423

Methylation Reactions, the Redox Balance and Atherothrombosis: The Search for a Link with Hydrogen Sulfide Roberta Lupoli, MD1 Alessandro Di Minno, PharmD2 Gaia Spadarella, MD1 Massimo Franchini, MD3 Raffaella Sorrentino, PhD2 Giuseppe Cirino, PhD2 Giovanni Di Minno, MD, PharmD1

University, Naples, Italy 2 Department of Pharmacy, “Federico II” University, Naples, Italy 3 Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, Mantova, Italy

Address for correspondence Giovanni Di Minno, MD, Dipartimento di Medicina Clinica e Chirurgia, Via S. Pansini 5, 80131, Napoli, Italy (e-mail: [email protected]).

Semin Thromb Hemost 2015;41:423–432.

Abstract

Keywords

► ► ► ► ►

oxidative stress epigenetics cell proliferation inflammation translational medicine

It is now clear that homocysteine (Hcy) is irreversibly degraded to hydrogen sulfide (H2S), an endogenous gasotransmitter that causes in vivo platelet activation via upregulation of phospholipase A2 and downstream boost of the arachidonate cascade. This mechanism involves a transsulfuration pathway. Based on these new data, clinical and experimental models on the relationships between Hcy and folate pathways in vascular disease and information on the Hcy controversy have been reanalyzed in the present review. Most interventional trials focused on Hcy lowering by folate administration did not exclude patients routinely taking the arachidonate inhibitor aspirin. This may have influenced the results of some of these trials. It is also clear that nutritional intake of folate affects several enzymatic reactions of the methionine–Hcy cycle and associated one-carbon metabolism and, thereby, both methylation reactions and redox balance. Hence, it is conceivable that the abnormally high Hcy levels seen in pathologic states reflect a poorly elucidated perturbation of such reactions and of such balance. While it is unknown whether there is an interplay between H2S, methylation reactions, and redox balance, measuring the sole reduction of blood Hcy that follows folate administration may well be an oversimplified approach to a complex biologic perturbation. The need to investigate this complex framework is thoroughly discussed in this article.

In 1969, Kilmer McCully first observed that an infant with severe hyperhomocysteinemia (HHcy) leading to homocystinuria as a result of a rare condition of abnormal cobalamin metabolism exhibited severe diffuse arteriosclerosis indistinguishable from the lesions seen in some cases of homocystinuria caused by cystathionine β-synthase (CBS) deficiency.1 Because severely elevated plasma homocysteine (Hcy) concentrations were the only common metabolic abnormality in patients with similar vascular lesions and different inborn errors of Hcy metabolism, he postulated that Hcy, or one of its

derivatives, was toxic for the vascular wall (the “Hcy theory”). Seven years later, Wilcken and Wilcken showed that the concentration of Hcy-cysteine–mixed disulfide after a methionine load was slightly higher in coronary heart disease (CAD) patients than in age- and sex-matched controls.2 Major advances in the understanding of disorders of Hcy metabolism followed over the next 50 years, encompassing the rare homozygous enzyme deficiencies and the more common milder abnormalities.3 In addition, accelerated atherosclerosis and endothelial dysfunction have been associated with

published online May 14, 2015

Copyright © 2015 by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA. Tel: +1(212) 584-4662.

Issue Theme Hot Topics VII; Guest Editor: Emmanuel J. Favaloro, PhD, FFSc (RCPA).

DOI http://dx.doi.org/ 10.1055/s-0035-1549848. ISSN 0094-6176.

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

1 Department of Clinical Medicine and Surgery, “Federico II”

The “Homocysteine Theory” Revisited

Lupoli et al.

HHcy,4–6 which, in turn, acts as a prothrombotic marker.7 Moreover, over the last few decades, retrospective and prospective studies have consistently shown that (1) mildly elevated total Hcy (tHcy) levels (by 15 µM) are independently associated with a high risk of cardiovascular disease8; (2) a positive correlation exists between Hcy plasma levels and cerebrovascular disorders9,10; and (3) in patients with CBS deficiency chronically treated with Hcy-lowering therapy, a dramatic decrease in the tendency to arterial and venous thrombosis occurs.11 Together, such evidence argues for tHcy as being an independent graded risk factor for vascular disease in both men and women. However, the relation between elevated plasma tHcy and vascular disease is stronger in retrospective than in prospective studies,12 and a prolonged correction of HHcy by folate administration little affects cardiovascular risk in large prospective trials.13 As a consequence, there has been a debate on whether tHcy elevation is truly a risk factor or an epiphenomenon of a more complex picture.14 Here, we examine some models that provide knowledge about the relationships between the Hcy and folate pathways and vascular disease, with emphasis on the oxidative stress hypothesis. A direct connection between Hcy and the susceptibility to atherothrombosis is also provided. In vivo, Hcy is irreversibly degraded to hydrogen sulfide (H2S), a gaseous transmitter that leads to in vivo platelet activation via upregulation of phospholipase A2 and downstream boost of the arachidonic acid cascade. Platelet activation is one of the major pathways underlying an abnormally high cardiovascular risk.15 Under physiological conditions, the homeostatic equilibrium between oxidative and reductive stress (as well as between hypo- and hypermethylation) modulates and is modulated by the methionine–Hcy cycle.16 Since one-carbon

Fig. 1 Homocysteine Metabolism.

Seminars in Thrombosis & Hemostasis

Vol. 41

No. 4/2015

metabolism dependent on folate availability regulates both redox and methylation status, HHcy may be presently viewed as a marker of perturbation of one-carbon metabolism.17 The relevance of such novel paradigm with respect to the data from interventional studies with folate will also be discussed.

Homocysteine Metabolism: Relationships with the Redox Balance In vivo, Hcy is irreversibly degraded either via the transsulfuration pathway or through a reaction catalyzed by 5,10methylenetetrahydrofolate reductase (MTHFR) and is remethylated back to methionine (the methionine–Hcy cycle).18 Severe HHcy (tHcy >50 µM) may be due to defects in either remethylation or transsulfuration. When circulating Hcy levels exceed 80 to 100 µM, homocystinuria occurs. Abnormal remethylation is caused by MTHFR and/or methionine synthase (MTR) deficiency due to mutations in their respective genes. MTR can also be dysfunctional due to defects in cobalamin metabolism. In MTR deficiency or dysfunction, 5-methylTHF cannot cycle through MTR, leading to 5-methyl-THF accumulation (at the expense of the other folates), and, in turn, hampering the synthesis of purines and thymidine. Remethylation defects result in high Hcy and low methionine levels. As a consequence, rapidly dividing cells (e.g., bone marrow cells) will be affected, resulting in megaloblastic anemia and pancytopenia (folate deficiency). MTHFR deficiency, which occurs in 5 to 7% of individuals, is associated with mild/moderate elevations of plasma Hcy (i.e., 15–50 μM) that neither limit the availability of folates for purines and thymidine synthesis nor lead to blood cell abnormalities. The 677TT MTHFR genotype (MTHFRþþ), associated with a thermolabile variant of the enzyme with

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

424

impaired activity and reduced 5-methyltetrahydrofolate (5-MTHF) availability,19 is the most common cause of inherited mild/moderate HHcy.20,21 A significant interaction between this MTHFR variant and folate and vitamin B12 levels is known,22 as vitamin availability by dietary intake or supplementation shows more relevant effects in the phenotypic expression of tHcy levels in subjects carrying the 677TT genotype.23 An alternate pathway of remethylation, which employs betaine as the methyl donor, is catalyzed by betaine homocysteine methyl transferase (BHMT).24 In the transsulfuration pathway (►Fig. 1), Hcy is irreversibly degraded to L-cysteine (L-Cys) and H2S through multiple steps involving CBS and cystathionine γ-lyase (CTH). The process is initiated by the irreversible conversion of Hcy and serine to cystathionine by the enzyme cystathionine β synthase (CBS), which utilizes vitamin B6 as a cofactor.25 CTH then catalyzes the conversion of cystathionine to cysteine with the release of α-ketobutyrate. Thereafter, the cysteine aminotransferase (CT) and 3-mercaptopyruvate sulfur transferase (3-MPST) enzymes convert L-Cys to generate H2S.26 CBS deficiency results in accumulation of Hcy, but in contrast to remethylation defects, methionine is increased. The major in vivo redox buffers are glutathione (γ-glutamyl cysteinyl glycine), cysteine, and thioredoxin; the thiol groups in these molecules are readily oxidized and, hence, can buffer changes in redox state. L-Cys is the major extracellular redox buffer and is present more in the oxidized form, with a ratio of reduced cysteine to oxidized cysteine disulfide in plasma of approximately 1:4, with a cytosolic ratio of reduced to oxidized glutathione disulfide (GSH/GSSG) of 50:1.27 Cysteine is derived from the transsulfuration pathway, from the diet, or from protein catabolism (in the fasting state). Cysteine synthesized via transsulfuration can be delivered to other tissues as reduced cysteine or is derived from the catabolism of glutathione. On the other hand, cysteine is a substrate for the formation of γ-glutamyl cysteine (i.e., for the rate-limiting step in glutathione synthesis). Hence, the methionine–Hcy cycle is the major system that regulates the concentration of the major intra- and extracellular redox buffers and can act as a buffer to regulate redox status. In addition to HHcy, abnormalities in the methionine–Hcy cycle, whether due to genetic, nutritional, or other factors, are associated with oxidant stress-induced (cardio)vascular pathology.28 How the remethylation and transsulfuration pathways (and the alternate pathway of remethylation) are coordinately regulated in health and disease (especially, how their regulation relates with oxidative stress) needs to be thoroughly investigated. Finally, cysteine can undergo catabolic conversion to the biologically essential compounds taurine and sulfate.

Reactive Oxygen Species and In Vivo Platelet Activation in HHcy Regardless of whether the defect is in the remethylation or the transsulfuration pathway, a high risk of arterial and venous thrombosis and juvenile atherosclerosis is frequent in patients with HHcy.29,30 In MTHFRþþ carriers with previ-

Lupoli et al.

ous early-onset (at age 50, 000 patients from high-quality randomized clinical trials document that lowering Hcy levels by low and high doses of folic acid and vitamin B12 (which are involved in remethylation of Hcy to methionine by MS) and vitamin B6 (which acts as a cofactor in the transsulfuration of Hcy to cystathionine and L-Cys) little affects such risk.48 The straightforward view that by decreasing Hcy levels addition of folic acid would reduce the risk of CAD thus appeared as an oversimplified approach to a complex biologic perturbation.49 However, the evidence from Nature’s randomized trials50 (in the setting of HHcy due to CBS deficiency, high prevalence of arterial and venous Seminars in Thrombosis & Hemostasis

Vol. 41

No. 4/2015

occlusive disease, dramatic decrease in vascular risk following Hcy-lowering treatments) is too strong to be ignored. As the first data from interventional trials appeared, several explanations for the poor impact of folic acid supplementation have been provided. First, such trials might have been conceivably shorter in duration than appropriate for modulating an established progressive vascular disease such as atherothrombosis.51 Second, several confounders might have affected the results of the observational (oldest) studies on Hcy. Individuals with pre-existing atherosclerosis have higher Hcy levels than those without; some drugs may increase Hcy levels (►Table 1)— suggesting the potential for attenuation of their clinical benefit—and the effect of some drugs on Hcy is reversible following treatment withdrawal. Confounding factors (e.g., the simultaneous use of statins, aspirin, and other drugs) might have led to bias and inappropriate interpretation of the data from randomized interventional trials with folate as well. Most of them did not exclude patients routinely taking the arachidonate inhibitor aspirin, an approach which may have influenced the results (in vivo platelet activation by H2S derived from Hcy degradation is mediated by enhanced release of arachidonic acid and in turn enhanced biosynthesis of platelet thromboxane A2). Third, in a meta-analysis, the effect of the MTHFRþþ variant on Hcy concentration was larger in low folate settings (Asia; difference between individuals with TT vs. CC genotype, 3·12 μmol/L; 95% confidence interval [CI], 2·23–4·01) than in areas with folate fortification (America, Australia, and New Zealand, high; 0·13 μmol/L, 0·85 to 1·11).52 The odds ratio for stroke was also higher in Asia (1·68; 95% CI, 1·44–1·97) than in America, Australia, and New Zealand (1·03; 95% CI, 0·84– 1·25). Most randomized Hcy-lowering trials have been performed in populations with high folate concentrations. The relative risk (RR) of stroke in trials of Hcy-lowering interventions (0·94; 95% CI, 0·85–1·04) was similar to that predicted for the same extent of Hcy reduction in large genetic studies in populations with similar folate status (predicted RR, 1·00; 95% CI, 0·90–1·11). While folate fortification programs in North America have been circumstantially associated with a reduction in stroke rates,53 folic acid supplementation was documented to be effective in stroke prevention in populations with no or partial folic acid fortification.54,55 Thus, the folate status of the population should be taken into account when evaluating the link between tHcy and CAD. On the other hand, the controversy over Hcy and cardiovascular risk called attention to the issue that nutritional intake of folate and of the sulfur-containing amino acids methionine and cysteine affects a variety of enzymatic reactions of the methionine–Hcy cycle and associated onecarbon metabolism and, thereby, both methylation reactions and redox balance.56 Under these circumstances, an abnormal plasma Hcy level may well reveal an underlying perturbation in the regulation of the methylation and/or the redox status that needs to be further characterized.

Folate-Dependent One-Carbon Metabolism Methyl (–CH3), methylene (–CH2–), and formyl (–CHO–) forms of folate are used in the transfer of one-carbon groups

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

426

The “Homocysteine Theory” Revisited

Lupoli et al.

427

Table 1 Common determinants of homocysteine plasma levels Genetic Transsulfuration defects Cystathionine β-synthase defect (chromosome 21)

Homozygote: 1/340,000 born Heterozygote: 0.5% whole population Heterozygote mutation 844ins68: 10–15% whole Population in association with other risk factors

Remethylation defects 5,10-Methylenetetrahydrofolate reductase (MTHFR) defect

Homozygote: 1/3,350,000 born Heterozygote: 0.5% whole population

Thermolabile variant of MTHFR C677T (50% activity)

Homozygote: 5–20% whole population

Cobalamin/methylcobalamin conversion defect (cbl C, D, E, F, G)

a

Age/Sex

Increasing age, male sex, menopause

Nutritional

Folate þ vitamin B12 deficiency (elderly, pregnancy, malignancy) Vitamin B6 deficiency Lifestyle: excessive coffee/alcohol intake

Diseases

Bowel: malabsorption of vitamin B12 Liver failure Renal failure; renal transplantation Psoriasis: folate reduction Lymphoblastic leukemia, malignancy Hypothyroidism, diabetes mellitus, hypertension

Pharmacological

Metformin Glitazones (some) Lipid-lowering drugs (colestipol, nicotinate, fibrates) Methotrexate: 5-methyl-tetrahydrofolate reduction Interference with folate: diuretics, Anticonvulsantsa Folate antagonists Vitamin B12 antagonists (e.g., nitrate) Vitamin B6 deficit: estrogens

For example, carbamazepine, isoniazid, phenytoin.

in several metabolic pathways.57 (1) MTHF is irreversibly converted by the Flavin Adenine dinucleotide (FAD)-dependent enzyme MTHFR to the methyl donor 5-MTHF (►Fig. 2). The latter provides the methyl group for the MS/cobalamin dependent remethylation of Hcy to methionine. The tetrahydrofolate (THF) thus generated can be converted to its immediate precursor (MTHF) by the enzyme serine hydroxyl methyl transferase with the concomitant conversion of serine to glycine. (2) MTHF donates the methyl group involved in the methylation of deoxy-uridine monophosphate to deoxythymidine monophosphate (catalyzed by thymidylate synthase). Dihydrofolate produced during thymidylate synthesis is reduced by dihydrofolate reductase to THF. (3) MTHF dehydrogenase catalyzes the conversion of MTHF to formyl THF and the transfer of the carbon atom as the formyl group during purine synthesis. Hence, via the transfer of one-carbon moieties, folate metabolism is both linked to DNA synthesis and DNA methylation. With one exception (in 102 patients with established CAD and low tHcy at baseline [2 standard deviations of controls. Significant correlations of 8-isoPGF2α with both urinary 11-dehydro-TXB2 excretion and fasting tHcy levels have been reported in that setting. The conflicting effects of H2S—both pro-thrombotic in regard to platelet activation while simultaneously protective against oxidative endothelial stress and as a vasodilator—help set the frame for evaluating whether, rather than (just) a cause, increased H2S is a marker of (and an important defense mechanism against) oxidative stress. Besides H2S, two other gasotransmitters are produced in vivo: NO and CO. CO is generated by the heme oxygenase (HO) family of enzymes (HO-1, HO-2, and HO-3).96 NO is synthesized by inducible NO synthase (iNOS), endothelial NO synthase (eNOS), and neuronal NO synthase (nNOS). The transcriptional nuclear factor kappa B (NF-κB)—a key determinant of the proliferation of vascular smooth cells whose signaling is activated by Hcy97—modulates NO production in response to Hcy.98 The activity of NO synthase is inhibited by asymmetric dimethylarginine.78 NO and H2S share many of the same regulatory roles, including vasodilation, promotion of angiogenesis, attenuation of apoptosis, and antioxidant actions. By upregulating PLA2, H2S endogenously generated within the platelets boosts the arachidonic acid cascade, the latter in turn acting as a downstream signal of the L-Cys/H2S pathway. Major effects of the L-arginine/NO pathway involve the arachidonic acid cascade as well.99 Although H2S and NO exhibit independent signaling, the cross-talk between the L-Cys/H2S and the L-arginine/NO pathways is a well-established event and is involved in multiple pathways.45,100 The extent to what such interplay is perturbed in MTHFRþþ should be clarified. Data from settings in which an oxidative stress induces depletion of B vitamins or antioxidants (e.g., prolonged states of inflammation or immune activation101 or Alzheimer disease102) support the possibility of vascular prevention by folate mechanisms independent of Hcy lowering. These involve the antioxidant potential of 5-MTHF, including the improvement of tetrahydrobiopterin (BH4) availability—a direct superoxide-scavenging capacity—and its ability to reduce superoxide generation by the “uncoupled” eNOS. By switching the enzyme to its “coupled” state, the later restores NO synthesis and NADPH oxidation in a BH4-dependent manner.103 In patients undergoing Seminars in Thrombosis & Hemostasis

Vol. 41

No. 4/2015

429

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

The “Homocysteine Theory” Revisited

The “Homocysteine Theory” Revisited

Lupoli et al.

coronary bypass surgery, the intravenous infusion of 5-MTHF on saphenous veins and internal mammary arteries prevents in vivo peroxynitrite-mediated BH4 oxidation and improves eNOS coupling.104 Conversely, diminished endogenous H2S results in reduced BH4 levels, eNOS uncoupling, and profound oxidative stress.105 The plasma level of the one-carbon metabolite dimethylglycine, an independent predictor of CAD, is linked to changes in lipoprotein assembly.106 This tertiary amine is produced from betaine during the remethylation of Hcy to methionine. High plasma dimethylglycine levels may thus reflect an altered flux through BHMT, influencing liver AdoMet levels107 and thereby the availability of methyl groups for transmethylation reactions, including phosphatidylcholine synthesis, the major phospholipid in very low-density lipoprotein particles. Through its scavenging potential, H2S inhibits in vitro the atherogenic modifications of purified low-density lipoproteins.108 The so far unclear relationship between H2S, lipids, one-carbon metabolism, and CAD deserves special attention and prompts for further studies in vascular medicine. In addition to its obvious pathophysiological relevance, research targeted at estimating the role of H2S within the framework of one-carbon metabolomics is likely to gradually provide values in the form of personalized medicine. However, major achievements take often more time than anticipated by the original enthusiasm.

5 Beard RS Jr, Bearden SE. Vascular complications of cystathionine

6

7 8

9

10

11

12

13

14 15 16

Conflict of interest The authors have served on advisory boards and/or have received fees as speakers at meetings unrelated to the content of the present report.

17

18

Note Roberta Lupoli and Alessandro Di Minno equally contributed to the present overview. 19

Acknowledgment Medical writing assistance was provided by Rosanna Scala.

20

21

References 1 McCully KS. Vascular pathology of homocysteinemia: implica-

tions for the pathogenesis of arteriosclerosis. Am J Pathol 1969; 56(1):111–128 2 Wilcken DE, Wilcken B. The pathogenesis of coronary artery disease. A possible role for methionine metabolism. J Clin Invest 1976;57(4):1079–1082 3 Blom HJ, Smulders Y. Overview of homocysteine and folate metabolism. With special references to cardiovascular disease and neural tube defects. J Inherit Metab Dis 2011;34(1): 75–81 4 Boushey CJ, Beresford SA, Omenn GS, Motulsky AG. A quantitative assessment of plasma homocysteine as a risk factor for vascular disease. Probable benefits of increasing folic acid intakes. JAMA 1995;274(13):1049–1057

Seminars in Thrombosis & Hemostasis

Vol. 41

No. 4/2015

22

23

24

β-synthase deficiency: future directions for homocysteine-tohydrogen sulfide research. Am J Physiol Heart Circ Physiol 2011; 300(1):H13–H26 Austin RC, Lentz SR, Werstuck GH. Role of hyperhomocysteinemia in endothelial dysfunction and atherothrombotic disease. Cell Death Differ 2004;11(Suppl 1):S56–S64 Fay WP. Homocysteine and thrombosis: guilt by association? Blood 2012;119(13):2977–2978 Ueland PM, Refsum H, Beresford SA, Vollset SE. The controversy over homocysteine and cardiovascular risk. Am J Clin Nutr 2000; 72(2):324–332 Kawamoto R, Kajiwara T, Oka Y, Takagi Y. An association between plasma homocysteine concentrations and ischemic stroke in elderly Japanese. J Atheroscler Thromb 2002;9(2):121–125 Towfighi A, Saver JL, Engelhardt R, Ovbiagele B. Factors associated with the steep increase in late-midlife stroke occurrence among US men. J Stroke Cerebrovasc Dis 2008;17(4):165–168 Yap S, Boers GH, Wilcken B, et al. Vascular outcome in patients with homocystinuria due to cystathionine beta-synthase deficiency treated chronically: a multicenter observational study. Arterioscler Thromb Vasc Biol 2001;21(12):2080–2085 Homocysteine Studies Collaboration. Homocysteine and risk of ischemic heart disease and stroke: a meta-analysis. JAMA 2002; 288(16):2015–2022 Di Minno MN, Tremoli E, Coppola A, Lupoli R, Di Minno G. Homocysteine and arterial thrombosis: challenge and opportunity. Thromb Haemost 2010;103(5):942–961 Brattström L, Wilcken DE. Homocysteine and cardiovascular disease: cause or effect? Am J Clin Nutr 2000;72(2):315–323 Davì G, Patrono C. Platelet activation and atherothrombosis. N Engl J Med 2007;357(24):2482–2494 Metes-Kosik N, Luptak I, Dibello PM, et al. Both selenium deficiency and modest selenium supplementation lead to myocardial fibrosis in mice via effects on redox-methylation balance. Mol Nutr Food Res 2012;56(12):1812–1824 Joseph J, Loscalzo J. Methoxistasis: integrating the roles of homocysteine and folic acid in cardiovascular pathobiology. Nutrients 2013;5(8):3235–3256 Davis SR, Stacpoole PW, Williamson J, et al. Tracer-derived total and folate-dependent homocysteine remethylation and synthesis rates in humans indicate that serine is the main one-carbon donor. Am J Physiol Endocrinol Metab 2004;286(2):E272–E279 Frosst P, Blom HJ, Milos R, et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nat Genet 1995;10(1):111–113 de Franchis R, Mancini FP, D’Angelo A, et al. Elevated total plasma homocysteine and 677C—>T mutation of the 5,10-methylenetetrahydrofolate reductase gene in thrombotic vascular disease. Am J Hum Genet 1996;59(1):262–264 D’Angelo A, Coppola A, Madonna P, et al. The role of vitamin B12 in fasting hyperhomocysteinemia and its interaction with the homozygous C677T mutation of the methylenetetrahydrofolate reductase (MTHFR) gene. A case-control study of patients with early-onset thrombotic events. Thromb Haemost 2000;83(4):563–570 Brattström L, Israelsson B, Lindgärde F, Hultberg B. Higher total plasma homocysteine in vitamin B12 deficiency than in heterozygosity for homocystinuria due to cystathionine beta-synthase deficiency. Metabolism 1988;37(2):175–178 Guttormsen AB, Ueland PM, Nesthus I, et al. Determinants and vitamin responsiveness of intermediate hyperhomocysteinemia (> or ¼ 40 micromol/liter). The Hordaland Homocysteine Study. J Clin Invest 1996;98(9):2174–2183 Finkelstein JD, Harris BJ, Kyle WE. Methionine metabolism in mammals: kinetic study of betaine-homocysteine methyltransferase. Arch Biochem Biophys 1972;153(1):320–324

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

430

Lupoli et al.

25 Mudd SH, Finkelstein JD, Irreverre F, Laster L. Transsulfuration in

47 d’Emmanuele di Villa Bianca R, Mitidieri E, Di Minno MND, et al.

mammals. Microassays and tissue distributions of three enzymes of the pathway. J Biol Chem 1965;240(11):4382–4392 Kimura H. Hydrogen sulfide: its production, release and functions. Amino Acids 2011;41(1):113–121 Ottaviano FG, Handy DE, Loscalzo J. Redox regulation in the extracellular environment. Circ J 2008;72(1):1–16 Wang X, Cui L, Joseph J, et al. Homocysteine induces cardiomyocyte dysfunction and apoptosis through p38 MAPK-mediated increase in oxidant stress. J Mol Cell Cardiol 2012;52(3):753–760 Mudd SH, Skovby F, Levy HL, et al. The natural history of homocystinuria due to cystathionine beta-synthase deficiency. Am J Hum Genet 1985;37(1):1–31 D’Angelo A, Selhub J. Homocysteine and thrombotic disease. Blood 1997;90(1):1–11 Coppola A, D’Angelo A, Fermo I, et al. Reduced in vivo oxidative stress following 5-methyltetrahydrofolate supplementation in patients with early-onset thrombosis and 677TT methylenetetrahydrofolate reductase genotype. Br J Haematol 2005;131(1):100–108 Dragani A, Falco A, Santilli F, et al. Oxidative stress and platelet activation in subjects with moderate hyperhomocysteinaemia due to MTHFR 677 C!T polymorphism. Thromb Haemost 2012; 108(3):533–542 Hajjar DP, Gotto AM Jr. Biological relevance of inflammation and oxidative stress in the pathogenesis of arterial diseases. Am J Pathol 2013;182(5):1474–1481 Moriarty-Craige SE, Jones DP. Extracellular thiols and thiol/ disulfide redox in metabolism. Annu Rev Nutr 2004;24:481–509 Patrono C, FitzGerald GA. Isoprostanes: potential markers of oxidant stress in atherothrombotic disease. Arterioscler Thromb Vasc Biol 1997;17(11):2309–2315 Awad JA, Morrow JD, Takahashi K, Roberts LJ II. Identification of noncyclooxygenase-derived prostanoid (F2-isoprostane) metabolites in human urine and plasma. J Biol Chem 1993;268(6):4161–4169 Violi F, Pignatelli P, Pignata C, et al. Reduced atherosclerotic burden in subjects with genetically determined low oxidative stress. Arterioscler Thromb Vasc Biol 2013;33(2):406–412 Reilly MP, Praticò D, Delanty N, et al. Increased formation of distinct F2 isoprostanes in hypercholesterolemia. Circulation 1998;98(25):2822–2828 Voutilainen S, Morrow JD, Roberts LJ II, et al. Enhanced in vivo lipid peroxidation at elevated plasma total homocysteine levels. Arterioscler Thromb Vasc Biol 1999;19(5):1263–1266 Eberhardt RT, Forgione MA, Cap A, et al. Endothelial dysfunction in a murine model of mild hyperhomocyst(e)inemia. J Clin Invest 2000;106(4):483–491 Whitfield NL, Kreimier EL, Verdial FC, Skovgaard N, Olson KR. Reappraisal of H2S/sulfide concentration in vertebrate blood and its potential significance in ischemic preconditioning and vascular signaling. Am J Physiol Regul Integr Comp Physiol 2008; 294(6):R1930–R1937 Jha S, Calvert JW, Duranski MR, Ramachandran A, Lefer DJ. Hydrogen sulfide attenuates hepatic ischemia-reperfusion injury: role of antioxidant and antiapoptotic signaling. Am J Physiol Heart Circ Physiol 2008;295(2):H801–H806 Bucci M, Papapetropoulos A, Vellecco V, et al. Hydrogen sulfide is an endogenous inhibitor of phosphodiesterase activity. Arterioscler Thromb Vasc Biol 2010;30(10):1998–2004 d’Emmanuele di Villa Bianca R, Sorrentino R, Coletta C, et al. Hydrogen sulfide-induced dual vascular effect involves arachidonic acid cascade in rat mesenteric arterial bed. J Pharmacol Exp Ther 2011;337(1):59–64 King AL, Lefer DJ. Cytoprotective actions of hydrogen sulfide in ischaemia-reperfusion injury. Exp Physiol 2011;96(9):840–846 di Villa Bianca Rd, Coletta C, Mitidieri E, et al. Hydrogen sulphide induces mouse paw oedema through activation of phospholipase A2. Br J Pharmacol 2010;161(8):1835–1842

Hydrogen sulphide pathway contributes to the enhanced human platelet aggregation in hyperhomocysteinemia. Proc Natl Acad Sci U S A 2013;110(39):15812–15817 Martí-Carvajal AJ, Solà I, Lathyris D, Karakitsiou DE, SimancasRacines D. Homocysteine-lowering interventions for preventing cardiovascular events. Cochrane Database Syst Rev 2013;1: CD006612 Loscalzo J. Homocysteine trials—clear outcomes for complex reasons. N Engl J Med 2006;354(15):1629–1632 Hingorani A, Humphries S. Nature’s randomised trials. Lancet 2005;366(9501):1906–1908 Selhub J. The many facets of hyperhomocysteinemia: studies from the Framingham cohorts. J Nutr 2006;136(6, Suppl): 1726S–1730S Holmes MV, Newcombe P, Hubacek JA, et al. Effect modification by population dietary folate on the association between MTHFR genotype, homocysteine, and stroke risk: a meta-analysis of genetic studies and randomised trials. Lancet 2011;378(9791):584–594 Yang Q, Botto LD, Erickson JD, et al. Improvement in stroke mortality in Canada and the United States, 1990 to 2002. Circulation 2006;113(10):1335–1343 Saposnik G, Ray JG, Sheridan P, McQueen M, Lonn E; Heart Outcomes Prevention Evaluation 2 Investigators. Homocysteine-lowering therapy and stroke risk, severity, and disability: additional findings from the HOPE 2 trial. Stroke 2009;40(4):1365–1372 Huo Y, Qin X, Wang J, et al. Efficacy of folic acid supplementation in stroke prevention: new insight from a meta-analysis. Int J Clin Pract 2012;66(6):544–551 Guéant JL, Namour F, Guéant-Rodriguez RM, Daval JL. Folate and fetal programming: a play in epigenomics? Trends Endocrinol Metab 2013;24(6):279–289 Løland KH, Bleie Ø, Strand E, et al. Effect of folic acid supplementation on levels of circulating Monocyte Chemoattractant Protein-1 and the presence of intravascular ultrasound derived virtual histology thin-cap fibroatheromas in patients with stable angina pectoris. PLoS ONE 2013;8(7):e70101 Lange H, Suryapranata H, De Luca G, et al. Folate therapy and instent restenosis after coronary stenting. N Engl J Med 2004; 350(26):2673–2681 Løland KH, Bleie O, Blix AJ, et al. Effect of homocysteinelowering B vitamin treatment on angiographic progression of coronary artery disease: a Western Norway B Vitamin Intervention Trial (WENBIT) substudy. Am J Cardiol 2010; 105(11):1577–1584 Blacher J, Czernichow S, Paillard F, Ducimetiere P, Hercberg S, Galan P; SU.FOL.OM3 Study Research Group. Cardiovascular effects of B-vitamins and/or N-3 fatty acids: the SU.FOL.OM3 trial. Int J Cardiol 2013;167(2):508–513 Obeid R. The metabolic burden of methyl donor deficiency with focus on the betaine homocysteine methyltransferase pathway. Nutrients 2013;5(9):3481–3495 Joseph J. Fattening by deprivation: methyl balance and perinatal cardiomyopathy. J Pathol 2011;225(3):315–317 Yeo EJ, Briggs WT, Wagner C. Inhibition of glycine N-methyltransferase by 5-methyltetrahydrofolate pentaglutamate. J Biol Chem 1999;274(53):37559–37564 Reik W, Dean W, Walter J. Epigenetic reprogramming in mammalian development. Science 2001;293(5532):1089–1093 Dean W, Lucifero D, Santos F. DNA methylation in mammalian development and disease. Birth Defects Res C Embryo Today 2005;75(2):98–111 Feinberg AP, Tycko B. The history of cancer epigenetics. Nat Rev Cancer 2004;4(2):143–153 Turunen MP, Ylä-Herttuala S. Epigenetic regulation of key vascular genes and growth factors. Cardiovasc Res 2011;90(3):441–446 Goldberg AD, Allis CD, Bernstein E. Epigenetics: a landscape takes shape. Cell 2007;128(4):635–638

26 27 28

29

30 31

32

33

34 35

36

37

38

39

40

41

42

43

44

45 46

48

49 50 51 52

53

54

55

56

57

58

59

60

61

62 63

64 65

66 67 68

Seminars in Thrombosis & Hemostasis

Vol. 41

No. 4/2015

431

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

The “Homocysteine Theory” Revisited

The “Homocysteine Theory” Revisited

Lupoli et al.

69 Chang PY, Lu SC, Lee CM, et al. Homocysteine inhibits arterial

70 71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

endothelial cell growth through transcriptional downregulation of fibroblast growth factor-2 involving G protein and DNA methylation. Circ Res 2008;102(8):933–941 Issa JP. CpG-island methylation in aging and cancer. Curr Top Microbiol Immunol 2000;249:101–118 Zaina S, Lindholm MW, Lund G. Nutrition and aberrant DNA methylation patterns in atherosclerosis: more than just hyperhomocysteinemia? J Nutr 2005;135(1):5–8 Lund G, Andersson L, Lauria M, et al. DNA methylation polymorphisms precede any histological sign of atherosclerosis in mice lacking apolipoprotein E. J Biol Chem 2004;279(28):29147–29154 Liu C, Xu D, Sjöberg J, Forsell P, Björkholm M, Claesson HE. Transcriptional regulation of 15-lipoxygenase expression by promoter methylation. Exp Cell Res 2004;297(1):61–67 Post WS, Goldschmidt-Clermont PJ, Wilhide CC, et al. Methylation of the estrogen receptor gene is associated with aging and atherosclerosis in the cardiovascular system. Cardiovasc Res 1999;43(4):985–991 Kim J, Kim JY, Song KS, et al. Epigenetic changes in estrogen receptor beta gene in atherosclerotic cardiovascular tissues and in-vitro vascular senescence. Biochim Biophys Acta 2007;1772(1):72–80 Castro R, Rivera I, Struys EA, et al. Increased homocysteine and S-adenosylhomocysteine concentrations and DNA hypomethylation in vascular disease. Clin Chem 2003;49(8):1292–1296 Böger RH, Bode-Böger SM, Sydow K, Heistad DD, Lentz SR. Plasma concentration of asymmetric dimethylarginine, an endogenous inhibitor of nitric oxide synthase, is elevated in monkeys with hyperhomocyst(e)inemia or hypercholesterolemia. Arterioscler Thromb Vasc Biol 2000;20(6):1557–1564 Loscalzo J. Adverse effects of supplemental L-arginine in atherosclerosis: consequences of methylation stress in a complex catabolism? Arterioscler Thromb Vasc Biol 2003;23(1):3–5 Figueiredo JC, Grau MV, Wallace K, et al. Global DNA hypomethylation (LINE-1) in the normal colon and lifestyle characteristics and dietary and genetic factors. Cancer Epidemiol Biomarkers Prev 2009;18(4):1041–1049 Pufulete M, Al-Ghnaniem R, Khushal A, et al. Effect of folic acid supplementation on genomic DNA methylation in patients with colorectal adenoma. Gut 2005;54(5):648–653 Rampersaud GC, Kauwell GP, Hutson AD, Cerda JJ, Bailey LB. Genomic DNA methylation decreases in response to moderate folate depletion in elderly women. Am J Clin Nutr 2000;72(4):998–1003 Newby AC. Metalloproteinase expression in monocytes and macrophages and its relationship to atherosclerotic plaque instability. Arterioscler Thromb Vasc Biol 2008;28(12):2108–2114 Rodríguez-Salvador J, Armas-Pineda C, Perezpeña-Diazconti M, et al. Effect of sodium butyrate on pro-matrix metalloproteinase9 and -2 differential secretion in pediatric tumors and cell lines. J Exp Clin Cancer Res 2005;24(3):463–473 Young DA, Lakey RL, Pennington CJ, et al. Histone deacetylase inhibitors modulate metalloproteinase gene expression in chondrocytes and block cartilage resorption. Arthritis Res Ther 2005; 7(3):R503–R512 Vacek TP, Kalani A, Voor MJ, Tyagi SC, Tyagi N. The role of homocysteine in bone remodeling. Clin Chem Lab Med 2013; 51(3):579–590 Litynski P, Loehrer F, Linder L, Todesco L, Fowler B. Effect of low doses of 5-methyltetrahydrofolate and folic acid on plasma homocysteine in healthy subjects with or without the 677C—> T polymorphism of methylenetetrahydrofolate reductase. Eur J Clin Invest 2002;32(9):662–668 Arnadottir M, Brattström L, Simonsen O, et al. The effect of highdose pyridoxine and folic acid supplementation on serum lipid and plasma homocysteine concentrations in dialysis patients. Clin Nephrol 1993;40(4):236–240 Pattini A, Di Salvo V, Schena F, Le Grazie C. Iron values and haematological parameters of cross-country skiers. Effect of iron

Seminars in Thrombosis & Hemostasis

Vol. 41

No. 4/2015

89 90

91 92 93

94

95

96

97

98

99

100 101

102

103

104

105

106

107

108

and 5-methyltetrahydrofolic acid (Prefolic) supplementation. Clin Trials J 1988;25:296–307 Shane B. Folate chemistry and metabolism. In: Bayley LB, ed. Folate in Health and Disease. New York: Marcel Dekker Inc.; 1995:1–22 Kim JI, Choi SH, Jung KJ, Lee E, Kim HY, Park KM. Protective role of methionine sulfoxide reductase A against ischemia/reperfusion injury in mouse kidney and its involvement in the regulation of transsulfuration pathway. Antioxid Redox Signal 2013;18(17):2241–2250 Tang G, Wu L, Wang R. Interaction of hydrogen sulfide with ion channels. Clin Exp Pharmacol Physiol 2010;37(7):753–763 Predmore BL, Lefer DJ, Gojon G. Hydrogen sulfide in biochemistry and medicine. Antioxid Redox Signal 2012;17(1):119–140 Mosharov E, Cranford MR, Banerjee R. The quantitatively important relationship between homocysteine metabolism and glutathione synthesis by the transsulfuration pathway and its regulation by redox changes. Biochemistry 2000;39(42):13005–13011 Di Minno G, Davì G, Margaglione M, et al. Abnormally high thromboxane biosynthesis in homozygous homocystinuria. Evidence for platelet involvement and probucol-sensitive mechanism. J Clin Invest 1993;92(3):1400–1406 Davì G, Di Minno G, Coppola A, et al. Oxidative stress and platelet activation in homozygous homocystinuria. Circulation 2001; 104(10):1124–1128 Calvert JW, Coetzee WA, Lefer DJ. Novel insights into hydrogen sulfide—mediated cytoprotection. Antioxid Redox Signal 2010; 12(10):1203–1217 Osanai T, Fujiwara N, Sasaki S, et al. Novel pro-atherogenic molecule coupling factor 6 is elevated in patients with stroke: a possible linkage to homocysteine. Ann Med 2010;42(1): 79–86 Welch GN, Upchurch GR Jr, Farivar RS, et al. Homocysteineinduced nitric oxide production in vascular smooth cells by NFκB dependent transcriptional activity of Nos2. Proc Assoc Am Physicians 1998;110:22–31 Salvemini D, Kim SF, Mollace V. Reciprocal regulation of the nitric oxide and cyclooxygenase pathway in pathophysiology: relevance and clinical implications. Am J Physiol Regul Integr Comp Physiol 2013;304(7):R473–R487 Review Kimura H. The physiological role of hydrogen sulfide and beyond. Nitric Oxide 2014;41:4–10 Fuchs D, Jaeger M, Widner B, Wirleitner B, Artner-Dworzak E, Leblhuber F. Is hyperhomocysteinemia due to the oxidative depletion of folate rather than to insufficient dietary intake? Clin Chem Lab Med 2001;39(8):691–694 McCaddon A, Regland B, Hudson P, Davies G. Functional vitamin B (12) deficiency and Alzheimer disease. Neurology 2002;58(9): 1395–1399 Stroes ESG, van Faassen EE, Yo M, et al. Folic acid reverts dysfunction of endothelial nitric oxide synthase. Circ Res 2000; 86(11):1129–1134 Antoniades C, Shirodaria C, Warrick N, et al. 5-methyltetrahydrofolate rapidly improves endothelial function and decreases superoxide production in human vessels: effects on vascular tetrahydrobiopterin availability and endothelial nitric oxide synthase coupling. Circulation 2006;114(11):1193–1201 Polhemus DJ, Lefer DJ. Emergence of hydrogen sulfide as an endogenous gaseous signaling molecule in cardiovascular disease. Circ Res 2014;114(4):730–737 Svingen GF, Ueland PM, Pedersen EK, et al. Plasma dimethylglycine and risk of incident acute myocardial infarction in patients with stable angina pectoris. Arterioscler Thromb Vasc Biol 2013; 33(8):2041–2048 Strakova J, Gupta S, Kruger WD, et al. Inhibition of betainehomocysteine S-methyltransferase in rats causes hyperhomocysteinemia and reduces liver cystathionine β-synthase activity and methylation capacity. Nutr Res 2011;31(7):563–571 Laggner H, Muellner MK, Schreier S, et al. Hydrogen sulphide: a novel physiological inhibitor of LDL atherogenic modification by HOCl. Free Radic Res 2007;41(7):741–747

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

432

Copyright of Seminars in Thrombosis & Hemostasis is the property of Thieme Medical Publishing Inc. and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use.

Methylation reactions, the redox balance and atherothrombosis: the search for a link with hydrogen sulfide.

It is now clear that homocysteine (Hcy) is irreversibly degraded to hydrogen sulfide (H(2)S), an endogenous gasotransmitter that causes in vivo platel...
348KB Sizes 0 Downloads 8 Views