Skip to main content
Log in

Induction of pro-apoptotic and cell cycle-inhibiting genes in chromium (VI)-treated human lung fibroblasts: Lack of effect of ERK

  • Published:
Molecular and Cellular Biochemistry Aims and scope Submit manuscript

Abstract

Cell proliferation and apoptosis are controlled by tightly orchestrated signaling pathways that culminate in transcriptional activation/repression of multiple proteins. Dysregulation of cell cycle and/or apoptosis control may lead to genomic instability, neoplastic transformation and tumor progression. Under certain conditions, some hexavalent chromium [Cr(VI)] compounds are toxic and carcinogenic in the human respiratory tract, and we have shown that they induce apoptosis and/or cell cycle arrest in a p53-dependent fashion. There is increasing evidence linking extracellular signal-regulated kinase (ERK) activation with the DNA damage response, by both p53-dependent and -independent mechanisms. Here, the aim was to study the effect of Cr(VI) transcriptional regulation of key cell cycle inhibitors and pro- and anti-apoptotic proteins, as well as the role of ERK activation in the Cr(VI) genotoxic response. Diploid human lung fibroblasts were incubated with 3–9 uM Na2CrO4, and RNA was isolated at 4, 8, and 24 h, as well as 24 h after Cr(VI) exposure was terminated (recovery). mRNA expression was quantitated by RNase protection assay with a 32P-labeled multi-transcript probe containing gene sequences for the cdk inhibitors, p21waf1/cip1, p27kip1, p16INK4a, p15INK4b; the pro-apoptotic proteins bcl-XS and bax; the anti-apoptotic proteins bcl-W, bcl-XL, and bcl2, GADD45, and cyclin A. In general, bcl-W and bcl-XL expression were both downregulated after Cr exposure, to around 50% at 24 h, which was more pronounced after the recovery period. At Cr(VI) concentrations ≤ 6 uM, bcl2 expression was upregulated. Of particular interest is that bax expression was reduced, in a dose and time-dependent fashion, however that of bcl-XS was elevated by nearly 3-fold after 8 h, and declined to control levels at the end of the recovery period. Expression of GADD45 and p21 were both upregulated by 2-fold at 8 h, but declined to control levels during recovery. Neither the expression of p27 nor that of p16 were apparently affected by Cr(VI) exposure, however the expression of p15 was markedly increased after exposure to all concentrations of Cr(VI). Finally, the expression of cyclin A was decreased after 24 h Cr(VI) exposure. Cr(VI) induced a transient burst of ERK activity (2–6-fold over control) around 0.5-3 h after exposure. However, inhibition of ERK activation with PD98059 had no effect on the Cr-induced alterations in gene expression. Moreover, Cr(VI)-induced clonogenic lethality, as assessed after 24 h exposure to 1 and 2 uM Cr(VI), was also not affected by ERK inhibition. These data suggest that both p53-dependent and -independent apoptotic and growth-inhibitory pathways are markedly affected by Cr(VI) exposure. However, the ability of Cr(VI) to affect key apoptotic and growth arresting genes, and thus clonogenic lethality, appears to be independent of ERK. Continued investigation into the cellular and molecular mechanisms of Cr(VI)-induced cell cycle and apoptosis control should further the understanding of Cr(VI)-associated carcinogenesis.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Hayes RB: Review of occupational epidemiology of chromium chemicals and respiratory cancer. Science Total Environ 71: 331–339, 1988

    Google Scholar 

  2. Langard S: One hundred years of chromium and cancer: A review of epidemiological evidence and selected case reports. Am J Ind Med 17: 189–215, 1990

    Google Scholar 

  3. International Agency for Research on Cancer: Chromium, nickel and welding. In: IARC. Monographs on the Evaluation of the Carcinogenic Risk to Humans, Vol. 49. Lyon, 1990, pp 1–648

    Google Scholar 

  4. De Flora S: Threshold mechanisms and site specificity in chromium (VI) carcinogenesis. Carcinogenesis 21: 533–541, 2000

    Google Scholar 

  5. Miller CA III, Costa M: Characterization of DNA-protein complexes induced in intact cells by the carcinogen chromate. Mol Carcinogen 1: 125–133, 1988

    Google Scholar 

  6. Standeven AM, Wetterhahn KE: Chromium (VI) toxicity: Uptake, reduction, and DNA damage. J Am Cell Toxicol 8: 1275–1283, 1989

    Google Scholar 

  7. Singh J, Mclean JA, Pritchard DE, Montaser A, Patierno SR: Sensitive quantitation of chromium-DNA adducts by inductively coupled plasma mass spectrometry with a direct injection high-efficiency nebulizer. Toxicol Sci 46: 260–265, 1998

    Google Scholar 

  8. Xu J, Manning FC, Patierno SR: Preferential formation and repair of chromium-induced DNA adducts and DNA-protein crosslinks in nuclear matrix DNA. Carcinogenesis 15: 1443–1450, 1994

    Google Scholar 

  9. Xu J, Bubley GJ, Detrick B, Blankenship LJ, Patierno SR: Chromium (VI) treatment of normal human lung cells results in guanine-specific DNA polymerase arrest, DNA-DNA cross-links and S-phase blockade of cell cycle. Carcinogenesis 17: 1511–1517, 1996

    Google Scholar 

  10. Singh J, Bridgewater LC, Patierno SR: Differential sensitivity of chromium-mediated DNA interstrand crosslinks and DNA-protein crosslinks to disruption by alkali and EDTA. Toxicol Sci 45: 72–76, 1998

    Google Scholar 

  11. Bridgewater LC, Manning FC, Patierno SR: Base-specific arrest of in vitro DNA replication by carcinogenic chromium: Relationship to DNA interstrand crosslinking. Carcinogenesis 15: 2421–2427, 1994

    Google Scholar 

  12. Singh J, Carlisle DL, Pritchard DE, Patierno SR: Chromium-induced genotoxicity and apoptosis: Relationship to chromium carcinogenesis (review). Oncol Rep 5: 1307–1318, 1998

    Google Scholar 

  13. Bridgewater LC, Manning FC, Woo ES, Patierno SR: DNA polymerase arrest by adducted trivalent chromium. Mol Carcinogen 9: 122–133, 1994

    Google Scholar 

  14. Manning FC, Xu J, Patierno SR: Transcriptional inhibition by carcinogenic chromate: Relationship to DNA damage. Mol Carcinogen 6: 270–279, 1992

    Google Scholar 

  15. Patierno SR, Banh D, Landolph JR: Transformation of C3H/10T1/2 mouse embryo cells to focus formation and anchorage independence by insoluble lead chromate but not soluble calcium chromate: Relationship to mutagenesis and internalization of lead chromate particles. Cancer Res 48: 5280–5288, 1988

    Google Scholar 

  16. Patierno SR, Landolph JR: Soluble vs. insoluble hexavalent chromate. Relationship of mutation to in vitro transformation and particle uptake. Biol Trace Element Res 21: 469–474, 1989

    Google Scholar 

  17. Zhitkovich A, Song Y, Quievryn G, Voitkun V: Non-oxidative mechanisms are responsible for the induction of mutagenesis by reduction of Cr(VI) with cysteine: Role of ternary DNA adducts in Cr(III)-dependent mutagenesis. Biochemistry 40: 549–560, 2001

    Google Scholar 

  18. Tully DB, Collins BJ, Overstreet JD, Smith CS, Dinse GE, Mumtaz MM, Chapin RE: Effects of arsenic, cadmium, chromium, and lead on gene expression regulated by a battery of 13 different promoters in recombinant HepG2 cells. Toxicol Appl Pharmacol 168: 79–90, 2000

    Google Scholar 

  19. Wetterhahn KE, Hamilton JW: Molecular basis of hexavalent chromium carcinogenicity: Effect on gene expression. Sci Total Environ 86: 113–129, 1989

    Google Scholar 

  20. Hamilton JW, Kaltreider RC, Bajenova OV, Ihnat MA, McCaffrey J, Turpie BW, Rowell EE, Oh J, Nemeth MJ, Pesce CA, Lariviere JP: Molecular basis for effects of carcinogenic heavy metals on inducible gene expression. Environ Health Perspect 106(suppl 4): 1005–1015, 1998

    Google Scholar 

  21. Singh J, Pritchard DE, Carlisle DL, Mclean JA, Montaser A, Orenstein JM, Patierno SR: Internalization of carcinogenic lead chromate particles by cultured normal human lung epithelial cells: Formation of intracellular lead-inclusion bodies and induction of apoptosis. Toxicol Appl Pharmacol 161: 240–248, 1999

    Google Scholar 

  22. Wise JP Sr, Stearns DM, Wetterhahn KE, Patierno SR: Cell-enhanced dissolution of carcinogenic lead chromate particles: The role of individual dissolution products in clastogenesis. Carcinogenesis 15: 2249–2254, 1994

    Google Scholar 

  23. Flatt PM, Polyak K, Tang LJ, Scatena CD, Westfall MD, Rubinstein LA, Yu J, Kinzler KW, Vogelstein B, Hill DE, Pietenpol JA: p53-dependent expression of PIG3 during proliferation, genotoxic stress, and reversible growth arrest. Cancer Lett 156: 63–72, 2000

    Google Scholar 

  24. Levine AJ: p53, the cellular gatekeeper for growth and division. Cell 88: 323–331, 1997

    Google Scholar 

  25. Carlisle DL, Pritchard DE, Singh J, Patierno SR: Chromium(VI) induces p53-dependent apoptosis in diploid human lung and mouse dermal fibroblasts. Mol Carcinogen 28: 111–118, 2000

    Google Scholar 

  26. Carlisle DL, Pritchard DE, Singh J, Owens BM, Blankenship LJ, Orenstein JM, Patierno SR: Apoptosis and P53 induction in human lung fibroblasts exposed to chromium (VI): Effect of ascorbate and tocopherol. Toxicol Sci 55: 60–68, 2000

    Google Scholar 

  27. Ye J, Wang S, Leonard SS, Sun Y, Butterworth L, Antonini J, Ding M, Rojanasakul Y, Vallyathan V, Castranova V, Shi X: Role of reactive oxygen species and p53 in chromium(VI)-induced apoptosis. J Biol Chem 274: 34974–34980, 1999

    Google Scholar 

  28. Wang S, Shi X: Mechanisms of Cr(VI)-induced p53 activation: The role of phosphorylation, mdm2 and ERK. Carcinogenesis 22: 757–762, 2001

    Google Scholar 

  29. Ha L, Ceryak S, Patierno SR: Chromium (VI) activates ataxia telangiectasia mutated (ATM) protein: Requirement of ATM for both apoptosis and recovery from terminal growth arrest. J Biol Chem 278: 17885–17894, 2003

    Google Scholar 

  30. Ballif BA, Blenis J: Molecular mechanisms mediating mammalian mitogen-activated protein kinase (MAPK) kinase (MEK)-MAPK cell survival signals. Cell Growth Differ 12: 397–408, 2001

    Google Scholar 

  31. Thomas G: MAP kinase by any other name smells just as sweet. Cell 68: 3–6, 1992

    Google Scholar 

  32. Rivard N, Boucher MJ, Asselin C, L'Allemain G: MAP kinase cascade is required for p27 downregulation and S phase entry in fibroblasts and epithelial cells. Am J Physiol 277: C652–C664, 1999

    Google Scholar 

  33. Tang D, Wu D, Hirao A, Lahti JM, Liu L, Mazza B, Kidd VJ, Mak TW, Ingram AJ: ERK activation mediates cell cycle arrest and apoptosis after DNA damage independently of p53. J Biol Chem 277: 12710–12717, 2002

    Google Scholar 

  34. Persons DL, Yazlovitskaya EM, Cui W, Pelling JC: Cisplatin-induced activation of mitogen-activated protein kinases in ovarian carcinoma cells: Inhibition of extracellular signal-regulated kinase activity increases sensitivity to cisplatin. Clin Cancer Res 5: 1007–1014, 1999

    Google Scholar 

  35. Buckley S, Driscoll B, Barsky L, Weinberg K, Anderson K, Warburton D: ERK activation protects against DNA damage and apoptosis in hyperoxic rat AEC2. Am J Physiol 277: L159–L166, 1999

    Google Scholar 

  36. DeHaan RD, Yazlovitskaya EM, Persons DL: Regulation of p53 target gene expression by cisplatin-induced extracellular signal-regulated kinase. Cancer Chemother Pharmacol 48: 383–388, 2001

    Google Scholar 

  37. Chuang SM, Liou GY, Yang JL: Activation of JNK, p38 and ERK mitogen-activated protein kinases by chromium(VI) is mediated through oxidative stress but does not affect cytotoxicity. Carcinogenesis 21: 1491–1500, 2000

    Google Scholar 

  38. Pritchard DE, Ceryak S, Ha L, Fornsaglio JL, Hartman SK, O'Brien TJ, Patierno SR: Mechanism of apoptosis and determination of cellular fate in chromium(VI)-exposed populations of telomerase-immortalized human fibroblasts. Cell Growth Differ 2001

  39. Canman CE, Lim DS, Cimprich KA, Taya Y, Tamai K, Sakaguchi K, Appella E, Kastan MB, Siliciano JD: Activation of the ATM kinase by ionizing radiation and phosphorylation of p53. Science 281: 1677–1679, 1998

    Google Scholar 

  40. Alessi DR, Cuenda A, Cohen P, Dudley DT, Saltiel AR: PD 098059 is a specific inhibitor of the activation of mitogen-activated protein kinase kinase in vitro and in vivo. J Biol Chem 270: 27489–27494, 1995

    Google Scholar 

  41. Ye J, Shi X: Gene expression profile in response to chromium-induced cell stress in A549 cells. Mol Cell Biochem 222: 189–197, 2001

    Google Scholar 

  42. Pritchard DE, Singh J, Carlisle DL, Patierno SR: Cyclosporin A inhibits chromium(VI)-induced apoptosis and mitochondrial cytochrome c release and restores clonogenic survival in CHO cells. Carcinogenesis 21: 2027–2033, 2000

    Google Scholar 

  43. Korsmeyer SJ, Gross A, Harada H, Zha J, Wang K, Yin XM, Wei M, Zinkel S: Death and survival signals determine active/inactive conformations of pro-apoptotic BAX, BAD, and BID molecules. Cold Spring Harb Symp Quant Biol 64: 343–350, 1999

    Google Scholar 

  44. Nicolli A, Basso E, Petronilli V, Wenger RM, Bernardi P: Interactions of cyclophilin with the mitochondrial inner membrane and regulation of the permeability transition pore, and cyclosporin A-sensitive channel. J Biol Chem 271: 2185–2192, 1996

    Google Scholar 

  45. O'Brien TJ, Fornsaglio JL, Ceryak S, Patierno SR: Effects of hexavalent chromium on the survival and cell cycle distribution of DNA repair-deficient S. cerevisiae. DNA Rep 1: 617–627, 2002

    Google Scholar 

  46. Sionov RV, Haupt Y: The cellular response to p53: The decision between life and death. Oncogene 18: 6145–6157, 1999

    Google Scholar 

  47. Banin S, Moyal L, Shieh S, Taya Y, Anderson CW, Chessa L, Smorodinsky NI, Prives C, Reiss Y, Shiloh Y, Ziv Y: Enhanced phosphorylation of p53 by ATM in response to DNA damage. Science 281: 1674–1677, 1998

    Google Scholar 

  48. Saito S, Goodarzi AA, Higashimoto Y, Noda Y, Lees-Miller SP, Appella E, Anderson CW: ATM mediates phosphorylation at multiple p53 sites, including Ser(46), in response to ionizing radiation. J Biol Chem 277: 12491–12494, 2002

    Google Scholar 

  49. Persons DL, Yazlovitskaya EM, Pelling JC: Effect of extracellular signal-regulated kinase on p53 accumulation in response to cisplatin. J Biol Chem 275: 35778–35785, 2000

    Google Scholar 

  50. Hayakawa J, Ohmichi M, Kurachi H, Kanda Y, Hisamoto K, Nishio Y, Adachi K, Tasaka K, Kanzaki T, Murata Y: Inhibition of BAD phosphorylation either at serine 112 via extracellular signal-regulated protein kinase cascade or at serine 136 via Akt cascade sensitizes human ovarian cancer cells to cisplatin. Cancer Res 60: 5988–5994, 2000

    Google Scholar 

  51. Hayakawa J, Ohmichi M, Kurachi H, Ikegami H, Kimura A, Matsuoka T, Jikihara H, Mercola D, Murata Y: Inhibition of extracellular signal-regulated protein kinase or c-Jun N-terminal protein kinase cascade, differentially activated by cisplatin, sensitizes human ovarian cancer cell line. J Biol Chem 274: 31648–31654, 1999

    Google Scholar 

  52. Greulich H, Erikson RL: An analysis of Mek1 signaling in cell proliferation and transformation. J Biol Chem 273:13280–13288, 1998

    Google Scholar 

  53. Moelling K, Schad K, Bosse M, Zimmermann S, Schweneker M: Regulation of Raf-Akt Cross-talk. J Biol Chem 277: 31099–31106, 2002

    Google Scholar 

  54. Murphy LO, Smith S, Chen RH, Fingar DC, Blenis J: Molecular interpretation of ERK signal duration by immediate early gene products. Nat Cell Biol 4: 556–564, 2002

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ceryak, S., Zingariello, C., O'Brien, T. et al. Induction of pro-apoptotic and cell cycle-inhibiting genes in chromium (VI)-treated human lung fibroblasts: Lack of effect of ERK. Mol Cell Biochem 255, 139–149 (2004). https://doi.org/10.1023/B:MCBI.0000007270.82431.3e

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1023/B:MCBI.0000007270.82431.3e

Navigation