throbber
Identification of the enzymatic mechanism of
`nitroglycerin bioactivation
`
`Zhiqiang Chen*†, Jian Zhang†, and Jonathan S. Stamler*†‡§
`
`*Howard Hughes Medical Institute, Departments of †Medicine and §Biochemistry, Duke University Medical Center, Durham, NC 27710
`
`Communicated by Irwin Fridovich, Duke University Medical Center, Durham, NC, April 15, 2002 (received for review March 26, 2002)
`
`Nitroglycerin (glyceryl trinitrate, GTN), originally manufactured by
`Alfred Nobel, has been used to treat angina and heart failure for
`over 130 years. However, the molecular mechanism of GTN bio-
`transformation has remained a mystery and it is not well under-
`stood why ‘‘tolerance’’ (i.e., loss of clinical efficacy) manifests over
`time. Here we purify a nitrate reductase that specifically catalyzes
`the formation of 1,2-glyceryl dinitrate and nitrite from GTN, lead-
`ing to production of cGMP and relaxation of vascular smooth
`muscle both in vitro and in vivo, and we identify it as mitochondrial
`aldehyde dehydrogenase (mtALDH). We also show that mtALDH is
`inhibited in blood vessels made tolerant by GTN. These results
`demonstrate that the biotransformation of GTN occurs predomi-
`nantly in mitochondria through a novel reductase action of
`mtALDH and suggest that nitrite is an obligate intermediate in
`generation of NO bioactivity. The data also indicate that attenu-
`ated biotransformation of GTN by mtALDH underlies the induction
`of nitrate tolerance. More generally, our studies provide new
`insights into subcellular processing of NO metabolites and suggest
`new approaches to generating NO bioactivity and overcoming
`nitrate tolerance.
`
`It is generally assumed that glyceryl trinitrate (GTN) is con-
`
`verted in vascular smooth muscle cells to NO or an NO
`congener (S-nitrosothiol, SNO), which activates guanylate cy-
`clase and thus relaxes vascular smooth muscle (1, 2). However,
`the molecular mechanism involved is not known, and there is no
`unifying explanation for the pharmacological profile of the drug
`(e.g., the preferential effects on capacitance vessels and sulfhy-
`dryl requirement for relaxation) or the phenomenon of nitrate
`tolerance (i.e., the loss of clinical efficacy over time), which
`leaves patients vulnerable to ischemia and may contribute to
`other deleterious consequences of long-term nitrate use (3, 4).
`GTN biotransformation is tissue and cell specific and dose
`dependent, and it yields 1,2-glyceryl dinitrate (1,2-GDN), 1,3-
`GDN, inorganic nitrite, and NO (or SNO) in differing amounts
`and ratios. In vascular smooth muscle cells, 1,2-GDN is the
`predominant dinitrate metabolite (2, 5, 6) and the vasorelax-
`ation-related formation of 1,2-GDN is diminished in GTN-
`tolerant blood vessels (7, 8). These data suggest that an enzyme
`with product specificity for 1,2-GDN (over 1,3-GDN) generates
`NO bioactivity from GTN and that loss of this activity at least
`partly accounts for tolerance. Several enzymatic mechanisms of
`vascular smooth muscle relaxation by GTN have been proposed
`(9 –15), and candidate enzymes include glutathione S-
`transferases, cytochrome P450 reductase, cytochrome P450,
`xanthine oxidoreductase, and an unidentified microsomal pro-
`tein. But none of these enzymes catalyzes the selective formation
`of 1,2-GDN (12, 15, 16) or is inhibited in tolerant vessels, and the
`roles of these enzymes in generating NO bioactivity remain
`controversial. Here we report on the purification of a GTN
`reductase that specifically generates 1,2-GDN, identify it as
`mitochondrial aldehyde dehydrogenase (mtALDH), and estab-
`lish its central role in GTN bioactivation, vasorelaxation, and
`tolerance.
`
`Materials Methods
`Chemicals and Drugs. GTN, 1,2-GDN, and 1,3-GDN standards
`were bought from Radian International (Austin, TX). GTN was
`
`synthesized according to ref. 17 and purified by TLC. 1,2-GDN
`and 1,3-GDN was prepared by hydrolysis of GTN and purified
`by TLC (18). [2-14C] GTN (55 mCi兾mmol) was obtained from
`American Radiolabeled Chemicals (St. Louis). Disulfiram, chlo-
`ral hydrate, cyanamide, acetaldehyde, phenylephrine, sodium
`nitroprusside (SNP), Q-Sepharose, DEAE-cellulose, and butyl-
`Sepharose were obtained from Sigma. Hydroxyapatite and
`protease inhibitor (mixture set III) were from CalBiochem.
`The cGMP assay (125I) kit was purchased from Amersham
`Pharmacia.
`
`Enzyme Purification. Mouse RAW264.7 cells (50-g pellet; ⬇1010
`cells) were disrupted by sonication in 30 mM phosphate buffer
`(KPi), pH 7.5 containing 1 mM DTT, 0.5 mM EDTA, and
`protease inhibitors (1:200 dilution). The 100,000-g supernatant
`was loaded onto a DEAE-cellulose column, and the flow-
`through fractions, which contained the enzyme activity, were
`pooled and concentrated by ultrafiltration (Amicon, 10-kDa
`cut-off membrane). After 3-fold dilution with cold water, the
`fractions were loaded onto a Q-Sepharose column, and the
`activity was eluted by a phosphate gradient (10 mM to 150 mM,
`pH 7.5); the active fractions were pooled and solid potassium
`chloride (KCl) was added to 1.25 M. The sample was then loaded
`onto a butyl-Sepharose column, and the enzyme activity was
`eluted by decreasing the salt concentration. The concentrated
`active fractions were then diluted and loaded onto a hydroxy-
`apatite column. After washing the column with 10 mM KPi兾0.4
`M KCl, the enzyme was eluted by a phosphate gradient from 10
`mM KPi to 150 mM KPi, pH 7.5. All steps were performed at
`4°C, and the elution buffers contained 1 mM DTT and 0.5 mM
`EDTA unless otherwise specified.
`
`GTN Biotransformation. 1,2-GDN and 1,3-GDN formation were
`measured by the TLC and liquid scintillation spectrometry
`method as described by Brien et al. (19) with a few modifications.
`During protein purification, the assay mixture (1 ml) contained:
`100 mM KPi (pH 7.5), 0.5 mM EDTA, 1 mM NADH, 1 mM
`NADPH, 0.1 or 1 ␮M GTN, and protein (with DTT) from
`column fractions. After incubation at 37°C for 10–30 min, the
`reaction was stopped (on dry ice or 4°C) and GTN and its
`metabolites were extracted with 3 ⫻ 4 ml ether and pooled, and
`the solvent was evaporated by a stream of nitrogen. The final
`volume was kept to less than 100 ␮l in ethanol for subsequent
`TLC separation and scintillation counting. For the activation of
`dialyzed enzyme samples and assay of the activity of purified
`enzyme, the assay mixture contained: 100 mM KPi (pH 7.5), 0.5
`mM EDTA, 0–1 mM DTT or other reductants (0.5 mM DTT for
`the bovine enzyme), and 1 ␮M GTN. For assays in tissues such
`as rabbit aorta, rings were blotted and weighed after sitting for
`1 h in Krebs solution (control) (composition described below).
`When used, inhibitors were added for 20 min before the addition
`
`Abbreviations: GTN, glyceryl trinitrate; GDN, glyceryl dinitrate; mtALDH, mitochondrial
`aldehyde dehydrogenase; SNO, S-nitrosothiol; SNP, sodium nitroprusside.
`
`See commentary on page 7816.
`‡To whom reprint requests should be addressed. E-mail: STAML001@mc.duke.edu.
`
`8306 – 8311 兩 PNAS 兩
`
`June 11, 2002 兩 vol. 99 兩 no. 12
`
`www.pnas.org兾cgi兾doi兾10.1073兾pnas.122225199
`
`Page 1 of 6
`
` Insys Exhibit 2027
`CFAD v. Insys
`IPR2015-01800
`
`

`
`Table 1. Purification of GTN reductase from mouse macrophage RAW264.7 cells
`
`Procedures
`
`100,000 g supernatant
`DEAE-cellulose
`Q-Sepharose
`Butyl-Sepharose
`Hydroxyapatite
`
`Total units,
`nmol兾h
`
`Total protein,
`mg
`
`Specific activity,
`nmol兾h per mg
`
`837.4
`756.0
`528.5
`205.4
`120.9
`
`2,265
`345
`123
`10.9
`3.6
`
`0.37
`2.19
`4.29
`18.81
`33.36
`
`Yield,
`%
`
`100
`90.3
`63.1
`24.5
`14.4
`
`Fold
`purification
`
`1.0
`5.9
`11.6
`50.8
`90.2
`
`Starting material: 50-g cell pellet.
`
`MEDICALSCIENCES
`
`of 1 ␮M GTN; the mixture (1 ml) was then kept at 37°C for 5 min.
`The extraction and TLC-liquid scintillation spectrometry anal-
`ysis were as described above. Buffer control (Krebs buffer plus
`GTN) and nonspecific biotransformation (heat-inactivated rings
`plus GTN) activities were also measured, and the results were
`corrected accordingly.
`
`Aortic Ring Bioassays. New Zealand White rabbits (2.5–3 kg) were
`killed by carbon dioxide inhalation. Thoracic aorta were re-
`moved, cleaned of fat and connecting tissue, and cut into 3-mm
`rings. The rings were mounted under 2 g of resting tension in
`tissue baths (25 ml) filled with Krebs solution (37°C) containing:
`118 mM NaCl, 4.8 mM KCl, 1.2 mM MgSO4, 1.2 mM KH2PO4,
`2.5 mM CaCl2, 25 mM NaHCO3, and 11 mM glucose, pH 7.4.
`The solution was bubbled with 20% O2, 5% CO2, and balance N2.
`Changes in isometric tension were recorded with Statham (Hato
`Rey, PR) transducers and a Grass Instruments (Quincy, MA)
`polygraph, and contractions were initiated with phenylephrine.
`Tolerance to GTN was induced by incubating rings with 0.3 mM
`GTN for 30 min. Rings were then washed several times with
`Krebs solution and left for 1 h before being further tested.
`
`Nitrite and Nitrate. Nitrite and nitrate concentrations (in the
`aqueous phase after ether extraction, see GTN biotransforma-
`tion) were determined in an NO analyzer (model 280, Sievers,
`Boulder, CO) according to the manufacturer’s instructions.
`
`cGMP Assay. Aortic rings were blotted and weighed before being
`incubated in aerated bioassay chambers for 1 h. Then, after
`treatment with different inhibitors, the rings were exposed to 1
`␮M GTN for 1 min and immediately frozen in liquid nitrogen
`and stored at ⫺80°C until the time of analysis. cGMP extraction
`and measurements were performed according to the manufac-
`turer’s protocol.
`
`ALDH Assay. Rabbit aortic rings were homogenized with a Kontes
`tissue grinder in 30 mM KPi buffer (deoxygenated with nitrogen
`gas), pH 7.5, and the homogenate was then sonicated and
`centrifuged at 10,000 g for 10 min. ALDH activity in the
`supernatant was monitored at room temperature by following
`NADH formation at 340 nm. The assay mixture (1 ml) con-
`tained: 100 mM Tris䡠HCl (pH 8.5), 1 mM NAD⫹, 1 mM
`propionaldehyde, and 1 mM 4-methylpyrazole.
`
`GTN Infusions inVivo. New Zealand White rabbits (2.5–3 kg) were
`anesthetized with isoflurane (1.5%). The carotid artery and
`jugular vein were isolated via cut down and cannulated. The
`blood pressure was monitored continuously with a Viggo Spec-
`tramed pressure transducer (AD Instruments, Grand Junction,
`CO) attached to a Gould (Cleveland) recorder. Drugs were
`infused via the jugular vein. mtALDH inhibitors (⬇5 mM) were
`infused 20 min before initiating studies with GTN. Cyanamide
`had no effect on mean arterial blood pressure, whereas chloral
`hydrate had a modest hypotensive effect. Similar studies were
`performed in Sprague–Dawley rats.
`
`Statistical Analysis. Results were statistically analyzed by the
`Student’s t test, and two sets of data were considered statistically
`different when P ⬍ 0.05.
`
`Results and Discussion
`We reasoned that the difficulty in isolating the enzyme respon-
`sible for GTN biotransformation from blood vessels may reflect
`a lack of starting material and兾or exposure to excessive concen-
`trations of GTN (under which conditions GTN metabolism may
`not represent biotransformation), and we thus screened alter-
`native animal tissues and cell lines by monitoring 1,2-GDN
`formation from physiological amounts of GTN (0.1 ␮M). We
`found that mouse macrophage RAW264.7 cells, which can be
`grown in large numbers, resemble vascular smooth muscle cells
`in generating mainly 1,2-GDN, and that after cellular disruption
`by sonication, the enzyme activity was present predominantly in
`the 100,000-g supernatant. We then developed a four-step
`purification procedure to isolate the enzyme from RAW264.7
`cells (Table 1). The enzyme activity passed freely through a
`DEAE-cellulose column and was preserved as a single peak after
`each of the following chromatographic steps:
`ion exchange
`(Q-Sepharose), hydrophobicity (butyl-Sepharose), and hydroxy-
`apatite column chromatography. At the end of the procedure the
`protein had been purified to near homogeneity (90-fold), with
`14.4% overall yield (Fig. 1a); its estimated subunit mass on SDS
`gel is about 53 kDa. N-terminal sequence of 30 aa residues
`(SAAATSAVPAPNHQPEVFXNQIFINNEWHD) was ob-
`tained by Edman degradation and matched exactly with mouse
`mtALDH (the unidentified residue 19 is a cysteine in the
`database). To confirm that this enzyme is located in mitochon-
`dria and responsible for the production of 1,2-GDN, we assayed
`purified preparations of mitochondria (20) that we had isolated
`from macrophage cells and found that they generated 1,2-GDN
`from GTN, whereas the mitochondria-free supernatant had no
`residual activity (data not shown). We then pretreated the
`macrophage cells with known ALDH inhibitors that either
`modify the active site residue (disulfiram or cyanamide) or
`compete for substrate binding (chloral hydrate) and found that
`all of the inhibitors, tested at concentrations from 50 ␮M to 1
`mM, blocked formation of 1,2-GDN (see below).
`In addition to the classical NAD⫹-dependent dehydrogena-
`tion activity, ALDH possesses an esterase activity (21), which is
`initiated by nucleophilic attack of the active site cysteine on
`substrate esters, such as p-nitrophenyl acetate, and is followed by
`hydrolysis to generate the acetic acid product. The analogous
`reaction with GTN would be expected to yield an E-S-NO2 active
`site intermediate that would then hydrolyze to yield nitrate
`⫺). However, we could not detect any nitrate formation.
`(NO3
`Rather, the purified enzyme catalyzed the stoichiometric for-
`⫺) from GTN. Specifically,
`mation of 1,2-GDN and nitrite (NO2
`the rates of 1,2-GDN formation were linear with physiological
`GTN concentrations in the range of 0.1 to 10 ␮M (data not
`shown), whereas 1,3-GDN was below detection limits, and nitrite
`was generated in stoichiometric amounts. The product ratio of
`1,2-GDN兾nitrite (0.83 ⫾ 0.14, n ⫽ 3) was similar to that of
`
`Chen et al.
`
`PNAS 兩
`
`June 11, 2002 兩 vol. 99 兩 no. 12 兩 8307
`
`Page 2 of 6
`
`

`
`Scheme 1.
`
`Km (acetaldehyde) and Ki (chloral hydrate) for dehydrogenase
`activity] reflect the absence of NAD⫹, which increases substrate
`binding (26). As expected, cyanamide did not have inhibitory
`effects on the purified enzyme because it first requires biocon-
`version in vivo.
`The classical sulfhydryl requirement for relaxation of vascular
`smooth muscle by GTN (2, 27) has never been satisfactorily
`explained. It has been suggested that thiols potentiate GTN
`activity through formation of bioactive SNOs and兾or protect
`against nitrate tolerance that results from depletion of intracel-
`lular thiols. However, the formation of SNO and depletion of
`intracellular thiol have not been confirmed (28), and alternative
`hypotheses, such as oxygen radical scavenging by thiol (29), have
`more recently found favor. We found that the purified mtALDH
`could catalyze 1,2-GDN formation only when DTT or 2-
`mercaptoethanol (2ME) were present (although these concen-
`trations of thiol had no effect on their own). Further, catalytic
`activity that was lost by dialysis could be fully restored by
`addition of DTT or 2ME (to a lesser extent) but not by
`glutathione.
`Human mtALDH has two cysteines immediately adjacent to
`the active site thiol. The formation of an intramolecular disulfide
`bond has been demonstrated by MS (30, 31) and accounts for the
`‘‘inactive form’’ of ALDH that is produced by ALDH inhibitors.
`Notably, active site participation in a disulfide bond has been
`previously implicated in the mechanism-based inhibition of
`mtALDH by NO (32), SNO (33), and GTN (21, 34), accounting
`for disulfram-like reactions in GTN-treated patients ingesting
`alcohol. But whereas the generation of disulfide has been viewed
`as indicative of enzyme inactivation (i.e., loss of dehydrogenase
`and esterase activities), the new data suggest that it can represent
`a reaction intermediate in a novel reductase reaction requiring
`thiol (and perhaps other reductant) cofactor (Scheme 1). That
`is, GTN is an inhibitor of mtALDH activity because it serves as
`a substrate for its reductase activity. Collectively, our data
`indicate that mtALDH is in fact a GTN reductase.
`The specific conversion of GTN to 1,2-GDN and dependence
`on thiol cofactor (as reductant) make mtALDH a compelling
`candidate for the elusive enzymatic system that is responsible for
`catalyzing the bioactivation of GTN in the vasculature. We tested
`this hypothesis by using an aortic ring bioassay. Preincubation of
`blood vessels with mtALDH inhibitors chloral hydrate and
`cyanamide or with the mtALDH substrate acetaldehyde (a
`competitive inhibitor of GTN metabolizing activity) produced a
`rightward shift in the GTN relaxation curves (Fig. 2 a1–c1) that
`depended on inhibitor concentration (Fig. 2a1 and data not
`shown). The mtALDH inhibitor disulfiram also blocked GTN
`relaxations, but interpretation of these data was confounded by
`a direct affect on vessel tone. By contrast, preincubation with the
`mtALDH inhibitors did not attenuate relaxation induced by the
`nitrosovasodilator SNP (Fig. 2 a2–c2), by NO solutions or
`
`(a) Purification of mtALDH from mouse macrophage RAW264.7 cells.
`Fig. 1.
`The gradual enrichment of the 53-kDa band is shown on a 7% SDS polyacryl-
`amide gel. Lane A, protein molecular mass marker (kDa); lane B, 100,000-g
`supernatant; lane C, after DEAE-cellulose; lane D, after Q-Sepharose; lane E,
`after butyl-Sepharose; lane F, after hydroxyapatite. (b) Kinetic analysis of
`purified GTN reductase in the absence of NAD⫹ (KM ⫽ 12 ␮M; Vmax ⫽ 3
`nmol兾mg per min).Vmax in the presence of NAD⫹ ⫽ ⬇30 nmol兾mg per min.
`
`⫺ ⫹ Eox.
`
`GTN ⫹ Ered
`
`1,2-GDN兾NOx (nitrite plus nitrate) (0.73 ⫾ 0.05, n ⫽ 3). Thus,
`the overall reaction can be expressed as:
`3 1,2-GDN ⫹ NO2
`To show that the GTN metabolizing activity and enantiomeric
`selectivity of mtALDH is not a function of cell type or species,
`we purified the enzyme from bovine liver (22). Bovine mtALDH
`also specifically catalyzes 1,2-GDN formation (1,2-GDN兾1,3-
`GDN ⫽ ⬇8:1) from GTN (0.1 to 10 ␮M), with classical
`Michaelis–Menten kinetics (Km ⫽ 11.98 ⫾ 3.04 ␮M and Vmax ⫽
`3.03 ⫾ 0.20 nmol兾min per mg) (Fig. 1b) [and with the same pH
`dependence as the dehydrogenase activity (optimum pH of 9.0)].
`Addition of NAD⫹ (20–1,000 ␮M) to the reaction mixture
`increases the rate of 1,2-GDN formation by about 10-fold (i.e.,
`Vmax ⬇ 30 nmol兾min per mg), easily adequate for NO bioactivity
`such as vasodilation. By comparison, the purified bovine enzyme
`has a specific ALDH activity of 0.54 ␮mol兾min per mg. It is
`important to appreciate that whereas the GTN reductase activity
`is ⬇15- to 20-fold lower than the classical dehydrogenase activity
`of mtALDH, bioactive NO concentrations are as much as
`10,000-fold lower than circulating or tissue acetaldehyde con-
`centrations (23), and the mitochondrial GTN reductase activity
`is large from the standpoint of NO biology, being significantly
`greater than that of mitochondrial NO synthase activity (24, 25).
`We reasoned that acetaldehyde (the natural enzyme sub-
`strate) should competitively inhibit GTN turnover by mtALDH,
`and we also tested the effects of the classical ALDH inhibitors
`on GTN-metabolizing activity of the purified bovine enzyme.
`Both chloral hydrate and acetaldehyde inhibited 1,2-GDN for-
`mation, with IC50 ⫽ 99.5 and 815.5 ␮M, respectively. The
`relatively high IC50s in the reductase assay [as compared with the
`
`8308 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.122225199
`
`Chen et al.
`
`Page 3 of 6
`
`

`
`Effects of inhibitor treatment and GTN tolerance on GTN-induced (Left) and SNP-induced (Right) relaxation in rabbit aorta. The dose–response curves
`Fig. 2.
`of control rings are shown as F, and inhibitor-treated rings (1 mM) are shown as E. (a) Chloral hydrate dose–response (E, 1 mM; Œ, 5 mM). (b) Cyanamide. (c)
`Acetaldehyde. (d) GTN tolerant. All inhibitor curves (a–c) are significantly different from GTN control (P ⬍ 0.01), but none are different from SNP control (P ⫽
`not significant). The GTN tolerance curve (d) is significantly different from control to P ⬍ 0.05. Data are expressed as the means ⫾ SD of 4 – 6 aortic rings.
`
`MEDICALSCIENCES
`
`verapamil, which works through a NO-independent mechanism
`(not shown). These results suggest a specific inhibition of the
`organic nitrate ester reductase activity in the arterial vascula-
`ture. Indeed, we confirmed that 1,2-GDN generation in the
`aortic rings was diminished by chloral hydrate, cyanamide, and
`acetaldehyde (1 mM of each) to 27.6%, 56.9%, and 52.5% of the
`control activity, respectively (Fig. 3a). The GTN reductase
`inhibitors and competitive substrate also significantly decreased
`cGMP accumulation in aortic rings (Fig. 4). For example, chloral
`hydrate and acetaldehyde completely blocked the increase in
`cGMP produced by GTN (1 ␮M for 1 min) and cyanamide
`abrogated the cGMP increase by 46%. Thus, we conclude that
`the reductase activity of mtALDH is largely responsible for
`the increase in cGMP that at least partly mediates vasorelaxation
`by GTN.
`It has been reported that vascular rings also generate some
`1,3-GDN from GTN, and we verified that a small amount
`of 1,3-GDN was produced under our assay conditions (1,2-
`GDN兾1,3-GDN ⫽ 4.98 ⫾ 0.64, n ⫽ 6). However, 1,3-GDN
`
`formation was not mechanism based, because heat-inactivated
`rings generated the same amounts.
`Chronic therapy with organic nitrates produces ‘‘tolerance,’’ a
`desensitization of blood vessels that compromises antianginal
`efficacy. This poorly understood phenomenon is at least partly
`attributable to impaired bioactivation of GTN in both small
`animals and humans and is readily recapitulated in vitro by
`lengthy high-dose exposures. We induced tolerance with a
`regimen of 0.3 mM GTN for 30 min, followed by washout.
`Whereas these conditions had no effect on SNP-induced rela-
`tions (Fig. 2d2), the EC50 value for GTN increased significantly
`(Fig. 2d1). Furthermore, in aorta made tolerant to GTN, both
`GTN reductase activity (Fig. 3b), as measured by 1,2-GDN-
`formation, and classical mtALDH activity (Fig. 3c) decreased
`significantly (and comparably), and cGMP accumulation was
`abolished (Fig. 4). Reports that ALDH (dehydrogenase activity)
`is also potently inhibited in patients undergoing chronic treat-
`ment with GTN and other nitrate esters (34) provide strong
`support for the clinical relevance of these findings.
`
`Chen et al.
`
`PNAS 兩
`
`June 11, 2002 兩 vol. 99 兩 no. 12 兩 8309
`
`Page 4 of 6
`
`

`
`Inhibition of cGMP accumulation in rabbit aorta by ALDH inhibitors
`Fig. 4.
`(1 mM ⫻ 20 min, followed by washout) and tolerance (Tol)-inducing amounts
`of GTN (0.3 mM ⫻ 30 min, followed by washout). Rings were (⫹) or were not
`(⫺) exposed to 1 ␮M GTN for 1 min. All bars are significantly less than control
`(Con) ⫹ GTN to P ⬍ 0.01, except cyanamide (Cya) ⫹ GTN, where P ⬍ 0.05. Data
`are presented as the means ⫾ SD of three aortic rings. Chl, chloral hydrate;
`Ace, acetaldehyde.
`
`(iv) offered alternative interpretations for the effects of thiols on
`GTN bioactivity, and (v) suggested new mechanisms of tolerance
`(29). There is even recent evidence to suggest that GTN acti-
`vation of guanylate cyclase may be independent of NO (38).
`None of these explanations, however, can reconcile the entire set
`of data. In contrast,
`the identification of
`the sulfhydryl-
`dependent mtALDH with GTN metabolic activity provides a
`viewpoint from which the models of Needleman et al. and
`Ignarro et al. are revived and shown to be in line with the original
`ideas of Murad et al. (1) as well as the newer data on enzymatic
`
`Inhibition of GTN (1 ␮M) biotransformation by ALDH inhibitors and
`Fig. 3.
`tolerance induction in rabbit aorta. (a) Concentration-dependent inhibition
`of 1,2-GDN formation by chloral hydrate (F), cyanamide (I), and acetalde-
`hyde (Œ). Activities are presented as the percentage of control rings (without
`inhibitor treatment). Data are the average of two experiments. (b) 1,2-GDN
`formation is reduced in tolerant vessels relative to control (P ⬍ 0.01). Data are
`the means ⫾ SD of three experiments. (c) mtALDH activity is inhibited in
`tolerant vessels (P ⬍ 0.01). Data are the means ⫾ SD of three experiments.
`
`As a further test of the physiological role of mtALDH in
`bioactivation of GTN, we assessed the effects of the mtALDH
`inhibitors cyanamide and chloral hydrate on the systemic effects
`of GTN infusions in anesthetized rabbits. Both inhibitors sig-
`nificantly attenuated the hypotensive effects of GTN (Fig. 5a),
`whereas they had little or no effect on SNP-mediated (i.e., NO
`dependent) or adenosine-mediated (i.e., NO independent) (not
`shown) hypotension (Fig. 5b). Similar results were obtained in
`rats, suggesting a generalized role for mtALDH in mammalian
`GTN metabolism (data not shown).
`The classical studies by Needleman et al. (27) and Ignarro et
`al. (2, 35), suggesting a sulfhydryl requirement for relaxation by
`GTN, have been interpreted in terms of a ‘‘thiol receptor model’’
`for organic nitrates in vascular smooth muscle, which provided
`for the possibility of an enzyme that generated inorganic nitrite
`as an intermediate and rationalized nitrate tolerance in terms of
`a depletion of intracellular thiols (27). Almost every feature of
`this model has been challenged in recent years by studies that
`have: (i) implicated alternative metalloenzymes in the mecha-
`nism of GTN bioactivation (11, 12); (ii) failed to detect a
`depletion of intracellular thiol
`in tolerant vessels (28); (iii)
`rejected the role of inorganic nitrite as an intermediate (36, 37);
`
`Fig. 5. mtALDH inhibitors block the hypotensive effects of GTN in vivo. (a)
`i.v. infusions of GTN in rabbits produce dose-dependent decrements in blood
`pressure (before treatment, F) that are attenuated significantly (P ⬍ 0.05) by
`both cyanamide (17 mg兾kg, E) and chloral hydrate (66 mg兾kg, ‚) (n ⫽ 5–7). (b)
`Cyanamide and chloral hydrate have no effect on SNP-mediated decreases in
`blood pressure (n ⫽ 7) (data for cyanamide treatment are shown). F, before
`treatment; E, after treatment.
`
`8310 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.122225199
`
`Chen et al.
`
`Page 5 of 6
`
`

`
`MEDICALSCIENCES
`
`biotransformation of GTN and impairment of this mechanism in
`tolerant vessels. In particular, our findings are strongly sup-
`ported by recent studies in GTN-treated patients, which have
`shown a profound decrease in mitochondrial-type (low Km)
`ALDH activity in the blood (34) and lower amounts of 1,2-GDN
`in tolerant vessels (8).
`Several lines of evidence have been offered against the original
`proposal (2, 39) that GTN acts via liberation of inorganic nitrite,
`including studies showing that GTN is readily transformed into
`NO bioactivity within cellular homogenates and subcellular
`fractions whereas nitrite is not (36, 37). However, these previous
`studies did not consider the possibility of biotransformation
`within the mitochondria, which is highly competent for nitrite
`bioactivation to NO. It has been shown that members of mito-
`chondria electron transport chain, cytochrome bc1 complex
`(complex III) and cytochrome c oxidase (complex IV) can
`reduce nitrite to NO (40, 41), and nitrite might also be trans-
`ported to the intermembrane space where the higher proton
`concentration may facilitate its conversion to NO or SNO.
`Mitochondria can also reduce NO to HNO, which has been
`shown to produce both reversible [e.g., intramolecular disulfide
`(32)] and irreversible [e.g., sulfinamide (33)] modifications of the
`active site cysteine in mtALDH and thus may explain why
`complete reversal of nitrate tolerance requires new protein
`synthesis (42). Interestingly, the organic nitrate nicorandil [N-(2-
`hydroxyethyl)nicotinamide nitrate ester] was recently shown to
`generate NO in rat myocardial mitochondria (43). It is therefore
`conceivable that other nitrates and endogenous nitrogen oxides
`may be ALDH substrates. More generally, our studies provide
`new insights into subcellular processing of NO metabolites and
`suggest new approaches for generating NO bioactivity and
`overcoming nitrate tolerance.
`
`1. Murad, F., Mittal, C. K., Arnold, W. P., Katsuki, S. & Kimura, H. (1978) Adv.
`Cyclic Nucleotide Res. 9, 145–158.
`2. Ignarro, L. J., Lippton, H., Edwards, J. C., Baricos, W. H., Hyman, A. L.,
`Kadowitz, P. J. & Gruetter, C. A. (1981) J. Pharmacol. Exp. Ther. 218, 739–749.
`3. Munzel, T. (2001) J. Am. Coll. Cardiol. 38, 1102–1105.
`4. Nakamura, Y., Moss, A. J., Brown, M. W., Kinoshita, M. & Kawai, C. (1999)
`Am. Heart J. 138, 577–585.
`5. Brien, J. F., McLaughlin, B. E., Kobus, S. M., Kawamoto, J. H., Nakatsu, K.
`& Marks, G. S. (1988) J. Pharmacol. Exp. Ther. 244, 322–327.
`6. Kawamoto, J. H., McLaughlin, B. E., Brien, J. F., Marks, G. S. & Nakatsu, K.
`(1990) J. Cardiovasc. Pharmacol. 15, 714–719.
`7. Slack, C. J., McLaughlin, B. E., Brien, J. F., Marks, G. S. & Nakatsu, K. (1989)
`Can. J. Physiol. Pharmacol. 67, 1381–1385.
`8. Sage, P. R., de la Lande, I. S., Stafford, I., Bennett, C. L., Phillipov, G.,
`Stubberfield, J. & Horowitz, J. D. (2000) Circulation 102, 2810–2815.
`9. Tsuchida, S., Maki, T. & Sato, K. (1990) J. Biol. Chem. 265, 7150–7157.
`10. Yeates, R. A., Schmid, M. & Leitold, M. (1989) Biochem. Pharmacol. 38,
`1749–1753.
`11. Millar, T. M., Stevens, C. R., Benjamin, N., Eisenthal, R., Harrison, R. & Blake,
`D. R. (1998) FEBS Lett. 427, 225–228.
`12. McDonald, B. J. & Bennett, B. M. (1990) Can. J. Physiol. Pharmacol. 68,
`1552–1557.
`13. McDonald, B. J. & Bennett, B. M. (1993) Biochem. Pharmacol. 45, 268–270.
`14. Seth, P. & Fung, H. L. (1993) Biochem. Pharmacol. 46, 1481–1486.
`15. McGuire, J. J., Anderson, D. J., McDonald, B. J., Narayanasami, R. & Bennett,
`B. M. (1998) Biochem. Pharmacol. 56, 881–893.
`16. Nigam, R., Anderson, D. J., Lee, S. F. & Bennett, B. M. (1996) J. Pharmacol.
`Exp. Ther. 279, 1527–1534.
`17. Dean, T. W. & Baun, D. C. (1975) Am. J. Hosp. Pharm. 32, 1036–1038.
`18. Crew, M. & Dicarlo, F. (1968) J. Chromatogr. 35, 506–512.
`19. Brien, J. F., McLaughlin, B. E., Breedon, T. H., Bennett, B. M., Nakatsu, K.
`& Marks, G. S. (1986) J. Pharmacol. Exp. Ther. 237, 608–614.
`20. Madden, E. A. & Storrie, B. (1987) Anal. Biochem. 163, 350–357.
`21. Mukerjee, N. & Pietruszko, R. (1994) J. Biol. Chem. 269, 21664–21669.
`22. Ikawa, M., Impraim, C. C., Wang, G. & Yoshida, A. (1983) J. Biol. Chem. 258,
`6282–6287.
`23. Weiner, H. (1979) in Biochemistry and Pharmacology of Ethanol, eds. Maj-
`chrowicz, E. & Noble, E. P. (Plenum, New York), Vol. 1, pp. 125–144.
`
`Residual, albeit attenuated, relaxations by GTN were ob-
`served in the presence of biotransformation inhibitors that
`completely blocked the GTN-mediated increase in cGMP. Like-
`wise, the relaxations in tolerant blood vessels appeared to be
`partly cGMP independent. These data are consistent with a
`number of studies showing that the mechanism of smooth muscle
`relaxation by NO and GTN is partly mediated by a direct effect
`on ion channels (44–46).
`Our discovery that GTN is metabolized within mitochondria
`by the actions of ALDH has implications for patient care. It has
`long been a mystery why nitrates do not provide survival
`benefits, unlike agents that increase NO synthase activity, and
`the possibility that they may impair mitochondrial metabolism
`would provide an explanation. Indeed, this notion is supported
`by the findings of mitochondrial damage, impaired respiration,
`and increased production of proatherogenic superoxide in GTN-
`exposed tissues (29, 47). In addition, our studies suggest that
`certain classes of drugs such as sulfonylurea hypoglycemics,
`chloral hydrate, and acetaminophen (48), which inhibit
`mtALDH activity, may be contraindicated in patients taking
`organic nitrates esters for the treatment of acute ischemic
`syndromes and congestive heart failure. Finally, a polymorphism
`in the mtALDH gene (ALDH2) has been linked to impairments
`in ethanol metabolism and risk of developing cancer and de-
`mentia (48), and it follows that the ALDH genotype may predict
`GTN responsiveness and even therapeutic efficacy. A variety of
`new approaches to providing NO bioactivity and overcoming
`tolerance are suggested by our results.
`
`We thank Drs. D. Hess, T. McMahon, A. Hausladen, J. Eu, and L. Liu
`for their support and many valuable discussions and M. Dewhirst for
`assistance.
`
`24. Kobzik, L., Stringer, B., Balligand, J. L., Reid, M. B. & Stamler, J. S. (1995)
`Biochem. Biophys. Res. Commun. 211, 375–381.
`25. Lacza, Z., Puskar, M., Figueroa, J. P., Zhang, J., Rajapakse, N. & Busija, D. W.
`(2001) Free Radical Biol. Med. 31, 1609–1615.
`26. Feldman, R. I. & Weiner, H. (1972) J. Biol. Chem. 247, 267–272.
`27. Needleman, P., Jakschik, B. & Johnson, E. M., Jr. (1973) J. Pharmacol. Exp.
`Ther. 187, 324–331.
`28. Boesgaard, S., Aldershvile, J., Poulsen, H. E., Loft, S., Anderson, M. E. &
`Meister, A. (1994) Circ. Res. 74, 115–120.
`29. Munzel, T., Sayegh, H., Freeman, B. A., Tarpey, M. M. & Harrison, D. G.
`(1995) J. Clin. Invest. 95, 187–194.
`30. Vallari, R. C. & Pietruszko, R. (1982) Science 216, 637–639.
`31. Shen, M. L., Lipsky, J. J. & Naylor, S. (2000) Biochem. Pharmacol. 60, 947–953.
`32. DeMaster, E. G., Redfern, B., Quast, B. J., Dahlseid,

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket