1
|
Li M, Lulla AR, Wang Y, Tsavaschidis S, Wang F, Karakas C, Nguyen TD, Bui TN, Pina MA, Chen MK, Mastoraki S, Multani AS, Fowlkes NW, Sahin A, Marshall CG, Hunt KK, Keyomarsi K. Low-Molecular Weight Cyclin E Confers a Vulnerability to PKMYT1 Inhibition in Triple-Negative Breast Cancer. Cancer Res 2024; 84:3864-3880. [PMID: 39186665 PMCID: PMC11567801 DOI: 10.1158/0008-5472.can-23-4130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 06/11/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
Cyclin E is a regulatory subunit of CDK2 that mediates S phase entry and progression. The cleavage of full-length cyclin E (FL-cycE) to low-molecular weight isoforms (LMW-E) dramatically alters substrate specificity, promoting G1-S cell cycle transition and accelerating mitotic exit. Approximately 70% of triple-negative breast cancers (TNBC) express LMW-E, which correlates with poor prognosis. PKMYT1 also plays an important role in mitosis by inhibiting CDK1 to block premature mitotic entry, suggesting it could be a therapeutic target in TNBC expressing LMW-E. In this study, analysis of tumor samples of patients with TNBC revealed that coexpression of LMW-E and PKMYT1-catalyzed CDK1 phosphorylation predicted poor response to neoadjuvant chemotherapy. Compared with FL-cycE, LMW-E specifically upregulates PKMYT1 expression and protein stability, thereby increasing CDK1 phosphorylation. Inhibiting PKMYT1 with the selective inhibitor RP-6306 (lunresertib) elicited LMW-E-dependent antitumor effects, accelerating premature mitotic entry, inhibiting replication fork restart, and enhancing DNA damage, chromosomal breakage, apoptosis, and replication stress. Importantly, TNBC cell line xenografts expressing LMW-E showed greater sensitivity to RP-6306 than tumors with empty vector or FL-cycE. Furthermore, RP-6306 exerted tumor suppressive effects in LMW-E transgenic murine mammary tumors and patient-derived xenografts of LMW-E-high TNBC but not in the LMW-E null models examined in parallel. Lastly, transcriptomic and immune profiling demonstrated that RP-6306 treatment induced interferon responses and T-cell infiltration in the LMW-E-high tumor microenvironment, enhancing the antitumor immune response. These findings highlight the LMW-E/PKMYT1/CDK1 regulatory axis as a promising therapeutic target in TNBC, providing the rationale for further clinical development of PKMYT1 inhibitors in this aggressive breast cancer subtype. Significance: PKMYT1 upregulation and CDK1 phosphorylation in triple-negative breast cancer expressing low-molecular weight cyclin E leads to suboptimal responses to chemotherapy but sensitizes tumors to PKMYT1 inhibitors, proposing a personalized treatment strategy.
Collapse
Affiliation(s)
- Mi Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amriti R. Lulla
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yan Wang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Fuchenchu Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cansu Karakas
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tuyen D.T. Nguyen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tuyen N. Bui
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marc A. Pina
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mei-Kuang Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sofia Mastoraki
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Asha S. Multani
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Natalie W. Fowlkes
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Aysegul Sahin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Kelly K. Hunt
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Khandan Keyomarsi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
2
|
Gottumukkala SB, Ganesan TS, Palanisamy A. Comprehensive molecular interaction map of TGFβ induced epithelial to mesenchymal transition in breast cancer. NPJ Syst Biol Appl 2024; 10:53. [PMID: 38760412 PMCID: PMC11101644 DOI: 10.1038/s41540-024-00378-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/29/2024] [Indexed: 05/19/2024] Open
Abstract
Breast cancer is one of the prevailing cancers globally, with a high mortality rate. Metastatic breast cancer (MBC) is an advanced stage of cancer, characterised by a highly nonlinear, heterogeneous process involving numerous singling pathways and regulatory interactions. Epithelial-mesenchymal transition (EMT) emerges as a key mechanism exploited by cancer cells. Transforming Growth Factor-β (TGFβ)-dependent signalling is attributed to promote EMT in advanced stages of breast cancer. A comprehensive regulatory map of TGFβ induced EMT was developed through an extensive literature survey. The network assembled comprises of 312 distinct species (proteins, genes, RNAs, complexes), and 426 reactions (state transitions, nuclear translocations, complex associations, and dissociations). The map was developed by following Systems Biology Graphical Notation (SBGN) using Cell Designer and made publicly available using MINERVA ( http://35.174.227.105:8080/minerva/?id=Metastatic_Breast_Cancer_1 ). While the complete molecular mechanism of MBC is still not known, the map captures the elaborate signalling interplay of TGFβ induced EMT-promoting MBC. Subsequently, the disease map assembled was translated into a Boolean model utilising CaSQ and analysed using Cell Collective. Simulations of these have captured the known experimental outcomes of TGFβ induced EMT in MBC. Hub regulators of the assembled map were identified, and their transcriptome-based analysis confirmed their role in cancer metastasis. Elaborate analysis of this map may help in gaining additional insights into the development and progression of metastatic breast cancer.
Collapse
Affiliation(s)
| | - Trivadi Sundaram Ganesan
- Department of Medical Oncology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Anbumathi Palanisamy
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, India.
| |
Collapse
|
3
|
Yip HYK, Shin SY, Chee A, Ang CS, Rossello FJ, Wong LH, Nguyen LK, Papa A. Integrative modeling uncovers p21-driven drug resistance and prioritizes therapies for PIK3CA-mutant breast cancer. NPJ Precis Oncol 2024; 8:20. [PMID: 38273040 PMCID: PMC10810864 DOI: 10.1038/s41698-024-00496-y] [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: 09/09/2022] [Accepted: 12/21/2023] [Indexed: 01/27/2024] Open
Abstract
Utility of PI3Kα inhibitors like BYL719 is limited by the acquisition of genetic and non-genetic mechanisms of resistance which cause disease recurrence. Several combination therapies based on PI3K inhibition have been proposed, but a way to systematically prioritize them for breast cancer treatment is still missing. By integrating published and in-house studies, we have developed in silico models that quantitatively capture dynamics of PI3K signaling at the network-level under a BYL719-sensitive versus BYL719 resistant-cell state. Computational predictions show that signal rewiring to alternative components of the PI3K pathway promote resistance to BYL719 and identify PDK1 as the most effective co-target with PI3Kα rescuing sensitivity of resistant cells to BYL719. To explore whether PI3K pathway-independent mechanisms further contribute to BYL719 resistance, we performed phosphoproteomics and found that selection of high levels of the cell cycle regulator p21 unexpectedly promoted drug resistance in T47D cells. Functionally, high p21 levels favored repair of BYL719-induced DNA damage and bypass of the associated cellular senescence. Importantly, targeted inhibition of the check-point inhibitor CHK1 with MK-8776 effectively caused death of p21-high T47D cells, thus establishing a new vulnerability of BYL719-resistant breast cancer cells. Together, our integrated studies uncover hidden molecular mediators causing resistance to PI3Kα inhibition and provide a framework to prioritize combination therapies for PI3K-mutant breast cancer.
Collapse
Affiliation(s)
- Hon Yan Kelvin Yip
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Sung-Young Shin
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Annabel Chee
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Ching-Seng Ang
- Bio21 Mass Spectrometry and Proteomics Facility, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Fernando J Rossello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, 3052, Australia
- Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC, 3052, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Lee Hwa Wong
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Lan K Nguyen
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia.
| | - Antonella Papa
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
4
|
Gondal MN, Chaudhary SU. Navigating Multi-Scale Cancer Systems Biology Towards Model-Driven Clinical Oncology and Its Applications in Personalized Therapeutics. Front Oncol 2021; 11:712505. [PMID: 34900668 PMCID: PMC8652070 DOI: 10.3389/fonc.2021.712505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/26/2021] [Indexed: 12/19/2022] Open
Abstract
Rapid advancements in high-throughput omics technologies and experimental protocols have led to the generation of vast amounts of scale-specific biomolecular data on cancer that now populates several online databases and resources. Cancer systems biology models built using this data have the potential to provide specific insights into complex multifactorial aberrations underpinning tumor initiation, development, and metastasis. Furthermore, the annotation of these single- and multi-scale models with patient data can additionally assist in designing personalized therapeutic interventions as well as aid in clinical decision-making. Here, we have systematically reviewed the emergence and evolution of (i) repositories with scale-specific and multi-scale biomolecular cancer data, (ii) systems biology models developed using this data, (iii) associated simulation software for the development of personalized cancer therapeutics, and (iv) translational attempts to pipeline multi-scale panomics data for data-driven in silico clinical oncology. The review concludes that the absence of a generic, zero-code, panomics-based multi-scale modeling pipeline and associated software framework, impedes the development and seamless deployment of personalized in silico multi-scale models in clinical settings.
Collapse
Affiliation(s)
- Mahnoor Naseer Gondal
- Biomedical Informatics Research Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
| | - Safee Ullah Chaudhary
- Biomedical Informatics Research Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
5
|
Rougny A, Paulevé L, Teboul M, Delaunay F. A detailed map of coupled circadian clock and cell cycle with qualitative dynamics validation. BMC Bioinformatics 2021; 22:240. [PMID: 33975535 PMCID: PMC8114686 DOI: 10.1186/s12859-021-04158-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/21/2021] [Indexed: 12/16/2022] Open
Abstract
Background The temporal coordination of biological processes by the circadian clock is an important mechanism, and its disruption has negative health outcomes, including cancer. Experimental and theoretical evidence suggests that the oscillators driving the circadian clock and the cell cycle are coupled through phase locking. Results We present a detailed and documented map of known mechanisms related to the regulation of the circadian clock, and its coupling with an existing cell cycle map which includes main interactions of the mammalian cell cycle. The coherence of the merged map has been validated with a qualitative dynamics analysis. We verified that the coupled circadian clock and cell cycle maps reproduce the observed sequence of phase markers. Moreover, we predicted mutations that contribute to regulating checkpoints of the two oscillators. Conclusions Our approach underlined the potential key role of the core clock protein NR1D1 in regulating cell cycle progression. We predicted that its activity influences negatively the progression of the cell cycle from phase G2 to M. This is consistent with the earlier experimental finding that pharmacological activation of NR1D1 inhibits tumour cell proliferation and shows that our approach can identify biologically relevant species in the context of large and complex networks. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-021-04158-9.
Collapse
Affiliation(s)
- Adrien Rougny
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Aomi, Tokyo, Japan.,Computational Bio Big Data Open Innovation Laboratory (CBBD-OIL), AIST, Aomi, Tokyo, Japan
| | - Loïc Paulevé
- Bordeaux INP, CNRS, LaBRI, UMR5800, Univ. Bordeaux, 33400, Talence, France
| | | | | |
Collapse
|
6
|
Yousuf M, Shamsi A, Queen A, Shahbaaz M, Khan P, Hussain A, Alajmi MF, Rizwanul Haque QM, Imtaiyaz Hassan M. Targeting cyclin-dependent kinase 6 by vanillin inhibits proliferation of breast and lung cancer cells: Combined computational and biochemical studies. J Cell Biochem 2021; 122:897-910. [PMID: 33829554 DOI: 10.1002/jcb.29921] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022]
Abstract
Cyclin-dependent kinase 6 (CDK6) is a member of serine/threonine kinase family, and its overexpression is associated with cancer development. Thus, it is considered as a potential drug target for anticancer therapies. This study showed the CDK6 inhibitory potential of vanillin using combined experimental and computational methods. Structure-based docking and 200 ns molecular dynamics simulation studies revealed that the binding of vanillin stabilizes the CDK6 structure and provides mechanistic insights into the binding mechanism. Enzyme inhibition and fluorescence-binding studies showed that vanillin inhibits CDK6 with an half maximal inhibitory concentration = 4.99 μM and a binding constant (K) 4.1 × 107 M-1 . Isothermal titration calorimetry measurements further complemented our observations. Studies on human cancer cell lines (MCF-7 and A549) showed that vanillin decreases cell viability and colonization properties. The protein expression studies have further revealed that vanillin reduces the CDK6 expression and induces apoptosis in the cancer cells. In conclusion, our study presents the CDK6-mediated therapeutic implications of vanillin for anticancer therapies.
Collapse
Affiliation(s)
- Mohd Yousuf
- Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Anas Shamsi
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Aarfa Queen
- Department of Chemistry, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Mohd Shahbaaz
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, Cape Town, South Africa.,Laboratory of Computational Modeling of Drugs, South Ural State University, Chelyabinsk, Russia
| | - Parvez Khan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Afzal Hussain
- Department of Pharmacognosy, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed F Alajmi
- Department of Pharmacognosy, King Saud University, Riyadh, Saudi Arabia
| | | | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| |
Collapse
|
7
|
Agapito G, Pastrello C, Jurisica I. Comprehensive pathway enrichment analysis workflows: COVID-19 case study. Brief Bioinform 2020. [PMCID: PMC7799312 DOI: 10.1093/bib/bbaa377] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) outbreak due to the novel coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been classified as a pandemic disease by the World Health Organization on the 12th March 2020. This world-wide crisis created an urgent need to identify effective countermeasures against SARS-CoV-2. In silico methods, artificial intelligence and bioinformatics analysis pipelines provide effective and useful infrastructure for comprehensive interrogation and interpretation of available data, helping to find biomarkers, explainable models and eventually cures. One class of such tools, pathway enrichment analysis (PEA) methods, helps researchers to find possible key targets present in biological pathways of host cells that are targeted by SARS-CoV-2. Since many software tools are available, it is not easy for non-computational users to choose the best one for their needs. In this paper, we highlight how to choose the most suitable PEA method based on the type of COVID-19 data to analyze. We aim to provide a comprehensive overview of PEA techniques and the tools that implement them.
Collapse
Affiliation(s)
| | - Chiara Pastrello
- Krembil Research Institute, University Health Network, Toronto, Canada
| | - Igor Jurisica
- Departments of Medical Biophysics and Computer Science, University of Toronto, Canada
| |
Collapse
|
8
|
Yousuf M, Khan P, Shamsi A, Shahbaaz M, Hasan GM, Haque QMR, Christoffels A, Islam A, Hassan MI. Inhibiting CDK6 Activity by Quercetin Is an Attractive Strategy for Cancer Therapy. ACS OMEGA 2020; 5:27480-27491. [PMID: 33134711 PMCID: PMC7594119 DOI: 10.1021/acsomega.0c03975] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 09/30/2020] [Indexed: 06/11/2023]
Abstract
Cyclin-dependent kinase 6 (CDK6) is a potential drug target that plays an important role in the progression of different types of cancers. We performed in silico and in vitro screening of different natural compounds and found that quercetin has a high binding affinity for the CDK6 and inhibits its activity with an IC50 = 5.89 μM. Molecular docking and a 200 ns whole atom simulation of the CDK6-quercetin complex provide insights into the binding mechanism and stability of the complex. Binding parameters ascertained by fluorescence and isothermal titration calorimetry studies revealed a binding constant in the range of 107 M-1 of quercetin to the CDK6. Thermodynamic parameters associated with the formation of the CDK6-quercetin complex suggested an electrostatic interaction-driven process. The cell-based protein expression studies in the breast (MCF-7) and lung (A549) cancer cells revealed that the treatment of quercetin decreases the expression of CDK6. Quercetin also decreases the viability and colony formation potential of selected cancer cells. Moreover, quercetin induces apoptosis, by decreasing the production of reactive oxygen species and CDK6 expression. Both in silico and in vitro studies highlight the significance of quercetin for the development of anticancer leads in terms of CDK6 inhibitors.
Collapse
Affiliation(s)
- Mohd Yousuf
- Department
of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Parvez Khan
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Anas Shamsi
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohd Shahbaaz
- South
African Medical Research Council Bioinformatics Unit, South African
National Bioinformatics Institute, University
of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
- Laboratory
of Computational Modeling of Drugs, South
Ural State University, 76 Lenin Prospekt, Chelyabinsk 454080, Russia
| | - Gulam Mustafa Hasan
- Department
of Biochemistry, College of Medicine, Prince
Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | - Alan Christoffels
- South
African Medical Research Council Bioinformatics Unit, South African
National Bioinformatics Institute, University
of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
| | - Asimul Islam
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Md. Imtaiyaz Hassan
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| |
Collapse
|
9
|
Serhan CN, Gupta SK, Perretti M, Godson C, Brennan E, Li Y, Soehnlein O, Shimizu T, Werz O, Chiurchiù V, Azzi A, Dubourdeau M, Gupta SS, Schopohl P, Hoch M, Gjorgevikj D, Khan FM, Brauer D, Tripathi A, Cesnulevicius K, Lescheid D, Schultz M, Särndahl E, Repsilber D, Kruse R, Sala A, Haeggström JZ, Levy BD, Filep JG, Wolkenhauer O. The Atlas of Inflammation Resolution (AIR). Mol Aspects Med 2020; 74:100894. [PMID: 32893032 PMCID: PMC7733955 DOI: 10.1016/j.mam.2020.100894] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Acute inflammation is a protective reaction by the immune system in response to invading pathogens or tissue damage. Ideally, the response should be localized, self-limited, and returning to homeostasis. If not resolved, acute inflammation can result in organ pathologies leading to chronic inflammatory phenotypes. Acute inflammation and inflammation resolution are complex coordinated processes, involving a number of cell types, interacting in space and time. The biomolecular complexity and the fact that several biomedical fields are involved, make a multi- and interdisciplinary approach necessary. The Atlas of Inflammation Resolution (AIR) is a web-based resource capturing an essential part of the state-of-the-art in acute inflammation and inflammation resolution research. The AIR provides an interface for users to search thousands of interactions, arranged in inter-connected multi-layers of process diagrams, covering a wide range of clinically relevant phenotypes. By mapping experimental data onto the AIR, it can be used to elucidate drug action as well as molecular mechanisms underlying different disease phenotypes. For the visualization and exploration of information, the AIR uses the Minerva platform, which is a well-established tool for the presentation of disease maps. The molecular details of the AIR are encoded using international standards. The AIR was created as a freely accessible resource, supporting research and education in the fields of acute inflammation and inflammation resolution. The AIR connects research communities, facilitates clinical decision making, and supports research scientists in the formulation and validation of hypotheses. The AIR is accessible through https://air.bio.informatik.uni-rostock.de.
Collapse
Affiliation(s)
- Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Shailendra K Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Mauro Perretti
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Catherine Godson
- Diabetes Complications Research Centre, Conway Institute & School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eoin Brennan
- Diabetes Complications Research Centre, Conway Institute & School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Yongsheng Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Oliver Soehnlein
- Department of Physiology and Pharmacology (FyFA), Karolinska Institutet, 17177, Stockholm, Sweden; German Center for Cardiovascular Research (DZHK), München, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University, 80336, München, Germany
| | - Takao Shimizu
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, 113-0033 Tokyo, Japan; National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, Japan
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743, Jena, Germany
| | - Valerio Chiurchiù
- Institute of Translational Pharmacology, National Research Council, 00133, Rome, Italy; Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Angelo Azzi
- School of Graduate Biomedical Pharmacology and Drug Development Program at Tufts University, Boston, MA 02111, USA
| | - Marc Dubourdeau
- Ambiotis, Canal Biotech 2 - 3 Rue des Satellites, 31400, Toulouse, France
| | - Suchi Smita Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Patrick Schopohl
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Matti Hoch
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Dragana Gjorgevikj
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Faiz M Khan
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - David Brauer
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Anurag Tripathi
- CSIR - Indian Institute of Toxicology Research, 226001, Lucknow, India
| | | | - David Lescheid
- Department of Medical Affairs & Research, Heel GmbH, 76532, Baden-Baden, Germany
| | - Myron Schultz
- Department of Medical Affairs & Research, Heel GmbH, 76532, Baden-Baden, Germany
| | - Eva Särndahl
- iRiSC - Inflammatory Response and Infection Susceptibility Centre, Faculty of Medicine and Health, Örebro University, SE-701 82, Örebro, Sweden; School of Medical Sciences, Örebro University, SE-701 82, Örebro, Sweden
| | - Dirk Repsilber
- School of Medical Sciences, Örebro University, SE-701 82, Örebro, Sweden
| | - Robert Kruse
- iRiSC - Inflammatory Response and Infection Susceptibility Centre, Faculty of Medicine and Health, Örebro University, SE-701 82, Örebro, Sweden; Department of Clinical Research Laboratory, Faculty of Medicine and Health, Örebro University, SE-701 82, Örebro, Sweden
| | - Angelo Sala
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milano, and IRIB, C.N.R, 90146, Palermo, Italy
| | - Jesper Z Haeggström
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, SE-171 77, Stockholm, Sweden
| | - Bruce D Levy
- Brigham and Women's Hospital, Department of Medicine, Pulmonary and Critical Care Medicine and Harvard Medical School, Boston, MA, 02115, USA
| | - János G Filep
- Department of Pathology and Cell Biology, University of Montreal, and Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, H1T 2M4, Canada
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany; Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Research Centre at Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
10
|
Serhan CN, Gupta SK, Perretti M, Godson C, Brennan E, Li Y, Soehnlein O, Shimizu T, Werz O, Chiurchiù V, Azzi A, Dubourdeau M, Gupta SS, Schopohl P, Hoch M, Gjorgevikj D, Khan FM, Brauer D, Tripathi A, Cesnulevicius K, Lescheid D, Schultz M, Särndahl E, Repsilber D, Kruse R, Sala A, Haeggström JZ, Levy BD, Filep JG, Wolkenhauer O. WITHDRAWN: The Atlas of Inflammation Resolution (AIR). Mol Aspects Med 2020:100893. [PMID: 32873427 DOI: 10.1016/j.mam.2020.100893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The Publisher regrets that this article is an accidental duplication of an article that has already been published, https://doi.org/10.1016/j.mam.2020.100894. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Shailendra K Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Mauro Perretti
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Catherine Godson
- Diabetes Complications Research Centre, Conway Institute & School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eoin Brennan
- Diabetes Complications Research Centre, Conway Institute & School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Yongsheng Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Oliver Soehnlein
- Department of Physiology and Pharmacology (FyFA), Karolinska Institutet, 17177, Stockholm, Sweden; German Center for Cardiovascular Research (DZHK), München, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University, 80336, München, Germany
| | - Takao Shimizu
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, 113-0033, Tokyo, Japan; National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, Japan
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743, Jena, Germany
| | - Valerio Chiurchiù
- Institute of Translational Pharmacology, National Research Council, 00133, Rome, Italy; Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Angelo Azzi
- School of Graduate Biomedical Pharmacology and Drug Development Program at Tufts University, Boston, MA, 02111, USA
| | - Marc Dubourdeau
- Ambiotis, Canal Biotech 2 - 3 Rue des Satellites, 31400, Toulouse, France
| | - Suchi Smita Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Patrick Schopohl
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Matti Hoch
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Dragana Gjorgevikj
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Faiz M Khan
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - David Brauer
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Anurag Tripathi
- CSIR - Indian Institute of Toxicology Research, 226001, Lucknow, India
| | | | - David Lescheid
- Department of Medical Affairs & Research, Heel GmbH, 76532, Baden-Baden, Germany
| | - Myron Schultz
- Department of Medical Affairs & Research, Heel GmbH, 76532, Baden-Baden, Germany
| | - Eva Särndahl
- IRiSC - Inflammatory Response and Infection Susceptibility Centre, Faculty of Medicine and Health, Örebro University, SE-701 82, Örebro, Sweden
| | - Dirk Repsilber
- School of Medical Sciences, University of Örebro, SE-701 82, Örebro, Sweden
| | - Robert Kruse
- IRiSC - Inflammatory Response and Infection Susceptibility Centre, Faculty of Medicine and Health, Örebro University, SE-701 82, Örebro, Sweden; Department of Clinical Research Laboratory, Faculty of Medicine and Health, Örebro University, SE-701 82, Örebro, Sweden
| | - Angelo Sala
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milano, and IRIB, C.N.R, 90146, Palermo, Italy
| | - Jesper Z Haeggström
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, SE-171 77, Stockholm, Sweden
| | - Bruce D Levy
- Brigham and Women's Hospital, Department of Medicine, Pulmonary and Critical Care Medicine and Harvard Medical School, Boston, MA, 02115, USA
| | - János G Filep
- Department of Pathology and Cell Biology, University of Montreal, Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, H1T 2M4, Canada
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany; Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Research Centre at Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
11
|
Ravel JM, Monraz Gomez LC, Sompairac N, Calzone L, Zhivotovsky B, Kroemer G, Barillot E, Zinovyev A, Kuperstein I. Comprehensive Map of the Regulated Cell Death Signaling Network: A Powerful Analytical Tool for Studying Diseases. Cancers (Basel) 2020; 12:E990. [PMID: 32316560 PMCID: PMC7226067 DOI: 10.3390/cancers12040990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 03/10/2020] [Indexed: 12/25/2022] Open
Abstract
The processes leading to, or avoiding cell death are widely studied, because of their frequent perturbation in various diseases. Cell death occurs in three highly interconnected steps: Initiation, signaling and execution. We used a systems biology approach to gather information about all known modes of regulated cell death (RCD). Based on the experimental data retrieved from literature by manual curation, we graphically depicted the biological processes involved in RCD in the form of a seamless comprehensive signaling network map. The molecular mechanisms of each RCD mode are represented in detail. The RCD network map is divided into 26 functional modules that can be visualized contextually in the whole seamless network, as well as in individual diagrams. The resource is freely available and accessible via several web platforms for map navigation, data integration, and analysis. The RCD network map was employed for interpreting the functional differences in cell death regulation between Alzheimer's disease and non-small cell lung cancer based on gene expression data that allowed emphasizing the molecular mechanisms underlying the inverse comorbidity between the two pathologies. In addition, the map was used for the analysis of genomic and transcriptomic data from ovarian cancer patients that provided RCD map-based signatures of four distinct tumor subtypes and highlighted the difference in regulations of cell death molecular mechanisms.
Collapse
Affiliation(s)
- Jean-Marie Ravel
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005 Paris, France; (J.-M.R.); (L.C.M.G.); (N.S.); (L.C.); (E.B.); (A.Z.)
- Laboratoire de génétique médicale, CHRU-Nancy, F-54000 Nancy, France
- Inserm, NGERE, Université de Lorraine, F-54000 Nancy, France
| | - L. Cristobal Monraz Gomez
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005 Paris, France; (J.-M.R.); (L.C.M.G.); (N.S.); (L.C.); (E.B.); (A.Z.)
| | - Nicolas Sompairac
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005 Paris, France; (J.-M.R.); (L.C.M.G.); (N.S.); (L.C.); (E.B.); (A.Z.)
- Centre de Recherches Interdisciplinaires, Université Paris Descartes, 75006 Paris, France
| | - Laurence Calzone
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005 Paris, France; (J.-M.R.); (L.C.M.G.); (N.S.); (L.C.); (E.B.); (A.Z.)
| | - Boris Zhivotovsky
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia;
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177 Stockholm, Sweden
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France;
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805 Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou 215163, China
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Emmanuel Barillot
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005 Paris, France; (J.-M.R.); (L.C.M.G.); (N.S.); (L.C.); (E.B.); (A.Z.)
| | - Andrei Zinovyev
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005 Paris, France; (J.-M.R.); (L.C.M.G.); (N.S.); (L.C.); (E.B.); (A.Z.)
| | - Inna Kuperstein
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, 75005 Paris, France; (J.-M.R.); (L.C.M.G.); (N.S.); (L.C.); (E.B.); (A.Z.)
| |
Collapse
|
12
|
Haprolid Inhibits Tumor Growth of Hepatocellular Carcinoma through Rb/E2F and Akt/mTOR Inhibition. Cancers (Basel) 2020; 12:cancers12030615. [PMID: 32155915 PMCID: PMC7139901 DOI: 10.3390/cancers12030615] [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: 12/01/2019] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) represents a major health burden with limited curative treatment options. There is a substantial unmet need to develop innovative approaches to impact the progression of advanced HCC. Haprolid is a novel natural component isolated from myxobacteria. Haprolid has been reported as a potent selective cytotoxin against a panel of tumor cells in recent studies including HCC cells. The aims of this study are to evaluate the antitumor effect of haprolid in HCC and to understand its underlying molecular mechanisms. METHODS The efficacy of haprolid was evaluated in human HCC cell lines (Huh-7, Hep3B and HepG2) and xenograft tumors (NMRI-Foxn1nu mice with injection of Hep3B cells). Cytotoxic activity of haprolid was determined by the WST-1 and crystal violet assay. Wound healing, transwell and tumorsphere assays were performed to investigate migration and invasion of HCC cells. Apoptosis and cell-cycle distribution were measured by flow cytometry. The effects of haprolid on the Rb/E2F and Akt/mTOR pathway were examined by immunoblotting and immunohistochemistry. RESULTS haprolid treatment significantly inhibited cell proliferation, migration and invasion in vitro. The epithelial-mesenchymal transition (EMT) was impaired by haprolid treatment and the expression level of N-cadherin, vimentin and Snail was downregulated. Moreover, growth of HCC cells in vitro was suppressed by inhibition of G1/S transition, and partially by induction of apoptosis. The drug induced downregulation of cell cycle regulatory proteins cyclin A, cyclin B and CDK2 and induced upregulation of p21 and p27. Further evidence showed that these effects of haprolid were associated with Rb/E2F downregulation and Akt/mTOR inhibition. Finally, in vivo nude mice experiments demonstrated significant inhibition of tumor growth upon haprolid treatment. CONCLUSION Our results show that haprolid inhibits the growth of HCC through dual inhibition of Rb/E2F and Akt/mTOR pathways. Therefore, haprolid might be considered as a new and promising candidate for the palliative therapy of HCC.
Collapse
|
13
|
Rahmati S, Abovsky M, Pastrello C, Kotlyar M, Lu R, Cumbaa CA, Rahman P, Chandran V, Jurisica I. pathDIP 4: an extended pathway annotations and enrichment analysis resource for human, model organisms and domesticated species. Nucleic Acids Res 2020; 48:D479-D488. [PMID: 31733064 PMCID: PMC7145646 DOI: 10.1093/nar/gkz989] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/11/2019] [Accepted: 11/13/2019] [Indexed: 12/14/2022] Open
Abstract
PathDIP was introduced to increase proteome coverage of literature-curated human pathway databases. PathDIP 4 now integrates 24 major databases. To further reduce the number of proteins with no curated pathway annotation, pathDIP integrates pathways with physical protein-protein interactions (PPIs) to predict significant physical associations between proteins and curated pathways. For human, it provides pathway annotations for 5366 pathway orphans. Integrated pathway annotation now includes six model organisms and ten domesticated animals. A total of 6401 core and ortholog pathways have been curated from the literature or by annotating orthologs of human proteins in the literature-curated pathways. Extended pathways are the result of combining these pathways with protein-pathway associations that are predicted using organism-specific PPIs. Extended pathways expand proteome coverage from 81 088 to 120 621 proteins, making pathDIP 4 the largest publicly available pathway database for these organisms and providing a necessary platform for comprehensive pathway-enrichment analysis. PathDIP 4 users can customize their search and analysis by selecting organism, identifier and subset of pathways. Enrichment results and detailed annotations for input list can be obtained in different formats and views. To support automated bioinformatics workflows, Java, R and Python APIs are available for batch pathway annotation and enrichment analysis. PathDIP 4 is publicly available at http://ophid.utoronto.ca/pathDIP.
Collapse
Affiliation(s)
- Sara Rahmati
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Department of Medicine, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada
- Department of Medicine, Memorial University of Newfoundland, Saint John's, NL A1B 3V6, Canada
| | - Mark Abovsky
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Chiara Pastrello
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Max Kotlyar
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Richard Lu
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Christian A Cumbaa
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Proton Rahman
- Department of Medicine, Memorial University of Newfoundland, Saint John's, NL A1B 3V6, Canada
| | - Vinod Chandran
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Department of Medicine, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada
- Department of Medicine, Memorial University of Newfoundland, Saint John's, NL A1B 3V6, Canada
- Department of Medicine, Division of Rheumatology, University of Toronto, Toronto, ON M5G 2C4, Canada
- Department of Laboratory Medicine and Pathobiology (LMP), Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Igor Jurisica
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Department of Medical Biophysics, University of Toronto, ON M 5G 1L7, Canada
- Department of Computer Science, University of Toronto, ON M5S 1A4, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
14
|
Cohen DPA, Lebsir D, Benderitter M, Souidi M. A systems biology approach to propose a new mechanism of regulation of repetitive prophylaxis of stable iodide on sodium/iodide symporter (NIS). Biochimie 2019; 162:208-215. [PMID: 31071356 DOI: 10.1016/j.biochi.2019.04.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/29/2019] [Indexed: 01/27/2023]
Abstract
Our group showed that repetitive dose of potassium iodide (KI) for eight days offers an efficient protection for exposure to repeated radioactive emissions without adverse effects on adult rats. However, differential expression of genes implicated in Wolff-Chaikoff effect was observed. To understand the Wolff-Chaikoff regulation and its molecular constituents during repetitive administration of KI, a biochemical reaction network was constructed as a "geographical" map of the thyrocyte depicting iodide and thyroid hormone synthesis. Path analysis of the network has been performed to investigate the presence of a regulatory circuit of the node iodide to the node "nis transcription". NIS is responsible for the uptake of KI and plays an important role in the Wolff-Chaikoff effect. The map is a source for the most updated information about iodide and thyroid hormone metabolism. Based on this map, we propose a hypothesis that shows a putative mechanism behind NIS regulation and KI uptake.
Collapse
Affiliation(s)
- David P A Cohen
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE/SESANE/LRTOX, 92262, Fontenay-aux-Roses, France
| | - Dalila Lebsir
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE/SESANE/LRTOX, 92262, Fontenay-aux-Roses, France
| | - Marc Benderitter
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PRP-SANTE/SERAMED, 92262, Fontenay-aux-Roses, France
| | - Maâmar Souidi
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PRP-SANTE/SERAMED, 92262, Fontenay-aux-Roses, France.
| |
Collapse
|
15
|
Gutiérrez-González M, Latorre Y, Zúñiga R, Aguillón JC, Molina MC, Altamirano C. Transcription factor engineering in CHO cells for recombinant protein production. Crit Rev Biotechnol 2019; 39:665-679. [PMID: 31030575 DOI: 10.1080/07388551.2019.1605496] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The continuous increase of approved biopharmaceutical products drives the development of more efficient recombinant protein expression systems. Chinese hamster ovary (CHO) cells are the mainstay for this purpose but have some drawbacks, such as low levels of expression. Several strategies have been applied to increase the productivity of CHO cells with different outcomes. Transcription factor (TF) engineering has emerged as an interesting and successful approach, as these proteins can act as master regulators; the expression and function of a TF can be controlled by small molecules, and it is possible to design tailored TFs and promoters with desired features. To date, the majority of studies have focused on the use of TFs with growth, metabolic, cell cycle or endoplasmic reticulum functions, although there is a trend to develop new, synthetic TFs. Moreover, new synthetic biological approaches are showing promising advances for the development of specific TFs, even with tailored ligand sensitivity. In this article, we summarize the strategies to increase recombinant protein expression by modulating and designing TFs and with advancements in synthetic biology. We also illustrate how this class of proteins can be used to develop more robust expression systems.
Collapse
Affiliation(s)
| | - Yesenia Latorre
- b Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso , Valparaíso , Chile
| | - Roberto Zúñiga
- a Centro de InmunoBiotecnología, Universidad de Chile , Santiago , Chile
| | | | | | - Claudia Altamirano
- b Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso , Valparaíso , Chile
| |
Collapse
|
16
|
Kondratova M, Sompairac N, Barillot E, Zinovyev A, Kuperstein I. Signalling maps in cancer research: construction and data analysis. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2018; 2018:4964960. [PMID: 29688383 PMCID: PMC5890450 DOI: 10.1093/database/bay036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/19/2018] [Indexed: 12/22/2022]
Abstract
Generation and usage of high-quality molecular signalling network maps can be augmented by standardizing notations, establishing curation workflows and application of computational biology methods to exploit the knowledge contained in the maps. In this manuscript, we summarize the major aims and challenges of assembling information in the form of comprehensive maps of molecular interactions. Mainly, we share our experience gained while creating the Atlas of Cancer Signalling Network. In the step-by-step procedure, we describe the map construction process and suggest solutions for map complexity management by introducing a hierarchical modular map structure. In addition, we describe the NaviCell platform, a computational technology using Google Maps API to explore comprehensive molecular maps similar to geographical maps and explain the advantages of semantic zooming principles for map navigation. We also provide the outline to prepare signalling network maps for navigation using the NaviCell platform. Finally, several examples of cancer high-throughput data analysis and visualization in the context of comprehensive signalling maps are presented.
Collapse
Affiliation(s)
- Maria Kondratova
- 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
| | - Nicolas Sompairac
- 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
| | - Andrei Zinovyev
- 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
| | - Inna Kuperstein
- 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
| |
Collapse
|
17
|
Zhou S, Appleman VA, Rose CM, Jun HJ, Yang J, Zhou Y, Bronson RT, Gygi SP, Charest A. Chronic platelet-derived growth factor receptor signaling exerts control over initiation of protein translation in glioma. Life Sci Alliance 2018; 1:e201800029. [PMID: 30456354 PMCID: PMC6238596 DOI: 10.26508/lsa.201800029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/29/2018] [Accepted: 05/29/2018] [Indexed: 01/23/2023] Open
Abstract
Using phospho-proteomics in a new model of malignant glioma, we reveal that clinically relevant, chronic PDGFRα signaling differs considerably from acute receptor stimulation and unveils previously unrecognized control over key elements of the translation initiation machinery. Activation of the platelet-derived growth factor receptors (PDGFRs) gives rise to some of the most important signaling pathways that regulate mammalian cellular growth, survival, proliferation, and differentiation and their misregulation is common in a variety of diseases. Herein, we present a comprehensive and detailed map of PDGFR signaling pathways assembled from literature and integrate this map in a bioinformatics protocol designed to extract meaningful information from large-scale quantitative proteomics mass spectrometry data. We demonstrate the usefulness of this approach using a new genetically engineered mouse model of PDGFRα-driven glioma. We discovered that acute PDGFRα stimulation differs considerably from chronic receptor activation in the regulation of protein translation initiation. Transient stimulation activates several key components of the translation initiation machinery, whereas the clinically relevant chronic activity of PDGFRα is associated with a significant shutdown of translational members. Our work defines a step-by-step approach to extract biologically relevant insights from global unbiased phospho-protein datasets to uncover targets for therapeutic assessment.
Collapse
Affiliation(s)
- Shuang Zhou
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Vicky A Appleman
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Hyun Jung Jun
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Juechen Yang
- Department of Computer Science, North Dakota State University, Fargo, ND, USA
| | - Yue Zhou
- Department of Statistics, North Dakota State University, Fargo, ND, USA
| | - Roderick T Bronson
- Rodent Histopathology Core, Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Steve P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Al Charest
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
Rougny A, Gloaguen P, Langonné N, Reiter E, Crépieux P, Poupon A, Froidevaux C. A logic-based method to build signaling networks and propose experimental plans. Sci Rep 2018; 8:7830. [PMID: 29777117 PMCID: PMC5959848 DOI: 10.1038/s41598-018-26006-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 04/27/2018] [Indexed: 11/15/2022] Open
Abstract
With the dramatic increase of the diversity and the sheer quantity of biological data generated, the construction of comprehensive signaling networks that include precise mechanisms cannot be carried out manually anymore. In this context, we propose a logic-based method that allows building large signaling networks automatically. Our method is based on a set of expert rules that make explicit the reasoning made by biologists when interpreting experimental results coming from a wide variety of experiment types. These rules allow formulating all the conclusions that can be inferred from a set of experimental results, and thus building all the possible networks that explain these results. Moreover, given an hypothesis, our system proposes experimental plans to carry out in order to validate or invalidate it. To evaluate the performance of our method, we applied our framework to the reconstruction of the FSHR-induced and the EGFR-induced signaling networks. The FSHR is known to induce the transactivation of the EGFR, but very little is known on the resulting FSH- and EGF-dependent network. We built a single network using data underlying both networks. This leads to a new hypothesis on the activation of MEK by p38MAPK, which we validate experimentally. These preliminary results represent a first step in the demonstration of a cross-talk between these two major MAP kinases pathways.
Collapse
Affiliation(s)
- Adrien Rougny
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Aomi, Tokyo, 135-0064, Japan.,Laboratoire de Recherche en Informatique UMR CNRS 8623, Université Paris-Sud, Université Paris-Saclay, Orsay Cedex, 91405, France
| | - Pauline Gloaguen
- PRC, INRA, CNRS, Université François Rabelais-Tours, 37380, Nouzilly, France
| | - Nathalie Langonné
- PRC, INRA, CNRS, Université François Rabelais-Tours, 37380, Nouzilly, France.,CNRS; Université François-Rabelais de Tours, UMR 7292, 37032, Tours, France
| | - Eric Reiter
- PRC, INRA, CNRS, Université François Rabelais-Tours, 37380, Nouzilly, France
| | - Pascale Crépieux
- PRC, INRA, CNRS, Université François Rabelais-Tours, 37380, Nouzilly, France
| | - Anne Poupon
- PRC, INRA, CNRS, Université François Rabelais-Tours, 37380, Nouzilly, France.
| | - Christine Froidevaux
- Laboratoire de Recherche en Informatique UMR CNRS 8623, Université Paris-Sud, Université Paris-Saclay, Orsay Cedex, 91405, France
| |
Collapse
|
19
|
Abstract
A major endeavor of systems biology is the construction of graphical and computational models of biological pathways as a means to better understand their structure and function. Here, we present a protocol for a biologist-friendly graphical modeling scheme that facilitates the construction of detailed network diagrams, summarizing the components of a biological pathway (such as proteins and biochemicals) and illustrating how they interact. These diagrams can then be used to simulate activity flow through a pathway, thereby modeling its dynamic behavior. The protocol is divided into four sections: (i) assembly of network diagrams using the modified Edinburgh Pathway Notation (mEPN) scheme and yEd network editing software with pathway information obtained from published literature and databases of molecular interaction data; (ii) parameterization of the pathway model within yEd through the placement of 'tokens' on the basis of the known or imputed amount or activity of a component; (iii) model testing through visualization and quantitative analysis of the movement of tokens through the pathway, using the network analysis tool Graphia Professional and (iv) optimization of model parameterization and experimentation. This is the first modeling approach that combines a sophisticated notation scheme for depicting biological events at the molecular level with a Petri net-based flow simulation algorithm and a powerful visualization engine with which to observe the dynamics of the system being modeled. Unlike many mathematical approaches to modeling pathways, it does not require the construction of a series of equations or rate constants for model parameterization. Depending on a model's complexity and the availability of information, its construction can take days to months, and, with refinement, possibly years. However, once assembled and parameterized, a simulation run, even on a large model, typically takes only seconds. Models constructed using this approach provide a means of knowledge management, information exchange and, through the computation simulation of their dynamic activity, generation and testing of hypotheses, as well as prediction of a system's behavior when perturbed.
Collapse
|
20
|
Zhu J, Yan F, Tao J, Zhu X, Liu J, Deng S, Zhang X. Cdc37 facilitates cell survival of colorectal carcinoma via activating the CDK4 signaling pathway. Cancer Sci 2018; 109:656-665. [PMID: 29288563 PMCID: PMC5834791 DOI: 10.1111/cas.13495] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/12/2017] [Accepted: 12/20/2017] [Indexed: 01/03/2023] Open
Abstract
Cell division cycle 37 (Cdc37) is an important partner for heat shock protein 90 (HSP90), assisting in molecular chaperone activities, particularly with regard to the regulation of protein kinases. Given its influence on cell growth pathways, Cdc37 has been discussed as a potential intermediate in carcinogenesis. However, to date, the potential functional roles and molecular mechanisms by which Cdc37 regulates cell survival in colorectal carcinoma (CRC) remain unclear. Here, we investigated the expression of Cdc37 and its clinical significance in CRC, and systematically explored the role and the underlying mechanism of Cdc37 in CRC cell survival both in vitro and in vivo. Our results showed that Cdc37 was remarkably up-regulated in CRC, which facilitated cell survival mainly by promoting cell proliferation, G1-S transition, and inhibiting cell apoptosis. Our data further indicated that Cdc37 increased the stability of cyclin-dependent kinase 4 (CDK4) to activate the retinoblastoma 1 (RB1) signaling pathway, followed by increased expression of Bcl-2 and Bcl-xL, which ultimately promoted cell survival in CRC. Moreover, knockdown of CDK4 reversed the Cdc37-mediated effect in promoting the progression of CRC. Our findings showed that Cdc37 played a critical role in promoting CRC cell survival by increasing CDK4 stability to activate the RB1 signaling pathway. Thereby, Cdc37 might serve as a potential therapeutic target in CRC patients.
Collapse
Affiliation(s)
- Jianjun Zhu
- Department of Human Anatomy and Sichuan Key Laboratory of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Fang Yan
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Jia Tao
- Department of Pathology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaohua Zhu
- Department of Human Anatomy and Sichuan Key Laboratory of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Jiayou Liu
- Department of Human Anatomy and Sichuan Key Laboratory of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Shishan Deng
- Department of Human Anatomy and Sichuan Key Laboratory of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Xiaoming Zhang
- Sichuan Key Laboratory of Medical Imaging and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
21
|
Lages J, Shepelyansky DL, Zinovyev A. Inferring hidden causal relations between pathway members using reduced Google matrix of directed biological networks. PLoS One 2018; 13:e0190812. [PMID: 29370181 PMCID: PMC5784915 DOI: 10.1371/journal.pone.0190812] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 12/20/2017] [Indexed: 12/18/2022] Open
Abstract
Signaling pathways represent parts of the global biological molecular network which connects them into a seamless whole through complex direct and indirect (hidden) crosstalk whose structure can change during development or in pathological conditions. We suggest a novel methodology, called Googlomics, for the structural analysis of directed biological networks using spectral analysis of their Google matrices, using parallels with quantum scattering theory, developed for nuclear and mesoscopic physics and quantum chaos. We introduce analytical "reduced Google matrix" method for the analysis of biological network structure. The method allows inferring hidden causal relations between the members of a signaling pathway or a functionally related group of genes. We investigate how the structure of hidden causal relations can be reprogrammed as a result of changes in the transcriptional network layer during cancerogenesis. The suggested Googlomics approach rigorously characterizes complex systemic changes in the wiring of large causal biological networks in a computationally efficient way.
Collapse
Affiliation(s)
- José Lages
- Institut UTINAM, Observatoire des Sciences de l'Univers THETA, CNRS, Université de Franche-Comté, 25030 Besançon, France
| | - Dima L Shepelyansky
- Laboratoire de Physique Théorique du CNRS, IRSAMC, Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Andrei Zinovyev
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, F-75005, Paris, France
- Laboratory of advanced methods for high-dimensional data analysis, Lobachevsky University, Nizhni Novgorod, Russia
| |
Collapse
|
22
|
Csabai L, Ölbei M, Budd A, Korcsmáros T, Fazekas D. SignaLink: Multilayered Regulatory Networks. Methods Mol Biol 2018; 1819:53-73. [PMID: 30421399 DOI: 10.1007/978-1-4939-8618-7_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Biological networks are graphs used to represent the inner workings of a biological system. Networks describe the relationships of the elements of biological systems using edges and nodes. However, the resulting representation of the system can sometimes be too simplistic to usefully model reality. By combining several different interaction types within one larger multilayered biological network, tools such as SignaLink provide a more nuanced view than those relying on single-layer networks (where edges only describe one kind of interaction). Multilayered networks display connections between multiple networks (i.e., protein-protein interactions and their transcriptional and posttranscriptional regulators), each one of them describing a specific set of connections. Multilayered networks also allow us to depict cross talk between cellular systems, which is a more realistic way of describing molecular interactions. They can be used to collate networks from different sources into one multilayered structure, which makes them useful as an analytic tool as well.
Collapse
Affiliation(s)
| | - Márton Ölbei
- Earlham Institute, Norwich Research Park, Norwich, UK.,Quadram Institute, Norwich Research Park, Norwich, UK
| | - Aidan Budd
- Earlham Institute, Norwich Research Park, Norwich, UK
| | - Tamás Korcsmáros
- Eötvös Loránd University, Budapest, Hungary. .,Earlham Institute, Norwich Research Park, Norwich, UK. .,Quadram Institute, Norwich Research Park, Norwich, UK.
| | - Dávid Fazekas
- Eötvös Loránd University, Budapest, Hungary.,Earlham Institute, Norwich Research Park, Norwich, UK
| |
Collapse
|
23
|
Khan FM, Sadeghi M, Gupta SK, Wolkenhauer O. A Network-Based Integrative Workflow to Unravel Mechanisms Underlying Disease Progression. Methods Mol Biol 2018; 1702:247-276. [PMID: 29119509 DOI: 10.1007/978-1-4939-7456-6_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Unraveling mechanisms underlying diseases has motivated the development of systems biology approaches. The key challenges for the development of mathematical models and computational tool are (1) the size of molecular networks, (2) the nonlinear nature of spatio-temporal interactions, and (3) feedback loops in the structure of interaction networks. We here propose an integrative workflow that combines structural analyses of networks, high-throughput data, and mechanistic modeling. As an illustration of the workflow, we use prostate cancer as a case study with the aim of identifying key functional components associated with primary to metastasis transitions. Analysis carried out by the workflow revealed that HOXD10, BCL2, and PGR are the most important factors affected in primary prostate samples, whereas, in the metastatic state, STAT3, JUN, and JUNB are playing a central role. The identified key elements of each network are validated using patient survival analysis. The workflow presented here allows experimentalists to use heterogeneous data sources for the identification of diagnostic and prognostic signatures.
Collapse
Affiliation(s)
- Faiz M Khan
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany
| | - Mehdi Sadeghi
- Research Institute for Fundamental Sciences (RIFS), University of Tabriz, Tabriz, Iran
| | - Shailendra K Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany.,Chhattisgarh Swami Vivekanand Technical University, Bhilai, Chhattisgarh, India
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051, Rostock, Germany. .,Chhattisgarh Swami Vivekanand Technical University, Bhilai, Chhattisgarh, India. .,Stellenbosch Institute of Advanced Study (STIAS), Wallenberg Research Centre, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
24
|
Pathak RK, Baunthiyal M, Pandey N, Pandey D, Kumar A. Modeling of the jasmonate signaling pathway in Arabidopsis thaliana with respect to pathophysiology of Alternaria blight in Brassica. Sci Rep 2017; 7:16790. [PMID: 29196636 PMCID: PMC5711873 DOI: 10.1038/s41598-017-16884-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 11/08/2017] [Indexed: 01/01/2023] Open
Abstract
The productivity of Oilseed Brassica, one of the economically important crops of India, is seriously affected by the disease, Alternaria blight. The disease is mainly caused by two major necrotrophic fungi, Alternaria brassicae and Alternaria brassicicola which are responsible for significant yield losses. Till date, no resistant source is available against Alternaria blight, hence plant breeding methods can not be used to develop disease resistant varieties. Jasmonate mediated signalling pathway, which is known to play crucial role during defense response against necrotrophs, could be strengthened in Brassica plants to combat the disease. Since scanty information is available in Brassica-Alternaria pathosystems at molecular level therefore, in the present study efforts have been made to model jasmonic acid pathway in Arabidopsis thaliana to simulate the dynamic behaviour of molecular species in the model. Besides, the developed model was also analyzed topologically for investigation of the hubs node. COI1 is identified as one of the promising candidate genes in response to Alternaria and other linked components of plant defense mechanisms against the pathogens. The findings from present study are therefore informative for understanding the molecular basis of pathophysiology and rational management of Alternaria blight for securing food and nutritional security.
Collapse
Affiliation(s)
- Rajesh Kumar Pathak
- Department of Biotechnology, Govind Ballabh Pant Institute of Engineering & Technology, Pauri Garhwal, 246194, Uttarakhand, India
| | - Mamta Baunthiyal
- Department of Biotechnology, Govind Ballabh Pant Institute of Engineering & Technology, Pauri Garhwal, 246194, Uttarakhand, India.
| | - Neetesh Pandey
- Centre for Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute (IASRI), Pusa, 110012, New Delhi, India
| | - Dinesh Pandey
- Department of Molecular Biology & Genetic Engineering, College of Basic Sciences & Humanities, G. B. Pant University of Agriculture & Technology, Pantnagar, 263145, India
| | - Anil Kumar
- Department of Molecular Biology & Genetic Engineering, College of Basic Sciences & Humanities, G. B. Pant University of Agriculture & Technology, Pantnagar, 263145, India.
| |
Collapse
|
25
|
Systems biology analysis of longitudinal functional response of endothelial cells to shear stress. Proc Natl Acad Sci U S A 2017; 114:10990-10995. [PMID: 28973892 DOI: 10.1073/pnas.1707517114] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Blood flow and vascular shear stress patterns play a significant role in inducing and modulating physiological responses of endothelial cells (ECs). Pulsatile shear (PS) is associated with an atheroprotective endothelial phenotype, while oscillatory shear (OS) is associated with an atheroprone endothelial phenotype. Although mechanisms of endothelial shear response have been extensively studied, most studies focus on characterization of single molecular pathways, mainly at fixed time points after stress application. Here, we carried out a longitudinal time-series study to measure the transcriptome after the application of PS and OS. We performed systems analyses of transcriptional data of cultured human vascular ECs to elucidate the dynamics of endothelial responses in several functional pathways such as cell cycle, oxidative stress, and inflammation. By combining the temporal data on differentially expressed transcription factors and their targets with existing knowledge on relevant functional pathways, we infer the causal relationships between disparate endothelial functions through common transcriptional regulation mechanisms. Our study presents a comprehensive temporally longitudinal experimental study and mechanistic model of shear stress response. By comparing the relative endothelial expressions of genes between OS and PS, we provide insights and an integrated perspective into EC function in response to differential shear. This study has significant implications for the pathogenesis of vascular diseases.
Collapse
|
26
|
Unraveling a tumor type-specific regulatory core underlying E2F1-mediated epithelial-mesenchymal transition to predict receptor protein signatures. Nat Commun 2017; 8:198. [PMID: 28775339 PMCID: PMC5543083 DOI: 10.1038/s41467-017-00268-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 06/15/2017] [Indexed: 12/18/2022] Open
Abstract
Cancer is a disease of subverted regulatory pathways. In this paper, we reconstruct the regulatory network around E2F, a family of transcription factors whose deregulation has been associated to cancer progression, chemoresistance, invasiveness, and metastasis. We integrate gene expression profiles of cancer cell lines from two E2F1-driven highly aggressive bladder and breast tumors, and use network analysis methods to identify the tumor type-specific core of the network. By combining logic-based network modeling, in vitro experimentation, and gene expression profiles from patient cohorts displaying tumor aggressiveness, we identify and experimentally validate distinctive, tumor type-specific signatures of receptor proteins associated to epithelial-mesenchymal transition in bladder and breast cancer. Our integrative network-based methodology, exemplified in the case of E2F1-induced aggressive tumors, has the potential to support the design of cohort- as well as tumor type-specific treatments and ultimately, to fight metastasis and therapy resistance.Deregulation of E2F family transcription factors is associated with cancer progression and metastasis. Here, the authors construct a map of the regulatory network around the E2F family, and using gene expression profiles, identify tumour type-specific regulatory cores and receptor expression signatures associated with epithelial-mesenchymal transition in bladder and breast cancer.
Collapse
|
27
|
Tang J, Xie Y, Xu X, Yin Y, Jiang R, Deng L, Tan Z, Gangarapu V, Tang J, Sun B. Bidirectional transcription of Linc00441 and RB1 via H3K27 modification-dependent way promotes hepatocellular carcinoma. Cell Death Dis 2017; 8:e2675. [PMID: 28300839 PMCID: PMC5386573 DOI: 10.1038/cddis.2017.81] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 01/16/2017] [Accepted: 01/24/2017] [Indexed: 02/07/2023]
Abstract
The retinoblastoma gene (RB1), a known tumor-suppressor gene (TSG), was decreased in multiple cancers including hepatocellular carcinoma (HCC). Here we focused on the bidirectional transcripted long noncoding RNA (Linc00441) with neighbor gene RB1 to investigate whether Linc00441 is involved in the suppression of RB1 in HCC. We found that aberrant upregulated intranuclear Linc00441 was reversely correlated with RB1 expression in human HCC samples. The gain- and loss-of-function investigation revealed that Linc00441 could promote the proliferation of HCC cells in vitro and in vivo with an apoptosis suppression and cell cycle rearrangement. Furthermore, RNA pull-down assay indicated the decreased level of RB1 induced by Linc00441 was associated with the incidental methylation by DNMT3A recruited by Linc00441. On the contrary, the transcription factor (TCF-4) enhanced H3K27 acetylation and direct transcription factor for Linc00441 was responsible for the upregulation of Linc00441 in HCC. In conclusion, the epigenetic interaction between Linc00441 and bidirectional transcripted neighbor RB1 may be a de novo theory cutting-point for the inactivation of RB1 in HCC and may serve as targeting site for tumor therapy in the future.
Collapse
Affiliation(s)
- Junwei Tang
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yu Xie
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiaoliang Xu
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yin Yin
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Runqiu Jiang
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lei Deng
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhongming Tan
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Venkatanarayana Gangarapu
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jinhai Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Beicheng Sun
- Liver Transplantation Center, The First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
28
|
Cho SH, Park SM, Lee HS, Lee HY, Cho KH. Attractor landscape analysis of colorectal tumorigenesis and its reversion. BMC SYSTEMS BIOLOGY 2016; 10:96. [PMID: 27765040 PMCID: PMC5072344 DOI: 10.1186/s12918-016-0341-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 10/10/2016] [Indexed: 02/08/2023]
Abstract
Background Colorectal cancer arises from the accumulation of genetic mutations that induce dysfunction of intracellular signaling. However, the underlying mechanism of colorectal tumorigenesis driven by genetic mutations remains yet to be elucidated. Results To investigate colorectal tumorigenesis at a system-level, we have reconstructed a large-scale Boolean network model of the human signaling network by integrating previous experimental results on canonical signaling pathways related to proliferation, metastasis, and apoptosis. Throughout an extensive simulation analysis of the attractor landscape of the signaling network model, we found that the attractor landscape changes its shape by expanding the basin of attractors for abnormal proliferation and metastasis along with the accumulation of driver mutations. A further hypothetical study shows that restoration of a normal phenotype might be possible by reversely controlling the attractor landscape. Interestingly, the targets of approved anti-cancer drugs were highly enriched in the identified molecular targets for the reverse control. Conclusions Our results show that the dynamical analysis of a signaling network based on attractor landscape is useful in acquiring a system-level understanding of tumorigenesis and developing a new therapeutic strategy. Electronic supplementary material The online version of this article (doi:10.1186/s12918-016-0341-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sung-Hwan Cho
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sang-Min Park
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Ho-Sung Lee
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hwang-Yeol Lee
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Kwang-Hyun Cho
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea. .,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
29
|
Tumor apelin, not serum apelin, is associated with the clinical features and prognosis of gastric cancer. BMC Cancer 2016; 16:794. [PMID: 27733135 PMCID: PMC5062883 DOI: 10.1186/s12885-016-2815-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 09/26/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND To study the association between Apelin expression and the clinical features and postoperative prognosis in patients with gastric cancer (Int J Cancer 136:2388-2401, 2015). METHODS Tumor samples and matched adjacent normal tissues were collected from 270 patients with GC receiving surgical resection. The tumor and serum Apelin levels were determined by immunohistochemistry and ELISA methods, respectively. GC cell lines were cultured for migration and invasive assays. RESULTS Our data showed that tumor Apelin expression status, instead of serum Apelin level, was closely associated with more advance clinical features including tumor differentiation, lymph node and distant metastases. Moreover, patients with high tumor Apelin level had a significantly shorter overall survival period compared to those with low Apelin expression and those with or negative Apelin staining. Our in vitro study revealed that the Apelin regulated the migration and invasion abilities of GC cell lines, accompanied by up-regulations of a variety of cytokines associated with tumor invasiveness. CONCLUSION Our data suggest that tumor Apelin can be used as a marker to evaluate clinical characteristics and predict prognosis in GC patients.
Collapse
|
30
|
Abstract
There is a need for formalised diagrams that both summarise current biological pathway knowledge and support modelling approaches that explain and predict their behaviour. Here, we present a new, freely available modelling framework that includes a biologist-friendly pathway modelling language (mEPN), a simple but sophisticated method to support model parameterisation using available biological information; a stochastic flow algorithm that simulates the dynamics of pathway activity; and a 3-D visualisation engine that aids understanding of the complexities of a system’s dynamics. We present example pathway models that illustrate of the power of approach to depict a diverse range of systems. This Community Page presents a biologist-friendly method for compiling detailed graphical models of biological pathways from the literature, including their subsequent parameterization for use in dynamic simulations of their activity.
Collapse
|
31
|
Long J, Liu Z, Wu X, Xu Y, Ge C. Identification of disease-associated pathways in pancreatic cancer by integrating genome-wide association study and gene expression data. Oncol Lett 2016; 12:537-543. [PMID: 27347177 PMCID: PMC4906788 DOI: 10.3892/ol.2016.4637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/17/2016] [Indexed: 12/02/2022] Open
Abstract
In order to additionally understand the pathogenesis of pancreatic cancer (PC), the present study conducted pathway analysis based on genome-wide association study (GWAS) and gene expression data to predict genes that are associated with PC. GWAS data (accession no., pha002874.1) were downloaded from National Center for Biotechnology Information (NCBI) database of Genotypes and Phenotypes, which included data concerning 1,896 patients with PC and 1,939 control individuals. Gene expression data [accession no., GSE23952; human pancreatic carcinoma Panc-1 transforming growth factor-β (TGF-β) treatment assay] were downloaded from NCBI Gene Expression Omnibus. Gene set enrichment analysis was used to identify significant pathways in the GWAS or gene expression profiles. Meta-analysis was performed based on pathway analysis of the two data sources. In total, 58 and 280 pathways were identified to be significant in the GWAS and gene expression data, respectively, with 7 pathways significant in both the data profiles. Hsa 04350 TGF-β signaling pathway had the smallest meta P-value. Other significant pathways in the two data sources were negative regulation of DNA-dependent transcription, the nucleolus, negative regulation of RNA metabolic process, the cellular defense response, exocytosis and galactosyltransferase activity. By constructing the gene-pathway network, 5 pathways were closely associated, apart from exocytosis and galactosyltransferase activity pathways. Among the 7 pathways, 11 key genes (2.9% out of a total of 380 genes) from the GWAS data and 43 genes (10.5% out of a total of 409 genes) from the gene expression data were differentially expressed. Only Abelson murine leukemia viral oncogene homolog 1 from the nucleolus pathway was significantly expressed in by both data sources. Overall, the results of the present analysis provide possible factors for the occurrence of PC, and the identification of the pathways and genes associated with PC provides valuable data for investigating the pathogenesis of PC in future studies.
Collapse
Affiliation(s)
- Jin Long
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhe Liu
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xingda Wu
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuanhong Xu
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Chunlin Ge
- Department of General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
32
|
Rougny A, Froidevaux C, Calzone L, Paulevé L. Qualitative dynamics semantics for SBGN process description. BMC SYSTEMS BIOLOGY 2016; 10:42. [PMID: 27306057 PMCID: PMC4910245 DOI: 10.1186/s12918-016-0285-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 06/02/2016] [Indexed: 01/08/2023]
Abstract
Background Qualitative dynamics semantics provide a coarse-grain modeling of networks dynamics by abstracting away kinetic parameters. They allow to capture general features of systems dynamics, such as attractors or reachability properties, for which scalable analyses exist. The Systems Biology Graphical Notation Process Description language (SBGN-PD) has become a standard to represent reaction networks. However, no qualitative dynamics semantics taking into account all the main features available in SBGN-PD had been proposed so far. Results We propose two qualitative dynamics semantics for SBGN-PD reaction networks, namely the general semantics and the stories semantics, that we formalize using asynchronous automata networks. While the general semantics extends standard Boolean semantics of reaction networks by taking into account all the main features of SBGN-PD, the stories semantics allows to model several molecules of a network by a unique variable. The obtained qualitative models can be checked against dynamical properties and therefore validated with respect to biological knowledge. We apply our framework to reason on the qualitative dynamics of a large network (more than 200 nodes) modeling the regulation of the cell cycle by RB/E2F. Conclusion The proposed semantics provide a direct formalization of SBGN-PD networks in dynamical qualitative models that can be further analyzed using standard tools for discrete models. The dynamics in stories semantics have a lower dimension than the general one and prune multiple behaviors (which can be considered as spurious) by enforcing the mutual exclusiveness between the activity of different nodes of a same story. Overall, the qualitative semantics for SBGN-PD allow to capture efficiently important dynamical features of reaction network models and can be exploited to further refine them. Electronic supplementary material The online version of this article (doi:10.1186/s12918-016-0285-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adrien Rougny
- Laboratoire de Recherche en Informatique UMR CNRS 8623, Université Paris-Sud, Université Paris-Saclay, Orsay Cedex, 91405, France
| | - Christine Froidevaux
- Laboratoire de Recherche en Informatique UMR CNRS 8623, Université Paris-Sud, Université Paris-Saclay, Orsay Cedex, 91405, France
| | - Laurence Calzone
- Institut Curie, PSL Research University, INSERM, U900, Mines Paris Tech, Paris, F-75005, France
| | - Loïc Paulevé
- Laboratoire de Recherche en Informatique UMR CNRS 8623, Université Paris-Sud, Université Paris-Saclay, Orsay Cedex, 91405, France.
| |
Collapse
|
33
|
Jagannadham J, Jaiswal HK, Agrawal S, Rawal K. Comprehensive Map of Molecules Implicated in Obesity. PLoS One 2016; 11:e0146759. [PMID: 26886906 PMCID: PMC4757102 DOI: 10.1371/journal.pone.0146759] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/22/2015] [Indexed: 01/22/2023] Open
Abstract
Obesity is a global epidemic affecting over 1.5 billion people and is one of the risk factors for several diseases such as type 2 diabetes mellitus and hypertension. We have constructed a comprehensive map of the molecules reported to be implicated in obesity. A deep curation strategy was complemented by a novel semi-automated text mining system in order to screen 1,000 full-length research articles and over 90,000 abstracts that are relevant to obesity. We obtain a scale free network of 804 nodes and 971 edges, composed of 510 proteins, 115 genes, 62 complexes, 23 RNA molecules, 83 simple molecules, 3 phenotype and 3 drugs in "bow-tie" architecture. We classify this network into 5 modules and identify new links between the recently discovered fat mass and obesity associated FTO gene with well studied examples such as insulin and leptin. We further built an automated docking pipeline to dock orlistat as well as other drugs against the 24,000 proteins in the human structural proteome to explain the therapeutics and side effects at a network level. Based upon our experiments, we propose that therapeutic effect comes through the binding of one drug with several molecules in target network, and the binding propensity is both statistically significant and different in comparison with any other part of human structural proteome.
Collapse
Affiliation(s)
- Jaisri Jagannadham
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida [UP]-201 307, India
| | - Hitesh Kumar Jaiswal
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida [UP]-201 307, India
| | - Stuti Agrawal
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida [UP]-201 307, India
| | - Kamal Rawal
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida [UP]-201 307, India
| |
Collapse
|
34
|
Mizuno S, Ogishima S, Kitatani K, Kikuchi M, Tanaka H, Yaegashi N, Nakaya J. Network Analysis of a Comprehensive Knowledge Repository Reveals a Dual Role for Ceramide in Alzheimer's Disease. PLoS One 2016; 11:e0148431. [PMID: 26849355 PMCID: PMC4752297 DOI: 10.1371/journal.pone.0148431] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/18/2016] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of senile dementia. Many inflammatory factors such as amyloid-β and pro-inflammatory cytokines are known to contribute to the inflammatory response in the AD brain. Sphingolipids are widely known to have roles in the pathogenesis of inflammatory diseases, where the precise roles for sphingolipids in inflammation-associated pathogenesis of AD are not well understood. Here we performed a network analysis to clarify the importance of sphingolipids and to model relationships among inflammatory factors and sphingolipids in AD. In this study, we have updated sphingolipid signaling and metabolic cascades in a map of AD signaling networks that we named “AlzPathway,” a comprehensive knowledge repository of signaling pathways in AD. Our network analysis of the updated AlzPathway indicates that the pathways related to ceramide are one of the primary pathways and that ceramide is one of the important players in the pathogenesis of AD. The results of our analysis suggest the following two prospects about inflammation in AD: (1) ceramide could play important roles in both inflammatory and anti-inflammatory pathways of AD, and (2) several factors such as Sphingomyelinase and Siglec-11 may be associated with ceramide related inflammation and anti-inflammation pathways in AD. In this study, network analysis of comprehensive knowledge repository reveals a dual role for ceramide in AD. This result provides a clue to clarify sphingolipids related inflammatory and anti-inflammatory pathways in AD.
Collapse
Affiliation(s)
- Satoshi Mizuno
- Department of Clinical Informatics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Clinical Record Informatics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- * E-mail: (SM); (SO)
| | - Soichi Ogishima
- Department of Clinical Record Informatics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- * E-mail: (SM); (SO)
| | - Kazuyuki Kitatani
- Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Masataka Kikuchi
- Department of Genome Informatics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hiroshi Tanaka
- Department of Clinical Record Informatics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Nobuo Yaegashi
- Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jun Nakaya
- Department of Clinical Informatics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
35
|
Kawakami E, Singh VK, Matsubara K, Ishii T, Matsuoka Y, Hase T, Kulkarni P, Siddiqui K, Kodilkar J, Danve N, Subramanian I, Katoh M, Shimizu-Yoshida Y, Ghosh S, Jere A, Kitano H. Network analyses based on comprehensive molecular interaction maps reveal robust control structures in yeast stress response pathways. NPJ Syst Biol Appl 2016; 2:15018. [PMID: 28725465 PMCID: PMC5516916 DOI: 10.1038/npjsba.2015.18] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 09/08/2015] [Accepted: 10/13/2015] [Indexed: 11/09/2022] Open
Abstract
Cellular stress responses require exquisite coordination between intracellular signaling molecules to integrate multiple stimuli and actuate specific cellular behaviors. Deciphering the web of complex interactions underlying stress responses is a key challenge in understanding robust biological systems and has the potential to lead to the discovery of targeted therapeutics for diseases triggered by dysregulation of stress response pathways. We constructed large-scale molecular interaction maps of six major stress response pathways in Saccharomyces cerevisiae (baker's or budding yeast). Biological findings from over 900 publications were converted into standardized graphical formats and integrated into a common framework. The maps are posted at http://www.yeast-maps.org/yeast-stress-response/ for browse and curation by the research community. On the basis of these maps, we undertook systematic analyses to unravel the underlying architecture of the networks. A series of network analyses revealed that yeast stress response pathways are organized in bow-tie structures, which have been proposed as universal sub-systems for robust biological regulation. Furthermore, we demonstrated a potential role for complexes in stabilizing the conserved core molecules of bow-tie structures. Specifically, complex-mediated reversible reactions, identified by network motif analyses, appeared to have an important role in buffering the concentration and activity of these core molecules. We propose complex-mediated reactions as a key mechanism mediating robust regulation of the yeast stress response. Thus, our comprehensive molecular interaction maps provide not only an integrated knowledge base, but also a platform for systematic network analyses to elucidate the underlying architecture in complex biological systems.
Collapse
Affiliation(s)
- Eiryo Kawakami
- Laboratory for Disease Systems Modeling, RIKEN-IMS, Kanagawa, Japan
| | | | | | - Takashi Ishii
- Laboratory for Disease Systems Modeling, RIKEN-IMS, Kanagawa, Japan
| | | | | | | | | | | | | | | | | | - Yuki Shimizu-Yoshida
- Laboratory for Disease Systems Modeling, RIKEN-IMS, Kanagawa, Japan.,Sony Computer Science Laboratories, Inc., Tokyo, Japan
| | | | - Abhay Jere
- LABS, Persistent Systems Limited, Pune, India
| | - Hiroaki Kitano
- Laboratory for Disease Systems Modeling, RIKEN-IMS, Kanagawa, Japan.,The Systems Biology Institute, Tokyo, Japan.,Sony Computer Science Laboratories, Inc., Tokyo, Japan.,Integrated Open Systems Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| |
Collapse
|
36
|
AlzPathway, an Updated Map of Curated Signaling Pathways: Towards Deciphering Alzheimer's Disease Pathogenesis. Methods Mol Biol 2016; 1303:423-32. [PMID: 26235082 DOI: 10.1007/978-1-4939-2627-5_25] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder in which loss of neurons and synaptic function causes dementia in the elderly. To clarify AD pathogenesis and develop drugs for AD, thousands of studies have elucidated signaling pathways involved. However, knowledge of AD signaling pathways has not been compiled as a pathway map. In this chapter, we introduce the manual construction of a pathway map in AD which we call "AlzPathway", that comprehensively catalogs signaling pathways in the field of AD. We have collected and manually curated over 100 review articles related to AD, and have built the AD pathway map. AlzPathway is currently composed of thousands of molecules and reactions in neurons, brain blood barrier, presynaptic, postsynaptic, astrocyte, and microglial cells, with their cellular localizations. AlzPathway provides a systems-biology platform of comprehensive AD signaling and related pathways which is expected to contribute to clarification of AD pathogenesis and AD drug development.
Collapse
|
37
|
Csikász-Nagy A, Mura I. Role of Computational Modeling in Understanding Cell Cycle Oscillators. Methods Mol Biol 2016; 1342:59-70. [PMID: 26254917 DOI: 10.1007/978-1-4939-2957-3_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The periodic oscillations in the activity of the cell cycle regulatory program, drives the timely activation of key cell cycle events. Interesting dynamical systems, such as oscillators, have been investigated by various theoretical and computational modeling methods. Thanks to the insights achieved by these modeling efforts we have gained considerable insights about the underlying molecular regulatory networks that can drive cell cycle oscillations. Here we review the basic features and characteristics of biological oscillators, discussing from a computational modeling point of view their specific architectures and the current knowledge about the dynamics that the life evolution selected to drive cell cycle oscillations.
Collapse
Affiliation(s)
- Attila Csikász-Nagy
- Randall Division of Cell and Molecular Biophysics, King's College London, Strand, London, SE1 1UL, UK,
| | | |
Collapse
|
38
|
Remy E, Rebouissou S, Chaouiya C, Zinovyev A, Radvanyi F, Calzone L. A Modeling Approach to Explain Mutually Exclusive and Co-Occurring Genetic Alterations in Bladder Tumorigenesis. Cancer Res 2015; 75:4042-52. [PMID: 26238783 DOI: 10.1158/0008-5472.can-15-0602] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 06/24/2015] [Indexed: 11/16/2022]
Abstract
Relationships between genetic alterations, such as co-occurrence or mutual exclusivity, are often observed in cancer, where their understanding may provide new insights into etiology and clinical management. In this study, we combined statistical analyses and computational modeling to explain patterns of genetic alterations seen in 178 patients with bladder tumors (either muscle-invasive or non-muscle-invasive). A statistical analysis on frequently altered genes identified pair associations, including co-occurrence or mutual exclusivity. Focusing on genetic alterations of protein-coding genes involved in growth factor receptor signaling, cell cycle, and apoptosis entry, we complemented this analysis with a literature search to focus on nine pairs of genetic alterations of our dataset, with subsequent verification in three other datasets available publicly. To understand the reasons and contexts of these patterns of associations while accounting for the dynamics of associated signaling pathways, we built a logical model. This model was validated first on published mutant mice data, then used to study patterns and to draw conclusions on counter-intuitive observations, allowing one to formulate predictions about conditions where combining genetic alterations benefits tumorigenesis. For example, while CDKN2A homozygous deletions occur in a context of FGFR3-activating mutations, our model suggests that additional PIK3CA mutation or p21CIP deletion would greatly favor invasiveness. Furthermore, the model sheds light on the temporal orders of gene alterations, for example, showing how mutual exclusivity of FGFR3 and TP53 mutations is interpretable if FGFR3 is mutated first. Overall, our work shows how to predict combinations of the major gene alterations leading to invasiveness through two main progression pathways in bladder cancer.
Collapse
Affiliation(s)
- Elisabeth Remy
- Aix Marseille Université, CNRS, Centrale Marseille, Marseille, France
| | - Sandra Rebouissou
- Institut Curie, PSL Research University, Paris, France. CNRS, UMR 144, Oncologie Moléculaire, Equipe Labellisée Ligue Contre le Cancer, Institut Curie, Paris, France
| | | | - Andrei Zinovyev
- Institut Curie, PSL Research University, Paris, France. INSERM, U900, Paris, France. Ecole des Mines ParisTech, Fontainebleau, France
| | - François Radvanyi
- Institut Curie, PSL Research University, Paris, France. CNRS, UMR 144, Oncologie Moléculaire, Equipe Labellisée Ligue Contre le Cancer, Institut Curie, Paris, France
| | - Laurence Calzone
- Institut Curie, PSL Research University, Paris, France. INSERM, U900, Paris, France. Ecole des Mines ParisTech, Fontainebleau, France.
| |
Collapse
|
39
|
Tripathi S, Flobak Å, Chawla K, Baudot A, Bruland T, Thommesen L, Kuiper M, Lægreid A. The gastrin and cholecystokinin receptors mediated signaling network: a scaffold for data analysis and new hypotheses on regulatory mechanisms. BMC SYSTEMS BIOLOGY 2015. [PMID: 26205660 PMCID: PMC4513977 DOI: 10.1186/s12918-015-0181-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background The gastrointestinal peptide hormones cholecystokinin and gastrin exert their biological functions via cholecystokinin receptors CCK1R and CCK2R respectively. Gastrin, a central regulator of gastric acid secretion, is involved in growth and differentiation of gastric and colonic mucosa, and there is evidence that it is pro-carcinogenic. Cholecystokinin is implicated in digestion, appetite control and body weight regulation, and may play a role in several digestive disorders. Results We performed a detailed analysis of the literature reporting experimental evidence on signaling pathways triggered by CCK1R and CCK2R, in order to create a comprehensive map of gastrin and cholecystokinin-mediated intracellular signaling cascades. The resulting signaling map captures 413 reactions involving 530 molecular species, and incorporates the currently available knowledge into one integrated signaling network. The decomposition of the signaling map into sub-networks revealed 18 modules that represent higher-level structures of the signaling map. These modules allow a more compact mapping of intracellular signaling reactions to known cell behavioral outcomes such as proliferation, migration and apoptosis. The integration of large-scale protein-protein interaction data to this literature-based signaling map in combination with topological analyses allowed us to identify 70 proteins able to increase the compactness of the map. These proteins represent experimentally testable hypotheses for gaining new knowledge on gastrin- and cholecystokinin receptor signaling. The CCKR map is freely available both in a downloadable, machine-readable SBML-compatible format and as a web resource through PAYAO (http://sblab.celldesigner.org:18080/Payao11/bin/). Conclusion We have demonstrated how a literature-based CCKR signaling map together with its protein interaction extensions can be analyzed to generate new hypotheses on molecular mechanisms involved in gastrin- and cholecystokinin-mediated regulation of cellular processes. Electronic supplementary material The online version of this article (doi:10.1186/s12918-015-0181-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sushil Tripathi
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), N-7489, Trondheim, Norway.
| | - Åsmund Flobak
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), N-7489, Trondheim, Norway.
| | - Konika Chawla
- Department of Biology, Norwegian University of Science and Technology (NTNU), N-7491, Trondheim, Norway.
| | - Anaïs Baudot
- I2M, Marseilles Institute of Mathematics CNRS - AMU, Case 907, 13288, Marseille, Cedex 9, France.
| | - Torunn Bruland
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), N-7489, Trondheim, Norway.
| | - Liv Thommesen
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), N-7489, Trondheim, Norway. .,Department of Technology, Sør-Trøndelag University College, N-7004, Trondheim, Norway.
| | - Martin Kuiper
- Department of Biology, Norwegian University of Science and Technology (NTNU), N-7491, Trondheim, Norway.
| | - Astrid Lægreid
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), N-7489, Trondheim, Norway. .,Institute of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), N-7489, Trondheim, Norway.
| |
Collapse
|
40
|
Kuperstein I, Bonnet E, Nguyen HA, Cohen D, Viara E, Grieco L, Fourquet S, Calzone L, Russo C, Kondratova M, Dutreix M, Barillot E, Zinovyev A. Atlas of Cancer Signalling Network: a systems biology resource for integrative analysis of cancer data with Google Maps. Oncogenesis 2015; 4:e160. [PMID: 26192618 PMCID: PMC4521180 DOI: 10.1038/oncsis.2015.19] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 05/29/2015] [Accepted: 06/08/2015] [Indexed: 02/07/2023] Open
Abstract
Cancerogenesis is driven by mutations leading to aberrant functioning of a complex network of molecular interactions and simultaneously affecting multiple cellular functions. Therefore, the successful application of bioinformatics and systems biology methods for analysis of high-throughput data in cancer research heavily depends on availability of global and detailed reconstructions of signalling networks amenable for computational analysis. We present here the Atlas of Cancer Signalling Network (ACSN), an interactive and comprehensive map of molecular mechanisms implicated in cancer. The resource includes tools for map navigation, visualization and analysis of molecular data in the context of signalling network maps. Constructing and updating ACSN involves careful manual curation of molecular biology literature and participation of experts in the corresponding fields. The cancer-oriented content of ACSN is completely original and covers major mechanisms involved in cancer progression, including DNA repair, cell survival, apoptosis, cell cycle, EMT and cell motility. Cell signalling mechanisms are depicted in detail, together creating a seamless ‘geographic-like' map of molecular interactions frequently deregulated in cancer. The map is browsable using NaviCell web interface using the Google Maps engine and semantic zooming principle. The associated web-blog provides a forum for commenting and curating the ACSN content. ACSN allows uploading heterogeneous omics data from users on top of the maps for visualization and performing functional analyses. We suggest several scenarios for ACSN application in cancer research, particularly for visualizing high-throughput data, starting from small interfering RNA-based screening results or mutation frequencies to innovative ways of exploring transcriptomes and phosphoproteomes. Integration and analysis of these data in the context of ACSN may help interpret their biological significance and formulate mechanistic hypotheses. ACSN may also support patient stratification, prediction of treatment response and resistance to cancer drugs, as well as design of novel treatment strategies.
Collapse
Affiliation(s)
- I Kuperstein
- 1] Institut Curie, Paris, France [2] INSERM, U900, Paris, France [3] Mines ParisTech, Fontainebleau, France
| | - E Bonnet
- 1] Institut Curie, Paris, France [2] INSERM, U900, Paris, France [3] Mines ParisTech, Fontainebleau, France
| | - H-A Nguyen
- 1] Institut Curie, Paris, France [2] INSERM, U900, Paris, France [3] Mines ParisTech, Fontainebleau, France
| | - D Cohen
- 1] Institut Curie, Paris, France [2] INSERM, U900, Paris, France [3] Mines ParisTech, Fontainebleau, France
| | | | - L Grieco
- 1] Institut Curie, Paris, France [2] INSERM, U900, Paris, France [3] Mines ParisTech, Fontainebleau, France [4] Ecole Normale Supérieure, IBENS, Paris, France [5] CNRS, UMR8197, Paris, France [6] INSERM, U1024, Paris, France
| | - S Fourquet
- 1] Institut Curie, Paris, France [2] INSERM, U900, Paris, France [3] Mines ParisTech, Fontainebleau, France
| | - L Calzone
- 1] Institut Curie, Paris, France [2] INSERM, U900, Paris, France [3] Mines ParisTech, Fontainebleau, France
| | - C Russo
- 1] Institut Curie, Paris, France [2] INSERM, U900, Paris, France [3] Mines ParisTech, Fontainebleau, France
| | - M Kondratova
- 1] Institut Curie, Paris, France [2] INSERM, U900, Paris, France [3] Mines ParisTech, Fontainebleau, France
| | - M Dutreix
- 1] Institut Curie, Paris, France [2] CNRS, UMR3347, Orsay, France [3] INSERM, U1021, Orsay, France
| | - E Barillot
- 1] Institut Curie, Paris, France [2] INSERM, U900, Paris, France [3] Mines ParisTech, Fontainebleau, France
| | - A Zinovyev
- 1] Institut Curie, Paris, France [2] INSERM, U900, Paris, France [3] Mines ParisTech, Fontainebleau, France
| |
Collapse
|
41
|
Pyysalo S, Ohta T, Rak R, Rowley A, Chun HW, Jung SJ, Choi SP, Tsujii J, Ananiadou S. Overview of the Cancer Genetics and Pathway Curation tasks of BioNLP Shared Task 2013. BMC Bioinformatics 2015; 16 Suppl 10:S2. [PMID: 26202570 PMCID: PMC4511510 DOI: 10.1186/1471-2105-16-s10-s2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Since their introduction in 2009, the BioNLP Shared Task events have been instrumental in advancing the development of methods and resources for the automatic extraction of information from the biomedical literature. In this paper, we present the Cancer Genetics (CG) and Pathway Curation (PC) tasks, two event extraction tasks introduced in the BioNLP Shared Task 2013. The CG task focuses on cancer, emphasizing the extraction of physiological and pathological processes at various levels of biological organization, and the PC task targets reactions relevant to the development of biomolecular pathway models, defining its extraction targets on the basis of established pathway representations and ontologies. RESULTS Six groups participated in the CG task and two groups in the PC task, together applying a wide range of extraction approaches including both established state-of-the-art systems and newly introduced extraction methods. The best-performing systems achieved F-scores of 55% on the CG task and 53% on the PC task, demonstrating a level of performance comparable to the best results achieved in similar previously proposed tasks. CONCLUSIONS The results indicate that existing event extraction technology can generalize to meet the novel challenges represented by the CG and PC task settings, suggesting that extraction methods are capable of supporting the construction of knowledge bases on the molecular mechanisms of cancer and the curation of biomolecular pathway models. The CG and PC tasks continue as open challenges for all interested parties, with data, tools and resources available from the shared task homepage.
Collapse
Affiliation(s)
- Sampo Pyysalo
- Department of Information technology, University of Turku, Turku, Finland
| | | | - Rafal Rak
- National Centre for Text Mining and School of Computer Science, University of Manchester, Manchester, UK
| | - Andrew Rowley
- National Centre for Text Mining and School of Computer Science, University of Manchester, Manchester, UK
| | - Hong-Woo Chun
- Software Research Center, Korea Institute of Science and Technology Information (KISTI), Daejeon, South Korea
| | - Sung-Jae Jung
- Software Research Center, Korea Institute of Science and Technology Information (KISTI), Daejeon, South Korea
- Department of Applied Information Science, University of Science and Technology (UST), Daejeon, South Korea
| | - Sung-Pil Choi
- Department of Library and Information Science, Kyonggi University, Suwon, South Korea
| | | | - Sophia Ananiadou
- National Centre for Text Mining and School of Computer Science, University of Manchester, Manchester, UK
| |
Collapse
|
42
|
Bonnet E, Viara E, Kuperstein I, Calzone L, Cohen DPA, Barillot E, Zinovyev A. NaviCell Web Service for network-based data visualization. Nucleic Acids Res 2015; 43:W560-5. [PMID: 25958393 PMCID: PMC4489283 DOI: 10.1093/nar/gkv450] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/24/2015] [Indexed: 12/17/2022] Open
Abstract
Data visualization is an essential element of biological research, required for obtaining insights and formulating new hypotheses on mechanisms of health and disease. NaviCell Web Service is a tool for network-based visualization of ‘omics’ data which implements several data visual representation methods and utilities for combining them together. NaviCell Web Service uses Google Maps and semantic zooming to browse large biological network maps, represented in various formats, together with different types of the molecular data mapped on top of them. For achieving this, the tool provides standard heatmaps, barplots and glyphs as well as the novel map staining technique for grasping large-scale trends in numerical values (such as whole transcriptome) projected onto a pathway map. The web service provides a server mode, which allows automating visualization tasks and retrieving data from maps via RESTful (standard HTTP) calls. Bindings to different programming languages are provided (Python and R). We illustrate the purpose of the tool with several case studies using pathway maps created by different research groups, in which data visualization provides new insights into molecular mechanisms involved in systemic diseases such as cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Eric Bonnet
- Institut Curie, 26 rue d'Ulm, 75248 Paris, France INSERM, U900, 75248 Paris, France Mines ParisTech, 77300 Fontainebleau, France
| | | | - Inna Kuperstein
- Institut Curie, 26 rue d'Ulm, 75248 Paris, France INSERM, U900, 75248 Paris, France Mines ParisTech, 77300 Fontainebleau, France
| | - Laurence Calzone
- Institut Curie, 26 rue d'Ulm, 75248 Paris, France INSERM, U900, 75248 Paris, France Mines ParisTech, 77300 Fontainebleau, France
| | - David P A Cohen
- Institut Curie, 26 rue d'Ulm, 75248 Paris, France INSERM, U900, 75248 Paris, France Mines ParisTech, 77300 Fontainebleau, France
| | - Emmanuel Barillot
- Institut Curie, 26 rue d'Ulm, 75248 Paris, France INSERM, U900, 75248 Paris, France Mines ParisTech, 77300 Fontainebleau, France
| | - Andrei Zinovyev
- Institut Curie, 26 rue d'Ulm, 75248 Paris, France INSERM, U900, 75248 Paris, France Mines ParisTech, 77300 Fontainebleau, France
| |
Collapse
|
43
|
Matsuoka Y, Funahashi A, Ghosh S, Kitano H. Modeling and simulation using CellDesigner. Methods Mol Biol 2014; 1164:121-45. [PMID: 24927840 DOI: 10.1007/978-1-4939-0805-9_11] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In silico modeling and simulation are effective means to understand how the regulatory systems function in life. In this chapter, we explain how to build a model and run the simulation using CellDesigner, adopting the standards such as SBML and SBGN.
Collapse
Affiliation(s)
- Yukiko Matsuoka
- The Systems Biology Institute, 5-6-9 Shirokanedai, Minato-ku, Tokyo, 108-0071, Japan
| | | | | | | |
Collapse
|
44
|
Kriete A, Noguchi E, Sell C. Introductory review of computational cell cycle modeling. Methods Mol Biol 2014; 1170:267-75. [PMID: 24906317 DOI: 10.1007/978-1-4939-0888-2_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recent advances in the modeling of the cell cycle through computer simulation demonstrate the power of systems biology. By definition, systems biology has the goal to connect a parts list, prioritized through experimental observation or high-throughput screens, by the topology of interactions defining intracellular networks to predict system function. Computer modeling of biological systems is often compared to a process of reverse engineering. Indeed, designed or engineered technical systems share many systems-level properties with biological systems; thus studying biological systems within an engineering framework has proven successful. Here we review some aspects of this process as it pertains to cell cycle modeling.
Collapse
Affiliation(s)
- Andres Kriete
- School of Biomedical Engineering, Science and Health Systems, Bossone Research Center, Drexel University, 3141 Chestnut Street, Philadelphia, PA, 19104, USA,
| | | | | |
Collapse
|
45
|
Niarakis A, Bounab Y, Grieco L, Roncagalli R, Hesse AM, Garin J, Malissen B, Daëron M, Thieffry D. Computational modeling of the main signaling pathways involved in mast cell activation. Curr Top Microbiol Immunol 2014; 382:69-93. [PMID: 25116096 DOI: 10.1007/978-3-319-07911-0_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A global and rigorous understanding of the signaling pathways and cross-regulatory processes involved in mast cell activation requires the integration of published information with novel functional datasets into a comprehensive computational model. Based on an exhaustive curation of the existing literature and using the software CellDesigner, we have built and annotated a comprehensive molecular map for the FcεRI signaling network. This map can be used to visualize and interpret high-throughput expression data. Furthermore, leaning on this map and using the logical modeling software GINsim, we have derived a qualitative dynamical model, which recapitulates the most salient features of mast cell activation. The resulting logical model can be used to explore the dynamical properties of the system and its responses to different stimuli, in normal or mutant conditions.
Collapse
Affiliation(s)
- Anna Niarakis
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Fisher CP, Plant NJ, Moore JB, Kierzek AM. QSSPN: dynamic simulation of molecular interaction networks describing gene regulation, signalling and whole-cell metabolism in human cells. Bioinformatics 2013; 29:3181-90. [PMID: 24064420 PMCID: PMC3842758 DOI: 10.1093/bioinformatics/btt552] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 09/03/2013] [Accepted: 09/18/2013] [Indexed: 12/11/2022] Open
Abstract
MOTIVATION Dynamic simulation of genome-scale molecular interaction networks will enable the mechanistic prediction of genotype-phenotype relationships. Despite advances in quantitative biology, full parameterization of whole-cell models is not yet possible. Simulation methods capable of using available qualitative data are required to develop dynamic whole-cell models through an iterative process of modelling and experimental validation. RESULTS We formulate quasi-steady state Petri nets (QSSPN), a novel method integrating Petri nets and constraint-based analysis to predict the feasibility of qualitative dynamic behaviours in qualitative models of gene regulation, signalling and whole-cell metabolism. We present the first dynamic simulations including regulatory mechanisms and a genome-scale metabolic network in human cell, using bile acid homeostasis in human hepatocytes as a case study. QSSPN simulations reproduce experimentally determined qualitative dynamic behaviours and permit mechanistic analysis of genotype-phenotype relationships. AVAILABILITY AND IMPLEMENTATION The model and simulation software implemented in C++ are available in supplementary material and at http://sysbio3.fhms.surrey.ac.uk/qsspn/.
Collapse
Affiliation(s)
- Ciarán P Fisher
- Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, Surrey GU2 7XH, UK
| | | | | | | |
Collapse
|
47
|
Wierstra I. The transcription factor FOXM1 (Forkhead box M1): proliferation-specific expression, transcription factor function, target genes, mouse models, and normal biological roles. Adv Cancer Res 2013; 118:97-398. [PMID: 23768511 DOI: 10.1016/b978-0-12-407173-5.00004-2] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
FOXM1 (Forkhead box M1) is a typical proliferation-associated transcription factor, which stimulates cell proliferation and exhibits a proliferation-specific expression pattern. Accordingly, both the expression and the transcriptional activity of FOXM1 are increased by proliferation signals, but decreased by antiproliferation signals, including the positive and negative regulation by protooncoproteins or tumor suppressors, respectively. FOXM1 stimulates cell cycle progression by promoting the entry into S-phase and M-phase. Moreover, FOXM1 is required for proper execution of mitosis. Accordingly, FOXM1 regulates the expression of genes, whose products control G1/S-transition, S-phase progression, G2/M-transition, and M-phase progression. Additionally, FOXM1 target genes encode proteins with functions in the execution of DNA replication and mitosis. FOXM1 is a transcriptional activator with a forkhead domain as DNA binding domain and with a very strong acidic transactivation domain. However, wild-type FOXM1 is (almost) inactive because the transactivation domain is repressed by three inhibitory domains. Inactive FOXM1 can be converted into a very potent transactivator by activating signals, which release the transactivation domain from its inhibition by the inhibitory domains. FOXM1 is essential for embryonic development and the foxm1 knockout is embryonically lethal. In adults, FOXM1 is important for tissue repair after injury. FOXM1 prevents premature senescence and interferes with contact inhibition. FOXM1 plays a role for maintenance of stem cell pluripotency and for self-renewal capacity of stem cells. The functions of FOXM1 in prevention of polyploidy and aneuploidy and in homologous recombination repair of DNA-double-strand breaks suggest an importance of FOXM1 for the maintenance of genomic stability and chromosomal integrity.
Collapse
|
48
|
Kuperstein I, Cohen DPA, Pook S, Viara E, Calzone L, Barillot E, Zinovyev A. NaviCell: a web-based environment for navigation, curation and maintenance of large molecular interaction maps. BMC SYSTEMS BIOLOGY 2013; 7:100. [PMID: 24099179 PMCID: PMC3851986 DOI: 10.1186/1752-0509-7-100] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 09/20/2013] [Indexed: 11/24/2022]
Abstract
Background Molecular biology knowledge can be formalized and systematically represented in a computer-readable form as a comprehensive map of molecular interactions. There exist an increasing number of maps of molecular interactions containing detailed and step-wise description of various cell mechanisms. It is difficult to explore these large maps, to organize discussion of their content and to maintain them. Several efforts were recently made to combine these capabilities together in one environment, and NaviCell is one of them. Results NaviCell is a web-based environment for exploiting large maps of molecular interactions, created in CellDesigner, allowing their easy exploration, curation and maintenance. It is characterized by a combination of three essential features: (1) efficient map browsing based on Google Maps; (2) semantic zooming for viewing different levels of details or of abstraction of the map and (3) integrated web-based blog for collecting community feedback. NaviCell can be easily used by experts in the field of molecular biology for studying molecular entities of interest in the context of signaling pathways and crosstalk between pathways within a global signaling network. NaviCell allows both exploration of detailed molecular mechanisms represented on the map and a more abstract view of the map up to a top-level modular representation. NaviCell greatly facilitates curation, maintenance and updating the comprehensive maps of molecular interactions in an interactive and user-friendly fashion due to an imbedded blogging system. Conclusions NaviCell provides user-friendly exploration of large-scale maps of molecular interactions, thanks to Google Maps and WordPress interfaces, with which many users are already familiar. Semantic zooming which is used for navigating geographical maps is adopted for molecular maps in NaviCell, making any level of visualization readable. In addition, NaviCell provides a framework for community-based curation of maps.
Collapse
|
49
|
Matsuoka Y, Matsumae H, Katoh M, Eisfeld AJ, Neumann G, Hase T, Ghosh S, Shoemaker JE, Lopes TJS, Watanabe T, Watanabe S, Fukuyama S, Kitano H, Kawaoka Y. A comprehensive map of the influenza A virus replication cycle. BMC SYSTEMS BIOLOGY 2013; 7:97. [PMID: 24088197 PMCID: PMC3819658 DOI: 10.1186/1752-0509-7-97] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/24/2013] [Indexed: 02/05/2023]
Abstract
Background Influenza is a common infectious disease caused by influenza viruses. Annual epidemics cause severe illnesses, deaths, and economic loss around the world. To better defend against influenza viral infection, it is essential to understand its mechanisms and associated host responses. Many studies have been conducted to elucidate these mechanisms, however, the overall picture remains incompletely understood. A systematic understanding of influenza viral infection in host cells is needed to facilitate the identification of influential host response mechanisms and potential drug targets. Description We constructed a comprehensive map of the influenza A virus (‘IAV’) life cycle (‘FluMap’) by undertaking a literature-based, manual curation approach. Based on information obtained from publicly available pathway databases, updated with literature-based information and input from expert virologists and immunologists, FluMap is currently composed of 960 factors (i.e., proteins, mRNAs etc.) and 456 reactions, and is annotated with ~500 papers and curation comments. In addition to detailing the type of molecular interactions, isolate/strain specific data are also available. The FluMap was built with the pathway editor CellDesigner in standard SBML (Systems Biology Markup Language) format and visualized as an SBGN (Systems Biology Graphical Notation) diagram. It is also available as a web service (online map) based on the iPathways+ system to enable community discussion by influenza researchers. We also demonstrate computational network analyses to identify targets using the FluMap. Conclusion The FluMap is a comprehensive pathway map that can serve as a graphically presented knowledge-base and as a platform to analyze functional interactions between IAV and host factors. Publicly available webtools will allow continuous updating to ensure the most reliable representation of the host-virus interaction network. The FluMap is available at http://www.influenza-x.org/flumap/.
Collapse
Affiliation(s)
- Yukiko Matsuoka
- JST ERATO Kawaoka infection-induced host responses project, Minato-ku, Tokyo 108-8639, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Gupta MK, Singh DB, Shukla R, Misra K. A comprehensive metabolic modeling of thyroid pathway in relation to thyroid pathophysiology and therapeutics. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2013; 17:584-93. [PMID: 24044365 DOI: 10.1089/omi.2013.0007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The thyroid pathway represents a complex interaction of different glands for thyroid hormone synthesis. Thyrotropin releasing hormone is synthesized in the hypothalamus and regulates thyrotropin stimulating hormone gene expression in the pituitary gland. In order to understand the complexity of the thyroid pathways, and using experimental data retrieved from the biomedical literature (e.g., NCBI, HuGE Navigator, Protein Data Bank, and KEGG), we constructed a metabolic map of the thyroid hormone pathway at a molecular level and analyzed it topologically. A total of five hub nodes were predicted in regards to the transcription thyroid receptor (TR), cAMP response element-binding protein (CREB), signal transducer and activator of transcription 3 (STAT 3), nuclear factor kappa-light-chain-enhancer of activated B cells (NFkB), and activator protein 1 (AP-1) as being potentially important in study of thyroid disorders and as novel putative therapeutic drug targets. Notably, the thyroid receptor is a highly connected hub node and currently used as a therapeutic target in hypothyroidism. Our analysis represents the first comprehensive description of the thyroid pathway, which pertains to understanding the function of the protein and gene interaction networks. The findings from this study are therefore informative for pathophysiology and rational therapeutics of thyroid disorders.
Collapse
Affiliation(s)
- Manish Kumar Gupta
- 1 Department of Bioinformatics, University Institute of Engineering and Technology, Chhatrapati Shahu Ji Maharaj University, Kanpur, 208024, Uttar Pradesh, India
| | | | | | | |
Collapse
|