AU2004201624A1 - Oxidant Scavengers - Google Patents

Oxidant Scavengers Download PDF

Info

Publication number
AU2004201624A1
AU2004201624A1 AU2004201624A AU2004201624A AU2004201624A1 AU 2004201624 A1 AU2004201624 A1 AU 2004201624A1 AU 2004201624 A AU2004201624 A AU 2004201624A AU 2004201624 A AU2004201624 A AU 2004201624A AU 2004201624 A1 AU2004201624 A1 AU 2004201624A1
Authority
AU
Australia
Prior art keywords
sod
mice
bond
cells
mntbap
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2004201624A
Inventor
Ines Batinic-Haberle
James D. Crapo
Brian J. Day
Rodney J. Folz
Bruce A. Freeman
Irwin Fridovich
Tim Oury
Michael P. Trova
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
UAB Research Foundation
Duke University
Original Assignee
UAB Research Foundation
Duke University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU63870/96A external-priority patent/AU725602B2/en
Priority claimed from AU53511/00A external-priority patent/AU769217B2/en
Application filed by UAB Research Foundation, Duke University filed Critical UAB Research Foundation
Publication of AU2004201624A1 publication Critical patent/AU2004201624A1/en
Abandoned legal-status Critical Current

Links

Description

1. 1
AUSTRALIA
Patents Act 1990 COMPLETE SPECIFICATION FOR A STANDARD PATENT Name of Applicants: DUKE UNIVERSITY.
UNIVERSITY OF ALABAMA BIRMINGHAM RESEARCH FOUNDATION Actual Inventors: Michael P. T CRAPO, Irwin OURY, Brian J.
FOLZ, Bruce A.
BATINIC-HABERLE
ROVA, James D.
FRIDOVICH, Tim DAY, Rodney J.
FREEMAN and Ines Address for Service: CULLEN CO., Patent Trade Mark Attorneys, 239 George Street, Brisbane, Qld. 4000, Australia.
Invention Title: OXIDANT SCAVENGERS The following statement is a full description of this invention, including the best method of performing it known to us: OXIDANT SCAVENGERS This is a divisional application of Australian Patent Application No. 63870/96.
TECHNICAL FIELD The present invention relates, in general, to a method of modulating physiological and pathological processes and, in particular, to a method of modulating intra- and extracellular levels of oxidants such as superoxide radicals and hydrogen peroxide and thereby processes in which such radicals are a participant. The invention also relates to compounds and compositions suitable for use in such methods.
BACKGROUND
Oxidants are produced as part of the normal metabolism of all cells but also are an important component of the pathogenesis of many disease processes.
Reactive oxygen species, for example, are critical elements of the pathogenesis of diseases of the lung, the central nervous system and skeletal muscle. Oxygen free radicals also play a role in modulating the effects of nitric oxide In this context, they contribute to the pathogenesis of vascular disorders, inflammatory diseases and the aging process.
S A critical balance of defensive enzymes against oxidants is required to maintain normal cell and organ function. Superoxide dismutases (SODs), a family of metalloenzymes which catalyze the intra- and extracellular conversion of 02- into H202 plus 02, and represent the first line of defense against the detrimental effects of superoxide radicals. .Mammals produce three distinct SODs. One is a dimeric copperand zinc-containing enzyme (CuZn SOD) found in the cytosol of all cells. A second is a tetrameric manganese-containing SOD (Mn SOD) found within mitochondria, and the third is a tetrameric, glycosylated, copper- and zinc-containing enzyme (EC-SOD) found in the extracellular fluids and bound to the extracellular matrix. Several other important antioxidant enzymes are known to exist within cells, including catalase and glucathione peroxidase. While extracellular fluids and the extracellular matrix contain only small amounts of these enzymes, other extracellular antioxidants are known to exist, including radical scavengers and inhibitors of lipid peroxidation, such as ascorbic acid, uric acid, and a-tocopherol (Halliwell et al, Arch. Biochem. Biophys. 280:1 (1990)) The relative lack of extracellular antioxidanz enzymes may reflect the possible function of extraceliular reactive oxygen species as bioeffector molecules (Halliwell et al, Arch. Biochem. Biophys. 280:1 (1990)).
The relative deficiency of such enzymes may also result in greater susceptibility to extracellular oxidant stresses- The enzyme EC-SOD, in many extracellular locations, exists only at low concentrations. While its physiologic role in vivo is yet -to be defined, in many extracellular locations, EC-SOD is not thought to .function as a bulk scavenger of 02". As indicated above, EC-SOD is a tetrameric Cu/Zn-ccntaining glycoprotein with a subunit molecular weight of 30,000 (Marklund, Proc. Natl. Acad. Sci. USA 79:7634 (1982); Tibell et al, Proc. Natl. Acad. Sci. USA 84:6634 (1987); see also USP 5,130,245 and WO 91/04315). Biochemical data suggest that EC-SOD binds to heparan sulfate proteoglycans on endothelial cells, where it has been speculated to serve as a "protective coat" (Marklund,
J.
Clin. Invest. 74:1398 (1984); Karlsson et Biochem.
J. 255:223 (1988)). Endothelial cells secrete both 02- 3 (Halliwell, Free Radical Res. Commun. 5:315 (1989)) and endothelium-derived relaxing factor, putatively identified as nitric oxide (Noak and Murphy, in Oxidative Stress Oxidants and Antioxidants, eds Sies, H.
(Academic, San Diego), pp. 445-489 (1991)). NOfunctions as a vasoregulator and as a regulator of neurotransmission (Schuman and Madison, Science 254:1503 (1991)). NO- can, however, be toxic to neurons in some situations (Dawson et al, Proc. Natl. Acad. Sci. USA 88:6368 (1991)). 02- is known to inactivate NO'-induced vasorelaxation (Gryglewski et al, Nature 320:454 (1986); Rubanyi and Vanhoutte. Am. J. Physiol. 250:H822 f1986); Rubanyi and Vanhoutte, Am. J. Physiol. 250:H813 (1986); Bult et al, Br. J. Pharmacol. 95:1308 (1988); Nucci et al, Proc. Natl. Acad. Sci. USA 85:2334 (1988)). Thus, a possible function for EC-SOD is to protect NO- released from cells from 02--mediated inactivation.
The reaction of 02- with NO- is also known to produce a potentially toxic intermediate in the form of the peroxynitrite anion (ONOO-) (Beckman et al, Proc.
0 Natl. Acad. Sci. USA 87:1620 (1990); Mulligan e- al, Proc. Natl. Acad. Sci. USA 88:6338 (1991); Hogg et a, Biochem: J. 281:419 (1992); Matheis et al, Am. J.
Physiol. 262:H616 (1992)). Thus EC-SOD may alsc function to prevent the formation of ONOO-.
Surprisingly, it has been found that EC-SOD increases, rather than decreases, central nervous system 02 toxicity and that this effect of EC-SOD occurs through modulation of NO-. This result implicates
NO-
as an important mediator in 02 toxicity. The invention thus relates to methods of manipulating nitric oxide function that involve the use of extracellular antioxidants.
In addition to superoxide radicals, hydrogen peroxide is an oxidant species that is produced under a wide variety of conditions of oxidant stress. The invention thus also provides a method of manipulating hydrogen peroxide levels.
The methods of the invention find application in various disease and non-disease states in which oxidative stress plays a role, including inflammation.
In a broader sense, the invention relates generally to methods of modulating intra- and extracellular processes in which an oxidant such as 02- or hydrogen peroxide is a participant.
SUMMARY OF THE INVENTION According to one aspect of the present invention there is provided a method of treating a pathological condition of a patient resulting from peroxynitrite accumulation comprising administering to said patient an effective amount of a compound having the activity of a catalytic antioxidant under conditions such that said treatment is effected.
According to a second aspect of the present invention there is provided method of treating a disorder resulting from aberrant smooth muscle function in a patient in need of such treatment comprising administering to said patient an effective amount of a mimetic of SOD, EC-SOD, catalase or peroxidase under conditions such that said treatment is effected.
According to a third aspect of the present invention there is provided an oxidant scavenger of the formula: 2 3 R3- R2-
R
20 1 5 R R 2 R 3
N
ON N I H 14 1 R RR
R
3 R2- Ri R R e2 R3 or a pharmaceutically acceptable salt thereof, or metal complex thereof wherein said metal is selected from the group consisting of manganese, copper and iron, wherein: each Ri' is independently a bond, -cor-, 9 00 0 C01H _CC-NH-, or 0CSY wherein Y" is an alkyl group, and wherein indicates bonding to R 2 at any position and indicates bonding to R2' and the Ri' phenyl substituent at any position; each R2' is independently a bond, or -(CH 2 )nwherein n is 1-4, each R 3 is independently -OH, OY", -NO 2 -CN, -NH 2 -COOH, -COY", -COO, or a heterocyclic group, wherein Y" is as defined above and is a primary, secondary, tertiary or quaternary amine, 00 wherein when Ri' is Rc- R 3 is not COOH, COY" or COO-, 0 00 0 wherein when Ri' is CN C, or c
R
3 is not -NO 2 and wherein collectively, are not -H, S' OOH (CH)3 Or C CH According to a fourth aspect of the present invention there is provided a method of modulating NO* function as a neurotransmitter in a mammal comprising contacting said mammal with an amount of an oxidant scavenger of the formula, R R2- Ri R- R2- R 3 is
H
bn j /N H N
N
R3~ R2- RI A
R
Ry- R3 13 12 or a pharmaceutically acceptable salt thereof, wherein:
R
1 is a bond, -CO N' NO, 7 so 3 H, 7S 03 x or _aY wherein X is a halogen and Y is an alkyl group and wherein indicates bonding to R 2 at any position and indicates bonding to R2 and the substituent at any position; and R2 is a bond, -(CY' 2
(CY'
2 -CY'=CY')n
(CY'
2 -CY'2-CH=CH)n-, or wherein Y' is hydrogen or an alkyl group and wherein n is I to 8; and R3 is -OH, -NH 2 -COOH, -COO-,
SO
3 H, -S03-, -C-PO 3
H
2 or -C-PO3H-, wherein Y" is an alkyl group, optionally complexed with a metal selected from the group consisting of manganese, copper and iron, sufficient to effect said modulation.
According to a fifth aspect of the present invention there is provided an oxidant scavenger of the formula: r3 'r,2 2 3
N\
P H 1 P S N
N
H P
N
R3- R2- R, R1 R. R 3 P P or a pharmaceutically acceptable salt thereof, wherein: R1i is a bond, C- No 2 0 sOH S X or Y wherein X is a 2 0 halogen and Y is an alkyl group and wherein indicates bonding to R2 at any position and indicates bonding to R2 and the substituent at any position; and each R 2 is independently, a bond,
(CY'
2 )nr -(CY'2-CY' 2 -CH=CH)n-, or
(CY'
2 -CO)n wherein Y' is hydrogen or an alkyl group and wherein n is 1 to 8; and each R 3 is, independently -OH, -NH 2
N
3 -COOH, -COO, -S03H, -SO3-, -C-PO3H 2 or -C-P03H-, wherein Y" is an alkyl group, each P is, independently, an electron withdrawing group or hydrogen wherein at least one P is not hydrogen, or
IN
P P or a pharmaceutically acceptable salt thereof, wherein each R 1 is independently a bond, ~cf H I COH or r 3, wherein Y" is an alkyl group, and wherein indicates bonding to R2' at any position and indicates bonding to R 2 and the Ri' phenyl substituent at any position; each R2' is independently a bond, or -(CH2)nwherein n is 1-4, each R3' is independently -OH, OY", -NO 2 -CN, -NH 2 -COOH, -COY", -COO, or a heterocyclic group, wherein Y" is as defined above and is a primary, secondary, tertiary or quaternary amine, each P is independently, an electron withdrawing group or hydrogen wherein at least one P is not hydrogen; wherein said oxidant scavenger is a metal complex wherein said metal is selected from the group consisting of manganese, copper or iron, wherein when said metal is copper and all P's are Br, and when RI is and R2 is a bond, R3 is not -CH 3 BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 shows the EC-SOD expression vector used to construct transgenic mice. Transgenic mice were generated with the Eco-RI-Xbal fragment. IVS1: Intervening sequence 1 from the human p-actin promoter.
Figure 2 shows the Northern analysis of tissues from transgenic mice. Twenty Vpg of total RNA from the tissues of transgenic mice were denatured with glyoxal and electrophoresed through a 1.2% agarose gel and blotted onto nitrocellulose. -The filter was probed with the entire human EC-SOD cDNA. The 2.5 Kb band corresponds to mRNA of the human EC-SOD transgene containing the 1 Kb intervening sequence (see Figure 1).
The 1.5 Kb band corresponds to the fully processed mRNA of the human EC-SOD transgene.
Figure 3 shows the percent survival of transgenic and nontransgenic mice exposed to 6 ATA oxygen for minutes. Mice were injected with saline or given mg/kg N-t-nitro-L-arginine (LNNA) i.p. 10 minutes 0 before compression. 400 mg/kg of diethyldithiocarbamate (DDC) in saline was injected i.p. 55 min before compression. *pcO.017 tested by X2 with Bonferroni correction, compared to transgenic saline treated mice.
Figure 4 shows time to onset of first seizure in transgenic and nontransgenic mice exposed to 6 ATA oxygen. Mice were injected with saline or given mg/kg N-1-nitro-L-arginine (LNNA) i.p. 10 minutes before beginning compression. 400 mg/kg diethyldithiocarbamate (DDC) was injected i.p. 55 minutes prior to compression. Results are expressed as mean S.D. of time to first seizure with zero time taken once chamber reached 6 ATA. *p<0.05 tested by analysis of variance with the Scheffe F-test compared to nontransgenic saline treated mice.
Figure 5 shows the effecc of diethyldithiocarbamate and 9-mercaproethanol on survival in 6 ATA oxygen for minutes. (C57BL/6 X C3H)Fl mice were injected i.p.
Ii with saline, 180 mg/kg E-mercapcoethanol or 400 mg/kg diethyldithiocarbamate (DDC) in saline 55 min.
before compression. *p<0.025 tested by X2 with Bonferroni correction compared to saline treated mice.
Figure 6 shows the seizure latency in wild-type mice exposed to 6 ATA oxygen after being treated with saline or 20 mg/kg N-1-nitro-L-arginine (LNNA) cr mg/kg N-1-nicro-L-arginine plus 50 mg/kg L-arginine (LNNA L-Arg). *p<0.05 tested by analysis of variance with a paired Student's t-test compared to saline treated mice.
Figure 7 shows the percent survival in wild-type mice exposed to 6 ATA oxygen. Mice were given an i.p.
injection of normal saline (0.008 cc/g) or 20 mg/kg N-L-nitro-L-arginine (LNNA) (0.008 cc/g) 15 minuoes prior to compression. The mice were exposed Ctc ATA of oxygen for 20 minutes (n=10, saline only), 25 minutes both groups), 30 minutes (n=10, saline only), minutes LNNA only), 75 minutes (n=12, LNNA 0 only), 90 minutes (n=14, LNNA only), 105 minutes (n=6 LNNA only) and 120 minutes LNNA only) and percent survival was measured for each group.
Figure 8 shows the survival dose response curve for N-[-nitro-L-arginine (LNNA). Wild-type mice were given an i.p. injection of normal saline (0.008 cc/g) or 0, 2, 20, or 30 mg/kg LNNA (0.008 cc/g) 15 minutes prior to compression and then exposed to 75 minutes a: 6 ATA oxygen. Percent survival was calculated for each treatment group.
Figure 9 shows the percent survival in wild-type mice pretreated with saline, 20 mg/kg N-o-nitro-Larginine (LNNA), or 20 mg/kg N- -nitro-L-arginine plus mg/kg L-arginine (LNNA L-Arg) and then exposed to minutes of 6 ATA oxygen. *p<0.05 tested with a xsquare test with Bonferroni correction.
Figure 10 shows the percent survival in transgenic LO and nontransgenic mice exposed to 6 ATA oxygen for minutes. Mice were injected with saline or given mg/kg N-I-nitro-L-arginine (LNNA) i.p. 10 minutes before compression. *p<0.05 tested by X 2 compared to nontransgenic saline treated mice. p<0.05 tested by
X
2 compared to transgenic saline treated mice.
Figure 11 shows the comparison of edema formation in EC-SOD transgenic mice to edema formation in nontransgenic littermates after cold-induced injury to the right cerebral hemisphere as well as in non-injured mice. Values are presented as mean standard error.
*p<0.05 compared to Edema Index of respective nontransgenic controls using a paired Student's t-test.
Figure 12 shows the effect of augmented levels of EC-SOD on vascular permeability changes after coldinduced brain injury. Vascular permeability is demonstrated as Evan's blue leakage in the injuried right 7 cerebral hemispheres of nontransgenic (control) and EC- SOD transgenic mice.
Figure 13 shows a Western blot analysis of rh-EC-SOD and a human lung homogenate to demonstrate antibody specificity. Proteins were separated on a 0.75 mm SDS-pclyacrylamide gel and transferred to nitrocellulose. Proteins were hybridized with the antibody to recombinant human EC-SOD (4.3 pg/ml) and the antibody was detected by hybridization with 125 1-Protein-A followed by autoradiography. The EC-SOD lane contained 0.05 pg of pure recombinant human type C EC-SOD lane protein. The lung lane contained 10 Ag of a 20,000 x g supernatant of a human lung homogenate.
Figures 14A-14C show the light microscopic immunchistochemical localization of EC-SOD in human lung. Tissues were labeled using the antibody to recombinant human EC-SOD (5.4 mg/ml; anti-EC-SOD) or the same antibody in which the anti-EC-SOD IgG was absorbed out using purified recombinant EC-SOD attached to CNBrsepharose (EC-SOD absorbed). Antibody was detected using a biotin/streptavidin-hoseradish peroxidase labeling technicue. A, Large elastic pulmonary artery labeled with anti-EC-SOD. Note labeling around smooth muscle cells beneath the endothelium and beneath the elastic layer of the vessel (short arrow), and the lack of labeling for EC-SOC on the surface of endothelial cells (open arrow) and on elastin (long arrow).
B,
Muscular pulmonary artery labeled with anti-EC-SOD.
Note high amount of labeling in the connective tissue matrix surrounding the vessel and lymphatics (long arrow), in the matrix surrounding smooth muscle cells (short arrow), and the lack of labeling on the surface of endothelial cells (open arrow). C, Muscular pulmonary artery labeled with EC-SOD absorbed antisera.
The absorption of anti-EC-SOD IgG abolished all labeling in the muscular vessel. (Bars 50 gm).
Figures 15A-15C show the immunohistochemical 0 localization of EC-SOD in human lung. Tissues were labeled using the antibody to recombinant human EC-SOD (5.4 mg/ml; anti-EC-SOD) Antibody was detected using a biotin/streptavidin-horseradish peroxidase labeling technique. A, Large cartilaginous airway labeled with anti-EC-SOD. Note the intense labeling for EC-SOD in the matrix around smooth muscle cells (short arrow), between the epithelial cells (long arrow), and the lack of labeling on the surface of the epithelial cells (open arrows), and in the matrix of cartilage (asterisk). B, Noncartilaginous airway labeled with anti-EC-SOD. Note the intense labeling for EC-SOD throughout the entire matrix beneath the epithelium (short arrow), and the lack of labeling on the surface of the epithelium (open arrow). C, Lung parenchyma labeled with anti-EC-SOD.
EC-SOD labeling is primarily at alveolar septal cips (short arrow), and in the matrix surrounding small vessels (long arrow). No labeling for EC-SOD was seen on the surface of alveolar epithelial cells (open arrow) (Bars 50 gm).
Figures 16A-16C show the electron microscopic immunolocalization of EC-SOD in vascular connec:ive tissue. Tissues were labeled using the antibody to recombinant human EC-SOD (40 gg/ml; anti-EC-SOD; or the same antibody after the anti-EC-SOD IgG was absorbed out using purified recombinant EC-SOD attached to CXBrsepharose (EC-SOD absorbed). Antibody was deteczed using 10 nm protein-A gold. A, Vascular collagen labeled with anti-EC-SOD, B, Vascular elastir labeled with anti-EC-SOD. C, Vascular collagen labeled with EC-SOD absorbed antisera. Note the intense labeling of EC-SOD in association with type I collagen and the lack of labelinc in association with elastin addition, absorption of anti-EC-SOD antibody aboclished all labeling for EC-SOD in association with type I collagen. (Bars 200 nm).
Figure 17 shows the electron microscopic immunolocalization of EC-SOD around vascular smooth muscle.
Tissues were labeled using the antibody to recombinant human EC-SOD (40 pg/ml). Antibody was detected using nm protein-A gold. There is a high degree of labeling in the connective tissue matrix around the vascular smooth muscle cell in association with type I collagen (short arrow), and other unidentified matrix elements (long arrow). (Bars 200 nm).
Figures S1A-18B show the electron miscropic immunolocalization of EC-SOD on the surface of pulmonary endothelial cells. Tissues were labeled using the antibody to recombinant human EC-SOD (40 gg/ml).
Antibody was detected using 10 nm protein-A gold. A, Endothelial cell from a small muscular pulmonary artery, B, Endothelial cell from a pulmonary capillary. No labeling for EC-SOD was on the surface of the endothelial cells (short arrows). EC-SOD is seen in the plasma and is associated with extracellular matrix proteins beneath the endothelium (long arrows).
(Bars 200 nm) 0 Figure 19 shows the electron microscopic immunolocalization of EC-SOD around bronchial epithelial cells. Tissues were labeled using the antibody to recombinant human EC-SOD (40 pg/ml) Antibody was detected using 10 nm procein-A gold. EC-SOD was found in the junction between the epithelial cells (arrow) and was also seen to some extent inside the cells.
(Bars 200 nm).
Figures 20A-20D show a partial restriction map, sequencing strategy, genomic structure, and protein structure of human EC-SOD Clone Fig. 20A, a partial restriction map of human EC-SOD genomic clone #7 is shown in the 5' to 3' orientation. A 1 kb size marker is indicated.. B, BamH I; H, Hind III; P, Pst I; S,, Sal I; K, Kpn I; E, EcoR I. In Fig. 20B, the subclonina and sequencing strategy is shown. Various size overlapping restriction fragments were subcloned into the plasmid vector pGEM3Zf(+) for subsequent DNA sequence analysis. All DNA was sequenced on both strands using Sequenase (USB) and double-stranded
DNA
template, except for -2 kb of the 3' 7K36 fragment in which only one orientation was sequenced. In Fig. the exon/intron structure of the human EC-SOD gene is shown. The position of the coding region for preEC-SOD in exon 3 is shown by the dashed lines. In Fig. the four structural domains of human EC-SOD protein are diagrammed. The signal peptide is indicated by an arrow. This is followed by the mature glycosylated (CHO) amino terminal peptide domain. A third region has very high amino acid sequence homology to human CuZn- SOD. The carboxy terminal domain has multiple charged basic residues which are critical for binding heparin glycosamincglycan.
Figures 21A-21! show human multiple tissue Northern blots of EC-SOD. Fig. 21A, two gg of poly mRNA from eight different human tissues were electronhoresed on a denaturing agarose gel, transferred to a charged nylon membrane, and probed with 32 PI-labeled antisense human EC-SOD cRNA. RNA molecular size markers (kilobases) are shcwn on the right. Quantitative transfer was monitored by ethidium bromide staining.
The results demonszrate a unique 1.4 kb mRNA present in all eight tissues examined. Interestingly skeletal muscle demonstrates a second, larger mRNA of -4.2 kb, while brain shows a faint approximately 2.2 kb band. In Fig. 21B, bands corresponding to EC-SOD mRNA were quantitated by laser densitometric scanning, normalized to the 1.4 kb brain band, and expressed as relative absorbance units.
Figures 22A-22B show analysis of the transcription initiation site. The 5' rapid amplification of cDNA ends RACE) technique was used to identify the site of transcription initiation for the human EC-SOD gene.
In Fig. 22A, a schematic diagram illustrates the annealing sites for the various oligonucleotides. The dark line represents first-strand reverse transcribed 0 cDNA of human heart poly mRNA which has been primed with EC7 (an EC-SOD gene specific primer) and pcly C tailed using terminal deoxynucleotidyl transferase (TdT). HECI, HEC2, EC4, and EC7 are human EC-SOD gene specific primers. The anchor primer is supplied with the 5' RACE kit (GIBCO BRL) and hybridized to the poly C tail. In Fig. 22B, PCR was used to amplify segments of DNA using [anchor EC4] or [EEC1 EC7] as primers and either pcly C tailed (+TdT, lanes 1 4) or non-poly C tailed (-TdT, lanes 2 5) cDNA as template.
Lane 3 includes PCR amplified DNA using [HEC1 EC7] as primers and a full-length human EC-SOD cDNA as template.
The resulting amplified DNAs were eleczrophoresed on a 2% agarose gel, transferred to charged nylon membranes, and probed with [32p]-labeled HEC2, a 5' nested gene 3 specific EC-SOD primer. DNA molecular weight markers were run between lanes 2 and 3. The expected size of the PCR amplified region in lanes 3, 4 and 5 is 217 bp.
Only a single band is seen in lane 1, with a molecular size of approximately 185 to 200 bp.
7 Figure 23 shows genomic Southern blot analysis of the human EC-SOD gene. Ten micrograms of human genomic DNA were completely digested with each of the restriction enzymes shown, electrophoresed on a 1% agarose gel and transferred to charged nylon membranes.
The blots were proved with a 3 2 p]-labeled EC-SCD partial length cRNA which corresponds to the first approximate 1050 nucleotides and autoradiographed. The specific restriction endonuclease is shown at the top of each lane. DNA molecular size markers (in kilobases) are shown on the right.
Figure 24 shows the nucleotide sequence and deduced amino acid sequence of the human EC-SOD gene. The complete nucleotide sequence of the human gene is shown.
The deduced amino acid sequence of the signal peptide and mature protein is indicated using the single letter amino acid code.
Figure 25 shows a Lineweaver-Burk plot demonstrating non-competitive inhibition of xanchine oxidase by MnTBAP.
Figure 25 shows the protection of pulmonary arcery endothelial cells from xanthine oxidase-induced injury by MnTBAP. Control MnTBAP Figure 27 shows the procection of lung epithelial cells from paraquat-induced injury of SOD mimetics.
Figure 28 shows the protection of pulmonary artery endothelial cells from paraquat-induced injury by MnTBAP.
Figure 29 shows the lack of protection of pulmonary artery endothelial cells from paraquat-induced injury by ZnTBAP.
Figure 30 shows the protection of MnTBAP against paraquat-induced lung injury.
Figure 31 shows a second order rate constant plot .0 for catalase mimetics.
Figure 32 shows the effect of MnTBAP on H 2 0 2 -induced endothelial injury.
Figures 33A and 333 show the reduction by MnTBAP and MnTmPyP of endothelial cell injuries caused by exposure to glucose oxidase-produced hydrogen peroxide.
Figure 33C shows that ZnTBAP does not reduce hydrogen peroxide-induced endothelial cell injury. Figure 33D shows that endothelial cells are not protected from hydrogen peroxide-induced injury by CuZnSOD.
0 Figures 34A and 343 show the time course of NMDAand KA-induced inactivation of aconitase. Fig. 34A.
Cortical cells treated with vehicle or 50 AM NMDA for 0, 15, 30, 60 and 240 min. and aconitase activity measured in cell lysates. Each point represents the mean S.E.M. Fig. 34B. Cortical cells treated with vehicle or 300 gM kainate for 0, 60, 240, and 360 min. and aconitase activity measured in cell lysates.
Each point represents the mean S.E.M. Figures 35A-35C show the correlation of toxicity with aconitase inactivation. Concentration-dependence of KA (Fig. 35A), PQ- (Fig. 35B) and NMDA (Fig. 35C) to induce toxicity (left axis) and aconitase inactivation (right axis) were determined. Values normalized as a percent LDH release or aconitase inhibition and plotted to assess the correlation between LDH release and aconitase inactivation. Closed squares represent LDH and open squares represent aconitase. Curves were computergenerated using non-linear regression analysis using the equation Y= Bottom (Top-Bottom)/i1+0 og so-x (GraphPad Prism). Each point represents the mean value 4-6).
Figures 36A and 36B show the blockade of aconitase inactivation and neurotoxicity by MnTBAP. Fig. 36A.
Cortical cells were treated with 150 uM PQ- for 3 hr.
(solid bars), 50 pM NMDA for 1 hr. (open bars) or 300 pM KA for 6 hr. (hatched bars) in the presence or absence of 200 jM MnTBAP (present 15 min. prior to and throughout the duration of treatment) and aconitase activity measured in cell lysates. Bars represent mean S.E.M. Asterisk indicates a difference from all other treatments (p<0.05, one-way ANOVA). Fig. 36B.
Cortical cells were treated with 150 gM PQ" (solid bars), 50 gM NMDA (open bars) or 300 gM KA (hatched bars) in the presence of varying concentrations of MnTBAP (present 15 min. prior to and throughout the duration of treatment) for 18 hr. and LDE release measured in the medium. Asterisk represents a S difference from controls (agonist in the absence of MnTBAP; p< 0.05 Dunnet's test). Bars revresen: mean S.E.M. Figure 37 shows the inhibition of NMDA toxicity by MnTBAP. Cortical cells were treated with varying concentrations of NMDA in the presence and absence of 200 jiM MnTBAP for 18 hr. and LDH measured in media. Each points represents mean n=3-4.
Figures 38A-38C show the differential effect of MnTBAP and ZnTBAP on cell death. Cortical cells were treated with 150 gMPQ" (Fig. 38A) 50 iM NMDA (Fig.
38B) or 300 pM KA (Fig- 38C) in the presence or absence of varying concentrations of MnTBAP (open squares) or ZnTBAP (closed squares) and dead cells stained with EthD-l. Images of EthD-1 -positive cells were stored and counted in randomly selected fields using a digital image analyzer. Data are expressed as the number of dead cells per field. Each point represents measurements made from 1200-1500 cells.
Figure 39 shows the effect on learning of removal of the EC SOD gene in a mouse model.
|I
Figure 40. Effect of Mn(III)cetrakis (4-benzoic acid) porphyrin (MnTBAP) (3-100 AM) on the oxidation of dihydrorhodamine 123 to rhodamine 123 in response to peroxynitrite (5 AM). Data are expressed as means s.e.m. of triplicate determinations.
Figure 41. Suppression by the peroxynitrite (Fig. 41A) and the NO donor compounds S-nitroso- N-acetyl-DL-penicillamine (SNAP, 3 mM) (Fig. 4132) and diethylamine:NO NONOate (DNO) (Fig. 41C) on mitochondrial respiration (expressed as percent of respiration of unstimulated cells) in J774 macrophages, and the protective effect of Mn(III)tetrakis (4-benzoic acid) porphyrin (MnTBAP) (10-300 AM)- against this suppression. Data are expressed as means of n=12 wells. *p<0.01 represents significant effect of SNAP when compared to control values; reresent significant protective effects cf MnTBAP (p<0.05 and p<0.01, respectively).
Figures 42A-D show synthezic reaction schemes.
DE7A r.7D :=C-n?r70NOF Th- INVCN-rON The nresent inventi-on relates to methods O: prcectinc aaans: the delet:erious effects C: oxzans particularly, superoxide radicals, hydrogen peroxi;de and peroxmnitrize, an-,d t o methods of prevering and treating aisease states that involve or result- from cxican: st ress- The invention also relates methods of modulating biological processes involving oxidant-s, including suneroxiac radi'cals, hydrogen peroxide, ni--tric oxide and eoyirt. T-he inveotion furt- r-= 2 ates to comoounds and coosincludina low molecu-lar weigh: antioxidants (eg mzeisof scavencers of reactive oxygen: sp-ecies-, including mimecics off SODs, catalases and ceroxi:das es) and form=ulations thnereo f, sLuitable for use in su.cs- metnhocs.
Mimetios c:f soavencrers or-- reactive oxygen soec ies aioorovriate oruse 'I te p~resentL methods include manaanic crvtesc: s ueczrzns o r p harm=a c~r e,:ti v a cc e z -ahl e s alt--s t h er2:eof:. Th..e metnt-ine substitue nts can- be selected so as to fcltt electron exc nance betwgenth m~mer-ic metal (ea M< ant-d the oxy.gen r az-ca' Sth bs::tuents that withdraw electrons fromn tn-e rz':na7 assist in delc#-z-'gth charge o-f the metal and thereby enhance catalyt icactivity. Acccrdi nzlv, tesubstit--uents can-7 be selectecso as to modu' ate th-e readox voterta 1 of th--eponie Substicuents can also be selecr-ec so as to render the por-phyrin reasistant to degradation by hemeoxygenase.
iemeoxyge-nase, a key enzy-me inr normal porp--hyri4n degradation and an enzyme that plays a role in the regulation of inflammation, attacks at the methine bridge carbons. By designing compounds not susceptible to attack (eg by introducing substituents at the methine bridge carbons), the half life of the porphyrin can be increased. Such compounds have the further advantage that they do not interfere with normal porphyrin metabolism. Selection of substituents can also be made based on a result sought to be achieved. For example, when passage through cell membranes is advantageous in a given treatment regimen, non-polar substituents can be selected so as to render the mimetic lipid soluble.
Substituents can also be selected so as to render the mimetic capable of binding to cell surface or extracellular matrix elements. Such substituents can be selected so as to target the mimetic on the basis of charge, shape, structure, etc. Targeting substituents can be specific, for example, for certain cell surface receptors (eg mannose receptors found on epithelial cells) or for certain sugars or lectins presen: on the cell surface.
In one embodiment, the mimetics are of the formula: R" R3- R 2 R- R3- R 2
R
R R- R R4 P P wherein: RI is a bond, Pt, No _SOH 0 x or wherein X is a halogen and Y is an alkyl group and wherein a indicates bonding to R2 at any position and Z indicates bonding to R 2 and the substituent at any position; and R2 is a bond, -(CY'2-CY'=CY')n -(CY'2 CY'2-CH=CH) or -(CY-'2C)n wherein Y' is hydrogen or an alkyl group and wherein n is 1 to 8; 0 R3 is a bond, hydrogen, -OH, 3 -COO-, -COO-, -S0 3 -S03 -C-P0 3 H- or -C-PC 3
H-,
wherein Y" is an alkyl group, and R4 is nothing, hydrogen, a cell surface or extracellular matrix targeting moiety or a linker-cell surface or extracellular matrix targeting moiety (wherein a "linker" is a moiety that links the mimetic core (porphyrin ring and R 1
R
2
R
3 to the targeting moiety).
In a more specific embodiment,
R
1 is a bond, No, SoH
S
x o r Y wherein X is Cl or Br and Y is a C 1 4 alkyl group;
R
2 is a bond,
-(CY
2 (CY 2-C =C
Y
-(CY CY'2-CH=C?)n or n wh y n 2 n rein Y' is hydrogen or a C 1 4 alkyl group and wherein n is to 4;
R
3 is a bond, hydrogen, -OH, -COO-, -COO-, -SO3-, -S03-, -C-PO3H- or wherein Y" is a C1.4 alkyl group, and
R
4 is nothing, hydrogen, a cell surface or extracellular matrix targeting moiety or a linker-cell surface or extracellular matrix targeting moiety.
In a further specific embodiment, R, is a bond,
N:,
S0 1 H x or Y wherein X is Cl or Br and Y is methyl or ethyl, and
R
2 is a bond, -(CY'2)n
-(CY'
2 -CY,=CY')n -(CY'2 CY'2-CH=CH)n-, or wherein is hydrogen or methyl or ethyl and wherein n is 1 or 2; and
R
3 is a bond, hydrogen, methyl, ethyl, -OH, -Ni-,
-N+(CH
3 3
-N+(CH
2 CH3)3, -COO-, -COO-, -SO3-, -S0 3 -C-PO3H- or -C-PO3H-, and
R
4 is nothing, hydrogen, a cell surface or extracellular matrix targeting moiety, or a linker-cell surface or extracellular matrix targeting moiety.
In another specific embodiment,
R
1 is a bond, -C
N
o 2 -so 3 H 7 s or Y wherein Y is alkyl, preferably, C 1 -4 alkyl, more preferably methyl or ethyl, O R2 is a bond, -(CY' 2 n or -(CY' 2 it wherein Y' is hydrogen or alkyl (preferably C 1 _4 alkyl, more preferably methyl or ethyl) and wherein n is 1 to 4 (preferably 1 or and
R
3 is a bond, hydrogen, C_4 alkyl (preferably methyl or ethyl), -OH, -N+(CH 3 3 (CH2CE3) 3 -COO-, -COO", -SO0 3 -S03", -C-P0 3 H- or -C-PO 3 and
R
4 is nothing, hydrogen, a cell surface or extracellular matrix targeting moiety or a linker-cell surface or extracellular matrix targeting moiety.
In yet another specific erLbodiment,
R
1 is a bond, -No, SO H or
R
2 is a bond, -(CY' 2 n or n wherein Y' is hydrogen or alkyl (preferably C.-4 alkyl, more preferably methyl or ethyl) and wherein n is 1 to 4 (preferably 1 or 2);
R
3 is a bond, hydrogen, C-,4 alkyl (preferably methyl or ethyl), -OH, -N*(CH 3 3
-N+(CH
2
CH
3 3 -COO-, -COO-, -SO 3 -S03~, -C-PO 3 E- or -C-PO 3 and
R
4 is nothing, hydrogen, a cell surface or extracellular matrix targeting moiety or a linker-cell surface or extracellular matrix targeting moiety.
In addition to the substituents described above, one or more of the pyrrole rings of the porphyrin can be substituted (at any or all of carbons 2, 4, 7, 8, 12, 13, 17 or 18) with an electron withdrawing group (designated for example, each P being, independently, a NO, group, a halogen (eg CI, Br or a nitrile, a vinyl group, or a formyl group. Such substituents alter the redox potential of the porphyrin and thus enhance its ability to scavenge oxygen radicals. Each P can, independently, also be hydrogen.
Specific mimetics suitable for use in the present methods include Mn(III) tetrakis (1-methy-4pyridyl)porphyrin (MnTMPyP), Mn(III) tetrakis (4trimethyl-aminophenyl)porphyrin (MnTMAP) and Mn(III) tetrakis (4-benzoic acid)porphyrin (MnTBAP), with or without a cell surface or extracellular matrix targeting moiety or a linker-cell surface or extracellular matrix targeting moiety a: the R, position.
Although the foregoing mimetics are described as manganese chelates, metals other than manganese, such as iron (III) and copper can also be used. The present invention also relates to the metal-free nitrogen-containing macro cyclic ligand. It will be appreciated that the metal selected may have various valence states, for example, manganese II, III or V can be used. The change in charge will be dependent upon the acceptance or release of electrons.
In addition to the foregoing mimetics, the invention also includes compounds (and targeted forms thereof as described above, including linker-cell surface or extracellular matrix targeted forms) of the formula: R R2- R 1, 20 s/ R-2- R 3 N7 N N i H p R R R'
R
1 R2 R3 P P or a pharmaceutically acceptable salt thereof, or metal complex thereof wherein the metal is, for example, manganese, copper or iron, wherein: 0 each is independently a bond, cCO2Y' V--H c 2 H C-NH-,
C-C-
o eor C(Y)3 wherein Y" is an alky! group(eg and wherein indicates bonding to R 2 at any position and _Z indicates bonding to R 2 and the R. phenyl substituent at any position; each is independently a bond, or -(CH wherein n is 1-4, each R3' is independently -OH, -OY",
-NO
2 -CN, -NH2, -COOH, -COY", -COO-, or a heterocyclic group, wherein Y" is as defined above and is a primary, secondary, tertiary or quaternary amine (preferably, an alkyl amine wherein the alkyl groups are, for example, C,-Cs alkyls) 00 wherein when is
R
3 is not COOH, COY" or COO-, 0 00 0 wherein when R is C-NH-, c-or R3 is not -NO 2 In certain embodiments of the invention, 00
R
3 collectively, are not Q-N*(Yn)3 -ccoo- or for example, when Y" is a methyl.
As indicated above, R 3 can represent a heterocyclic moiety. Possible heterocyclics include substituted or unsubstituted tetrazoles, furans, thiophenes, indoles, imadazoles, pyridines, oxadiazoles and quinolines. Possible substituents on such moieties include halogen (eg Br or Cl), -NO 2
C,.
4 alkyl, and C .4 alkyl alcohol groups.
As above, one or more of the pyrrole rings of the porphyrin can be substituted (at any or all of carbons 2, 4, 7, 8, 12, 13, 17 or 18) with an electron withdrawing group (designated for example, each P being, independently, a NO, group, a halogen (eg Cl, Br or F), a nitrile, a vinyl group, or a formyl group. P can also be hydrogen.
Where rotational isomers are possible, all such isomers of the herein described mimetics (oxidant scavengers) are within the scope of the invention.
In addition to the compounds described above, the present invention includes within its scope porphine mimetics (and targeted forms thereof) bearing any of the above substituents in any of the possible combinations.
For example R, can be selected from the group described above with respect to R. or R' R2 can be selected from the group described above with respect to R 2 and and R3 can be selected from the group described above with respect to R 3 or Targeted forms of the mimetics can be produced by coupling the porphine directly or via a linker to a cell surface or extracellular matrix targeting moiety, for example, as indicated above in the definition of R 4 The targeted mimetics can be used, for example, to mimic EC-SOD. Since the sequence of human EC-SOD is known (Hjalmarsson et al, Proc. Natl. Acad. Sci. USA 84:6340 (1987)), the C-terminal oligopeptide can be prepared and attached to the "mimetic core" (eg, a Mn(III)-porphyrin) via, for example, a free amine or carboxy group with a 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide
(EDC)
coupling reaction (Yamada et al, Biochemistry 20:4836 (1981); Davis and Preston, Anal. Biochem. 116:402.
(1981)). This simple coupling procedure makes it possible to attach a wide variety of different binding domains in order to target the mimetics of the invention. Heparin binding affinity of a mimetic can be assessed by passing the mimetic over a heparin-sepharose CL-6B column (Karlsson et al, Biochem. J. 256:29 (1988)).
Candidate targeting moieties suitable for attaching to the mimetics of the invention to confer GAG binding properties include the following: i) the A+ helix of protein C inhibitor (Boissinot et al, Biochem. Biophys. Res. Commun. 190:250 (1993)) NH2 His Arg His His Pro Arg Glu Met Lys Lys Arg Val Glu Asp Leu COOK; ii) the C-terminal end of human EC-SOD (heparin binding domain) (Karlsson et al, Biochem. J. 255:223 (1988)) NH2 Arg Glu His Ser Glu Arg Lys Lys Arg Arg Arg Glu Ser Glu Cys Lys Ala Ala COOH; iii) variants of the C-terminal end of human EC-SOD having heparin binding affinity (Sandstram et al, J. Biol. Chem. 267:18205 (1992)) a. NH2 Arg Glu His Ser Glu Arg Lys Lys Arg Arg Arg Glu COOK; b. NH2 Arg Glu His Ser Glu Arg Lys Lys Arg Arg Arg Ala COOH; c. NH 2 Arg Glu His Ser Glu Arg Lys Lys Arg Arg Arg Ala Ser Glu Cys Lys Ala Ala COOH; d. NH2 Arg Glu His Ser Glu Arg Lys Lys Arg Arg Arg Glu Ser Glu Ala Lys Ala Ala COOH; e. NH 2 Arg Glu His Ser Glu Arg Lys Lys Arg Arg. Arg Glu Ser Glu Cys Ala Ala Ala COOH; f. NH 2 Arg Glu His Ser Glu Arg Lys Lys Arg Arg Arg Ala Ser Ala Cys Lys Ala Ala COOH; g. NH 2 Arg Glu His Ser Glu Arg Lys Lys Arg Arg Arg .r Ala Ser Glu Cys Ala Ala Ala COOH; h. NH 2 Arg Glu His Ser Glu Arg Lys Lys Gly Arg Arg Ala Ser Glu Cys Ala Ala Ala COOH; iv) any combination of repeating positively changed amino acids (ie poly (Arg) n poly (Lys)n, poly (Arg)n (Lys)n or poly (Arg Lys)n or poly (Lys Arg)n, poly (Lys Lys Arg Arg)n, wherein n is, preferably, in the range of 1 to 12, more preferably, 1 to 8, and, most preferably, 1 to 6, with or without a C-terminal Glu or Ala (Sandstr6m et al, J. Biol. Chem. 267:18205 (1992)); 0 and v) polyethyleneimine, e.g. (NH-CH2-CH,-NH)nH, wherein n is 1 to 6.
In addition to the foregoing, targeting moieties also include heparin binding proteins generally and sugars, including mannose and oligosaccharides.
o c Appropriate linkers include -C-NH- or -NH-C-, -S0 2
-PO
3 or -P0 3 Specific examples of suitable mimetics oi- the invention are set forth below: R~R NtCH 3 :Rr=
COOH
R
NH
2 R=RR/R =CONHR' R /O
NHCOR'
H2 -AracGluHisSerGlurazysLysA-c-.,-a~g GIuSerGluCysLysAaa.-
COOH
S,lO,15,20-Tetra Uis CR-group] manganese (111) porphyrins
CON(CHJ,
AM
3 N\'CH3 Mn-C B-TMPyP Mn-OC-TMPyP *C H 3
CH
3 Mn-C F-TM PyP Mn-ON-TMPyP Mn-OB-TBAPMnC-fA Mn-OC-TBAP M-F-APMn-ON-TBAP Mn-ON-TBAP Mn-OB-TPSP Mn-OC-TPSP Mn-OF-TPSP Mn-OF-TPSPMn-ON -TP SP Mimetics suitable for use in the present methods can be selected by assaying for SOD, catalase and/or oeroxidase activity and stability. The selective, reversible and SOD-sensitive inactivation of aconitase by known generators can be used as a marker of 0' generation. Thus, suitable mimetics can be selected by assaying for the ability to protect aconitase activity.
SOD activity can be monitored in the presence and absence of EDTA using the method of McCord and Fridovich L Biol. Chem. 244:6049 (1969)). The efficacy of a mimetic can be determined by measuring the effect of the mimetic on the growth of a SOD null E. coli strain versus a wild type strain. Specifically, wild type E.
coli (AB1157) and SOD null E. coli. (JI132) can be grown in M9 medium containing 0.2% casamino acids and 0.2% glucose at pH 7.0 and 37 0 C; growch can be monitored in terms of turbidity followed at 700 nm spectrophotometrically. Active mimetics can be tested for toxicity in mammalian cell culture by measuring 0 lactate dehydrogenase (LDH) release. Specifically, rat L2 cells (a lung Type II like cell; (Kaighn and Douglas, J. Cell Biol. 59:160a (1973))can be grown in Ham's F-12 medium with 10% fetal calf serum supplement at pH 7.4 and 37 0 C; cells can be seeded at equal densities in 24 well culture dishes and grown to approximately confluence; SOD mimetics can be added to the cells at log doses (eg micromolar doses in minimal essenzial medium (MEM)) and incubated for 24 hours. Toxicity can be assessed by morphology and by measuring the release 0 of the cytosolic injury marker, LDH (eq on a thermokinetic plate reader), as described by Vassault (In: Methods of Enzymatic Analysis, Bergmeyer (ed) PP. 118-26 (1983); oxidation of NADH is measured at 340 nm). Efficacy of active mimerics can be assessed by determining their ability to protect mammalian cells against methylviologen (paraquat)-induced toxicity.
Specifically, rat L2 cells grown as described above and seeded into 24 well dishes can be pre-incubated with various concentrations of the SOD mimetic and then incubated with a concentration of methylviologen previously shown to produce an LC 75 in control L2 cells.
Efficacy of the mimetic can be correlated with a decrease in the methylviologen-induced LDH release (St. Clair et al, FEBS Lett. 293:199 (1991)). The efficacy of SOD mimetics can be tested in vivo with mouse and/or rat models using both aerosol administration and parenteral injection. For example, male Balb/c mice can be randomized into 4 groups of 8 mice each to form a standard 2X2 contingency statistical model. Animals can be treated with either paraquat mg/kg, ip) or saline and treated with SOD mimetic or vehicle control. Lung injury can be assessed 48 hours after paraquat treatment by analysis of bronchoalveolar lavage fluid (BALF) damage parameters (LDH, protein and 0 PMN) as previously described (Hampson et Tox.
Appl. Pharm. 98:206 (1989); Day et al, J. Pharm. Methods 24:1 (1990)). Lungs from 2 mice of each group can be instillation-fixed with 4% paraformaldehyde and processed for hiscopachology at the light microscopic 0 level.
Catalase activity can be monitored by measuring absorbance at 240nm in the presence of hydrogen peroxide (see Beers and Sizer, J. Biol. Chem. 195:133 (1952)) or by measuring oxygen evolution with a Clark oxygen electrode (Del Rio et al, Anal. Biochem. 80:409 -(1977)).
Peroxidase activity can be measured spectrophotometrically as previously described by Putter and Becker: Peroxidases. In: Methods of Enzymatic Analysis, H.U. Bergmeyer Verlag Chemie, Weinheim, pp. 286-292 (1983. Aconitase activity can be measured as described in Example XI below.
Table IX below provides a summary of the activities of various oxidant scavengers of the invention. The footnote to that Table provides details of the assays used.
Synthesis of mimetics suitable for use in the present method can be effected using art-recognized protocols. Example XIV includes a detailed description of the synthesis of four specific mimetics. In the case of Mn(III)-porphyrin mimetics, porphyrin rings with various methine bridge carbon side groups can be purchased commercially and the Mn(III) metal ion inserted into the porphyrin ring by methods including the following: admixture of Mn(II) acetate with porphyrin in the presence of oxygen, under which condition selective stabilization of Mn(III) by the porphyrin causes autooxidation of Mn(II); preparation of Mn(III) (OH) 3 by a modification of the Winkler method (Sastry et al, Anal. Chem. 41:857 (1969)) followed by reaction with the porphyrin; stirring Mn0 2 with the porphyrin in the presence of NH 2 0H, which serves to reduce the Mn(IV) to Mn(III), which is then trapped by the porphyrin; or a method modified from Pasternack et al (Biochemistry 22:2406 (1983)) which refluxes excess MnC1 3 with the porphyrin. Mn(III)porphyrin complexes can be precipitated from solution with sodium perchlorate, washed and residue perchlorate removed by strong anionic exchange resin. Formation of the Mn(III)-porphyrin can be followed spectrophotometrically by monitoring a characteristic Soret band at 468 nm. Synthesis of compounds of the invention bearing electron-withdrawing groups at one or more pyrrole carbons can be carried out as described by Richards et al, Inorg. Chem. 35:1940 (1996).
Purification of the mimetics can be effected using artrecognized techniques such as recrystallization, chromatography, etc. Coupling of a binding domain to the "mimetic core" can be carried out as described above.
One embodiment of the present invention results, at least in part, from the realization that
EC-SOD
specifically regulates NO- function. In addition, the invention is based on the realization that EC-SOD is synthesized by epithelial cells and is primarily localized in the interstitium, on matrix elements and collagen and around smooth muscle cells (particularly lung airways and vasculature). NO- is an intercellular signal and, as such, NO- must traverse the extracellular matrix to exert its effects. NO., however, is highly sensitive to inactivation mediated by present in the extracellular spaces. EC-SOD is thus an enzyme ideally suited to increase the bioavailability of NO- by preventing its degradation by 02 One embodiment of the present invention relates to a method of regulating extracellular NO- levels using polypeptides having EC-SOD activity. As indicated above, the invention also relates to mimetics of EC-SOD that can be targeted to strategic locations and to the use of such mimetics in manipulating extracellular tI levels of NO'. The invention, however, is not limited to NO- manipulation as the sole mechanism of action of the compounds, mimetics, etc, of the invention. Rather, the invention relates to oxygen radical, hydrogen peroxide and peroxynitrite scavenging generally.
The present invention relates, in a further specific embodiment, to a method of inhibiting production of superoxide radicals. In this embodiment, the mimetics of the invention are used to inhibit oxidases, such as xanthine oxidase, that are responsible for production of superoxide radicals (see Example VII).
The ability of a mimetic to protect mammalian cells from xanthine/xanthine oxidase-induced injury can be assessed, for example, by growing rat L2'cells in 24well dishes. Cells can be pre-incubated with various concentrations of a mimetic and then xanthine oxidase (XO) can be added to the culture along with xanthine The appropriate amount of XO/X used in the study can be pre-determined for each cell line by performing a dose-response curve for injury. X/XO can be used in an amount that produces approximately an LC 75 in the culture. Efficacy of the mimetic can be correlated with a decrease in XO/X-induced LDH release. The ability of the mimetics to inhibit the production of such radicals makes possible the use the mimetics as therapeutics for the treatment of gout and reperfusion injuries.
The mimetics of the invention can be used as catalytic scavengers of reactive oxygen species to protect against ischemia reperfusion injuries associated with myocardial infarction, stroke, acute head trauma, organ reperfusion following transplantation, bowel ischemia, pulmonary infarction, surgical occlusion of blood flow, and soft tissue injury. The mimetics can further be used to protect against skeletal muscle reperfusion injuries. The mimetics can also be used to protect against damage to the eye due to sunlight (and to the skin) as well as glaucoma, and macular degeneration of the eye. Diseases of the bone are also amenable to treatment with the mimetics. Further, connective tissue disorders associated with defects in collagen synthesis or degradation can be expected to be susceptible to treatment with the present mimemics.
In addition to.scavenging superoxide, the ability of the mimetics of the invention to scavenge hydrogen peroxide would protect from the possible formation of the highly reactive hydroxyl radical by interfering with Fenton chemistry (Aruoma and Halliwell, Biochem. J.
241:273 (1987); Mello Filho et al, Biochem. J. 218:273 (1984); Rush and Bielski, J. Phys. Chem. 89:5062 (1985).
These metalloporphyrins have been shown to scavenge peroxynitrite as demonstrated indirectly by inhibition of the oxidation of dihydrorhodamine 123 to rhodamine 123 (see Example XIII) and directly by accelerating peroxynitrite degradation by stop flow analysis.
In addition to the above, the mimetics of the invention can be used as catalytic scavengers of reactive oxygen species to increase the very limited storage viability of transplanted hearts, kidneys, skin and other organs and tissues. The invention also provides methods of inhibiting damage due to autoxidation of substances resulting in the formation of 0" including food products, pharmaceuticals, stored blood, etc. To effect this end, the mimetics of the invention are added to food products, pharmaceuticals, stored blood and the like, in -an amount sufficient to inhibit or prevent oxidation damage and thereby to inhibit or prevent the degradation associated with the autoxidation reactions. (For other uses of the mimetics of the invention, see USP 5,227,405). The amount of mimetic to be used in a particular treatment or to be associated with a particular substance can be determined by one skilled in the art.
The availability of the mimetics of the invention also makes possible studies of processes mediated by O2-, hydrogen peroxide, nitric oxide and peroxynitrite.
In addition to the above, the present invention relates to diagnostic protocols made possible by the availability of the EC-SOD gene sequence (see Example V and Figure 24). A defect in the EC-SOD gene is more likely to occur than a defect in nitric oxide synthase due to the nature and number of physiological functions served by NO-. Detection of an EC-SOD gene defect would signal the need for measures to be undertaken to elevate levels of functional EC-SOD, or related superoxide scavenging compounds, at strategic locations to correct NO- imbalances and thus disorders involving oxidazive stress.
To effect modulation of the efficacy of extracellular NO-, eg, in relaxing smooth muscle, molecules (agents) having EC-SOD activity are administered under conditions such that levels of extracellular 0- are altered.
Molecules suitable for use in this method include forms of SOD that bind heparin sulfate or other glycosaminoglycans (GAG), for example, by virtue of the fact that they contain positively charged amino acids near their carboxy terminal end. Proteinaceous agents suitable for use in the present method include forms of EC-SOD C described in WO 91/04315, as well as additional polypeptides defined and described therein as havina comparable or enhanced binding to heparin as compared to recombinant EC-SOD C (eg, polypeptides G! and SA216; note also polypeptide SA219 which has the same heparin binding as recombinant EC-SOD C. Further proteinaceous agents suitable for use in the present method include chimeric proteins with targeted binding and SOD activity, for example, Cu/Zn SOD linked to an EC-SOD binding sequence (see also Boissinot et al, Biochem.
Biophys. Res. Commun. 190:250 (1993)).
Proteinaceous molecules suitable for use in the present method can be synthesized chemically or 0 recombinantly using art-recognized protocols (see WO 91/04315). Non-glycosylated recombinant peptides can be produced, for example, using host cells (ie, E. coli cells) that are incapable of effecting glycosylation, or using DNA sequences encoding functional proteins lacking glycosylation sites.
In addition to polypeptides, molecules suitable for use in the present method include mimetics of EC-SOD (eg targeted mimetics), including those described above.
The general requirements of such mimetics are that they: be stable enough to retain the ligated metal (eg Cu or Mn) in the presence of the multiple chelating agents present in living systems, be active enough that reasonable doses can serve to significantly augment the total SOD activity in the extracellular spaces, be able to adhere to the surfaces of cells or extracellular matrix elements (eg collagen) when protection against extracellular sources of 02- is needed, and d) be of low toxicity. Examples of suitable mimetics include nitrocen-containing macrocyclic ligands effective as catalysts for dismutating superoxide, including Mn(III) complexes of porphyrins with bulky cationic substituents on the methine bridge carbons, such as those described above (eg MnTMAP and MnTMPyP) Such complexes are very active and are stable enough to retain full activity in the presence of excess EDTA or in the presence of tissue extracts.
The polypeptides and mimetics described above can be formulated into pharmaceutical compositions suitable for use in the present methods. Such compositions include the active agent (polypeptide or mimetic)
MMMM
together with a pharmaceutically acceptable carrier, excipient or diluent. The composition can be present in dosage unit form for example, tablets, capsules or suppositories. The composition can also be in the form of a sterile solution suitable for injection or nebulization. Compositions can also be in a form suitable for opthalmic use. The invention also includes compositions formulated for topical administration, such compositions taking the form, for example, of a lotion, cream, gel or ointment. The concentration of aczive agent to be included in the composition can be selected based on the nature of the agent, the dosage regimen and the result sought.
The dosage of the composition of the invention to be administered can be determined without undue experimentation and will be dependent upon various factors including the nature of the active agent, the route of administration, the patient, and the result sought to be achieved. A suitable dosage of protein administered IV can be expected to be in the range of about 10-1000 mg/day. For topical treatment, it is expected that lower doses will be recuired (see WO 91/04315); for aerosol administration, it is expected that doses will be in the range of 1 to 10 mg/kg.
Suitable doses of mimetics will vary, for example, with the mimetic and with the result sought. The results of Faulkner et al Biol. Chem. 269:23471 (1994)) indicate that the in vivo oxidoreductase activity of the mimetics is such that a pharmaceutically effective dose 0 will be low enough to avoid problems of toxicity. Doses that can be used include those in the range of 1 to mg/kg.
In addition to compositions of the types described above, the present invention also includes compositions suitable for use in gene therapy types of protocols.
For example, the invention includes DNA sequences encoding proteins having EC-SOD activity and formulated so as to be incorporated into cells (eg, lung cells) upon contact therewith (eg, via inhalation). The 1' sequence can be present in a vector, eg, a viral vector, and/or the sequence can be present in a delivery vehicle, such as a liposome. The amounts of such compositions to be administered can be readily determined by one skilled in the art.
Further examples of diseases or disorders appropriate for treatment using the compounds and compositions of the present invention include diseases of the central nervous system (including AIDS dementia, stroke, amyotrophic lateral sclerosis (ALS), Parkinson's disease and Huntington's disease) and diseases of the musculature (including diaphramic diseases (eg respiratory fatigue in emphysema, bronchitis and cystic fibrosis), cardiac fatigue of congestive heart failure, muscle weakness syndromes associated with myopathies, ALS and multiple sclerosis). Many neurologic disorders (including stroke, Huntington's disease, Parkinson's disease, ALS, Alzheimer's and AIDS dementia) are associated with an over stimulation of the major subtype of glutamate receptor, the NMDA (or N-methyl-Daspartate) subtype. On stimulation of the NMDA
I
receptor, excessive neuronal calcium concentrations contribute to a series of membrane and cytoplasmic events leading to production of oxygen free radicals and nitric oxide Interactions between oxygen free radicals and NO' have been shown to contribute to neuronal cell death. Well-established neuronal cortical culture models of NMDA-toxicity have been developed and used as the basis for drug development. In these same systems, the mimetics of the invention inhibit
NMDA
induced injury. The results presented in Example
XI
demonstrate that the formation of radicals is an obligate step in the intracellular events culminating in excitotoxic death of cortical neurons and further demonstrate that the mimetics of the invention can be used to scavenge radicals and thereby serve as protectants against excitotoxic injury. Compound 10303 (see Table IX) decreases NMDA- and kainate-induced excitctoxicity in rat cortical cells in a dose dependent manner with 100% protection against NMDA-induced 0 excitotoxicity (50 M) being achieved at 25 AM compound 10303, 100 AM compound 10303 in the case of kainateinduced excitotoxicity (300 gM).
The present invention also relates to methods of treating arthritis, systemic hypertension, atherosclerosis, edema, septic shock, pulmonary hypertension, including primary pulmonary hypertension, impotence, infertility, endometriosis, premature uterine contractions, microbial infections, gout and in the treatment of Type II diabetes mellitus. The scavengers of the invention can be used to ameliorate the toxic effects associated with endotoxin, for example, by preserving vascular tone and preventing multi-organ system damage.
The invention further relates to methods of treating memory disorders. While not wishing to be bound by theory, it is believed that nitric oxide is a neurotransmitter involved in long-term memory potentiation. Using an EC-SOD knocked-out mouse model, it can be shown that learning impairment correlates with pI reduced superoxide scavenging in extracellular spaces of the brain (see Example XII). Reduced scavenging results in higher extracellular O- levels. is believed to react with nitric oxide thereby preventing or inhibiting nitric oxide-medicated neurotransmission and thus longterm memory potentiation. The mimetics of the invention can be used to treat dementias and memory/learning disorders.
Therapeutic regimens, including mode of administration, appropriate for effecting treatment of 0 the conditions described above can be readily determined by one skilled in the art.
Inflammations, particularly inflammations of the lung, are amenable to treatment using the present invention (note particularly the inflammatory based disorders of asthma, ARDS including oxygen toxicity, pneumonia (especially AIDS-related pneumonia), cystic fibrosis, chronic sinusitis and autoimmune diseases (such as rheumatoid arthritis)). EC-SOD is localized in the interstitial spaces surrounding airways and 3 vasculature smooth muscle cells. EC-SOD and mediate the antiinflammatory proinflammatory balance in the alveolar septum. NO- released by alveolar septal cells acts to suppress inflammation unless it reacts with O.to form ONOO-. By scavenging 02-, EC-SOD tips the balance in the alveolar septum against inflammation.
Significant amounts of ONOO- will form only when EC-SOD is deficient or when there is greatly increased 0,release. EC-SOD mimetics, such as those described herein, can be used to protect against destruction caused by hyperoxia. Appropriate therapeutic regimens can be readily established by one skilled in the art.
As indicated above, it is expected that defects in the EC-SOD gene, that are manifest in the protein itself, may in fact be the cause of pathologic problems related to NO- function, rather than defects in nitric oxide synthase. Thus, in a further embodiment, the present invention relaces to diagnostic protocols suitable for use in identifying EC-SOD gene defects.
This aspect of the invention is based on the availability of the EC-SOD gene sequence. The present invention includes within its scope the gene sequence presented in Figure 24 as well as portions of non-coding regions of that sequence of at least 15 bases, preferably, at least 50 bases, more preferably, at least 100 bases and most preferably, at least 500 bases. Such portions, and the complements thereof, which are also within the scope of the invention, can be used as probes or primers in protocols including those described in Example
VI.
Screening of subjects for defects in the EC-SOD gene can be done using the approach used to identify mutations on the S-adrenergic receptor gene (Reihaus et al, Am. J. Respir. Cell. Mol. Biol. 8:334 (1993)). That approach is similar to the one used by Rosen et al- (Nature 262:59 (1993)) (see Example VI).
The following are predicted sites of important gene mutations: Positions 1-558: This represents a 5' flanking to region which contains transcriptional regulatory elements. Mutations here can be expected to lead to deficient levels of EC-SOD or defective enhancement or reduction in EC-SOD levels under conditions which reqcuire manipulating the EC-SOD concentration. The following regions have been identified as putative regulatory regions. Mutations in these regions can be expected to result in deficient levels of EC-SOD: 89 95 metal regulatory response element 121 126 cyclic AMP responsive element 0 370 375 glucocorticoid response element 238 244 skeletal muscle trans-activating factor response element 251 256 cis responsive element in the induction of the c-fos proto-oncogene 162 168 TPA reponsive element 171 179 SV40 enhancer region Positions 560-570: Mutations here can be expected to lead to an inability co splice out intron 1. This would result in no EC-SOD production (or much reduced) due to initiation of translation at multiple cryptic ATG sites located in intron 1 which are upstream of the EC-SOD ATG start codon. For example, base pair 653, 656, 720, 743, 748, etc, would potentially initiate., translation.
Positions 564-1135: Intron 1 contains DNA sequence unique to EC-SOD. In addition, there are poten:ial transcription regulatory regions within this DNA stretch which are listed below; mutations in intron 1 would lead to deficient levels of EC-SOD or defective enhancement or reduction in EC-SOD levels under conditions that require manipulating the EC-SOD concentration: 1085 1095 Xenobiotic responsive region 650 661 Antioxidant responsive element Positions 71-95: Mutations here can be expected to lead to an inability to splice out intron 1. This would result in no EC-SOD production (or much reduced) due to initiation of translation at multiple cryptic ATG sites located in intron 1 that are upstream of the EC-SOD ATG start codon. For example, base pair 653, 656, 720, 743, 748, etc, would potentially initiate translation.
Positions 1211-1230: Mutations here can be expected to lead to an inability to splice out intron 2.
This would result in no EC-SOD production (or much reduced) due to initiation of translation at multiple cryptic ATG sites located in intron 2 that are upstream of the EC-SOD ATG start codon. Examples of upstream ATG translation start sites can be found at 1339 and 1518, Positions 0SS5-5080: Mutations here would lead to an inability to splice out intron 2. This would result in no EC-SOD production (or much reduced) due to initiation of translation at multiple cryptic ATG sites located in intron 2 which are upstream of the EC-SOD ATG start codon. Examples of upstream ATG translation start sites can be found at 1339 and 1518.
L' Positions 5085-5138: Mutations here would (1) interfere with efficiency of translation of EC-SOD resulting in deficient levels of the enzyme (2) interfere with targeting of EC-SOD to the endoplasmic reticulum which is required for secretion of EC-SOD, (3) interfere with co-translational signal peptide processing (ie, removal of the signal peptide) that may lead to deficient levels due to inability to proteolytically cleave the signal peptide from the mature protein which in turn would result in the protein .0 being trapped in the endoplasmic reticulum, (4) interfere with post-translational processing (specifically glycosylation) which may result in defective levels due to the synthesis of poorly soluble protein.
Positions 5139-5150: Mutations here may interfere with the signal peptidase cleavage site resulting in a mutant EC-SOD which would contain an altered amino terminus leading to defective EC-SOD function of deficient levels.
Positions 5403-5405: Mutations here can be expected to result in loss of glycosylation which may result in defective levels due to the synthesis of poorly soluble protein.
Positions 5424-5720: Mutations here can be expected to result in defective EC-SOD activity. This region is critical for binding to the substrate and catalyzing the dismutation of superoxide anion radical.
In addition, this region would also affect any other activities of this enzyme including the reduction of other species such as nitric oxide, etc.
Positions 5721-5804: Mutations in this region would cause defects in binding of EC-SOD to target tissues such as type I collagen in the extracellular matrix, and around smooth muscle cells in both vessels and bronchi. Such mutations here are highly likely to cause disease.
Positions 6385-6395: Mutations here can be expected to result in defective polyadenylation that can lead to deficient levels of EC-SOD due to a decreased half-life of EC-SOD mRNA species.
The present invention relates not only to the entire EC-SOD gene sequence, but to portions thereof for use, for example, as probes or primers in detecting mutations in regions of the gene including those noted above. Such portions can be expected to be, for example, 18-1500 bases in length, preferably 18-22 bases in length, 50-75 bases in length, 100-400 bases in length or 500 to 1500 bases in length.
Certain details of the present invention are described in greater detail in the non-limiting Examples that follow.
Example I Preparation and Characterization of Transgenic Mice i) Construction of Transgenic Mice Construction of the human EC-SOD expression vector: The EC-SOD expression vector (Figure was constructed as follows: The entire human EC-SOD cDNA fragment (Hjalmarrson et Proc. Natl. Acad. Sci. USA 84:6340 (1987); Hendrickson et al, Genomics 8:736 (1990)) flanked by EcoRI restriction sites was converted with mung bean nuclease to form blunt-ends, ligated to Sall linkers, digested with Sall, and then inserted into the Sall site of the human S-actin expression vector pHEAPr-1. The EcoRI-HindilI fragment of the resultant plasmid containing the human B-actin promoter (provided by Dr. Larry Kedes of the University of Southern California, Los Angeles, California), intron, and EC-SOD cDNA was isolated- In addition, the Hpal site of at position 2666 in plasmid pMSG (Pharmacia LKB Biotechnology, Piscataway, New Jersey) was converted to a HindIII site by linker ligation and the HindIII-PstI fragment containing the polyadenylation site of the early region was isolated. These two DNA fragments were then ligated to an EcoRI plus PstI digested pKS vector (Stratogene, La Jolla, California). The EcoR!-XbaI fragment containing the entire expression construct free of plasmid sequences was isolated and used to establish transgenic mice. All of the recombinant DNA procedures were done according to established methods (Sambrook et al, Molecular Cloning: A Laboratory Manual 3, Cold Spring Harbor; Cold Spring Harbor Laboratory, 1989).
Deveoment of transenic rnice: Purified DNA at pg/ml in 5 mM Tris-HC!, pH 7.4, 0.1 mM EDTA was injected into the pronuclei of fertilized eggs isolated from mice ((C57BL/6 X C3H)F1 X (C57BL/6
X
C3H)FI) ((C57BL/6 X C3H) Fa mice were purchased from Charles River). Mouse eggs surviving microinjection were then implanted into the oviducts of pseudopregnant foster mothers
(CD
1 (CDI mice were purchased from Charles River) following procedures described by Hogan et al (Hogan et al, Manipulating the Mouse Embryo, Cold Spring Harbor; Cold Spring Harbor Laboratory 1986, 32).
Mice carrying the transgene were identified by Southern blot analysis of tail DNA probed with the entire human EC-SOD cDNA. Transgenic founders were found the first litter screened. These mice were bred with (C57BL/G
X
C3H)FI to produce offspring for further studies. (In all of the following experiments with the
EC-SOD
transgenic mice, the nontransgenic mice refer to littermates of the transgenic mice that did not contain the transgene for the human EC-SOD. In experiments in which EC-SOD transgenic mice were not used, wild type (C57BL/6 X C3H)FI were used.) Production of homozyaous EC-SOD_ transcrenic mice: Homozygous transgenic mice were produced by an F! cross of heterozygous transgenic mice. Tail DNA from F 2 mice were isolated and treated with RNAase. 10 pg of DNA from each mouse was cut with PstI and then electrophoresed through a 1.2% agarose gel. Southern blot analysis of tail DNA probed with the entire human 0 EC-SOD cDNA was then done. The human EC-SOD cDNA did not cross-react with the mouse EC-SOD gene. Band intensity was compared visually to determine which mice were homozygous, heterozygous, or negative for the human EC-SOD transgene.
ii) Characterization of Transgenic Mice Northern analysis: Transgenic mice and nontransgenic littermates were killed with an overdose of pentobarbital. Tissues were quickly excised and frozen in liauid nitrogen until ready for further processing. Total RNA was then isolated by the CsCI procedure was described (Sambrook et al, Molecular Cloning: A Laboratory Manual. 3. Cold Spring Harbor, Cold Spring Harbor Laboratory, 1989). Twenty yg of total RNA from the tissues of transgenic mice and nontransgenic littermates and an RNA ladder were then denatured with glyoxal, electrophoresed through a 1.2% agarose gel and blotted to nitrocellulose as described (Sambrook et al, Molecular Cloning: A Laboratory Manual. 3. Cold Spring Harbor, Cold Spring Harbor Laboratory, 1989). The blots were then probed with the entire human EC-SOD cDNA.
s Seasration of SOD isoenzvmes by concanavalin q senharos chrcmatograhy: Tissues taken from 3 mice were weighed, then combined and homogenized in volumes of ice-cold 50 mM potassium phosphate, pH 7.4, with 0.3 M KBr, 3 mM diethylenetriaminepentaacetic .0 acid, and 0.5 mM phenylmethylsulfonyl fluoride.
Separation of EC-SOD from CuZn SOD and Mr SOD was accomplished by passing tissue homogenates over a concanavalin A sepharose column as described (Marklund et Clin. Chim. Acta 126:4 (1982)).
SOD actirviv: EC-SOD activity and total SOD activity (CuZn SOD and Mn SOD) remaininc after EC-SOD extraction were measured by inhibition of cytochrome
C
reduction at pH 10, as previously described (Crapo et al, Methods Enzymol. 53:382 (1978)). Total protein was determined by the BCA protein assay (Pierce, Rockford, IL). The SOD activities were then expressed as units/mg total protein.
i) EC-SOD Transgenic Mice Characterization of transaenic mice: Mice carrying the human EC-SOD transgene were detected by Southern blot analysis. Northern analysis of various tissues from the Fl of one mouse found to carry the transgene is shown in Figure 2. High levels of message for human EC-SOD were detected in the heart, skeletal muscle, and brain of transgenic mice, with little or no message 3 observed in the lung, liver, and spleen. No message was detectable in nontransaenic littermates.
Homozygous mice were generated by breeding two heterozygous Fl mice. Homozygous mice were detected by differential band intensities found using Southern blot analysis of equal amounts of PstI digested DNA from the offspring. EC-SOD activity in the mice was found to increase in response to the total copies of the EC-SOD transgene (Table I).
TABLE I EC-SOD activity in tissues of nontransgenic, heterozygous transgenic, and homozygous transgenic mice.
Tissues from 3 mice were combined for each measurement.
Activity is expressed as Units/g tissue wet weight.
Tissue Nontranscenic Heterozyaous Homozvyous Brain 18 38 Heart 35 69 102 Example II Central Nervous System Oxygen Toxicity Protocols: Oxven exosures: 7-8 week old mice were exposed to hyperbaric oxygen five at a time in a small-animal chamber (Bethlehem, Pennsylvania). After flushing the chamber with pure oxygen, compression to 50 meters (6 ATA) was performed within 5 minutes. The oxygen concentration in the chamber was monitored continuously with a Servomex oxygen analyzer (model 572, Sybron, Norwood, Massachusetts) and maintained at 99%. The carbon dioxide concentration was analyzed from intermittent samples of chamber gas with an IR detector (IR Industries, Santa Barbara, California) and was not allowed to rise above The chamber temperature was maintained a: 25-26°C, but the compression of oxygen in the chamber raised the temperature to 30-320C transiently, but an environmental control system restored the normal chamber temperature within 3 minutes. The exposures lasted 25 to 75 minutes and were followed by decompression for 5 minutes. The mice were observed continuously for signs of oxygen toxicity from the beginning of the exposure until 4 hours after removal from the chamber. The time to the first generalized convulsion (seizure latency) and the time to death were recorded. These exposure conditions are designed to cause CNS oxygen toxicity without appreciable evidence of pulmonary oxygen toxicity.
Treatment with die chvdi thiocarbamaee: One hour prior to exposure to 6 ATA oxygen, mice were given either i.p. injections of either 0.008 cc/g saline or 400 mg/kg diethyldithiocarbamate dissolved in normal saline (0.008 cc/g). The mice were then exposed to 6 ATA oxygen for 25 mintues as described above.
To determine the extent of EC-SOD and CuZn SOD inhibition by diethyldithiocarbamate, mice were given diethyldithiocarbamate and sacrificed one hour later.
The brains were removed and assayed for EC-SOD and CuZn SOD activity as described above.
Treatment with S-mercaotoethanol: One hour prior to exposure to 6 ATA oxygen, mice were given either i.p.
injections of 0.008 cc/g saline or 180 mg/kg Zmercaptoethanol (0.008 cc/g). This dose of Smercaptoethanol was selected because it contains an equal number of reducing thiols as the dose of diethyldithiocarbamate. The mice were then exposed to 6 ATA oxygen for 30 minutes as described above.
Treatment with N-a-nitro-L-arainine. an inhibitor of nitric oxide syvchasge: Ten minutes prior to beginning compression, 0.008 cc/g saline or 20 mg/kg (0.008 cc/g) N-w-nitro-L-arginine dissolved in szerile water was given i.p to the transgenic and nontransgenic mice. Mice were then exposed at 6 ATA oxygen for 25 or minutes as described above.
Statistical anavlys: A paired Student's t-zest was used to compare enzyme activities in transgenic and nontransgenic mice. A X 2 test with Bonferroni correction was used to assess significance in survival differences to hyperbaric exposures. Analysis of variance with a Scheffe F-test was used to comoare differences in seizure latency in the different crouos of mice.
Hvperbaric oxvaen exposures: To test the effects of increased brain EC-SOD levels on CNS oxygen toxicity, both transgenic and nontransgenic mice (see Examnle I) were exposed to 6 ATA oxygen for 25 minutes. Transgenic mice were more susceptible (83% mortality) to CNS oxygen toxicity than nontransgenic mice (33% mortality) (Figure 3).
Transgenic and nontransgenic mice were subsequently treated with an inhibitor of CuZn SOD, diethyldithiocarbamate, to confirm that the increased sensitivity of transgenic mice to CNS oxygen toxicity was the result of increased SOD activity. In both transgenic and nontransgenic mice, treatment with 400 mg/kg diethyldithiocarbamate resulted in inhibition of EC-SOD and 60% inhibition of CuZn SOD in the brain. This is consistent with previous findings (Frank et al, Biochem. Pharmacol. 27:251 (1978); Heikkila et al, J. Biol. Chem. 251:2182 (1976)).
Treatment with diethyldithiocarbamate conferred increased resistance to CNS oxygen toxicity for both transgenic and nontransgenic mice. Survival increased to 100% in transgenic mice and 93% in nontransgenic mice (Figure The onset of seizures was also delayed four-fold in mice treated with diethyldithiocarbamaze (Figure 4).
To evaluate whether or not diethyldithiocarbamate protects against CNS oxygen toxicity by acting as a reducing agent rather than as an inhibitor of SOD activity, mice were treated with an equimolar amounr of reducing thiols in the form of S-mercaptoethanol and exposed to hyperbaric oxygen. Figure 5 shows that Smercapcoethanol did not protect against CNS oxygen toxicity.
One possibility that might explain why EC-SOD exacerbates CNS oxygen toxicity would be that nitric
MMWW
oxide is a mediator of CNS oxygen toxicity and EC-SOD is protecting nitric oxide from superoxide mediated inactivation. To test the hypothesis that nitric oxide contributes to CNS oxygen toxicity, wild-type (C57BL/6 X C3H)F1 mice were treated with an inhibitor of nitric oxide synthase, N-m-nitro-L-arginine. Figure 6 shows the effects of N-c-nitro-L-arginine on seizure latency in mice. Pretreatment with N-u-nitro-L-arginine resulted in a significant increase in seizure latency 0 (13.50+5.6 min) when compared to saline treated mice (2.75±1 min). Figure 7 shows that nitric oxide synthase inhibition also significantly increased survival after exposure to hyperbaric oxygen. Mice given the inhibitor of nitric oxide synthase displayed 50% mortality after exposure to 90 minutes of 6 ATA oxygen and 100% mortality was not obtained until after 2 hours of this exposure. Saline treated mice, however, had a mortality after only 25 minutes of exposure, with 100% mortality after only 30 minutes at 6 ATA of oxygen.
Figure 8 shows that the percent survival in hyperbaric oxygen was dependent on the dose of the inhibitor given.
The protection offered by this competitive inhibitor of nitric oxide synthase could be reversed when an excess of L-arginine was given (Figure 6 and Figure 9).
The effects of the nitric oxide synthase inhibitor, N-a-nitro-L-arginine, upon CNS oxygen toxicity was then studied in the transgenic mice. This treatment dramatically reduced CNS oxygen toxicity in both transgenic and nontransgenic mice. Survival after a minute exposure to 6 ATA oxygen increased to 100% in both groups (Figure Seizure latency was also significantly delayed (Figure The exposure time was then increased to 75 minutes to investigate whether transgenic mice were still more sensitive than nontransgenic mice to hyperbaric oxygen. The results in Figure 10 indicate that treatment with N-c-nitro-Larginine abolished the difference in sensitivity that was observed between untreated transgenic and nontransgenic mice during the 25 minute exposure shown in Figure 3.
Example III Cold-Induced Brain Edema Protocols: Injury model: Young (6-7 week) mice (see Example I) were anesthetized with 60 mg/kg pentobarbital (Nembutal, Abbott Laboratories, Chicago, Illinois). An incision was then made in the scalp and a steel rod cm long, 3 mm in diameter, equilibrated in liquid nitrogen with an 8 cm bath of liquid nitrogen 4 cm from the end of the rod, was placed on the skull over the right cerebral hemisphere for 30 seconds. The skin incision was then sutured.
Two hours after the injury the mouse was given an additional dose of pentobarbital. The chest cavity was opened, the lungs were excised, and the mouse was then perfused with 20 ml saline through the left ventricle of the heart. The brain was then removed and the cerebellum was excised. The right and left (L) cerebral hemispheres were separated and immediately weighed (wet weight, Each hemisphere was then dried at 70 0 C for 2-3 days in a hot air oven until a constant weight was achieved (dry weight, An index of edema was then calculated as shown in equation 13.
I=(W/D R W/D L) X 100 [13] This calculation allowed the left hemisphere to serve as an internal control for the injured right hemisphere in each mouse.
Chemical treatments: Six groups of experiments were conducted to investigate the importance of extracellular superoxide, iron, and nitric oxide in cold-induced brain edema. In all groups, drugs were dissolved in saline and injected at 0.008 cc/g minutes prior to cold injury. In Group 1, edema formation of EC-SOD transgenic mice was comDared with that of nontransgenic littermates. Group 2 compared 0 edema formation between wild-type (C57BL/6 X C3H)Fl mice treated with saline and (C57BL/6 X C3H)Fl mice treated with 0.33 mg/g deferoxamine (0.51 pmoles/g). Group 3 compared (C57BL/6 X C3H)F1 mice treated with saline to (C57BL/6 X C3H)F1 mice treated with 0.51 gmoles/g Fe 3 saturated deferoxamine. Group 4 consisted of (C57BL/6 X C3H)F1 mice treated with saline and (C57BL/6 X C3H)F1 mice treated with 0.02 mg/g N-w-nitro- L-arginine methyl ester. Group 5 consisted of (C57BL/6 X C3H)F1 mice treated with saline and (C57BL/6 X C3H)F1 mice treated with 0.02 mg/g N-u-nitro- L-arginine methyl ester plus 0.05 mg/g L-arginine.
Group 6 compared edema formation between nontransgenic mice, EC-SOD transgenic mice treated with saline, and EC-SOD transgenic mice treated with 0.02 mg/g N-a-nitro- L-arginine methyl ester.
Iron saturated deferoxamine was made by dissolving equimolar amounts of deferoxamine and then ferric chloride in saline. Saturation of deferoxamine with .0 ferric iron was determined spectrophotometrically be measuring the absorbance at 425 nm (e=2500 M-1 cm-1 for Fe3+-deferoxamine) (Monzyk and Crumbliss, J. Amer. Chem.
Soc. 104:4921 (1982)) Evan's blue treatment: One hour and 50 minutes after cold injury, 5 ml/kg of 1% Evan's Blue in saline was injected into the femoral artery of transgenic and nontransgenic mice. The mice were sacrificed 1: minutes later. The lungs were then excised and the mice were then perfused with normal saline through the left ventricle until there was no more blue color in the effluent. The brains were then removed and photographed.
Statistical analvyis: A paired Student's t-test was used to compare significance of edema develcoment compared to nontransgenic mice or saline treated mice for each of the groups examined. Analysis of variance with a Fisher PLSD test was used to compare significance in Group 6. P-value less than 0-05 were considered to be significant.
Results: Cold-induced Brain Edema: When transgenic mice and nontransgenic littermates were subjected to cold-induced injury to the right cerebral hemisphere it was found that the transgenic mice were significantly protected against edema formation compared to nontransgenic littermates (Figure 11). Percent edema was 44% less in 0 transgenic mice then in nontransgenic littermates and Evan's blue dye extravasation was visibly less in transgenic mice compared to nontransgenic littermates (Figure 12).
To test the contribution of iron to edema formation in this model, mice were precreated with i.p. injections of deferoxamine or saline prior to cold-induced injury.
Table II shows that pretreatment with deferoxamine resulted in 43% less edema formation compared to mice only given saline. Mice were then pretreated with i.p.
3 injections of iron-saturated deferoxamine or saline before cold-induced injury to see if the iron chelating properties of this compound were truly necessary for protection against edema formation. Table IV shows that, even when deferoxamine was saturated with iron, it 3 was still capable of protecting against edema formation and resulted in 32-48% less edema than that found in saline treated controls. The absolute values for the edema index were found to quite variable, however, repeated experiments consistently show the same signficant trends in protection against edema formation in the various treatments examined.
Table II Evaluation of the effect of the ferric iron chelator deferoxamine on edema formation after cold-induced brain injury. Wild-type (C57BL/S X C3H)F1 mice were treated with deferoxamine to determine what effect the iron chelating properties of this compound had on edema formation. Values are presented as mean standard error.
Treatment n Edema Index Saline 7 5.92±0.62 Deferoxamine 7 3.41±0.49* Run 1: Saline 6 8.24±0.53 Fe-Deferoxamine 6 5.63±0.63* Run 2: Saline 5 7.65+1.30 Fe-Deferoxamine 5 3.97+0.59* *p<0.05 compared to Edema Index of respective saline treated controls using a paired Student's t-test.
These results indicate that deferoxamine is capable of protecting against edema formation by a mechanism independent of its ability to scavenge iron. Because deferoxamine is capable of scavenging both the peroxynitrite anion (Radi et al, Arch. Biochem. Biophys.
288(2):481 (1991)) as well as the hydroxyl radical (Hoe et al, Chemico-Biological Interactions 41:75 (1982)), it was hypothesized that it is these properties of deferoxamine that enable it to protect against vasogenic edema.
To test this hypothesis, the synthesis of nitric oxide was inhibited with N-o-nitro-L-arginine methyl ester, a competitive inhibitor of the enzyme nitric oxide synthase, to determine if this would result in protection against edema formation after a cold-induced injury. Table III shows that treatment with N-o-nitro- L-arginine methyl ester significantly protected mice against edema formation resulting in 37% less edema formation than that occurring in saline treated controls. This protection by N-w-nitro-L-arginine methyl ester was reversed by simultaneous administration of an excess of L-arginine to the mice (Table
III)
Table III The effect of inhibition of nitric oxide synthesis on edema formation after cold-induced brain injury. Wildtype (C57BL/6 X C3H)Fl mice were treated with the competitive inhibitor of nitric oxide synthase, N-wnitro-L-arginine methyl ester (LNAME) to determine what effect nitric oxide had on vasogenic edema. Mice were also given N-W-nitro-L-arginine methyl ester plus an excess of L-arginine (LNAME L-Arg) to see if the effects seen with LNAME alone could be reversed. Values are presented as mean standard error.
Treatment n Edema Index Saline 6 5.77+0.29 LNAME 6 3.65±0.51* Saline 6 6.56±0.21 LNAME L-Arg 6 6.03±0.71 *p<0.05 compared to Edema Index of respective saline treated controls using a paired Student's t-test.
In the final experiments, EC-SOD transgenic mice were treated with either saline or N-w-nitro-L-arginine methyl ester to determine if there was an additive effect in preventing edema formation in mice which have both increased levels of EC-SOD as well as the inhibitor of nitric oxide synthase. Table IV shows that when EC-SOD transgenic mice were given the inhibitor of nitric oxide synthase, no added protection against edema formation was detected relative to transgenic mice protected only by increased levels of EC-SOD in the brain.
Table IV Evaluation of the effect of inhibition of nitric oxide synthesis on edema formation in transgenic mice.
Comparison of edema formation in nontransgenic mice to edema formation in transgenic mice with elevated levels of brain EC-SOD activity, and to edema formation in transgenic mice treated with an inhibitor of nitric oxide synthesis (20 mg/kg N-a-nitro-L-arginine; Transgenic LNAME) 15 minutes prior to cold-induced injury. Values are presented as mean standard error and were compared using analysis of variance with a Fisher PLSD test. No significant difference was seen between transgenic and transgenic LNAME mice.
Treatment n Edema Index Ncntransgenic 6 7.91±0.67 Transgenic 6 4.91±0.78* Transgenic LNAME 6 4.30±0.96* *p<0.05 compared to Edema Index of nontransgenic mice.
Example IV Immunolocalization of EC-SOD Protocols: Human 1ufl: Five human lung samples were obtained to evaluate the distribution of EC-SOD in human lung tissue. One sample was obtained from a surgical pathology specimen of a right upper lobe resected from a 43 year old white female with a 50 pack year smoking history (equivalent to one pack per day for one year) and a solitary nodule found on chest X-ray. The patient was diagnosed with squamous cell carcinoma. Tissue from a region not involved in the carcinoma from this lobe was used in the studies presented here. A second lung was obtained from a right upper lobe surgical pathology specimen resected from a 51 year old white male wizh a pack year smoking history found to have an isolated nodule on X-ray. The patient had no other illness and was diagnosed with squamous cell carcinoma. Lung tissue not involved in the carcinoma from this specimen was 2 used for the localization of EC-SOD. A third lung was obtained from a rapid autopsy (tissue obtained 6 hours after death) of a 66 year old white male with demenzia, but no history of smoking or lung disease. The fourth lung examined was obtained from excess lung tissue of a lung too large for the recipient of a lung transplant.
The lung was donaced from a 45 year old white female with no history of smoking or lung disease. The fifth lung examined in these studies was also from excess lung tissue used for lung transplantation from a 39 year old white male with no history of smoking or lung disease.
Notably, no differences in labeling patterns were seen between the surgical pathology specimens, the autopsy tissues from donors for lung transplantation.
The tissues were fixed in 2%s paraformaldehyde/0.2% gluteraldehyde in 0.01 M phosphate buffered saline (PBS; 1.2 g NaH2PO 4 8 g NaCl, 0.2 g KC1, in 1 liter pH 7.3) for 1 hour followed by overnight fixation in 4% paraformaldehyde at 4 0 C and then in O.C.T. compound.
The tissues were frozen in liquid nitrogen chilled hexane and stored at -700C until they were sectioned for light microscopic studies.
For electron microscopic studies, lung tissues were processed as in the light microscopic studies up to the equilibration in sucrose. After equilibration in sucrose, the lung tissues were infiltrated with gelatin at 37 0 C for 10 minutes. The tissue slices, in gelatin, were then solidified on ice, cut into 2 mm/side cubes, and then cryoprotected in 4% polyvinyl alcohol containing 2 M sucrose overnight. These samples were then mounted onto stubs, flash frozen in licuid nitrogen, and.then stored in liquid nitrogen until they were sectioned for electron microscopic studies.
Characterization of anrtibcdv co human recombinant SC-SOD: Human recombinant EC-SOD (furnished by S.L.
Marklund, Umea, Sweden; Tibell et al Proc. Natl. Acad.
Sci. USA 84:6634 (1987)) and the 20,000 x g supernatant of a human lung hcmogenate were denatured in the presence of S-mercap:oethanol and sodium dodecyl sulface 3 by boiling for 5 minutes and then electrophoresed through 12% polyacrylamide gel in the presence of sodium dodecy! sulfate. The protein was then electrophoretically transferred to nitrocellulose. The blot was then incubated with 4.3 gg/ml of an IgG purified fraction of rabbit anti-rh-EC-SOD (furnished by S.M. Marklund, Umea University Hospital, Umea, Sweden) affinity purified with rh-EC-SOD followed by incubation with 125I-protein A and autoradiography.
Absorption of anti-EC-SOD IG: CNBr activated sepharose was swollen in PBS. Swollen gel was mixed with PBS so that the settled gel occupied 50% of the volume. The gel was suspended and 100 1 l was mixed with 100 gg pure rh-EC-SOD for 2 hours at room temperature while gently agitating. The gel was then washed 4 times with PBS 1% bovine serum albumin (BSA) and made up 100 pg with PBS 1% BSA. 100 pl of rabbit anti-rh- EC-SOD at two times the concentration used for C immunolabeling was then added and mixed for 2 hours with gentle agitation at room temperature. Non-immune rabbit IgG was then added to the supernatant in a concentration equivalent to the predicted concentration of anti-rh- EC-SOD IgG removed by the procedure. This supernatant was then used for subsequent immunolabeling.
Liaht microsconic immunohisochemistry: 4 gm serial sections of O.C.T. embedded tissue were cut on a cryostat at -20 0 C and put on poly-L-lysine-coated slides (3 sections/slide). Sections were stored ac -70 0 °C until labeling was done. Sections were then labeled for EC-SOD using an indirect immunopercxidase method (Milde et al, J. Histochem. Cytochem. 37:1609 (1989); Randell et al, Am. J. Respir. Cell. Mol. Biol. 4:544 (1991)) with a biotinylated goat anti-rabbit IgG and streptavidin-horseradish peroxidase (Jackson, ImmunoResearch Laboratoreis (West Grove, Pennsylvania)) (Table To reduce background staining, the sections were incubated in 1% H 2 0 2 in methanol to inactivate endogenous peroxidases, 10 mM borohydride' to block aldehydes, and nonspecific binding was blocked by incubation with 5% normal goat serum (NGS), 5% milk, and 1% BSA in PBS. The optimal primary and secondary antibody dilutions were determined empirically and made in PBS with 1% milk plus 1% BSA (milk was not included in the streptavidin solution). The slides were developed using diaminobenzidine (10 mg diaminobenzidine, 50 ml 0.05 M Tris-Cl, pH 7.6, 100 Ul 3% H 2 0 2 and counterstained with 1% methyl green. As a control, LO serial sections were separately labeled with either rabbit anti-rh-EC-SOD (EC-SOD), non-immune rabbit IgG, or rabbit anti-rh-EC-SOD from which EC-SOD binding IgG had been absorbed out (EC-SOD absorbed; see above).
TABLE V Staining procedures for light microscopic immunohistochemistry. All incubations were performed in a humidified chamber at room temperature.
Incubacion Time Step 1 11 H 2 0 2 in mecthanol minutes Step 2 10 mM borchydride in PBS .0 30 minutes (glu7eraldehyde fixea tissue only: Step 3 5% NGS, 5% milk, BSA/PBS minutes Step 4 Primary antibcdy 1%e milk, 1% BSA/PBS 1 hour (various dilutions) Step 5 Biotin-labeled goac a-.zi-rabbi IgG 1 hour (1:6000 in 1% milk, I% BSA/PBS) Step 6 Streptavidin.Horse radish peroxidase 1 hour :0 (1:2000 in 1% BSA/PBS Step 7 Diaminobenzidi.e 15 minutes Step S 1% Methyl Green in water 1S minutes Electron microscooic iniunocvcocheminstrv: Ultrathin cryo sections (70 nm) of human lung tissue were immunolabeled with rabbit anti rh-EC-SOD and protein A-gold as prevsicusly described (Crapo et al, Proc. Nati. Acad. Sci. USA 89:10405 (1992)) (Table VI).
Briefly, sections were first incubated three times for five minutes at room temperature in 0.15% glycine in PBS 0 to block aldehyde groups. Nonspecific binding was further blocked by incubation in 1% BSA in PBS for minutes. Primary and secondary antibody dilutions were determined empirically and made in PBS containing 1% BSA. Sections were stained with uranyl oxalaze and uranyl acetate in methyl cellulose as previously described (Crapo et al, Proc. Natl. Acad. Sci. USA 89:10405 (1992)). Control groups were as described for light microscopy above.
TABLE VI Staining procedures for electron microscopic immunohistochemistry. All incubations were performed at room temperature.
Incubation Time Step 1 PBS O.15% glycine 3 x 5 minues Step 23 Primary anibody ir BSA/BS 5 minutes Step 3 Primary antibody in Ik BSA/PBS minutes Step 4 Protein -A gold minutes Step 5 Uranvy oxalate 5 minutes Step 6 Uranyl acetate/methyl cellulose 10 minutes Results: Characteristic of EC-SOD antibody: The antibody to rh-EC-SOD was characterized by Western blot analysis of rh-EC-SOD and a human lung homogenate. Figure 13 shows that the antibody reacted with both the EC-SOD type C (top band) and the type A (bottom band) subunits (Sandstr6m et al, Biochem. J. 267:18205 (1992) in a human lung homogenate. The type A subunit does not exist in the interstium of tissues in vivo (Sandstrom et 3 al, Biochem. J. 290:623 (1993)). The antibody reacted with three bands in the lane containing purified type C rh-EC-SOD. The two lowest molecular weight species in Figure 13 are due to partial insufficient glycosylation of the rh-ECSOD in the heavily overproducing CEO-cells.
Liaht microscoPic immunohistochemist2r: Using an antibody to rh-EC-SOD, this protein was immunolocalized in human lungs. Light microscopic immunchistochemistry revealed with EC-SOD is mainly associated with the connective tissue matrix around vessels and airways in the lung (Figure 14a and b, Figure 15a, b, and c) EC-SOD was found in close proximity to vascular and airway smooth muscle (Figure 14a and b, and Figure EC-SOD was also seen in connective tissue of alveolar septal tips (Figure 15c) suggesting an affinity of EC-SOD for connective tissue matrix. No labeling was seen in association with vascular endothelial cells in large elastic arteries, medium-sized vessels, or canillaries, (Figure 14a and EC-SOD was notably absent from the epithelial cell surfaces of airways (Figure 15a and b) and was also not present in cartilage (Figure The antibody to EC-SOD was an IgG polyclonal rabbit antibody which was affinity purified using rh-EC-SOD.
To test the specificity of the labeling for EC-SOD, IgG soecific for EC-SOD was absorbed out of the antisera using pure rh-EC-SOD bound to cyanogen bromide sepharose. Nonimmune rabbit IgG was then added to this absorbed antibody in a sufficient amount to replace the absorbed IgG. Labeling lung tissues with this preabsorbed antibody preparation resulted in the absence of labeling in all areas of the lung including the pulmonary vasculature (Figure 14c). Labeling lung tissue with nonimmune IgG alone also resulted in the absence of labeling in all areas of the lung. The controls indicate that the labeling observed with the primary antibody is specific for EC-SOD in the lung.
Electron microscopic immunocvtchemisrry:
A
summary of the labeling for EC-SOD in the lung found using electron microscopic immunocytochemistry is summarized in Table VII. EC-SOD was mainly associated with extracellular matrix proteins in all regions of the lung. In particular, a high degree of labeling was seen in areas rich in type I collagen (Figure 16) and in association with other unidentified proceoglycans in extracellular matrix (Figure 17). Notably, no labeling for EC-SOD was seen in elastin-rich areas (Figure 16).
A high degree of labeling was observed near the surface of smooth muscle cells and in the connective tissue matrix surrounding smooth muscle cells in vessels (Figure 17) and airways. Labeling was notably absent from the surface of endothelial cells on small, medium and large vessels (Figures 18a and The lack of endothelial cell labeling found with the light microscopic immunohistochemistry support the electron microscopic findings. Labeling of EC-SOD was also seen in plasma within the lumen of blood vessels (Figure 18a). The localization of EC-SOD in plasma is expected as this protein was first discovered in plasma (Marklund, Acta Physiol. Scand., 5492:19 (1980)).
Labeling for EC-SOD was observed in the intercellular junctions between bronchial epithelial cells (Figure 19), but was absent from the apical surface of these cells. Finally, EC-SOD labeling was absent from the surface of type I and type II cells. A moderate, but consistent amount of intracellular EC-SOD was found in type II epithelial cells and in bronchial epithelial cells (Figure 19).
Table VII Distribution of EC-SOD in human lung. indicates presence of labeling for EC-SOD and indicates no labeling for EC-SOD. represents areas where low low amounts of labeling for EC-SOD were inconsistently observed.
Location
EC-SOD
Cell Surfaces Endothelial Type I cell Type II cell Smooth muscle cell Fibroblast Extracellular Matrix Type I collagen Elastin Cartilage Unidentified matrix elements Intracellular 0 Endothelial cell Type I cell Type II cell Bronchial epithelial cell Smooth muscle cell Fibroblast Blood Plasma Red Blood Cell Controls done by absorbing EC-SOD specific antibody out of the primary antibody and replacing this absorbed antibody with nonimmune rabbit IgG resulted in the absence of labeling in all areas of the lung including areas rich in type I collagen as seen in Figure 15c. In addition, use of nonimmune rabbit IgG instead of the primary antisera also resulted in the absence of .labeling in all areas of the lung. The lack of labeling with these controls indicates that the labeling observed with the primary antisera is specific for EC-SOD in the lung.
The localization of EC-SOD on the surface of smooch muscle cells and in the extracellular matrix around these cells in both blood vessels and airways indicates that EC-SOD may have an important function in this location. Superoxide is known to rapidly react with nitric oxide and inactivate its properties as a smooth muscle relaxant. Therefore, the presence of EC-SOD along the diffusion oath of nitric oxide to smooth muscle cells should increase the half life of this short lived intercellular messenger in this particular region and thus increase its potency as a vasodilator. The high labeling for EC-SOD seen around vascular and airway smooth muscle cells indicates a function for EC-SOD as a mediator of nitric oxide activity in the maintenance, of low pulmonary vascular pressures and airway resistence.
In addition to the labeling of EC-SOD in association with smooth muscle cells, EC-SOD also appears to strongly colocalize with type I collagen.
mx Collagen has previously been demonstrated to be susceptible to attack by reactive oxygen species such as the superoxide anion. In addition, the superoxide anion may be capable of activating latent collagenases from polymorphonuclear leukocytes (PMN) which can lead to further collagen degradation. Because collagen fragments have been shown to chemoattract and activate PMN's, any increased produced of superoxide that results in collagen degradation may accelerate inflammatory 0 reactions and tissue destruction through PMN recruitment and activation. Consequently, the association of EC-SOD with collagen may be importanc in preventing superoxide mediated degradation of collagen and therefore, represent a means of controlling inflammatory responses.
ORMW
Example V Human EC-SOD Gene Protocols: Materials and radiochemicals: [a-35SjdATP (-1400 Ci/mmol), Ly- 3 2P]ATP (3000 Ci/mmol), and [a- 3 5 P1CTP (800 Ci/mmol), were purchased from New England Nuclear. Human genomic DNA, T 7
T
3 and SP6 RNA polymerase, RNasin, and the pGEM3Zf(+) plasmid were obtained from Promega Biotec. Secuenase 0 sequencing kit (V 2.0) was from United States Biochemicals Corporation. Human poly A+ RNA was acquired from Clontech. SeaPlaque GTG agarose was from FMC BioProducts. Restriction enzymes were from New England Biolabs. All other reagents used were molecular biology grade. Oligonucleotides were synthesized using an Applied Biosystems 380B or 392 by the Duke University, Department of Botany DNA synthesis facility.
Charged nylon membranes (GeneScreen Plus) were from DuPont.
2 Human Northern blot or analysis: Two pg poly A+ RNA were purified from eight different human tissues. These mRNAs were electrophoresed on a denaturing formaldehyde 1.2% agarose gel and transferred to a charge-modified nylon membrane followed by UV irradiation fixation. The membrane was prehybridized in 50% formamide, 0.25 M NaPO 4 (pH 0.25 M NaCI, I mM EDTA, 7% SDS, and polyethylene glycol (molecular weight 8000). The blot was hybridized in the same buffer overnight a: S o C with I X 106 cpm/ml of [32P] -labeled human EC-SOD RNA generated by transcription of the full-length cVNA using
T
3 RNA polymerase in the presence of [a- 32 PICTp. The blot was washed in 0.25 M NaPO 4 (pH 1% SDS, and ImM EDTA at 75°C followed by a second wash using 0.04 M NaPO 4 (pH 1% SDS, and 1 mM EDTA at 750C for LO 30 minutes. This was followed by exposure to XAR-5 film using a Lightening Plus intensifier screen at -70 0
C.
The autoradiogram was scanned using an LKB Ultrascan
XL
laser densitomer, and the peaks were quantitated by integration using the internal digital integrator of the densitometer or by cutting out the peak from a =rinter tracing and weighing.
Racid amo.ifciation of cDNA ends: gg of poly A+ mRNA from human heart was reversed transcribed using 2 pmoles of EC7, a 5' EC-SOD 0 gene specific anti-sense oligonucleotide ATGACCTCCTGCCAGATCTCC-3'), following a protocol by GISCO BRL RACE System). The RNA template was degraded by the addition of RNase H at 55 0 C for 10 minutes. The resulting cDNA was isolated using a GlassMAX DNA (GIBCO BRL) isolation spin cartridge. The purified cDNA was dC-tailed using terminal deoxynucleotidyl transferase (TdT, 0.5 units/ul). 200 M dCTP, 10 mM Tris-HCl (pH 25 mM KC!, 1.25 mM MgC1 2 and 50 gg/ml bovine serum albumin for 10 minutes at 37°C. The TdT was heat inactivated for 10 minutes at Products of this reaction were next PCR amplified using the "anchor" primer (GIBCO BRL), which hybridizes to the homopolymeric tail, and EC4 (a nested internal EC-SOD gene specific anti-sense oligonucleotide, 5'-AGGCAGGAACACAGTAGC-3'). Alternatively, the dC-tailed products were PCR amplified using EC7 and HEC1 (a sensestrand EC-SOD gene specific primer, in 5'-TGGGTGCAGCTCTCTTTTCAGG-3'). The final composicion of the reaction included 20 mM Tris-HC1 (pH 50 mM KC1, 2.5 mM MgCl 2 100 pg/ml bovine serum albumin, 400 nM Anchor primer, 200 nM gene-specific primer, and 200 AM each of dATP, dCTP, dGTP, dTTP. After incubating the PCR reaction for 5 minutes at 94°C, amplitaq (Perkin Elmer Cetus) was added at a final concentration of 0.04 units/l!. PCR cycling was performed on a Perkin Elmer 9600 for 35 cycles with melting at 94°C for 45 seconds and annealing at 53°C for 15 seconds and extension at 720C for 90 seconds. The full-length EC-SOD cDNA (6 ng) was used as a positive control in the PCR reaction. The PCR products were electrophoresed in a 2% SeaPlaque GTG agarose gel, transferred to charged nylon membranes by the method of Southern (Southern, J. Mol. Biol. 98:503 (1975)) using the alkaline transfer protocol (Reed. et al, Nuc. Acids Res. 13:7207 (1985)). The DNA was fixed to the membrane by baking at 80°C in a vacuum oven for 2 hours. The subsequent blot was hybridized to [32p] end-labeled HEC2 (an internal, nested EC-SOD specific i0 primer, 5'-TCCAGCTCCTCCAAGAGAGC-3') overnight at 370C.
The blot was washed at increasing stringencies until background hybridization was removed. This was followed by exposure to XAR-5 film using a Lightening Plus intensifier screen at -70 0
C.
Human genomic Southern blot analysis: Ten Ag human genomic DNA were digested BamH I, EcoR I, Kpn I, and Psc I restriction endonuclease enzymes until completion. The DNA was then electrophoresed on 1% agarose gel and transferred to a 0 charged nylon membrane by the Southern technique (Southern, J. Mol. Biol. 98:503 (1975)), after alkaline denaturation (Reed et al, Nuc. Acids Res. 13:7207 (1985)). The DNA was fixed to the membrane by heating to 80 0 C in a vacuum oven for 2 hours. 32 p]CTP-labeled human EC-SOD antisense strand cRNA was synthesized using the EC-SOD cDNA which had been linearized with Stu I.
The blot was hybridized (500 X 10 3 cpm/ml) in formamide, 0.25 M NaPC4 (pH 0.25 M NaC1, 1 mM EDTA, 7% SDS, and 5% polyethylene glycol (molecular weight 8000), at 50 0 C. Following overnight hybridization, they were washed in 0.25 M NaPO 4 (pH 2% SDS, and 1 mM EDTA followed by 0.04 M NaPO 4 (pH 1% SDS, and 1 mM EDTA at increasing stringencies until background hybridization was minimized. The blot was exposed to XAR-5 film using a Lightening Plus intensifier screen at -700C.
Isolation of the human gene for EC-SOD: A human adult female leukocyte genomic library constructed in the EMBL-3 vector was obtained from Clontech. Approximately 1 X 106 pfu were screened at a density of -50,000 pfu/plate using 32 p]CTP-labeled human EC-SOD cRNA (1 X 106 dpm/ml). The primary through tertiary screens identified approximately 7 unique putative positive plaques. Individual plaques were isolated and lambda DNA purified using LambdaSorb phage 0 adsorbent (Promega Biotec). The size of the insert DNA from each clone was assessed by Sal I restriction endonuclease digestion followed by electrophoresis in 0.7% agarose. Selected clones underwent extensive restriction endonuclease mapping. Based on the restriction mapping results and asymmetric hybridization using 5' and 3' annealing EC-SOD oligonucleotides, Clone #7 was selected for all subsequent DNA sequence analysis. Clone #7 contains an approximate 18-20 kb fragment.
DNA sequencing of the human EC-SOD aene: The overall strategy used for sequencing clone #7 is illustrated in Figure 20. Various size restriction endonuclease DNA fragments from clone #7 were subcloned into the pGEM3Zf(+) vector DNA. The dideoxy sequencing method using double-stranded DNA (Ausubel et al, Current Protocols in Molecular Biology, Green Publishing Assoc.
and Wiley Interscience, New York (1992)) as template and Sequenase enzyme (United States Biochemicals) were employed (Sanger et al, Proc. Natl. Acad. Sci. USA 74:5463 (1977)). Both the Universal and -40 M13 sequencing primers were used to initiate DNA sequencing for each subcloned fragment. Oligonucleotides derived from this initial sequencing data were synthesized approximately every 250 base pairs until the complete nucleotide sequence was obtained. Sequencing data were obtained from both strands as shown in Figure 203 except at the 3' portion of the gene where DNA sequence was obtained on one strand only.
-0 Computer-assisted sequence analysis and transcriptional database search: The IntelliGenetics Geneworks program (Version 2.2) was used for organizing the DNA sequence data. Eomology searching was performed at the NCBI using the BLAST (Altschul et al, J. Mol. Biol. 215:403 (1990)) network service and the non-redundant nucleotide sequence database (GenBank(77.0) EMBL(35.0) EMBLUpdate GBUdate). Transcriptional factor database searching was performed using both the SIGNAL SCAN 3.0 algorithm 0 (Prestridge et al, CABIOS 9:113 (1993)) as well as the FINDPATTERNS program of the GCG Package (V 7.2) using Release 6.3 of the Transcription Factor Database (Gosh, Nuc. Acids Res. 18:1749 (1990)). For a prediction of the site of signal peptide cleavage, the programs SIGSEQ1 (Folz et al, J. Biol. Chem. 261:14752 (1986)) and SIGSEQ2 were employed (Folz et al, Biochem. Biophys.
Res. Commun. 146:870 (1987)).
MMMMW
Tissue specific extression of human EC-SOD: To investigate the expression of human EC-SOD, poly mRNA from eight different human tissues was fractionated on a denaturing agarose gel and transferred to a charged nylon membrane. Because a previous paper reported long exposures times in order to identify EC- SOD specific bands during genomic Southern analysis (Hendrickson et al, Genomics 8:736 (1990)), a Lu radiolabeled antisense cRNA probe derived from fulllength human EC-SOD cDNA was used (Oury et al, Proc.
Nat. Acad. Sci. USA 89:9715 (1992)). A discrete band of approximately 1.4 kb can be seen in all eight human tissues analyzed (Figure 21A). In addition, skeletal muscle contains an approximate 4.2 kb message, not detected in the other tissues. By densitometric scanning of the 4.2 and 1.4 kb bands, it can be calculated that larger message to make up about 32% of the total skeletal muscle EC-SOD message. In brain, a very faint band of 2.2 kb can be seen. This band was too weak for quantitation by laser densitometer.
Quantitation of these bands was performed by laser densitometry and integration of peaks of autoradiograms obtained in the linear range of exposure (Figure 213).
After normalizing to the brain, the heart showed the most expression with 10.1 times brain. This was followed by the placenta, pancreas, and lung which gave 13.6, 10.2, and 7.5, respectively. Expression in skeletal muscle was 4.7 for the 1.4 kb band or 6.9 for both 1.4 kb and 4.2 kb message, while the kidney and liver gave 6.3 and 4.1 time expression over brain.
These patterns of expression have been reproduced based on probing an additional independent multiple tissue Northern blot. The bands are specific based on the relatively high stringencies of washing and from data using a sense strand EC-SOD cRNA as a probe which showed no hybridization under the conditions given.
Mapina the site transcricpion initiation: Initially, mapping of the site of transcription initiation was attempted using the primer extension method. Using several different end-labeled oligonucleotides and bobh human lung and human heart poly A+ mRNA as well as total RNA isolated from human foreskin fibroblasts, a positive signal was not obtained even after long exposure times. This did not seem to be due to technique since it was not possible to get positive signals using RNA generated by in vitro transcription of the EC-SOD cDNA. Whether lack of !0 success using this technique was due to very low abundance of mRNA encoding EC-SOD or some other problem(s) is unclear. Working under the assumption of low abundance mRNA, the technique of rapid amplification of cDNA ends in order to PCR amplify this signal was attempted. The EC-SOD gene specific primer EC7 was used for hybridization and reverse transcription of human heart poly A+ mRNA as shown in Figure 22. Half of this reaction was 3' dC tailed using terminal deoxynucleocidyl transferase and the remaining half was not. These templates were then subjected to PCR amplification using the gene specific primers HECi EC7 as well as the anchor primer EC4. The products of these reactions were fractioned by agarose electrophoresis, transfered to nylon membranes, and probed with the interal nested gene specific primer HEC2. An autoradicgram of this experiment is shown in Figure 22A. Using EC-SOD cDNA as a control template and HECI EC7, a band of 217 bp is expected (lane 3 of 1" Figure 22A). Since the primers HECI and EC7 are expected to amplify independant of dC tailing, bands of equal intensity in lanes 4 and 5, which are also of the same size as the EC-SOD control, are seen. Using the anchor primer (which hybridizes to the dC tail) and EC4, only one band of -190 bp was seen (lane Since the template was not poly C tailed, lane 2 shows no signal as expected. By subtracting 48 bp (the size of the anchor primer), the size of the reverse transcribed DNA would correspond to -136 bp 5' of the EC4 primer. This analysis would predict that there is approximately 6 base pairs of additional 5' sequence on the cDNA clone and that transcription initiation starts about 6 bp upstream of the first intron (indicated by a dashed box). Although eukaryotic transcription initiation usually begins at an adenosine residue, it is expected that it will begin at a G (Breathnach et al, Ann. Rev.
Biochem. 50:349 (1981)) Genomic Southern blot analysis: To begin to charcterize the human EC-SOD gene, pg of the total human genomic DNA was restriction digested and the reaction products electrophoresed on an agarose gel followed by transfer to a nylon membrane.
The blot was probed with a (32p]-labeled partial EC-SOD cRNA. An autoradiogram of this blot is shown in Figure 23. As can be seen for each lane, there are unique bands associated with each restriction digest. No shadow bands which might suggest pseudogenes were seen.
When a full-length cRNA probe was used for Kpn 7 digested DNA, an additional band of -4000 bt was seen which corresponds to the 3' end of the gene. :n addition, the Kpn I lane shows a 0.5 kb band which was better seen on other blots. This banding pattern was similar to a restriction map of the human EC-SOD clone #7 (see Figure Isolation and characterization of the hm;a n EC-SOD by DNA secuencing: Multiple independent positive clones were identified from a human adult leukocyte genomic library constructed in EMBL-3. These clones underwent extensive restriction endonuclease mapping and were probed with EC-SOD specific 5' and 3' oligonucleotides in order to determine the relative orientation of the inserts.
Based on these results, clone #7 was picked for further analysis. Clone #7 is about 18 to 20 kb and contains at least 5000 bp of 5' flanking DNA and at least 4000 bp of 3' flanking DNA. Restriction mapping of clone #7 is shown in Figure 20A. This map is similar to the results obtained with genomic Southern blot analysis data indicating that clone #7 contains the EC-SOD gene. The strategy for sub1coning and sequencing clone #7 is shown in Figure 20B. Various size continguous and overlapping restriction fragments were subcloned into the plasmid vector pGEM32f(+) (Figure 20B). The DNA inserts were sequenced on both strands using a combination of primer walking and vector specific, universal sequencing primers. The 3' half of 7K36 insert was sequenced on Ul one strand only. Published sequence data for the human EC-SOD cDNA (Hjalmarsson ec al, Proc. Nat!. Acad. Sci.
USA 84:6340 (1987)) as well as DNA sequence information obtained from an independant cDNA clone which contained additional 5' untranslated data (Hendrickson et al, Genomics 8:736 (1990)) were used to determine the genomic intron/exon structure. Based on a comparison of these data with the genomic sequence information, the human EC-SOD gene was determined to contain three exons and two introns (Figure 20C) Excn 1 contains at least 5 base pairs and is probably larger (by about 6 base paris), since the exact start of transcription initiation was not determined (note below). Exon 2 contains 84 bp and is separated from exon 1 by a 572 bp intervening sequence marked as intron 1. Exon 3 is separated from exon 2 by intron 2, a 3849 bp segment.
Exon 3 contains a total of 1336 bp and at 17 bp into this exon starts the beginning of the complete coding sequence for preEC-SOD (Figure 20D). This includes an 18 amino acid signal peptide that precedes the 222 amino \0 acid mature protein sequence. There are no introns separating the various structural domains of EC-SOD.
These domains are shown schematically in Figure 20D and include amino acids 1-95 which contain a glycosylated Asn-89 and show no sequence homology with other proteins. Resides 96-193 show strong homology with CuZn-SOD protein sequences with preservation of critical amino acids important in enzyme catalysis and structure.
Amino acids 194-222 contain multiple charged resides which have been shown to be important in binding to sulfated proteoglycans. 558 bp of the region containing putative regulatory elements and 3675 bp of the 3'-flanking region were also sequenced. The exonic DNA sequence data are in agreement with the published cDNA sequence (Hjalmarsson et al, Proc. Natl.
Acad. Sci. USA-84:6340 (1987)). The intron-exon boundries are shown in Table VIII and conform to the eukaryotic consensus splice sequence (Senapathy et al, Methods Enzymol. 183:252 (1990)). Both introns split sequences in the 5'-nontranslated region of the EC-SOD gene.
100 Table VIII Sequences at incron/exon splice junctions The size of the introns and exons are shown in base pairs The uppercase letters indicate exon sequence while the lowercase letters indicate intron sequence. The splice junctions shown conform to previously published concensus sequences for splice junctions (Senapathy et Methods Enzymol. 183:252 (1990)) 0 Donor Intron Acceptor Exon size (bp) TGCGGG g: ggac 572 gccc ag GCTCCA 84 GGAAAG gt gggt 3849 ccgc ag GTGCCC :336 Figure 24 shows the entire sequence for the human EC-SOD gene. Exonic sequences are shown in boxed uppercase letters while intronic, and 3'-flanking sequence are shown in lowercase. Exon 3 contains the entire uninterrupted coding region for EC-SOD and the protein sequence is shown using the single letter amino acid code. The 18 amino acid signal peptide and 222 amino acid mature protein sequence are highlighted. The identification of the signal peptide cleavage site is consistent with computer algorithms which predict the site of eukaryotic signal peptide cleavage (Folz et al, Biochem. Biophys. Res. Comm. 146:870 (1987)); Von Heijne, Eur. J. Biochem. 133:17 (1983)).
Transcriptional factor database searching was used to putatively identify transcriptional regulatory elements. Although almost all eukaryotic promoters utilize a TATA box element to fix the position of transcription initiation, an obvious TATA box cannot be discerned for the EC-SOD gene. Two CAAT box elements were identified. One is in the reverse orientation and located about 20 bp upstream of the first exon, while the second can be found about 335 bp upstream. The putative signal for polyadenylation is shown and the site of poly A adenylation is indicated.
Transcriptional factor database searching of the '0 nontranslated region and first intron identified several potential regulatory elements. A cAMP responsive element (CREB) (TGACGT) which is similar to the adenovirus transcription virus (ATF) element can be found starting at 121 bp (Fink et al, Proc. NatI. Acad.
Sci. USA 85:6662 (1988); Sassone-Corsi Proc. Natl. Acad.
Sci. USA 85:7192 (1988)). A half site for the glucocorticoid response element (GRE) (TGTCCT) is located at 370 bp (Karin et Nature 308:513 (1984)).
A skeletal muscle specific trans-activaning factor 3 response element (M-CAT) (CATTCCT) is found in the reverse orientation beginning at position 238 (Mar et al, Mol. Cell. Biol. 10:4271 (1990)). A xenobiotic responsive element (XRE) (CACGCW) is found within the first intron at position 1085 bp (Rushmore et al, J.
Bicl. Chem. 265:14648 (1990)). A metal regulatory element (MRE) (TGCRCYC) is found at position 89 (Culotta et al, Mol. Cell. Biol. 9:1376 (1989)). Two putative antioxidant response elements (ARE) (RGTGACNNNGC) are found at position 650 and 5022 (Rushmore et al, J. Biol.
Chem. 266:11632 (1991)) A sis responsive element (SIF) (CCCGTC) important in the induction of the c-fos protooncogene is found in the reverse orientation at position 251 (Wagner et al, EMBO J. 9:4477 (1990)). There is an API binding site or TPA responsive element (TRE) (TGACTCA) found at position 162 (Risse et al, EMBO.J.
8:3825 (1989)). The SV40 enhancer region AP4 (CAGCTGTGG) can be found at position 171 (Jones et al, Genes Dev. 2:267 (1988)).
Example VI 0 Screening Patients for Gene Defects in EC-SOD Preparation of leukocyte derived aenomic DNA from patients: Normal healthy control patients and patients with asthma, primary pulmonary hypertension, and secondary pulmonary hypertension will be identified.
Genomic DNA will be purified utilizing a Qiagen Slood PCR Kit. One ml of blood containing -107 leukocytes/ml will be- collected in sodium citrace from each patient or control subject. The blood is placed into a QIAGEN-spin column and the leukocytes are entrapped in the resin by 3 brief centrifugation, while erythrocytes and hemoglobin are washed through. The leukocytes are lysed by the addition of 0.7 ml of lysis buffer and incubated at room temperature for 30 minutes. DNA that is released, binds to the resin in the tube. Remaining cellular debris is 3 washed away by multiple wash/spin cycles. The DNA is eluted by the addition of 1.2 M KC1, 50 mM MOPS, ethanol, pH 8.3. This typically yields -10 Pg of 103 genomic DNA (Reihsaus et al, Am. J. Respir. Cell. Mol.
Biol. 8:334 (1993)).
Primer design and PCR amplification of EC-SOD exonic sequences: Sense and antisense oligonucleotide primers (or use primers already obtained from sequencing the genomic DNA) will be designed containing a 3'GC clamp (Sheffeld et al, Proc. Natl., Acad. Sci. USA 86:232 (1989)). These primers will encode the intronless coding region of the EC-SOD gene. A 172 bp region in 0 the 3' untranslated region has been amplified using DNA sequencing primers and human genomic DNA. PCR conditions are as described (Reihause et al, Am. J.
Respir. Cell. Mol. Biol. 8:334 (1993); Innis et al (eds) Academic Press San Diego pp. 1-12 (1990)) using Taq polymerase, with temperature cycling as follows: initial denaturation a: 95°C for 5 min followed by cycles of 940°C denazuring for 20 sec, 57°C annealing for 15 sec, and 720C elongation for 45 sec. Because of the GC composition and actual primer sequence, it will be necessary to experimentally optimize conditions for PCR amplification using each set of primers. Three sets of primers will be used to encompass the entire coding region.
Identification of mutations with single-strand conformational polymorghism (SSCP) analysis: SSCP analysis has been used to detect single base pair mismatch (Orita et al, Genomics 5:874 (1989)).- Temperature-gradient gel electrophoresis (TGGE) will be 104 used to detect differences in mobility (Wartell et al, Nuc. Acids Res. 18:2699 (1990)). Samples for TGC-G will be prepared by heat denaturing the PCR produce at 980C for 5 min, then renaturing at 50°C for 15 min with the corresponding wild-type DNA derived from PCR of thecloned gene. Elecurophoresis will be carried out on a acrylamide, 8 M urea gel over a temperature gradient.
The temperature gradient will be optimized for each of the EC-SOD DNA segments. Typical gradients for the detection of g2-adrenergic receptor mutations were between 35CC to 60C, and required 4 to 6 hours of run time (Rosen, Nature 262:59 (1993)).
All PCR samples found to be positive for mutations by TGGE will be sequenced directly using the dideoxy technique (Sanger es al, Proc. Natl. Acad. Sci. USA 74:5463 (1977)).
Example VII Inhibiionr of Xanthine Oxidase In a firs: study, assays were performed in a 1 ml quartz cuve=te containing 50 mM carbonate buffer, pH 0.1 mM EDTA, 1 r-M xanthine oxidase (Boehringer Mannheim) at 250C. Xanthine oxidase activity was measured spectrophotomezrically by following the loss of xanthine over time at 295 nm. Four concentrations of xanchine 50, 250, 500 gM) and 2 concentrations of MnTBAP pM) were used. Two inhibition constants were then derived from the curve's intercepts (Kii=5.5 gM) and 105 slopes (Kis=15 gM) The results presented in Figure show that MnTBA_ inhibits xanthine oxidase in a noncompetitive manner.
In a second study, calf pulmonary artery endothelial cell cultures (CPA-47 (Tissue and Cell~ 10:535 (1978)) were grown to confluence in Ham's F-12K medium with 10% fetal bovine serum at pH 7.4 and 37 0
C.
Cells were then trypsinized and seeded at equal densities in 24 well plates and grown to 90% confluence.
Cells were washed and pre-incubated for 1 hour with gM of MnTBAP in minimum essential medium (MEM) or MEM only. Varing amounts of xanthine oxidase (XC) plus 200 pM xanthine were added and allowed to incubate for 24 hours. Cell injury was quantitiated by measuring the release of cellular lactate dehydrogenase (LDH) into the medium. The efficacy of MnTBAP is shown in Figure 26 by the decrease in XO/X-induced LDH release.
Example VII SOD Mimetic Affords Cellular Protection From Paraquat-Induced Injury Rat pulmonary epithelial cell cultures (L2 (Kaighn and Douglas J. Cell Biol. 59:160a (1973)) were grown to confluence in Ham's F-12K medium with 10% fetal bovine serum at pH 7.4 and 37 0 C. Cells were then trypsinized and seeded at equal densities in 24 well plates and grown to 90% confluence. Cells were washed and preincubated for 1 hour with 100 pM of MnTBAP or MnTMPyP in 106
NOMOW
MEM or MEM only. Paraquat (2.5 mM) was added and allowed to incubate for 48 hours. Cell injury was quantitiated by measuring the release of cellular lactate dehydrogenase (LDH) into the medium. Figure 27 shows that MnTPyP (hatched bars) and MnTBAP (grey bars) decrease paraquat-induced LDH release.
In a further study, calf pulmonary artery endothelial cell cultures (CPA-47 (Tissue and Cell 10:535 (1987)) were grown to confluence in Ham's F-12K medium with 10% fetal bovine serum at pH 7.4 and 370C.
Cells were then trypsinized and seeded at equal densities in 24 well plates and grown to 90% confluence.
Cells were washed and pre-incubated for 1 hour with varing concentrations of MnTBAP in MEM or MEM only.
Paraquat (2 mM) was added and allowed to incubate for 24 hours. Cell injury was cuantitiated by measuring the release of cellular lactate dehydrogenase (LDH) into the medium. MnTBAP decreases paraquat-induced LDH release in a dose-dependent manner (see Figure 28).
.0 In contrast to MnTBAP, ZnTBAP does not protect against paraquat-induced injury. Calf pulmonary artery endothelial cell cultures (CPA-47) were grown to confluence in Ham's F-12K medium with 10% fetal bovine serum at pH 7.4 and 37 0 C. Cells were then trypsinized and seeded at equal densities in 24 well plates and grown to 90% confluence. Cells were washed and preincubated for 1 hour with varing concentrations of ZnTBAP in MEM or MEM only. Paraquat (2 mM) was added and allowed to incubate for 24 hours. Cell injury was 0 quantitiated by measuring the release of cellular 107 Ino lactate dehydrogenase (LDH) into the medium. The results presented in Figure 29 demonstrate that ZnTBAP does not possess SOD-like activity. ZnTBAP can be used as a negative control to show that the redox metal is important in the protection against paraquat toxicity.
Example IX Protection by MnTBAP Against Paraquat-Induced Lung Injury Mice were treated with either paraquat (PQ, 45 mg/kg, ip) or saline (10 ml/kg, ip) and exposed to MnTBAP (2.5 mg/ml, nebulized into a 2 1 chamber at 2 L/min for 30 minutes twice daily for 2 days) or room air. Mice were killed 48 hours after start of treatment and lung injury was assessed by analysis of bronchoalveolar lavage fluid (BALF). BALF damage markers used were lactate dehydrogenase (LDH, as units/L), procein concentration (as mg/dl) and percent of polymorphonuclear leukocytes (PMN) MnTBAP treatment provided partial protection against paraquat-induced lung injury (see Figure Example X Catalase activity was measured by means of a Clark oxygen electrode using a modified assay previously described by Del Rio et al, Anal. Biochem. 80:409 (1977) Briefly, reactions were performed in a nitrogen degassed phosphate buffer (50 mM, pH 7.8) containing 0.1 mM EDTA at 25CC. Three concentrations of hydrogen peroxide (1-4 mM) and four metalloporphyrin concentrations (0.5-50 pM) were used to determine second order rate constants. The rate of oxygen evolution was followed for 2 minutes. The results are shown in Figure 31.
Calf pulmonary endothelial cell (CPA-47) line was grown to near confluence in 12-well plates with F-12K 0 medium containing 10% fetal calf serum. CPA-47 cells were loaded with Cr" as previously described by Simon et al J. Clin. Invest. 78:1375 (1986) in high glucose minimum essential medium (DMEM). Cells were pretreated.
with MnTBAP (100 gM) for hour and then exposed to 3 various concentrations of hydrogen peroxide generator, glucose oxidase, for 4 hours. Cell injury was then quantitated as the specific Cr 5 release from CPA-47 cells which has been adjusted for spontaneous Cr., release. The results are shown in Figure 32.
Calf pulmonary endothelial cell (CPA-47) line was grown to near confluence in 12-well plates with F-12K medium containing 10% fetal calf serum. CPA-47 cells were loaded with Cr 5 as previously described by Simon et al Clin. Invest. 78:1375 (1986)) in high glucose minimum essential medium (DMEM). Cells were pretreated with various concentrations of either: MnTBAP; (B) MnTMPyP; ZnTBAP; or CuZnSOD for 1 hour and then exposed to the hydrogen peroxide generator, glucose oxidase (100 Mu/ml), for 4 hours. Cell injury was then quantitated as the specific Cr" release from CPA-47 109 cells which has been adjusted for spontaneous CrK release. The results are shown in Figure 33A-33D.
Example XI Mimetics as Protectants Against Excitotoxic Cell Death Experimental procedures Tissue culture: Mixed neuronal and glial cultures were prepared from embryonic day-18 rat cerebral cortices (Sprague-Dawley, Zivic Miller). Briefly, the cerebral cortices were dissected and enzymatically dissociated by incubation in Mg"-free Hank's balanced salt solution (HBSS) supplemented with 10 mM HEPES and 0.25 k trypsin for 20 min. at 37 0 C. The tissue was rinsed and dispersed into single cell suspension by gentle passage through a fire polished Pasteur pipette.
The cell suspension was centrifuged and resuspended in Minimum Essential Media (MEM), containing Earle's salts supplemented with 3 g/L glucose, 5% horse serum and fetal bovine serum (growth media). The cells were plated in poly-D-lysine-coated multi-well plates: 12-well plates for accnitase measurement, and 24 well plates for toxicity experiments. Cells were maintained ac 37=C in a humidified incubator with 5% CO 2 /95% air in growth media. Media was not replaced so as to reduce glial overgrowth and neuronal loss. Mature cells (14-17 days in vitro) were used for all experiments.
110 Cell treatment: The growth media was replaced with MEM supplemented with 25 mM glucose (MEM-g). For both neurotoxicity studies and aconitase measurement, cells were incubated in the designated treatment for the indicated length of time at 37 0 C. Unless otherwise.
specified, antagonists or SOD-mimetics were added min. prior to agonists. For measurement of neurotoxicity, cells were incubated with treatments for 18 hr at 37°C in MEM-g. To assess the ability of Ln antagonists to inhibit acute NMDA toxicity or rescue cells from an ongoing NMDA insult, an alternate paradigm of NMDA treatment was used in addition to the one described above. In this paradigm, growth media was replaced with HBSS- Mg--free Hank's Balanced Salt Solution supplemented with 2 mM CaCd 2 1 mM NaHCO 3 10 mM HEPES and 5 uM glycine), cells were treated with vehicle or 100 M NMDA for 15 min. after which HBSS- was replaced with MEM-g and cells were returned to the incubator for 18 additional hrs. Antagonists or SOD-mimetics were 0 added to the cells 15 min. before NMDA exposure or or 60 min. following final media replacement. For determination of Ca--dependence, cells were incubated in KBSS- with no added When kainate (KA) was used as an agonist, 100 AM D-APV was routinely included to block secondary NMDA receptor activation. Catalase (100 U/ml) was always included when xanthine plus xanthine oxidase (X+XO) was used as a treatment to rule out the contribution of hydrogen peroxide.
111 Neurotoxicity studies: Neurotoxicity was determined by the measurement of LDH released in the supernatant media as previously described (Patel et a, Toxicol. Appl. Pharmacol. 115:124 (1991) and Neurotoxicology 14:35 (1992)). LDH was measured by the method of Vassault, Lactate dehydrogenase. In: Methods of Enzymatic Analysis, Bergmeyer HU Verlag Chemie, Weinheim, pp. 118-126 (1983). Additionally, cell death was confirmed with ethidium-l homodimer (EthD-1).
Briefly, the cells were washed with HESS, loaded with pM EthD-1 for 30 min., rinsed and viewed through fluorescence optics. The number of dead cells labeled with EthD-1 were counted in 4-6 randomly selected fields, the numbers averaged to give an estimate of cell death and the experiment was repeated 3 times.
Aconitase measurement: For the measurement of aconitase, following treatment with agents, media was removed and cells lysed in ice-cold 50 mM Tris/HC1, pH 7.4 containing 0.6 mM MnCI, 1 mM L-cysteine, 1 mM 0 citrate and 0.5% Triton-X 100. The aconitase activity of cell lysates was measured spectrophotometrically by monitoring the formation of cis-aconitate from isocitrate at 240 nm in 50 mM Tris/HC1, pH 7.4, containing 0.6 mM MnC1, and 20 mM isocitrate at (Kreb and Holzach, Biochem. J. 52:527 (1952); Gardner and Fridovich, J. Biol. Chem. 267:8757 (1992)). Inactive aconitase was reactivated by a 30 min. incubation of cortical cell lysates (90pl) with 0.5 M DTT (10 ll), mM NaS (1 l) and 20 mM ferrous ammonium sulfate(1pil) 112 in 50 mM Tris/HCl, pH 8.0. at 37°C. Aconitase activity in cortical cell lysates was inhibited by 0.1 mM potassium ferricyanide or by boiling of sample.
Statistical analyses: One-way analysis of variance (ANOVA) was used to compare three or more treatments and Dunnet's test for comparing multiple treatment groups with a control group. Tukey-Kramer multiple comparisons test was used to detect differences between treatments.
Student's t-test was used for comparison of two treatments.
Results Validation of aconicase activity as a marker of 0; radical formation in cortical cell culture: With the use of agents that are known to generate O0" (X+XO and aconitase activity was established as a valid marker of The 02- generators selectively and reversibly inactivated aconitase and did so in a SODpreventable manner.
Aconicase inactivation produced by PQ", NMDA and KA correlate with cell death: Cortical cells were treated with varying concentrations of KA, NMDA, and PQ" for 18 hr. and the media analyzed for LDH activity. Sister cultures were treated with varying concentrations of KA, NMDA, or PQ" for 6, 1 or 3 hr., respectively, and cell lysates analyzed for aconitase activity; a shorter time of incubation was assessed for aconitase activity 113 because death of neurons leads to irreversible inactivation of aconitase activity, presumably due to protein degradation. Therefore, an earlier time point was selected for measuring aconitase activity based upon the time of approximating the steady state of reversible inactivation (Fig. 34 A and B for 50 AM NMDA and 300 AM KA, respectively).
KA treatment produced proportionate decreases in aconitase activity and cell death as monitored by LDH release (Fig. 35A). Likewise, PQ" treatment produced a proportionate decrease of aconitase activity and the amount of LDH release (Fig. 35 At higher concentrations (30-1000 AM), NMDA produced proportionate decrease in aconitase activity and also cell death.
However, low concentrations (3 and 10 AM) of NMDA produced cell death (LDH release) but no detectable aconitase inactivation (Fig. 35 C).
MnTBAP prevents aconitase inactivation and cell death produced by PQ, NMDA and KA: To determine whether 0 MnTBAP can prevent aconitase inactivation and cell death produced by NMDA, PQ" and KA, cortical cells were incubated with PQ-, NMDA and KA in the absence and presence of 200 gM MnTBAP. PQ", NMDA, and KA produced a 40 and 42% decrease in aconitase activity after 3, 1 and 6 hrs., respectively; 200 AM MnTBAP markedly inhibited the NMDA- and KA-mediated decreases of aconitase activity (Fig. 36 Parallel effects were evident on cell death as measured by LDH release.
MnTBAP (200 AM) markedly inhibited NMDA- and KA- 0 WOW mediated cell death (Fig. 36 The effects of MnTBAP on cell death were concentration dependent (Fig. 36 B).
MnTBAP (200 gM) produced a right-ward and down-ward shift in the concentration-response curve for NMDA, providing complete protection at low concentrationi of NMDA (Fig 37).
The possibility that MnTBAP simply prevented activation of NMDA or AMPA receptors was examined and eliminated.
To strengthen the possibility that MnTBAP decreased aconitase inactivation and prevented cell death by its SOD-mimetic action, the effects of the structurally related congener, ZnTBAP, with diminished SOD activity (ie 10-fold less) were examined. EthD-1 was used to quantitate cell death in these experiments because preliminary studies showed that ZnTBAP interfered with the measurement of LDH (MnTBAP was found to have no effect on LDH measurement). Concentration-response curves for NMDA and KA demonstrated a strong correlation 0 between cell death measured using LDH release and dead cells quantitated by EthD-1. Concentration-response curves disclosed that MnTBAP exhibited markedly increased neuroprotective effects in comparison to ZnTBAP against PQ", NMDA and KA toxicities (Fig. 38).
To determine whether addition of MnTBAP after initiation of the excitocoxic insult exerted neuroprotective effects, 100 gM NMDA was included for min. in HBSS- Mg"-free HBSS containing 5.6 mM glucose and supplemented with 2 mM CaCI., 1 mM NaHCO 3 3 mM HEPES and 5 M glycine). This acute exposure 115 paradigm induced delayed neuronal death (measured 18 hr.
later) and allowed determination of the temporal relationship between MnTBAP exposure and neuroprotection. The neuroprotective effects of MnTBAP were assessed when it was: 1) present 15 min. prior .to and during a 15 min. NMDA application (but not for the 18 hr. period after media change) (condition 2) present 15 min. prior to and during a 15 min. NMDA application and for the ensuing 18 hr. (condition "pre 0 post"); 3) added 15, 30 or 60 min. following a 15 min.
incubation with 100 pM NMDA and left in the media for the ensuing 18 hr. Condition "pre" resulted in a 25 reduction of LDH release measured 18 hr. later; using an identical time course of application of 100 pM D-APV in condition "pre", a complete blockade of LDH release was observed. In contrast, MnTBAP incubated as in condition "pre post" resulted in a 51% reduction of LDE release. Addition of 200 AM MnTBAP 15, 30 or min. following a 15 min. exposure to 100 gM NMDA resulted in a 38%, 30% and 25% reduction of LDH release respectively. In contrast, addition of 10 gM MK801 min. after NMDA treatment had a modest protective effect when added 15 min. after the NMDA insult. In order to investigate the possibility that NMDA-induced aconitase inactivation resulted from peroxynitrite formation, the effects of nitric oxide synthase (NOS) inhibitors on NMDA-induced aconitase inactivation and cell death were examined. NMDA-induced aconitase inactivation and LDH release were unchanged by the presence of 1 mM of either WN-nitro-L-arginine methyl 116
EWNW
ester, Nt-nitro-L-arginine or NI-monomethyl-L-arginine.
NOS, however, was expressed in the cortical neuronal preparation used. Thus, peroxynitrite would not appear to play an etiological role in NMDA-induced aconitase inactivaticn or cell death under the conditions used.
Example XII Learning Impairment in EC-SOD Knocked-Out Mice EC-SOD knocked-out mice were obtained from Dr. Lena Carlsson and Dr. Stephan Marklund, UmeA, Sweden.
0 Learning was assessed in knock-out mice and control mice (controls were derived from litter mates bearing normal levels of EC-SOD) using an eight arm radical maze in which food was placed in a well at the end of each arm. Food was withheld from the mice for a period of eigh: hours and then the mice were placed into the maze.
A count was made of the number of arms the mice went down to recover food before they went down any arm for a second time. Naive animals randomly went down between four and five different arms of the maze before 0 beginning to repeat.
The results presented in Figure 39 demonstrate that when the experiment is done on 24 successive days, control mice learn the pattern and are able to increase the number of arms that they go down to find food without error. The EC-SOD knock-out animals failed to show any significant learning over the entire 24 sessions.
117 Example XIII Scavenging of Peroxynitrite by Mimetics Experimental Protocols Cell culture: J774 macrophages were cultured in DMEM medium, supplemented with L-glutamine (3.5 mmol/L) and 10% fetal calf serum. Cells were cultured in 96well plates (200 g! medium/well) until confluence. To induce the inducible isoform of nitric oxide synthase (iNOS), fresh culture medium containing E. coli LPS (011:B4; 10 gg/ml) alone or in combination with murine y-interferon (y-IFN, 10 U/ml) was added in the presence or absence of the NOS inhibitor, the SOD mimetic of the combination of the two compounds for 24h. Moreover, cells were exposed to the NO donor compounds S-nitroso-N-acetyl-DL-penicillamine (SNAP) (3 mM) and diethylamine:NO NONOate (DNO) (3 mM) for 24h or to authentic peroxynitrite (1 mM) for 1h. Nitrite/nitrate concentration in the medium and mitochondrial respiration were then measured as described below.
0 Measurement of nitrice/nitrate production: Nitrite/nitrate production, an indicator of NO synthesis, was measured in the supernatant. First, nitrate in the culture medium was reduced to nitrite by incubation with nitrate reductase (670 mU/ml) and NADPH 3 (160 gM) at room temperacure for 2h. After 2h, nitrite concentration in the samples was measured by the Griess reaction, by adding 100 Al of Griess reagent (1% 118 sulfanilamide and 0.1% naphthylethylenediamide in phosphoric acid) to 100 Al samples of conditioned medium. The optical density at 550 nm (0D 5 5 0 was measured using a Speccramax 250 microplate reader (Molecular Devices, Sunnyvale, CA). Nitrate concentrations were calculated by comparison with OD 550 of standard solutions of sodium nitrate prepared in culture medium. All measurements were corrected for the interference of MnTBAP at this wavelength. MnTBAP (up to 300 pM) did not scavenge nitrite or nitrate and did not interfere with the activity of nitrate reduccase.
Measurement of mitochondrial respiration: Cell respiration was assessed by the mitochondrial-dependent reduction of 3-(4,5-dimethylthiazol-2-yl)bromide (MTT) to formazan.
Cells in 96-well plates were incubated at 37 0 C with MTT (0.2 mg/ml) for 1 hour. Culture medium was removed by aspiration and the cells were solubilized in DMSO (100 The extent of reduction of MTT to formazan within cells was quantitated by the measurement of
OD
550 All measurements were corrected for the interference of MnTBAP at this wavelength.
Measurement of peroxynitrite-induced oxidation of dihydrorhodamine 123: The peroxynitrite-dependent oxidation of dihydrorhodamine 123 to rhodamine 123, was measured based on the principles of the method described by Kooy et al, Free Radical Biol. Med. 16:149 (1994).
Briefly, peroxynitrite at 5 M was added into phosphate- 119 buffered saline containing 10 AM dihydrorhodamine 123, in the absence or presence of MnTBAP (3-100 MM) Afcer a 10 min incubation at 22°C, the fluorescence of rhodamine 123 was measured using a Perkin-Elmer fluorimeter (Model LS50B; Perkin-Elmer, Norwalk, CT) at an excitation wavelength of 500 mn, emission wavelength of 536 nm (slit widths 2.5 and 3.0 nm, respectively).
Measurement of NO-induced vascular relaxations: New Zealand white rabbits weighing 3-4 kg were anesthetized with pentobarbital (30 mg/kg). The descending thoracic aorta was isolated, removed, cleaned and placed in Krebs buffer (pH Vessels were cut into 5 mm rings and hung on stirrups connected to force transducers. Rings were suspended in 20 ml jacketed baths which were kept at 370C and bubbled with 95% 02 and 5% CO2. Rings were equilibrated with 2g resting tension for lh before use and contracted with phenylephrine. Saturated NO solution was prepared by bubbling compressed NO gas through a NaOH trap and then into anaerobic deionized water. Aliquots of the nitric oxide solutions (final concentration: 75-300 nM) were added to the rings (in the presence or absence of 100 fM MnTBAP) and relaxant responses were-recorded.
Data analysis: All values are expressed as mean standard error of the mean of n observations, where n represents the number of wells studied (12 wells from 2- 3 independent experiments). Data sets were examined by analysis of variance and individual group means were 120 then compared with Student's unpaired t-test. A p-value less than 0.05 was considered significant.
Results MnTBAP is not a scavenger of nitric oxide: MnTBAP did not inhibit the relaxations in vascular rings in response to authentic NO. Moreover, MnTBAP, at 300 pM, did not inhibit nitrite/nitrate accumulation in the culture medium in response to the NO donor compound SNAP and caused a slight inhibition in response to DNO.
These data, and the finding that NO does not affect the spectral changes of MnTBAP soret band, indicate that NO does not complex with the manganese in MnTBAP.
MnTBAP inhibits peroxynitrite-mediated oxidation: Peroxynitrite induced significant oxidation of dihydrorhodamine 123 to rhodamine 123, which was dosedependently inhibited by MnTBAP with 50% inhibition at AM (Fig. 40). This suggests that MnTBAP, like cysteine, urate, ascorbate and alpha-tocopherol, inhibits the oxidations caused by peroxynitrite.
MnTBAP inhibits the suppression of mitochondrial respiration by authentic peroxynitrite in J774 cells.
Mitochondrial respiration was profoundly inhibited by exposure to 1 mM peroxynitrite at 1h (Fig. 41A). This effect was partially and dose dependently prevented by MnTBAP.
MnTBAP inhibits the suppression of mitochondrial respiration by NO donors in J774 cells: Mitochondrial respiration was also inhibited by exposure to the NO donor compound SNAP (Fig. 41B) and DNO for 24h (Fig. 41C). This effect was partially and dose dependently prevented by MnTBAP.
Effects of MnTBAP on NO production and suppression of mitochondrial respiration in immunostimulated J774 macrophages: Immunostimulation of the cells by LO lipopolysaccharide (LPS; 10 pg/ml) alone, and, more pronouncedly, in the presence of gamma interferon (IFN; U/ml), resulted in nitrite/nitrate formation and a pronounced inhibition of the mitochondrial respirazion.
Administration of IFN alone did not induce detec:able nitrite/nitrate production and only caused a mild suppression of respiration (n=12).
MnTBAP caused a dose-dependent inhibition of nitrite/nitrate production in cells stimulated with LPS.
However, in cells immunostimulated with the combination !0 of LPS and IFN, MnTBAP caused a less pronounced inhibition of nitrite, nitrate accumulation (300 AM).
For instance, at 100 AM and 300 MM, MnTBAP caused a significant, 63 and 86% inhibition, of the LPS-induced nitrite/nitrate accumulation. When given in the combined presence of LPS and IFN, 100 AM MnTBAP only caused a 25% inhibition of nitrite/nitrate accumulation and pronounced inhibition was only observed at the highest concentration of MnTBAP tested (300 AM) L-NMA (NG-methyl-L-arginine) caused a near-complete inhibition 122 mv of the production of NO in cells stimulated with LPS or LPS and IFN; and MnTBAP had no additional effect on nitrite/nitrate formation in the presence of L-NMA. The inhibition of nitrite/nitrate accumulation by MnTBAP in LPS-stimulated macrcphages diminished by more than when the agent was applied 6h after LPS, whereas in the case of the inhibition seen with L-NMA, the extent of inhibition seen was similar when the compound was given together with LPS or at 6h thereafter MnTBAP caused a dose-dependent, partial restoration of the immunostimulation-induced suppression of the mitochondrial respiration in both LPS-treated and (less potently) LPS and IFN-treated cells. Inhibition of NOS with L-NMA caused a restoration of the respiration to an extent comparable with that of 300 pM MnTBAP. The combined administration of 300 pM MnTBAP and 3mM L-NMA caused an additional restoration of the mitochondrial respiration. In the presence of L-NMA and MnTBAP, respiration was restored to initial levels in the LPSstimulated cells, but remained below normal in cells stimulated with LPS and IFN.
Example XIV Synthetic Reaction Schemes Synthesis of 10303 (Fic. 42A) 1. Methyl 2-methoxy-3-methyl benzoate (1) To a magnetically stirred solution of 2-hydroxy-3methyl benzoic acid (2.22 g, 14.6 mmol), finely ground anhydrous K2CO3 (8.06 g, 58.3 mmol) and acetone (150 mL) was added (CH 3 0) 2
SO
2 (2.9 mL, 30.6 mmol). The solution was stirred at room temperature for 18 hours then heated at reflux until analysis by TLC indicated the reaction was complete (1-2 hours). The reaction mixture was cooled to room temperature, filtered and the excess K2CO 3 cake was thoroughly washed with acetone. The filtrate was evaporated and the residue was redissolved in EtOAc (100-125 mL) and criethylamine mL) was added. The reaction mixture was stirred at room temperature for 30 minutes, transferred to a separatory funnel and washed successively with H 2 0 (100 mL), 2N HC1 (until slightly acidic), H20 (100 mL) and brine (100 mL), then dried (Na 2
SCO
4 filtered and evaporated under reduced pressure. Chromatography of the residue on silica gel (ht of silica: 15 cm, diameter: 3 cm, eluent: 1:1 hexanes/Et20) provided 2.50 g of pure 2. Methyl 2-methoxy-4-(a,a-dibromomethyl) benzoate (2) A magnetically stirred solution of 1 (24.43 g, 135.6 mmol), NLS (54.41 g, 305.7 mmol) and CC14 (IL) was exposed to a 100 watt lamp for 6 hours. Analysis by TLC indicated that the reaction mixture was composed of the mono, 'di and tribrominated benzoates, wherein the dibromide was the major product. The reaction mixture was quenched with H20 (200 mL) then saturated Na 2
S
2 0 3 (500 mL) was added to destroy Br 2 The mixture was transferred to a separatory funnel, vigorously shaken then separated. The organic layer was dried (Na 2
SO
4 filtered and evaporated under reduced pressure. The crude 'H NMR spectrum indicated that methyl-2-methoxy-4- (a,a-dibromo-methyl) benzoate was the major product, and was used without further purification.
3. Methyl 4-formyi-2-methoxy-benzoate The crude product 2I was dissolved in acetone/O 2 0 (200 mL, 83:17) then AgNO 3 (47.25 g, 278.2 mmole) was added. The flask was covered with foil to avoid .0 decomposition of the AgNO 3 by light. The reaction mixture was szirred a: room temperature for 2-3 hours, then the AgBr salts were filtered off from the solution.
The filtrate was diluted with EtOAc (400 mL), transferred to a separatory funnel, then washed with saturated NaHCO 3 (300 mL) to extract acid away from the aldehyde. The organic layer was washed successively with H 2 0 (300 mL) and brine (300 mL), dried (Na 2
SO
4 filtered and evaporated under reduced pressure.
Chromatography of the residue on silica gel (ht of L silica: 15 cm, diameter: 5cm, eluent: 1:1 hexanes/Et 2 0) provided 11.92 g (42k) of pure aidehyde 4. Methyl 4-formyl-2-hydroxy benzoate (4) To a magnetically stirred solution of methyl 4formyl-2-methcxy benzoate (1.82 g, 9.37 mmol) in
CH
2 C1 2 (35 mL) at 0°C and under N 2 was added dropwise, 1M BC1 3 (18.7 mL, 18.7 mmole). The reaction mixture was stirred at 0°C for 10-30 minutes then quenched with H 2 0 (SO mL) Ether (100 mL) was added to the mixture then transferred to a separatory funnel and layered. The aqueous layer was extracted with ether (35 mL). The combined organic extracts were washed with brine (100 mL), dried (Na 2
SO
4 filtered, and evaporated under reduced pressure. Chromatography on silica gel (ht of silica: 12.5 cm, diameter: 5 cm, eluent: 1:l hexanes/Et 2 provided 1.60 g of pure methyl 4formyl-2-hydroxy benzoate as an oil.
Tetramethyl 2'''-tetrahydroxy-4,4', 4", 41''-(21H, 23H-porphine-5,10,15,20-terrayl)tetrabenzoate In a foil-covered, IL 3 neck round-bottomed flask equipped with a reflux condenser, magnetic stirrer and a
N
2 inlet was added methyl 4-formyl-2-hydroxy benzoate (1.01 g, 5.6 mmole), pyrrole (397 AL, 5.6 mmole) and dry CH2C1 2 (560 mL) The reaction mixture was stirred at room temperature for 10-30 minutes then B? 3 -OEt 2 (69 uL, 0.56 mmole) was added. The extent of reaction was followed by uv-vis spectrophotometry. After 1.5 hours at room temperature tetrachloro-1,4-benzoc-inone (1.03 g, 4.2 mmcle) was added and the reaction mixture was heated at reflux for 3.5 hours. The reaction mixture was allowed to cool to room temperature then the solvent was evaporated under reduced pressure. After 2 chromatographic purifications on silica gel, 0.38 g of was obtained as a violet solid.
126 General procedure for chromatographic purification of E.
On a 1 g scale of aldehyde, the crude product was usually combined with 2-3 g silica gel to make.a mixture which was then divided into 2 batches for 2 separate chromatographic purifications on silica gel (ht of silica: 12-15 cm, diameter 5 cm). The first eluent used was ether 1:1 hexanes/Et 2 0 or 4:1 hexanes/EtOAc (depending on how pcrphyrin behaves on TLC) to remove hydroquinone by-products and other non-polar impurities.
After all non-polar impurities were removed, the eluent was changed to either CH2C1 2 or CHC13 to elute the porphyrin.
6. Trimethyl 2" '-tetrahydroxy-4-benzoic acid-4', 41'' (porphine -5,10,15,20-tetrayl)tribenzoate manganese (III) chloride A solution of 5 (0.38 g, 0.42 mmole) and MnC12 (0.26 g, 2.1 mmole) in DMF (25 mL) was heated at reflux for 1.5 hours, then air was bubbled for 1.5 hrs as the solution was being cooled to room temperature. The DMF was evaporaced under reduced pressure, then the residue was combined with silica gel (1 g) and CH 2
CI
2 (10 mL) to make a slurry. The CH2C12 was evaporated leaving a solid mixture which was loaded dry into a column packed with silica gel (ht of column: 12.5 cm, diameter: 5 cm, eluent: 3% MeOH/CH 2 Ci 2 The preceeding purification yielded 124 mg of pure 6 as a dark green solid.
(mp >300OC; UV-VIS Xmax 4 6 7 nm (131,000); FAB-MS calculated for C,.H.
4 MnN.
4 C. 949, found .949) 127
I
Synthesis of 10204 (Ficr. 42R) 7. 23H-porphine-5,10,15,20-te:rayl) tetrakis(benzonitrile) in a foil-covered 2L 3-neck round-bottomed flask equipped with a reflux condenser, magnetic stirrer and a
N
2 inlet was added 4-formyl benzonitrile (1.13 g, 8.6 mmole), pyrrole (0.6 ml, 8.6 mmole) and CH 2 C1 2 (850 ml).
The reaction mixture was stirred for 15 minutes at RT then BF.-0Et 2 (105 ul, 0.85 mmole) was added. The extent .0 of reaction was followed by uv-vis spectrophotometry.
After 2 hours, tetrachloro-1,4-benzoquinone (1.56 g, 6.34 mmole) was added and the reaction mixture was heated at reflux for 2 hours. The reaction mixture was evaporated to a volume of 50-100 ml, then added to 2.8 g of silica gel. The rest of the solvent was evaporated under reduced pressure to provide a solid mixture for chromatographic purifications. After 3 senarate purifications, 700 mg of pure 7 was obtained as a powdery violet solid.
8. 1,1 1' -Tetra- (21:,23H-porphine-5,10,15,20-tetravl)tetrakisbenzene A solution of 2 (0.30 g, 0.42 mmole), NaN3 (0.24 g, 3.69 mmole), NH4C1 (0.18 g, 3.37 mmole) and DMF (25 ml) was heated at 1200°C for 3 days. Additional NaN, (0.17 g, 2.59 mmole) and NE 4 Cl (0.11 g, 2.05 mmole) were added in portions during day 2 to drive the reaction to completion. The DMF was evaporated under reduced 128 pressure then cold H 2 0 (10 ml) was added. The resulting solution was acidified with 6N HC1 then CH 2 Cl, (10 ml) was added. The resulting precipitate was collected and dried under vacuum to provide 0.37 g of 2 as a greenish solid which was used without further purification.
9. 1,1',l",l1 '-Tetra-(iH-tezrazol-5-yl)-4,4',4",4' (porphine-5,10,15,20-tetrayl) -tetrakisbenzene manganese (III) chloride .0 A solution of Z (0.35 g, 0.4 mmole) and MnCl 2 (0.25 g, 2 mmole) in DMF (20 mL) was heated at reflux for 4-5 hours. The reaction mixture was allowed to cool to room temperature then DMF was evaporated under reduced pressure. The crude product was adsorbed onto 1.5 g of silica gel with 10-15 mL CH2CI 2 The C-2C1 2 was evaporated leaving a solid which was loaded dry into a column packed with silica gel slurry (ht of silica: 12.5 cm, diameter: 5 cm, eiuen:: 6 CHC1 2 /3 MeOH/1 Chromatographic purification provided 0.19 g 0 of pure 2 as a dark green solid. (mp >320°C; UV- VIS Xma x 468 nm (42,000); FA3-MS calculated for
C
4
H,
2 MnN 20 939, found 939) Synthesis of 10305 (Fic. 42C) Methyl 3-methoxy-4-methyl benzoate (O0).
To a magnetically stirred solution of 3-hydroxy-4methyl benzoic acid (15.1 g, 99.2 mmole), finely ground anhydrous K 2
CO
3 (43.7 g, 317 mmole) and acetone (250 mL) 129 was added (MeO) 2
SO
2 (21 mL, 222 mmole). The reaction mixture was stirred at room temperature for 24 hours then excess K 2
CO
3 was filtered off. The acetone was removed by evaporation and the residue was redissolved in EtOAc (250 mL) then Et 3 N (15 mL) was added. The solution was stirred at.room temperature for 30 minutes, transferred to a separacory funnel and washed successively with H 2 0 (100 mL), IN HC1 (until slightly acidic), NaHCO 3 (100 mL), H 2 0 (100 mL) and brine (100 mL), dried (Na 2
SO
4 filtered and evaporated under reduced pressure. Chromatography of the residue on silica gel (ht of silica gel: 25 cm, diameter: 4 cm, eluent: 1:1 CH 2 C12/hexanes) provided 16.84 g of pure 11. Methyl 4-formyl-3-methoxy benzoate (jL).
To a magnetically stirred solution of 12 (5.87 g, 32.6 mmol) in 1:1 C-3CN/H 2 0 (250 mL) was added consecutively CuSO 4
-SH
2 0 (8.13 g, 32.6 mmol) and K 2
S
2 0 8 (26.4 g, 97.7 mmole). The reaction mixture was heated 0 at reflux for 50 minutes then transferred to a separatory funnel and extracted with EtOAc (2 x 100 mL).
The combined organic extracts were dried over Na 2
SO
4 filtered and evaporated. Purification on silica gel by gravity chromatography (ht of silica gel: 33 cm, diameter: 4 cm, eluent: 5:1 hexanes/EtOAc then 4:1 hexanes/EtOAc) provided 1.47 g of pure 11.
12. Tetramethyl 3, 3'''-tetramethoxy-4, 4,4", 4' 23 H-porphine-5,10,15,20-tetrayl)tetrabenzoate (12).
In a foil-covered 2L 3 neck round-bottomed flask equipped with a reflux condenser, magnetic stirrer and a
N
2 inlet was added methyl 4-formyl-3-methoxy benzoate (11) (1.23 g, 6.3 mmole), pyrrole (448 4l, 6.3 mmole) and dry CH 2 C1 2 (630 mL) The reaction mixture was stirred for 15 minutes then BF 3 -OEt (78 pL, 0.63 mmole) 0 was added. The reaction was monitored by uv-vis spectrophotometry. After 3 hours, tetrachloro-1,4benzoquinone (1.17 g, 4.7 mmole) was added and the reaction mixture was heated at reflux for 2.5 hours.
The reaction mixture was allowed to cool to room temperature then the solvent was evaporated in vacuo.
The crude product was added onto 2.6 g silica gel with 10-15 mL, CH 2
CI
2 The CH 2 C1 2 was evaporated and the resulting mixture was divided into 3 equal oorcions for three separate chromotographic purifications. Each portion was separately loaded dry into a column oacked with silica gel (ht of silica gel: 15 cm, diamecer: cm, eluent: 1:1 hexanes/Et 2 0 then CH2C1 2 The combined product 0.461 g of pure 12 was obtained as a mixture of rocational isomers.
13. Tetramethyl 3, '-tetramethoxy-4,4',4", 4 (porphine-5,10,15,20-tetrayl)-tetrabenzoate manganese (III) chloride (1a) A solution of 12 (0.32 g, 0.33 mmole) and MnCl 2 (0.20 g, 1.56 mmole) in DMF (32 mL) was heated at reflux for 6.5 hours. Air was then bubbled into the solution for 1.25 hours as the reaction mixture cooled to room temperature. The DMF was evaporated off and the crude mixture was adsorbed onto 1.5 g silica gel with 10-15 mL -0 CH2C1 2 The CH 2 C1 2 was evaporated and the resulting solid mixture was loaded dry into a column packed with silica gel (ht of silica: 12.5 cm, diameter: 5 cm, eluent: 5% MeOH in CH 2 Cl 2 Chromatographic purification provided 294 mg of pure 13 as a green-black solid. (mp >3000°C; UV-VIS max 467 nm (145,000) FAB-MS calculated for C 5 H1,MnNO 2 1019, found 1019).
14. 3,3' '-Tetramethcxy-4,4',4",41 ''-(pcrphine- 5,10,15,20-tetray) tetrakis(benzoic acid) (14).
.0 A magnetically stirred solution of manganated tetrabenzoate 13 in 10 mL Claisen's case (KOH/MeOH/H 2 0) was heated at reflux for 40-60 minutes. The reaction was cooled to room temperature then to 000C. The cold solution was then acidified with 6N HC1 then MeOH was evaporated under reduced pressure. The solids were filtered away from the acidic solution and was washed thoroughly with cold water. The solids were collected and dried under vacuum at 800°C overnight provide 96 mg (87k) of pure 14 as a dark green solid (mpo >300°C; uv- 132 vis; Amax 467 nm (150,000); FAB-MS calculated for Cs 2 EaGMnNIOu 963, found 963).
Synthesis of 10109 (Fia. 42D) 4 (Porphine-5,10,15,20-tetrayl) tecrakis(benzoylchloride) manganese (III) chloride Mn TBAP (500 mg, 0.57 mmole) was suspended in dry benzene (125 ml) in a 3-neck round bottom flask equipped with a reflux condenser and a N. inlet. Thionylchloride ml; 617 mmol) was added to the mixture and the reaction mixture was heated a: reflux for 22 hours. The benzene and the unreacted SOC12 was removed by distillation. The residue was washed with dry benzene ml) and evaporated on rotary evaporator to dryness.
This washing and evaporating was repeated twice. The dark green residue was dried under high vacuum to yield 0.54 g (100%).
16. Tetra(N,N-dimethyl) 4 4 4 ",4'''-(porphine- 5,10,15,20-tetrayl)-tetrakis(benzamide) manganese (III) chloride (1i) To compound 15 (340 mg, 0.36 mmole) in THF (20 mL) was added dimethylamine in THEF solution (10 ml, 2M solution). The reaction mixture was refluxed for 6 hours. TLC analysis (eluent: 1 methanol:3 chloroform) showed complete conversion of the acid-chloride to the product. After cooling to room temperature, deionized water (100 ml) was added to the reaction mixture and the solid was filtered off. The crude solid was purified by 133 column chromatography on silica gel (methylene chloride:merhanol 4: 1 eluent). The combined fractions (Rf 0.75 in CH2Cl:MeOH 4:1) were evaporated, and dried under high vacuum, to provide the product as a green solid 0.11 g (UV-VIS max 467 nm.
FAB-MS calculated for
C
56 H4 9 MnNsO 4 952, found 952.) e o f 1 2 106 2 11 00 1 00 an d ii Insofar as compounds 10402, 10602, 11001, 11002, 11003, and 11103 are concerned, synthesis can be carried out essentially, as described in Example XIV sections and 6; the following starting material substitutions are required: 4 -hydroxybenzaidehyde for methyl 4 -or-my-2hydroxy benzoate in the case of 10402, 4 -hydroxy-3nitrobenzaldehyde for methyl 4 -formyl-2-hydroxy benzoate in the case of 10602, indole-3-carboxaldehyde for methyl 4-formy-2-hydroxy benzoate in the case of 11001, 4quinolinecarboxaidehyde for methyl 4 -formyl-2-hydroxy benzoate in the case of 11002, 3-quincinecarboxaldehyde for metuhyl 4icco dey L. m- -2-hydroxy benzoate in the case of 0 11003 and 3-fluro-4-methoxybenzaldehyde for methyl 4-formyl-2-hydroxy benzoate in the case of 11103.
Synrhesis of 10207 and 025 Acetonitrile is reacted with hydroxylamine to prepare the amide oxime. An excess of the amide oxime is allowed to react with the product described in Example XIV, section 15, to provide a mixture of 10207 134 and 10208. Chromatographic purification of the reaction mixture provides pure 10207 and 10208.
Synthesis of 10209 Commercially available 4-methylbenzoic acid is transformed into its acid chloride by reaction with oxalyl chloride or thionyl chloride. The resulting acid chloride is heated in the presence of cuprous cyanide to provide the a-ketonitrile derivative. The a-ketonitrile is hydrolyzed with anhydrous methanol saturated with .0 anhydrous hydrogen chloride gas to give the a-ketonitrile ester derivative. The aromatic methyl group is efficiently oxidized to the aldehyde oxidation state by first reaction with N-bromosuccinimide and subsequent hydrolysis with silver nitrate. Synthesis of 10209 can be carried out essentially as described in Example XIV, sections 5 and 6; the substitution of starting material of methyl 4-formyl-2-hydroxy benzoate with the aldehyde described hereinabove being reouired.
J All documents cited above are hereby incorporated in their entirety by reference.
One skilled in the art will appreciate from a reading of this disclosure that various changes in form and detail can be made withouc departing from the true scope of the invention. In addition to the compounds described herein, compounds disclosed in the following 135 M references can also be used as oxidant scavengers consistent with the invention: USP 5,227,405; Nagele et al, Biochem. Pharmacol. (UK) 47:555-562 (1994); Baudry et al, Biochem. Biophys. Res. Comm. 192:964-968 (1993); Duran et al, Cancer Lett (IRE) 69:167-172 (1993); Itami et al, Biochem. Biophys. Res. Comm. 197:536-541 (1993); Kitajima et al, Inorg. Chem. 32:1879-1880 (1993); Riley et al, Free Radical Biol. Med. 15:514 (1993); Weiss et al, J. Biol Chem. (US) 268:23049-23054 (1993); Foye, Ann. Pharmacother. 26:1144-1147 (1992); Haseloff et al, J. Biolumin. Chemilumin. (UK) 7:171-175 (1992) Pelletier, Biochem. Pharmacol. 43:1061-1066 (1992); Yaping et al, J. Free Radic. BiolMed 13:533-541 (1992); Schechinger et al, Biol. Met. 1:112 (1988); Linss et al, Inorc. Chim. Acta 125:117 (1986); and Weser et al, Adv.
Exp. Med. Biol. 264:51 (1990).
136 Footnote to Table IX 'Porphine carbon numbers refer to the methine bridge carbons 10, 15, 20) at which each R group is attached, or to the pyrrole carbon 4, 7, 8, 12, 13, 17 or 18), at which P is attached. P is hydrogen unless otherwise defined.
2 SOD activity determined as described by McCord and Fridovich, J. Biol. Chem. 244:6049 (1969).
3 Catalase activity determined as described by -Del Rio et al, Anal. Biochem. 80:409 (1977). Metalloporphyrins were assayed at various concentrations in the presence of 1 mM hydrogen peroxide for catalase-like activity. A Clark electrode was used to measure the formation of oxygen from the breakdown of hydrogen peroxide over a period of 2 minuces.
4 Rates of growth of cultures of null SOD E. coli (JIi32 scrain) were followed turbidimetrically at 700 nm to minimize interferences from the absorbance of test compounds. Growth medium contained 0.2% glucose, 0.2% casamino acids, 30 mg/L thiamine, 30 mg/L pantotheni.c acid, and M9 salts in water, pH adjusted to 7.0. Test compounds were filter-sterilized before adding to media.
(Faulkner et al, J. Biol. Chem. 269:23471 (1994)).
137 sHuman umbilicord vein endothelial cells (HUVEC) (ATCC #CRL-1730) (Moldow et Methods in Enzymology 105:378- 385 (1984)) were grown in Ham's F-12K medium supplemented with 10% fetal bovine serum, 0.1 mc heparin and 0.03 mg/ml endothelial cell growth factor in culture flasks in a humidified atmosphere containing carbon dioxide and 95% air. Cells were plated at x 101 cells/well in 24-well plates. Experiments were performed 48 hours later. Medium was changed to 0 EMEM without serum supplement but including heparin and endothelial cell growth factor at the start of each study. Stock solutions of D-aminoacid oxidase and metalloporphyrins were diluted into (EMEM) and passed through a 0.2cc filter before use. HUVEC cells were 3 labelled with Cr" 5 by incubating them with EMEM containing radiolabeled sodium chromate for 4 hrs ac an isotope concentration of 20Ci/10 cells.
Unincorporated Cr 5 was removed by repeated washes in EMEM. Injury was initiated by incubation of endcthelial cells for 5 hours with a hydrogen peroxide generating system consisting of D-alanine (1 mM) and 40 mU/ml of D-aminoacid oxidase in EMEM. D-amino acid oxidase is found in PMNs (polymorphonuclear leucocytes) and is thought to be part of the oxidative burst which is a key part of the PMN mediated inflammatory response (Robinson et J. Cell Biology, 77:59 (1978); Cline et al, Microbiology 62:756 (1969); De Chatelet J. Reticul. Soc.
24:73 (1978)). Supernatanc from each well was collected ac the end of the incubation period, cells were washed with PBS and then lysed with 0.4 N sodium hydroxide.
138 Radioactivity in both supernatants and cell lysates were measured separately in a gamma counter. A set of wells in each plate were used to measure basal release of Cr 51 from HUVEC incubated in EMEM alone. Cr sl release was 3 expressed as a percent of radioactivity in supern.aant to total radioactivity [(Cr s 5 in supernatant+ CR 5 in lysate) x 100].
139 ACTIVITY (IF COMPOUNDS Compound (metal complexes) (Mn) 10101 (Mn) 10104 10105 (Mn) Porphyrin Carbon number (a) 5,10,15, 20 5,10, 15,20 5,10,15 5,20 10,1iS
RI
(or R0) (or P,) 00 R2 (or R") TABLE IX R3 aide (or RI group o r Rcila s sa Nil, cationic Biochemistry SOD' Catalasel pg/USOD min-' 213 3.74 -In Vitro Hull q(OD' bioass_ j I1, HUVEjZC' r 1" I I O ?CNH 0 CO\/I 0 9NH1 Bond Bond -W(II 10 -Ni CMI)I
COOH-
cationic cat ion ic 349 445 N. D.
1. 11 N. D.
N. D.
I t I I- I 10106 (Mn)
COOH
(Cll,).
cationic 140 0.53 1. 1 1 1 Compound (metal complexes) 10107 (Mn) 10108 (Mn) (Mn) 10109 (Mn) 10201 (M11) 10202 (Mn) .10203 -(Mn) 10204 Porphyrin Carbon number (s) 15 10,20 5 10,15,20 5110,15,20 5%110,15,20 (or P 1 0 R2 (orR,) Bond -(C1[2)2 Bond Bond Bond R3 (orH)
(CHO,
-W(CM,)
1 -Cool! -N wCM,), coolI side group class cationic cationic ca tionic Biochemistry SOD 2 Catalase' F9/USOD mirr'1 172 1.69 In Vitro Hull SO C l l i H bioassay
RVC
375 470 0.88 0.59 anionic 33 0.8 I I I I I I I L 5, 10, 15,20 I cq'c, Bond if I anionic 10, 15, 20 HIond Bond -so, ani -i anionic inactive 0.33 I I Compound Porphyrin (metal Carbon complexes) number (s) (Mn) 10205 (Mn) 10206 10207 (Mn) 5,10,15,20 5,10.15,20
RI
(o r R~ (or Pj
A-
R2 (or
R
1 Bond Bond 10,20 Biochemis try 10208 (Mn) (Mn) 10209 (Mn-) 10301i (Mn) 10302 5,15 20 5,10,15,20 5,10,15,20 s5io,,15,20 Bon d Bond Bond 03 (or Rd 0 0 011
OH
-OCI')
anionic aide group class
SOD'
Pg/USOn 28 anionic Iinactive I inactive Catalano, 1.8 anionic I- Bond 00o Bond Bond anionic anionic chelator Chelator inactive N.
D.
in Vitro Mull SOD 4 E. Coli bioassay HIJVKC' inactive 314 0.95 37 2.9 I T T Compound (metal complexes) Porphyrin Carbon number (s) Ri (or R') (or P,) R2 (or R,) R3 (or R 1 side group class Diochemis try SOD' Catalase' pg/USOD min-' In Vitro Null BODS H. Col
HI
bioassay
HUVEC'
-tI-I I 10303 (Mn) _q co'
I_
AR
Bond Bond chelator 10,15,20 I 1 1 1 1 I (Mn) 10304 10305- (Mn) 10306 5,10,15,20 5,10, 15,20 5, 10, 15,20 CR- colc..
OP -CV11, Bond Bond Bond chelator ppt
-OCM
3 00% chelator chelator inactive 1 1 I I 1* I I (Mn) 10401 (Mn) 10402 5,10,15,20 5, 10, 15, 20 C(Cetd, 0q Bond antioxidant Insol insol N. D.
N. D.
1 I I 1- I .4 Bond antioxidant inactive I t t 1 4 1 1 (Mn) 10601 (Mn) 10602 5,10, 15,20 O Cu, Biond NO, nitrio 1 1 t F 4 1 1 5,10,15,20 Oa3-1 Bond NO, n i tro 1070 1 0,15,2 Bond i n 96 2.1 (Mn01 {51<1~ 0 i subse it Compound (metal complexes) (Mn) 10901 (Mn] 11001 (Mn) 11002 (Mn) 11003 11101
(CU)
Porphyrin Carbon number Cs) 5,10,15,20 5,10,15,20 5,10,15,20 5,10,15,20 5,10,15,20 2,31,7,0, 12, 13, 17, 18 5,10,15,20 2,3, 7,8, 12; 13, 17, .18 5,10, L5, 20 Ri (or (or or P) (R2 1% Q Bond R 3 Bond IBiochemistry Bond Bond Bond Bond Bond Q105 Bond -Br O N0 Bond -Br 3 O ri 3011(
_CM
Sll -cP, side group class c yano heterocyclic heterocyclic heterocyclic halogena ted
SOD
1 pg/JSOD 169 404 min"' 3.2 0.9 22 106 1 .02 inactive In Vitro =Null SOD, b Ioa HVC'
N.D.
N. D.
N. D.
N. D. inactive N.
D.
1I10 2 (Mn) halIogenated (M11) 11103 4.6 2.A N.
D.
N.
D.
halogena ted N.
D.
N.
D.
P
PT

Claims (6)

1. A method of treating a pathological condition of a patient resulting from peroxynitrite accumulation comprising administering to said patient an effective amount of a compound having the activity of a catalytic antioxidant under conditions such that said treatment is effected.
2. A method of treating a disorder resultihg from aberrant smooth muscle function in a patient in need of such treatment comprising administering to said patient an effective amount of a mimetic of SOD, EC-SOD, catalase or peroxidase under conditions such that said treatment is effected.
3. A method of modulating NO- function as a neurotransmitter in a mammal comprising contacting said mammal with an amount of an oxidant scavenger of the formula, R i' R2 R i 20 R j- R2 R3 N N N N H R3 R 2 R R- R2 R3 or a pharmaceutically acceptable salt thereof, wherein: R is a bond, N" N O 0, SO 3 H, x or Y wherein X is a halogen and Y is an alkyl group and wherein indicates bonding to R 2 at any position and indicates bonding to R 2 and the substituent at any position; and R 2 is a bond, -(CY'2)n -(CY 2-CY n, (CY'2-CY'2-CH=CH)n-, or -(CY' 2 wherein Y' is hydrogen or an alkyl group and wherein n is 1 to 8; and R 3 is -OH, -NH 2 -COOH, -COO-, SO 3 H, -SO3-, -C-PO0 3 H 2 or -C-PO3H-, wherein Y" is an alkyl group, optionally complexed with a metal selected from the group consisting of manganese, copper and iron, sufficient to effect said modulation.
An oxidant scavenger of the formula: R3- R2- or a pharmaceutically acceptable salt thereof, wherein: R, i s a bond, 7, NO, 2 e 3s 0 x 7e or Y wherein X is a halogen and Y is an alkyl group and wherein indicates bonding to R 2 at any position and indicates bonding to R 2 and the substituent at any position: and each R2 is independently, a bond, (CY' 2 -(CY' 2 -CY' 2 -CH=CH)n-, or (CY' 2 wherein Y' is hydrogen or an alkyl group and wherein n is 1 to 8; and each R3 is, independently -OH, -NH 2 -COOH, -COO, -S0 3 H, -S03-, -C-PO3H 2 or -C-PO3H-, wherein Y" is an alkyl group, each P is, independently, an electron withdrawing group or hydrogen wherein at least one P is not hydrogen, or P P R- R2- R R R 2 R3 N P H P N N P H P N, R3 R- R- R R3 P P or a pharmaceutically acceptable salt thereof, wherein each R 1 is independently a bond, wherein Y" is an alkyl group, and wherein indicates bonding to R2' at any position and indicates bonding to R 2 and the Ri' phenyl substituent at any position; each R 2 is independently a bond, or -(CHz)n- wherein n is 1-4, each R 3 is independently -OH, OY", -N02, -CN, -NH 2 -COOH, -COY", -COO-, or a heterocyclic group, wherein Y" is as defined above and is a primary, secondary, tertiary or quaternary amine, each P is independently, an electron withdrawing group or hydrogen wherein at least one P is not hydrogen; wherein said oxidant scavenger is a metal complex wherein said metal is selected from the group consisting of manganese, copper or iron, wherein when said metal is copper and all P's are Br, and when RI is -NL and R 2 is a bond, R 3 is not -CH 3
5. The oxidant scavenger according to claim 4 wherein said scavenger is a manganese, copper or iron complex of octobromo octochloro octofluro octonitro octobromo octochloro octofluro octonitro octobromo octochloro octofluro octonitro TBAP TBAP TBAP TBAP TMPyP TMPyP TMPyP TMPyP tetrakis 4-phenyl sulfonate porphyrin tetrakis 4-phenyl sulfonate porphyrin tetrakis 4-phenyl sulfonate porphyrin tetrakis 4-phenyl sulfonate porphyrin
6. A method of protecting a biological substance from oxidative stress or a method of maintaining biological function of NO-, or active forms thereof, in an intracellular or extracellular space of a tissue or organ comprising contacting said substance or said tissue 148 or organ with an amount of said scavenger according to claim 10 sufficient to effect said protection or said maintenance. DATED this 2 0 t h day of April 2004 DUKE UNIVERSITY UNIVERSITY OF ALABAMA AT BIRMINGHAM RESEARCH FOUNDATION By their Patent Attorneys CULLEN CO.
AU2004201624A 1995-06-07 2004-04-20 Oxidant Scavengers Abandoned AU2004201624A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US08/476866 1995-06-07
US08/613418 1996-03-11
AU63870/96A AU725602B2 (en) 1995-06-07 1996-06-07 Oxidant scavengers
AU53511/00A AU769217B2 (en) 1995-06-07 2000-08-21 Oxidant scavengers

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU53511/00A Division AU769217B2 (en) 1995-06-07 2000-08-21 Oxidant scavengers

Publications (1)

Publication Number Publication Date
AU2004201624A1 true AU2004201624A1 (en) 2004-05-13

Family

ID=34275174

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2004201624A Abandoned AU2004201624A1 (en) 1995-06-07 2004-04-20 Oxidant Scavengers

Country Status (1)

Country Link
AU (1) AU2004201624A1 (en)

Similar Documents

Publication Publication Date Title
US6583132B1 (en) Oxidant scavengers
AU702596B2 (en) Superoxide dismutase and mimetics thereof
AU725602B2 (en) Oxidant scavengers
US5994339A (en) Oxidant scavengers
Bonetta Potential Therapeutic Applications of MnSODs and SOD‐Mimetics
Sheng et al. Effects of metalloporphyrin catalytic antioxidants in experimental brain ischemia
JP5312404B2 (en) Substituted porphyrins
Duarte et al. NRF2 and hypoxia-inducible factors: key players in the redox control of systemic iron homeostasis
JPH10500671A (en) Method of using peroxynitrite decomposition catalyst and pharmaceutical composition therefor
US6605619B1 (en) Nitroxides as protectors against oxidatives stress
Subramanian et al. Mitochondrially targeted antioxidants for the treatment of cardiovascular diseases
Valerio Jr Mammalian iron metabolism
AU769217B2 (en) Oxidant scavengers
Moore The biochemistry of heme synthesis in porphyria and in the porphyrinurias
AU2004201624A1 (en) Oxidant Scavengers
CA2614621A1 (en) Superoxide dismutase and mimetics thereof
ES2378896T3 (en) Combination of drugs containing Probucol and a tetrazolyl alkoxy dihydrocarbostiryl derivative with superoxide inhibitory effects
US7989420B2 (en) Smooth muscle relaxation
Ward et al. Iron and its role in neurodegenerative diseases
Shopova et al. Ffects Of Mntnhex-2-Pyp On Markers Of Inflammation And Lipid Peroxidation In Asthma Mice Model
Chakraborty Insights into iron trafficking and handling in eukaryotic cells

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application