Skip to main content
Frontiers in Pharmacology logoLink to Frontiers in Pharmacology
. 2018 Aug 20;9:935. doi: 10.3389/fphar.2018.00935

Commentary: Usage of Mitogen-Activated Protein Kinase Small Molecule Inhibitors: More Than Just Inhibition!

Marius Pollet 1, Jean Krutmann 1,2, Thomas Haarmann-Stemmann 1,*
PMCID: PMC6110190  PMID: 30177882

Steffen Meurer and Ralf Weiskirchen recently published an interesting and important study concerning the off-target effects of so-called “specific” protein kinase inhibitors (PKI) that are frequently applied in both, basic research and clinical applications (Meurer and Weiskirchen, 2018). A PKI-dependent inhibition of non-targeted protein kinases may occur due to the usage of concentrations that exceed the respective IC50 value by multiple factors. Meurer and Weiskirchen instead describe a PKI-mediated activation of non-targeted protein kinases. Specifically, the authors observed that a treatment of hepatic stellate cells, hepatocytes and portal myofibroblasts with a chemical inhibitor for a certain mitogen-activated protein kinase (MAPK) led to an activation of other members of the MAPK network. The authors called this phenomenon “activation by inhibition” and “cross-activation” (Meurer and Weiskirchen, 2018), terms which well describe the experimental observations but not the underlying molecular mechanism.

Regarding the latter one, we realized that four of the five MAPK inhibitors tested in the respective study are known to interfere with the activity of the aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor and key regulator of xenobiotic metabolism (Murray et al., 2014). In its inactive form, the AHR is trapped in a cytosolic multiprotein complex. Upon binding of small molecular weight compounds, this complex dissociates and the AHR shuttles in the nucleus, dimerizes with its partner molecule ARNT and induces gene expression (Murray et al., 2014). The probably best-examined AHR target genes encode for the xenobiotic-metabolizing monooxygenases cytochrome P450 (CYP) 1A1, CYP1A2, and CYP1B1, which, in most cases, oxidize the invading chemicals to enhance their polarity and facilitate their excretion (Mescher and Haarmann-Stemmann, 2018). Importantly, the ligand-driven activation of AHR is frequently accompanied by a stimulation of other cellular signaling pathways, including NF-κB, epidermal growth factor receptor (EGFR) and MAPK signal transduction (Haarmann-Stemmann et al., 2009; Puga et al., 2009; Tian, 2009). The list of AHR ligands encompasses infamous environmental pollutants, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and benzo[a]pyrene, plant polyphenols, microbiota-derived indoles and phenazines as well as several pharmaceuticals (Murray et al., 2014). Interestingly, more than a dozen PKI, including the four MAPK inhibitors SB203580, U0126, PD98059, and SP600125 tested by Meurer and Weiskirchen, have been identified to date to modulate AHR activity and downstream gene expression (e.g., of CYP1A1) in either a positive or a negative manner (Table 1). Several PKI, such as the phosphoinositide 3-kinase inhibitor LY294002, bind to the AHR protein and antagonize its activation by the prototype ligand TCDD (Guo et al., 2000), whereas others, including the aforementioned U0126 and SB203580, were shown to interact with AHR and increase its transcriptional activity (Andrieux et al., 2004; Korashy et al., 2011). In this context, it is interesting to know that an activation of AHR by different ligands has been reported to stimulate the phosphorylation of ERK1/2, p38 MAPK, JNK, and upstream receptor tyrosine kinases in various human and rodent cells (Haarmann-Stemmann et al., 2009; Puga et al., 2009). In fact, Fumio Matsumura and his team have been among the first reporting a direct impact of AHR activation on protein kinase activity. Specifically, they observed an increased activity of protein kinase C and EGFR in hepatic tissue of rodents treated with TCDD (Madhukar et al., 1984; Bombick et al., 1985). Further examples for an impact of AHR activation on the signal transduction network, are the TCDD-induced phosphorylation of p38 MAPK observed in hepatoma cells (Weiss et al., 2005) and macrophages (Park et al., 2005), as well as the activation of EGFR and downstream ERK1/2 signaling by the AHR agonists 6-formylindolo[3,2-b]carbazole and TCDD in keratinocytes and colon cancer cells, respectively (Fritsche et al., 2007; Xie et al., 2012). It is thus tempting to speculate that at least some of the PKI-induced off-target effects observed by Meurer and Weiskirchen, such as the phosphorylation of ERK1/2 and JNK by the p38 MAPK inhibitor SB203580 or the activation of JNK and p38 MAPK by the MEK1/2 blocker U0126, were due to a stimulation of AHR activity. One may describe this phenomenon as an effect of a certain PKI on a non-kinase target (Munoz, 2017) or simply as the recognition of a foreign compound by the cellular defense system against xenobiotics. This notion is supported by the fact that some PKI have been found to interact with other xenobiotic receptors as well. The MEK1/2 inhibitor U0126, for instance, was shown to induce the expression of CYP3A4 in human hepatoma cells by binding to the pregnane X receptor (PXR) (Smutny et al., 2014). In addition, five out of nine tested clinically relevant PKI (erlotinib, gefitinib, nilotinib, sorafenib, and vandetanib) induced the expression of the ATP-binding cassette transporter P-glycoprotein in a PXR-dependent manner in human colon cancer cells (Harmsen et al., 2013).

Table 1.

Overview on protein kinase inhibitors known to manipulate AHR signaling.

Inhibitor Target kinase IC50
target kinase [μM]a
AHR
modulation
Tested cells/cell-lines/tissue IC50/EC50 AHR-specific endpoint [μM] References
AG-494 EGFR 1 Inhibitorb Human Caco-2 colon cancer cells Not available Kasai and Kikuchi, 2010
Akti-1/2 AKT1
AKT2
0.05
0.21
Inhibitorc Human MCF-7 breast cancer cells IC50: 5.86 Gilot et al., 2010
LY294002 PI3Kα
PI3Kβ
PI3Kδ
0.5
0.97
0.57
Antagonist Human MCF-10A mammary
epithelial cells
IC50: 35 Guo et al., 2000
PD98059 MEK 2 Antagonist MCF-10A cells IC50: 1–4 Reiners et al., 1998
PP2 SFK Fyn
SFK Hck
SFK Lck
SFK Src
0.005
0.005
0.004
0.1
Agonist Human HepG2 hepatoma cells, human NCTC 2544 keratinocytes Not available Frauenstein et al., 2015
SB203580 p38 0.0003–0.0005 Agonist Murine Hepa1c1c7 hepatoma cells, HepG2 Not available Korashy et al., 2011
SB216763 GSK3 0.034 Partial agonist Hepa1c1c7 cells, murine PW531 hepatoma cells, murine primary hepatocytes Not available Braeuning and Buchmann, 2009
SP600125 JNK1/2
JNK3
0.04
0.09
Antagonist/partial agonist Hepa1c1c7 cells, HepG2 cells, rat liver, human primary hepatocytes IC50: 1.5–7
EC50:
0.005–1.89
Joiakim et al., 2003 Dvorak et al., 2008
STO-609 CaMKKα/β 0.027 Agonist MCF-7 cells, human primary macrophages, human A549 lung cancer cells EC50: 0.043–3.4 Monteiro et al., 2008
SU11248 c-Kit
CSF1R
FGFR1
FLT3
PDGFRα
PDGFRβ
RET
VEGFR1/2
0.001–0.01
0.05–0.1
0.88
0.25
0.069
0.039
0.05
0.004
Inducerc MCF-7 cells Not available Maayah et al., 2013
TSU-16/SU5416 VEGFR2 1.2 Agonist HepG2 cells, human primary hepatocytes, human 101L hepatoma cells, rat 5L hepatoma cells, murine primary splenocytes EC50: 0.007–9.8 Mezrich et al., 2012
Matsuoka-Kawano et al., 2010
TSU-68/SU6668 AURKB
AURKC
FGFR1
PDGFRβ
VEGFR2
0.035
0.21
3
0.06
2.43
Inducerb Human primary hepatocytes,
rat liver
Not available Kitamura et al., 2008a
Kitamura et al., 2008b
U0126 MEK1
MEK2
0.07
0.06
Agonist Human B16A2 hepatoma cells, rat primary hepatocytes EC50: 2.5 Andrieux et al., 2004
a

IC50 values for target kinase(s) and respective references are provided by Cayman Chemical (www.caymanchem.com) and Selleckchem (www.selleckchem.com), respectively. The IC50 values for SU11248 are from Heng and Kollmannsberger (2010).

b

Mode of action not clear.

c

Ligand-independent mode of action.

AURKB, aurora kinase B; AURKC, aurora kinase C; AKT, protein kinase B; CaMKK, calcium/calmodulin-dependent protein kinase kinase; CSF1R, colony stimulating factor 1 receptor; c-kit, mast/stem cell growth factor receptor; EGFR, epidermal growth factor receptor; FGFR, fibroblast growth factor receptor; FLT3, Fms-like tyrosine kinase 3; GSK3, glycogen synthase kinase 3; JNK, c-Jun N-terminal kinase; MEK, mitogen-activated protein kinase kinase; PI3K, phosphoinositide-3-kinase; RET, rearranged during transfection; SFK, Src family kinases; p38 MAPK, p38 mitogen-activated protein kinase; PDGFR, platelet-derived growth factor receptor; VEGFR, vascular endothelial growth factor receptor.

The majority of PKI interact with the ATP-binding cleft of the target enzyme (Bain et al., 2007; Wu et al., 2015). Given that the 3D structure of the ATP-binding cleft is highly conserved amongst eukaryotic protein kinases, these inhibitors are limited in both their size and structural diversity (Bain et al., 2007; Wu et al., 2015). As implied by the growing list of PKI identified to interact with AHR and PXR, the structural prerequisites to bind to the ATP-binding cleft of protein kinases seem to resemble those required to interact with the ligand-binding domain of the xenobiotic receptors. Indeed, AHR and PXR contain a relatively large ligand-binding domain with a cavity volume of ~840 Å3 (Denison et al., 2002) and ~1,150 Å3 (Watkins et al., 2001), respectively, and share an extreme structural diversity of ligands (Denison and Faber, 2017). Notably, the known IC50/EC50 values of some PKI to modulate AHR activity are indeed in the range of the IC50 for their target kinases (Table 1). However, apart from ligand-binding, certain PKI may indirectly interfere with AHR signaling, for instance by inhibiting protein kinases contributing to AHR's nuclear translocation (Haarmann-Stemmann et al., 2009).

We agree with the authors that a detailed knowledge of the off-target effects induced by widely used PKI is urgently required, not only for the proper interpretation of experimental data, but in particular, to better forecast potential drug-drug interactions during therapy. When working with so-called “specific” PKI, one should be aware that these compounds will be at first recognized by the chemical defense system, i.e. by xenobiotic receptors, drug-metabolizing enzymes and drug transporters, of the exposed cells. The interaction with one or more xenobiotic receptor(s) is determined by the structural and physiochemical properties of a given PKI and may not only affect its own metabolism, but also the metabolism of eventually co-administered drugs, and the activity of other signaling pathways that might be tightly interconnected with the addressed chemosensory receptor.

Author contributions

MP performed literature research and created the table. JK revised the manuscript critically for important intellectual content. TH-S performed literature research and wrote the manuscript. All authors approved the final version of the manuscript.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

We thank Melina Mescher for critically reading the manuscript.

References

  1. Andrieux L., Langouët S., Fautrel A., Ezan F., Krauser J. A., Savouret J. F., et al. (2004). Aryl hydrocarbon receptor activation and cytochrome P450 1A induction by the mitogen-activated protein kinase inhibitor U0126 in hepatocytes. Mol. Pharmacol. 65, 934–943. 10.1124/mol.65.4.934 [DOI] [PubMed] [Google Scholar]
  2. Bain J., Plater L., Elliott M., Shpiro N., Hastie C. J., Mclauchlan H., et al. (2007). The selectivity of protein kinase inhibitors: a further update. Biochem. J. 408, 297–315. 10.1042/BJ20070797 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Bombick D. W., Madhukar B. V., Brewster D. W., Matsumura F. (1985). TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) causes increases in protein kinases particularly protein kinase C in the hepatic plasma membrane of the rat and the guinea pig. Biochem. Biophys. Res. Commun. 127, 296–302. 10.1016/S0006-291X(85)80158-3 [DOI] [PubMed] [Google Scholar]
  4. Braeuning A., Buchmann A. (2009). The glycogen synthase kinase inhibitor 3-(2,4-dichlorophenyl)-4-(1-methyl-1H-indol-3-yl)-1H-pyrrole-2,5-dione (SB216763) is a partial agonist of the aryl hydrocarbon receptor. Drug Metab. Dispos. 37, 1576–1580. 10.1124/dmd.109.027821 [DOI] [PubMed] [Google Scholar]
  5. Denison M. S., Faber S. C. (2017). And now for something completely different: diversity in ligand-dependent activation of Ah receptor responses. Curr Opin Toxicol 2, 124–131. 10.1016/j.cotox.2017.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Denison M. S., Pandini A., Nagy S. R., Baldwin E. P., Bonati L. (2002). Ligand binding and activation of the Ah receptor. Chem. Biol. Interact. 141, 3–24. 10.1016/S0009-2797(02)00063-7 [DOI] [PubMed] [Google Scholar]
  7. Dvorak Z., Vrzal R., Henklova P., Jancova P., Anzenbacherova E., Maurel P., et al. (2008). JNK inhibitor SP600125 is a partial agonist of human aryl hydrocarbon receptor and induces CYP1A1 and CYP1A2 genes in primary human hepatocytes. Biochem. Pharmacol. 75, 580–588. 10.1016/j.bcp.2007.09.013 [DOI] [PubMed] [Google Scholar]
  8. Frauenstein K., Tigges J., Soshilov A. A., Kado S., Raab N., Fritsche E., et al. (2015). Activation of the aryl hydrocarbon receptor by the widely used Src family kinase inhibitor 4-amino-5-(4-chlorophenyl)-7-(dimethylethyl)pyrazolo[3,4-d]pyrimidine (PP2). Arch. Toxicol. 89, 1329–1336. 10.1007/s00204-014-1321-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Fritsche E., Schäfer C., Calles C., Bernsmann T., Bernshausen T., Wurm M., et al. (2007). Lightening up the UV response by identification of the arylhydrocarbon receptor as a cytoplasmatic target for ultraviolet B radiation. Proc. Natl. Acad. Sci. U.S.A. 104, 8851–8856. 10.1073/pnas.0701764104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Gilot D., Giudicelli F., Lagadic-Gossmann D., Fardel O. (2010). Akti-1/2, an allosteric inhibitor of Akt 1 and 2, efficiently inhibits CaMKIalpha activity and aryl hydrocarbon receptor pathway. Chem. Biol. Interact. 188, 546–552. 10.1016/j.cbi.2010.08.011 [DOI] [PubMed] [Google Scholar]
  11. Guo M., Joiakim A., Reiners J. J., Jr. (2000). Suppression of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-mediated aryl hydrocarbon receptor transformation and CYP1A1 induction by the phosphatidylinositol 3-kinase inhibitor 2-(4-morpholinyl)-8-phenyl-4H-1- benzopyran-4-one (LY294002). Biochem. Pharmacol. 60, 635–642. 10.1016/S0006-2952(00)00379-8 [DOI] [PubMed] [Google Scholar]
  12. Haarmann-Stemmann T., Bothe H., Abel J. (2009). Growth factors, cytokines and their receptors as downstream targets of arylhydrocarbon receptor (AhR) signaling pathways. Biochem. Pharmacol. 77, 508–520. 10.1016/j.bcp.2008.09.013 [DOI] [PubMed] [Google Scholar]
  13. Harmsen S., Meijerman I., Maas-Bakker R. F., Beijnen J. H., Schellens J. H. (2013). PXR-mediated P-glycoprotein induction by small molecule tyrosine kinase inhibitors. Eur. J. Pharm. Sci. 48, 644–649. 10.1016/j.ejps.2012.12.019 [DOI] [PubMed] [Google Scholar]
  14. Heng D. Y. C., Kollmannsberger C. (2010). Sunitinib in Small Molecules in Oncology ed Martens U. M. (Berlin: Springer; ), 71–82. [Google Scholar]
  15. Joiakim A., Mathieu P. A., Palermo C., Gasiewicz T. A., Reiners J. J., Jr. (2003). The Jun N-terminal kinase inhibitor SP600125 is a ligand and antagonist of the aryl hydrocarbon receptor. Drug Metab. Dispos. 31, 1279–1282. 10.1124/dmd.31.11.1279 [DOI] [PubMed] [Google Scholar]
  16. Kasai S., Kikuchi H. (2010). The inhibitory mechanisms of the tyrosine kinase inhibitors herbimycin a, genistein, and tyrphostin B48 with regard to the function of the aryl hydrocarbon receptor in Caco-2 cells. Biosci. Biotechnol. Biochem. 74, 36–43. 10.1271/bbb.90438 [DOI] [PubMed] [Google Scholar]
  17. Kitamura R., Asanoma H., Nagayama S., Otagiri M. (2008a). Identification of human liver cytochrome P450 isoforms involved in autoinduced metabolism of the antiangiogenic agent (Z)-5-[(1,2-dihydro-2-oxo-3H-indol-3-ylidene)methyl]-2,4-dimethyl-1H-pyrrole-3-pr opanoic acid (TSU-68). Drug Metab. Dispos. 36, 1003–1009. 10.1124/dmd.107.019877 [DOI] [PubMed] [Google Scholar]
  18. Kitamura R., Matsuoka K., Nagayama S., Otagiri M. (2008b). Time-dependent induction of rat hepatic CYP1A1 and CYP1A2 expression after single-dose administration of the anti-angiogenic agent TSU-68. Drug Metab. Pharmacokinet. 23, 421–427. 10.2133/dmpk.23.421 [DOI] [PubMed] [Google Scholar]
  19. Korashy H. M., Anwar-Mohamed A., Soshilov A. A., Denison M. S., El-Kadi A. O. (2011). The p38 MAPK inhibitor SB203580 induces cytochrome P450 1A1 gene expression in murine and human hepatoma cell lines through ligand-dependent aryl hydrocarbon receptor activation. Chem. Res. Toxicol. 24, 1540–1548. 10.1021/tx200141p [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Maayah Z. H., El Gendy M. A., El-Kadi A. O., Korashy H. M. (2013). Sunitinib, a tyrosine kinase inhibitor, induces cytochrome P450 1A1 gene in human breast cancer MCF7 cells through ligand-independent aryl hydrocarbon receptor activation. Arch. Toxicol. 87, 847–856. 10.1007/s00204-012-0996-y [DOI] [PubMed] [Google Scholar]
  21. Madhukar B. V., Brewster D. W., Matsumura F. (1984). Effects of in vivo-administered 2,3,7,8-tetrachlorodibenzo-p-dioxin on receptor binding of epidermal growth factor in the hepatic plasma membrane of rat, guinea pig, mouse, and hamster. Proc. Natl. Acad. Sci. U.S.A. 81, 7407–7411. 10.1073/pnas.81.23.7407 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Matsuoka-Kawano K., Yoshinari K., Nagayama S., Yamazoe Y. (2010). TSU-16, (Z)-3-[(2,4-dimethylpyrrol-5-yl)methylidenyl]-2-indolinone, is a potent activator of aryl hydrocarbon receptor and increases CYP1A1 and CYP1A2 expression in human hepatocytes. Chem. Biol. Interact. 185, 33–41. 10.1016/j.cbi.2010.02.014 [DOI] [PubMed] [Google Scholar]
  23. Mescher M., Haarmann-Stemmann T. (2018). Modulation of CYP1A1 metabolism: from adverse health effects to chemoprevention and therapeutic options. Pharmacol. Ther. 187, 71–87. 10.1016/j.pharmthera.2018.02.012 [DOI] [PubMed] [Google Scholar]
  24. Meurer S. K., Weiskirchen R. (2018). Usage of mitogen-activated protein kinase small molecule inhibitors: more than just inhibition! Front. Pharmacol. 9:98. 10.3389/fphar.2018.00098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Mezrich J. D., Nguyen L. P., Kennedy G., Nukaya M., Fechner J. H., Zhang X., et al. (2012). SU5416, a VEGF receptor inhibitor and ligand of the AHR, represents a new alternative for immunomodulation. PLoS ONE 7:e44547. 10.1371/journal.pone.0044547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Monteiro P., Gilot D., Langouet S., Fardel O. (2008). Activation of the aryl hydrocarbon receptor by the calcium/calmodulin-dependent protein kinase kinase inhibitor 7-oxo-7H-benzimidazo[2,1-a]benz[de]isoquinoline-3-carboxylic acid (STO-609). Drug Metab. Dispos. 36, 2556–2563. 10.1124/dmd.108.023333 [DOI] [PubMed] [Google Scholar]
  27. Munoz L. (2017). Non-kinase targets of protein kinase inhibitors. Nat. Rev. Drug Discov. 16, 424–440. 10.1038/nrd.2016.266 [DOI] [PubMed] [Google Scholar]
  28. Murray I. A., Patterson A. D., Perdew G. H. (2014). Aryl hydrocarbon receptor ligands in cancer: friend and foe. Nat. Rev. Cancer 14, 801–814. 10.1038/nrc3846 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Park S. J., Yoon W. K., Kim H. J., Son H. Y., Cho S. W., Jeong K. S., et al. (2005). 2,3,7,8-Tetrachlorodibenzo-p-dioxin activates ERK and p38 mitogen-activated protein kinases in RAW 264.7 cells. Anticancer Res. 25, 2831–2836. [PubMed] [Google Scholar]
  30. Puga A., Ma C., Marlowe J. L. (2009). The aryl hydrocarbon receptor cross-talks with multiple signal transduction pathways. Biochem. Pharmacol. 77, 713–722. 10.1016/j.bcp.2008.08.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Reiners J. J., Jr., Lee J. Y., Clift R. E., Dudley D. T., Myrand S. P. (1998). PD98059 is an equipotent antagonist of the aryl hydrocarbon receptor and inhibitor of mitogen-activated protein kinase kinase. Mol. Pharmacol. 53, 438–445. 10.1124/mol.53.3.438 [DOI] [PubMed] [Google Scholar]
  32. Smutny T., Bitman M., Urban M., Dubecka M., Vrzal R., Dvorak Z., et al. (2014). U0126, a mitogen-activated protein kinase kinase 1 and 2 (MEK1 and 2) inhibitor, selectively up-regulates main isoforms of CYP3A subfamily via a pregnane X receptor (PXR) in HepG2 cells. Arch. Toxicol. 88, 2243–2259. 10.1007/s00204-014-1254-2 [DOI] [PubMed] [Google Scholar]
  33. Tian Y. (2009). Ah receptor and NF-kappaB interplay on the stage of epigenome. Biochem. Pharmacol. 77, 670–680. 10.1016/j.bcp.2008.10.023 [DOI] [PubMed] [Google Scholar]
  34. Watkins R. E., Wisely G. B., Moore L. B., Collins J. L., Lambert M. H., Williams S. P., et al. (2001). The human nuclear xenobiotic receptor PXR: structural determinants of directed promiscuity. Science 292, 2329–2333. 10.1126/science.1060762 [DOI] [PubMed] [Google Scholar]
  35. Weiss C., Faust D., Dürk H., Kolluri S. K., Pelzer A., Schneider S., et al. (2005). TCDD induces c-jun expression via a novel Ah (dioxin) receptor-mediated p38-MAPK-dependent pathway. Oncogene 24, 4975–4983. 10.1038/sj.onc.1208679 [DOI] [PubMed] [Google Scholar]
  36. Wu P., Nielsen T. E., Clausen M. H. (2015). FDA-approved small-molecule kinase inhibitors. Trends Pharmacol. Sci. 36, 422–439. 10.1016/j.tips.2015.04.005 [DOI] [PubMed] [Google Scholar]
  37. Xie G., Peng Z., Raufman J. P. (2012). Src-mediated aryl hydrocarbon and epidermal growth factor receptor cross talk stimulates colon cancer cell proliferation. Am. J. Physiol. Gastrointest. Liver Physiol. 302, G1006–G1015. 10.1152/ajpgi.00427.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Frontiers in Pharmacology are provided here courtesy of Frontiers Media SA

RESOURCES