2,756
Views
14
CrossRef citations to date
0
Altmetric
Review

Unraveling cellular pathways contributing to drug-induced liver injury by dynamical modeling

, , &
Pages 5-17 | Received 24 Jun 2016, Accepted 02 Sep 2016, Published online: 23 Sep 2016

References

  • Kaplowitz N. Drug-Induced Liver Injury. Clin Infect Dis. 2004;38:S44–S48.
  • Lee WM. Acute liver failure in the United States. Ann Intern Med. 2003;23:217–226.
  • Fung M, Thornton A, Mybeck K, et al. evaluation of the characteristics of safety withdrawal of prescription drugs from worldwide pharmaceutical markets-1960 to 1999. Drug Inf J. 2001;35:293–317.
  • Hussaini SH, Farrington E. Idiosyncratic drug-induced liver injury: an overview. Expert Opin Drug Saf. 2007;6:673–684.
  • Wink S, Hiemstra S, Huppelschoten S, et al. Quantitative High Content Imaging of Cellular Adaptive Stress Response Pathways in Toxicity for Chemical Safety Assessment. Chem Res Toxicol. 2014;27(3):338–355.
  • National Research Council. Toxicity testing in the 21st century: a vision and a strategy. Vol. 2007. Washington, DC: National Academy of Sciences; 2007.
  • Vinken M. Adverse outcome pathways and drug-induced liver injury testing. Chem Res Toxicol. 2015;28:1391–1397.
  • Bhattacharya S, Shoda LKM, Zhang Q, et al. Modeling drug- and chemical-induced hepatotoxicity with systems biology approaches. Front Physiol. 2012;3:1–18.
  • Przybylak KR, Cronin MT. In silico models for drug-induced liver injury – current status. Expert Opin Drug Metab Toxicol. 2012;8:201–217.
  • Chen M, Bisgin H, Tong L, et al. Toward predictive models for drug-induced liver injury in humans: are we there yet? Biomark Med. 2014;8:201–213.
  • Wink S, Hiemstra S, Herpers B, et al. High-content imaging-based BAC-GFP toxicity pathway reporters to assess chemical adversity liabilities. Arch Toxicol. 2016. doi: 10.1007/s00204-016-1781-0. [Epub ahead of print]
  • OECD. Guidance document on developing and accessing adverse outcome pathways. Series on testing and assessment. Vol. 2013. Paris-France: OECD Publications Office; 2013. p. 1–45.
  • Ekins S, Mestres J, Testa B. In silico pharmacology for drug discovery: methods for virtual ligand screening and profiling. Br J Pharmacol. 2007;152:9–20.
  • Danhof M, Ecm DL, Della Pasqua OE, et al. Mechanism-based pharmacokinetic-pharmacodynamic (PK-PD) modeling in translational drug research. Trends Pharmacol Sci. 2008;29:186–191.
  • Beard DA, Qian H. Chemical biophysics: quantitative analysis of cellular systems. Cambridge: Cambridge University Press, 2008.
  • Keener J, Sneyd J. Mathematical physiology: I: cellular physiology. New York: Springer Science + Business Media, 2009.
  • Van Riel NAW. Dynamic modelling and analysis of biochemical networks: mechanism-based models and model-based experiments. Brief Bioinform. 2006;7:364–374.
  • Kitano H. Systems biology: a brief overview. Science. 2002;295:1662–1665.
  • Kitano H. Computational systems biology. Nature. 2002;420:206–210.
  • Zhang Q, Andersen ME. Dose response relationship in anti-stress gene regulatory networks. PLoS Comput Biol. 2007;3:0345–63.
  • Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007;35:495–516.
  • Leclerc E, Hamon J, Claude I, et al. Investigation of acetaminophen toxicity in HepG2/C3a microscale cultures using a system biology model of glutathione depletion. Cell Biol Toxicol. 2015;31:173–185.
  • Peper A, Grimbergen CA, Spaan JAE, et al. A mathematical model of the hsp70 regulation in the cell. Int J Hyperthermia. 1998;14:97–124.
  • Erguler K, Pieri M, Deltas C. A mathematical model of the unfolded protein stress response reveals the decision mechanism for recovery, adaptation and apoptosis. BMC Syst Biol. 2013;7:16.
  • Dolan DWP, Zupanic A, Nelson G, et al. Integrated stochastic model of DNA damage repair by non-homologous end joining and p53/p21-mediated early senescence signalling. PLoS Comput Biol. 2015;11:1–19.
  • Han D, Shinohara M, Ybanez MD, et al. Signal transduction pathways involved in drug-induced liver injury derick. Adverse Drug Reactions, Handb Exp Pharmacol. 2010;196:267–310.
  • Bryan HK, Olayanju A, Goldring CE, et al. The Nrf2 cell defence pathway: Keap1-dependent and -independent mechanisms of regulation. Biochem Pharmacol. 2013;85:705–717.
  • Zhang Q, Pi J, Woods CG, et al. Phase I to II cross-induction of xenobiotic metabolizing enzymes: a feedforward control mechanism for potential hormetic responses. Toxicol Appl Pharmacol. 2009;237:345–356.
  • Zhang Q, Pi J, Woods CG, et al. A systems biology perspective on Nrf2-mediated antioxidant response. Toxicol Appl Pharmacol. 2010;244:84–97.
  • Hamon J, Jennings P, Bois FY. Systems biology modeling of omics data: effect of cyclosporine a on the Nrf2 pathway in human renal cells. BMC Syst Biol. 2014;8:76.
  • Wilmes A, Limonciel A, Aschauer L, et al. Application of integrated transcriptomic, proteomic and metabolomic profiling for the delineation of mechanisms of drug induced cell stress. J Proteomics. 2013;79:180–194.
  • Chen X, Chen J, Gan S, et al. DNA damage strength modulates a bimodal switch of p53 dynamics for cell-fate control. BMC Biol. 2013;11:73.
  • Chen M, Suzuki A, Borlak J, et al. Drug-induced liver injury: interactions between drug properties and host factors. J Hepatol. 2015;63:503–514.
  • Newton K, Dixit VM. Signaling in innate immunity and inflammation. Cold Spring Harb Perspect Biol. 2012;4:1–19.
  • Ghosh S, May MJ, Kopp EB. NF-kappaB AND REL PROTEINS: evolutionarily conserved mediators of immune responses. Annu Rev Imunol. 1998;16:225–260.
  • Williams RA, Timmis J, Qwarnstrom EE. Computational models of the NF-KB signalling pathway. Computation. 2014;2:131–158.
  • Williams RA, Timmis J, Qwarnstrom EE. The rise in computational systems biology approaches for understanding NF-kappaB signaling dynamics. Sci Signal. 2015;8:13–15.
  • Carlotti F, Dower SK, Qwarnstrom EE. Dynamic shuttling of nuclear factor kappaB between the nucleus and cytoplasm as a consequence of inhibitor dissociation. J Biol Chem. 2000;275:41028–41034.
  • Hoffmann A, Levchenko A, Scott ML, et al. The IkappaB-NF-kappaB signaling module: temporal control and selective gene activation. Science. 2002;298:1241–1245.
  • Tsui R, Kearns JD, Lynch C, et al. IkappaBbeta enhances the generation of the low-affinity NF-kappaB/RelA homodimer. Nat Commun. 2015;6:7068.
  • Junkin M, Kaestli Alicia J, Cheng Z, et al. High-content quantification of single-cell immune dynamics. Cell Rep. 2016;15:411–422.
  • Lee TK, Denny EM, Sanghvi JC, et al. A noisy paracrine signal determines the cellular NF-kappaB response to lipopolysaccharide. Sci Signal. 2009;2:ra65.
  • Tay S, Hughey JJ, Lee TK, et al. Single-cell NF-kappaB dynamics reveal digital activation and analogue information processing. Nature. 2010;466:267–271.
  • Turner D, Paszek P, Woodcock DJ, et al. Physiological levels of TNFalpha stimulation induce stochastic dynamics of NF-kappaB responses in single living cells. J Cell Sci. 2010;123:2834–2843.
  • Krishna S, Jensen MH, Sneppen K. Minimal model of spiky oscillations in NF-kappaB signaling. Proc Natl Acad Sci U S A. 2006;103:10840–10845.
  • Yde P, Mengel B, Jensen MH, et al. Modeling the NF-kappaB mediated inflammatory response predicts cytokine waves in tissue. BMC Syst Biol. 2011;5:115.
  • Longo DM, Selimkhanov J, Kearns JD, et al. Dual delayed feedback provides sensitivity and robustness to the NF-kB signaling module. PLoS Comput Biol. 2013;9:e1003112.
  • Zambrano S, Bianchi ME, Agresti A. A simple model of NF-kappaB dynamics reproduces experimental observations. J Theor Biol. 2014;347:44–53.
  • West S, Bridge LJ, White MRH, et al. A method of ‘speed coefficients’ for biochemical model reduction applied to the NF-kappaB system. J Math Biol. 2014;70(3):591–620.
  • Ramadori G, Armbrust T. Cytokines in the liver. Eur J Gastroenterol Hepatol. 2001;13:777–784.
  • Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8:519–529.
  • Brown MK, Naidoo N. The endoplasmic reticulum stress response in aging and age-related diseases. Front Physiol. 2012;3:263.
  • Dara L, Ji C, Kaplowitz N. The contribution of endoplasmic reticulum stress to liver diseases. Hepatology. 2011;53:1752–1763.
  • Walter P, Ron D. The unfolded protein response: from stress pathway to homeostatic regulation. Science. 2011;334:1081–1086.
  • Yoshida H, Matsui T, Yamamoto A, et al. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell. 2001;107:881–891.
  • Vattem KM, Wek RC. Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells. Proc Natl Acad Sci U S A. 2004;101:11269–11274.
  • Harding HP, Novoa I, Zhang Y, et al. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol Cell. 2000;6:1099–1108.
  • Haze K, Yoshida H, Yanagi H, et al. Mammalian transcription factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis in response to endoplasmic reticulum stress. Mol Biol Cell. 1999;10:3787–3799.
  • Hetz C. The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Biol. 2012;13:89–102.
  • Novoa I, Zeng H, Harding HP, et al. Feedback inhibition of the unfolded protein response by GADD34-mediated dephosphorylation of eIF2alpha. J Cell Biol. 2001;153:1011–1022.
  • Royle K, Kontoravdi C. A systems biology approach to optimising hosts for industrial protein production. Biotechnol Lett. 2013;35:1961–1969.
  • Rutkowski DT, Arnold SM, Miller CN, et al. Adaptation to ER stress is mediated by differential stabilities of pro-survival and pro-apoptotic mRNAs and proteins. PLoS Biol. 2006;4:e374.
  • Pincus D, Chevalier MW, Aragón T, et al. BiP binding to the ER-stress sensor Ire1 tunes the homeostatic behavior of the unfolded protein response. PLoS Biol. 2010;8:e1000415.
  • Trusina A, Papa FR, Tang C. Rationalizing translation attenuation in the network architecture of the unfolded protein response. Proc Natl Acad Sci. 2008;105:20280–20285.
  • Trusina A, Tang C. The unfolded protein response and translation attenuation: a modelling approach. Diabetes Obes Metab. 2010;12:27–31.
  • Schmitz O, Brock B, Hollingdal M, et al. High frequency insulin pulsatility and type 2 diabetes: from physiology and pathophysiology to clinical pharmacology. Diabetes Metab. 2002;28:4S14–4S20.
  • Curtu R, Diedrichs D. Small-scale modeling approach and circuit wiring of the unfolded protein response in mammalian cells. In: Arabnia RH, editor. Advances in computational biology. New York: Springer; 2010. p. 261–274.
  • Little E, Lee AS. Generation of a mammalian cell line deficient in glucose-regulated protein stress induction through targeted ribozyme driven by a stress-inducible promoter. J Biol Chem. 1995;270:9526–9534.
  • Zhang T, Brazhnik P, Tyson JJ. Computational analysis of dynamical responses to the intrinsic pathway of programmed cell death. Biophys J. 2009;97:415–434.
  • Kregel KC. Molecular biology of thermoregulation invited review: heat shock proteins: modifying factors in physiological stress responses and acquired thermotolerance. J Appl Physiol. 2002;92:2177–2186.
  • Wilson AGE. New horizons in predictive toxicology: current status and application. Cambridge: The Royal Society of Chemistry; 2011.
  • Ikeyama S, Kusumoto K, Miyake H, et al. A non-toxic heat shock protein 70 inducer, geranylgeranylacetone, suppresses apoptosis of cultured rat hepatocytes caused by hydrogen peroxide and ethanol. J Hepatol. 2001;35:53–61.
  • McMillan DR, Xiao X, Shao L, et al. Targeted disruption of heat shock transcription factor 1 abolishes thermotolerance and protection against heat-inducible apoptosis. J Biol Chem. 1998;273:7523–7528.
  • Petre I, Mizera A, Hyder CL, et al. A simple mass-action model for the eukaryotic heat shock response and its mathematical validation. Nat Comput. 2011;10:595–612.
  • Scheff JD, Stallings JD, Reifman J, et al. Mathematical modeling of the heat-shock response in HeLa cells. Biophys J. 2015;109:182–193.
  • Akerfelt M, Morimoto RI, Sistonen L. Heat shock factors: integrators of cell stress, development and lifespan. Nat Rev Mol Cell Biol. 2010;11:545–555.
  • Lecomte S, Reverdy L, Le Quément C, et al. Unraveling complex interplay between heat shock factor 1 and 2 splicing isoforms. PLoS One. 2013;8:e56085.
  • Helleday T, Eshtad S, Nik-Zainal S. Mechanisms underlying mutational signatures in human cancers. Nat Rev Genet. 2014;15:585–598.
  • Hussain SP, Hofseth LJ, Harris CC. Radical causes of cancer. Nat Rev Cancer. 2003;3:276–285.
  • Nebert DW, Dalton TP. The role of cytochrome P450 enzymes in endogenous signalling pathways and environmental carcinogenesis. Nat Rev Cancer. 2006;6:947–960.
  • Spassova MA, Miller DJ, Nikolov AS. Kinetic modeling reveals the roles of reactive oxygen species scavenging and DNA repair processes in shaping the dose-response curve of KBrO 3 -induced DNA damage. Oxid Med Cell Longev. 2015;2015:764375.
  • Roos WP, Thomas AD, Kaina B. DNA damage and the balance between survival and death in cancer biology. Nat Rev Cancer. 2015;16:20–33.
  • Lev Bar-Or R, Maya R, Segel LA, et al. Generation of oscillations by the p53-Mdm2 feedback loop: a theoretical and experimental study. Proc Natl Acad Sci U S A. 2000;97:11250–11255.
  • Batchelor E, Loewer A, Mock C, et al. Stimulus-dependent dynamics of p53 in single cells. Mol Syst Biol. 2011;7:488.
  • Batchelor E, Mock CS, Bhan I, et al. Recurrent initiation: a mechanism for triggering p53 pulses in response to DNA damage. Mol Cell. 2008;30:277–289.
  • Cai X, Yuan Z-M. Stochastic modeling and simulation of the p53-MDM2/MDMX loop. J Comput Biol. 2009;16:917–933.
  • Cucinotta FA, Pluth JM, Anderson J, et al. Biochemical kinetics model of DSB repair and induction of gamma-H2AX foci by non-homologous end joining. Radiat Res. 2008;169:214–222.
  • Dolan D, Nelson G, Zupanic A, et al. Systems modelling of NHEJ reveals the importance of redox regulation of Ku70/80 in the dynamics of DNA damage foci. PLoS One. 2013;8:e55190.
  • Eliaš J, Dimitrio L, Clairambault J, et al. The p53 protein and its molecular network: modelling a missing link between DNA damage and cell fate. Biochim Biophys Acta. 2014;1844:232–247.
  • Li Y, Qian H, Wang Y, et al. A stochastic model of DNA fragments rejoining. PLoS One. 2012;7:1–9.
  • Ma L, Wagner J, Rice JJ, et al. A plausible model for the digital response of p53 to DNA damage. Proc Natl Acad Sci U S A. 2005;102:14266–14271.
  • Passos JF, Nelson G, Wang C, et al. Feedback between p21 and reactive oxygen production is necessary for cell senescence. Mol Syst Biol. 2010;6:347.
  • Talemi SR, Kollarovic G, Lapytsko A, et al. Development of a robust DNA damage model including persistent telomere-associated damage with application to secondary cancer risk assessment. Sci Rep. 2015;5:13540.
  • Stracker TH, Roig I, Knobel PA, et al. The ATM signaling network in development and disease. Front Genet. 2013;4:1–19.
  • Sun T, Yang W, Liu J, et al. Modeling the basal dynamics of P53 system. PLoS One. 2011;6:1–9.
  • Taleei R, Nikjoo H. The non-homologous end-joining (NHEJ) pathway for the repair of DNA double-strand breaks: I. Mathematical Model Radiat Res. 2013;179:530–539.
  • Mihalas GI, Simon Z, Balea G, et al. Possible oscillatory behavior in p53-mdm2 interaction computer simulation. J Biol Syst. 2000;8:21–29.
  • Toledo F, Wahl GM. Regulating the p53 pathway: in vitro hypotheses, in vivo veritas. Nat Rev Cancer. 2006;6:909–923.
  • Lahav G, Rosenfeld N, Sigal A, et al. Dynamics of the p53-Mdm2 feedback loop in individual cells. Nat Genet. 2004;36:147–150.
  • Parrish AB, Freel CD, Kornbluth S. Cellular mechanisms controlling caspase activation and function. Cold Spring Harb Perspect Biol. 2013 June 1;5(6):a008672.
  • Würstle ML, Zink E, Prehn JHM, et al. From computational modelling of the intrinsic apoptosis pathway to a systems-based analysis of chemotherapy resistance: achievements, perspectives and challenges in systems medicine. Cell Death Dis. 2014;5:e1258.
  • Schleich K, Lavrik IN. Mathematical modeling of apoptosis. Cell Commun Signal. 2013;11:44.
  • Lavrik IN. Systems biology of apoptosis signaling networks. Curr Opin Biotechnol. 2010;21:551–555.
  • Lavrik IN, Eils R, Fricker N, et al. Understanding apoptosis by systems biology approaches. Mol Biosyst. 2009;5:1105–1111.
  • Huber H, Bullinger E, Rehm M. Systems biology approaches to the study of apoptosis. In: Dong Z, Yin X-M, eds. Essentials of apoptosis: a guide for basic and clinical research. Totowa, NJ: Humana Press; 2009. p. 283–297.
  • Spencer SL, Sorger PK. Measuring and modeling apoptosis in single cells. Cell. 2011;144:926–939.
  • Huber HJ, Duessmann H, Wenus J, et al. Mathematical modelling of the mitochondrial apoptosis pathway. Biochim Biophys Acta. 2011;1813:608–615.
  • Rehm M, Prehn JHM. Systems modelling methodology for the analysis of apoptosis signal transduction and cell death decisions. Methods. 2013;61:165–173.
  • Fussenegger M, Bailey JE, Varner J. A mathematical model of caspase function in apoptosis. Nat Biotechnol. 2000;18:768–774.
  • Eissing T, Conzelmann H, Gilles ED, et al. Bistability analyses of a caspase activation model for receptor-induced apoptosis. J Biol Chem. 2004;279:36892–36897.
  • Chaves M, Eissing T, Allgöwer F. Regulation of apoptosis via the NFκB pathway: modeling and analysis. In: Ganguly N, Deutsch A, Mukherjee A, editors. Dynamics on and of complex networks: applications to biology, computer science, and the social sciences. Boston (MA): Birkhäuser; 2009. p. 19–33.
  • Lipniacki T, Paszek P, Brasier AR, et al. Mathematical model of NF-kappaB regulatory module. J Theor Biol. 2004;228:195–215.
  • Basak S, Behar M, Hoffmann A. Lessons from mathematically modeling the NF-kappaB pathway. Immunol Rev. 2012;246:221–238.
  • Ramaiahgari SC, Den Braver MW, Herpers B, et al. A 3D in vitro model of differentiated HepG2 cell spheroids with improved liver-like properties for repeated dose high-throughput toxicity studies. Arch Toxicol. 2014;88(5):1083–1095.
  • Messner S, Agarkova I, Moritz W, et al. Multi-cell type human liver microtissues for hepatotoxicity testing. Arch Toxicol. 2013;87(1):209–213.
  • Bhatia SN, Ingber DE. Microfluidic organs-on-chips. Nat Biotech. 2014 08//print;32(8):760–772.
  • Wardyn JD, Ponsford AH, Sanderson CM. Dissecting molecular cross-talk between Nrf2 and NF-κB response pathways. Biochem Soc Trans. 2015;43:621–626.
  • Herpers B, Wink S, Fredriksson L, et al. Activation of the Nrf2 response by intrinsic hepatotoxic drugs correlates with suppression of NF-kappaB activation and sensitizes toward TNF??-induced cytotoxicity. Arch Toxicol. 2016;90:1163–1179.
  • Reichard JF, Motz GT, Puga A. Heme oxygenase-1 induction by NRF2 requires inactivation of the transcriptional repressor BACH1. Nucleic Acids Res. 2007;35:7074–7086.