Vous êtes sur la page 1sur 11

Clinical Biochemistry 36 (2003) 431– 441

Hyperhomocysteinemia and its role in the development of


atherosclerosis
A.B. Lawrence de Koning, Geoff H. Werstuck, Ji Zhou, Richard C. Austin*
Department of Pathology and Molecular Medicine, McMaster University and the Henderson Research Centre, Hamilton, Ontario, Canada

Received 31 December 2002; received in revised form 25 April 2003; accepted 25 April 2003

Abstract
Numerous epidemiological studies have demonstrated that hyperhomocysteinemia (HHcy) is a strong and independent risk factor for
cardiovascular disease. HHcy can result from a deficiency in the enzymes or vitamin cofactors required for homocysteine metabolism.
Several hypotheses have been proposed to explain the cellular mechanisms by which HHcy promotes cardiovascular disease, including
oxidative stress, endoplasmic reticulum (ER) stress and the activation of pro-inflammatory factors. Studies using genetic- and diet-induced
animal models of HHcy have now demonstrated a direct causal relationship between HHcy, endothelial dysfunction and accelerated
atherosclerosis. These recently established animal models of HHcy provide investigators with important in vivo tools to (i) further
understand the cellular mechanisms by which HHcy contributes to endothelial dysfunction and atherosclerosis, and (ii) develop therapeutic
agents useful in the treatment of cardiovascular disease. © 2003 The Canadian Society of Clinical Chemists. All rights reserved.

Keywords: Hyperhomocysteinemia; Atherosclerosis; Endothelial dysfunction; Endoplasmic reticulum stress

1. Introduction ischemic symptoms, acute coronary syndromes usually re-


sult from lesion rupture and thrombosis [1– 4].
Atherosclerosis, the principal cause of cardiovascular Conventional risk factors for cardiovascular disease, includ-
disease and stroke, is a complex, chronic process that is ing hypercholesterolemia, hypertension, smoking and diabetes,
initiated at sites of endothelial cell injury and culminates in account for approximately 50% of all cases [1,2,12]. Evidence
the formation of stratified lesions of the arterial wall [1–5]. now indicates that HHcy, which occurs in approximately 5 to
Subendothelial infiltration of monocytic cells, proliferation 7% of the general population, is an important, independent risk
and migration of smooth muscle cells, cholesterol deposi- factor for atherosclerosis and thrombotic disease [13–20]. Fur-
tion, and elaboration of extracellular matrix are hallmark thermore, up to 40% of patients diagnosed with premature
features of atherosclerotic lesions. Cholesterol-laden coronary artery disease, peripheral vascular disease or recur-
smooth muscle cells and macrophages, morphologically rent venous thrombosis have HHcy [13–18].
recognized as foam cells, are observed at all stages of lesion In this review article, we will summarize the genetic and
development [6,7]. The importance of cholesterol and its nutritional factors that induce HHcy and further examine the
oxidized derivatives in the pathogenesis of atherosclerosis is clinical evidence implicating HHcy as an independent risk
supported by studies demonstrating the presence of choles- factor for cardiovascular disease. In addition, potential
terol within the lesions from humans and animals [8 –12]. mechanisms by which homocysteine accelerates atheroscle-
Traditionally, cholesterol and its oxidized derivatives are rosis will be discussed in light of the important findings
thought to accumulate in atherosclerotic lesions, thereby recently reported for both genetic- and diet-induced animal
contributing to the formation of advanced, multilayered models of HHcy.
atheromas. Although advanced atherosclerotic lesions cause
progressive narrowing of the vessel lumen that can lead to
2. Genetic and nutritional factors causing HHcy
* Corresponding author. Tel.: ⫹1-905-527-2299 ext. 42628; fax: ⫹1-
905-575-2646. Homocysteine is a thiol-containing amino acid that is
E-mail address: raustin@thrombosis.hhscr.org (R.C. Austin). formed during the metabolic conversion of methionine to

0009-9120/03/$ – see front matter © 2003 The Canadian Society of Clinical Chemists. All rights reserved.
doi:10.1016/S0009-9120(03)00062-6
432 A.B. Lawrence de Koning et al. / Clinical Biochemistry 36 (2003) 431– 441

It has been estimated that inadequate intake of B-vita-


mins and folate may account for approximately two thirds
of all cases of HHcy [28]. The decision of both American
and Canadian governments to enrich cereal grain flour with
folate in the late 1990s to prevent neural tube defects
(NTDs) was thus seen as having great potential in prevent-
ing HHcy in the general public. Cohort studies have since
showed that folate fortification of cereal flour products has
been effective in reducing total plasma homocysteine levels
in the general population [32–34]. It is not yet known,
however, if lowering of total plasma homocysteine via fo-
late supplementation is effective in reducing the risk of
developing atherosclerosis and thrombosis [32]. Prospective
studies are currently underway to examine this association.
Fig. 1. Metabolism of homocysteine. Dietary methionine is converted to
Regardless of the underlying condition leading to HHcy, the
the methyl donor S-adenosylmethionine (SAM) and is demethylated to relationship between elevated plasma homocysteine levels
S-adenosylhomocysteine (SAH), which is subsequently cleaved into aden- and vascular disease persists.
osine and homocysteine. Through the transsulphuration pathway, homo-
cysteine is converted to cystathionine and cysteine by the enzyme cysta-
thionine ␤-synthase (CBS) and the cofactor vitamin B6 (methylcobalamin).
Homocysteine can also be remethylated through the folate cycle. This
3. Homocysteine and vascular disease
pathway depends on the enzyme methionine synthase (MS) and vitamin
B12 as well as the enzyme 5,10-methylenetetrahydrofolate reductase Patients suffering from homocystinuria develop exten-
(MTHFR) and folate, which enters the cycle as tetrahydrofolate (THF). In sive arterial intimal thickening and fibrous plaques rich in
liver and kidney, homocysteine is also remethylated by the enzyme betaine smooth muscle cells and collagen [20,23,35]. These fibrous
homocysteine methyltransferase (BHMT), which transfers a methyl group
to homocysteine via demethylation of betaine to dimethylglycine (DMG).
lesions greatly outnumber fatty atherosclerotic lesions in the
major arteries of homocystinuric patients and, combined
with abnormally accelerated thrombosis, lead to tissue in-
cysteine (Figure 1). Once synthesized, homocysteine may farction and death at an early age [35]. Venous thrombosis
either be metabolized to cysteine by the transsulphuration and, to a lesser extent, arterial thrombosis are common in
pathway or converted back to methionine by the remethy- patients with homocystinuria [36]. Homocysteine-induced
lation pathway [16,19,20]. Mutations in genes responsible thrombosis seems to result from disturbances in the throm-
for homocysteine metabolism can result in homocystinuria, botic potential of endothelial cells rather than changes in
a severe form of HHcy [20]. The most common genetic platelet physiology. This leads to the development of a
cause of homocystinuria, homozygous cystathionine ␤-syn- prothrombotic phenotype consistent with vascular injury
thase (CBS) deficiency, results in plasma homocysteine (reviewed in [19,20]).
concentrations of up to 400 ␮mol/L, compared to normal It is estimated that 5 to 7% of the general population has
plasma levels of ⬃10 ␮mol/L [16,19,20]. Homozygous mild to moderate HHcy [13–16] and are at an increased risk
CBS deficiency is inherited as an autosomal recessive dis- of developing coronary atherosclerosis and vascular throm-
order with pleiotropic clinical manifestations, including bosis in later life. Individuals with mild HHcy also have an
mental retardation, ectopia lentis, osteoporosis, skeletal ab- increased prevalence of carotid artery stenosis, premature
normalities and hepatic steatosis [16,20]. Furthermore, pa- peripheral and cerebral vascular atherosclerosis [19 –21,
tients are at higher risk for premature atherosclerosis and 23]. Epidemiological data has generally showed a strong
thrombotic disease, which is the major cause of death association between plasma homocysteine and the prema-
[16,19,21–23]. While homozygous CBS deficiency is rare, ture development of atherosclerotic cardiovascular disease.
heterozygous CBS deficiency occurs in approximately 1% Many case-control and cross-sectional studies have identi-
of the general population and is associated with premature fied an association between total plasma homocysteine lev-
atherosclerosis and thrombotic disease in phenotypically els and the development of carotid, coronary, peripheral and
normal individuals [13,15,16,21–23]. Deficiency of 5,10- aortic atherosclerotic disease [37]. The multicentre Euro-
methylenetetrahydrofolate reductase (MTHFR), the enzyme pean Concerted Action Project (750 patients, 800 controls),
involved in folate-dependent remethylation of homocys- for example, revealed that HHcy confers an independent
teine to methionine, also causes severe HHcy and can lead risk similar to smoking and hypercholesterolemia for devel-
to premature atherosclerosis and thrombotic disease [24 – oping occlusive arterial disease [38]. Additionally, this
26]. Additionally, nutritional deficiencies in other vitamin study suggested that mild HHcy may have a synergistic
cofactors required for homocysteine metabolism, namely effect when combined with other risk factors, such as dia-
folate, vitamin B6 (pyridoxal phosphate), B12 (methylco- betes and hypertension. Control of mild HHcy may there-
balamin), and B2 (riboflavin) can promote HHcy [27–31]. fore be critically important in attenuating lesion formation
A.B. Lawrence de Koning et al. / Clinical Biochemistry 36 (2003) 431– 441 433

and preventing atherosclerosis when other established risk damage by HHcy is the impairment of vasodilation [48 –
factors are present. Unlike case-control studies, however, 52]. Abnormal vasomotor response is believed to be an
prospective cohort studies have not shown a robust or con- early step in the formation of atherosclerotic lesions [53].
sistent association between HHcy and atherosclerosis [39]. Mild to moderate HHcy in both human patients and animal
This seems to be especially true for prospective studies of models has been shown to impair normal endothelium-
healthy individuals with no previous history of cardiovas- dependent vasodilation [49,51,54]. B vitamin supplementa-
cular disease. Contrasting findings from prospective studies tion, which has been shown to lower plasma homocysteine
have been suggested to be a result of differing experimental levels, is effective in restoring normal endothelial function
designs and endpoints or perhaps even genetic or nutritional [55]. The additional observation that antioxidants prevent
heterogeneity of test populations, resulting in different car- impairment in endothelium-dependent relaxation suggests
diovascular risk profiles in the populations studied [40]. the involvement of reactive oxygen species (ROS) [47–54].
Despite these conflicting prospective studies, mild HHcy is The thiol group of homocysteine readily undergoes auto-
generally regarded as a causal, independent risk factor for oxidation in plasma to generate ROS, and it has been sug-
occlusive vascular disease [18,41,42]. gested that homocysteine induces cell injury/dysfunction
via a mechanism involving oxidative stress [50,56]. How-
ever, this hypothesis fails to explain why cysteine, which is
4. Potential cellular mechanisms by which present in plasma at 20 to 30 fold higher concentrations than
homocysteine promotes atherosclerosis homocysteine and is more readily auto-oxidized, does not
cause endothelial cell injury and is not considered a risk
4.1. Inflammatory response factor for cardiovascular disease [57–59]. Recent studies
have also demonstrated that homocysteine does not signif-
In general, the development and progression of athero- icantly increase the production of ROS via auto-oxidation
sclerosis is considered to be a form of chronic inflammation and is largely involved in antioxidant and reductive cellular
[1–3,11]. In support of these findings, in vitro studies have biochemistry [60]. Based on these findings, a re-examina-
demonstrated that homocysteine enhances the production of tion of the role of oxidative stress due to the auto-oxidation
several pro-inflammatory cytokines. Expression of mono- of homocysteine is warranted.
cyte chemoattractant protein 1 (MCP-1) is increased in Homocysteine-induced oxidative stress may occur
cultured human vascular endothelial cells, smooth muscle through other mechanisms. Various ex vivo studies using
cells and monocytes treated with homocysteine [43– 45]. vascular tissues have implicated HHcy in causing abnormal
MCP-1 is known to enhance monocyte endothelial binding vascular relaxation responses by enhancing the intracellular
and recruitment to the subendothelial cell space, a critical production of superoxide anion (O2⫺) [53,61,62]. O2⫺ is
early step in the formation of fatty streaks [11]. Homocys- believed to react with and decrease the availability of en-
teine has also been shown to increase expression of IL-8 dothelial nitric oxide (NO) to yield peroxynitrite, thereby
[43], a T-lymphocyte and neutrophil chemoattractant, in limiting normal vasodilation responses [63,64]. O2⫺ and
cultured endothelial cells. Homocysteine-induced expres- peroxynitrite are also known to contribute to the oxidative
sion of MCP-1 and IL-8 in monocytes and endothelial cells modification of tissues, resulting in the formation of lipid
has been shown to occur through activation of NF-␬B, a peroxides and nitrosated end products such as 3-nitroty-
transcription factor involved in mediating downstream in- rosine. The observations that homocysteine decreases the
flammatory processes [46]. Active NF-␬B, which is found expression of a wide range of antioxidant enzymes [65,66]
in atherosclerotic lesions, stimulates production of cyto- and impairs endothelial NO bioavailability by inhibiting
kines, chemokines, interferons, leukocyte adhesion mole- glutathione peroxidase activity [52,67] raises the possibility
cules, hemopoietic growth factors and major histocompati- that homocysteine sensitizes cells to the cytotoxic effects of
bility (MHC) class I molecules—all of which are thought to agents or conditions known to generate ROS. Decreased NO
influence vascular inflammation and, ultimately, atherogen- bioavailability has also been shown ex vivo to increase the
esis [46]. Recent data substantiating the in vivo activation of expression of MCP-1, which may enhance intravascular
NF-␬B and downstream inflammatory marker expression in monocyte recruitment and lead to accelerated lesion forma-
atherosclerotic lesions in hyperhomocysteinemic apoli- tion [68].
poprotein E (apoE)-deficient mice further supports the con-
cept that homocysteine contributes to the development of 4.3. Endoplasmic reticulum stress
atherosclerosis by causing vascular inflammation [47].
In eukaryotic cells, the endoplasmic reticulum (ER) is
4.2. Oxidative stress the cellular organelle where secretory proteins or proteins
destined for the plasma membrane undergo a variety of
Homocysteine is injurious to vascular endothelial cells modifications, including disulphide bond formation, glyco-
and impairs normal cellular function. One of the most rec- sylation, folding and oligomeric assembly (see reviews
ognizable physiological changes associated with endothelial [69 –71]). To assist in the correct folding of newly synthe-
434 A.B. Lawrence de Koning et al. / Clinical Biochemistry 36 (2003) 431– 441

facilitating the activation of IRE-1, ATF-6 and PERK [72–


74]. Activation of both IRE-1 and ATF-6 increase the ex-
pression of ER-resident chaperones. IRE-1 is a stress-acti-
vated transmembrane protein kinase having
endoribonuclease activity. Following ER stress, IRE-1
dimerizes and is autophosphorylated, thereby allowing
IRE-1 to act as an endoribonuclease in the alternative splic-
ing of XBP-1 mRNA. The removal of a 26 base pair intron
results in a translation frameshift that permits XBP-1 to act
as a transcriptional activator of genes containing upstream
ER stress response elements (ERSE). ATF-6 is a transcrip-
tion factor that is localized to the ER membrane in un-
stressed cells. Upon ER stress, ATF-6 is transported to the
Golgi where the cytosolic transactivation domain of ATF-6
is cleaved from the membrane by specific proteases (S1P
and S2P) that also recognize and cleave sterol regulatory
element-binding proteins (SREBPs) under conditions of ste-
Fig. 2. Proximal sensors IRE-1, ATF-6 and PERK co-ordinately regulate rol starvation. Following release, the transactivation domain
UPR signaling in eukaryotes. ER transmembrane protein IRE-1 dimerizes of ATF-6 localizes to the nucleus where it binds to ERSE,
and autophosphorylates upon release of GRP78 to bind unfolded proteins.
Endoribonuclease activity of IRE-1 cleaves mammalian XBP-1 mRNA to
thereby activating transcription of numerous UPR-respon-
remove a 26-base pair intron. Translation of alternatively spliced XBP-1 sive genes, including GRP78, GADD153 and ERp72. Con-
mRNA yields XBP-1 protein containing a novel C-terminus that binds to comitant with an increase in the transcriptional activation of
ER stress response element (ERSE) promoters, increasing transcription of UPR-responsive genes, ER stress also leads to a rapid,
ER chaperone genes. ATF-6 migrates to the Golgi following GRP78 general decrease in protein synthesis, a cellular process
release and is cleaved by site 1 and site 2 (S1/S2) proteases. The released
50 to 60 kDa ATF-6 transactivation domain migrates to the nucleus and
mediated by the transmembrane protein kinase, PERK. Ac-
binds to ERSE, increasing transcription of ER chaperones. PERK is acti- tivation of PERK causes phosphorylation of eukaryotic ini-
vated via dimerization and autophosphorylation following GRP78 release. tiation factor-2␣ (eIF-2␣), which effectively blocks mRNA
PERK catalyzes the phosphorylation of eukaryotic initiation factor-2␣ translation initiation to help relieve the unfolded protein
(eIF-2␣), which prevents eIF-2␣ from initiating translation, thereby limit- burden on the ER. Recent studies have also demonstrated
ing the unfolded protein load on the ER. Activation of the PERK pathway
has also been shown to mediate the transcriptional activation of ER stress
that PERK-dependent eIF-2␣ phosphorylation is required
response genes. for transcriptional activation of a wide range of UPR-re-
sponsive genes [75,76]. As a result, the UPR co-ordinately
enhances cell survival by ensuring that the adverse effects
sized proteins and to prevent aggregation of folding inter- of ER stress are dealt with in a timely and efficient manner.
mediates, the ER contains a high concentration of molecular Failure to elicit a functional UPR following ER stress can
chaperones including the 78-kDa glucose-regulated protein result in programmed cell death via specific ER-dependent
(GRP78), GRP94, calnexin, calreticulin, protein disulphide pathways and contribute to the pathogenesis of a number of
isomerase and ERp72. These chaperones act as a quality human diseases, including diabetes, Alzheimer’s disease,
control system by ensuring that only correctly folded pro- Parkinson’s disease and cancer [71].
teins are allowed to enter the Golgi for further processing A recently proposed mechanism of vascular injury in-
and secretion. The observation that these chaperones are volves the ability of homocysteine to cause ER stress by
induced in response to a variety of pathological agents disrupting disulphide bond formation and causing misfold-
and/or conditions that cause ER stress [70] suggests that ing of proteins traversing the ER [65,66,77–79]. We as well
they play a critical role in quality processes during protein as others have previously reported that elevated levels of
folding and processing. intracellular homocysteine increase the expression of sev-
ER stress results in the activation of the unfolded protein eral ER stress response genes, including GRP78, GRP94,
response (UPR), an intracellular signaling pathway essential Herp and RTP [65,66,77,78,80]. In addition, homocysteine
for survival of cells undergoing ER stress (Figure 2). In induces the expression of GADD153 [65,66,80], a basic
mammalian cells, the UPR is mediated via three ER-resi- region leucine zipper transcription factor involved in ER
dent sensors: a type-I ER transmembrane protein kinase stress-induced cell death [81]. These effects in gene expres-
(IRE-1), the activating transcription factor 6 (ATF-6) and sion directly involve the UPR because homocysteine treat-
the PKR like ER kinase (PERK). Activation of these three ment has been shown to induce the activation of both PERK
pathways is mediated by GRP78, which is bound to each [82,83] and IRE-1 [84,85].
sensor in the absence of ER stress. During the accumulation Transient and chronic ER stress elicited by homocysteine
of unfolded proteins in the ER, GRP78 dissociates from its has been shown to adversely affect several cellular func-
respective sensor to interact with unfolded proteins, thereby tions involved in the development and progression of ath-
A.B. Lawrence de Koning et al. / Clinical Biochemistry 36 (2003) 431– 441 435

erosclerosis, including lipid dysregulation, programmed cell as well as their in vivo relevance to the development and
death and inflammation. We have recently demonstrated progression of atherosclerosis.
that homocysteine-induced ER stress causes dysregulation Previous work by Pahl and colleagues [97–100] has
of lipid biosynthesis by activating the SREBPs [80], ER- demonstrated that the in vitro activation of NF-␬B occurs in
resident transcription factors responsible for the induction cells undergoing ER stress. This suggests that homocys-
of genes in the cholesterol/triglyceride biosynthesis and teine-induced ER stress could contribute to the development
uptake pathways [86,87]. The observation that stable over- of atherosclerosis by directly enhancing an inflammatory
expression of GRP78, which protects cells from agents response via NF-␬B activation. Recent studies have now
and/or conditions known to cause ER stress, inhibits homo- demonstrated that HHcy enhances activation of NF-␬B in
cysteine-induced cholesterol gene expression in cultured atherosclerotic lesions and in selective tissues from apoE-
human cells further supports an association between ER deficient mice having diet-induced HHcy [47]. Although the
stress and lipid metabolism [80]. Additional studies have mechanism responsible for this proinflammatory effect of
demonstrated that activation of the UPR increases lipid HHcy is unknown, homocysteine-induced ER stress could
biosynthesis in yeast [88,89] and dolichol biosynthesis [90], enhance the phosphorylation and activation of JNK, thereby
which is a component of the cholesterol biosynthesis path- leading to further inflammation and programmed cell death
way in cultured mammalian cells. Our findings suggest that [92]. Based on these recent findings, homocysteine-induced
activation of the UPR by homocysteine promotes the over- ER stress may prove to be an important mechanism that not
production of ER lipid components, including cholesterol only contributes to the development of atherosclerosis but
and triglycerides, resulting in the accumulation of lipids in also influences other specific diseases, including diabetes
affected cell types, including hepatocytes, smooth muscle and hypertension [71].
cells and macrophages. This dysregulation in lipid biosyn-
thesis and uptake may explain the paradox as to why lipid- 4.4. Additional mechanisms
rich atherosclerotic lesions develop in hyperhomocysteine-
mic patients with normal serum lipid profiles. In addition to the established mechanisms described
Recent studies have now shown that homocysteine in- above, other studies have suggested that elevated levels of
duces programmed cell death in cultured human vascular homocysteine can elicit other effects that contribute to
endothelial cells and that this effect involves activation of atherogenesis and thrombotic disease. It has been reported
the UPR [85]. Homocysteine-induced cell death was mim- that homocysteine stimulates the proliferation of cultured
icked by other ER stress agents and was dependent on vascular smooth muscle cells [101–103] and upregulates
IRE-1 signaling. Activation of IRE-1 by homocysteine smooth muscle cell collagen [104], findings consistent with
leads to a rapid and sustained activation of JNK protein the observation that patients with severe HHcy have wide-
kinases [91], a result consistent with the finding that acti- spread premature atherosclerosis with intimal thickening
vation of JNK by ER stress involves binding of IRE-1 to and collagen-rich, fibrous lesions [15]. Several potential
TRAF2 [92]. Because persistent activation of JNK corre- mechanisms have also been proposed to explain endothelial
lates with cell death [93], these studies provide further cell dysfunction during HHcy, including direct cell injury
support for a mechanism involving homocysteine-induced [50,56], growth arrest [65] and altered cellular methylation
programmed cell death. In addition, caspase-3 or a caspase- status [105]. In addition to causing endothelial cell dysfunc-
like protease was shown to be essential for homocysteine- tion, elevated levels of homocysteine have been shown to
induced programmed cell death, a result consistent with the increase the procoagulant activity of cultured endothelial
ability of homocysteine thiolactone, a cyclic thioester de- cells by enhancing tissue factor [106 –108] and factor V
rivative of homocysteine synthesized by specific aminoacyl- [109] activities, inhibiting cell surface thrombomodulin
tRNA synthetases [94], to induce programmed cell death in [110] and protein C [111], and impairing tissue plasmino-
HL-60 cells [95]. Although caspase-7 mediated caspase-12 gen activator binding to its endothelial cell receptor, an-
activation has been implicated in the coupling of ER stress nexin II [112].
to programmed cell death [96], there are presently no re- Homocysteine thiolactone, the cyclic thioester of homo-
ported studies examining the effect of homocysteine on the cysteine, has recently received attention for its potential role
activation of these caspases. Given that programmed cell in atherosclerosis and thrombotic disease [94,113]. In addi-
death has been widely documented to occur in animal and tion to the transsulphuration and remethylation pathways,
human atherosclerotic lesions, and that programmed cell intracellular homocysteine can be converted by methionyl-
death and ER stress are increased in atherosclerotic lesions tRNA synthase into a homocysteine-AMP complex. This
from mice fed hyperhomocysteinemic diets (Zhou and Aus- complex is subsequently catabolised to homocysteine thio-
tin, unpublished results), it is tempting to suggest that ho- lactone, thereby preventing the incorporation of homocys-
mocysteine could adversely affect the stability and/or teine into nascent polypeptide chains. However, homocys-
thrombogenicity of atherosclerotic lesions through its abil- teine thiolactone has unique reactive properties that can lead
ity to elicit ER stress. Further studies are, however, required to the homocysteinylation of lysine residues and free amine
to identify the pro-apoptotic factors involved in this process groups on numerous cellular proteins, thereby resulting in
436 A.B. Lawrence de Koning et al. / Clinical Biochemistry 36 (2003) 431– 441

decreased biological activity and premature degradation early atherosclerotic lesions [119]. This is significant, given
[113]. In addition, homocysteine thiolactone secreted into that plasma lipid levels are normal in these animals. Al-
the circulation may induce widespread modification of though the arterial lipid inclusions are not as elaborate as
plasma proteins that could potentially contribute to the de- lesions observed in murine models of atherosclerosis, such
velopment of atherosclerosis and thrombotic disease. Re- as apoE-deficient or LDL receptor-deficient mice, they are
cent studies have demonstrated that homocysteine thiolac- the first observations showing that mild HHcy is involved in
tone decreases paraoxonase activity associated with HDL, the formation of vascular lesions.
thereby rendering HDL less protective against oxidative Studies in our laboratory examined the molecular and
damage and against toxicity of homocysteine thiolactone cellular consequences of diet-induced HHcy in wild-type
[114]. Additional studies using physiologically relevant in and heterozygous CBS-deficient mice [80]. Both mild and
vivo models of HHcy are necessary to further understand the severe HHcy were shown to induce ER stress and abnormal
atherogenic role of homocysteine thiolactone. hepatic accumulation of lipid in each strain of mice. ER
stress was shown to promote SREBP activation, thereby
resulting in increased expression of enzymes encoding
5. Animal models of hyperhomocysteinemia genes for cholesterol and triglyceride biosynthesis, as well
as the LDL receptor. SREBP dysregulation resulted in he-
Recent findings from genetic- and diet-induced animal patic steatosis, a characteristic feature observed in human
models of HHcy have significantly enhanced the status of homocystinuric patients. Interestingly, plasma lipid levels
homocysteine as a risk factor for atherosclerosis. They have remained unchanged despite increases in hepatic VLDL-
also supported and extended many of the proposed in vitro triglyceride secretion rates. These observations suggest that
mechanisms and have provided a more physiological per- hepatic steatosis in HHcy does not result from impaired
spective on the role of homocysteine in the induction of lipid export, but instead is due to increased lipid biosynthe-
cardiovascular disease. sis coupled with enhanced uptake of LDL by the liver.
Manipulation of plasma homocysteine can be accom- Should this effect of homocysteine on lipid metabolism and
plished by dietary and/or genetic approaches. The addition uptake be confirmed in vascular cells involved in athero-
of methionine and/or depletion of B vitamins and folate in genesis, this could potentially explain the lipid rich arterial
the diet can be used to induce mild to severe HHcy. Genet- lesions associated with mild HHcy in patients with normal
ically engineered mice deficient in CBS or MTHFR have lipid profiles.
also been used as animal models of HHcy. Homozygous One of the earliest detectable cellular responses in the
CBS-deficient mice have total plasma homocysteine levels development of atherosclerotic lesions is the binding of
40 times greater than normal and suffer from severe growth monocytic cells to the vascular endothelium. Recent studies
retardation, hepatic steatosis and ectopia lentis – phenotypic have now demonstrated that the number of monocytes
changes also observed in patients with homozygous CBS present on the surface of aortic endothelium is significantly
deficiency [115]. Heterozygous CBS-deficient mice, how- elevated in rats with diet-induced HHcy, although typical
ever, do not suffer from the same developmental defects as atherosclerotic lesions were not observed [120]. A signifi-
homozygotes. These animals have twice the normal concen- cant increase in the expression of MCP-1, VCAM-1 and
tration of plasma homocysteine, making them ideal models E-selectin was also observed in the aortic endothelium of
to study mild HHcy. Recently, the chronic, mild HHcy hyperhomocysteinemic rats, thereby providing a potential
present in heterozygous CBS-deficient mice has been shown cellular mechanism responsible for the increase in mono-
to cause endothelial dysfunction by decreasing vascular NO cyte binding and recruitment. The additional observation
bioavailability, thereby leading to impaired vasorelaxation that folate supplementation prevented an increase in plasma
[48,52]. This association is not unique to CBS-deficient homocysteine levels, decreased monocyte binding to the
mice, as endothelial dysfunction has also been shown in endothelium and inhibited the expression of MCP-1,
rabbits and cynomolgus monkeys (Macaca fascicularis) VCAM-1 and E-selectin, emphasizes a potential antiathero-
having diet-induced HHcy [53,116 –118]. Although CBS- genic effect of lowering plasma homocysteine.
deficient mice and nonmurine models of HHcy exhibit en- Since the development and progression of atherosclero-
dothelial dysfunction, it is important to note that they do not sis is limited in some animal models of HHcy, we and other
develop atherosclerosis [115–118]. researchers have generated dietary HHcy in genetically al-
MTHFR-deficient mice have been recently developed to tered mice prone to developing atherosclerosis. An estab-
examine the effects of HHcy resulting from genetic defi- lished model of spontaneous atherosclerosis is the apoE-
ciencies in the remethylation pathway [119]. Although deficient mouse [121]. These mice develop
MTHFR-deficient mice share basic phenotypic similarities hypercholesterolemia because of impaired reverse choles-
with CBS-deficient mice, they are unique in ways that may terol transport to the liver, thereby leading to the accumu-
predispose them to developing atherosclerosis. Mature het- lation of cholesterol-rich remnants in plasma. This persis-
erozygous and homozygous MTHFR-deficient mice accu- tent and severe hypercholesterolemia (up to 5 times normal)
mulate lipid in their proximal aorta which is reminiscent of leads to the premature development of complex atheroscle-
A.B. Lawrence de Koning et al. / Clinical Biochemistry 36 (2003) 431– 441 437

NF-␬B and enhance vascular inflammatory processes may


in part be related to its ability to cause ER stress [97–99].
Our finding that markers of ER stress are increased in the
livers [80] and atherosclerotic lesions (Zhou and Austin,
unpublished results) of mice with diet-induced HHcy pro-
vides support for this concept. Lesions with increased ex-
pression of the collagen degrading matrix metalloprotein-
ase-9 were also observed, which could potentially promote
lesion instability and rupture. The increased collagen in the
lesions observed by Zhou et al. [122] is in contrast to the
prediction of plaque instability by Hofmann et al. [47]. Both
studies, however, are in agreement that diet induced HHcy
in the presence of a hyperlipidemic state accelerates the
development of atherosclerosis. A recent study by Wang et
al. [124], using apoE and CBS double knockout mice,
showed that atherosclerotic lesion area and lesion lipid
content increased with plasma homocysteine concentra-
tions, independent of diet. Overall, these data strongly sug-
gest that HHcy amplifies proatherogenic cellular processes
when combined with hyperlipidemia. As previously men-
Fig. 3. Atherosclerotic lesions in the aortic root of mice fed control (A),
high homocysteine (B) or high methionine diet (C). Lesion sizes in the
tioned, case-control epidemiological studies seem to impli-
hyperhomocysteinemic mice (B, C) were significantly larger, compared to cate HHcy in compounding the overall risk of cardiovascu-
the control mice (A). Atrophy of the tunica media and rupture of the elastic lar disease when in the presence of additional risk factors
laminae were often observed (arrow in B). All sections were stained with such as hypertension and diabetes [35,37,38]. Supporting
Orcein to reveal the elastic lamina. Data were kindly provided by Drs. Ji this hypothesis, Matthias et al. [125] showed that sponta-
Zhou and Erling Falk.
neously hypertensive rats fed high doses of methionine
developed more complex arterial lesions compared to rats
rotic lesions [121]. Two separate studies published in the fed normal diets.
last two years demonstrate that mild HHcy accelerates Collectively, these studies have helped elucidate the
atherogenesis in apoE-deficient mice. Zhou et al. [122] causal role of homocysteine in the development of endothe-
showed that apoE-deficient mice fed methionine- or homo- lial dysfunction and atherosclerosis. Furthermore, these an-
cysteine-enriched diets developed larger, more complex and imal models are valuable in vivo tools to further examine
more numerous arterial lesions, compared to mice fed con- potential therapeutic approaches in lowering plasma homo-
trol diets (Figure 3). An important feature of these patho- cysteine while decreasing the prevalence of cardiovascular
logical changes was that they occurred without a significant disease.
elevation in the concentration of plasma lipids. Many of
these lesions were rich in collagen, probably owing to the
ability of homocysteine to stimulate SMC proliferation and 6. Conclusions and questions
extracellular matrix deposition [104,123]. Zhou et al. fur-
ther demonstrated that the differences in lesion size and HHcy is an independent risk factor for cardiovascular
frequency were seen only between groups following 3 disease. Recent studies have identified three major cellular
months of dietary treatment compared to 12 months, sug- mechanisms by which HHcy may contribute to the devel-
gesting that HHcy mediates the early stages of atherogene- opment of endothelial dysfunction and atherosclerosis.
sis [122]. In a separate study using apoE-deficient mice fed They include the induction of pro-inflammatory factors,
diets supplemented with methionine to induce HHcy, Hof- oxidative stress, and ER stress. Genetic- and diet-induced
mann et al. [47] found significant increases in atheroscle- animal models of HHcy have now demonstrated a causal
rotic lesion area, compared to mice fed control diets. Fur- relationship between HHcy and accelerated atherosclerosis.
thermore, diet-induced HHcy promoted NF-␬B activation in They have also provided important insights into the role of
aortic tissue, leading to the increased expression of the HHcy in endothelial dysfunction and the development of
pro-inflammatory adhesion molecule VCAM-1 as well as atherosclerosis. These studies, however, raise some inter-
the pro-inflammatory mediators RAGE and EN-RAGE. In- esting and relevant questions. Although HHcy seems to be
creased expression of pro-inflammatory genes in the aorta very important in early lesion development, what role does
of hyperhomocysteinemic animals is also consistent with it play in plaque stability and/or thrombogenicity? Can
the finding of increased plasma concentrations of TNF-␣ HHcy, in the absence of hypercholesterolemia or other sig-
and increased numbers of lesion-resident macrophages. As nificant risk factors, enhance lesion development in hu-
discussed previously, the ability of homocysteine to activate mans? Does dietary enrichment of vitamins essential for the
438 A.B. Lawrence de Koning et al. / Clinical Biochemistry 36 (2003) 431– 441

metabolism of homocysteine protect against cardiovascular [13] Clarke R, Daly L, Robinson K, et al. Hyperhomocysteinemia: an
disease? Answering these and other important questions independent risk factor for vascular disease. N Engl J Med 1991;
324:1149 –55.
will certainly enhance our understanding of the role of
[14] den Heijer M, Koster T, Blom HJ, et al. Hyperhomocysteinemia as
HHcy in the development and progression of atherosclero- a risk factor for deep-vein thrombosis. N Engl J Med 1996;334:
sis. 759 – 62.
[15] McCully K.S. Homocysteine and vascular disease. Nat Med 1996;
2:386 –9.
[16] Mudd SH, Levy HL, Skovby F. Disorders of transsulfation. In:
Acknowledgments Scriver CR, Beadet AL, Sly WS, Vallee D, editors. The Metabolic
Basis for Inherited Disease. New York, NY: McGraw-Hill, 1989. p.
The author’s work is supported by Research Grants from 693.
the Heart and Stroke Foundation of Ontario (T-4005) and [17] Selhub J, Jacques PF, Bostom AG, et al. Association between
the Canadian Institutes of Health Research (MOP-49417). plasma homocysteine concentrations and extracranial carotid-artery
stenosis. N Engl J Med 1995;332:286 –91.
R.C. Austin is a Career Investigator of the Heart and Stroke [18] Ueland PM, Refsum H, Beresford SAA, Vollset SE. The contro-
Foundation of Ontario. We apologize to many of our col- versy over homocysteine and cardiovascular risk. Am J Clin Nutr
leagues whose work could not be directly acknowledged 2000;72:324 –32.
due to space limitations. [19] Welch GN, Loscalzo JN. Homocysteine and atherothrombosis.
N Engl J Med 1998;338:1042–50.
[20] Wilcken DEL, Dudman NPB. Homocystinuria and atherosclerosis.
In: Lusis AJ, Rotter JI, Sparkes RS, editors. Molecular genetics of
Note added to proof coronary artery disease; candidate genes and process in atheroscle-
rosis. Monograms in human genetics. New York, NY: Karger, 1992.
Recent studies by Ji and Kaplowitz [126] have demon- p. 311.
strated increased HHcy, ER stress and liver injury in an [21] Mudd SH, Havlik R, Levy HL, McKusick VA, Feinleib M. Cardio-
vascular risk in heterozygotes for homocystinuria. Am J Hum Genet
alcohol-fed mouse model of HHcy. 1982;34:1018 –21.
[22] Ueland PM, Refsum H. Plasma homocysteine, a risk factor for
vascular disease: plasma levels in health, disease, and drug therapy.
References J Lab Clin Med 1989;114:473–501.
[23] McCully KS. Vascular pathology of homocysteinemia: implications
for the pathogenesis of arteriosclerosis. Am J Pathol 1969;56:111–
[1] Ross R. Atherosclerosis-an inflammatory disease. N Engl J Med
28.
1999;340:115–26.
[24] Frosst P, Blom HJ, Milos R, et al. A candidate genetic risk factor for
[2] Ross R. The pathogenesis of atherosclerosis: a perspective for the
vascular disease: a common mutation in methylenetetrahydrofolate
1990s. Nature 1993;362:801–9.
reductase. Nat Genet 1995;10:111–3.
[3] Berliner JA, Navab M, Gofelman AM, et al. Atherosclerosis: basic
[25] Goyette P, Sumner JS, Milos R, et al. Human methylenetetrahydro-
mechanisms. Oxidation, inflammation, and genetics. Circulation
folate reductase: isolation of cDNA, mapping and mutation identi-
1995;91:2488 –96.
fication. Nat Genet 1994;7:195–200.
[4] Navab M, Berliner JA, Watson AD, et al. The Yin and Yang of
[26] Rosenblatt DS. Inherited disorders of folate transport and metabo-
oxidation in the development of the fatty streak. Arterioscler
lism. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The
Thromb Vasc Biol 1996;16:831– 42.
[5] Spady DK. Reverse cholesterol transport and atherosclerosis regres- Metabolic Basis of Inherited Disease 6th edition. New York, NY:
sion. Circulation 1999;100:576 – 8. McGraw-Hill, 1989. p. 2049.
[6] Stary HC, Chandler AB, Dinsmore RE, et al. A definition of ad- [27] Franken DG, Boers GHJ, Blom HJ, Trijibels FJM, Kloppenborg
vanced types of atherosclerotic lesions and a histological classifica- PW. Treatment of mild hyperhomocysteinemia in vascular disease
tion of atherosclerosis. A report from the Committee on Vascular patients. Arterioscler Thromb 1994;14:465–70.
Lesions of the Council on Arteriosclerosis, Am Heart Association. [28] Selhub J, Jacques PF, Wilson PWF, Rush D, Rosenberg IH. Vitamin
Circulation 1995;92:1355–74. status and intake as primary determinants of homocysteinemia in an
[7] Stary HC, Chandler AB, Glagov S, et al. A definition of initial, fatty elderly population. JAMA 1995;270:2693– 8.
streak, and intermediate lesions of atherosclerosis. A report from the [29] Hustad S, Ueland PM, Vollset SE, et al. Riboflavin as a determinant
Committee on Vascular Lesions of the Council on Arteriosclerosis, of plasma total homocysteine: effect modification by the methyl-
Am Heart Association. Circulation 1994;89:2462–78. enetetrahydrofolate reductase C677T polymorphism. Clin Chem
[8] Avogaro P, Bon GB, Cazzolato G, et al. Presence of a modified low 2000;46:1065–71.
density lipoprotein in humans. Arteriosclerosis 1988;8:79 – 87. [30] Ubbink JB, van der Merwe A, Delport R. The effect of a subnormal
[9] Palinski W, Rosenfeld ME, Yla-Herttuala S, et al. Low density vitamin B-6 status on homocysteine metabolism. J Clin Invest 1996;
lipoprotein undergoes oxidative modification in vivo. Proc Natl 98:177– 84.
Acad Sci USA 1989;86:1372– 6. [31] Ubbink JB, Vermaak WJ, van der Merwe A, Becker PJ. Vitamin
[10] Suarna C, Dean RT, May J, Stocker R, et al. Human atherosclerotic B-12, vitamin B-6, and folate nutritional status in men with hyper-
plaque contains both oxidized lipids and relatively large amounts of homocysteinemia. Am J Clin Nutr 1993;57:47–53.
alpha-tocopherol and ascorbate. Arterioscler Thromb Vasc Biol [32] Bostom AG, Jacques PF, Liaugaudas G, Rogers G, Rosenberg IH,
1995;15:1616 –24. Selhub J. Total homocysteine lowering treatment among coronary
[11] Yla-Herttuala S, Palinski W, Rosenfeld ME, et al. Evidence for the artery disease patients in the era of folic acid-fortified cereal grain
presence of oxidatively modified low density lipoprotein in athero- flour. Arteriocler Thromb Vasc Biol 2002;22:488 –91.
sclerotic lesions of rabbit and man. J Clin Invest 1989;84:1086 –95. [33] Rader, JI. Folic acid fortification, folate status and plasma homo-
[12] Lusis AJ. Atherosclerosis. Nature 2000;407:233– 41. cysteine. J Nutr 2002;132:2466S-70S.
A.B. Lawrence de Koning et al. / Clinical Biochemistry 36 (2003) 431– 441 439

[34] Anderson JL, Horne BD, Carlquist JF, et al. Effect of implementa- [56] Starkebaum G, Harlan JM. Endothelial cell injury due to copper-
tion of folic acid fortification of food on homocysteine concentra- catalyzed hydrogen peroxide generation from homocysteine. J Clin
tions in subjects with coronary artery disease. Am J Cardiol 2002; Invest 1986;77:1370 –76.
90:536 –9. [57] Jacobsen DW. Cellular mechanisms of homocysteine pathogenesis
[35] Refsum H, Ueland PM, Nygard O, Vollset SE. Homocysteine and in atherosclerosis. In: Jacobsen DW, editor. Homocysteine in health
cardiovascular disease. Annu Rev Med 1998;49:31– 62. and disease. New York, NY: Cambridge University Press, 2001. p.
[36] Brattstrom L, Wilcken DEL. Homocysteine and cardiovascular dis- p. 425–50.
ease: cause or effect? Am J Clin Nutr 2000;72:315–23. [58] Jacobsen DW. Hyperhomocysteinemia and oxidative stress: time for
[37] Eikelboom JW, Lonn E, Genest J, Hanjey G, Yusuf S. Homocys- a reality check? Arterioscler Thromb Vasc Biol 2000;20:1182– 4.
t(e)ine and cardiovascular disease: a critical review of the epidemi- [59] Sengupta S, Wehbe C, Majors AK, Ketterer ME, DiBello PM,
ologic evidence. Ann Intern Med 1999;131:363–75. Jacobsen DW. Relative roles of albumin and ceruloplasmin in the
[38] Graham IM, Daly LE, Refsum HM, et al. Plasma homocysteine as formation of homocystine, homocysteine-cysteine-mixed disulfide,
a risk factor for vascular disease. The European Concerted Action and cysteine in circulation. J Biol Chem 2001;276:46896 –904.
Project. JAMA 1997;277:1775– 81. [60] Zappacosta B, Mordente A, Persichilli S, et al. Is homocysteine a
[39] The homocysteine studies collaboration. Homocysteine and risk of pro-oxidant? Free Radic Res 2001;35:499 –505.
ischemic heart disease and stroke: a meta analysis. JAMA 2002; [61] Duan J, Murohara T, Ikeda T, et al. Hyperhomocysteinemia impairs
288:2015–22. angiogenesis in response to hindlimb ischemia. Arterioscler Thromb
[40] Cattaneo M. Hyperhomocysteinemia, atherosclerosis, and thrombo- Vasc Biol 2000;20:2579 – 85.
sis. Thromb Haemost 1999;81:165–76. [62] Franken DG, Boers GHJ, Blom HJ, Trijibels FJM, Kloppenborg
[41] McCully KS. The biomedical significance of homocysteine. J Sci PW. Treatment of mild hyperhomocysteinemia in vascular disease
Expl 2001;15:5–20. patients. Arterioscler Thromb 1994;14:465–70.
[42] Scott JM. Homocysteine and cardiovascular risk. Am J Clin Nutr [63] Gryglewski RJ, Palmer RM, Moncada S. Superoxide anion is in-
2000;72:333– 4. volved in the breakdown of endothelium-derived vascular relaxing
[43] Poddar R, Sivasubramanian N, Dibello PM, Robinson K, Jacobsen factor. Nature 1986;320:454 – 6.
D. Homocysteine induces expression and secretion of monocyte [64] Heinecke JW, Rosen H, Suzuki LA, Chait A. The role of sulfur-
chemoattractant protein-1 and interleukin-8 in human aortic endo- containing amino acids in superoxide production and modification
of low density lipoprotein by arterial smooth muscle cells. J Biol
thelial cells: implications for vascular disease. Circulation 2001;103:
Chem 1987;262:10098 –103.
2717–3.
[65] Outinen PA, Sood SK, Pfeifer SI, et al. Homocysteine-induced
[44] Wang G, O K. Homocysteine stimulates the expression of monocyte
endoplasmic reticulum stress and growth arrest leads to specific
chemoattractant protein-1 receptor (CCR2) in human monocytes:
changes in gene expression in human vascular endothelial cells.
possible involvement of oxygen free radicals. Biochem J 2001;357:
Blood 1999;94:959 – 67.
233– 40.
[66] Outinen PA, Sood SK, Liaw PCY, et al. Characterization of the
[45] Wang G, Siow YL, O K. Homocysteine induces monocyte chemoat-
stress-inducing effects of homocysteine. Biochem J 1998;332:213–
tractant protein-1 expression by activating NF-kappa B in THP-1
21.
macrophages. Am J Physiol Heart Circ Physiol 2001;280:H2840 –7.
[67] Upchurch GR, Welch GN, Fabian AN, et al. Homocyst(e)ine de-
[46] Collins T, Cybulsky MI. NF-kappaB: pivotal mediator or innocent
creases bioavailable nitric oxide by a mechanism involving gluta-
bystander in atherogenesis? J Clin Invest 2001;107:255– 64.
thione peroxidase. J Biol Chem 1997;272:17012–7.
[47] Hofmann MA, Lalla E, Lu Y, et al. Hyperhomocysteinemia en-
[68] Tsao PS, Wang B, Buitrago R, Shyy JY, Cooke JP. Nitric oxide
hances vascular inflammation and accelerates atherosclerosis in a regulates monocyte chemotactic protein-1. Circulation 1997;96:
murine model. J Clin Invest 2001;107:675– 83. 934 – 40.
[48] Eberhardt RT, Forgione MA, Cap A, et al. Endothelial dysfunction [69] Ron D. Translational control in the endoplasmic reticulum stress
in a murine model of mild hyperhomocyst(e)inemia. J Clin Invest response. J Clin Invest 2002;110:1383– 8.
2000;106:483–91. [70] Lee AS. The glucose-regulated proteins: stress induction and clinical
[49] Lentz SR, Sobey CG, Piegors DJ, et al. Vascular dysfunction in applications. Trends Biochem Sci 2001;26:504 –10.
monkeys with diet-induced hyperhomocyst(e)inemia. J Clin Invest [71] Kaufman RJ. Orchestrating the unfolded protein response in health
1996;98:24 –9. and disease. J Clin Invest 2002;110:1389 –98.
[50] Loscalzo J. The oxidant stress of hyperhomocyst(e)inemia. J Clin [72] Shen J, Chen X, Hendershot L, Prywes R. ER stress regulation of
Invest 1996;98:5–7. ATF6 localization by dissociation of BiP/GRP78 binding and un-
[51] Tawakol A, Omland T, Gerhard M, Wu JT, Creager MA. Hyper- masking of golgi localization signals. Dev Cell 2002;3:99 –111.
homocyst(e)inemia is associated with impaired endothelium-depen- [73] Liu CY, Schroder M, Kaufman RJ. Ligand-independent dimeriza-
dent vasodilation in humans. Circulation 1997;95:1119 –21. tion activates the stress response kinases IRE1 and PERK in the
[52] Weiss N, Heydrick S, Zhang YY, Bierl C, Cap A, Loscalzo J. lumen of the endoplasmic reticulum. J Biol Chem 2000;275:
Cellular redox state and endothelial dysfunction in mildly hyperho- 24881–5.
mocysteinemic cystathionine beta-synthase-deficient mice. Arterio- [74] Liu CY, Wong HN, Schauerte JA, Kaufman RJ. The protein kinase/
scler Thromb Vasc Biol 2002;22:34 – 41. endoribonuclease IRE1a that signals the unfolded protein response
[53] Lang D, Kredan MB, Moat SJ, et al. Homocysteine-induced inhi- has a lumenal amino-terminal ligand-independent dimerization do-
bition of endothelium-dependent relaxation in rabbit aorta: role for main. J Biol Chem 2002;277:18346 –56.
superoxide anions. Arterioscler Thromb Vasc Biol 2000;20:422–7. [75] Scheuner D, Song B, McEwen E, et al. Translational control is
[54] Chambers JC, McGregor A, Jean-Marie J, Mbeid OA, Kooner JS. required for the unfolded protein response and in vivo glucose
Demonstration of rapid onset vascular endothelial dysfunction after homeostasis. Molecular Cell 2001;7:1165–76.
hyperhomocysteinemia: an effect reversible with vitamin C therapy. [76] Harding HP, Novoa I, Zhang Y, et al. Regulated translational initi-
Circulation 1999;99:1156 – 60. ation controls stress-induced gene expression in mammalian cells.
[55] Chambers JC, Ueland PM, Obeid OA, Wrigley J, Refsum H, Kooner Molecular Cell 2000;6:1099 –1108.
JS. Improved vascular endothelial function after oral B vitamins: an [77] Kokame K, Agarwala KL, Kato H, Miyata T. Herp, a new ubiquitin-
effect mediated through reduced concentrations of free plasma ho- like membrane protein induced by endoplasmic reticulum stress.
mocysteine. Circulation 2000;102:2479 – 83. J Biol Chem 2000;275:32846 –53.
440 A.B. Lawrence de Koning et al. / Clinical Biochemistry 36 (2003) 431– 441

[78] Kokame K, Kato H, Miyata T. Homocysteine-respondent genes in [97] Pahl HL, Sester M, Burgert HG, Baeuerle PA. Activation of tran-
vascular endothelial cells identified by differential display analysis. scription factor NF-kappa B by the adenovirus E3/19K protein
GRP78/BiP and novel genes. J Biol Chem 1996;271:29659 – 65. requires its ER retention. J Cell Biol 1996;132:511–22.
[79] Ron D. Hyperhomocysteinemia and function of the endoplasmic [98] Pahl HL, Baeuerle PA. Expression of influenza virus hemagglutinin
reticulum. J Clin Invest 2001;107:1221–2. activates transcription factor NF-kappa B. J Virol 1995;69:1480 – 4.
[80] Werstuck GH, Lentz SR, Dayal S, et al. Homocysteine-induced [99] Pahl HL, Baeuerle PA. A novel signal transduction pathway from
endoplasmic reticulum stress causes dysregulation of the cholesterol the endoplasmic reticulum to the nucleus is mediated by transcrip-
and triglyceride biosynthetic pathways. J Clin Invest 2001;107: tion factor NF-kappa B. EMBO J 1995;14:2580 – 8.
1263–73. [100] Meyer MWH, Caselmann V, Schluter R, Schreck PH, Hofschneider
[81] Zinszner H, Kuroda M, Wang XZ, et al. CHOP is implicated in PH, Baeuerle PA. Hepatitis B virus transactivator MHBst: activation
programmed cell death in response to impaired function of the of NF-kappa B, selective inhibition by antioxidants and integral
endoplasmic reticulum. Genes Dev 1998;12:982–95. membrane localization. EMBO J 1992;11:2991–3001.
[82] Ma K, Vattem KM, Wek RC. Dimerization and release of molecular [101] Taha S, Azzi A, Ozner NK. Homocysteine induces DNA synthesis
chaperone inhibition facilitate activation of eukaryotic initiation and proliferation of vascular smooth muscle cells by a hydrogen
factor-2 kinase in response to endoplasmic reticulum stress. J Biol peroxide-independent mechanism. Antioxid Redox Signal 1999;1:
Chem 2002;277:18728 –35. 365-9.
[83] Nonaka K, Tsujino T, Watari Y, Emoto N, Yokoyama M. Taurine [102] Tsai JC, Perrella MA, Yoshizumi M, et al. Promotion of vascular
prevents the decrease in expression and secretion of extracellular smooth muscle cell growth by homocysteine: a link to atheroscle-
superoxide dismutase induced by homocysteine: amelioration of rosis. Proc Natl Acad Sci 1994;91:6369 –73.
homocysteine-induced endoplasmic reticulum stress by taurine. Cir- [103] Tsai JC, Wang H, Perrella MA, et al. Induction of cyclin A gene
culation 2001;104:1165–70. expression by homocysteine in vascular smooth muscle cells. J Clin
[84] Zhang C, Kawauchi J, Adachi MT, et al. Activation of JNK and Invest 1996;97:146 –53.
transcriptional repressor ATF3/LRF1 through the IRE1/TRAF2 [104] Majors AK, Sengupta S, Jacobsen DW, Pyeritz RE. Upregulation of
pathway is implicated in human vascular endothelial cell death by smooth muscle cell collagen production by homocysteine-insight
homocystiene. Biochem Biophys Res Commun 2001;289:719 –24. into the pathogenesis of homocystinuria. Mol Genet Metab 2002;
[85] Zhang C, Cai Y, Adachi MT, et al. Homocysteine induces pro- 76:92–9.
grammed cell death in human vascular endothelial cells through [105] Lee ME, Wang H. Homocysteine and hypomethylation-a novel link
activation of the unfolded protein response. J Biol Chem 2001;276: to vascular disease. Trends Cardiovasc Med 1999;9:49 –54.
35867–74. [106] Khajuria A, Houston DS. Induction of monocyte tissue factor ex-
[86] Shimano H, Horton JD, Hammer RE, et al. Overproduction of pression by homocysteine: a possible mechanism for thrombosis.
cholesterol and fatty acids causes massive liver enlargement in Blood 2000;96:966 –72.
transgenic mice expressing truncated SREBP-1a. J Clin Invest 1996; [107] Fryer RH, Wilson BD, Gubler DB, Fitzgerald LA, Rodgers GM.
98:1575– 84. Homocysteine, a risk factor for premature vascular disease and
[87] Horton JD, Shimano H, Hamilton RL, Brown MS, Goldstein JL. thrombosis, induces tissue factor activity in endothelial cells. Arte-
Disruption of LDL receptor gene in transgenic SREBP-1a mice rioscler Thromb 1993;13:1327–33.
unmasks hyperlipidemia resulting from production of lipid-rich [108] Durand P, Lussier-Cacan S, Blache D. Acute methionine load-
VLDL. J Clin Invest 1999;103:1067–76. induced hyperhomocysteinemia enhances platelet-aggregation,
[88] Cox JS, Chapman RE, Walter P. The unfolded protein response thromboxane biosynthesis, and macrophage-derived tissue factor
coordinates the production of endoplasmic reticulum protein and activity in rats. FASEB J 1997;11:1157– 68.
endoplasmic reticulum membrane. Mol Biol Cell 1997;8:1805–14. [109] Rodgers GM, Kane WH. Activation of endogenous factor V by a
[89] Travers KJ, Patil CK, Wodicka L, Lockhart DJ, Weissman JS, homocysteine-induced vascular endothelial cell activator. J Clin
Walter P. Functional and genomic analyses reveal an essential co- Invest 1986;77:1909 –16.
ordination between the unfolded protein response and ER-associated [110] Lentz SR, Sadler JE. Inhibition of thrombomodulin surface expres-
degradation. Cell 2000;101:249 –58. sion and protein C activation by the thrombogenic agent homocys-
[90] Doerrler WT, Lehrman MA. Regulation of the dolichol pathway in teine. J Clin Invest 1991;88:1906 –14.
human fibroblasts by the endoplasmic reticulum unfolded protein [111] Rodgers GM, Conn MT. Homocysteine: an atherogenic stimulus,
response. Proc Natl Acad Sci USA 1999;96:13050 –5. reduces protein C activation by arterial, and venous endothelial
[91] Cai Y, Zhang C, Nawa T, et al. Homocysteine-responsive ATF3 cells. Blood 1990;75:895–901.
gene expression in human vascular endothelial cells: activation of [112] Hajjar KA, Mauri L, Jacovina AT, et al. Tissue plasminogen acti-
c-Jun NH(2)-terminal kinase and promoter response element. Blood vator binding to the annexin II tail domain: direct modulation by
2000;96:2140 – 8. homocysteine. J Biol Chem 1998;273:9987–93.
[92] Urano F, Wang X, Bertolotti A, et al. Coupling of stress in the ER [113] Jakubowski H, Ambrosius WT, Pratt HJ. Genetic determinants of
to activation of JNK protein kinases by transmembrane protein homocysteine thiolactonase activity in humans: implications for
kinase IRE1. Science 2000;287:664 – 6. atherosclerosis. FEBS 2001;491:35–9.
[93] Chen Y-T, Meyer CF, Tan TH. Persistent activation of c-Jun N- [114] Ferretti G, Bacchetti T, Marotti E, Curatola G. Effect of homocys-
terminal kinase 1 (JNK1) in gamma radiation-induced apoptosis. teinylation on human high-density lipoproteins: a correlation with
J Biol Chem 1996;271:631– 4. paraoxonase activity. Metabolism 2003;52:146 –51.
[94] Jakubowski H. Metabolism of homocysteine thiolactone in human [115] Watanabe M, Osada J, Aratani Y, et al. Mice deficient in cystathi-
cell cultures. Possible mechanism for pathological consequences of onine beta-synthase: animal models for mild and severe homocys-
elevated homocysteine levels. J Biol Chem 1997;272:1935– 42. t(e)inemia. Proc Natl Acad Sci U S A 1995;92:1585– 89.
[95] Huang RFS, Huang SM, Lin BS, Wei JS, Liu T-Z. Homocysteine [116] Lentz SR, Erger RA, Dayal S, et al. Folate dependence of hyperho-
thiolactone induces apoptotic DNA damage mediated by increased mocysteinemia and vascular dysfunction in cystathionine beta-syn-
intracellular hydrogen peroxide and caspase 3 activation in HL-60 thase-deficient mice. Am J Physiol Heart Circ Physiol 2000;279:
cells. Life Sci 2001;68:2799 – 811. H970 –75.
[96] Rao RV, Hermel E, Castro-Obregon S, et al. Coupling endoplasmic [117] Lentz SR, Malinow RM, Piegors DJ, et al. Consequences of hyper-
reticulum stress to the cell death program. Mechanism of caspase homocyst(e)inemia on vascular function in atherosclerotic monkeys.
activation. J Biol Chem 2001;276:33869 –74. Arterioscler Thromb Vasc Biol 1997;17:2930 – 4.
A.B. Lawrence de Koning et al. / Clinical Biochemistry 36 (2003) 431– 441 441

[118] Lentz SR, Piegors DJ, Malinow RM, Heistad DD. Supplementation plaque rupture in ApoE-deficient mice. Arterioscler Thromb Vasc
of atherogenic diet with B vitamins does not prevent atheroscle- Biol 2001;21:1470 – 6.
rosis or vascular dysfunction in monkeys. Circulation 2001;103: [123] Majors A, Ehrhart LA, Pezacka EH. Homocysteine as a risk factor
1006 –11. for vascular disease. Enhanced collagen production and accumula-
[119] Chen Z, Karaplis AC, Ackerman SL, et al. Mice deficient in meth- tion by smooth muscle cells. Arterioscler Thromb Vasc Biol 1997;
ylenetetrahydrofolate reductase exhibit hyperhomocysteinemia and 17:2074 – 81.
decreased methylation capacity, with neuropathology and aortic [124] Wang H, Jiang X, Yang F, et al. Hyperhomocystinemia accelerates
lipid deposition. Hum Mol Genet 2001;10:433– 43. atherosclerosis in cystathionine ␤-synthase and apolipoprotein E
[120] Wang G, Woo CWH, Sung FL, Siow YL, OK. Increased monocyte double knockout mice with and without dietary pertubation. Blood
adhesion to aortic endothelium in rats with hyperhomocysteinemia: 2002; in press.
role of chemokine and adhesion molecules. Arterioscler Thromb [125] Matthias D, Becker CH, Riezler R, Kindling PH. Homocysteine
Vasc Biol 2002;22:1777– 83. induced arteriosclerosis-like alterations of the aorta in normotensive
[121] Zhang SH, Reddick RL, Piedrahita JA, Maeda N. Spontaneous and hypertensive rats following application of high doses of methi-
hypercholesterolemia and arterial lesions in mice lacking apoli- onine. Atherosclerosis 1996;122:201–16.
poprotein E. Science 1992;258:468 –71. [126] Ji C, Kaplowitz N. Betaine decreases hyperhomocysteinemia, endo-
[122] Zhou J, Moller J, Danielsen CC, et al. Dietary supplementation with plasmic reticulum stress, and liver injury in alcohol-fed mice. Gas-
methionine and homocysteine promotes early atherosclerosis but not troenterology 2003;124:1488-99.

Vous aimerez peut-être aussi