Você está na página 1de 7

Review TRENDS in Pharmacological Sciences Vol.22 No.

4 April 2001 195

Biological and clinical implications of


the MTHFR C677T polymorphism
Per Magne Ueland, Steinar Hustad, Jørn Schneede, Helga Refsum and Stein Emil Vollset

The enzyme methylenetetrahydrofolate reductase (MTHFR) directs folate plasma tHcy in the general population8. Its
species either to DNA synthesis or to homocysteine (Hcy) remethylation. The prevalence is related to ethnicity: the frequency of
common MTHFR C677T polymorphism affects the activity of the enzyme and homozygosity for the T allele (TT genotype) is ~10% in
hence folate distribution. Under conditions of impaired folate status, the caucasians, ~20% in some Italian populations and
homozygous TT genotype has been regarded as harmful because it is only a few per cent in Afro–Americans9.
associated with a high concentration of plasma total Hcy, increased risk of The first studies investigating the clinical
neural tube defects and colorectal neoplasias, and can also predispose relevance of the MTHFR C677T polymorphism were
individuals to adverse effects from drugs with antifolate effects. The MTHFR based on the simplistic view that this base transition
C677T polymorphism shows no consistent correlation with cardiovascular risk caused a defective enzyme leading to elevated tHcy.
and longevity but, in combination with positive folate balance, the TT genotype Accordingly, most studies focused on conditions
is associated with decreased risk of colorectal neoplasias. Because of the high known to be related to elevated tHcy such as vascular
prevalence of this polymorphism in most populations, the TT variant might disease and pregnancy complications. However, the
represent an ancestral genetic adaptation to living constraints (tissue injury or combined evidence from a large series of case-control
unbalanced vitamin intake) that has become a determinant of disease profiles studies showed that an increased concentration of
in modern times. tHcy attributable to the TT genotype conferred
essentially no cardiovascular risk enhancement8,10.
The flavin adenine dinucleotide (FAD)-dependent This article reviews three aspects of the MTHFR
enzyme 5,10-methylenetetrahydrofolate reductase C677T polymorphism: first, the metabolic effects,
(MTHFR, EC 1.5.1.20) catalyzes the irreversible including altered folate distribution and tHcy
conversion of 5,10-methylenetetrahydrofolate to concentrations, and the interrelationship between the
5-methyltetrahydrofolate, which serves as a methyl concentration of tHcy and lifestyle factors and
donor in the remethylation of homocysteine (Hcy) to diseases; second, its association with risk of diseases,
methionine. The enzyme resides at a metabolic with emphasis on cardiovascular diseases, colorectal
branch point directing the folate pool towards Hcy cancer, birth defects and pregnancy complications;
remethylation at the expense of DNA and RNA and finally, the possible consequences of this
biosynthesis1 (Figs 1 and 2). polymorphism for drug therapy.
The enzyme MTHFR received much interest after
the pivotal discovery of Kang et al.2, who reported Metabolic effects
that a thermolabile enzyme variant was associated Hcy and folate
with increased cardiovascular risk and an increased Several reports have shown consistently that the
concentration of plasma total Hcy (tHcy). Plasma T allele is associated with a high concentration of
tHcy has now been identified as a risk factor for plasma tHcy. The effect on the concentration of tHcy
occlusive disease in the coronary, cerebral and is most pronounced in homozygous TT subjects with
peripheral arteries and for venous thrombosis3, and low folate concentrations8. This is envisaged by a
has also been related to the occurrence of neural tube steeper slope of the curve of the inverse relationship
and other birth defects4 and pregnancy between plasma tHcy and serum folate in subjects
complications5. tHcy concentrations are elevated in with the TT compared with the CC genotype11, which
some diseases and are also determined by genetic and suggests that the C677T transition confers increased
physiological factors, lifestyle and intake of folate responsiveness. In line with this, folic acid at
B-vitamins3 such as folates, cobalamin, vitamin B6 daily doses of 0.5–2.0 mg caused a marked decrease in
Per Magne Ueland*
and riboflavin6. The effects of vitamins are explained tHcy in TT subjects who obtained the same tHcy
Steinar Hustad by their functions as co-factors or co-substrates in Hcy concentration as subjects with the CC genotype1,12,13.
Jørn Schneede metabolism (Fig. 1). The concentration of erythrocyte folate varies
Helga Refsum In 1995, Frosst and colleagues reported on the according to genotype, but the variations are related
Stein Emil Vollset
LOCUS for homocysteine
C677T polymorphism in the MTHFR gene. The to the folate assay used14. This is explained by
and related vitamins, phenotype of this genetic variant is characterized by differential detection by different assays of various
Armauer Hanssens hus, reduced catalytic activity and thermolability in vitro, intracellular folate species, the distribution of which
University of Bergen,
and elevated tHcy under conditions of impaired folate is related to the MTHFR genotype. The latter
5021 Bergen, Norway.
*e-mail: status7. Later studies confirmed that this observation was made by Bagley and Selhub15, who
per.ueland@ikb.uib.no polymorphism is a common genetic determinant of reported that in the homozygous TT subjects,

http://tips.trends.com 0165-6147/01/$ – see front matter © 2001 Elsevier Science Ltd. All rights reserved. PII: S0165-6147(00)01675-8
196 Review TRENDS in Pharmacological Sciences Vol.22 No.4 April 2001

formylated tetrahydrofolate polyglutamates


CH3DNA Protein RNA DNA accumulated at the expense of 5-methyl-
tetrahydrofolate species, the prevailing forms in
Purines dTMP
CHOTHF
subjects with the CC genotype. Thus, the
DHF thermolabile enzyme variant causes a retention of the
MAT DHFR
AdoMet Met THF
folate species committed to purine and pyrimidine
TS CHTHF
synthesis (Fig. 2).
Serine dUMP
R N,N-dimethyl-
glycine Vitamins B2 and B12
SHMT CH2THF
MT BHMT MS Recently, it has been shown that the concentration of
B12
R-CH3 Betaine riboflavin is inversely related to the plasma
Glycine concentration of tHcy, and this relationship has been
Hcy demonstrated in healthy subjects with both low and
AdoHcy CH3THF
SAHH high concentrations of serum folate. The relationship
CBS is genotype dependent and is essentially confined to
B6 MTHFR
individuals with the C677T transition6. A possible
Cysta FAD
regulation of folate distribution and tHcy
Ado concentration by riboflavin status and the C677T
CL B6
polymorphism could be explained by the function of
Cys FAD as a cofactor for MTHFR (Ref. 6), and it gains
TRENDS in Pharmacological Sciences further support from the observation that a mutation
in the bacterial MTHFR that is homologous to the
Fig. 1. Homocysteine (Hcy) formation, remethylation and trans-sulfuration, and the enzymes and C677T transition affects FAD dissociation kinetics16.
B-vitamins involved. Hcy is formed from S-adenosylhomocysteine (AdoHcy). Remethylation to Serum cobalamin is an established determinant of,
methionine (Met) is in most tissues catalyzed by the ubiquitous methionine synthase (MS), which
requires cobalamin (B12) as a cofactor and 5-methyltetrahydrofolate (CH3THF) as a substrate. CH3THF
and is inversely related to, plasma tHcy. The negative
is formed by the action of the flavine adenine dinucleotide (FAD)-dependent enzyme slope of the relationship between cobalamin and tHcy
5,10-methylenetetrahydrofolate reductase (MTHFR), which resides at a crucial metabolic locus was found to be more pronounced in subjects with the
directing the folate pool (green) to Hcy remethylation at the expense of folate used for DNA and RNA
TT genotype than those with CT or CC genotypes17.
biosynthesis. Abbreviations: Ado, adenosine; AdoMet, S-adenosylmethionine; BHMT, betaine-
homocysteine S-methyltransferase; CBS, cystathionine β-synthase; CH2THF, 5,10- Notably, the tHcy–cobalamin relationship was
methylenetetrahydrofolate; CH3DNA, methylated DNA; CHOTHF, formyltetrahydrofolate; maintained after folate supplementation, which
CHTHF, methenyltetrahydrofolate; CL, cystathionine lyase; Cys, cysteine; Cysta, cystathionine; makes confounding from folate status unlikely17. The
DHF, dihydrofolate; DHFR, dihydrofolate reductase; dTMP, deoxythymdine 5′-monophosphate;
mechanism responsible for this interaction is not
dUMP, deoxyuridine 5′-monophosphate; MAT, methionine adenosyltransferase; MT, methyltransferase;
SAHH, S-adenosylhomocysteine hydrolase; SHMT, serine hydroxymethyltransferase; readily understood but could reflect the propensity of
THF, tetrahydrofolate; TS, thymidylate synthase. subjects with the TT genotype to respond with a large
tHcy increment to several factors that cause
hyperhomocysteinemia.
FAD
Hcy MTHFR dUMP Lifestyle
THF CH3THF CH2THF DHF As part of the Hordaland Homocysteine Study,
subjects with markedly elevated tHcy concentrations
CHTHF
(≥40 µmol l−1) were investigated. These individuals
represented the upper 0.4% of the tHcy distribution
CHOTHF
Met AdoMet dTMP from a general population sample of ~18 000 men and
THF women. More than 70% of these 67 individuals
CC Purines possessed the TT genotype combined with folate
50%
deficiency. In addition, they were also more frequently
Protein CH3DNA TT RNA DNA heavier smokers, consumed more coffee and had a
10% CT
40% sedentary lifestyle compared with CT or CC
genotypes11. All these factors are established
TRENDS in Pharmacological Sciences determinants of tHcy (Ref. 18). These findings
suggest that an unhealthy lifestyle interacts with the
Fig. 2. The 5,10-methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism affects the TT genotype and provokes a markedly elevated tHcy.
distribution between folate species (green) used for DNA and RNA syntheses and
5-methyltetrahydrofolate required for homocysteine (Hcy) remethylation and thereby protein DNA methylation
synthesis. The pie chart in the center indicates the genotype prevalence often found in caucasian
populations and the size of the associated vertical arrows indicates the MTHFR activity according
A recent study by Stern et al. showed that the TT
to the genotype. Abbreviations: AdoMet, S-adenosylmethionine; CHOTHF, formyltetrahydrofolate; genotype is associated with lower DNA methylation
CHTHF, methenyltetrahydrofolate; CH2THF, 5,10-methylenetetrahydrofolate; CH3DNA, in peripheral leukocytes compared with the CC
methylated DNA; CH3THF, 5-methyltetrahydrofolate; DHF, dihydrofolate; dTMP, deoxythymidine
genotype. A possible reason for this is the reduced
5′-monophosphate; dUMP, deoxyuridine 5′-monophosphate; FAD, flavine adenine dinucleotide;
Hcy, homocysteine; Met, methionine; THF, tetrahydrofolate. CC and TT are the homozygous availability of 5-methyltetrahydrofolate required for
genotypes and CT is the heterozygous genotype. Modified, with permission, from Ref. 10. S-adenosylmethionine biosynthesis19. This

http://tips.trends.com
Review TRENDS in Pharmacological Sciences Vol.22 No.4 April 2001 197

observation is particularly important because it negatively affect the clinical outcome in kidney
demonstrates that altered folate distribution in failure25 and has also been associated with
subjects with the TT genotype has secondary nephropathy in diabetics with a low concentration of
metabolic effects in addition to folate28. Further studies are required to determine
hyperhomocysteinemia. In addition, DNA whether elevation of the concentration of tHcy
methylation has been considered to be an important attributable to the TT genotype confers increased risk
factor in carcinogenesis20. of cardiovascular disease.

Disease Congenital abnormalities and pregnancy outcome


The relationship between the MTHFR C677T The discovery that periconceptional folic acid
polymorphism and disease involves two aspects. supplementation markedly reduces the occurrence
First, the disease might influence tHcy and recurrence of neural tube defects represents a
concentrations and there might be effect modification major achievement of the past century in preventing
by the MTHFR polymorphism. Second, the congenital malformations. Neural tube defects have
genotype might be associated with disease risk, been related to a low blood concentration of folate and
possibly mediated by altered metabolism of folates an elevated concentration of tHcy in mothers4.
and Hcy. Most of the studies on this subject have shown
that the TT genotype in the mother and baby (but not
Cardiovascular disease in the father) is associated with increased risk of
The observation that the MTHFR TT genotype confers neural tube defects. A recent meta-analysis showed
essentially no cardiovascular disease risk that the TT genotype in the mother or child confers
enhancement has been taken as evidence that an overall risk enhancement corresponding to an
elevated tHcy concentrations do not cause vascular odds ratio of about 29, but a recent large study on
disease. It also appears to support the view that 271 baby–mother pairs indicates that the
hyperhomocysteinemia is a mere epiphenomenon predominant MTHFR-related effect occurs in the
related to subclinical nephrosclerosis21. However, the developing embryo29. A strong nutrient–gene
original studies and the meta-analyses published so interaction is suggested by the observation that a low
far do not have sufficient statistical power to detect the concentration of maternal blood folate30 and no
small risk enhancement related to the mean tHcy periconceptional vitamin supplementation31 enhance
increment of 2.6 µmol l−1 detected in subjects with the the risk associated with the T allele. The
TT compared with the CC genotype8,10. Furthermore, combination of the TT genotype and a low
there is evidence to show that elevated tHcy interacts concentration of folate increases the concentration of
with other risk factors and might provoke the acute Hcy and impairs methionine formation in the
vascular events in subjects with an underlying disease embryo, and it is thought that both factors are
or thrombophilia. Notably, the observation that the TT involved in the genesis of incomplete neural tube
genotype is a strong risk factor for cardiovascular closure. (The relationship between the MTHFR
disease in Japanese populations highlights the C677T polymorphism and congenital anomalies,
significance of genetic background10. Finally, a recent including neural tube defects, was the subject of a
study showed that the TT genotype and recent comprehensive review article9.)
hyperhomocysteinemia are associated with opposite The MTHFR TT genotype of the mother has been
preclinical modification of carotid artery geometry, shown to increase the risk of Down’s syndrome by
which suggests that this genotype might even protect approximately twofold32,33. Further risk
against occlusive vascular disease22. enhancement has been observed with the combined
presence in the mothers of the TT genotype and the
Renal failure A66G polymorphism in the gene encoding methionine
Renal disease is associated with increased risk of synthase reductase33, which is another enzyme
cardiovascular disease and markedly elevated tHcy involved in the methionine cycle. This observation not
(Ref. 23). Recently, renal patients with the TT only addresses the multigenic origin of Down’s
genotype were found to be more susceptible to syndrome but also emphasizes the importance of
hyperhomocysteinemia than those with the CC folate status in the preconceptional period33.
genotype24. As in healthy subjects, the concentration Hyperhomocysteinemia and the TT genotype have
of serum folate was lower and the regression line been associated with pre-eclampsia, spontaneous
relating tHcy to folate was steeper in the patients abortion and placental abruption5. These conditions
with the TT genotype25, but patients with this are related to impaired function of the placental
genotype readily responded to high-dose folic acid vascular bed, and it is thought that elevated tHcy
supplementation – which was evident from a might be responsible for the vasculopathy. However,
reduction in tHcy – and they more often attained recent publications have questioned the association
normal tHcy concentrations26,27. between the MTHFR C677T polymorphism and
In renal patients, tHcy predicts mortality and placenta-mediated diseases34–37. Conceivably, some
cardiovascular morbidity23. The TT genotype can points raised in the ongoing debate on MTHFR and

http://tips.trends.com
198 Review TRENDS in Pharmacological Sciences Vol.22 No.4 April 2001

cardiovascular disease10 might also be relevant for investigations of the possible relationship between
the issue of the correlation between pre-eclampsia the C677T polymorphism and diabetic complications
and MTHFR. have shown conflicting results24.

Cancer Drugs
Low folate status is a risk factor for colorectal cancer Several drugs that interfere with folate status
as well as some other forms of cancer 20. Folate status increase the concentration of tHcy (Ref. 18). Such
shows a strong interactive effect with the C677T drugs include not only the classical antifolate
polymorphism; this has been most convincingly methotrexate, trimethoprim52, but also sulfasalazine
demonstrated for colorectal cancer and its clinical and the antiepileptic drugs phenytoin,
precursor, colorectal adenomas. In folate-replete carbamazepine and valproic acid.
subjects, the TT genotype affords 50% risk reduction, In rheumatoid patients treated with methotrexate
whereas in subjects with low folate status, the TT or sulfasalazine 53 and in epileptics receiving
genotype confers no protection or probably risk anticonvulsant medication54, the tHcy concentrations
enhancement38–41. One study has suggested that are higher in TT than in CC subjects. In
smoking, which is an established risk factor of hypercholesterolemic children, treatment with
colorectal neoplasias, might be a particularly strong cholestyramine was associated with a significant
interactive factor. Furthermore, the study identified increase in tHcy concentrations, but only in those
two high-risk categories for premalignant colorectal children with one or two T alleles55.
adenomas: smokers with low folate and the L-Dihydroxyphenylalanine (L-dopa) increases Hcy
TT genotype, and smokers with the CC genotype production by serving as a substrate for catechol
and high folate42. This observation has implications in O-methyltransferase56. In rats this produces a rise in
relation to the debate of folate fortification. plasma tHcy, which is superimposed on the
There is preliminary evidence to suggest that hyperhomocysteinemia induced by folate deficiency56.
individuals with the TT genotype have a decreased The tHcy response to L-dopa treatment is accentuated
risk of adult acute leukemia and increased risk of in patients with the MTHFR TT genotype57. Thus, for
endometrial cancer, cervical neoplasia and breast drugs that cause hyperhomocysteinemia either by
cancer43. These studies were based on small interference with folate metabolism or by enhanced
populations, and there was no assessment of folate Hcy production, there is a marked effect modification
status. Risk enhancement in one malignancy versus by the MTHFR C677T polymorphism, and the TT
protection in other malignancies might actually genotype enhances the Hcy response. The important
reflect the folate status of the study population. question is whether some of the side-effects caused by
Published data on the MTHFR C677T these drugs are mediated by Hcy or by altered folate
polymorphism and cancer risk indicate that the distribution, and whether subjects with the TT
T allele protects against cancer in folate-replete genotype are at increased risk. Some observations
subjects but increases the risk under conditions of suggest that this might be the case. A high
impaired folate status. The protection might be concentration of tHcy in rheumatoid patients treated
related to abundant purines and pyrimidines with methotrexate has been associated with adverse
available for DNA synthesis, leading to efficient DNA effects53. Furthermore, a preliminary report suggests
repair and essentially no uracil incorporation into that essentially all cancer patients experiencing
DNA. The combination of low folate and TT genotype severe toxicity from the drug combination
impairs Hcy remethylation to methionine; this could cyclophosphamide, methotrexate and fluorouracil
thereby cause DNA hypomethylation, which is known (CMF) possessed the TT genotype58.
to be involved in carcinogenesis38. The MTHFR C677T polymorphism might
modulate the effect of some drugs. The genotype could
Other diseases even interact with drugs to increase the risk of
A positive association between the TT genotype and diseases associated with hyperhomocysteinemia such
inflammatory bowel disease (IBD) has been observed as cardiovascular disease, birth defects and pregnancy
in both an Irish44 and a Danish45 population but not in complications. Therefore, because genotyping could be
an Italian population46. Such differences between useful for tailoring the dosage of some drugs, in
study populations could be explained by different particular antifolate agents, the pharmacogenetics of
folate status, which is in agreement with an elevated the MTHFR C677T polymorphism will be an
concentration of tHcy and a low concentration of important area for future research.
folate in the Irish patients. These observations are
relevant to the increased risk of colorectal cancer and Concluding remarks and a hypothesis
thromboembolic events in IBD patients44. The C677T transition has been implicated in several
The TT genotype has been shown to increase the diseases. For neural tube defects and some malignant
risk of psychiatric disorders24 such as dementia47, diseases, the TT genotype confers increased risk at
schizophrenia48,49 and depression, but this has been low folate concentrations. At high folate
contested in other studies24,47,50,51. In addition, concentrations, the TT genotype affords protection

http://tips.trends.com
Review TRENDS in Pharmacological Sciences Vol.22 No.4 April 2001 199

Box 1. The effect of MTHFR C677T polymorphism on folate distribution under different conditions – a hypothetical model
The enzyme 5,10-methylenetetrahydrofolate
reductase (MTHFR) is responsible for the Fig. I CC TT
irreversible conversion of 5,10-methylene-
tetrahydrofolate (CH2THF) to 5-methyl- Positive folate and riboflavin balance
tetrahydrofolate (CH3THF). Figure I depicts a
model that proposes a modification by the FAD FAD
MTHFR C677T polymorphism of the MTHFR MTHFR
Hcy Hcy
metabolic consequences of folate,
cobalamin or riboflavin deficiency. This
model integrates data on the metabolic
effects, regulation and genetics of the MTHFR
polymorphism – with some biological
Folate deficiency
observations – into a unifying hypothesis. FAD FAD
The model explains the propensity Hcy MTHFR MTHFR
Hcy
towards hyperhomocysteinemia in subjects
with the TT genotype, whereas folate
species used for DNA and RNA synthesis are
preserved. A high concentration of total
homocysteine can by itself have a
procoagulant effect and thereby predispose Cobalamin deficiency and folate trap
susceptible individuals to vascular occlusive
FAD FAD
disease, but folate sparing might also secure
Hcy MTHFR Hcy MTHFR
cell proliferation and tissue repair, which
could be beneficial in subjects who are
deficient in folate, riboflavin or cobalamin.
Additional support for the model can be
gained from the following observations. One
report described a patient with an inborn Riboflavin deficiency
error of cobalamin metabolism FAD FAD

(complementation group CblG) and the Hcy MTHFR Hcy MTHFR


TT genotype who did not have
megaloblastic anemia, which suggests that
the TT genotype protects the bone marrowb.
In addition, MTHFR is among the flavo
enzymes that are most sensitive to impaired Protein DNA Protein DNA
riboflavin statusc. Thus, riboflavin deficiency CH3DNA RNA CH3DNA RNA
could influence the tissue distribution and TRENDS in Pharmacological Sciences
economy of reduced folate by decreasing
MTHFR activityd. The FAD dissociation Fig. I. A hypothetical model for the effect of 5,10-methylenetetrahydrofolate reductase (MTHFR) C677T
kinetics of the enzyme variant associated polymorphism on folate distribution under normal conditions and in folate, cobalamin or riboflavin deficiency. The
folate homeostasis in vitamin-replete subjects (upper panel) ensures optimal homocysteine (Hcy) remethylation
with the T allelee might favor this adaptive and low plasma total Hcy (tHcy) concentrations in both genotypes. In folate deficiency (second panel) the amount
process. In experimental animals, riboflavin of all folate species is reduced. In the TT genotype, low MTHFR activity decreases 5-methyltetrahydrofolate
(CH3THF) formation and thereby Hcy remethylation, whereas there is a sparing effect on other one-carbon-
deficiency causes an increased oxidation
substituted folates. In cobalamin deficiency (third panel), secondary folate loss can develop as a result of folate
state of the folate pool and a reduction in the trappinga. The low MTHFR activity associated with the TT genotype could have a protective effect by antagonizing
relative amounts of 5-methyltetra- the loss of folate. Finally, the model predicts that riboflavin deficiency might have a similar sparing effect on one-
carbon-substituted folates other than CH3THF (fourth panel), as a result of depletion of the cofactor flavine adenine
hydrofolatec. Riboflavin deficiency does not
dinucleotide (FAD) and low MTHFR activity. In subjects with the TT genotype, this effect is enhanced because of
seem to be associated with overt lower cofactor affinity. The figures on the left- and right-hand sides demonstrate folate balance in subjects with the
megaloblastic anemiad, and this could be CC and the TT genotypes, respectively. The thickness of the black arrows represents the enzyme activity and the
metabolic flux through this pathway. The thickness of the green arrows indicates the size of and flux through the
due to a sparing effect on folate species used
folate pools, and the sizes of Hcy boxes represent the plasma tHcy concentrations. The sizes of the FAD circles and
for DNA and RNA synthesis. the overlap with the MTHFR boxes indicate FAD concentrations and binding to the MTHFR, respectively.

References c Bates, C.J. and Fuller, N.J. (1986) The effect of of critical flavoenzyme-dependent metabolic
a Shane, B. and Stokstad, E.L. (1985) Vitamin riboflavin deficiency on methylenetetrahydro- pathways. Biofactors 3, 185–190
B12–folate interrelationships. Annu. Rev. Nutr. 5, folate reductase (NADPH) (EC 1.5.1.20) and folate e Guenther, B.D. et al. (1999) The structure and
115–141 metabolism in the rat. Br. J. Nutr. 55, 455–464 properties of methylenetetrahydrofolate
b Kvittingen, E.A. et al. (1997) Methionine synthase [Erratum: Br. J. Nutr. (1986) 56, following 683] reductase from Escherichia coli suggest how folate
deficiency without megaloblastic anaemia. Eur. J. d Ross, N.S. and Hansen, T.P. (1992) Riboflavin ameliorates human hyperhomocysteinemia. Nat.
Pediatr. 156, 925–930 deficiency is associated with selective preservation Struct. Biol. 6, 359–365

http://tips.trends.com
200 Review TRENDS in Pharmacological Sciences Vol.22 No.4 April 2001

against colorectal neoplasias. However, despite procoagulant effect of elevated tHcy concentrations in
intensive research, no conclusion has been reached TT subjects enhances hemostasis. Furthermore,
regarding the association between the polymorphism sufficient folate for DNA synthesis could ensure
and cardiovascular disease. effective cell proliferation and tissue repair, which
Thus, the MTHFR TT genotype appears to protect could be advantageous under conditions of delivery
against some diseases and increase the risk of others, and tissue injury. The TT genotype could conserve
which is in accordance with observations showing no folate species used for DNA synthesis in individuals
correlation between genotype and longevity24,59. This on nutriture characterized by unbalanced or low
polymorphism might therefore influence the disease content of folate, riboflavin or cobalamin (Box 1).
profile in a population rather than the overall These features might have promoted survival or
morbidity and mortality. reproduction in ancient times, and the C667T
Acknowledgements The C667T polymorphism should not be regarded transition might therefore represent an example of
Our work was supported a priori as a genetic defect that causes disease but ecological genetics of hemostasis, tissue repair or
by EU Commission
Demonstration
rather as a genetic variant that could confer some nutrition. Thus, an ancestral genetic adaptation
Project Contract contingent advantages during the reproductive might have turned into a predictor of diseases in
BMH4-CT98-3549. period of life. For example, it is possible that the modern times.
References from low dose folic acid supplementation in women 25 Kimura, H. et al. (2000) The C677T
1 Bailey, L.B. and Gregory, J.F., III (1999) with unexplained recurrent miscarriages. J. Nutr. methylenetetrahydrofolate reductase gene
Polymorphisms of methylenetetrahydrofolate 128, 1336–1341 mutation in hemodialysis patients. J. Am. Soc.
reductase and other enzymes: metabolic 14 Molloy, A.M. et al. (1998) Whole-blood folate values Nephrol. 11, 885–893
significance, risks and impact on folate requirement. in subjects with different methylenetetrahydrofolate 26 Tremblay, R. et al. (2000) Hyperhomocysteinemia in
J. Nutr. 129, 919–922 reductase genotypes: differences between the hemodialysis patients: effects of 12-month
2 Kang, S-S. et al. (1993) Thermolabile defect of radioassay and microbiological assays. Clin. Chem. supplementation with hydrosoluble vitamins.
methylenetetrahydrofolate reductase in coronary 44, 186–188 Kidney Int. 58, 851–858
artery disease. Circulation 88, 1463–1469 15 Bagley, P.J. and Selhub, J. (1998) A common 27 Sunder-Plassmann, G. et al. (2000) Effect of high
3 Refsum, H. et al. (1998) Homocysteine and mutation in the methylenetetrahydrofolate dose folic acid therapy on hyperhomocysteinemia in
cardiovascular disease. Annu. Rev. Med. 49, 31–62 reductase gene is associated with an accumulation of hemodialysis patients: results of the Vienna
4 Finnell, R.H. et al. (1998) Neural tube and formylated tetrahydrofolates in red blood cells. Proc. multicenter study. J. Am. Soc. Nephrol. 11,
craniofacial defects with special emphasis on folate Natl. Acad. Sci. U. S. A. 95, 13217–13220 1106–1116
pathway genes. Crit. Rev. Oral Biol. Med. 9, 38–53 16 Guenther, B.D. et al. (1999) The structure and 28 Shpichinetsky, V. et al. (2000) The association
5 Ray, J.G. and Laskin, C.A. (1999) Folic acid and properties of methylenetetrahydrofolate reductase between two common mutations C677T and A1298C
homocyst(e)ine metabolic defects and the risk of from Escherichia coli suggest how folate ameliorates in human methylenetetrahydrofolate reductase
placental abruption, pre-eclampsia and spontaneous human hyperhomocysteinemia. Nat. Struct. Biol. 6, gene and the risk for diabetic nephropathy in type II
pregnancy loss: a systematic review. Placenta 20, 359–365 diabetic patients. J. Nutr. 130, 2493–2497
519–529 17 D’Angelo, A. et al. (2000) The role of vitamin B12 in 29 Shields, D.C. et al. (1999) The ‘thermolabile’variant
6 Hustad, S. et al. (2000) Riboflavin as a determinant fasting hyperhomocysteinemia and its interaction of methylenetetrahydrofolate reductase and neural
of plasma total homocysteine: effect modification by with the homozygous C677T mutation of the tube defects: an evaluation of genetic risk and the
the methylenetetrahydrofolate reductase C677T methylenetetrahydrofolate reductase (MTHFR) relative importance of the genotypes of the embryo
polymorphism. Clin. Chem. 46, 1065–1071 gene. A case-control study of patients with early- and the mother. Am. J. Hum. Genet. 64, 1045–1055
7 Frosst, P. et al. (1995) Identification of a candidate onset thrombotic events. Thromb. Haemost. 83, 30 Christensen, B. et al. (1999) Genetic polymorphisms
genetic risk factor for vascular disease: a common 563–570 in methylenetetrahydrofolate reductase and
mutation at the methylenetetrahydrofolate 18 Ueland, P.M. et al. (2000) Determinants of plasma methionine synthase, folate levels in red blood cells,
reductase locus. Nat. Genet. 10, 111–113 homocysteine. In Homocysteine and Vascular and risk of neural tube defects. Am. J. Med. Genet.
8 Brattstrom, L. et al. (1998) Common Disease (Robinson, K., ed.), pp. 59–84, Kluwer 84, 151–157
methylenetetrahydrofolate reductase gene Academic Publishers 31 Shaw, G.M. et al. (1998) Maternal vitamin use,
mutation leads to hyperhomocysteinemia but not to 19 Stern, L.L. et al. (2000) Genomic DNA genetic variation of infant methylene-
vascular disease: the result of a meta-analysis. hypomethylation, a characteristic of most cancers, is tetrahydrofolate reductase, and risk for spina bifida.
Circulation 98, 2520–2526 present in peripheral leukocytes of individuals who Am. J. Epidemiol. 148, 30–37
9 Botto, L.D. and Yang, Q. (2000) 5,10- are homozygous for the C677T polymorphism in the 32 James, S.J. et al. (1999) Abnormal folate metabolism
Methylenetetrahydrofolate reductase gene variants methylenetetrahydrofolate reductase gene. Cancer and mutation in the methylenetetrahydrofolate
and congenital anomalies: a HuGE review. Am. J. Epidemiol. Biomarkers Prev. 9, 849–853 reductase gene may be maternal risk factors for
Epidemiol. 151, 862–877 20 Kim, Y-I. (1999) Folate and carcinogenesis: evidence, Down syndrome. Am. J. Clin. Nutr. 70, 495–501
10 Ueland, P.M. et al. (2000) The controversy over mechanisms and implications. J. Nutr. Biochem. 10, 33 Hobbs, C.A. et al. (2000) Polymorphisms in genes
homocysteine and cardiovascular risk. Am. J. Clin. 66–88 involved in folate metabolism as maternal risk
Nutr. 72, 324–332 21 Brattstrom, L. and Wilcken, D.E. (2000) factors for Down syndrome. Am. J. Hum. Genet. 67,
11 Guttormsen, A.B. et al. (1996) Determinants and Homocysteine and cardiovascular disease: cause or 623–630
vitamin responsiveness of intermediate effect? Am. J. Clin. Nutr. 72, 315–323 34 van der Molen, E.F. et al. (2000) A common
hyperhomocysteinemia (≥40 µmol l−1). The 22 Demuth, K. et al. (1998) Opposite effects of plasma mutation in the 5,10-methylenetetrahydrofolate
Hordaland homocysteine study. J. Clin. Invest. 98, homocysteine and the methylenetetrahydrofolate reductase gene as a new risk factor for placental
2174–2183 reductase C677T mutation on carotid artery vasculopathy. Am. J. Obstet. Gynecol. 182,
12 Malinow, M.R. et al. (1997) The effects of folic acid geometry in asymptomatic adults. Arterioscler. 1258–1263
supplementation on plasma total homocysteine are Thromb. Vasc. Biol. 18, 1838–1843 35 Kobashi, G. et al. (2000) Absence of association
modulated by multivitamin use and 23 Bostom, A.G. and Culleton, B.F. (1999) between a common mutation in the
methylenetetrahydrofolate reductase genotypes. Hyperhomocysteinemia in chronic renal disease. methylenetetrahydrofolate reductase gene and
Arterioscler. Thromb. Vasc. Biol. 17, 1157–1162 J. Am. Soc. Nephrol. 10, 891–900 preeclampsia in Japanese women. Am. J. Med.
13 Nelen, W.L. et al. (1998) Methylenetetrahydrofolate 24 Fodinger, M. et al. (2000) Molecular biology of 5,10- Genet. 93, 122–125
reductase polymorphism affects the change in methylenetetrahydrofolate reductase. J. Nephrol. 36 Kaiser, T. et al. (2000) Methylenetetrahydrofolate
homocysteine and folate concentrations resulting 13, 20–33 reductase polymorphisms are not a risk factor for

http://tips.trends.com
Review TRENDS in Pharmacological Sciences Vol.22 No.4 April 2001 201

pre-eclampsia/eclampsia in Australian women. 44 Mahmud, N. et al. (1999) Increased prevalence of 52 Smulders, Y.M. et al. (1998) Trimethoprim and
Gynecol. Obstet. Invest. 50, 100–102 methylenetetrahydrofolate reductase C677T fasting plasma homocysteine. Lancet 352,
37 Laivuori, H. et al. (2000) 677 C→T polymorphism of variant in patients with inflammatory bowel 1827–1828 [Erratum: Lancet (1999) 353, 758]
the methylenetetrahydrofolate reductase gene and disease, and its clinical implications. Gut 45, 53 Haagsma, C.J. et al. (1999) Influence of
preeclampsia. Obstet. Gynecol. 96, 277–280 389–394 sulphasalazine, methotrexate, and the combination
38 Chen, J. et al. (1999) MTHFR polymorphism, 45 Nielsen, J.N. et al. (2000) Increased prevalence of of both on plasma homocysteine concentrations in
methyl-replete diets and the risk of colorectal methylenetetrahydrofolate reductase C677T patients with rheumatoid arthritis. Ann. Rheum.
carcinoma and adenoma among U.S. men and variant in patients with IBD. Gut 47, 456–457 Dis. 58, 79–84
women: an example of gene–environment 46 Vecchi, M. et al. (2000) Inflammatory bowel diseases 54 Yoo, J.H. and Hong, S.B. (1999) A common mutation
interactions in colorectal tumorigenesis. are not associated with major hereditary conditions in the methylenetetrahydrofolate reductase gene is
J. Nutr. 129, 560S–564S predisposing to thrombosis. Dig. Dis. Sci. 45, a determinant of hyperhomocysteinemia in epileptic
39 Levine, A.J. et al. (2000) The methylene- 1465–1469 patients receiving anticonvulsants. Metabolism 48,
tetrahydrofolate reductase 677C→T polymorphism 47 Yoo, J.H. et al. (2000) Pathogenicity of thermolabile 1047–1051
and distal colorectal adenoma risk. Cancer methylenetetrahydrofolate reductase for vascular 55 Tonstad, S. et al. (1998) The C677T mutation in the
Epidemiol. Biomarkers Prev. 9, 657–663 dementia. Arterioscler. Thromb. Vasc. Biol. 20, methylenetetrahydrofolate reductase gene
40 Slattery, M.L. et al. (1999) Methylene- 1921–1925 predisposes to hyperhomocysteinemia in children
tetrahydrofolate reductase, diet, and risk of colon 48 Wei, J. and Hemmings, G.P. (1999) Allelic with familial hypercholesterolemia treated with
cancer. Cancer Epidemiol. Biomarkers Prev. 8, association of the MTHFR gene with schizophrenia. cholestyramine. J. Pediatr. 132, 365–368
513–518 Mol. Psychiatry 4, 115–116 56 Daly, D. et al. (1997) The effect of L-dopa
41 Ulrich, C.M. et al. (1999) Colorectal adenomas and 49 Joober, R. et al. (2000) Association between the administration and folate deficiency on plasma
the C677T MTHFR polymorphism: evidence for methylenetetrahydrofolate reductase 677C→T homocysteine concentrations in rats. J. Nutr.
gene–environment interaction? Cancer Epidemiol. missense mutation and schizophrenia. Mol. Biochem. 8, 634–640
Biomarkers Prev. 8, 659–668 Psychiatry 5, 323–326 57 Yasui, K. et al. (2000) Plasma homocysteine and
42 Ulvik, A. et al. Smoking, folate and 50 Pollak, R.D. et al. (2000) The C677T mutation in the MTHFR C677T genotype in levodopa-treated
methylenetetrahydrofolate reductase status as methylenetetrahydrofolate reductase (MTHFR) patients with PD. Neurology 55, 437–440
interactive determinants of adenomatous and gene and vascular dementia. J. Am. Geriatr. Soc. 48, 58 Toffoli, G. et al. (2000) MTHFR gene polymorphism
hyperplastic polyps of colorectum. Am. J. Med. 664–668 and severe toxicity during adjuvant treatment of early
Genet. (in press) 51 Tysoe, C. et al. (1997) Analysis of α-1 breast cancer with cyclophosphamide, methotrexate,
43 Kim, Y.I. (2000) Methylenetetrahydrofolate antichymotrypsin, presenilin-1, angiotensin- and fluorouracil (CMF). Ann. Oncol. 11, 373–374
reductase polymorphisms, folate, and cancer risk: a converting enzyme, and methylenetetrahydrofolate 59 Brattstrom, L. et al. (1998) A common
paradigm of gene–nutrient interactions in reductase loci as candidates for dementia. Am. J. methylenetetrahydrofolate reductase gene mutation
carcinogenesis. Nutr. Rev. 58, 205–209 Med. Genet. 74, 207–212 and longevity. Atherosclerosis 141, 315–319

FGF and VEGF function in angiogenesis:


signalling pathways, biological
responses and therapeutic inhibition
Michael J. Cross and Lena Claesson-Welsh

Angiogenic growth factors such as fibroblast growth factors (FGFs) and is characterized by tight regulation both spatially and
vascular endothelial growth factors (VEGFs) are currently targets of intense temporally. Angiogenic factors, such as fibroblast
efforts to inhibit deregulated blood vessel formation in diseases such as cancer. growth factors (FGFs) and vascular endothelial growth
FGFs and VEGFs exert their effects via specific binding to cell surface- factors (VEGFs), stimulate endothelial cells to secrete
expressed receptors equipped with tyrosine kinase activity. Activation of the several proteases and plasminogen activators,
receptor kinase activity allows coupling to downstream signal transduction resulting in the degradation of the vessel basement
pathways that regulate proliferation, migration and differentiation of membrane, which in turn allows cells to invade the
endothelial cells. Inhibitors of FGF and VEGF signalling are currently in clinical surrounding matrix. The cells migrate, proliferate and
trials. In this article, the current knowledge of FGF- and VEGF-induced signal eventually differentiate to form a new, lumen-
transduction that leads to specific biological responses will be summarized. containing vessel. Finally, the endothelial cells deposit
Furthermore, the manner in which this knowledge is being exploited to a new basement membrane and secrete growth factors,
regulate angiogenesis will be discussed. such as platelet-derived growth factor (PDGF), which
attract supporting cells such as pericytes, ensuring the
Angiogenesis denotes the formation of new blood stability of the new vessel1. This is a complex process
vessels from pre-existing vessels. Physiological that involves the concerted action of several other
angiogenesis, which is required for embryonic factors, such as the angiopoietins and ephrins, that act
development, wound healing and the menstrual cycle, on specific receptors to regulate vessel stability2.

http://tips.trends.com 0165-6147/01/$ – see front matter © 2001 Elsevier Science Ltd. All rights reserved. PII: S0165-6147(00)01676-X

Você também pode gostar