Cerebral ischemia enhances polyamine oxidation: identification of enzymatically formed 3-aminopropanal as an endogenous mediator of neuronal and glial cell death

S Ivanova, G I Botchkina, Y Al-Abed, M Meistrell 3rd, F Batliwalla, J M Dubinsky, C Iadecola, H Wang, P K Gregersen, J W Eaton, K J Tracey, S Ivanova, G I Botchkina, Y Al-Abed, M Meistrell 3rd, F Batliwalla, J M Dubinsky, C Iadecola, H Wang, P K Gregersen, J W Eaton, K J Tracey

Abstract

To elucidate endogenous mechanisms underlying cerebral damage during ischemia, brain polyamine oxidase activity was measured in rats subjected to permanent occlusion of the middle cerebral artery. Brain polyamine oxidase activity was increased significantly within 2 h after the onset of ischemia in brain homogenates (15.8 +/- 0.9 nmol/h/mg protein) as compared with homogenates prepared from the normally perfused contralateral side (7.4 +/- 0.5 nmol/h/mg protein) (P <0.05). The major catabolic products of polyamine oxidase are putrescine and 3-aminopropanal. Although 3-aminopropanal is a potent cytotoxin, essential information was previously lacking on whether 3-aminopropanal is produced during cerebral ischemia. We now report that 3-aminopropanal accumulates in the ischemic brain within 2 h after permanent forebrain ischemia in rats. Cytotoxic levels of 3-aminopropanal are achieved before the onset of significant cerebral cell damage, and increase in a time-dependent manner with spreading neuronal and glial cell death. Glial cell cultures exposed to 3-aminopropanal undergo apoptosis (LD50 = 160 microM), whereas neurons are killed by necrotic mechanisms (LD50 = 90 microM). The tetrapeptide caspase 1 inhibitor (Ac-YVAD-CMK) prevents 3-aminopropanal-mediated apoptosis in glial cells. Finally, treatment of rats with two structurally distinct inhibitors of polyamine oxidase (aminoguanidine and chloroquine) attenuates brain polyamine oxidase activity, prevents the production of 3-aminopropanal, and significantly protects against the development of ischemic brain damage in vivo. Considered together, these results indicate that polyamine oxidase-derived 3-aminopropanal is a mediator of the brain damaging sequelae of cerebral ischemia, which can be therapeutically modulated.

Figures

Figure 1
Figure 1
Polyamine oxidase activity increases during cerebral ischemia, and is inhibited by aminoguanidine and chloroquine. Polyamine oxidase activity was measured in brain homogenates prepared as described in Methods. Data shown are mean ± SE; n = 3. Normal, sham-operated control brain homogenate; Ischemia Vehicle, homogenate prepared 2 h after the onset of middle cerebral artery occlusion; Ischemia AG, addition of aminoguanidine (1 mM) at time = −5 min before spermine; Ischemia CHLQ, addition of chloroquine (1 mM) at time = −5 min before spermine. *, P <0.05 versus normal; #, P <0.05 versus ischemia vehicle.
Figure 2
Figure 2
(a) 1H-NMR spectroscopy (DMSO-d6 and CDCl3, 270 MHz) of the products of reacting 3-aminopropanal with 2,4-dinitrophenylhydrazine. NMR revealed the presence of anti and syn isomers with resonance at δ8.83 and δ11.35. (Inset) Structure of the principle condensation products. (b) Electrospray ionization mass spectrum of synthetic dansylated 3-aminopropanal-2,4-dinitrophenylhydrazone. 3-aminopropanal was derivatized with 2,4-dinitrophenylhydrazine and dansyl chloride, and the reaction products were subjected to HPLC as outlined in Materials and Methods. The inset shows the HPLC profile of the separable geometric isomers. Note that the EIMS of the HPLC-purified fractions revealed the expected molecular ion at m/z 251. (c) EIMS of derivatized ischemic brain homogenate. Animals were subjected to permanent focal cerebral ischemia and after 25 h, brain tissue was obtained for homogenization and derivatization as described in Materials and Methods. The inset shows the HPLC profile, and the mass spectrum confirms the expected molecular ion of the HPLC-purified fractions at m/z 251.
Figure 2
Figure 2
(a) 1H-NMR spectroscopy (DMSO-d6 and CDCl3, 270 MHz) of the products of reacting 3-aminopropanal with 2,4-dinitrophenylhydrazine. NMR revealed the presence of anti and syn isomers with resonance at δ8.83 and δ11.35. (Inset) Structure of the principle condensation products. (b) Electrospray ionization mass spectrum of synthetic dansylated 3-aminopropanal-2,4-dinitrophenylhydrazone. 3-aminopropanal was derivatized with 2,4-dinitrophenylhydrazine and dansyl chloride, and the reaction products were subjected to HPLC as outlined in Materials and Methods. The inset shows the HPLC profile of the separable geometric isomers. Note that the EIMS of the HPLC-purified fractions revealed the expected molecular ion at m/z 251. (c) EIMS of derivatized ischemic brain homogenate. Animals were subjected to permanent focal cerebral ischemia and after 25 h, brain tissue was obtained for homogenization and derivatization as described in Materials and Methods. The inset shows the HPLC profile, and the mass spectrum confirms the expected molecular ion of the HPLC-purified fractions at m/z 251.
Figure 2
Figure 2
(a) 1H-NMR spectroscopy (DMSO-d6 and CDCl3, 270 MHz) of the products of reacting 3-aminopropanal with 2,4-dinitrophenylhydrazine. NMR revealed the presence of anti and syn isomers with resonance at δ8.83 and δ11.35. (Inset) Structure of the principle condensation products. (b) Electrospray ionization mass spectrum of synthetic dansylated 3-aminopropanal-2,4-dinitrophenylhydrazone. 3-aminopropanal was derivatized with 2,4-dinitrophenylhydrazine and dansyl chloride, and the reaction products were subjected to HPLC as outlined in Materials and Methods. The inset shows the HPLC profile of the separable geometric isomers. Note that the EIMS of the HPLC-purified fractions revealed the expected molecular ion at m/z 251. (c) EIMS of derivatized ischemic brain homogenate. Animals were subjected to permanent focal cerebral ischemia and after 25 h, brain tissue was obtained for homogenization and derivatization as described in Materials and Methods. The inset shows the HPLC profile, and the mass spectrum confirms the expected molecular ion of the HPLC-purified fractions at m/z 251.
Figure 3
Figure 3
Brain 3-aminopropanal levels increase during cerebral ischemia. Brain 3-aminopropanal levels were measured by HPLC in rats subjected to permanent focal cerebral ischemia. 3-Aminopropanal was not detected in sham-operated controls. Note that 3-aminopropanal tissue levels increased markedly within 2 h after middle cerebral artery occlusion, and continued to increase for at least 25 h. Data shown are mean ± SE, n = 3 animals/group. *, P <0.05 versus t = 0 h by analysis of variance.
Figure 4
Figure 4
Brain damaging effects of intracortically administered polyamines, and protection with polyamine oxidase inhibitors. (a) Brain damage mediated by intracortical microinjection of spermine, spermidine, and 3-aminopropanal, but not putrescine. Data shown are volume of brain damage (mm3) as measured by integrating the area of negative TTC staining over the entire brain hemisphere in animals injected with the polyamines shown; mean ± SE, n = 6–8/group. *, P <0.05 versus vehicle. (b) Aminoguanidine and chloroquine protection against intracortical spermine toxicity. All animals received intracortical spermine (25 μg in 2 μl) by stereotactically guided microinjection. Experimental animals were treated with aminoguanidine or chloroquine simultaneously with the intracortical spermine injectate in the following doses: systemic aminoguanidine was 320 mg/kg intraperitoneal 30-min pretreatment followed by subsequent doses of 110 mg/kg intraperitoneally each 8 h after intracortical spermine; intracortical aminoguanidine was administered by a single dose (320 mg/kg) given simultaneously with intracortical spermine; chloroquine was administered by a single intraperitoneal dose (25 mg/kg) 30 min before the spermine injection. Data shown are infarct volume (mm3) assessed quantitatively 48 h after the intracortical spermine injection (mean ± SE, n = 6–8/group). *, P <0.05 versus spermine/vehicle.
Figure 4
Figure 4
Brain damaging effects of intracortically administered polyamines, and protection with polyamine oxidase inhibitors. (a) Brain damage mediated by intracortical microinjection of spermine, spermidine, and 3-aminopropanal, but not putrescine. Data shown are volume of brain damage (mm3) as measured by integrating the area of negative TTC staining over the entire brain hemisphere in animals injected with the polyamines shown; mean ± SE, n = 6–8/group. *, P <0.05 versus vehicle. (b) Aminoguanidine and chloroquine protection against intracortical spermine toxicity. All animals received intracortical spermine (25 μg in 2 μl) by stereotactically guided microinjection. Experimental animals were treated with aminoguanidine or chloroquine simultaneously with the intracortical spermine injectate in the following doses: systemic aminoguanidine was 320 mg/kg intraperitoneal 30-min pretreatment followed by subsequent doses of 110 mg/kg intraperitoneally each 8 h after intracortical spermine; intracortical aminoguanidine was administered by a single dose (320 mg/kg) given simultaneously with intracortical spermine; chloroquine was administered by a single intraperitoneal dose (25 mg/kg) 30 min before the spermine injection. Data shown are infarct volume (mm3) assessed quantitatively 48 h after the intracortical spermine injection (mean ± SE, n = 6–8/group). *, P <0.05 versus spermine/vehicle.
Figure 5
Figure 5
Histology of 3-aminopropanal–induced cell death. Animals were anesthetized and perfused intracardially with 100 ml saline followed by 200–300 ml of 4% formaldehyde in PBS. Brains were removed, postfixed for 2 hr in the same fixative, and then incubated overnight in 30% sucrose at 4°C. Ten micrometer sections were prepared and airdried. TUNEL staining was performed as outlined in Methods. (A) Low magnification microphotograph demonstrating the site of microinjecting needle (large arrow) and areas of 3-aminopropanal–induced prominent cell death (small arrows). (B) High power microphotograph taken within the marked area showing coexistence of apoptosis (TUNEL-positive cell on the right) and necrosis (eosin-stained cell on the left). (C) High magnification of area indicated in A showing the extent of cellular degeneration.
Figure 6
Figure 6
Apoptosis in glial cells exposed to 3-aminopropanal. (a) DNA gel electrophoresis of glial-like and neuronal-like cells exposed to 3-aminopropanal for 13 h as described in Materials and Methods; 7.5 μg of DNA was loaded per lane for the glial cells (lanes B and C), and 15 μg was loaded per lane for the neuronal cells (lanes D and E). Lane A, 1 kb DNA size marker; lane B, 3-aminopropanal–treated (160 μM); lane C, vehicle control; lane D, vehicle-treated; lane E, 3-aminopropanal–treated (90 μM). (b) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated glial cells (160 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, 76% of the cell population stained TUNEL positive. (c) FACS® histogram of vehicle-treated glial cells revealed no evidence of apoptosis (0.13% TUNEL-positive cells). (d) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated neuronal cells (90 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, only 5.02% of the cell population of stained TUNEL positive, though a forward/side scatter of the same cell population revealed 55.7% cell death upon electronic gating (data not shown). (e) FACS® histogram of vehicle-treated neuronal cells showed no evidence of apoptosis (0.53% TUNEL-positive cells). Electronic gating for forward/side scatter showed 9.1% cell death under these conditions. (  f  ) Annexin V FITC/propidium iodide (PI) staining of 3-aminopropanal– treated neuronal cells, as above, revealed no evidence of apoptosis (1.8% apoptotic cells). Necrotic cells comprised 68.97% of the cell population. (g) Annexin V FITC/propidium iodide staining of vehicle-treated neuronal cells revealed no evidence of apoptosis (1.02% of the cell population) or necrosis (1.58% of the cell population).
Figure 6
Figure 6
Apoptosis in glial cells exposed to 3-aminopropanal. (a) DNA gel electrophoresis of glial-like and neuronal-like cells exposed to 3-aminopropanal for 13 h as described in Materials and Methods; 7.5 μg of DNA was loaded per lane for the glial cells (lanes B and C), and 15 μg was loaded per lane for the neuronal cells (lanes D and E). Lane A, 1 kb DNA size marker; lane B, 3-aminopropanal–treated (160 μM); lane C, vehicle control; lane D, vehicle-treated; lane E, 3-aminopropanal–treated (90 μM). (b) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated glial cells (160 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, 76% of the cell population stained TUNEL positive. (c) FACS® histogram of vehicle-treated glial cells revealed no evidence of apoptosis (0.13% TUNEL-positive cells). (d) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated neuronal cells (90 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, only 5.02% of the cell population of stained TUNEL positive, though a forward/side scatter of the same cell population revealed 55.7% cell death upon electronic gating (data not shown). (e) FACS® histogram of vehicle-treated neuronal cells showed no evidence of apoptosis (0.53% TUNEL-positive cells). Electronic gating for forward/side scatter showed 9.1% cell death under these conditions. (  f  ) Annexin V FITC/propidium iodide (PI) staining of 3-aminopropanal– treated neuronal cells, as above, revealed no evidence of apoptosis (1.8% apoptotic cells). Necrotic cells comprised 68.97% of the cell population. (g) Annexin V FITC/propidium iodide staining of vehicle-treated neuronal cells revealed no evidence of apoptosis (1.02% of the cell population) or necrosis (1.58% of the cell population).
Figure 6
Figure 6
Apoptosis in glial cells exposed to 3-aminopropanal. (a) DNA gel electrophoresis of glial-like and neuronal-like cells exposed to 3-aminopropanal for 13 h as described in Materials and Methods; 7.5 μg of DNA was loaded per lane for the glial cells (lanes B and C), and 15 μg was loaded per lane for the neuronal cells (lanes D and E). Lane A, 1 kb DNA size marker; lane B, 3-aminopropanal–treated (160 μM); lane C, vehicle control; lane D, vehicle-treated; lane E, 3-aminopropanal–treated (90 μM). (b) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated glial cells (160 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, 76% of the cell population stained TUNEL positive. (c) FACS® histogram of vehicle-treated glial cells revealed no evidence of apoptosis (0.13% TUNEL-positive cells). (d) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated neuronal cells (90 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, only 5.02% of the cell population of stained TUNEL positive, though a forward/side scatter of the same cell population revealed 55.7% cell death upon electronic gating (data not shown). (e) FACS® histogram of vehicle-treated neuronal cells showed no evidence of apoptosis (0.53% TUNEL-positive cells). Electronic gating for forward/side scatter showed 9.1% cell death under these conditions. (  f  ) Annexin V FITC/propidium iodide (PI) staining of 3-aminopropanal– treated neuronal cells, as above, revealed no evidence of apoptosis (1.8% apoptotic cells). Necrotic cells comprised 68.97% of the cell population. (g) Annexin V FITC/propidium iodide staining of vehicle-treated neuronal cells revealed no evidence of apoptosis (1.02% of the cell population) or necrosis (1.58% of the cell population).
Figure 6
Figure 6
Apoptosis in glial cells exposed to 3-aminopropanal. (a) DNA gel electrophoresis of glial-like and neuronal-like cells exposed to 3-aminopropanal for 13 h as described in Materials and Methods; 7.5 μg of DNA was loaded per lane for the glial cells (lanes B and C), and 15 μg was loaded per lane for the neuronal cells (lanes D and E). Lane A, 1 kb DNA size marker; lane B, 3-aminopropanal–treated (160 μM); lane C, vehicle control; lane D, vehicle-treated; lane E, 3-aminopropanal–treated (90 μM). (b) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated glial cells (160 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, 76% of the cell population stained TUNEL positive. (c) FACS® histogram of vehicle-treated glial cells revealed no evidence of apoptosis (0.13% TUNEL-positive cells). (d) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated neuronal cells (90 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, only 5.02% of the cell population of stained TUNEL positive, though a forward/side scatter of the same cell population revealed 55.7% cell death upon electronic gating (data not shown). (e) FACS® histogram of vehicle-treated neuronal cells showed no evidence of apoptosis (0.53% TUNEL-positive cells). Electronic gating for forward/side scatter showed 9.1% cell death under these conditions. (  f  ) Annexin V FITC/propidium iodide (PI) staining of 3-aminopropanal– treated neuronal cells, as above, revealed no evidence of apoptosis (1.8% apoptotic cells). Necrotic cells comprised 68.97% of the cell population. (g) Annexin V FITC/propidium iodide staining of vehicle-treated neuronal cells revealed no evidence of apoptosis (1.02% of the cell population) or necrosis (1.58% of the cell population).
Figure 6
Figure 6
Apoptosis in glial cells exposed to 3-aminopropanal. (a) DNA gel electrophoresis of glial-like and neuronal-like cells exposed to 3-aminopropanal for 13 h as described in Materials and Methods; 7.5 μg of DNA was loaded per lane for the glial cells (lanes B and C), and 15 μg was loaded per lane for the neuronal cells (lanes D and E). Lane A, 1 kb DNA size marker; lane B, 3-aminopropanal–treated (160 μM); lane C, vehicle control; lane D, vehicle-treated; lane E, 3-aminopropanal–treated (90 μM). (b) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated glial cells (160 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, 76% of the cell population stained TUNEL positive. (c) FACS® histogram of vehicle-treated glial cells revealed no evidence of apoptosis (0.13% TUNEL-positive cells). (d) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated neuronal cells (90 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, only 5.02% of the cell population of stained TUNEL positive, though a forward/side scatter of the same cell population revealed 55.7% cell death upon electronic gating (data not shown). (e) FACS® histogram of vehicle-treated neuronal cells showed no evidence of apoptosis (0.53% TUNEL-positive cells). Electronic gating for forward/side scatter showed 9.1% cell death under these conditions. (  f  ) Annexin V FITC/propidium iodide (PI) staining of 3-aminopropanal– treated neuronal cells, as above, revealed no evidence of apoptosis (1.8% apoptotic cells). Necrotic cells comprised 68.97% of the cell population. (g) Annexin V FITC/propidium iodide staining of vehicle-treated neuronal cells revealed no evidence of apoptosis (1.02% of the cell population) or necrosis (1.58% of the cell population).
Figure 6
Figure 6
Apoptosis in glial cells exposed to 3-aminopropanal. (a) DNA gel electrophoresis of glial-like and neuronal-like cells exposed to 3-aminopropanal for 13 h as described in Materials and Methods; 7.5 μg of DNA was loaded per lane for the glial cells (lanes B and C), and 15 μg was loaded per lane for the neuronal cells (lanes D and E). Lane A, 1 kb DNA size marker; lane B, 3-aminopropanal–treated (160 μM); lane C, vehicle control; lane D, vehicle-treated; lane E, 3-aminopropanal–treated (90 μM). (b) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated glial cells (160 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, 76% of the cell population stained TUNEL positive. (c) FACS® histogram of vehicle-treated glial cells revealed no evidence of apoptosis (0.13% TUNEL-positive cells). (d) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated neuronal cells (90 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, only 5.02% of the cell population of stained TUNEL positive, though a forward/side scatter of the same cell population revealed 55.7% cell death upon electronic gating (data not shown). (e) FACS® histogram of vehicle-treated neuronal cells showed no evidence of apoptosis (0.53% TUNEL-positive cells). Electronic gating for forward/side scatter showed 9.1% cell death under these conditions. (  f  ) Annexin V FITC/propidium iodide (PI) staining of 3-aminopropanal– treated neuronal cells, as above, revealed no evidence of apoptosis (1.8% apoptotic cells). Necrotic cells comprised 68.97% of the cell population. (g) Annexin V FITC/propidium iodide staining of vehicle-treated neuronal cells revealed no evidence of apoptosis (1.02% of the cell population) or necrosis (1.58% of the cell population).
Figure 6
Figure 6
Apoptosis in glial cells exposed to 3-aminopropanal. (a) DNA gel electrophoresis of glial-like and neuronal-like cells exposed to 3-aminopropanal for 13 h as described in Materials and Methods; 7.5 μg of DNA was loaded per lane for the glial cells (lanes B and C), and 15 μg was loaded per lane for the neuronal cells (lanes D and E). Lane A, 1 kb DNA size marker; lane B, 3-aminopropanal–treated (160 μM); lane C, vehicle control; lane D, vehicle-treated; lane E, 3-aminopropanal–treated (90 μM). (b) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated glial cells (160 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, 76% of the cell population stained TUNEL positive. (c) FACS® histogram of vehicle-treated glial cells revealed no evidence of apoptosis (0.13% TUNEL-positive cells). (d) FACS® histogram showing TUNEL staining of 3-aminopropanal–treated neuronal cells (90 μM, 13 h) as outlined in Materials and Methods. As indicated by the marker, only 5.02% of the cell population of stained TUNEL positive, though a forward/side scatter of the same cell population revealed 55.7% cell death upon electronic gating (data not shown). (e) FACS® histogram of vehicle-treated neuronal cells showed no evidence of apoptosis (0.53% TUNEL-positive cells). Electronic gating for forward/side scatter showed 9.1% cell death under these conditions. (  f  ) Annexin V FITC/propidium iodide (PI) staining of 3-aminopropanal– treated neuronal cells, as above, revealed no evidence of apoptosis (1.8% apoptotic cells). Necrotic cells comprised 68.97% of the cell population. (g) Annexin V FITC/propidium iodide staining of vehicle-treated neuronal cells revealed no evidence of apoptosis (1.02% of the cell population) or necrosis (1.58% of the cell population).
Figure 7
Figure 7
Inhibition of caspase 1 but not of caspase 3 blocks 3-aminopropanal–induced glial apoptosis. Cells were pretreated with (a) the caspase 1 inhibitor (Ac-YVAD-CMK) or (b) the caspase 3 inhibitor (Ac-DEVD-CHO) at concentrations 0.4 (triangles) or 40 μM (circles) for 3 h, followed by treatment with 3-aminopropanal for an additional 5 h, and then were analyzed for cell viability by the 3-(4,5-dimethylthiazol-2-yl)- 2,5-diphenyltetrazolium bromide assay. Controls consisted of DMSO-treated cells (squares) to assess for nonspecific solvent effects. Data are mean ± SE, n = 3 wells/experiment.
Figure 7
Figure 7
Inhibition of caspase 1 but not of caspase 3 blocks 3-aminopropanal–induced glial apoptosis. Cells were pretreated with (a) the caspase 1 inhibitor (Ac-YVAD-CMK) or (b) the caspase 3 inhibitor (Ac-DEVD-CHO) at concentrations 0.4 (triangles) or 40 μM (circles) for 3 h, followed by treatment with 3-aminopropanal for an additional 5 h, and then were analyzed for cell viability by the 3-(4,5-dimethylthiazol-2-yl)- 2,5-diphenyltetrazolium bromide assay. Controls consisted of DMSO-treated cells (squares) to assess for nonspecific solvent effects. Data are mean ± SE, n = 3 wells/experiment.

References

    1. Zhang L, Xheng X, Paupard MC, Wang AP, Santchi L, Friedman LK, Zukin RS, Bennett MVL. Spermine potentiation of recombinant N-methyl-D-aspartate receptors is affected by subunit composition. Proc Natl Acad Sci USA. 1994;91:10883–10887.
    1. Harman RJ, Shaw GG. The spontaneous and evoked release of spermine from rat brain in vitro. . Br J Pharmacol. 1981;73:165–174.
    1. Bergeron RJ, Weimar WR, Wu Q, Feng Y, McManis JS. Polyamine analogue regulation of NMDA MK-801 binding: a structure–activity study. J Med Chem. 1996;39:5257–5266.
    1. Glantz L, Nates JL, Trembovler V, Bass R, Shohami E. Polyamines induce blood-brain barrier disruption and edema formation in the rat. J Basic Clin Physiol Pharmacol. 1996;7:1–10.
    1. Dempsey RJ, Roy MW, Meyer K, Tain HH, Olson JW. Polyamine and prostaglandin markers in focal cerebral ischemia. Neurosurgery (Baltim) 1985;17:635–640.
    1. Schmitz MP, Combs DJ, Dempsey RJ. Difluoromethylornithine decreases postischemic brain edema and blood-brain barrier breakdown. Neurosurgery (Baltim) 1993;33:882–888.
    1. Anderson DJ, Crossland J, Shaw GG. The actions of spermidine and spermine on the central nervous system. Neuropharmacology. 1975;14:571–577.
    1. Doyle KM, Shaw GG. The mechanism of the neurotoxic effects of spermidine. Biochem Soc Trans. 1994;22 (Suppl.):386S.
    1. Kindy MS, Hu Y, Dempsey RJ. Blockade of ornithine decarboxylase enzyme protects against ischemic brain damage. J Cereb Blood Flow Metab. 1994;14:1040–1045.
    1. Marton LJ, Pegg AE. Polyamines as targets for therapeutic intervention. Annu Rev Pharmacol Toxicol. 1995;35:55–91.
    1. Lövkvist-Wallström E, Stjernborg-Ulvsbäck L, Scheffler IE, Persson L. Regulation of mammalian ornithine decarboxylase. Studies on the induction of the enzyme by hypotonic stress. Eur J Biochem. 1995;231:40–44.
    1. Pegg AE, Shantz LM, Coleman CS. Ornithine decarboxylase: structure, function and translational regulation. Biochem Soc Trans. 1994;22:846–852.
    1. Paschen W. Polyamine metabolism in different pathological states of the brain. Mol Chem Neuropathol. 1992;16:241–271.
    1. Traynelis SF, Hartley M, Heinemann SF. Control of proton sensitivity of the NMDA receptor by RNA splicing and polyamines. Science. 1995;268:873–876.
    1. Traynelis SF, Cull-Candy SG. Pharmacological properties and H+ sensitivity of excitatory amino acid receptor channels in rat cerebellar granule neurones. J Physiol (Lond) 1991;433:727–763.
    1. Sullivan JM, Traynelis SF, Chen HS, Escobar W, Heinemann SF, Lipton SA. Identification of two cysteine residues that are required for redox modulation of the NMDA subtype of glutamate receptor. Neuron. 1994;13:929–936.
    1. Fahey JM, Pritchard GA, Miller LG. Polyamine neurotoxicity is antagonized by dizocilpine in cultured chick cortical neurons. Neurosci Lett. 1993;161:109–112.
    1. Paschen W, Schmidt-Kastner R, Djuricic B, Meese C, Linn F, Hossmann KA. Polyamine changes in reversible cerebral ischemia. J Neurochem. 1987;49:35–37.
    1. Paschen W. Polyamine metabolism in reversible cerebral ischemia. Cerebrovasc Brain Metab Rev. 1992;4:59–88.
    1. Morgan, D.M.L. 1989. Polyamine oxidases and oxidized polyamines. In The Physiology of Polyamines. U. Bachrach and Y.M. Heimer, editors. CRC Publications, Cleveland, OH. 203–229.
    1. Paschen W, Hallmayer J, Rohn G. Relationship between putrescine content and density of ischemic cell damage in the brain of mongolian gerbils: effect of nimodipine and barbiturate. Acta Neuropathol. 1988;76:388–394.
    1. Seiler N, Bolkenius FN. Polyamine reutilization and turnover in brain. Neurochem Res. 1985;10:529–544.
    1. Seiler N, Bolkenius N, Rennert OM. Interconversion, catabolism and elimination of the polyamines. Med Biol. 1981;59:334–346.
    1. Bolkenius FN, Seiler N. Developmental aspects of polyamine interconversion in rat brain. Int J Dev Neurosci. 1986;4:217–224.
    1. Bolkenius FN, 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;838:69–76.
    1. Seiler, N. 1995. Polyamine oxidase, properties and functions. Prog. Brain Res. 333–344.
    1. Morgan DML. Polyamines. Essays Biochem. 1987;23:82–115.
    1. Houen G, Bock K, Jensen AL. HPLC and NMR investigation of the serum amine oxidase catalyzed oxidation of polyamines. Acta Chem Scand. 1994;48:52–60.
    1. Bouzyk E, Rosiek O. Clastogenic and cytotoxic effects of spermine oxidation products in mouse lymphoma L5178Y cells. Cancer Lett. 1988;39:93–99.
    1. Brunton VG, Grant MH, Wallace HM. Spermine toxicity in BHK-21/C13 cells in the presence of bovine serum: the effect of aminoguanidine. Toxicol In Vitro. 1994;8:337–341.
    1. Gaugas JM, Dewey DL. Evidence for serum binding of oxidized spermine and its potent G1-phase inhibition of cell proliferation. Br J Cancer. 1978;39:548–557.
    1. Morgan DML, Bachrach U, Assaraf G, Harri E, Golenser J. The effect of purified aminoaldehydes produced by polyamine oxidation on the development in vitro of Plasmodium falciparumin normal and glucose-6-phosphate-dehydrogenase–deficient erythrocytes. J Biochem. 1986;236:97–101.
    1. Ferrante A, Rzepczyk CM, Saul AJ. Polyamine oxidase-mediated trypanosome killing: the role of hydrogen peroxide and aldehydes. J Immunol. 1984;133:2157–2162.
    1. Parchment RE, Pierce GB. Polyamine oxidation, programmed cell death, and regulation of melanoma in the murine embryonic limb. Cancer Res. 1989;49:6680–6686.
    1. Kurihara H, Matsuzaki S, Yamazaki H, Tsukahara T, Tamura M. Relation between tissue polyamine levels and malignancy in primary brain tumors. Neurosurgery (Baltim) 1993;32:372–375.
    1. Gahl WA, Pitot HC. Reversal by aminoguanidine of the inhibition of proliferation of human fibroblasts by spermidine and spermine. Chem-Biol Interact. 1978;22:91–98.
    1. Henle KJ, Moss AJ, Nagle WA. Mechanism of spermidine cytotoxicity at 37 degrees C and 43 degrees C in Chinese hamster ovary cells. Cancer Res. 1986;46:175–182.
    1. Milani D, Guidolin D, Facci L, Pozzan T, Buso M, Leon A, Skaper SD. Excitatory amino acid–induced alterations of cytoplasmic free Ca2+ in individual cerebellar granule neurons: role in neurotoxicity. J Neurosci Res. 1991;28:434–441.
    1. Zimmerman GA, Meistrell M, III, Bloom O, Cockroft KM, Bianchi M, Risucci D, Broome J, Farmer P, Cerami A, Vlassara H, Tracey KJ. Neurotoxicity of advanced glycation endproducts during focal stroke, and neuroprotective effects of aminoguanidine. Proc Natl Acad Sci USA. 1995;92:3744–3748.
    1. Cockroft KM, Meistrell M, III, Zimmerman GA, Risucci D, Bloom O, Cerami A, Tracey KJ. Cerebroprotective effects of aminoguanidine in a rodent model of stroke. Stroke. 1996;27:1393–1398.
    1. Bederson JB, Pitts LH, Germano SM, Nishimura MC, Davis RL, Bartkowski HM. Evaluation of 2,3,5-triphenyltetrazolium chloride as a stain for detection and quantification of experimental cerebral infarction in rats. Stroke. 1986;17:1304–1308.
    1. Whitmore WL, Slotkin TA. A simplified method for isocratic HPLC analysis of polyamines. Experientia (Basel) 1985;41:1209–1211.
    1. Bachrach U, Reches B. Enzymatic assay for spermine and spermidine. Anal Biochem. 1966;17:38–48.
    1. Dickinson, R., and N. Jacobsen. 1970. A new and sensitive test for detection of aldehydes: formation of 6-mercapto-3-substituted-s-triazolo(4,3-b-)-s-tetrazines. Chem. Commun. 1719–1720.
    1. Ponten J, MacIntyre EH. Long term culture of normal and neoplastic human glia. Acta Pathol Microbiol Scand. 1968;74:465–486.
    1. Bluestein HG. Neurocytotoxic antibodies in serum of patients with systemic lupus erythematosus. Proc Natl Acad Sci USA. 1978;75:3965–3969.
    1. Sieuwerts AM, Klijn JG, Peters HA, Foekens JA. The MTT tetrazolium salt assay scrutinized: how to use this assay reliably to measure metabolic activity of cell cultures in vitro for the assessment of growth characteristics, IC50-values and cell survival. Eur J Clin Chem Clin Biochem. 1995;33:813–823.
    1. Meistrell ME, III, Botchkina GI, Wang H, Di Santo E, Cockroft KM, Bloom O, Vishnubhakat JM, Ghezzi P, Tracey KJ. Tumor necrosis factor is a brain-damaging cytokine in cerebral ischemia. Shock. 1997;8:341–348.
    1. Holtta E. Oxidation of spermidine and spermine in rat liver: purification and properties of polyamine oxidase. Biochemistry. 1977;16:91–100.
    1. Flayeh KA. Spermidine oxidase activity in serum of normal and schizophrenic subjects. Clin Chem. 1988;34:401–403.
    1. Gahl WA, Pitot HC. Polyamine degradation in foetal and adult bovine serum. Biochem J. 1982;202:603–611.
    1. Seiler N. Polyamine metabolism. Digestion. 1990;46:319–330.
    1. Gavrieli Y, Sherman Y, Ben-Sasson SA. Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation. J Cell Biol. 1992;119:493–501.
    1. Furuya Y, Ohta S, Ito H. Apoptosis of androgen-independent mammary and prostate cell lines induced by topoisomerase inhibitors: common pathway of gene regulation. Anticancer Res. 1997;17:2089–2093.
    1. Bhat RV, DiRocco R, Marcy VR, Flood DG, Zhu Y, Dobrzanski P, Siman R, Scott R, Contreras PC, Miller M. Increased expression of IL-1beta converting enzyme in hippocampus after ischemia: selective localization in microglia. J Neurosci. 1996;16:4146–4154.
    1. Loddick SA, MacKenzie A, Rothwell NJ. An ICE inhibitor, z-VAD-DCB attenuates ischaemic brain damage in the rat. Neuroreport. 1996;7:1465–1468.
    1. Hara H, Friedlander RM, Gagliardini V, Ayata C, Fink K, Huang Z, Shimizu-Sasamata M, Yuan J, Moskowitz MA. Inhibition of interleukin 1beta converting enzyme family proteases reduces ischemic and excitotoxic neuronal damage. Proc Natl Acad Sci USA. 1997;94:2007–2012.
    1. Friedlander RM, Gagliardini V, Hara H, Fink KB, Li W, MacDonald G, Fishman MC, Greenberg AH, Moskowitz MA, Yuan J. Expression of a dominant negative mutant of interleukin-1β converting enzyme in transgenic mice prevents neuronal cell death induced by trophic factor withdrawal and ischemic brain injury. J Exp Med. 1997;185:933–940.
    1. Gillardon F, Bottiger B, Schmitz B, Zimmermann M, Hossmann KA. Activation of CPP-32 protease in hippocampal neurons following ischemia and epilepsy. Brain Res Mol Brain Res. 1997;50:16–22.
    1. Zhang F, Casey RM, Ross E, Iadecola C. Aminoguanidine ameliorates and L-arginine worsens brain damage from intraluminal middle cerebral artery occlusion. Stroke. 1996;27:317–323.
    1. Gilad GM, Gilad VH, Wyatt RJ. Accumulation of exogenous polyamines in gerbil brain after ischemia. Mol Chem Neuropathol. 1993;18:197–210.
    1. Harada J, Sugimoto M. Polyamines prevent apoptotic cell death in cultured cerebellar granule neurons. Brain Res. 1997;753:251–259.
    1. Xie X, Tome ME, Gerner EW. Loss of intracellular putrescine pool-size regulation induces apoptosis. Exp Cell Res. 1997;230:386–392.
    1. Casero RA, Jr, Pegg AE. Spermidine/spermine N1-acetyltransferase—the turning point in polyamine metabolism. FASEB (Fed Am Soc Exp Biol) J. 1993;7:653–661.
    1. Tipnis UR, He GY, Khan MF. Differential induction of polyamine oxidase activity in liver and heart of iron-overloaded rats. J Toxicol Environ Health. 1997;51:235–244.
    1. Li L, Hamilton RF, Jr, Taylor DE, Holian A. Acrolein-induced cell death in human alveolar macrophages. Toxicol Appl Pharmacol. 1997;145:331–339.
    1. Fernandez C, Sharrard RM, Talbot M, Reed BD, Monks N. Evaluation of the significance of polyamines and their oxidases in the aetiology of human cervical carcinoma. Br J Cancer. 1995;72:1194–1199.
    1. Coyle JT, Puttfarcken P. Oxidative stress, glutamate, and neurodegenerative disorders. Science. 1993;262:689–695.
    1. Rothwell NJ, Strijbos PJ. Cytokines in neurodegeneration and repair. Int J Dev Neurosci. 1995;13:179–185.
    1. Irikura K, Huang PL, Ma J, Lee WS, Dalkara T, Fishman MC, Dawson TM, Snyder SH, Moskowitz MA. Cerebrovascular alterations in mice lacking neuronal nitric oxide synthase gene expression. Proc Natl Acad Sci USA. 1995;92:6823–6827.
    1. Rothwell NJ, Relton JK. Involvement of interleukin-1 and lipocortin-1 in ischaemic brain damage. Cerebrovasc Brain Metab Rev. 1993;5:178–198.
    1. Taupin V, Toulmond S, Serrano A, Benavides J, Zavala F. Increase in IL-6, IL-1 and TNF levels in rat brain following traumatic lesion. Influence of pre- and post-traumatic treatment with Ro5 4864, a peripheral-type (p site) benzodiazepine ligand. J Neuroimmunol. 1993;42:177–185.
    1. Saito K, Suyama K, Nishida K, Sei Y, Basile AS. Early increases in TNF-alpha, IL-6 and IL-1 beta levels following transient cerebral ischemia in gerbil brain. Neurosci Lett. 1996;206:149–152.
    1. Choi DW. Excitotoxic cell death. J Neurobiol. 1992;23:1261–1276.
    1. Montague PR, Gancayco CD, Winn MJ, Marchase RB, Friedlander JJ. Role of NO production in NMDA receptor-mediated neurotransmitter release in cerebral cortex. Science. 1994;263:973–976.
    1. Hara H, Fink K, Endres M, Friedlander RM, Gagliardini V, Yuan J, Moskowitz MA. Attenuation of transient focal cerebral ischemic injury in transgenic mice expressing a mutant ICE inhibitory protein. J Cereb Blood Flow Metab. 1997;17:370–375.
    1. Zhang M, Caragine T, Wang H, Cohen PS, Botchkina G, Soda K, Bianchi M, Ulrich P, Cerami A, Sherry B, Tracey KJ. Spermine inhibits proinflammatory cytokine synthesis in human mononuclear cells: a counterregulatory mechanism that restrains the immune response. J Exp Med. 1997;185:1–10.
    1. Seiler N, Bolkenius FN, Knodgen B, Mamont P. Polyamine oxidase in rat tissues. Biochim Biophys Acta. 1980;615:480–488.

Source: PubMed

3
Abonnieren