Skip to main content
Biochemical Journal logoLink to Biochemical Journal
. 2003 Nov 15;376(Pt 1):1–14. doi: 10.1042/BJ20031327

A perspective of polyamine metabolism.

Heather M Wallace 1, Alison V Fraser 1, Alun Hughes 1
PMCID: PMC1223767  PMID: 13678416

Abstract

Polyamines are essential for the growth and function of normal cells. They interact with various macromolecules, both electrostatically and covalently and, as a consequence, have a variety of cellular effects. The complexity of polyamine metabolism and the multitude of compensatory mechanisms that are invoked to maintain polyamine homoeostasis argue that these amines are critical to cell survival. The regulation of polyamine content within cells occurs at several levels, including transcription and translation. In addition, novel features such as the +1 frameshift required for antizyme production and the rapid turnover of several of the enzymes involved in the pathway make the regulation of polyamine metabolism a fascinating subject. The link between polyamine content and human disease is unequivocal, and significant success has been obtained in the treatment of a number of parasitic infections. Targeting the polyamine pathway as a means of treating cancer has met with limited success, although the development of drugs such as DFMO (alpha-difluoromethylornithine), a rationally designed anticancer agent, has revolutionized our understanding of polyamine function in cell growth and provided 'proof of concept' that influencing polyamine metabolism and content within tumour cells will prevent tumour growth. The more recent development of the polyamine analogues has been pivotal in advancing our understanding of the necessity to deplete all three polyamines to induce apoptosis in tumour cells. The current thinking is that the polyamine inhibitors/analogues may also be useful agents in the chemoprevention of cancer and, in this area, we may yet see a revival of DFMO. The future will be in adopting a functional genomics approach to identifying polyamine-regulated genes linked to either carcinogenesis or apoptosis.

Full Text

The Full Text of this article is available as a PDF (258.6 KB).

Selected References

These references are in PubMed. This may not be the complete list of references from this article.

  1. Abdel-Monem M. M., Newton N. E., Weeks C. E. Inhibitors of polyamine biosynthesis. 1. Alpha-methyl-(plus or minus)-ornithine, an inhibitor of ornithine decarboxylase. J Med Chem. 1974 Apr;17(4):447–451. doi: 10.1021/jm00250a016. [DOI] [PubMed] [Google Scholar]
  2. Abdel-Monem M. M., Newton N. E., Weeks C. E. Inhibitors of polyamine biosynthesis. 3. (+/-)-5-Amino-2-hydrazine-2-methylpentanoic acid, an inhibitor of ornithine decarboxylase. J Med Chem. 1975 Sep;18(9):945–948. doi: 10.1021/jm00243a017. [DOI] [PubMed] [Google Scholar]
  3. Alhonen-Hongisto L., Seppänen P., Jänne J. Intracellular putrescine and spermidine deprivation induces increased uptake of the natural polyamines and methylglyoxal bis(guanylhydrazone). Biochem J. 1980 Dec 15;192(3):941–945. doi: 10.1042/bj1920941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Alm K., Berntsson P. S., Kramer D. L., Porter C. W., Oredsson S. M. Treatment of cells with the polyamine analog N, N11-diethylnorspermine retards S phase progression within one cell cycle. Eur J Biochem. 2000 Jul;267(13):4157–4164. doi: 10.1046/j.1432-1327.2000.01460.x. [DOI] [PubMed] [Google Scholar]
  5. Auvinen M., Paasinen A., Andersson L. C., Hölttä E. Ornithine decarboxylase activity is critical for cell transformation. Nature. 1992 Nov 26;360(6402):355–358. doi: 10.1038/360355a0. [DOI] [PubMed] [Google Scholar]
  6. Averill-Bates D. A., Agostinelli E., Przybytkowski E., Mondovi B. Aldehyde dehydrogenase and cytotoxicity of purified bovine serum amine oxidase and spermine in Chinese hamster ovary cells. Biochem Cell Biol. 1994 Jan-Feb;72(1-2):36–42. doi: 10.1139/o94-006. [DOI] [PubMed] [Google Scholar]
  7. Bacchi C. J., Garofalo J., Ciminelli M., Rattendi D., Goldberg B., McCann P. P., Yarlett N. Resistance to DL-alpha-difluoromethylornithine by clinical isolates of Trypanosoma brucei rhodesiense. Role of S-adenosylmethionine. Biochem Pharmacol. 1993 Aug 3;46(3):471–481. doi: 10.1016/0006-2952(93)90524-z. [DOI] [PubMed] [Google Scholar]
  8. Bacchi C. J. Resistance to clinical drugs in African trypanosomes. Parasitol Today. 1993 May;9(5):190–193. doi: 10.1016/0169-4758(93)90145-6. [DOI] [PubMed] [Google Scholar]
  9. Bacchi C. J., Yarlett N. Polyamine metabolism as chemotherapeutic target in protozoan parasites. Mini Rev Med Chem. 2002 Dec;2(6):553–563. doi: 10.2174/1389557023405549. [DOI] [PubMed] [Google Scholar]
  10. Bailey S., Smith K., Fairlamb A. H., Hunter W. N. Substrate interactions between trypanothione reductase and N1-glutathionylspermidine disulphide at 0.28-nm resolution. Eur J Biochem. 1993 Apr 1;213(1):67–75. doi: 10.1111/j.1432-1033.1993.tb17734.x. [DOI] [PubMed] [Google Scholar]
  11. Bardocz S. The role of dietary polyamines. Eur J Clin Nutr. 1993 Oct;47(10):683–690. [PubMed] [Google Scholar]
  12. Basu H. S., Smirnov I. V., Peng H. F., Tiffany K., Jackson V. Effects of spermine and its cytotoxic analogs on nucleosome formation on topologically stressed DNA in vitro. Eur J Biochem. 1997 Jan 15;243(1-2):247–258. doi: 10.1111/j.1432-1033.1997.0247a.x. [DOI] [PubMed] [Google Scholar]
  13. Basu H. S., Wright W. D., Deen D. F., Roti-Roti J., Marton L. J. Treatment with a polyamine analog alters DNA-matrix association in HeLa cell nuclei: a nucleoid halo assay. Biochemistry. 1993 Apr 20;32(15):4073–4076. doi: 10.1021/bi00066a031. [DOI] [PubMed] [Google Scholar]
  14. Beninati S., Gentile V., Caraglia M., Lentini A., Tagliaferri P., Abbruzzese A. Tissue transglutaminase expression affects hypusine metabolism in BALB/c 3T3 cells. FEBS Lett. 1998 Oct 16;437(1-2):34–38. doi: 10.1016/s0014-5793(98)01191-0. [DOI] [PubMed] [Google Scholar]
  15. Beppu T., Shirahata A., Takahashi N., Hosoda H., Samejima K. Specific depletion of spermidine and spermine in HTC cells treated with inhibitors of aminopropyltransferases. J Biochem. 1995 Feb;117(2):339–345. doi: 10.1093/jb/117.2.339. [DOI] [PubMed] [Google Scholar]
  16. Bettuzzi S., Davalli P., Astancolle S., Pinna C., Roncaglia R., Boraldi F., Tiozzo R., Sharrard M., Corti A. Coordinate changes of polyamine metabolism regulatory proteins during the cell cycle of normal human dermal fibroblasts. FEBS Lett. 1999 Mar 5;446(1):18–22. doi: 10.1016/s0014-5793(99)00182-9. [DOI] [PubMed] [Google Scholar]
  17. Bolkenius F. N., Bey P., Seiler N. Specific inhibition of polyamine oxidase in vivo is a method for the elucidation of its physiological role. Biochim Biophys Acta. 1985 Jan 28;838(1):69–76. doi: 10.1016/0304-4165(85)90251-x. [DOI] [PubMed] [Google Scholar]
  18. Bolkenius F. N., Seiler N. Acetylderivatives as intermediates in polyamine catabolism. Int J Biochem. 1981;13(3):287–292. doi: 10.1016/0020-711x(81)90080-x. [DOI] [PubMed] [Google Scholar]
  19. Brunton V. G., Grant M. H., Wallace H. M. Mechanisms of spermine toxicity in baby-hamster kidney (BHK) cells. The role of amine oxidases and oxidative stress. Biochem J. 1991 Nov 15;280(Pt 1):193–198. doi: 10.1042/bj2800193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Brunton V. G., Grant M. H., Wallace H. M. Spermine toxicity and glutathione depletion in BHK-21/C13 cells. Biochem Pharmacol. 1990 Oct 15;40(8):1893–1900. doi: 10.1016/0006-2952(90)90371-q. [DOI] [PubMed] [Google Scholar]
  21. Byers T. L., Kameji R., Rannels D. E., Pegg A. E. Multiple pathways for uptake of paraquat, methylglyoxal bis(guanylhydrazone), and polyamines. Am J Physiol. 1987 Jun;252(6 Pt 1):C663–C669. doi: 10.1152/ajpcell.1987.252.6.C663. [DOI] [PubMed] [Google Scholar]
  22. Casero R. A., Jr, Pegg A. E. Spermidine/spermine N1-acetyltransferase--the turning point in polyamine metabolism. FASEB J. 1993 May;7(8):653–661. [PubMed] [Google Scholar]
  23. Casero R. A., Jr, Woster P. M. Terminally alkylated polyamine analogues as chemotherapeutic agents. J Med Chem. 2001 Jan 4;44(1):1–26. doi: 10.1021/jm000084m. [DOI] [PubMed] [Google Scholar]
  24. Celano P., Berchtold C. M., Kizer D. L., Weeraratna A., Nelkin B. D., Baylin S. B., Casero R. A., Jr Characterization of an endogenous RNA transcript with homology to the antisense strand of the human c-myc gene. J Biol Chem. 1992 Jul 25;267(21):15092–15096. [PubMed] [Google Scholar]
  25. Chopra S., Wallace H. M. Induction of spermidine/spermine N1-acetyltransferase in human cancer cells in response to increased production of reactive oxygen species. Biochem Pharmacol. 1998 Apr 1;55(7):1119–1123. doi: 10.1016/s0006-2952(97)00601-1. [DOI] [PubMed] [Google Scholar]
  26. Clifford A., Morgan D., Yuspa S. H., Soler A. P., Gilmour S. Role of ornithine decarboxylase in epidermal tumorigenesis. Cancer Res. 1995 Apr 15;55(8):1680–1686. [PubMed] [Google Scholar]
  27. Coffino P. Antizyme, a mediator of ubiquitin-independent proteasomal degradation. Biochimie. 2001 Mar-Apr;83(3-4):319–323. doi: 10.1016/s0300-9084(01)01252-4. [DOI] [PubMed] [Google Scholar]
  28. Coleman C. S., Huang H., Pegg A. E. Structure and critical residues at the active site of spermidine/spermine-N1-acetyltransferase. Biochem J. 1996 Jun 15;316(Pt 3):697–701. doi: 10.1042/bj3160697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Coleman C. S., Pegg A. E. Proteasomal degradation of spermidine/spermine N1-acetyltransferase requires the carboxyl-terminal glutamic acid residues. J Biol Chem. 1997 May 2;272(18):12164–12169. doi: 10.1074/jbc.272.18.12164. [DOI] [PubMed] [Google Scholar]
  30. Coleman C. S., Stanley B. A., Viswanath R., Pegg A. E. Rapid exchange of subunits of mammalian ornithine decarboxylase. J Biol Chem. 1994 Feb 4;269(5):3155–3158. [PubMed] [Google Scholar]
  31. Coleman C. S., Wallace H. M. Polyamine excretion from human cancer cells. Biochem Soc Trans. 1990 Dec;18(6):1228–1229. doi: 10.1042/bst0181228. [DOI] [PubMed] [Google Scholar]
  32. Corti A., Dave C., Williams-Ashman H. G., Mihich E., Schenone A. Specific inhibition of the enzymic decarboxylation of S-adenosylmethionine by methylglyoxal bis(guanylhydrazone) and related substances. Biochem J. 1974 May;139(2):351–357. doi: 10.1042/bj1390351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Croghan M. K., Aickin M. G., Meyskens F. L. Dose-related alpha-difluoromethylornithine ototoxicity. Am J Clin Oncol. 1991 Aug;14(4):331–335. doi: 10.1097/00000421-199108000-00012. [DOI] [PubMed] [Google Scholar]
  34. Danzin C., Casara P., Claverie N., Metcalf B. W. alpha-Ethynyl and alpha-vinyl analogues of ornithine as enzyme-activated inhibitors of mammalian ornithine decarboxylase. J Med Chem. 1981 Jan;24(1):16–20. doi: 10.1021/jm00133a005. [DOI] [PubMed] [Google Scholar]
  35. Danzin C., Marchal P., Casara P. Irreversible inhibition of rat S-adenosylmethionine decarboxylase by 5'-([(Z)-4-amino-2-butenyl]methylamino)-5'-deoxyadenosine. Biochem Pharmacol. 1990 Oct 1;40(7):1499–1503. doi: 10.1016/0006-2952(90)90446-r. [DOI] [PubMed] [Google Scholar]
  36. Driscoll J., Frydman J., Goldberg A. L. An ATP-stabilized inhibitor of the proteasome is a component of the 1500-kDa ubiquitin conjugate-degrading complex. Proc Natl Acad Sci U S A. 1992 Jun 1;89(11):4986–4990. doi: 10.1073/pnas.89.11.4986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Dudley H. W., Rosenheim M. C., Rosenheim O. The Chemical Constitution of Spermine. I. The Isolation of Spermine from Animal Tissues, and the Preparation of its Salts. Biochem J. 1924;18(6):1263–1272. doi: 10.1042/bj0181263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Dudley H. W., Rosenheim O., Starling W. W. The Chemical Constitution of Spermine: Structure and Synthesis. Biochem J. 1926;20(5):1082–1094. doi: 10.1042/bj0201082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Duranton B., Holl V., Schneider Y., Carnesecchi S., Gossé F., Raul F., Seiler N. Cytotoxic effects of the polyamine oxidase inactivator MDL 72527 to two human colon carcinoma cell lines SW480 and SW620. Cell Biol Toxicol. 2002;18(6):381–396. doi: 10.1023/a:1020863506170. [DOI] [PubMed] [Google Scholar]
  40. Eloranta T. O., Khomutov A. R., Khomutov R. M., Hyvönen T. Aminooxy analogues of spermidine as inhibitors of spermine synthase and substrates of hepatic polyamine acetylating activity. J Biochem. 1990 Oct;108(4):593–598. doi: 10.1093/oxfordjournals.jbchem.a123248. [DOI] [PubMed] [Google Scholar]
  41. Fair W. R., Wehner N., Brorsson U. Urinary polyamine levels in the diagnosis of carcinoma of the prostate. J Urol. 1975 Jul;114(1):88–92. doi: 10.1016/s0022-5347(17)66951-9. [DOI] [PubMed] [Google Scholar]
  42. Fairlamb A. H., Henderson G. B., Bacchi C. J., Cerami A. In vivo effects of difluoromethylornithine on trypanothione and polyamine levels in bloodstream forms of Trypanosoma brucei. Mol Biochem Parasitol. 1987 Jun;24(2):185–191. doi: 10.1016/0166-6851(87)90105-8. [DOI] [PubMed] [Google Scholar]
  43. Flayeh K. A., Wallace H. M. Polyamine oxidase activity in a human colonic carcinoma cell line. Biochem Soc Trans. 1990 Dec;18(6):1225–1225. doi: 10.1042/bst0181225. [DOI] [PubMed] [Google Scholar]
  44. Fogel-Petrovic M., Shappell N. W., Bergeron R. J., Porter C. W. Polyamine and polyamine analog regulation of spermidine/spermine N1-acetyltransferase in MALME-3M human melanoma cells. J Biol Chem. 1993 Sep 5;268(25):19118–19125. [PubMed] [Google Scholar]
  45. Fraser Alison V., Woster Patrick M., Wallace Heather M. Induction of apoptosis in human leukaemic cells by IPENSpm, a novel polyamine analogue and anti-metabolite. Biochem J. 2002 Oct 1;367(Pt 1):307–312. doi: 10.1042/BJ20020156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Fredlund J. O., Oredsson S. M. Ordered cell cycle phase perturbations in Chinese hamster ovary cells treated with an S-adenosylmethionine decarboxylase inhibitor. Eur J Biochem. 1997 Oct 1;249(1):232–238. doi: 10.1111/j.1432-1033.1997.00232.x. [DOI] [PubMed] [Google Scholar]
  47. Gerner E. W., Garewal H. S., Emerson S. S., Sampliner R. E. Gastrointestinal tissue polyamine contents of patients with Barrett's esophagus treated with alpha-difluoromethylornithine. Cancer Epidemiol Biomarkers Prev. 1994 Jun;3(4):325–330. [PubMed] [Google Scholar]
  48. Ghoda L., van Daalen Wetters T., Macrae M., Ascherman D., Coffino P. Prevention of rapid intracellular degradation of ODC by a carboxyl-terminal truncation. Science. 1989 Mar 17;243(4897):1493–1495. doi: 10.1126/science.2928784. [DOI] [PubMed] [Google Scholar]
  49. Giffin B. F., McCann P. P., Bitonti A. J., Bacchi C. J. Polyamine depletion following exposure to DL-alpha-difluoromethylornithine both in vivo and in vitro initiates morphological alterations and mitochondrial activation in a monomorphic strain of Trypanosoma brucei brucei. J Protozool. 1986 May;33(2):238–243. doi: 10.1111/j.1550-7408.1986.tb05599.x. [DOI] [PubMed] [Google Scholar]
  50. Gilmour S. K., Verma A. K., Madara T., O'Brien T. G. Regulation of ornithine decarboxylase gene expression in mouse epidermis and epidermal tumors during two-stage tumorigenesis. Cancer Res. 1987 Mar 1;47(5):1221–1225. [PubMed] [Google Scholar]
  51. Giuseppina Monti M., Ghiaroni S., Barbieri D., Franceschi C., Marverti G., Moruzzi M. S. 2-deoxy-d-ribose-induced apoptosis in HL-60 cells is associated with the cell cycle progression by spermidine. Biochem Biophys Res Commun. 1999 Apr 13;257(2):460–465. doi: 10.1006/bbrc.1999.0492. [DOI] [PubMed] [Google Scholar]
  52. Grassilli E., Benatti F., Dansi P., Giammarioli A. M., Malorni W., Franceschi C., Desiderio M. A. Inhibition of proteasome function prevents thymocyte apoptosis: involvement of ornithine decarboxylase. Biochem Biophys Res Commun. 1998 Sep 18;250(2):293–297. doi: 10.1006/bbrc.1998.9291. [DOI] [PubMed] [Google Scholar]
  53. Grove J., Fozard J. R., Mamont P. S. Assay of alpha-difluoromethylornithine in body fluids and tissues by automatic amino-acid analysis. J Chromatogr. 1981 May 8;223(2):409–416. doi: 10.1016/s0378-4347(00)80114-0. [DOI] [PubMed] [Google Scholar]
  54. Guo J., Wu Y. Q., Rattendi D., Bacchi C. J., Woster P. M. S-(5'-deoxy-5'-adenosyl)-1-aminoxy-4-(methylsulfonio)-2-cyclopentene (AdoMao): an irreversible inhibitor of S-adenosylmethionine decarboxylase with potent in vitro antitrypanosomal activity. J Med Chem. 1995 May 12;38(10):1770–1777. doi: 10.1021/jm00010a021. [DOI] [PubMed] [Google Scholar]
  55. Haddox M. K., Magun B. E., Russell D. H. Ornithine decarboxylase induction during B1 progression of normal and Rous sarcoma virus-transformed cells. Cancer Res. 1980 Mar;40(3):604–608. [PubMed] [Google Scholar]
  56. Hahm H. A., Dunn V. R., Butash K. A., Deveraux W. L., Woster P. M., Casero R. A., Jr, Davidson N. E. Combination of standard cytotoxic agents with polyamine analogues in the treatment of breast cancer cell lines. Clin Cancer Res. 2001 Feb;7(2):391–399. [PubMed] [Google Scholar]
  57. Hamana K., Matsuzaki S. Polyamines as a chemotaxonomic marker in bacterial systematics. Crit Rev Microbiol. 1992;18(4):261–283. doi: 10.3109/10408419209113518. [DOI] [PubMed] [Google Scholar]
  58. Hampel K. J., Crosson P., Lee J. S. Polyamines favor DNA triplex formation at neutral pH. Biochemistry. 1991 May 7;30(18):4455–4459. doi: 10.1021/bi00232a012. [DOI] [PubMed] [Google Scholar]
  59. Harada J., Sugimoto M. Polyamines prevent apoptotic cell death in cultured cerebellar granule neurons. Brain Res. 1997 Apr 11;753(2):251–259. doi: 10.1016/s0006-8993(97)00011-5. [DOI] [PubMed] [Google Scholar]
  60. Harik S. I., Snyder S. H. Ornithine decarboxylase: inhibition by alpha-hydrazinoornithine. Biochim Biophys Acta. 1973 Dec 19;327(2):501–509. doi: 10.1016/0005-2744(73)90433-6. [DOI] [PubMed] [Google Scholar]
  61. Hayashi S., Murakami Y., Matsufuji S. Ornithine decarboxylase antizyme: a novel type of regulatory protein. Trends Biochem Sci. 1996 Jan;21(1):27–30. [PubMed] [Google Scholar]
  62. Hayashi S., Murakami Y. Rapid and regulated degradation of ornithine decarboxylase. Biochem J. 1995 Feb 15;306(Pt 1):1–10. doi: 10.1042/bj3060001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Heby O. Ornithine decarboxylase as target of chemotherapy. Adv Enzyme Regul. 1985;24:103–124. doi: 10.1016/0065-2571(85)90072-x. [DOI] [PubMed] [Google Scholar]
  64. Heby O., Persson L. Molecular genetics of polyamine synthesis in eukaryotic cells. Trends Biochem Sci. 1990 Apr;15(4):153–158. doi: 10.1016/0968-0004(90)90216-x. [DOI] [PubMed] [Google Scholar]
  65. Heby O. Role of polyamines in the control of cell proliferation and differentiation. Differentiation. 1981;19(1):1–20. doi: 10.1111/j.1432-0436.1981.tb01123.x. [DOI] [PubMed] [Google Scholar]
  66. Heljasvaara R., Veress I., Halmekytö M., Alhonen L., Jänne J., Laajala P., Pajunen A. Transgenic mice overexpressing ornithine and S-adenosylmethionine decarboxylases maintain a physiological polyamine homoeostasis in their tissues. Biochem J. 1997 Apr 15;323(Pt 2):457–462. doi: 10.1042/bj3230457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Hessels J., Kingma A. W., Ferwerda H., Keij J., van den Berg G. A., Muskiet F. A. Microbial flora in the gastrointestinal tract abolishes cytostatic effects of alpha-difluoromethylornithine in vivo. Int J Cancer. 1989 Jun 15;43(6):1155–1164. doi: 10.1002/ijc.2910430632. [DOI] [PubMed] [Google Scholar]
  68. Hibshoosh H., Johnson M., Weinstein I. B. Effects of overexpression of ornithine decarboxylase (ODC) on growth control and oncogene-induced cell transformation. Oncogene. 1991 May;6(5):739–743. [PubMed] [Google Scholar]
  69. Holley J. L., Mather A., Wheelhouse R. T., Cullis P. M., Hartley J. A., Bingham J. P., Cohen G. M. Targeting of tumor cells and DNA by a chlorambucil-spermidine conjugate. Cancer Res. 1992 Aug 1;52(15):4190–4195. [PubMed] [Google Scholar]
  70. Hurta R. A., Wright J. A. Ornithine decarboxylase gene expression is aberrantly regulated via the cAMP signal transduction pathway in malignant H-ras transformed cell lines. J Cell Physiol. 1994 Nov;161(2):383–391. doi: 10.1002/jcp.1041610224. [DOI] [PubMed] [Google Scholar]
  71. Hölttä E. Oxidation of spermidine and spermine in rat liver: purification and properties of polyamine oxidase. Biochemistry. 1977 Jan 11;16(1):91–100. doi: 10.1021/bi00620a015. [DOI] [PubMed] [Google Scholar]
  72. Hölttä E., Sistonen L., Alitalo K. The mechanisms of ornithine decarboxylase deregulation in c-Ha-ras oncogene-transformed NIH 3T3 cells. J Biol Chem. 1988 Mar 25;263(9):4500–4507. [PubMed] [Google Scholar]
  73. Inoue H., Kato Y., Takigawa M., Adachi K., Takeda Y. Effect of DL-alpha-hydrazino-delta-aminovaleric acid, an inhibitor of ornithine decarboxylase, on polyamine metabolism in isoproterenol-stimulated mouse parotid glands. J Biochem. 1975 Apr;77(4):879–893. doi: 10.1093/oxfordjournals.jbchem.a130796. [DOI] [PubMed] [Google Scholar]
  74. Ito H., Hibasami H., Shimura K., Nagai J., Hidaka H. Antitumor effect of dicyclohexylammonium sulfate, a potent inhibitor of spermidine synthase against P388 leukemia. Cancer Lett. 1982 Mar-Apr;15(3):229–235. doi: 10.1016/0304-3835(82)90123-9. [DOI] [PubMed] [Google Scholar]
  75. Ivanov I. P., Rohrwasser A., Terreros D. A., Gesteland R. F., Atkins J. F. Discovery of a spermatogenesis stage-specific ornithine decarboxylase antizyme: antizyme 3. Proc Natl Acad Sci U S A. 2000 Apr 25;97(9):4808–4813. doi: 10.1073/pnas.070055897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Jaggi R., Friis R., Groner B. Oncogenes modulate cellular gene expression and repress glucocorticoid regulated gene transcription. J Steroid Biochem. 1988 May;29(5):457–463. doi: 10.1016/0022-4731(88)90179-3. [DOI] [PubMed] [Google Scholar]
  77. Johnson T. D. Modulation of channel function by polyamines. Trends Pharmacol Sci. 1996 Jan;17(1):22–27. doi: 10.1016/0165-6147(96)81566-5. [DOI] [PubMed] [Google Scholar]
  78. Jänne J., Morris D. R. Inhibition of S-adenosylmethionine decarboxylase and diamine oxidase activities by analogues of methylglyoxal bis(guanylhydrazone) and their cellular uptake during lymphocyte activation. Biochem J. 1984 Mar 15;218(3):947–951. doi: 10.1042/bj2180947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Kallio A., McCann P. P., Bey P. DL-a-Monofluoromethylputrescine is a potent irreversible inhibitor of Escherichia coli ornithine decarboxylase. Biochem J. 1982 Jun 15;204(3):771–775. doi: 10.1042/bj2040771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Khomutov R. M., Hyvönen T., Karvonen E., Kauppinen L., Paalanen T., Paulin L., Eloranta T., Pajula R. L., Andersson L. C., Pösö H. 1-Aminooxy-3-aminopropane, a new and potent inhibitor of polyamine biosynthesis that inhibits ornithine decarboxylase, adenosylmethionine decarboxylase and spermidine synthase. Biochem Biophys Res Commun. 1985 Jul 31;130(2):596–602. doi: 10.1016/0006-291x(85)90458-9. [DOI] [PubMed] [Google Scholar]
  81. Kingsnorth A. N., Lumsden A. B., Wallace H. M. Polyamines in colorectal cancer. Br J Surg. 1984 Oct;71(10):791–794. doi: 10.1002/bjs.1800711019. [DOI] [PubMed] [Google Scholar]
  82. Kingsnorth A. N., Wallace H. M., Bundred N. J., Dixon J. M. Polyamines in breast cancer. Br J Surg. 1984 May;71(5):352–356. doi: 10.1002/bjs.1800710513. [DOI] [PubMed] [Google Scholar]
  83. Kingsnorth A. N., Wallace H. M. Elevation of monoacetylated polyamines in human breast cancers. Eur J Cancer Clin Oncol. 1985 Sep;21(9):1057–1062. doi: 10.1016/0277-5379(85)90291-3. [DOI] [PubMed] [Google Scholar]
  84. Korhonen V. P., Niiranen K., Halmekytö M., Pietilä M., Diegelman P., Parkkinen J. J., Eloranta T., Porter C. W., Alhonen L., Jänne J. Spermine deficiency resulting from targeted disruption of the spermine synthase gene in embryonic stem cells leads to enhanced sensitivity to antiproliferative drugs. Mol Pharmacol. 2001 Feb;59(2):231–238. doi: 10.1124/mol.59.2.231. [DOI] [PubMed] [Google Scholar]
  85. Kramer D. L., Chang B. D., Chen Y., Diegelman P., Alm K., Black A. R., Roninson I. B., Porter C. W. Polyamine depletion in human melanoma cells leads to G1 arrest associated with induction of p21WAF1/CIP1/SDI1, changes in the expression of p21-regulated genes, and a senescence-like phenotype. Cancer Res. 2001 Nov 1;61(21):7754–7762. [PubMed] [Google Scholar]
  86. Kramer D. L., Fogel-Petrovic M., Diegelman P., Cooley J. M., Bernacki R. J., McManis J. S., Bergeron R. J., Porter C. W. Effects of novel spermine analogues on cell cycle progression and apoptosis in MALME-3M human melanoma cells. Cancer Res. 1997 Dec 15;57(24):5521–5527. [PubMed] [Google Scholar]
  87. Kramer D. L., Khomutov R. M., Bukin Y. V., Khomutov A. R., Porter C. W. Cellular characterization of a new irreversible inhibitor of S-adenosylmethionine decarboxylase and its use in determining the relative abilities of individual polyamines to sustain growth and viability of L1210 cells. Biochem J. 1989 Apr 15;259(2):325–331. doi: 10.1042/bj2590325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Laitinen J., Hölttä E. Methylation status and chromatin structure of an early response gene (ornithine decarboxylase) in resting and stimulated NIH-3T3 fibroblasts. J Cell Biochem. 1994 Jun;55(2):155–167. doi: 10.1002/jcb.240550202. [DOI] [PubMed] [Google Scholar]
  89. Lamond S., Wallace H. M. Polyamine oxidase activity and growth in human cancer cells. Biochem Soc Trans. 1994 Nov;22(4):396S–396S. doi: 10.1042/bst022396s. [DOI] [PubMed] [Google Scholar]
  90. Levin V. A., Prados M. D., Yung W. K., Gleason M. J., Ictech S., Malec M. Treatment of recurrent gliomas with eflornithine. J Natl Cancer Inst. 1992 Sep 16;84(18):1432–1437. doi: 10.1093/jnci/84.18.1432. [DOI] [PubMed] [Google Scholar]
  91. Libby P. R., Ganis B., Bergeron R. J., Porter C. W. Characterization of human spermidine/spermine N1-acetyltransferase purified from cultured melanoma cells. Arch Biochem Biophys. 1991 Feb 1;284(2):238–244. doi: 10.1016/0003-9861(91)90291-p. [DOI] [PubMed] [Google Scholar]
  92. Libby P. R., Porter C. W. Inhibition of enzymes of polyamine back-conversion by pentamidine and berenil. Biochem Pharmacol. 1992 Aug 18;44(4):830–832. doi: 10.1016/0006-2952(92)90424-h. [DOI] [PubMed] [Google Scholar]
  93. Lindsay G. S., Wallace H. M. Changes in polyamine catabolism in HL-60 human promyelogenous leukaemic cells in response to etoposide-induced apoptosis. Biochem J. 1999 Jan 1;337(Pt 1):83–87. [PMC free article] [PubMed] [Google Scholar]
  94. Lipton A., Sheehan L. M., Kessler G. F., Jr Urinary polyamine levels in human cancer. Cancer. 1975 Feb;35(2):464–468. doi: 10.1002/1097-0142(197502)35:2<464::aid-cncr2820350225>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
  95. Lu L., Berkey K. A., Casero R. A., Jr RGFGIGS is an amino acid sequence required for acetyl coenzyme A binding and activity of human spermidine/spermine N1acetyltransferase. J Biol Chem. 1996 Aug 2;271(31):18920–18924. doi: 10.1074/jbc.271.31.18920. [DOI] [PubMed] [Google Scholar]
  96. Lu L., Stanley B. A., Pegg A. E. Identification of residues in ornithine decarboxylase essential for enzymic activity and for rapid protein turnover. Biochem J. 1991 Aug 1;277(Pt 3):671–675. doi: 10.1042/bj2770671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Luk G. D., Casero R. A., Jr Polyamines in normal and cancer cells. Adv Enzyme Regul. 1987;26:91–105. doi: 10.1016/0065-2571(87)90007-0. [DOI] [PubMed] [Google Scholar]
  98. Löser C., Fölsch U. R., Paprotny C., Creutzfeldt W. Polyamine concentrations in pancreatic tissue, serum, and urine of patients with pancreatic cancer. Pancreas. 1990 Mar;5(2):119–127. doi: 10.1097/00006676-199003000-00001. [DOI] [PubMed] [Google Scholar]
  99. Lövkvist Wallström E., Takao K., Wendt A., Vargiu C., Yin H., Persson L. Importance of the 3' untranslated region of ornithine decarboxylase mRNA in the translational regulation of the enzyme. Biochem J. 2001 Jun 1;356(Pt 2):627–634. doi: 10.1042/0264-6021:3560627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Mamont P. S., Danzin C., Kolb M., Gerhart F., Bey P., Sjoerdsma A. Marked and prolonged inhibition of mammalian ornithine decarboxylase in vivo by esters of (E)-2-(fluoromethyl)dehydroornithine. Biochem Pharmacol. 1986 Jan 15;35(2):159–165. doi: 10.1016/0006-2952(86)90509-5. [DOI] [PubMed] [Google Scholar]
  101. Marton L. J., Pegg A. E. Polyamines as targets for therapeutic intervention. Annu Rev Pharmacol Toxicol. 1995;35:55–91. doi: 10.1146/annurev.pa.35.040195.000415. [DOI] [PubMed] [Google Scholar]
  102. Marty C., Mori G., Sabini L., Rivarola V. Effects of alpha-difluoromethylornithine on the cyclin A expression in Hep-2 cells. Biocell. 2000 Apr;24(1):49–52. [PubMed] [Google Scholar]
  103. Matsufuji S., Matsufuji T., Miyazaki Y., Murakami Y., Atkins J. F., Gesteland R. F., Hayashi S. Autoregulatory frameshifting in decoding mammalian ornithine decarboxylase antizyme. Cell. 1995 Jan 13;80(1):51–60. doi: 10.1016/0092-8674(95)90450-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Matsui I., Pegg A. E. Effect of inhibitors of protein synthesis on rat liver spermidine N-acetyltransferase. Biochim Biophys Acta. 1981 Jul 17;675(3-4):373–378. doi: 10.1016/0304-4165(81)90028-3. [DOI] [PubMed] [Google Scholar]
  105. Matsui I., Pegg A. E. Increase in acetylation of spermidine in rat liver extracts brought about by treatment with carbon tetrachloride. Biochem Biophys Res Commun. 1980 Feb 12;92(3):1009–1015. doi: 10.1016/0006-291x(80)90802-5. [DOI] [PubMed] [Google Scholar]
  106. Matthews H. R. Polyamines, chromatin structure and transcription. Bioessays. 1993 Aug;15(8):561–566. doi: 10.1002/bies.950150811. [DOI] [PubMed] [Google Scholar]
  107. McCloskey D. E., Yang J., Woster P. M., Davidson N. E., Casero R. A., Jr Polyamine analogue induction of programmed cell death in human lung tumor cells. Clin Cancer Res. 1996 Mar;2(3):441–446. [PubMed] [Google Scholar]
  108. Melvin M. A., Melvin W. T., Keir H. M. Excretion of spermidine from BHK-21/C13 cells exposed to 6-thioguanosine. Cancer Res. 1978 Sep;38(9):3055–3058. [PubMed] [Google Scholar]
  109. Meyskens F. L., Jr, Emerson S. S., Pelot D., Meshkinpour H., Shassetz L. R., Einspahr J., Alberts D. S., Gerner E. W. Dose de-escalation chemoprevention trial of alpha-difluoromethylornithine in patients with colon polyps. J Natl Cancer Inst. 1994 Aug 3;86(15):1122–1130. doi: 10.1093/jnci/86.15.1122. [DOI] [PubMed] [Google Scholar]
  110. Milord F., Pépin J., Loko L., Ethier L., Mpia B. Efficacy and toxicity of eflornithine for treatment of Trypanosoma brucei gambiense sleeping sickness. Lancet. 1992 Sep 12;340(8820):652–655. doi: 10.1016/0140-6736(92)92180-n. [DOI] [PubMed] [Google Scholar]
  111. Mitchell J. L., Judd G. G., Bareyal-Leyser A., Ling S. Y. Feedback repression of polyamine transport is mediated by antizyme in mammalian tissue-culture cells. Biochem J. 1994 Apr 1;299(Pt 1):19–22. doi: 10.1042/bj2990019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Monti M. G., Ghiaroni S., Pernecco L., Barbieri D., Marverti G., Franceschi C. Polyamine depletion protects HL-60 cells from 2-deoxy-D-ribose-induced apoptosis. Life Sci. 1998;62(9):799–806. doi: 10.1016/s0024-3205(97)01181-8. [DOI] [PubMed] [Google Scholar]
  113. Morgan D. M. Uptake of polyamines by human endothelial cells. Characterization and lack of effect of agonists of endothelial function. Biochem J. 1992 Sep 1;286(Pt 2):413–417. doi: 10.1042/bj2860413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Morgan J. E., Blankenship J. W., Matthews H. R. Polyamines and acetylpolyamines increase the stability and alter the conformation of nucleosome core particles. Biochemistry. 1987 Jun 16;26(12):3643–3649. doi: 10.1021/bi00386a058. [DOI] [PubMed] [Google Scholar]
  115. Murakami Y., Fujita K., Kameji T., Hayashi S. Accumulation of ornithine decarboxylase-antizyme complex in HMOA cells. Biochem J. 1985 Feb 1;225(3):689–697. doi: 10.1042/bj2250689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Murakami Y., Matsufuji S., Hayashi S., Tanahashi N., Tanaka K. Degradation of ornithine decarboxylase by the 26S proteasome. Biochem Biophys Res Commun. 2000 Jan 7;267(1):1–6. doi: 10.1006/bbrc.1999.1706. [DOI] [PubMed] [Google Scholar]
  117. Murakami Y., Matsufuji S., Kameji T., Hayashi S., Igarashi K., Tamura T., Tanaka K., Ichihara A. Ornithine decarboxylase is degraded by the 26S proteasome without ubiquitination. Nature. 1992 Dec 10;360(6404):597–599. doi: 10.1038/360597a0. [DOI] [PubMed] [Google Scholar]
  118. Murray-Stewart Tracy, Wang Yanlin, Devereux Wendy, Casero Robert A., Jr Cloning and characterization of multiple human polyamine oxidase splice variants that code for isoenzymes with different biochemical characteristics. Biochem J. 2002 Dec 15;368(Pt 3):673–677. doi: 10.1042/BJ20021587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Muscari C., Guarnieri C., Stefanelli C., Giaccari A., Caldarera C. M. Protective effect of spermine on DNA exposed to oxidative stress. Mol Cell Biochem. 1995 Mar 23;144(2):125–129. doi: 10.1007/BF00944391. [DOI] [PubMed] [Google Scholar]
  120. Nairn L. M., Lindsay G. S., Woster P. M., Wallace H. M. Cytotoxicity of novel unsymmetrically substituted inhibitors of polyamine biosynthesis in human cancer cells. J Cell Physiol. 2000 Feb;182(2):209–213. doi: 10.1002/(SICI)1097-4652(200002)182:2<209::AID-JCP9>3.0.CO;2-8. [DOI] [PubMed] [Google Scholar]
  121. Nemoto T., Kamei S., Seyama Y., Kubota S. p53 independent G(1) arrest induced by DL-alpha-difluoromethylornithine. Biochem Biophys Res Commun. 2001 Jan 26;280(3):848–854. doi: 10.1006/bbrc.2000.4227. [DOI] [PubMed] [Google Scholar]
  122. Nichols C. G., Lopatin A. N. Inward rectifier potassium channels. Annu Rev Physiol. 1997;59:171–191. doi: 10.1146/annurev.physiol.59.1.171. [DOI] [PubMed] [Google Scholar]
  123. Nigro N. D., Bull A. W., Boyd M. E. Importance of the duration of inhibition on intestinal carcinogenesis by difluoromethylornithine in rats. Cancer Lett. 1987 May;35(2):153–158. doi: 10.1016/0304-3835(87)90039-5. [DOI] [PubMed] [Google Scholar]
  124. Niiranen Kirsi, Pietilä Marko, Pirttilä Terhi J., Järvinen Aki, Halmekytö Maria, Korhonen Veli-Pekka, Keinänen Tuomo A., Alhonen Leena, Jänne Juhani. Targeted disruption of spermidine/spermine N1-acetyltransferase gene in mouse embryonic stem cells. Effects on polyamine homeostasis and sensitivity to polyamine analogues. J Biol Chem. 2002 May 8;277(28):25323–25328. doi: 10.1074/jbc.M203599200. [DOI] [PubMed] [Google Scholar]
  125. Nilsson J., Grahn B., Heby O. Antizyme inhibitor is rapidly induced in growth-stimulated mouse fibroblasts and releases ornithine decarboxylase from antizyme suppression. Biochem J. 2000 Mar 15;346(Pt 3):699–704. [PMC free article] [PubMed] [Google Scholar]
  126. Nitta Takeshi, Igarashi Kazuei, Yamamoto Naoki. Polyamine depletion induces apoptosis through mitochondria-mediated pathway. Exp Cell Res. 2002 May 15;276(1):120–128. doi: 10.1006/excr.2002.5517. [DOI] [PubMed] [Google Scholar]
  127. Nowels K., Homma Y., Seidenfeld J., Oyasu R. Prevention of inhibitory effects of alpha-difluoromethylornithine on rat urinary bladder carcinogenesis by exogenous putrescine. Cancer Biochem Biophys. 1986 Oct;8(4):257–263. [PubMed] [Google Scholar]
  128. O'Brien T. G., Megosh L. C., Gilliard G., Soler A. P. Ornithine decarboxylase overexpression is a sufficient condition for tumor promotion in mouse skin. Cancer Res. 1997 Jul 1;57(13):2630–2637. [PubMed] [Google Scholar]
  129. O'Brien T. G., Simsiman R. C., Boutwell R. K. Induction of the polyamine-biosynthetic enzymes in mouse epidermis and their specificity for tumor promotion. Cancer Res. 1975 Sep;35(9):2426–2433. [PubMed] [Google Scholar]
  130. Oredsson S. M. Polyamine dependence of normal cell-cycle progression. Biochem Soc Trans. 2003 Apr;31(2):366–370. doi: 10.1042/bst0310366. [DOI] [PubMed] [Google Scholar]
  131. Packham G., Cleveland J. L. Ornithine decarboxylase is a mediator of c-Myc-induced apoptosis. Mol Cell Biol. 1994 Sep;14(9):5741–5747. doi: 10.1128/mcb.14.9.5741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Parchment R. E., Pierce G. B. Polyamine oxidation, programmed cell death, and regulation of melanoma in the murine embryonic limb. Cancer Res. 1989 Dec 1;49(23):6680–6686. [PubMed] [Google Scholar]
  133. Pegg A. E., Erwin B. G., Persson L. Induction of spermidine/spermine N1-acetyltransferase by methylglyoxal bis(guanylhydrazone). Biochim Biophys Acta. 1985 Oct 17;842(2-3):111–118. doi: 10.1016/0304-4165(85)90192-8. [DOI] [PubMed] [Google Scholar]
  134. Pegg A. E., Jones D. B., Secrist J. A., 3rd Effect of inhibitors of S-adenosylmethionine decarboxylase on polyamine content and growth of L1210 cells. Biochemistry. 1988 Mar 8;27(5):1408–1415. doi: 10.1021/bi00405a003. [DOI] [PubMed] [Google Scholar]
  135. Pegg A. E., McCann P. P. Polyamine metabolism and function. Am J Physiol. 1982 Nov;243(5):C212–C221. doi: 10.1152/ajpcell.1982.243.5.C212. [DOI] [PubMed] [Google Scholar]
  136. Pegg A. E. Polyamine metabolism and its importance in neoplastic growth and a target for chemotherapy. Cancer Res. 1988 Feb 15;48(4):759–774. [PubMed] [Google Scholar]
  137. Pegg A. E. Recent advances in the biochemistry of polyamines in eukaryotes. Biochem J. 1986 Mar 1;234(2):249–262. doi: 10.1042/bj2340249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Pegg A. E., Tang K. C., Coward J. K. Effects of S-adenosyl-1,8-diamino-3-thiooctane on polyamine metabolism. Biochemistry. 1982 Sep 28;21(20):5082–5089. doi: 10.1021/bi00263a036. [DOI] [PubMed] [Google Scholar]
  139. Penning L. C., Schipper R. G., Vercammen D., Verhofstad A. A., Denecker T., Beyaert R., Vandenabeele P. Sensitization of tnf-induced apoptosis with polyamine synthesis inhibitors in different human and murine tumour cell lines. Cytokine. 1998 Jun;10(6):423–431. doi: 10.1006/cyto.1997.0310. [DOI] [PubMed] [Google Scholar]
  140. Persson L., Pegg A. E. Studies of the induction of spermidine/spermine N1-acetyltransferase using a specific antiserum. J Biol Chem. 1984 Oct 25;259(20):12364–12367. [PubMed] [Google Scholar]
  141. Peña A., Reddy C. D., Wu S., Hickok N. J., Reddy E. P., Yumet G., Soprano D. R., Soprano K. J. Regulation of human ornithine decarboxylase expression by the c-Myc.Max protein complex. J Biol Chem. 1993 Dec 25;268(36):27277–27285. [PubMed] [Google Scholar]
  142. Piik K., Pösö H., Jänne J. Reversible inhibition of rat liver regeneration by 1,3-diamino-2-propanol, an inhibitor of ornithine decarboxylase. FEBS Lett. 1978 May 15;89(2):307–312. doi: 10.1016/0014-5793(78)80243-9. [DOI] [PubMed] [Google Scholar]
  143. Pines J. The cell cycle kinases. Semin Cancer Biol. 1994 Aug;5(4):305–313. [PubMed] [Google Scholar]
  144. Porter C. W., Bergeron R. J. Enzyme regulation as an approach to interference with polyamine biosynthesis--an alternative to enzyme inhibition. Adv Enzyme Regul. 1988;27:57–79. doi: 10.1016/0065-2571(88)90009-x. [DOI] [PubMed] [Google Scholar]
  145. Porter C. W., Cavanaugh P. F., Jr, Stolowich N., Ganis B., Kelly E., Bergeron R. J. Biological properties of N4- and N1,N8-spermidine derivatives in cultured L1210 leukemia cells. Cancer Res. 1985 May;45(5):2050–2057. [PubMed] [Google Scholar]
  146. Porter C. W., Ganis B., Libby P. R., Bergeron R. J. Correlations between polyamine analogue-induced increases in spermidine/spermine N1-acetyltransferase activity, polyamine pool depletion, and growth inhibition in human melanoma cell lines. Cancer Res. 1991 Jul 15;51(14):3715–3720. [PubMed] [Google Scholar]
  147. Porter C. W., McManis J., Casero R. A., Bergeron R. J. Relative abilities of bis(ethyl) derivatives of putrescine, spermidine, and spermine to regulate polyamine biosynthesis and inhibit L1210 leukemia cell growth. Cancer Res. 1987 Jun 1;47(11):2821–2825. [PubMed] [Google Scholar]
  148. Porter C. W., Miller J., Bergeron R. J. Aliphatic chain length specificity of the polyamine transport system in ascites L1210 leukemia cells. Cancer Res. 1984 Jan;44(1):126–128. [PubMed] [Google Scholar]
  149. Pyronnet S., Pradayrol L., Sonenberg N. A cell cycle-dependent internal ribosome entry site. Mol Cell. 2000 Apr;5(4):607–616. doi: 10.1016/s1097-2765(00)80240-3. [DOI] [PubMed] [Google Scholar]
  150. Pöso H., Jänne J. Inhibition of ornithine decarboxylase activity and spermidine accumulation in regenerating rat liver. Biochem Biophys Res Commun. 1976 Apr 19;69(4):885–892. doi: 10.1016/0006-291x(76)90456-3. [DOI] [PubMed] [Google Scholar]
  151. Ragione F. D., Pegg A. E. Purification and characterization of spermidine/spermine N1-acetyltransferase from rat liver. Biochemistry. 1982 Nov 23;21(24):6152–6158. doi: 10.1021/bi00267a020. [DOI] [PubMed] [Google Scholar]
  152. Ravanko K., Järvinen K., Paasinen-Sohns A., Hölttä E. Loss of p27Kip1 from cyclin E/cyclin-dependent kinase (CDK) 2 but not from cyclin D1/CDK4 complexes in cells transformed by polyamine biosynthetic enzymes. Cancer Res. 2000 Sep 15;60(18):5244–5253. [PubMed] [Google Scholar]
  153. Ray R. M., Zimmerman B. J., McCormack S. A., Patel T. B., Johnson L. R. Polyamine depletion arrests cell cycle and induces inhibitors p21(Waf1/Cip1), p27(Kip1), and p53 in IEC-6 cells. Am J Physiol. 1999 Mar;276(3 Pt 1):C684–C691. doi: 10.1152/ajpcell.1999.276.3.C684. [DOI] [PubMed] [Google Scholar]
  154. Redman C., Xu M. J., Peng Y. M., Scott J. A., Payne C., Clark L. C., Nelson M. A. Involvement of polyamines in selenomethionine induced apoptosis and mitotic alterations in human tumor cells. Carcinogenesis. 1997 Jun;18(6):1195–1202. doi: 10.1093/carcin/18.6.1195. [DOI] [PubMed] [Google Scholar]
  155. Ribeiro J. M., Carson D. A. Ca2+/Mg(2+)-dependent endonuclease from human spleen: purification, properties, and role in apoptosis. Biochemistry. 1993 Sep 7;32(35):9129–9136. doi: 10.1021/bi00086a018. [DOI] [PubMed] [Google Scholar]
  156. Roberts R. W., Crothers D. M. Stability and properties of double and triple helices: dramatic effects of RNA or DNA backbone composition. Science. 1992 Nov 27;258(5087):1463–1466. doi: 10.1126/science.1279808. [DOI] [PubMed] [Google Scholar]
  157. Rogers S., Wells R., Rechsteiner M. Amino acid sequences common to rapidly degraded proteins: the PEST hypothesis. Science. 1986 Oct 17;234(4774):364–368. doi: 10.1126/science.2876518. [DOI] [PubMed] [Google Scholar]
  158. Russell D. H., Durie B. G., Salmon S. E. Polyamines as predictors of success and failure in cancer chemotherapy. Lancet. 1975 Oct 25;2(7939):797–799. doi: 10.1016/s0140-6736(75)80009-2. [DOI] [PubMed] [Google Scholar]
  159. Russell D. H., Levy C. C., Schimpff S. C., Hawk I. A. Urinary polyamines in cancer patients. Cancer Res. 1971 Nov;31(11):1555–1558. [PubMed] [Google Scholar]
  160. Sakata K., Kashiwagi K., Igarashi K. Properties of a polyamine transporter regulated by antizyme. Biochem J. 2000 Apr 1;347(Pt 1):297–303. [PMC free article] [PubMed] [Google Scholar]
  161. Saydjari R., Alexander R. W., Upp J. R., Jr, Barranco S. C., Townsend C. M., Jr, Thompson J. C. Differential sensitivity of various human tumors to inhibition of polyamine biosynthesis in vivo. Int J Cancer. 1991 Jan 2;47(1):44–48. doi: 10.1002/ijc.2910470109. [DOI] [PubMed] [Google Scholar]
  162. Schipper R. G., Penning L. C., Verhofstad A. A. Involvement of polyamines in apoptosis. Facts and controversies: effectors or protectors? Semin Cancer Biol. 2000 Feb;10(1):55–68. doi: 10.1006/scbi.2000.0308. [DOI] [PubMed] [Google Scholar]
  163. Schuber F. Influence of polyamines on membrane functions. Biochem J. 1989 May 15;260(1):1–10. doi: 10.1042/bj2600001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Scorcioni F., Corti A., Davalli P., Astancolle S., Bettuzzi S. Manipulation of the expression of regulatory genes of polyamine metabolism results in specific alterations of the cell-cycle progression. Biochem J. 2001 Feb 15;354(Pt 1):217–223. doi: 10.1042/0264-6021:3540217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Seiler N., Delcros J. G., Moulinoux J. P. Polyamine transport in mammalian cells. An update. Int J Biochem Cell Biol. 1996 Aug;28(8):843–861. doi: 10.1016/1357-2725(96)00021-0. [DOI] [PubMed] [Google Scholar]
  166. Seiler N., Dezeure F. Polyamine transport in mammalian cells. Int J Biochem. 1990;22(3):211–218. doi: 10.1016/0020-711x(90)90332-w. [DOI] [PubMed] [Google Scholar]
  167. Sistonen L., Hölttä E., Lehväslaiho H., Lehtola L., Alitalo K. Activation of the neu tyrosine kinase induces the fos/jun transcription factor complex, the glucose transporter and ornithine decarboxylase. J Cell Biol. 1989 Nov;109(5):1911–1919. doi: 10.1083/jcb.109.5.1911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Soulet Denis, Covassin Laurence, Kaouass Mohammadi, Charest-Gaudreault René, Audette Marie, Poulin Richard. Role of endocytosis in the internalization of spermidine-C(2)-BODIPY, a highly fluorescent probe of polyamine transport. Biochem J. 2002 Oct 15;367(Pt 2):347–357. doi: 10.1042/BJ20020764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Stanek J., Caravatti G., Frei J., Furet P., Mett H., Schneider P., Regenass U. 4-Amidinoindan-1-one 2'-amidinohydrazone: a new potent and selective inhibitor of S-Adenosylmethionine decarboxylase. J Med Chem. 1993 Jul 23;36(15):2168–2171. doi: 10.1021/jm00067a014. [DOI] [PubMed] [Google Scholar]
  170. Stefanelli C., Bonavita F., Stanic' I., Pignatti C., Flamigni F., Guarnieri C., Caldarera C. M. Spermine triggers the activation of caspase-3 in a cell-free model of apoptosis. FEBS Lett. 1999 May 21;451(2):95–98. doi: 10.1016/s0014-5793(99)00549-9. [DOI] [PubMed] [Google Scholar]
  171. Stevens L. Ornithine decarboxylase activity in germinating conidia of Aspergillus nidulans. FEBS Lett. 1975 Nov 1;59(1):80–82. doi: 10.1016/0014-5793(75)80345-0. [DOI] [PubMed] [Google Scholar]
  172. Suzuki T., He Y., Kashiwagi K., Murakami Y., Hayashi S., Igarashi K. Antizyme protects against abnormal accumulation and toxicity of polyamines in ornithine decarboxylase-overproducing cells. Proc Natl Acad Sci U S A. 1994 Sep 13;91(19):8930–8934. doi: 10.1073/pnas.91.19.8930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Tabor C. W., Tabor H. Polyamines. Annu Rev Biochem. 1984;53:749–790. doi: 10.1146/annurev.bi.53.070184.003533. [DOI] [PubMed] [Google Scholar]
  174. Tadolini B. Polyamine inhibition of lipoperoxidation. The influence of polyamines on iron oxidation in the presence of compounds mimicking phospholipid polar heads. Biochem J. 1988 Jan 1;249(1):33–36. doi: 10.1042/bj2490033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Thomas T., Thomas T. J. Polyamines in cell growth and cell death: molecular mechanisms and therapeutic applications. Cell Mol Life Sci. 2001 Feb;58(2):244–258. doi: 10.1007/PL00000852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Thompson H. J., Meeker L. D., Herbst E. J., Ronan A. M., Minocha R. Effect of concentration of D,L-2-difluoromethylornithine on murine mammary carcinogenesis. Cancer Res. 1985 Mar;45(3):1170–1173. [PubMed] [Google Scholar]
  177. Tobias K. E., Kahana C. Intersubunit location of the active site of mammalian ornithine decarboxylase as determined by hybridization of site-directed mutants. Biochemistry. 1993 Jun 8;32(22):5842–5847. doi: 10.1021/bi00073a017. [DOI] [PubMed] [Google Scholar]
  178. Tome M. E., Fiser S. M., Payne C. M., Gerner E. W. Excess putrescine accumulation inhibits the formation of modified eukaryotic initiation factor 5A (eIF-5A) and induces apoptosis. Biochem J. 1997 Dec 15;328(Pt 3):847–854. doi: 10.1042/bj3280847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Tosaka Y., Tanaka H., Yano Y., Masai K., Nozaki M., Yomogida K., Otani S., Nojima H., Nishimune Y. Identification and characterization of testis specific ornithine decarboxylase antizyme (OAZ-t) gene: expression in haploid germ cells and polyamine-induced frameshifting. Genes Cells. 2000 Apr;5(4):265–276. doi: 10.1046/j.1365-2443.2000.00324.x. [DOI] [PubMed] [Google Scholar]
  180. Turchanowa Lyudmila, Shvetsov Alexander S., Demin Alexander V., Khomutov Alex R., Wallace Heather M., Stein Jürgen, Milovic Vladan. Insufficiently charged isosteric analogue of spermine: interaction with polyamine uptake, and effect on Caco-2 cell growth. Biochem Pharmacol. 2002 Aug 15;64(4):649–655. doi: 10.1016/s0006-2952(02)01225-x. [DOI] [PubMed] [Google Scholar]
  181. Uchida K., Seidenfeld J., Rademaker A., Oyasu R. Inhibitory action of alpha-difluoromethylornithine on N-butyl-N-(4-hydroxybutyl)nitrosamine-induced rat urinary bladder carcinogenesis. Cancer Res. 1989 Oct 1;49(19):5249–5253. [PubMed] [Google Scholar]
  182. Verma A. K., Boutwell R. K. Vitamin A acid (retinoic acid), a potent inhibitor of 12-O-tetradecanoyl-phorbol-13-acetate-induced ornithine decarboxylase activity in mouse epidermis. Cancer Res. 1977 Jul;37(7 Pt 1):2196–2201. [PubMed] [Google Scholar]
  183. Vujcic Slavoljub, Diegelman Paula, Bacchi Cyrus J., Kramer Debora L., Porter Carl W. Identification and characterization of a novel flavin-containing spermine oxidase of mammalian cell origin. Biochem J. 2002 Nov 1;367(Pt 3):665–675. doi: 10.1042/BJ20020720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Vujcic Slavoljub, Liang Ping, Diegelman Paula, Kramer Debora L., Porter Carl W. Genomic identification and biochemical characterization of the mammalian polyamine oxidase involved in polyamine back-conversion. Biochem J. 2003 Feb 15;370(Pt 1):19–28. doi: 10.1042/BJ20021779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Wallace H. M., Caslake R. Polyamines and colon cancer. Eur J Gastroenterol Hepatol. 2001 Sep;13(9):1033–1039. doi: 10.1097/00042737-200109000-00006. [DOI] [PubMed] [Google Scholar]
  186. Wallace H. M., Duthie J., Evans D. M., Lamond S., Nicoll K. M., Heys S. D. Alterations in polyamine catabolic enzymes in human breast cancer tissue. Clin Cancer Res. 2000 Sep;6(9):3657–3661. [PubMed] [Google Scholar]
  187. Wallace H. M., Fraser A. V. Polyamine analogues as anticancer drugs. Biochem Soc Trans. 2003 Apr;31(2):393–396. doi: 10.1042/bst0310393. [DOI] [PubMed] [Google Scholar]
  188. Wallace H. M., Keir H. M. Excretion of polyamines from baby hamster kidney cells (BHK-21/C13: effect of infection with Herpes Simplex Virus Type 1. J Gen Virol. 1981 Oct;56(Pt 2):251–258. doi: 10.1099/0022-1317-56-2-251. [DOI] [PubMed] [Google Scholar]
  189. Wallace H. M., Keir H. M. Factors affecting polyamine excretion from mammalian cells in culture. Inhibitors of polyamine biosynthesis. FEBS Lett. 1986 Jan 1;194(1):60–63. doi: 10.1016/0014-5793(86)80051-5. [DOI] [PubMed] [Google Scholar]
  190. Wallace H. M., Keir H. M. Uptake and excretion of polyamines from baby hamster kidney cells (BHK-21/C13). The effect of serum on confluent cell cultures. Biochim Biophys Acta. 1981 Aug 5;676(1):25–30. doi: 10.1016/0304-4165(81)90005-2. [DOI] [PubMed] [Google Scholar]
  191. Wallace H. M., Mackarel A. J. Regulation of polyamine acetylation and efflux in human cancer cells. Biochem Soc Trans. 1998 Nov;26(4):571–575. doi: 10.1042/bst0260571. [DOI] [PubMed] [Google Scholar]
  192. Wallace H. M., Nuttall M. E., Robinson F. C. Acetylation of spermidine and methylglyoxal bis(guanylhydrazone) in baby-hamster kidney cells (BHK-21/C13). Biochem J. 1988 Jul 1;253(1):223–227. doi: 10.1042/bj2530223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Wallace H. M. Polyamines in human health. Proc Nutr Soc. 1996 Mar;55(1B):419–431. doi: 10.1079/pns19960039. [DOI] [PubMed] [Google Scholar]
  194. Wallace H. M. Polyamines: specific metabolic regulators or multifunctional polycations? Biochem Soc Trans. 1998 Nov;26(4):569–571. doi: 10.1042/bst0260569. [DOI] [PubMed] [Google Scholar]
  195. Wang Y., Devereux W., Stewart T. M., Casero R. A., Jr Characterization of the interaction between the transcription factors human polyamine modulated factor (PMF-1) and NF-E2-related factor 2 (Nrf-2) in the transcriptional regulation of the spermidine/spermine N1-acetyltransferase (SSAT) gene. Biochem J. 2001 Apr 1;355(Pt 1):45–49. doi: 10.1042/0264-6021:3550045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Wang Y., Devereux W., Woster P. M., Stewart T. M., Hacker A., Casero R. A., Jr Cloning and characterization of a human polyamine oxidase that is inducible by polyamine analogue exposure. Cancer Res. 2001 Jul 15;61(14):5370–5373. [PubMed] [Google Scholar]
  197. Wang Y., Xiao L., Thiagalingam A., Nelkin B. D., Casero R. A., Jr The identification of a cis-element and a trans-acting factor involved in the response to polyamines and polyamine analogues in the regulation of the human spermidine/spermine N1-acetyltransferase gene transcription. J Biol Chem. 1998 Dec 18;273(51):34623–34630. doi: 10.1074/jbc.273.51.34623. [DOI] [PubMed] [Google Scholar]
  198. Webb H. K., Wu Z., Sirisoma N., Ha H. C., Casero R. A., Jr, Woster P. M. 1-(N-alkylamino)-11-(N-ethylamino)-4,8-diazaundecanes: simple synthetic polyamine analogues that differentially alter tubulin polymerization. J Med Chem. 1999 Apr 22;42(8):1415–1421. doi: 10.1021/jm980603+. [DOI] [PubMed] [Google Scholar]
  199. Weeks C. E., Herrmann A. L., Nelson F. R., Slaga T. J. alpha-Difluoromethylornithine, an irreversible inhibitor of ornithine decarboxylase, inhibits tumor promoter-induced polyamine accumulation and carcinogenesis in mouse skin. Proc Natl Acad Sci U S A. 1982 Oct;79(19):6028–6032. doi: 10.1073/pnas.79.19.6028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Weeks R. S., Vanderwerf S. M., Carlson C. L., Burns M. R., O'Day C. L., Cai F., Devens B. H., Webb H. K. Novel lysine-spermine conjugate inhibits polyamine transport and inhibits cell growth when given with DFMO. Exp Cell Res. 2000 Nov 25;261(1):293–302. doi: 10.1006/excr.2000.5033. [DOI] [PubMed] [Google Scholar]
  201. Williams K. Interactions of polyamines with ion channels. Biochem J. 1997 Jul 15;325(Pt 2):289–297. doi: 10.1042/bj3250289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Woster P. M., Black A. Y., Duff K. J., Coward J. K., Pegg A. E. Synthesis and biological evaluation of S-adenosyl-1,12-diamino-3-thio-9-azadodecane, a multisubstrate adduct inhibitor of spermine synthase. J Med Chem. 1989 Jun;32(6):1300–1307. doi: 10.1021/jm00126a026. [DOI] [PubMed] [Google Scholar]
  203. Wright R. K., Buehler B. A., Schott S. N., Rennert O. M. Spermine and spermidine, modulators of the cell surface enzyme adenylate cyclase. Pediatr Res. 1978 Aug;12(8):830–833. doi: 10.1203/00006450-197808000-00005. [DOI] [PubMed] [Google Scholar]
  204. Wrighton C., Busslinger M. Direct transcriptional stimulation of the ornithine decarboxylase gene by Fos in PC12 cells but not in fibroblasts. Mol Cell Biol. 1993 Aug;13(8):4657–4669. doi: 10.1128/mcb.13.8.4657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Wu Y., Woster P. M. S-(5'-deoxy-5'-adenosyl)-1-ammonio-4-(methylsulfonio)-2-cyclopentene: A potent, enzyme-activated irreversible inhibitor of S-adenosylmethionine decarboxylase. J Med Chem. 1992 Aug 21;35(17):3196–3201. doi: 10.1021/jm00095a015. [DOI] [PubMed] [Google Scholar]
  206. Wyatt I., Soames A. R., Clay M. F., Smith L. L. The accumulation and localisation of putrescine, spermidine, spermine and paraquat in the rat lung. In vitro and in vivo studies. Biochem Pharmacol. 1988 May 15;37(10):1909–1918. doi: 10.1016/0006-2952(88)90536-9. [DOI] [PubMed] [Google Scholar]
  207. Xiao L., Swank R. A., Matthews H. R. Photoaffinity polyamines: sequence-specific interactions with DNA. Nucleic Acids Res. 1991 Jul 11;19(13):3701–3708. doi: 10.1093/nar/19.13.3701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Zhu C., Lang D. W., Coffino P. Antizyme2 is a negative regulator of ornithine decarboxylase and polyamine transport. J Biol Chem. 1999 Sep 10;274(37):26425–26430. doi: 10.1074/jbc.274.37.26425. [DOI] [PubMed] [Google Scholar]

Articles from Biochemical Journal are provided here courtesy of The Biochemical Society

RESOURCES