Skip to main content

A Network-Based Integrative Workflow to Unravel Mechanisms Underlying Disease Progression

  • Protocol
  • First Online:
Systems Biology

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1702))

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.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Barabasi A-L, Gulbahce N, Loscalzo J (2011) Network medicine: a network-based approach to human disease. Nat Rev Genetics 12:56–68

    Article  CAS  PubMed  Google Scholar 

  2. Chuang H-Y, Lee E, Liu Y-T et al (2007) Network-based classification of breast cancer metastasis. Mol Syst Biol 3:140

    Article  PubMed  PubMed Central  Google Scholar 

  3. Csermely P, Korcsmáros T, Kiss HJM et al (2013) Structure and dynamics of molecular networks: a novel paradigm of drug discovery: a comprehensive review. Pharmacol Ther 138:333–408

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Alon U (2007) Network motifs: theory and experimental approaches. Nat Rev Genet 8:450–461

    Article  CAS  PubMed  Google Scholar 

  5. Kitano H (2007) A robustness-based approach to systems-oriented drug design. Nat Rev Drug Discov 6:202–210

    Article  CAS  PubMed  Google Scholar 

  6. Wolkenhauer O (2014) Why model? Front Physiol 5:21

    Article  PubMed  PubMed Central  Google Scholar 

  7. Le Novère N (2015) Quantitative and logic modelling of molecular and gene networks. Nat Rev Genet 16:146–158

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Voit EO (2009) A {systems-theoretical} framework for health and disease. Math Biosci 217:11–18

    Article  PubMed  Google Scholar 

  9. Sadeghi M, Ranjbar B, Ganjalikhany MR et al (2016) MicroRNA and transcription factor gene regulatory network analysis reveals key regulatory elements associated with prostate cancer progression. PLoS One 11:e0168760

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Ideker T, Galitski T, Hood L (2001) A new approach to decoding life: systems biology. Annu Rev Genomics Hum Genet 2:343–372

    Article  CAS  PubMed  Google Scholar 

  11. Kitano H (2002) Systems biology: a brief overview. Science (New York, NY) 295:1662–1664

    Article  CAS  Google Scholar 

  12. Funahashi A, Morohashi M, Kitano H et al (2003) CellDesigner: a process diagram editor for gene-regulatory and biochemical networks. Biosilico 1:159–162

    Article  Google Scholar 

  13. Shannon P, Markiel A, Ozier O et al (2003) Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13:2498–2504

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Junker BH, Klukas C, Schreiber F (2006) VANTED: a system for advanced data analysis and visualization in the context of biological networks. BMC Bioinformatics 7:109

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Ashyraliyev M, Fomekong-Nanfack Y, Kaandorp JA et al (2009) Systems biology: parameter estimation for biochemical models. FEBS J 276(4):886–902

    Article  CAS  PubMed  Google Scholar 

  16. Wittig U, Kania R, Golebiewski M et al (2012) SABIO-RK—database for biochemical reaction kinetics. Nucleic Acids Res 40:D790–D796

    Article  CAS  PubMed  Google Scholar 

  17. Li C, Donizelli M, Rodriguez N et al (2010) BioModels database: an enhanced, curated and annotated resource for published quantitative kinetic models. BMC Syst Biol 4:92

    Article  PubMed  PubMed Central  Google Scholar 

  18. Lee W-P, Tzou W-S (2009) Computational methods for discovering gene networks from expression data. Brief Bioinform 10:408–423

    CAS  PubMed  Google Scholar 

  19. Zahiri J, Bozorgmehr JH, Masoudi-Nejad A (2013) Computational prediction of protein–protein interaction networks: algo-rithms and resources. Curr Genomics 14:397–414

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Matsuoka Y, Matsumae H, Katoh M et al (2013) A comprehensive map of the influenza A virus replication cycle. BMC Syst Biol 7:97

    Article  PubMed  PubMed Central  Google Scholar 

  21. Wu G, Zhu L, Dent JE et al (2010) A comprehensive molecular interaction map for rheumatoid arthritis. PLoS One 5:e10137

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Caron E, Ghosh S, Matsuoka Y et al (2010) A comprehensive map of the mTOR signaling network. Mol Syst Biol 6:453

    Article  PubMed  PubMed Central  Google Scholar 

  23. Calzone L, Gelay A, Zinovyev A et al (2008) A comprehensive modular map of molecular interactions in RB/E2F pathway. Mol Syst Biol 4:173

    Article  PubMed  PubMed Central  Google Scholar 

  24. Oda K, Matsuoka Y, Funahashi A et al (2005) A comprehensive pathway map of epidermal growth factor receptor signaling. Mol Syst Biol 1:2005.0010

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Ritz A, Poirel CL, Tegge AN et al (2016) Pathways on demand: automated reconstruction of human signaling networks. Syst Biol Appl 2:16002

    Article  Google Scholar 

  26. Supper J, Spangenberg L, Planatscher H et al (2009) BowTieBuilder: modeling signal transduction pathways. BMC Syst Biol 3:67

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Gursoy A, Keskin O, Nussinov R (2008) Topological properties of protein interaction networks from a structural perspective. Biochem Soc Trans 36:1398–1403

    Article  CAS  PubMed  Google Scholar 

  28. Zhang Z, Zhang J (2009) A big world inside small-world networks. PLoS One 4:e5686

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Jeong H, Mason SP, Barabási A-L et al (2001) Lethality and centrality in protein networks. Nature 411:41–42

    Article  CAS  PubMed  Google Scholar 

  30. Kotlyar M, Fortney K, Jurisica I (2012) Network-based characterization of drug-regulated genes, drug targets, and toxicity. Methods 57:499–507

    Article  CAS  PubMed  Google Scholar 

  31. Mitra K, Carvunis A-R, Ramesh SK et al (2013) Integrative approaches for finding modular structure in biological networks. Nat Rev Genet 14:719–732

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wang J, Lu M, Qiu C et al (2010) TransmiR: a transcription factor-microRNA regulation database. Nucleic Acids Res 38:D119–D122

    Article  CAS  PubMed  Google Scholar 

  33. Oti M, Brunner HG (2007) The modular nature of genetic diseases. Clin Genet 71:1–11

    Article  CAS  PubMed  Google Scholar 

  34. Yeger-Lotem E, Sattath S, Kashtan N et al (2004) Network motifs in integrated cellular networks of transcription–regulation and protein–protein interaction. Proc Natl Acad Sci U S A 101:5934–5939

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Tyson JJ, Novák B (2010) Functional motifs in biochemical reaction networks. Annu Rev Phys Chem 61:219–240

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhang Y, Xuan J, de Los Reyes BG et al (2008) Network motif-based identification of breast cancer susceptibility genes. Conference proceedings: annual international conference of the IEEE engineering in medicine and biology society. IEEE engineering in medicine and biology society annual conference, 2008, pp 5696–5699

    Google Scholar 

  37. Wang X, Gulbahce N, Yu H (2011) Network-based methods for human disease gene prediction. Brief Funct Genomics 10:280–293

    Article  CAS  PubMed  Google Scholar 

  38. Barabási A-L, Oltvai ZN (2004) Network biology: understanding the cell’s functional organization. Nat Rev Genet 5:101–113

    Article  PubMed  CAS  Google Scholar 

  39. Guebel DV, Schmitz U, Wolkenhauer O et al (2012) Analysis of cell adhesion during early stages of colon cancer based on an extended multi-valued logic approach. Mol BioSyst 8:1230–1242

    Article  CAS  PubMed  Google Scholar 

  40. Voit EO (2016) The inner workings of life: vignettes in systems biology. Cambridge University Press, Cambridge, NY

    Book  Google Scholar 

  41. Bezručko BP, Smirnov DA (2010) Extracting knowledge from time series: an introduction to nonlinear empirical modeling. Springer, New York, NY

    Book  Google Scholar 

  42. Vera J, González-Alcón C, Marín-Sanguino A et al (2010) Optimization of biochemical systems through mathematical programming: methods and applications. Comput Oper Res 37:1427–1438

    Article  Google Scholar 

  43. Tyson JJ, Chen KC, Novak B (2003) Sniffers, buzzers, toggles and blinkers: dynamics of regulatory and signaling pathways in the cell. Curr Opin Cell Biol 15:221–231

    Article  CAS  PubMed  Google Scholar 

  44. Zi Z, Klipp E (2007) Constraint-based modeling and kinetic analysis of the smad dependent TGF-β signaling pathway. PLoS One 2:e936

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Raia V, Schilling M, Böhm M et al (2011) Dynamic mathematical modeling of IL13-induced signaling in Hodgkin and primary mediastinal B-cell lymphoma allows prediction of therapeutic targets. Cancer Res 71:693–704

    Article  CAS  PubMed  Google Scholar 

  46. Vera J, Schmitz U, Lai X et al (2013) Kinetic modeling-based detection of genetic signatures that provide chemoresistance via the E2F1-p73/DNp73-miR-205 network. Cancer Res 73:3511–3524

    Article  CAS  PubMed  Google Scholar 

  47. Samaga R, Saez-Rodriguez J, Alexopoulos LG et al (2009) The logic of EGFR/ErbB signaling: theoretical properties and analysis of high-throughput data. PLoS Comput Biol 5:e1000438

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Schlatter R, Philippi N, Wangorsch G et al (2012) Integration of Boolean models exemplified on hepatocyte signal transduction. Brief Bioinform 13:365–376

    Article  PubMed  Google Scholar 

  49. Bornholdt S (2005) Less is more in modeling large genetic networks. Science 310:449–451

    Article  CAS  PubMed  Google Scholar 

  50. Saez-Rodriguez J, Simeoni L, Lindquist JA et al (2007) A logical model provides insights into T cell receptor signaling. PLoS Comput Biol 3:e163

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Schlatter R, Schmich K, Avalos Vizcarra I et al (2009) ON/OFF and beyond—a boolean model of apoptosis. PLoS Comput Biol 5:e1000595

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Saadatpour A, Wang R-S, Liao A et al (2011) Dynamical and structural analysis of a T cell survival network identifies novel candidate therapeutic targets for large granular lymphocyte leukemia. PLoS Comput Biol 7:e1002267

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Chowdhury S, Pradhan RN, Sarkar RR (2013) Structural and logical analysis of a comprehensive Hedgehog signaling pathway to identify alternative drug targets for glioma, colon and pancreatic cancer. PLoS One 8:e69132

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Assmann SM, Albert R (2009) Discrete dynamic modeling with asynchronous update, or how to model complex systems in the absence of quantitative information. Methods Mol Biol (Clifton, NJ) 553:207–225

    Article  CAS  Google Scholar 

  55. Kauffman SA (1969) Metabolic stability and epigenesis in randomly constructed genetic nets. J Theor Biol 22:437–467

    Article  CAS  PubMed  Google Scholar 

  56. Terfve C, Cokelaer T, Henriques D et al (2012) CellNOptR: a flexible toolkit to train protein signaling networks to data using multiple logic formalisms. BMC Syst Biol 6:133

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Helikar T, Kowal B, McClenathan S et al (2012) The cell collective: toward an open and collaborative approach to systems biology. BMC Syst Biol 6:96

    Article  PubMed  PubMed Central  Google Scholar 

  58. Klamt S, Saez-Rodriguez J, Gilles ED (2007) Structural and functional analysis of cellular networks with CellNetAnalyzer. BMC Syst Biol 1:2

    Article  PubMed  PubMed Central  Google Scholar 

  59. Chaouiya C, Naldi A, Thieffry D (2012) Logical modelling of gene regulatory networks with GINsim. Methods Mol Biol (Clifton, NJ) 804:463–479

    Article  CAS  Google Scholar 

  60. Müssel C, Hopfensitz M, Kestler HA (2010) BoolNet—an R package for generation, reconstruction and analysis of Boolean networks. Bioinformatics (Oxford) 26:1378–1380

    Article  CAS  Google Scholar 

  61. Albert I, Thakar J, Li S et al (2008) Boolean network simulations for life scientists. Source Code Biol Med 3:16

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Zheng J, Zhang D, Przytycki PF et al (2010) SimBoolNet—a cytoscape plugin for dynamic simulation of signaling networks. Bioinformatics (Oxford) 26:141–142

    Article  CAS  Google Scholar 

  63. Di Cara A, Garg A, De Micheli G et al (2007) Dynamic simulation of regulatory networks using SQUAD. BMC Bioinformatics. 8:462

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Hinkelmann F, Brandon M, Guang B et al (2011) ADAM: analysis of discrete models of biological systems using computer algebra. BMC Bioinformatics 12:295

    Article  PubMed  PubMed Central  Google Scholar 

  65. Swat M, Kel A, Herzel H (2004) Bifurcation analysis of the regulatory modules of the mammalian G1/S transition. Bioinformatics (Oxford) 20:1506–1511

    Article  CAS  Google Scholar 

  66. Saal LH, Johansson P, Holm K et al (2007) Poor prognosis in carcinoma is associated with a gene expression signature of aberrant PTEN tumor suppressor pathway activity. Proc Natl Acad Sci U S A 104:7564–7569

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Pützer BM, Engelmann D (2013) E2F1 apoptosis counterattacked: evil strikes back. Trends Mol Med 19:89–98

    Article  PubMed  CAS  Google Scholar 

  68. Polager S, Ginsberg D (2009) p53 and E2f: partners in life and death, Nature Reviews. Cancer 9:738–748

    CAS  PubMed  Google Scholar 

  69. Mirschel S, Steinmetz K, Rempel M et al (2009) ProMoT: modular modeling for systems biology. Bioinformatics 25:687–689

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Hennessy BT, Smith DL, Ram PT et al (2005) Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov 4:988–1004

    Article  CAS  PubMed  Google Scholar 

  71. Hallstrom TC, Mori S, Nevins JR (2008) An E2F1-dependent gene expression program that determines the balance between proliferation and cell death. Cancer Cell 13:11–22

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Khan FM, Schmitz U, Nikolov S et al (2014) Hybrid modeling of the crosstalk between signaling and transcriptional networks using ordinary differential equations and multi-valued logic. Biochim Biophys Acta 1844:289–298

    Article  CAS  PubMed  Google Scholar 

  73. Ramachandran S, Liu P, Young AN et al (2005) Loss of HOXC6 expression induces apoptosis in prostate cancer cells. Oncogene 24:188–198

    Article  CAS  PubMed  Google Scholar 

  74. Alfieri R, Bartocci E, Merelli E et al (2011) Modeling the cell cycle: from deterministic models to hybrid systems. Biosystems 105:34–40

    Article  CAS  PubMed  Google Scholar 

  75. Kristensen VN, Lingjaerde OC, Russnes HG et al (2014) Principles and methods of integrative genomic analyses in cancer. Nat Rev Cancer 14:299–313

    Article  CAS  PubMed  Google Scholar 

  76. Storey JD, Tibshirani R (2003) SAM thresholding and false discovery rates for detecting differential gene expression in DNA Microarrays. In: Irizarry RA (ed) The analysis of gene expression data: methods and software. Springer, New York, NY, pp 272–290

    Chapter  Google Scholar 

  77. Kerr MK, Martin M, Churchill G (2000) Analysis of variance for gene expression microarray data. J Comput Biol 7:819–837

    Article  CAS  PubMed  Google Scholar 

  78. Baldi P, Long AD (2001) A Bayesian framework for the analysis of microarray expression data: regularized t-test and statistical inferences of gene changes. Bioinformatics 17:509–519

    Article  CAS  PubMed  Google Scholar 

  79. Wu TD (2001) Analysing gene expression data from DNA microarrays to identify candidate genes. J Pathol 195(1):53–65

    Article  CAS  PubMed  Google Scholar 

  80. Tomczak K, Czerwińska P, Wiznerowicz M (2015) The Cancer Genome Atlas (TCGA): an immeasurable source of knowledge. Contemp Oncol (Pozn) 19(1A):A68–A77

    Google Scholar 

  81. Hudson TJ, Anderson W, Aretz A et al (2010) International network of cancer genome projects. Nature 464:993–998

    Article  CAS  PubMed  Google Scholar 

  82. Siegel RL, Miller KD, Jemal A (2016) Cancer statistics. CA Cancer J Clin 66:7–30

    Article  PubMed  Google Scholar 

  83. Varambally S, Dhanasekaran SM, Zhou M et al (2002) The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature 419:624–629

    Article  CAS  PubMed  Google Scholar 

  84. Barton BE, Karras JG, Murphy TF et al (2004) Signal transducer and activator of transcription 3 (STAT3) activation in prostate cancer: Direct STAT3 inhibition induces apoptosis in prostate cancer lines. Mol Cancer Ther 3:11–20

    Article  CAS  PubMed  Google Scholar 

  85. Abdulghani J, Gu L, Dagvadorj A et al (2008) Stat3 promotes metastatic progression of prostate cancer. Am J Pathol 172:1717–1728

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Nair S, Barve A, Khor T-O et al (2010) Regulation of Nrf2- and AP-1-mediated gene expression by epigallocatechin-3-gallate and sulforaphane in prostate of Nrf2-knockout or C57BL/6J mice and PC-3 AP-1 human prostate cancer cells. Acta Pharmacol Sin 31:1223–1240

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Mehta HH, Gao Q, Galet C et al (2011) IGFBP-3 is a metastasis suppression gene in prostate cancer. Cancer Res 71:5154–5163

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Taylor BS, Schultz N, Hieronymus H et al (2011) Integrative genomic profiling of human prostate cancer. Cancer Cell 18:11–22

    Article  CAS  Google Scholar 

  89. Bengtsson H, Wirapati P, Speed TP (2009) A single-array preprocessing method for estimating full-resolution raw copy numbers from all Affymetrix genotyping arrays including GenomeWideSNP 5 & 6. Bioinformatics 25:2149–2156

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Ritchie ME, Phipson B, Wu D et al (2015) limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 43:e47

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Matys V, Fricke E, Geffers R et al (2003) TRANSFAC: transcriptional regulation, from patterns to profiles. Nucleic Acids Res 31:374–378

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Chou C-H, Chang N-W, Shrestha S et al (2016) miRTarBase 2016: updates to the experimentally validated miRNA-target interactions database. Nucleic Acids Res 44(D1):D239–D247

    Article  CAS  PubMed  Google Scholar 

  93. Betel D, Koppal A, Agius P et al (2010) Comprehensive modeling of microRNA targets predicts functional non-conserved and non-canonical sites. Genome Biol 11:R90

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Agarwal V, Bell GW, Nam J-W et al (2015) Predicting effective microRNA target sites in mammalian mRNAs. eLife 4:PMC4532895

    Article  Google Scholar 

  95. Cai Y, Yu X, Hu S et al (2009) A brief review on the mechanisms of miRNA regulation. Genomics Proteomics Bioinformatics 7:147–154

    Article  CAS  PubMed  Google Scholar 

  96. Lorio MV, Croce CM (2012) MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Mol Med 4:143–159

    Article  CAS  Google Scholar 

  97. Lai X, Schmitz U, Gupta SK et al (2012) Computational analysis of target hub gene repression regulated by multiple and cooperative miRNAs. Nucleic Acids Res 40:8818–8834

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Wang L, Chen S, Xue M et al (2012) Homeobox D10 gene, a candidate tumor suppressor, is downregulated through promoter hypermethylation and associated with gastric carcinogenesis. Mol Med 18:389–400

    CAS  PubMed  Google Scholar 

  99. Yu Y, Liu L, Xie N et al (2013) Expression and function of the progesterone receptor in human prostate stroma provide novel insights to cell proliferation control. J Clin Endocrinol Metab 98:2887–2896

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Bonkhoff H, Fixemer T, Hunsicker I et al (2001) Progesterone receptor expression in human prostate cancer: correlation with tumor progression. Prostate 48:285–291

    Article  CAS  PubMed  Google Scholar 

  101. Li M, Ma H, Yang L et al (2016) Mangiferin inhibition of proliferation and induction of apoptosis in human prostate cancer cells is correlated with downregulation of B-cell lymphoma-2 and upregulation of microRNA-182. Oncol Lett 11:817–822

    PubMed  Google Scholar 

  102. Esber N, Le Billan F, Resche-Rigon M et al (2015) Ulipristal acetate inhibits progesterone receptor isoform A-mediated human breast cancer proliferation and BCl2-L1 expression. PLoS One 10:e0140795

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Yin P, Lin Z, Cheng Y-H et al (2007) Progesterone receptor regulates Bcl-2 gene expression through direct binding to its promoter region in uterine leiomyoma cells. J Clin Endocrinol Metab 92:4459–4466

    Article  CAS  PubMed  Google Scholar 

  104. Horvath CM (2000) STAT proteins and transcriptional responses to extracellular signals. Trends Biochem Sci 25:496–502

    Article  CAS  PubMed  Google Scholar 

  105. Abell K, Watson CJ (2005) The Jak/Stat pathway: a novel way to regulate PI3K activity. Cell Cycle (Georgetown, TX) 4:897–900

    Article  CAS  Google Scholar 

  106. Ho HH, Ivashkiv LB (2006) Role of STAT3 in type I interferon responses: negative regulation of stat1-dependent inflammatory gene activation. J Biol Chem 281:14111–14118

    Article  CAS  PubMed  Google Scholar 

  107. Jochum W, Passegue E, Wagner EF (2001) AP-1 in mouse development and tumorigenesis. Oncogene 20:2401–2412

    Article  CAS  PubMed  Google Scholar 

  108. Karin M, Liu Z, Zandi E (1997) AP-1 function and regulation. Curr Opin Cell Biol 9:240–246

    Article  CAS  PubMed  Google Scholar 

  109. Tu WH, Thomas TZ, Masumori N et al (2003) The loss of TGF-β signaling promotes prostate cancer metastasis. Neoplasia (New York, NY) 5:267–277

    Article  CAS  Google Scholar 

  110. Thomsen MK, Bakiri L, Hasenfuss SC et al (2015) Loss of JUNB/AP-1 promotes invasive prostate cancer. Cell Death Differ 22:574–582

    Article  CAS  PubMed  Google Scholar 

  111. Birner P, Egger G, Merkel O et al (2015) JunB and PTEN in prostate cancer: “loss is nothing else than change”. Cell Death Differ (4):22, 522–523

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Olaf Wolkenhauer .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer Science+Business Media LLC

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Khan, F.M., Sadeghi, M., Gupta, S.K., Wolkenhauer, O. (2018). A Network-Based Integrative Workflow to Unravel Mechanisms Underlying Disease Progression. In: Bizzarri, M. (eds) Systems Biology. Methods in Molecular Biology, vol 1702. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-7456-6_12

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-7456-6_12

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-7455-9

  • Online ISBN: 978-1-4939-7456-6

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics