Literature
首页医源资料库在线期刊动脉硬化血栓血管生物学杂志2006年第26卷第11期

Tetrahydrobiopterin and Cardiovascular Disease

来源:《动脉硬化血栓血管生物学杂志》
摘要:【摘要】Tetrahydrobiopterin(BH4)isanessentialcofactorforthearomaticaminoacidhydroxylases,whichareessentialintheformationofneurotransmitters,andfornitricoxidesynthase。Itispresentlyusedclinicallytotreatsomeformsofphenylketonuria(PKU)thatcanbeameliora......

点击显示 收起

【摘要】  Tetrahydrobiopterin (BH 4 ) is an essential cofactor for the aromatic amino acid hydroxylases, which are essential in the formation of neurotransmitters, and for nitric oxide synthase. It is presently used clinically to treat some forms of phenylketonuria (PKU) that can be ameliorated by BH 4 supplementation. Recent evidence supports potential cardiovascular benefits from BH 4 replacement for the treatment of hypertension, ischemia-reperfusion injury, and cardiac hypertrophy with chamber remodeling. Such disorders exhibit BH 4 depletion because of its oxidation and/or reduced synthesis, which can result in functional uncoupling of nitric oxide synthase (NOS). Uncoupled NOS generates more oxygen free radicals and less nitric oxide, shifting the nitroso-redox balance and having adverse consequences on the cardiovascular system. While previously difficult to use as a treatment because of chemical instability and cost, newer methods to synthesize stable BH4 suggest its novel potential as a therapeutic agent. This review discusses the biochemistry, physiology, and evolving therapeutic potential of BH4 for cardiovascular disease.

Tetrahydrobiopterin (BH 4 ) is an essential cofactor for the aromatic amino acid hydroxylases, which are essential in the formation of neurotransmitters, and for nitric oxide synthase (NOS). BH 4 replacement may help treat hypertension, ischemia-reperfusion injury, and cardiac hypertrophy with chamber remodeling, by restoring functional NOS. This review discusses BH 4 biochemistry, physiology, and evolving uses to treat cardiovascular disease.

【关键词】  tetrahydrobiopterin nitric oxide synthase atherosclerosis inflammation


Introduction


In 1963, a naturally occurring coenzyme for phenylalanine hydroxylase (PAH) was discovered to be the unconjugated pterin 5,6,7,8-tetrahydrobiopterin (BH 4 ). 1 BH 4 was subsequently found to be an essential cofactor for several other aromatic amino acid hydroxylases (tyrosine 2 and tryptophane 3 ) involved with neurotransmitter biosynthesis, glyceryl-ether mono-oxygenase, and nitric oxide synthase (NOS). To be functional, BH 4 must be in its fully reduced form, and depletion and/or BH 4 oxidation to BH 3 and BH 2 reduces its activity. For the cardiovascular system, the role of BH 4 in NOS activity is particularly relevant. Reduced BH 4 was first shown to contribute to vascular pathophysiology and hypertension, whereas more recent studies have found important roles in cardiac hypertrophy and remodeling, and ischemia/reperfusion physiology. Development of genetic mouse models that modulate BH 4 synthesis have greatly advanced understanding of its role to normal NOS and vascular function. Here we briefly review the pharmacology, physiology, and therapeutic potential of BH 4.


BH 4 Biosynthesis


BH 4 is formed by either a de novo or salvage pathway ( Figure 2 ). De novo synthesis starts with guanidine triphosphate cyclohydrolase (GTPCH) in a magnesium, zinc, and NADPH-dependent reaction, and continues through 2 intermediates (7,8-dihydroneopterin triphosphate and 6-pyruvoyl-5,6,7, 8-tetrahydropterin) mediated by 6-pyruvoyl-tetrahydropterin synthase and sepiapterin reductase. 4 GTPCH is the rate limiting enzyme and is under negative feedback regulation by GTPCH feedback regulatory protein (GFRP) and BH 4 itself, and positive feedback by phenylalanine. 5 GTPCH is also regulated at the expression level, being increased by calcium 6 and 3-hydroxy-3-methylglutaryl (HMG)-coenzyme A (CoA) reductase inhibition, 7 and by cytokines such as interferon-, tumor necrosis factor-, and interleukin-1ß. Cytokine activation may involve coordinated activation of NF- B and the Jak2/Stat pathway, 8 and can increase BH 4 levels by increasing GTPCH-1 expression, 9-12 reducing GFRP expression, 5 and increasing PTPS expression. 12 BH 4 synthesis is also stimulated by insulin via a phosphatidylinositol-3-kinase-dependent activation of GTPCH-1, 13 whereas insulin-resistant states impair this mechanism. 14-17 Suppressors of GTPCH-1 activity include glucocorticoids 18,19 and cyclic GMP, the latter generated by short-term treatment with NO donors or sodium nitroprusside 20 and high levels of 7,8 BH 2. 21 These and other factors are summarized in the Table.


Figure 2. BH 4 biosynthesis and metabolism of BH 4. BH 4 can be formed by both a de novo pathway and a salvage pathway. The de novo pathway starts from guanidine triphosphate (GTP) and is regulated by the enzymes GTP cyclohydrolase (GTPCH), 6-pyruvoyltetrahydropterin synthase (PTPS) and sepiapterin reductase (SR). The salvage pathway starts from sepiapterin (Sep) and is mediated by the enzymes SR and dihydrofolate reductase (DHFR).


Influencing Factors of GTPCH


The salvage pathway generates BH 4 from oxidized forms via sepiapterin and sepiapterin reductase 22 but cannot compensate for defects in biosynthesis or recycling. 22-25 Two other enzymes are also involved with regenerating reduced BH 4 from oxidized forms, dihydrofolate reductase and dihyrdopterine reductase. Dihydrofolate reductase is mainly involved in folate metabolism and converts inactive 7,8-BH 2 back to BH 4, and plays an important role in the metabolism of exogenously administered BH 4. Recently, Chalupsky et al 26 demonstrated the role of dihydrofolate reductase in the regulation of BH 4 and NO bioavailability in the endothelium. Endothelial NAD(P)H oxidase-derived H 2 O 2 downregulated dihydrofolate reductase expression in response to angiotensin II, resulting in BH 4 deficiency and uncoupling of eNOS. Dihydropteridine reductase catalyzes BH 4 regeneration from qBH2 formed under oxidative stress.


BH 4 and NOS Function


BH 4 is an essential cofactor for all 3 NOS isoforms, 21,27,28 and basal enzyme activity correlates with the amount of BH 4 bound tightly to the protein. NOS is a homodimeric oxidoreductase containing iron protoporphyrin IX (heme), flavin adenine dinucleotide, flavin mononucleotide, and BH 4. 29,30 The flavin-containing reductase domain and a heme-containing oxygenase domain are connected by a regulatory calmodulin-binding domain. Binding of Ca 2+ /calmodulin orients the other domains to allow NADPH-derived electrons generated in the reductase domain to flow to the oxygenase domain, 31 ultimately resulting in the conversion of L-arginine to NO and L-citrulline. This occurs if BH 4 is bound 32,33 in the dimer interface, where it interacts with amino acid residues from both monomers to stabilize NOS dimerization and participate in arginine oxidation through the N-hydroxyl- L -arginine intermediate and the subsequent generation of NO.


The functional influence of BH 4 on NOS occurs at several levels. BH 4 can shift the NOS heme iron to a high spin state, increasing arginine binding and stabilizing the active dimeric form. 34-36 NOS-bound BH 4 may act as a redox-active cofactor via an unknown mechanism. 34 BH 4 increases substrate affinity of NOS 21,35,37 and participates in the electron transfer process, being converted to BH 3. radical during the NOS catalytic cycle and then restored to BH 4. The best-characterized structural effect of BH 4 is its stabilization of NOS dimers, particularly striking for inducible NOS (iNOS). 38 Under certain conditions iNOS dimerization strictly depends on BH 4. However, dimeric forms of all 3 isoforms can be obtained in the absence of BH 4. 39,40 Functional dimerization is thought to be a general requirement for normal NOS activity by biophysical alignment of the 2 oxidase domains linked to the opposing monomer reductase domain, thus this influence is thought to impact on enzyme function. Reduction of the ferric iron of endothelial NOS (eNOS) results in formation of an FeII-dioxygen complex, which would yield superoxide. However, BH 4 donates an electron to form an iron-oxy species (FeII-O) that in turn participates in arginine hydroxylation and NO generation. BH 4 also critical effects on the heme including the shift of the ferric iron spin state equilibrium toward a high spin state, 41-43 altering the stability of the Fe(II)O2 complex 44 and stabilizing 6-coordinate forms of NOS-ferrous-CO and ferrous-NO complexes. 40,45 Lastly, BH 4 has some modest antioxidant effects and can scavenge NOS derived reactive nitrogen and oxygen species. 37,46


When BH 4 bioavailability declines, NOS undergoes multiple changes. The dimer architecture is altered possibly because of malrotation of the oxidase domains to yield "molecular" uncoupling, 47,48 and the catalytic activity becomes "functionally" uncoupled. In the latter situation, the stoichiometric coupling between the reductase domain and L-arginine at the active site is lost, resulting in formation of superoxide and/or hydrogen peroxide. While increased generation of superoxide by uncoupled eNOS has become general accepted, it should be noted that these findings are all based on in vitro measurements and that this remains to be confirmed by in vivo real-time measurements.


The importance of GTPCH to BH 4 levels and NOS activity have been elegantly explored both in vitro and in vivo. Cai et al 49 showed in endothelial cells that GTPCH gene transfer increases BH 4 10-fold over baseline, accompanied by a 25% increase in NOS3-dependent NO production. In the control cells, NOS3 was principally monomeric, whereas GTPCH gene transfer induced a 3-fold increase of NOS3 dimerization. Alp et al reported on a transgenic mouse with human GTPCH overexpression targeted to endothelial cells under control of the mouse Tie2 promoter. 48 Theses mice demonstrated a 3-fold increase in vascular BH 4, reduced endothelial superoxide production, and preserved NO bioavailability comp with wild-type littermates in a streptozotocin model of diabetic vascular disease. These investigators also revealed enhanced NOS activity by gene transfer of GTPCH, and evidence of tight stoichiometry between BH 4 and NOS enzyme levels using combined GTPCH-transgenic and NOS3 knockout models. 50 A hph-1 mouse 51 has decreased hepatic GTPCH activity and defective BH 4 biosynthesis. These mice display pulmonary hypertension with right heart hypertrophy, and enhanced sensitivity to chronic hypoxia. 52


BH 4 Bioavailability: Role of Oxidant Stress


BH 4 bioavailability is potently influenced by oxidative stress, by decreasing expression of GTPCH, 53 depleting NADPH, which is required for de novo synthesis 54 and is involved with BH 4 recycling, 55 and by oxidation to inactive BH 2. 56,57 Oxidized BH 4 further augments superoxide anion synthesis from NOS3, increasing the synthesis of peroxynitrite (ONOO - ), which is a potent oxidizer of BH 4. 58,59 Angiotensin II reduces BH 4 by endothelial NAD(P)H oxidase-derived H 2 O 2 -dependent downregulation of DHFR, 26 an enzyme involved with reduction of BH 2 back to BH 4. This response is associated with a significant increase in endothelial O 2 - production 60,61 and impaired endothelial function and homeostasis. BH4 oxidation is observed in a number of vascular diseases, 48,62 and although it cannot act as an NO cofactor, it can exacerbate BH 4 availability by competitive binding to NOS. 63


BH 4 Bioavailability and Inflammation/Atherosclerosis


Unlike hypertension, hypertrophy, and oxidant stress stimulation, other stimuli such as inflammatory cytokines have been found to increase BH 4 biosynthesis, and this may play a role in atherosclerosis. For example, d?Uscio et al 64 detected elevated BH 4 in atherosclerotic aortas of apolipoprotein E-deficient mice caused by increased expression and enzyme activity of GTPCH. Upregulation of GTPCH and BH 4 synthesis has been linked to stimulation by certain inflammatory cytokines 8,10,65-68 such as tumor necrosis factor-, interferon-, and IL-1ß, and may in this setting serve as a counter response to enhance NO production. 64 In atherosclerotic vessels, total NOS activity is three times higher than in control arteries, 69 caused mostly by increased expression and activity of iNOS. 68 Additional support for upregulated BH 4 synthesis in the setting of inflammation comes from studies showing increased neopterin, a side-product of GTPCH-1 activity. 70,71 Intrinsic upregulation of BH 4 biosynthesis per se still does not rule out potential utility of exogenous BH 4 supplementation, because uncoupling is often still observed. 71


BH 4 Bioavailability: Role of Homocysteine, Folate, and Ascorbate


Increased vascular homocysteine is a potent risk factor for atherosclerosis and endothelial dysfunction, and some of this effect maybe mediated by its influence on BH 4. Homocysteine reduces intracellular BH 4 accompanied by apparent inhibition of de novo synthesis 72 likely by blunting sepiapterin reductase. BH 4 administration has beneficial effects on homocysteine-induced impairment of endothelial function, increased superoxide production, and impaired agonist-stimulated NO release. 73


Folic acid (folate) enhances the binding-affinity of BH 4 to NOS by a pteridine-binding domain serving as a locus through which the active form 5-methyl tetrahydrofolate (5MTHF) facilitates the electron transfer by BH 4 from the NOS reductase domain to the heme. 74 Folate also enhances regeneration of BH 4 from inactive BH 2 75 by stimulating DHFR, and it chemically stabilizes BH 4.


Ascorbic acid (Vitamin C) assists in BH 4 stabilization primarily through antioxidant and other effects. 76,77 Vitamin C also prevents formation of BH 2 from the BH 3. radical by facilitating the recycling to BH 4. 76 This may explain some of the benefits of ascorbate on endothelial function independent of superoxide scavenging. 78


BH4 Supplementation: Vascular Effects


Clinical data supporting vascular benefits of exogenous BH 4 are largely based on acute or subacute studies examining endothelium-dependent vasodilation by agonists or flow stimuli. BH 4 improves endothelial function in those who smoke, 79 diabetic subjects, 80 hypertensive subjects, 81 patients with hypercholesterolemia, 82 and those with coronary artery disease. 83,84 More recently, Setoguchi et al 85 showed BH 4 improves endothelial function in patients with systolic heart failure. Intracoronary administration of BH 4 to patients with cardiovascular risks but without flow-limiting coronary artery stenoses (<75%), enhanced endothelial-dependent vasodilation to acetylcholine. 83 Some studies contrasting acute BH 4 infusion versus more chronic treatment found beneficial effects on endothelial function only with the latter. 86 This supports changes in NOS3 coupling rather than a less specific antioxidant effect likely explain the response. Preliminary results of chronic treatment with BH 4 (400 mg twice daily, 4 weeks; Schirks Laboratories, Zurich, Switzerland) revealed benefits on endothelial dysfunction measured by acetylcholine response in forearm venous occlusion plethysmography in subjects with hypercholesterolemia. 87


BH 4 and the Heart


Reduced BH 4 likely represents an important cellular defect involved with both endothelial and myocyte dysfunction in hearts exposed to ischemia/reperfusion. BH 4 prevents ischemia/reperfusion cardiac dysfunction in vitro, 88 attenuating the normally observed rise in malondialdehyde levels, a marker of lipid peroxidation, and improving endothelial-dependent vasorelaxation. These changes appear independent of the intrinsic radical scavenging action of BH 4. 89 Takimoto et al 47 recently revealed the importance of BH 4 depletion and consequent NOS3 uncoupling in mice subjected to sustained pressure overload. In this model, myocardial and myocyte hypertrophy, interstitial fibrosis, and eventual cardiac dilation and dysfunction were linked to increased oxidant stress generated by uncoupled NOS3. Mice lacking NOS3 and exposed to the same pressure load developed more compensated concentric hypertrophy with preserved function, whereas control animals displayed marked dilation and dysfunction after 9 weeks of pressure stress. BH 4 50%, and BH 4 replacement therapy was able to reduced oxidative stress and inhibit cardiac dilation and depressed function in nonmutant controls. These data support potential benefits of BH 4 to the heart under conditions of stress, such as postinfarction remodeling, dilated myopathic remodeling, and hypertrophy.


Clinical Pharmacology


Exogenous BH 4 or its precursor sepiapterin first increases systemic BH 2 ( Figure 2 ) that is subsequently reduced to BH 4 90,91 by DHFR. Oral sapropterin hydrochloride, the synthetic form of 6R-BH4, at 2 mg/kg causes a 3-fold increase in BH 4 after 3 hours, returning to baseline at 24 hours. 92 Intracoronary infusion of 1 mg/min results in a rapid increase within 2 minutes raising coronary sinus BH 4 levels nearly 100-fold. 93 These doses are high and unlikely to be used as chronic therapy. They may also have amplified nonspecific antioxidants effects 94 of BH 4 independent of its role to NOS coupling and NO synthesis. Unfortunately, measurement of systemic (plasma) BH 4 has not been particularly useful for assessing local tissue levels and abnormal bioavailability. This has been shown to be true for coronary artery disease in which no significant differences were demonstrated compared with control population. 95 Shinozaki et al 96 demonstrated that patients with insulin resistance have lower ratios of plasma BH 4 :BH 2 and plasma BH4:total biopterin, whereas BH4 levels remained unchanged in patients with insulin resistance versus controls.


A potential disadvantage of BH 4 is that it might stimulate neuronal and inducible NOS activity, leading to excessive NO production and toxicity, particularly in inflammatory disorders. This remains conjectural. There are also some reports of elevated catecholamines with BH 4 induced by IL-2 treatment in cancer patients, 97 although studies in PKU patients receiving BH 4 have not reported this effect.


To date, the major factor limiting clinical BH 4 use has been its pharmacological preparation. BH 4 tablets have been large with an acidic taste and unstable as BH 4 is hygroscopic and easily oxidized. Thus, the medication had to be maintained frozen at -20°C to maintain long-term stability. However, BH 4 has recently been developed in the form of a thermostable and photostable tablet, with stability at room temperature of nearly 2 years (Biomarin, San Francisco, Calif). This development has opened up broader potential use for cardiovascular indications.


Conclusion


BH 4 plays a central role to normal NOS3 activity, yet remarkably it appears vulnerable to depletion, thereby providing a key mechanism underlying a number of cardiovascular disorders. This also opens up intriguing potential for replacement therapy, and new developments in BH 4 pharmaceutical preparation should facilitate larger scale testing of such efficacy. Such studies are being initiated now and we can anticipate new information regarding the therapeutic potential for BH 4 treatment of hypertension, vascular dysfunction, and cardiac remodeling in the relatively near future.


Figure 1. Biochemical structure of 5,6,7,8-tetrahydrobiopterin.


Acknowledgments


Sources of Funding


D.A.K. is supported by NHLBI P01: HL-59408 and the Peter Belfer Foundations. A.L.M. is supported by the American Heart Association Mid-Atlantic Affiliate Fellowship Grant, by the Belgian American Educational Foundation, and the University of Antwerp. D.A.K. is the recipient of a research grant from BioMarin, Calif.


Disclosures


None.

【参考文献】
  Kaufman S. The structure of the phenyl-alanine cofactor. Proc Natl Acad Sci U S A. 1963; 50: 1085-1093.

Brenneman AR, Kaufman S. Characteristics of the hepatic phenylalanine-hydroxylating system in newborn rats. J Biol Chem. 1965; 240: 3617-3622.

Jequier E, Robinson DS, Lovenberg W, Sjoerdsma A. Further studies on tryptophan hydroxylase in rat brainstem and beef pineal. Biochem Pharmacol. 1969; 18: 1071-1081.

Mayer B, Werner ER. In search of a function for tetrahydrobiopterin in the biosynthesis of nitric oxide. Naunyn Schmiedebergs Arch Pharmacol. 1995; 351: 453-463.

Gesierich A, Niroomand F, Tiefenbacher CP. Role of human GTP cyclohydrolase I and its regulatory protein in tetrahydrobiopterin metabolism. Basic Res Cardiol. 2003; 98: 69-75.

Hwang O, Choi HJ, Park SY. Up-regulation of GTP cyclohydrolase I and tetrahydrobiopterin by calcium influx. Neuroreport. 1999; 10: 3611-3614.

Hattori Y, Nakanishi N, Akimoto K, Yoshida M, Kasai K. HMG-CoA reductase inhibitor increases GTP cyclohydrolase I mRNA and tetrahydrobiopterin in vascular endothelial cells. Arterioscler Thromb Vasc Biol. 2003; 23: 176-182.

Huang A, Zhang YY, Chen K, Hatakeyama K, Keaney JF, Jr. Cytokine-stimulated GTP cyclohydrolase I expression in endothelial cells requires coordinated activation of nuclear factor-kappaB and Stat1/Stat3. Circ Res. 2005; 96: 164-171.

Hattori Y, Nakanishi N, Kasai K, Shimoda SI. GTP cyclohydrolase I mRNA induction and tetrahydrobiopterin synthesis in human endothelial cells. Biochim Biophys Acta. 1997; 1358: 61-66.

Katusic ZS, Stelter A, Milstien S. Cytokines stimulate GTP cyclohydrolase I gene expression in cultured human umbilical vein endothelial cells. Arterioscler Thromb Vasc Biol. 1998; 18: 27-32.

Linscheid P, Schaffner A, Blau N, Schoedon G. Regulation of 6-pyruvoyltetrahydropterin synthase activity and messenger RNA abundance in human vascular endothelial cells. Circulation. 1998; 98: 1703-1706.

Franscini N, Blau N, Walter RB, Schaffner A, Schoedon G. Critical role of IL-1beta for transcriptional regulation of endothelial 6-pyruvoyltetrahydropterin synthase. Arterioscler Thromb Vasc Biol. 2003; 23: e50-e53.

Ishii M, Shimizu S, Nagai T, Shiota K, Kiuchi Y, Yamamoto T. Stimulation of tetrahydrobiopterin synthesis induced by insulin: possible involvement of phosphatidylinositol 3-kinase. Int J Biochem Cell Biol. 2001; 33: 65-73.

Verma S, Arikawa E, Yao L, Laher I, McNeill JH. Insulin-induced vasodilation is dependent on tetrahydrobiopterin synthesis. Metabolism. 1998; 47: 1037-1039.

Viveros OH, Lee CL, bou-Donia MM, Nixon JC, NICHOL CA. Biopterin cofactor biosynthesis: independent regulation of GTP cyclohydrolase in adrenal medulla and cortex. Science. 1981; 213: 349-350.

Shinozaki K, Nishio Y, Okamura T, Yoshida Y, Maegawa H, Kojima H, Masada M, Toda N, Kikkawa R, Kashiwagi A. Oral administration of tetrahydrobiopterin prevents endothelial dysfunction and vascular oxidative stress in the aortas of insulin-resistant rats. Circ Res. 2000; 87: 566-573.

Meininger CJ, Marinos RS, Hatakeyama K, Martinez-Zaguilan R, Rojas JD, Kelly KA, Wu G. Impaired nitric oxide production in coronary endothelial cells of the spontaneously diabetic BB rat is due to tetrahydrobiopterin deficiency. Biochem J. 2000; 349: 353-356.

Mitchell BM, Webb RC. Impaired vasodilation and nitric oxide synthase activity in glucocorticoid-induced hypertension. Biol Res Nurs. 2002; 4: 16-21.

Johns DG, Dorrance AM, Tramontini NL, Webb RC. Glucocorticoids inhibit tetrahydrobiopterin-dependent endothelial function. Exp Biol Med (Maywood). 2001; 226: 27-31.

Shiraishi H, Kato T, Atsuta K, Sumi-Ichinose C, Ohtsuki M, Itoh M, Hishida H, Tada S, Udagawa Y, Nagatsu T, Hagino Y, Ichinose H, Nomura T. cGMP inhibits GTP cyclohydrolase I activity and biosynthesis of tetrahydrobiopterin in human umbilical vein endothelial cells. J Pharmacol Sci. 2003; 93: 265-271.

Klatt P, Schmid M, Leopold E, Schmidt K, Werner ER, Mayer B. The pteridine binding site of brain nitric oxide synthase. Tetrahydrobiopterin binding kinetics, specificity, and allosteric interaction with the substrate domain. J Biol Chem. 1994; 269: 13861-13866.

Nichol CA, Lee CL, Edelstein MP, Chao JY, Duch DS. Biosynthesis of tetrahydrobiopterin by de novo and salvage pathways in adrenal medulla extracts, mammalian cell cultures, and rat brain in vivo. Proc Natl Acad Sci U S A. 1983; 80: 1546-1550.

Duch DS, Lee CL, Edelstein MP, Nichol CA. Biosynthesis of tetrahydrobiopterin in the presence of dihydrofolate reductase inhibitors. Mol Pharmacol. 1983; 24: 103-108.

Gross SS, Levi R. Tetrahydrobiopterin synthesis. An absolute requirement for cytokine-induced nitric oxide generation by vascular smooth muscle. J Biol Chem. 1992; 267: 25722-25729.

Blau N, Thony B. Possible impact of tetrahydrobiopterin and sepiapterin on endothelial dysfunction. Arterioscler Thromb Vasc Biol. 2003; 23: 913-914.

Chalupsky K, Cai H. Endothelial dihydrofolate reductase: critical for nitric oxide bioavailability and role in angiotensin II uncoupling of endothelial nitric oxide synthase. Proc Natl Acad Sci U S A. 2005; 102: 9056-9061.

Marletta MA. Nitric oxide synthase structure and mechanism. J Biol Chem. 1993; 268: 12231-12234.

Stuehr DJ. Structure-function aspects in the nitric oxide synthases. Annu Rev Pharmacol Toxicol. 1997; 37: 339-359.

Stuehr D, Pou S, Rosen GM. Oxygen reduction by nitric-oxide synthases. J Biol Chem. 2001; 276: 14533-14536.

Knowles RG, Moncada S. Nitric oxide synthases in mammals. Biochem J. 1994; 298 (Pt 2): 249-258.

bu-Soud HM, Stuehr DJ. Nitric oxide synthases reveal a role for calmodulin in controlling electron transfer. Proc Natl Acad Sci U S A. 1993; 90: 10769-10772.

Schmidt HH, Hofmann H, Schindler U, Shutenko ZS, Cunningham DD, Feelisch M. No ·NO from NO synthase. Proc Natl Acad Sci U S A. 1996; 93: 14492-14497.

Wever RM, van DT, van Rijn HJ, de GF, Rabelink TJ. Tetrahydrobiopterin regulates superoxide and nitric oxide generation by recombinant endothelial nitric oxide synthase. Biochem Biophys Res Commun. 1997; 237: 340-344.

Gross S. Tetrahydrobiopterin: an essential cofactor of nitric oxide synthase with an elusive role. In: Nitric Oxide Biology and Pathobiology. San Diego, CA: Academic; 2005; 167-187.

Gorren AC, Schrammel A, Schmidt K, Mayer B. Effects of pH on the structure and function of neuronal nitric oxide synthase. Biochem J. 1998; 331 (Pt 3): 801-807.

Rusche KM, Marletta MA. Reconstitution of pterin-free inducible nitric-oxide synthase. J Biol Chem. 2001; 276: 421-427.

Kotsonis P, Frohlich LG, Shutenko ZV, Horejsi R, Pfleiderer W, Schmidt HH. Allosteric regulation of neuronal nitric oxide synthase by tetrahydrobiopterin and suppression of auto-damaging superoxide. Biochem J. 2000; 346 (Pt 3): 767-776.

Panda K, Rosenfeld RJ, Ghosh S, Meade AL, Getzoff ED, Stuehr DJ. Distinct dimer interaction and regulation in nitric-oxide synthase types I, II, and III. J Biol Chem. 2002; 277: 31020-31030.

Ghosh S, Wolan D, Adak S, Crane BR, Kwon NS, Tainer JA, Getzoff ED, Stuehr DJ. Mutational analysis of the tetrahydrobiopterin-binding site in inducible nitric-oxide synthase. J Biol Chem. 1999; 274: 24100-24112.

Abu-Soud HM, Wu C, Ghosh DK, Stuehr DJ. Stopped-flow analysis of CO and NO binding to inducible nitric oxide synthase. Biochemistry. 1998; 37: 3777-3786.

Rusche KM, Spiering MM, Marletta MA. Reactions catalyzed by tetrahydrobiopterin-free nitric oxide synthase. Biochemistry. 1998; 37: 15503-15512.

Mayer B, Wu C, Gorren AC, Pfeiffer S, Schmidt K, Clark P, Stuehr DJ, Werner ER. Tetrahydrobiopterin binding to macrophage inducible nitric oxide synthase: heme spin shift and dimer stabilization by the potent pterin antagonist 4-amino-tetrahydrobiopterin. Biochemistry. 1997; 36: 8422-8427.

Gerber NC, Nishida CR, Ortiz de Montellano PR. Characterization of human liver inducible nitric oxide synthase expressed in Escherichia coli. Arch Biochem Biophys. 1997; 343: 249-253.

Abu-Soud HM, Gachhui R, Raushel FM, Stuehr DJ. The ferrous-dioxy complex of neuronal nitric oxide synthase. Divergent effects of L-arginine and tetrahydrobiopterin on its stability. J Biol Chem. 1997; 272: 17349-17353.

Wang J, Stuehr DJ, Rousseau DL. Interactions between substrate analogues and heme ligands in nitric oxide synthase. Biochemistry. 1997; 36: 4595-4606.

Reif A, Frohlich LG, Kotsonis P, Frey A, Bommel HM, Wink DA, Pfleiderer W, Schmidt HH. Tetrahydrobiopterin inhibits monomerization and is consumed during catalysis in neuronal NO synthase. J Biol Chem. 1999; 274: 24921-24929.

Takimoto E, Champion HC, Li M, Ren S, Rodriguez ER, Tavazzi B, Lazzarino G, Paolocci N, Gabrielson KL, Wang Y, Kass DA. Oxidant stress from nitric oxide synthase-3 uncoupling stimulates cardiac pathologic remodeling from chronic pressure load. J Clin Invest. 2005; 115: 1221-1231.

Alp NJ, Mussa S, Khoo J, Cai S, Guzik T, Jefferson A, Goh N, Rockett KA, Channon KM. Tetrahydrobiopterin-dependent preservation of nitric oxide-mediated endothelial function in diabetes by targeted transgenic GTP-cyclohydrolase I overexpression. J Clin Invest. 2003; 112: 725-735.

Cai S, Alp NJ, McDonald D, Smith I, Kay J, Canevari L, Heales S, Channon KM. GTP cyclohydrolase I gene transfer augments intracellular tetrahydrobiopterin in human endothelial cells: effects on nitric oxide synthase activity, protein levels and dimerisation. Cardiovasc Res. 2002; 55: 838-849.

Bendall JK, Alp NJ, Warrick N, Cai S, Adlam D, Rockett K, Yokoyama M, Kawashima S, Channon KM. Stoichiometric relationships between endothelial tetrahydrobiopterin, endothelial NO synthase (eNOS) activity, and eNOS coupling in vivo: insights from transgenic mice with endothelial-targeted GTP cyclohydrolase 1 and eNOS overexpression. Circ Res. 2005; 97: 864-871.

McDonald JD, Cotton RG, Jennings I, Ledley FD, Woo SL, Bode VC. Biochemical defect of the hph-1 mouse mutant is a deficiency in GTP-cyclohydrolase activity. J Neurochem. 1988; 50: 655-657.

Khoo JP, Zhao L, Alp NJ, Bendall JK, Nicoli T, Rockett K, Wilkins MR, Channon KM. Pivotal role for endothelial tetrahydrobiopterin in pulmonary hypertension. Circulation. 2005; 111: 2126-2133.

Zheng JS, Yang XQ, Lookingland KJ, Fink GD, Hesslinger C, Kapatos G, Kovesdi I, Chen AF. Gene transfer of human guanosine 5'-triphosphate cyclohydrolase I restores vascular tetrahydrobiopterin level and endothelial function in low renin hypertension. Circulation. 2003; 108: 1238-1245.

NICHOL CA, Smith GK, Duch DS. Biosynthesis and metabolism of tetrahydrobiopterin and molybdopterin. Annu Rev Biochem. 1985; 54: 729-764.

Scott-Burden T. Regulation of nitric oxide production by tetrahydrobiopterin. Circulation. 1995; 91: 248-250.

Wever RM, Luscher TF, Cosentino F, Rabelink TJ. Atherosclerosis and the two faces of endothelial nitric oxide synthase. Circulation. 1998; 97: 108-112.

Kinoshita H, Tsutsui M, Milstien S, Katusic ZS. Tetrahydrobiopterin, nitric oxide and regulation of cerebral arterial tone. Prog Neurobiol. 1997; 52: 295-302.

Laursen JB, Somers M, Kurz S, McCann L, Warnholtz A, Freeman BA, Tarpey M, Fukai T, Harrison DG. Endothelial regulation of vasomotion in apoE-deficient mice: implications for interactions between peroxynitrite and tetrahydrobiopterin. Circulation. 2001; 103: 1282-1288.

Milstien S, Katusic Z. Oxidation of tetrahydrobiopterin by peroxynitrite: implications for vascular endothelial function. Biochem Biophys Res Commun. 1999; 263: 681-684.

Kase H, Hashikabe Y, Uchida K, Nakanishi N, Hattori Y. Supplementation with tetrahydrobiopterin prevents the cardiovascular effects of angiotensin II-induced oxidative and nitrosative stress. J Hypertens. 2005; 23: 1375-1382.

Taniyama Y, Griendling KK. Reactive oxygen species in the vasculature: molecular and cellular mechanisms. Hypertension. 2003; 42: 1075-1081.

Landmesser U, Dikalov S, Price SR, McCann L, Fukai T, Holland SM, Mitch WE, Harrison DG. Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension. J Clin Invest. 2003; 111: 1201-1209.

Gorren AC, Bec N, Schrammel A, Werner ER, Lange R, Mayer B. Low-temperature optical absorption spectra suggest a redox role for tetrahydrobiopterin in both steps of nitric oxide synthase catalysis. Biochemistry. 2000; 39: 11763-11770.

d?Uscio LV, Katusic ZS. Increased vascular biosynthesis of tetrahydrobiopterin in apolipoprotein E-deficient mice. Am J Physiol Heart Circ Physiol. 2006; 290: H2466-H2471.

Werner ER, Werner-Felmayer G, Wachter H. Tetrahydrobiopterin and cytokines. Proc Soc Exp Biol Med. 1993; 203: 1-12.

Sakai N, Kaufman S, Milstein S. Tetrahydrobiopterin is required for cytokine-induced nitric oxide production in a murine macrophage cell line (RAW 264). Mol Pharmacol. 1993; 43: 6-10.

Rosenkranz-Weiss P, Sessa WC, Milstien S, Kaufman S, Watson CA, Pober JS. Regulation of nitric oxide synthesis by proinflammatory cytokines in human umbilical vein endothelial cells. Elevations in tetrahydrobiopterin levels enhance endothelial nitric oxide synthase specific activity. J Clin Invest. 1994; 93: 2236-2243.

Kuhlencordt PJ, Chen J, Han F, Astern J, Huang PL. Genetic deficiency of inducible nitric oxide synthase reduces atherosclerosis and lowers plasma lipid peroxides in apolipoprotein E-knockout mice. Circulation. 2001; 103: 3099-3104.

d?Uscio LV, Milstien S, Richardson D, Smith L, Katusic ZS. Long-term vitamin C treatment increases vascular tetrahydrobiopterin levels and nitric oxide synthase activity. Circ Res. 2003; 92: 88-95.

Berdowska A, Zwirska-Korczala K. Neopterin measurement in clinical diagnosis. J Clin Pharm Ther. 2001; 26: 319-329.

Alp NJ, McAteer MA, Khoo J, Choudhury RP, Channon KM. Increased endothelial tetrahydrobiopterin synthesis by targeted transgenic GTP-cyclohydrolase I overexpression reduces endothelial dysfunction and atherosclerosis in ApoE-knockout mice. Arterioscler Thromb Vasc Biol. 2004; 24: 445-450.

Topal G, Brunet A, Millanvoye E, Boucher JL, Rendu F, Devynck MA, vid-Dufilho M. Homocysteine induces oxidative stress by uncoupling of no synthase activity through reduction of tetrahydrobiopterin. Free Radic Biol Med. 2004; 36: 1532-1541.

Dhillon B, Badiwala MV, Maitland A, Rao V, Li SH, Verma S. Tetrahydrobiopterin attenuates homocysteine induced endothelial dysfunction. Mol Cell Biochem. 2003; 247: 223-227.

Hyndman ME, Verma S, Rosenfeld RJ, Anderson TJ, Parsons HG. Interaction of 5-methyltetrahydrofolate and tetrahydrobiopterin on endothelial function. Am J Physiol Heart Circ Physiol. 2002; 282: H2167-H2172.

Kaufman S. Some metabolic relationships between biopterin and folate: implications for the "methyl trap hypothesis". Neurochem Res. 1991; 16: 1031-1036.

Kuzkaya N, Weissmann N, Harrison DG, Dikalov S. Interactions of peroxynitrite, tetrahydrobiopterin, ascorbic acid, and thiols: implications for uncoupling endothelial nitric-oxide synthase. J Biol Chem. 2003; 278: 22546-22554.

Patel KB, Stratford MR, Wardman P, Everett SA. Oxidation of tetrahydrobiopterin by biological radicals and scavenging of the trihydrobiopterin radical by ascorbate. Free Radic Biol Med. 2002; 32: 203-211.

Heller R, Unbehaun A, Schellenberg B, Mayer B, Werner-Felmayer G, Werner ER. L-ascorbic acid potentiates endothelial nitric oxide synthesis via a chemical stabilization of tetrahydrobiopterin. J Biol Chem. 2001; 276: 40-47.

Higman DJ, Strachan AM, Buttery L, Hicks RC, Springall DR, Greenhalgh RM, Powell JT. Smoking impairs the activity of endothelial nitric oxide synthase in saphenous vein. Arterioscler Thromb Vasc Biol. 1996; 16: 546-552.

Pieper GM. Acute amelioration of diabetic endothelial dysfunction with a derivative of the nitric oxide synthase cofactor, tetrahydrobiopterin. J Cardiovasc Pharmacol. 1997; 29: 8-15.

Cosentino F, Patton S, d?Uscio LV, Werner ER, Werner-Felmayer G, Moreau P, Malinski T, Luscher TF. Tetrahydrobiopterin alters superoxide and nitric oxide release in prehypertensive rats. J Clin Invest. 1998; 101: 1530-1537.

Stroes E, Kastelein J, Cosentino F, Erkelens W, Wever R, Koomans H, Luscher T, Rabelink T. Tetrahydrobiopterin restores endothelial function in hypercholesterolemia. J Clin Invest. 1997; 99: 41-46.

Setoguchi S, Mohri M, Shimokawa H, Takeshita A. Tetrahydrobiopterin improves endothelial dysfunction in coronary microcirculation in patients without epicardial coronary artery disease. J Am Coll Cardiol. 2001; 38: 493-498.

Maier W, Cosentino F, Lutolf RB, Fleisch M, Seiler C, Hess OM, Meier B, Luscher TF. Tetrahydrobiopterin improves endothelial function in patients with coronary artery disease. J Cardiovasc Pharmacol. 2000; 35: 173-178.

Setoguchi S, Hirooka Y, Eshima K, Shimokawa H, Takeshita A. Tetrahydrobiopterin improves impaired endothelium-dependent forearm vasodilation in patients with heart failure. J Cardiovasc Pharmacol. 2002; 39: 363-368.

Heitzer T, Brockhoff C, Mayer B, Warnholtz A, Mollnau H, Henne S, Meinertz T, Munzel T. Tetrahydrobiopterin improves endothelium-dependent vasodilation in chronic smokers : evidence for a dysfunctional nitric oxide synthase. Circ Res. 2000; 86: E36-E41.

Hurlimann D, Noll G, Gatti CD, Chenevard R, Eto M, Blau N, Alp N, Ruschitzka F, Enselelt F, Cannon K, Luscher TF, Cosentino F. Oral treatment with tetrahydrobiopterin reverses endothelial dysfunction and oxidative stress in hypercholesterolemia. Circulation. 2005; 112: U192-U193.

Verma S, Maitland A, Weisel RD, Fedak PW, Pomroy NC, Li SH, Mickle DA, Li RK, Rao V. Novel cardioprotective effects of tetrahydrobiopterin after anoxia and reoxygenation: identifying cellular targets for pharmacologic manipulation. J Thorac Cardiovasc Surg. 2002; 123: 1074-1083.

Yamashiro S, Noguchi K, Matsuzaki T, Miyagi K, Nakasone J, Sakanashi M, Koja K, Sakanashi M. Beneficial effect of tetrahydrobiopterin on ischemia-reperfusion injury in isolated perfused rat hearts. J Thorac Cardiovasc Surg. 2002; 124: 775-784.

Sawabe K, Wakasugi KO, Hasegawa H. Tetrahydrobiopterin uptake in supplemental administration: elevation of tissue tetrahydrobiopterin in mice following uptake of the exogenously oxidized product 7,8-dihydrobiopterin and subsequent reduction by an anti-folate-sensitive process. J Pharmacol Sci. 2004; 96: 124-133.

Hasegawa H, Sawabe K, Nakanishi N, Wakasugi OK. Delivery of exogenous tetrahydrobiopterin (BH4) to cells of target organs: role of salvage pathway and uptake of its precursor in effective elevation of tissue BH4. Mol Genet Metab. 2005; 86 (Suppl 1): S2-S10.

Ueda S, Matsuoka H, Miyazaki H, Usui M, Okuda S, Imaizumi T. Tetrahydrobiopterin restores endothelial function in long-term smokers. J Am Coll Cardiol. 2000; 35: 71-75.

Fukuda Y, Teragawa H, Matsuda K, Yamagata T, Matsuura H, Chayama K. Tetrahydrobiopterin restores endothelial function of coronary arteries in patients with hypercholesterolaemia. Heart. 2002; 87: 264-269.

Alp NJ, Channon KM. Regulation of endothelial nitric oxide synthase by tetrahydrobiopterin in vascular disease. Arterioscler Thromb Vasc Biol. 2004; 24: 413-420.

Walter R, Blau N, Kierat L, Schoedon G, Reinhart WH. Systemic tetrahydrobiopterin (BH4) levels and coronary artery disease. Cardiology. 2000; 94: 265-266.

Shinozaki K, Hirayama A, Nishio Y, Yoshida Y, Ohtani T, Okamura T, Masada M, Kikkawa R, Kodama K, Kashiwagi A. Coronary endothelial dysfunction in the insulin-resistant state is linked to abnormal pteridine metabolism and vascular oxidative stress. J Am Coll Cardiol. 2001; 38: 1821-1828.

Baker H, Marcus SL, Frank O, Petrylak DP, DeAngelis B, Dutcher JP, Wiernik PH. IL-2 enhances biopterins and catecholamines production during adoptive immunotherapy for various cancers. Cancer. 1989; 64: 1226-1231. <a href="/cgi/external_ref?access_num=10.1002/1097-0142(19890915)64:6


作者单位:Division of Cardiology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Md.

作者: An L. Moens; David A. Kass
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具