is challenging to define and quantify. Previous control centrality metrics, which aim to capture the ability of individual nodes to control the system, have been found to suffer from plausibility and applicability problems. Here we present a new approach to control centrality based on network convergence behaviour, implemented as an extension of our genetic regulatory network simulation framework Jimena ( http://stefan-karl.de/jimena). We distinguish three types of network control, and show how these mathematical concepts correspond to experimentally verified node functions and signalling pathways in immunity and cell differentiation: Total control centrality quantifies the impact of node mutations and identifies potential pharmacological targets such as genes involved in oncogenesis (e.g. zinc finger protein GLI2 or bone morphogenetic proteins in chondrocytes). Dynamic control centrality describes relaying functions as observed in signalling cascades (e.g. src kinase or Jak/Stat pathways). Value control centrality measures the direct influence of the value of the node on the network (e.g. Indian hedgehog as an essential regulator of proliferation in chondrocytes). Surveying random scale-free networks and biological networks, we find that control of the network resides in few high degree driver nodes and networks can be controlled best if they are sparsely connected.
Collapse
Affiliation(s)
- Stefan Karl
- Department of Bioinformatics, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
13
|
Rodriguez A, Crespo I, Fournier A, del Sol A. Discrete Logic Modelling Optimization to Contextualize Prior Knowledge Networks Using PRUNET. PLoS One 2015; 10:e0127216. [PMID: 26058016 PMCID: PMC4461287 DOI: 10.1371/journal.pone.0127216] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 04/13/2015] [Indexed: 01/09/2023] Open
Abstract
High-throughput technologies have led to the generation of an increasing amount of data in different areas of biology. Datasets capturing the cell's response to its intra- and extra-cellular microenvironment allows such data to be incorporated as signed and directed graphs or influence networks. These prior knowledge networks (PKNs) represent our current knowledge of the causality of cellular signal transduction. New signalling data is often examined and interpreted in conjunction with PKNs. However, different biological contexts, such as cell type or disease states, may have distinct variants of signalling pathways, resulting in the misinterpretation of new data. The identification of inconsistencies between measured data and signalling topologies, as well as the training of PKNs using context specific datasets (PKN contextualization), are necessary conditions to construct reliable, predictive models, which are current challenges in the systems biology of cell signalling. Here we present PRUNET, a user-friendly software tool designed to address the contextualization of a PKNs to specific experimental conditions. As the input, the algorithm takes a PKN and the expression profile of two given stable steady states or cellular phenotypes. The PKN is iteratively pruned using an evolutionary algorithm to perform an optimization process. This optimization rests in a match between predicted attractors in a discrete logic model (Boolean) and a Booleanized representation of the phenotypes, within a population of alternative subnetworks that evolves iteratively. We validated the algorithm applying PRUNET to four biological examples and using the resulting contextualized networks to predict missing expression values and to simulate well-characterized perturbations. PRUNET constitutes a tool for the automatic curation of a PKN to make it suitable for describing biological processes under particular experimental conditions. The general applicability of the implemented algorithm makes PRUNET suitable for a variety of biological processes, for instance cellular reprogramming or transitions between healthy and disease states.
Collapse
Affiliation(s)
- Ana Rodriguez
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg
| | - Isaac Crespo
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg
| | - Anna Fournier
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg
| | - Antonio del Sol
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg
| |
Collapse
|
14
|
Albert R, Thakar J. Boolean modeling: a logic-based dynamic approach for understanding signaling and regulatory networks and for making useful predictions. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2015; 6:353-69. [PMID: 25269159 DOI: 10.1002/wsbm.1273] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The biomolecules inside or near cells form a complex interacting system. Cellular phenotypes and behaviors arise from the totality of interactions among the components of this system. A fruitful way of modeling interacting biomolecular systems is by network-based dynamic models that characterize each component by a state variable, and describe the change in the state variables due to the interactions in the system. Dynamic models can capture the stable state patterns of this interacting system and can connect them to different cell fates or behaviors. A Boolean or logic model characterizes each biomolecule by a binary state variable that relates the abundance of that molecule to a threshold abundance necessary for downstream processes. The regulation of this state variable is described in a parameter free manner, making Boolean modeling a practical choice for systems whose kinetic parameters have not been determined. Boolean models integrate the body of knowledge regarding the components and interactions of biomolecular systems, and capture the system's dynamic repertoire, for example the existence of multiple cell fates. These models were used for a variety of systems and led to important insights and predictions. Boolean models serve as an efficient exploratory model, a guide for follow-up experiments, and as a foundation for more quantitative models.
Collapse
|
15
|
Cappuccio A, Tieri P, Castiglione F. Multiscale modelling in immunology: a review. Brief Bioinform 2015; 17:408-18. [PMID: 25810307 DOI: 10.1093/bib/bbv012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/30/2015] [Indexed: 01/26/2023] Open
Abstract
One of the greatest challenges in biomedicine is to get a unified view of observations made from the molecular up to the organism scale. Towards this goal, multiscale models have been highly instrumental in contexts such as the cardiovascular field, angiogenesis, neurosciences and tumour biology. More recently, such models are becoming an increasingly important resource to address immunological questions as well. Systematic mining of the literature in multiscale modelling led us to identify three main fields of immunological applications: host-virus interactions, inflammatory diseases and their treatment and development of multiscale simulation platforms for immunological research and for educational purposes. Here, we review the current developments in these directions, which illustrate that multiscale models can consistently integrate immunological data generated at several scales, and can be used to describe and optimize therapeutic treatments of complex immune diseases.
Collapse
Affiliation(s)
- Antonio Cappuccio
- Laboratory of Integrative biology of human dendritic cells and T cells, U932 Immunity and cancer, Institut Curie, 26 Rue d`Ulm, 75005 Paris, France
| | - Paolo Tieri
- Institute for Applied Mathematics (IAC), National Research Council of Italy (CNR), Via dei Taurini 19, 00185 Rome, Italy
| | - Filippo Castiglione
- Institute for Applied Mathematics (IAC), National Research Council of Italy (CNR), Via dei Taurini 19, 00185 Rome, Italy
| |
Collapse
|
16
|
Abou-Jaoudé W, Monteiro PT, Naldi A, Grandclaudon M, Soumelis V, Chaouiya C, Thieffry D. Model checking to assess T-helper cell plasticity. Front Bioeng Biotechnol 2015; 2:86. [PMID: 25674559 PMCID: PMC4309205 DOI: 10.3389/fbioe.2014.00086] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 12/20/2014] [Indexed: 12/03/2022] Open
Abstract
Computational modeling constitutes a crucial step toward the functional understanding of complex cellular networks. In particular, logical modeling has proven suitable for the dynamical analysis of large signaling and transcriptional regulatory networks. In this context, signaling input components are generally meant to convey external stimuli, or environmental cues. In response to such external signals, cells acquire specific gene expression patterns modeled in terms of attractors (e.g., stable states). The capacity for cells to alter or reprogram their differentiated states upon changes in environmental conditions is referred to as cell plasticity. In this article, we present a multivalued logical framework along with computational methods recently developed to efficiently analyze large models. We mainly focus on a symbolic model checking approach to investigate switches between attractors subsequent to changes of input conditions. As a case study, we consider the cellular network regulating the differentiation of T-helper (Th) cells, which orchestrate many physiological and pathological immune responses. To account for novel cellular subtypes, we present an extended version of a published model of Th cell differentiation. We then use symbolic model checking to analyze reachability properties between Th subtypes upon changes of environmental cues. This allows for the construction of a synthetic view of Th cell plasticity in terms of a graph connecting subtypes with arcs labeled by input conditions. Finally, we explore novel strategies enabling specific Th cell polarizing or reprograming events.
Collapse
Affiliation(s)
- Wassim Abou-Jaoudé
- Institut de Biologie de l’Ecole Normale Supérieure, Paris, France
- UMR CNRS 8197, Paris, France
- INSERM U1024, Paris, France
- Laboratoire d’Informatique de l’Ecole Normale Supérieure, Paris, France
| | - Pedro T. Monteiro
- INESC-ID, Lisboa, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Aurélien Naldi
- Centre Intégratif de Génomique, Université de Lausanne, Lausanne, Switzerland
| | | | - Vassili Soumelis
- Laboratoire d’Immunologie Clinique, Institut Curie, Paris, France
- INSERM U932, Paris, France
| | | | - Denis Thieffry
- Institut de Biologie de l’Ecole Normale Supérieure, Paris, France
- UMR CNRS 8197, Paris, France
- INSERM U1024, Paris, France
| |
Collapse
|
17
|
Carbo A, Hontecillas R, Andrew T, Eden K, Mei Y, Hoops S, Bassaganya-Riera J. Computational modeling of heterogeneity and function of CD4+ T cells. Front Cell Dev Biol 2014; 2:31. [PMID: 25364738 PMCID: PMC4207042 DOI: 10.3389/fcell.2014.00031] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Accepted: 07/10/2014] [Indexed: 12/19/2022] Open
Abstract
The immune system is composed of many different cell types and hundreds of intersecting molecular pathways and signals. This large biological complexity requires coordination between distinct pro-inflammatory and regulatory cell subsets to respond to infection while maintaining tissue homeostasis. CD4+ T cells play a central role in orchestrating immune responses and in maintaining a balance between pro- and anti- inflammatory responses. This tight balance between regulatory and effector reactions depends on the ability of CD4+ T cells to modulate distinct pathways within large molecular networks, since dysregulated CD4+ T cell responses may result in chronic inflammatory and autoimmune diseases. The CD4+ T cell differentiation process comprises an intricate interplay between cytokines, their receptors, adaptor molecules, signaling cascades and transcription factors that help delineate cell fate and function. Computational modeling can help to describe, simulate, analyze, and predict some of the behaviors in this complicated differentiation network. This review provides a comprehensive overview of existing computational immunology methods as well as novel strategies used to model immune responses with a particular focus on CD4+ T cell differentiation.
Collapse
Affiliation(s)
- Adria Carbo
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA ; Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA
| | - Raquel Hontecillas
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA ; Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA
| | - Tricity Andrew
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA ; Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA
| | - Kristin Eden
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA ; Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA
| | - Yongguo Mei
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA ; Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA
| | - Stefan Hoops
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA
| | - Josep Bassaganya-Riera
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA ; Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech Blacksburg, VA, USA ; Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech Blacksburg, VA, USA
| |
Collapse
|
18
|
Castiglione F, Pappalardo F, Bianca C, Russo G, Motta S. Modeling biology spanning different scales: an open challenge. BIOMED RESEARCH INTERNATIONAL 2014; 2014:902545. [PMID: 25143952 PMCID: PMC4124842 DOI: 10.1155/2014/902545] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/25/2014] [Indexed: 02/03/2023]
Abstract
It is coming nowadays more clear that in order to obtain a unified description of the different mechanisms governing the behavior and causality relations among the various parts of a living system, the development of comprehensive computational and mathematical models at different space and time scales is required. This is one of the most formidable challenges of modern biology characterized by the availability of huge amount of high throughput measurements. In this paper we draw attention to the importance of multiscale modeling in the framework of studies of biological systems in general and of the immune system in particular.
Collapse
Affiliation(s)
- Filippo Castiglione
- Institute for Applied Mathematics, National Research Council of Italy, Rome, Italy
| | | | - Carlo Bianca
- Theoretical Physics of Condensed Matter, Sorbonne Universities, UPMC Univ Paris 6, 75252 Paris Cedex 05, France
- UMR 7600 LPTMC, CNRS, 75252 Paris Cedex 05, France
| | - Giulia Russo
- Department of Pharmaceutical Sciences, University of Catania, Catania, Italy
| | - Santo Motta
- Department of Mathematics and Computer Science, University of Catania, 95125 Catania, Italy
| |
Collapse
|
19
|
Differentiation of Th subsets inhibited by nonstructural proteins of respiratory syncytial virus is mediated by ubiquitination. PLoS One 2014; 9:e101469. [PMID: 24992002 PMCID: PMC4081659 DOI: 10.1371/journal.pone.0101469] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 06/05/2014] [Indexed: 12/02/2022] Open
Abstract
Human respiratory syncytial virus (RSV), a major cause of severe respiratory diseases, constitutes an important risk factor for the development of subsequent asthma. However, the mechanism underlying RSV-induced asthma is poorly understood. Viral non-structural proteins NS1 and NS2 are critically required for RSV virulence; they strongly suppress IFN-mediated innate immunity of the host cells. In order to understand the effects of NS1 and NS2 on differentiation of Th subsets, we constructed lentiviral vectors of NS1 or NS2 to infect 16 HBE and analyzed the expression of HLA-DR, CD80 and CD86 and differentiation of Th1, Th2 and Th17 by Flow Cytometric Analysis and real-time PCR. The results showed that NS1 inhibited expression of HLA-DR, CD80 and CD86 and differentiation of Th1, Th2 and Th17 lymphocytes, which could be reversed by deleting elongin C binding domain. NS2 inhibited the differentiation of Th2 and Th17, which was reversed by proteasome inhibitors of PS-341. Our results indicated that NS1 inhibited the differentiation of T lymphocytes through its mono-ubiquitination to interacted proteins, while NS2 inhibited differentiation of Th2 and Th17 through ubiquitin-proteasome pathway, which may be related with the susceptibility to asthma after RSV infection.
Collapse
|
20
|
Li WZ, Wang J, Long R, Su GH, Bukhory DK, Dai J, Jin N, Huang SY, Jia P, Li T, Fan C, Liu K, Wang Z. Novel antibody against a glutamic acid-rich human fibrinogen-like protein 2-derived peptide near Ser91 inhibits hfgl2 prothrombinase activity. PLoS One 2014; 9:e94551. [PMID: 24728278 PMCID: PMC3984148 DOI: 10.1371/journal.pone.0094551] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 03/17/2014] [Indexed: 12/11/2022] Open
Abstract
Fibrinogen-like protein 2 (fgl2) is highly expressed in microvascular endothelial cells in diseases associated with microcirculatory disturbances and plays a crucial role in microthrombosis. Previous studies have demonstrated that the Ser89 residue is a critical site for mouse fgl2 prothrombinase activity. The aim of this study was to investigate the prothrombinase inhibitory ability of antibodies against an hfgl2-derived peptide. The peptide was termed NPG-12 because it is located at the N-terminus of membrane-bound hfgl2, contains 12 amino acid residues (corresponding to residues 76 to 87), and is rich in Glu. This peptide was selected as an antigenic determinant to produce antibodies in immunized rabbits using the DNAStar and HomoloGene software program. Abundant hfgl2 expression was induced in human umbilical vein endothelial cells through treatment with TNF-α. The generated anti-NPG-12 antibodies specifically recognize fgl2, as determined by ELISA, Western Blot and immunostaining. Moreover, one-stage clotting and thrombin generation tests provide evidence that the antibodies can reduce the hfgl2 prothrombinase activity without affecting the platelet-poor plasma prothrombin time (PT) or the activated partial thromboplastin time (APTT). In addition, the antibodies exerted undetectable influence on the proliferation or activation of bulk T cell populations. In conclusion, the selected peptide sequence NPG-12 may be a critical domain for hfgl2 prothrombinase activity, and the development of inhibitors against this sequence may be promising for research or management of hfgl2-associated microcirculatory disturbances.
Collapse
Affiliation(s)
- Wen-Zhu Li
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jue Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Long
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guan-Hua Su
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dinesh-Kumar Bukhory
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Dai
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nan Jin
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi-Yuan Huang
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Jia
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Li
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Fan
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Liu
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaohui Wang
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Mbodj A, Junion G, Brun C, Furlong EEM, Thieffry D. Logical modelling of Drosophila signalling pathways. MOLECULAR BIOSYSTEMS 2014; 9:2248-58. [PMID: 23868318 DOI: 10.1039/c3mb70187e] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A limited number of signalling pathways are involved in the specification of cell fate during the development of all animals. Several of these pathways were originally identified in Drosophila. To clarify their roles, and possible cross-talk, we have built a logical model for the nine key signalling pathways recurrently used in metazoan development. In each case, we considered the associated ligands, receptors, signal transducers, modulators, and transcription factors reported in the literature. Implemented using the logical modelling software GINsim, the resulting models qualitatively recapitulate the main characteristics of each pathway, in wild type as well as in various mutant situations (e.g. loss-of-function or gain-of-function). These models constitute pluggable modules that can be used to assemble comprehensive models of complex developmental processes. Moreover, these models of Drosophila pathways could serve as scaffolds for more complicated models of orthologous mammalian pathways. Comprehensive model annotations and GINsim files are provided for each of the nine considered pathways.
Collapse
Affiliation(s)
- Abibatou Mbodj
- Technological Advances for Genomics and Clinics (TAGC), INSERM UMR_S 1090, Aix-Marseille Université, Marseille, France.
| | | | | | | | | |
Collapse
|
22
|
Teku GN, Ortutay C, Vihinen M. Identification of core T cell network based on immunome interactome. BMC SYSTEMS BIOLOGY 2014; 8:17. [PMID: 24528953 PMCID: PMC3937033 DOI: 10.1186/1752-0509-8-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 02/05/2014] [Indexed: 12/03/2022]
Abstract
Background Data-driven studies on the dynamics of reconstructed protein-protein interaction (PPI) networks facilitate investigation and identification of proteins important for particular processes or diseases and reduces time and costs of experimental verification. Modeling the dynamics of very large PPI networks is computationally costly. Results To circumvent this problem, we created a link-weighted human immunome interactome and performed filtering. We reconstructed the immunome interactome and weighed the links using jackknife gene expression correlation of integrated, time course gene expression data. Statistical significance of the links was computed using the Global Statistical Significance (GloSS) filtering algorithm. P-values from GloSS were computed for the integrated, time course gene expression data. We filtered the immunome interactome to identify core components of the T cell PPI network (TPPIN). The interconnectedness of the major pathways for T cell survival and response, including the T cell receptor, MAPK and JAK-STAT pathways, are maintained in the TPPIN network. The obtained TPPIN network is supported both by Gene Ontology term enrichment analysis along with study of essential genes enrichment. Conclusions By integrating gene expression data to the immunome interactome and using a weighted network filtering method, we identified the T cell PPI immune response network. This network reveals the most central and crucial network in T cells. The approach is general and applicable to any dataset that contains sufficient information.
Collapse
Affiliation(s)
| | | | - Mauno Vihinen
- Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
23
|
Calçada D, Vianello D, Giampieri E, Sala C, Castellani G, de Graaf A, Kremer B, van Ommen B, Feskens E, Santoro A, Franceschi C, Bouwman J. The role of low-grade inflammation and metabolic flexibility in aging and nutritional modulation thereof: a systems biology approach. Mech Ageing Dev 2014; 136-137:138-47. [PMID: 24462698 DOI: 10.1016/j.mad.2014.01.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 01/10/2014] [Accepted: 01/13/2014] [Indexed: 02/07/2023]
Abstract
Aging is a biological process characterized by the progressive functional decline of many interrelated physiological systems. In particular, aging is associated with the development of a systemic state of low-grade chronic inflammation (inflammaging), and with progressive deterioration of metabolic function. Systems biology has helped in identifying the mediators and pathways involved in these phenomena, mainly through the application of high-throughput screening methods, valued for their molecular comprehensiveness. Nevertheless, inflammation and metabolic regulation are dynamical processes whose behavior must be understood at multiple levels of biological organization (molecular, cellular, organ, and system levels) and on multiple time scales. Mathematical modeling of such behavior, with incorporation of mechanistic knowledge on interactions between inflammatory and metabolic mediators, may help in devising nutritional interventions capable of preventing, or ameliorating, the age-associated functional decline of the corresponding systems.
Collapse
Affiliation(s)
- Dulce Calçada
- TNO, Microbiology and Systems Biology Group, Utrechtseweg 48, 3704 HE Zeist, The Netherlands; Wageningen University, Department of Human Nutrition, Wageningen, The Netherlands
| | - Dario Vianello
- University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine, Via San Giacomo 12, 40126 Bologna, Italy
| | - Enrico Giampieri
- University of Bologna, Department of Physics and Astronomy, 40127 Bologna, Italy
| | - Claudia Sala
- University of Bologna, Department of Physics and Astronomy, 40127 Bologna, Italy
| | - Gastone Castellani
- University of Bologna, Department of Physics and Astronomy, 40127 Bologna, Italy
| | - Albert de Graaf
- TNO, Microbiology and Systems Biology Group, Utrechtseweg 48, 3704 HE Zeist, The Netherlands
| | - Bas Kremer
- TNO, Microbiology and Systems Biology Group, Utrechtseweg 48, 3704 HE Zeist, The Netherlands
| | - Ben van Ommen
- TNO, Microbiology and Systems Biology Group, Utrechtseweg 48, 3704 HE Zeist, The Netherlands
| | - Edith Feskens
- Wageningen University, Department of Human Nutrition, Wageningen, The Netherlands
| | - Aurelia Santoro
- University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine, Via San Giacomo 12, 40126 Bologna, Italy
| | - Claudio Franceschi
- University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine, Via San Giacomo 12, 40126 Bologna, Italy; University of Bologna, Interdepartmental Centre "L. Galvani" (CIG), 40126 Bologna, Italy
| | - Jildau Bouwman
- TNO, Microbiology and Systems Biology Group, Utrechtseweg 48, 3704 HE Zeist, The Netherlands.
| |
Collapse
|
24
|
Tieri P, Prana V, Colombo T, Santoni D, Castiglione F. Multi-scale Simulation of T Helper Lymphocyte Differentiation. ADVANCES IN BIOINFORMATICS AND COMPUTATIONAL BIOLOGY 2014. [DOI: 10.1007/978-3-319-12418-6_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
25
|
Castiglione F, Tieri P, De Graaf A, Franceschi C, Liò P, Van Ommen B, Mazzà C, Tuchel A, Bernaschi M, Samson C, Colombo T, Castellani GC, Capri M, Garagnani P, Salvioli S, Nguyen VA, Bobeldijk-Pastorova I, Krishnan S, Cappozzo A, Sacchetti M, Morettini M, Ernst M. The onset of type 2 diabetes: proposal for a multi-scale model. JMIR Res Protoc 2013; 2:e44. [PMID: 24176906 PMCID: PMC3841357 DOI: 10.2196/resprot.2854] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 09/16/2013] [Indexed: 11/16/2022] Open
Abstract
Background Type 2 diabetes mellitus (T2D) is a common age-related disease, and is a major health concern, particularly in developed countries where the population is aging, including Europe. The multi-scale immune system simulator for the onset of type 2 diabetes (MISSION-T2D) is a European Union-funded project that aims to develop and validate an integrated, multilevel, and patient-specific model, incorporating genetic, metabolic, and nutritional data for the simulation and prediction of metabolic and inflammatory processes in the onset and progression of T2D. The project will ultimately provide a tool for diagnosis and clinical decision making that can estimate the risk of developing T2D and predict its progression in response to possible therapies.
Recent data showed that T2D and its complications, specifically in the heart, kidney, retina, and feet, should be considered a systemic disease that is sustained by a pervasive, metabolically-driven state of inflammation. Accordingly, there is an urgent need (1) to understand the complex mechanisms underpinning the onset of this disease, and (2) to identify early patient-specific diagnostic parameters and related inflammatory indicators. Objective We aim to accomplish this mission by setting up a multi-scale model to study the systemic interactions of the biological mechanisms involved in response to a variety of nutritional and metabolic stimuli and stressors. Methods Specifically, we will be studying the biological mechanisms of immunological/inflammatory processes, energy intake/expenditure ratio, and cell cycle rate. The overall architecture of the model will exploit an already established immune system simulator as well as several discrete and continuous mathematical methods for modeling of the processes critically involved in the onset and progression of T2D. We aim to validate the predictions of our models using actual biological and clinical data. Results This study was initiated in March 2013 and is expected to be completed by February 2016. Conclusions MISSION-T2D aims to pave the way for translating validated multilevel immune-metabolic models into the clinical setting of T2D. This approach will eventually generate predictive biomarkers for this disease from the integration of clinical data with metabolic, nutritional, immune/inflammatory, genetic, and gut microbiota profiles. Eventually, it should prove possible to translate these into cost-effective and mobile-based diagnostic tools.
Collapse
Affiliation(s)
- Filippo Castiglione
- Consiglio Nazionale delle Ricerche, Istituto per le Applicazioni del Calcolo "Mauro Picone", Roma, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Martínez-Sosa P, Mendoza L. The regulatory network that controls the differentiation of T lymphocytes. Biosystems 2013; 113:96-103. [PMID: 23743337 DOI: 10.1016/j.biosystems.2013.05.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 02/13/2013] [Accepted: 05/21/2013] [Indexed: 12/22/2022]
Abstract
There is a vast amount of molecular information regarding the differentiation of T lymphocytes, in particular regarding in vitro experimental treatments that modify their differentiation process. This publicly available information was used to infer the regulatory network that controls the differentiation of T lymphocytes into CD4(+) and CD8(+) cells. Hereby we present a network that consists of 50 nodes and 97 regulatory interactions, representing the main signaling circuits established among molecules and molecular complexes regulating the differentiation of T cells. The network was converted into a continuous dynamical system in the form of a set of coupled ordinary differential equations, and its dynamical behavior was studied. With the aid of numerical methods, nine fixed point attractors were found for the dynamical system. These attractors correspond to the activation patterns observed experimentally for the following cell types: CD4(-)CD8(-), CD4(+)CD8(+), CD4(+) naive, Th1, Th2, Th17, Treg, CD8(+) naive, and CTL. Furthermore, the model is able to describe the differentiation process from the precursor CD4(-)CD8(-) to any of the effector types due to a specific series of extracellular signals.
Collapse
Affiliation(s)
- Pablo Martínez-Sosa
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apartado Postal 70228, Ciudad Universitaria, CP04510 México, D.F., Mexico
| | | |
Collapse
|
27
|
Bérenguier D, Chaouiya C, Monteiro PT, Naldi A, Remy E, Thieffry D, Tichit L. Dynamical modeling and analysis of large cellular regulatory networks. CHAOS (WOODBURY, N.Y.) 2013; 23:025114. [PMID: 23822512 DOI: 10.1063/1.4809783] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The dynamical analysis of large biological regulatory networks requires the development of scalable methods for mathematical modeling. Following the approach initially introduced by Thomas, we formalize the interactions between the components of a network in terms of discrete variables, functions, and parameters. Model simulations result in directed graphs, called state transition graphs. We are particularly interested in reachability properties and asymptotic behaviors, which correspond to terminal strongly connected components (or "attractors") in the state transition graph. A well-known problem is the exponential increase of the size of state transition graphs with the number of network components, in particular when using the biologically realistic asynchronous updating assumption. To address this problem, we have developed several complementary methods enabling the analysis of the behavior of large and complex logical models: (i) the definition of transition priority classes to simplify the dynamics; (ii) a model reduction method preserving essential dynamical properties, (iii) a novel algorithm to compact state transition graphs and directly generate compressed representations, emphasizing relevant transient and asymptotic dynamical properties. The power of an approach combining these different methods is demonstrated by applying them to a recent multilevel logical model for the network controlling CD4+ T helper cell response to antigen presentation and to a dozen cytokines. This model accounts for the differentiation of canonical Th1 and Th2 lymphocytes, as well as of inflammatory Th17 and regulatory T cells, along with many hybrid subtypes. All these methods have been implemented into the software GINsim, which enables the definition, the analysis, and the simulation of logical regulatory graphs.
Collapse
Affiliation(s)
- D Bérenguier
- Institut de Mathématiques de Luminy, Marseille, France
| | | | | | | | | | | | | |
Collapse
|
28
|
Liao C, Lu T. A minimal transcriptional controlling network of regulatory T cell development. J Phys Chem B 2013; 117:12995-3004. [PMID: 23642089 DOI: 10.1021/jp402306g] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Regulatory T cells (Treg) are a subpopulation of T cells that are central to immune homeostasis and develop under the control of a complex regulatory network consisting of FoxP3 and its partner factors. A central question about this network is how does it enable T cells to robustly specify and stably maintain their states despite intrinsic and environmental fluctuations. Inspired by recent experimental advances, we propose here a minimal transcriptional controlling network and use it to illustrate the robustness and dynamic features of Treg development. Our study shows that the controlling network may exhibit distinct dynamics depending on its parameter regimes and that the maintenance of multistability requires the orchestration of both its positive and negative feedback loops. In addition, system volume contributes monotonically to the increase in the network's robustness. We further show that the dynamics of our model varies with the alteration of FoxP3-DNA binding affinity, consistent with recent experimental findings. This minimal model thereby offers new insights into the dynamics and robustness of Treg development and may serve as a platform for future exploration toward a more quantitative and systematic understanding of the immune system.
Collapse
Affiliation(s)
- Chen Liao
- Department of Bioengineering and Institute for Genomic Biology, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | | |
Collapse
|
29
|
Mendoza L. A Virtual Culture of CD4+ T Lymphocytes. Bull Math Biol 2013; 75:1012-29. [DOI: 10.1007/s11538-013-9814-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 01/09/2013] [Indexed: 12/11/2022]
|
30
|
Saadatpour A, Albert R. Boolean modeling of biological regulatory networks: a methodology tutorial. Methods 2012; 62:3-12. [PMID: 23142247 DOI: 10.1016/j.ymeth.2012.10.012] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 10/31/2012] [Indexed: 12/14/2022] Open
Abstract
Given the complexity and interactive nature of biological systems, constructing informative and coherent network models of these systems and subsequently developing efficient approaches to analyze the assembled networks is of immense importance. The integration of network analysis and dynamic modeling enables one to investigate the behavior of the underlying system as a whole and to make experimentally testable predictions about less-understood aspects of the processes involved. In this paper, we present a tutorial on the fundamental steps of Boolean modeling of biological regulatory networks. We demonstrate how to infer a Boolean network model from the available experimental data, analyze the network using graph-theoretical measures, and convert it into a predictive dynamic model. For each step, the pitfalls one may encounter and possible ways to circumvent them are also discussed. We illustrate these steps on a toy network as well as in the context of the Drosophila melanogaster segment polarity gene network.
Collapse
Affiliation(s)
- Assieh Saadatpour
- Department of Mathematics, The Pennsylvania State University, University Park, PA 16802, USA
| | | |
Collapse
|
31
|
Weinstein N, Mendoza L. Building Qualitative Models of Plant Regulatory Networks with SQUAD. FRONTIERS IN PLANT SCIENCE 2012; 3:72. [PMID: 22639661 PMCID: PMC3355663 DOI: 10.3389/fpls.2012.00072] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 03/28/2012] [Indexed: 05/24/2023]
Affiliation(s)
- Nathan Weinstein
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de MéxicoDistrito Federal, México
| | - Luis Mendoza
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de MéxicoDistrito Federal, México
| |
Collapse
|
32
|
Rodríguez A, Sosa D, Torres L, Molina B, Frías S, Mendoza L. A Boolean network model of the FA/BRCA pathway. ACTA ACUST UNITED AC 2012; 28:858-66. [PMID: 22267503 DOI: 10.1093/bioinformatics/bts036] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
MOTIVATION Fanconi anemia (FA) is a chromosomal instability syndrome originated by inherited mutations that impair the Fanconi Anemia/Breast Cancer (FA/BRCA) pathway, which is committed to the repair of DNA interstrand cross-links (ICLs). The disease displays increased spontaneous chromosomal aberrations and hypersensitivity to agents that create DNA interstrand cross-links. In spite of DNA damage, FA/BRCA-deficient cells are able to progress throughout the cell cycle, probably due to the activity of alternative DNA repair pathways, or due to defects in the checkpoints that monitor DNA integrity. RESULTS We propose a Boolean network model of the FA/BRCA pathway, Checkpoint proteins and some alternative DNA repair pathways. To our knowledge, this is the largest network model incorporating a DNA repair pathway. Our model is able to simulate the ICL repair process mediated by the FA/BRCA pathway, the activation of Checkpoint proteins observed by recurrent DNA damage, as well as the repair of DNA double-strand breaks and DNA adducts. We generated a series of simulations for mutants, some of which have never been reported and thus constitute predictions about the function of the FA/BRCA pathway. Finally, our model suggests alternative DNA repair pathways that become active whenever the FA/BRCA pathway is defective.
Collapse
Affiliation(s)
- Alfredo Rodríguez
- Departamento de Investigación en Genética Humana, Instituto Nacional de Pediatría, Posgrado en Ciencias Biológicas, México
| | | | | | | | | | | |
Collapse
|
33
|
Sankar M, Osmont KS, Rolcik J, Gujas B, Tarkowska D, Strnad M, Xenarios I, Hardtke CS. A qualitative continuous model of cellular auxin and brassinosteroid signaling and their crosstalk. ACTA ACUST UNITED AC 2011; 27:1404-12. [PMID: 21450717 DOI: 10.1093/bioinformatics/btr158] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
MOTIVATION Hormone pathway interactions are crucial in shaping plant development, such as synergism between the auxin and brassinosteroid pathways in cell elongation. Both hormone pathways have been characterized in detail, revealing several feedback loops. The complexity of this network, combined with a shortage of kinetic data, renders its quantitative analysis virtually impossible at present. RESULTS As a first step towards overcoming these obstacles, we analyzed the network using a Boolean logic approach to build models of auxin and brassinosteroid signaling, and their interaction. To compare these discrete dynamic models across conditions, we transformed them into qualitative continuous systems, which predict network component states more accurately and can accommodate kinetic data as they become available. To this end, we developed an extension for the SQUAD software, allowing semi-quantitative analysis of network states. Contrasting the developmental output depending on cell type-specific modulators enabled us to identify a most parsimonious model, which explains initially paradoxical mutant phenotypes and revealed a novel physiological feature. AVAILABILITY The package SQUADD is freely available via the Bioconductor repository at http://www.bioconductor.org/help/bioc-views/release/bioc/html/SQUADD.html.
Collapse
Affiliation(s)
- Martial Sankar
- Department of Plant Molecular Biology, University of Lausanne, Lausanne, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Demongeot J, Elena A, Noual M, Sené S, Thuderoz F. "Immunetworks", intersecting circuits and dynamics. J Theor Biol 2011; 280:19-33. [PMID: 21439971 DOI: 10.1016/j.jtbi.2011.03.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 03/17/2011] [Accepted: 03/17/2011] [Indexed: 12/21/2022]
Abstract
This paper proposes a study of biological regulation networks based on a multi-level strategy. Given a network, the first structural level of this strategy consists in analysing the architecture of the network interactions in order to describe it. The second dynamical level consists in relating the patterns found in the architecture to the possible dynamical behaviours of the network. It is known that circuits are the patterns that play the most important part in the dynamics of a network in the sense that they are responsible for the diversity of its asymptotic behaviours. Here, we pursue further this idea and argue that beyond the influence of underlying circuits, intersections of circuits also impact significantly on the dynamics of a network and thus need to be payed special attention to. For some genetic regulation networks involved in the control of the immune system ("immunetworks"), we show that the small number of attractors can be explained by the presence, in the underlying structures of these networks, of intersecting circuits that "inter-lock".
Collapse
Affiliation(s)
- Jacques Demongeot
- Université Joseph Fourier de Grenoble, AGIM, CNRS FRE 3405, 38700 La Tronche, France
| | | | | | | | | |
Collapse
|