ArticlePDF Available

Folic Acid A case for personalised preventive nutrition

Authors:

Abstract

Fortification of food with folic acid has resulted in significant declines in the occurrence of Neural Tube defect affected pregnancies; however, growing scientific evidence in the field suggests a possible association between high intake levels of folic acid and risks of cancer and other harmful effects. This article discusses the need to translate new epigenetic evidence into a more personalized folic acid supplementation approach
FOLIC ACID
A case for personalized preventive nutrition?
Fortification of food with folic
acid has resulted in significant
declines in the occurrence of
Neural Tube defect affected
pregnancies; however, growing
scientific evidence in the field
suggests a possible association
between high intake levels of
folic acid and risks of cancer and
other harmful effects. This article
discusses the need to translate
new epigenetic evidence into a
more personalized folic acid sup-
plementation approach.
Verhagen, M.C.M. 1 ; Brand, A 1;
Ambrosino, E 1; Haslberger, A.G. 2
FOLATE DEFICIENCY AND NTDS
The relationship between folate deficien-
cy and Neural Tube Defects (NTDs) occur-
rence was hypothesized as early as 1965
(Crider, Bailey, & Berry, 2011). Evidence
from scientific studies has since then
conclusively demonstrated that folic acid
supplementation can prevent the occur-
rence of NTDs (EFSA, 2009); supplemen-
tation of >400µg/day is found to prevent
as much as 70% of these defects (Van
den Veyver, 2002). This has led many
countries to recommend women plan-
ning to become pregnant to supplement
their diet with folic acid (synthetic form of
folate). In the EU, the recommended ref-
erence intake for folate is set by SCF (Sci-
entific Committee on Foods, EC) : 200μg
folate/day for adults, 400μg folate/day
for pregnancy (EFSA, 2009). Yet, Public
Health campaigns by countries to pro-
mote the awareness of this message and
promote voluntary supplement intake by
pregnant women have been unsuccessful
in most countries (EUROCAT, 2005); the
fact that the former target group needs
to consume folic acid in the 4 weeks be-
fore and 8 weeks after conception, makes
a voluntary intervention hard to imple-
ment effectively. This problem, together
with the discovery of another important
suspected effect of folic acid in the early
1990s: protection against cardiovascular
diseases later in life (Cornel, de Smit, &
de Jong-van den Berg, 2005), lead Public
Health prevention policies to develop in
two ways: some American and developing
countries choose to implement mandato-
ry fortification of staple foods (Cornel,de
Smit, & de Jong-van den Berg, 2005), like
flour, while voluntary fortification of food
with folic acid is permitted and obligated
in most European countries. Currently
no EU country has implemented manda-
tory fortification (EFSA, 2009). Manda-
tory folic acid food fortification has since
then resulted in significant declines in the
occurrence of NTD affected pregnan-
cies (EFSA, 2009). The percent declines
range from 28% to 46% in the USA and
Canada respectively (EFSA, 2009). Many
(European) countries are therefore con-
sidering whether to adopt this mandatory
fortification policy. American biomarker
studies postfortification showed dramatic
increases in population blood measure-
ments of folate, this raised concerns that
fortification exceeded the original daily
intake target by
as much as 2-fold
(Ulrich & Potter,
2006). At the
same time an ad-
vertising hyper-
bole of proactive
fortified health
foods market
seems to have
occurred, “where
a muddying of
waters can oc-
cur regarding the
ideal between too
little or too much
of any given nutrient” (Lucock & Yates,
2009). This is of concern, taking into ac-
count the growing scientific evidence that
has recently emerged suggesting a pos-
sible association between high intake lev-
els of folic acid and risks of cancer (EFSA,
2009) and various other harmful effects.
RESULTS FROM CLINICAL STUDIES
Effects of folic acid supplementation are
reviewed by Lucock & Yates (2009); the
main positive effects described are low-
ering of the risk for birth defects, lower
vasculotoxic, embryotoxic and neurotoxic
homocysteine with benefit to a range
of vascular conditions and Alzheimer’s
disease. Evidence from in vitro, animal,
and human studies has shown that fo-
late supplementation can act to prevent
tumour initiation (Smith, Kim, & Refsum,
2008), yet it seems to facilitate progres-
sion of precancerous lesions (Lucock &
Yates, 2009). A similar effect has also
been shown in vascular patients, where
supraphysiologic vitamins B6 and B12
along with folic acid did not lower recur-
rent cardiovascular disease after acute
myocardial infarction, with the authors
indicating a possible harmful effect of
combined vitamin B therapy (Reviewed
in: Lucock & Yates, 2009). Furthermore,
it now seems that synthetic folate satu-
rates human dihydrofolate reductase
(DHFR) leading to unmetabolized folate
in the circulation, and possibly masks the
irreversible pernicious anaemia of B12
deficiency, the former increasing risk for
cognitive impairment, while during preg-
nancy the same folate-B12 disposition
may increase insulin resistance and obe-
sity in offspring (Reviewed in: Lucock &
Yates, 2009). Folic acid also reduces natu-
ral killer cell cytotoxicity, may increase the
prevalence of positional plagiocephaly,
and increases multiple births after IVF
(Reviewed in: Lucock & Yates, 2009).
Since the 1940s antifolate drugs are being
used in cancer chemotherapy (Ulrich &
Potter, 2006), because removal of folate
or a blockade of its metabolism causes in-
hibition of tumor growth (Smith, Kim, &
Refsum, 2008). Recent evidence showed
that excess folic acid alters the efficacy
of antifolate drugs (Reviewed in: Lucock
& Yates, 2009). In general, “intervention
studies using folic acid have produced a
range of different results including ad-
verse effects; overall they do not support
the hypothesis that folic acid supplemen-
tation of human populations reduces the
chronic disease risk” (EFSA, 2009). It is
clear that disruptions in folate metabolism
increase risk for a variety of pathologies
(Stover & Caudill, 2008), yet while folate
supplementation can reduce the risk of
some disorders developing (Stover & Cau-
dill, 2008), it can itself likely disrupt the
folate metabolism as well. So far, the pre-
cise biochemical mechanisms underlying
folate-related pathologies have remained
elusive despite intensive investigation
(Stover & Caudill, 2008), yet new system
biological evidence is rapidly emerging.
NUTRIGENETIC AND
EPIGENETIC ASPECTS
Adequate folate status is essential for
DNA synthesis and cell division and low
folate status in humans is associated
with an increase in DNA strand breaks,
impaired DNA repair and increased mu-
tations (Smith, Kim, & Refsum, 2008).
Folate also plays a major role in the fo-
late-mediated one-carbon metabolism;
with a main function in the provision of
methyl-groups for the conversion of ho-
mocysteine to methionine (Smith, Kim, &
Refsum, 2008), which in turn can be ad-
enosylated to form S-adenosylmethionine
(SAM) (Stover & Caudill, 2008). SAM is
a molecule with many functions, includ-
ing methylation of cytosine residues in
DNA and of arginine and lysine residues
in histones, both of which are involved in
regulating gene expression (Smith, Kim,
& Refsum, 2008). By example, meth-
ylation of promoter related CpG islands
can suppress gene expression by caus-
ing chromatin condensation (“silencing”),
and could, for instance, silence tumor
supressor as well as a tumor oncogenes.
Therefore, folate plays a major role in epi-
genetic mechanisms; “any processs that
alters gene activity without changes of
the DNA sequence” (Haslberger, Varga,
& Karlic, 2006). Gene variants that en-
code folate-dependent enzymes and al-
ter the efficiency of nucleotide and SAM
biosynthesis can confer both, protection
and risk for specific pathologies; exam-
ples are the common SNPs in the methy-
lenetetrahydrofolate dehydrogenase
gene (MTHFD1), 1958G>A and methy-
lenetetrahydrofolate reductase gene
(MTHFR), 677C>T, which increases the
risk for NTDs (Stover & Caudill, 2008).
It is described that periconceptual expo-
sure to folic acid might genetically select
the latter gene,
and lead to
an increase
in prevalence
of individuals
with MTHFR,
677C>T. This
SNP is also as-
sociated with
d e ge ne r at iv e
disorders (Lu-
cock & Yates,
2009), however,
seems protec-
tive against co-
lon cancer in
folate-replete individuals (Stover & Cau-
dill, 2008). So far, “The functional role
of these polymorphisms in the etiology
of NTDs and cancer is unknown but is
likely related to DNA synthesis, repair,
and/or methylation mechanisms (Stover
& Caudill, 2008)”. “Low folate status
is often associated with impairment of
DNA methylation, but sometimes it leads
to hypermethylation and thus could af-
fect gene expression in complex ways. It
is not known whether an excess of fo-
late might have any adverse effects on
these functions (Smith, Kim, & Refsum,
2008)”. Changing the folate status in hu-
mans has been shown to influence DNA
methylation, but, it is not yet established
whether alterations in DNA methylation
after changes in folate status are harm-
ful in humans, for example, by regulating
the expression of oncogenes or tumor
suppressorgenes (Smith, Kim, & Refsum,
2008) and studies published have shown
many contradicting results. However,
animal studies on colorectal cancer have
shown that the timing and dose of folate
intervention are critical: If folate supple-
mentation is started before the establish-
ment of neoplastic foci, the development
and progression of the tumor is sup-
pressed, if started after, it enhances their
growth and progression (Smith, Kim, &
Refsum, 2008). This dichotomy has been
consistently shown for colorectal adeno-
mas, colorectal cancer rates, breast and
prostate cancers (Lucock & Yates, 2009).
Furthermore, a recent study by Berner, et
al. (2010) found that exposure to a high
concentration of folic acid enhanced
cancer cell growth, and concomitant in-
creased methylation of estrogen recep-
tor and tumor suppressor promotors was
observed, while a lower concentration of
folic acid decreased cell growth. Aber-
rant promotor hypermethylation that is
associated with inappropriate gene si-
lencing of tumor suppressor genes is hy-
pothesised to affect virtually every step
in tumor progression (Haslberger, Varga,
& Karlic, 2006).
It is important to note that Smith, Kim,
& Refsum (2008) stated that additional
biological explanations for contradictions
in study outcomes are also likely: “For ex-
ample, it is biologically plausible that any
effect of folate on carcinogenesis will in-
teract with a large number of other risk
factors and that the patterns of these
risk factors will differ between individu-
als. Observational studies have identified
many factors, apart from age and sex, that
might interact with folate in cancer risk,
including vitamin B-12, alcohol, smoking,
and polymorphisms in genes coding for
enzymes related to one-carbon metabo-
lism (Smith, Kim, & Refsum, 2008)”. Fur-
thermore, the influence of folate status
on DNA methylation in both animals and
humans is hypothesised to be tissue-, site-
, and gene-specific (Smith, Kim, & Ref-
sum, 2008). Overall, studies reviewed by
Smith, Kim, & Refsum (2008) show that it
is not justified to assume that the finding
of a protective effect of high folate in a
whole population necessarily applies to all
people within that population, moreover,
evidence provides cause for concern that
increasing folate levels in an entire popu-
lation may, in some people, increase the
risk of cancer.
A NEED FOR AN
INDIVIDUALIZED APPROACH?
Because the interactions between the
epigenome and folic acid methylation-
diets are complex and the fact that un-
derlying biochemical mechanisms remain
elusive, a population-based prevention
method like folic acid food fortification
is hazardous. “The highly complex and
critical biological importance of folic ac-
id-related molecular nutrition makes it a
difficult micronutrient to deploy as a sim-
ple intervention at a population level – it
has far too many biochemical spheres of
OH
OH
OH
O
O
O
N
N
NN
H
N
H
N
H2N
28 JEM Dezember 2011
Bericht & Report
29 JEM Dezember 2011
Bericht & Report
influence to predict effects in a general-
ized way” (Lucock & Yates, 2009). It re-
mains unclear whether the possible harm
of high folic acid levels outweighs the
known and potential benefits. Further-
more, this harm-benefit balance may dif-
fer across individuals and populations, by
genetic characteristics and by life stage
(Ulrich & Potter, 2006). Therefore, rec-
ommended folic acid supplementation
may need to be adapted to individual
genotypes (Van den Veyver, 2002), and
epigenetic DNA methylation profiles.
Yet, to individualize folic acid dietary
recommendations it seems necessary
to have a detailed understanding of all
genetic and physiological variables that
influence the interaction of folate with
the genome and their relationship to the
disease process (Stover & Garza, 2002).
Moreover, a thorough understanding of
the role of epigenetic variables in this in-
teraction seems crucial.
1 Institute for Public Health Genomics,
Maastricht University
2 Department for Nutritional Sciences,
University of Vienna, alexander.haslberg-
er@univie.ac.at
REFERENCES
Boccia, S. (2010). Methylenetetrahydrofolate Re-
ductase C677T and A1298C Polymorphisms and
Cancer Risk: A Review of the Published Meta-
Analyses. In A. G. Haslberger (Ed.), Epigenetics
and human health: Linking hereditary, environ-
mental and nutritional aspects (p. 42). Wein-
heim, Germany: Wiley-VCH Verlag GmbH &
Co. KgaA.
Cornel, M.C., de Smit, D.J., & de Jong-van den
Berg, L.T.W. (2005). Folic acid—the scientific
debate as a base for public health policy. Re-
productive Toxicology 20, 411–415. Retrieved
from http://www.sciencedirect.com/science/
article/pii/S089062380500119X
Crider, K.S., Bailey, L.B., & Berry, R.J. (2011).
Folic Acid Food Fortification—Its History, Ef-
fect, Concerns, and Future Directions. Nutri-
ents, 3(3), 370-384. doi:10.3390/nu3030370.
EFSA (2009). ESCO report prepared by the
EFSA Scientific Cooperation Working Group on
Analysis of Risks and Benefits of Fortification
of Food with Folic Acid. Retrieved from http://
www.efsa.europa.eu/en/scdocs/doc/3e.pdf
Eurocat (2009). Prevention of Neural Tube
Defects by Periconceptional Folic Acid Supple-
mentation in Europe (Updated version De-
cember 2009). Retrieved from http://www.
eurocat-network.eu/content/Special-Report-
NTD-3rdEd-Part-I.pdf
Berner, C., Aumüller, E., Gnauck , A., Nest-
elberger, M., Just, A., & Haslberger, A.G.
(2010). Epigenetic Control of Estrogen Recep-
tor Expression and Tumor Suppressor Genes
Is Modulated by Bioactive Food Compounds.
Ann Nutr Metab, 57(3-4), 183-189. doi:
10.1159/000321514
Haslberger, A., Varga, F., & Karlic, H. (2006).
Recursive causality in evolution: A model for
epigenetic mechanisms in cancer develop-
ment. Med Hypotheses, 67, 1448-1454. Re-
trieved from http://www.radioisotopos.ufrj.
br/radioiso/arquivos/haslberger_recursive _
causality.pdf
Mansuy, I.M., & Mohanna S. (2011). Epi-
genetics and the Human Brain: Where Nur-
ture Meets Nature. Cerebrum, May. Retrieved
from: http://dana.org/news/cerebrum/detail.
aspx?id=32670
Mathers, J.C. (2006). Nutritional modula-
tion of ageing: Genomic and epigenetic ap-
proaches. Mechanisms of Ageing and De-
velopment, 127(6), 584–589. doi: 10.1016/j.
mad.2006.01.018
Lucock, M., & Yates, Z. (2009). Folic acid
fortification: a double-edged sword. Cur-
rent Opinion in Clinical Nutrition & Meta-
bolic Care, 12(6), 555-564. doi: 10.1097/
MCO.0b013e32833192bc
Relton, C.L., & Smith, G.D. (2010). Epigen-
etic Epidemiology of Common Complex Dis-
ease: Prospects for Prediction, Prevention, and
Treatment. PLoS Med, 7(10), e1000356. doi:
10.1371/journal.pmed.1000356
Robitaille, J. (2009). Nutrigenomics and
Personalized Diet: What are the Antici-
pated Impacts for Research on Chronic
Diseases and Public Health. Current Phar-
macogenomics and Personalized Medicine,
7(2), 106-114(9). Retrieved from http://
www.ingentaconnect.com/content/ben /
cppm/2009/00000007/00000002/art00002
Schulte in den Baumen, T., & Brand, A. (2010).
Public Health Genomics: Integrating Genom-
ics and Epigenetics into national and European
health strategies and policies. In A. G. Hasl-
berger (Ed.), Epigenetics and human health:
Linking hereditary, environmental and nutri-
tional aspects (pp. 267-278). Weinheim, Ger-
many: Wiley-VCH Verlag GmbH & Co. KgaA.
Smith, A.D., Kim, Y.I., & Refsum, H. (2008). Is
folic acid good for everyone? Am J Clin Nutr,
87(3), 517-533. Retrieved from http://www.
ajcn.org/content/87/3/517.long
Stover, P.J., & Garza, C. (2002) Bringing Indi-
viduality to Public Health Recommendations.
The American Society for Nutritional Sciences
J. Nut.,132, 2476-2480. Retrieved from: http://
jn.nutrition.org/content/132/8/2476S.full
Stover, P.J., & Caudill, M.A. (2008). Genetic
and epigenetic contributions to human nutri-
tion and health: managing genome-diet inter-
actions. Journal of the American Dietetic As-
sociation, 108(9), 1480-1487. doi: 10.1016/j.
jada.2008.06.430
Ulrich, C.M., & Potter J.D. (2006). Folate Sup-
plementation: Too Much of a Good Thing?
Cancer Epidemiol Biomarkers Prev, 15(2); 189.
doi: 10.1158/1055-9965.EPI-06-0054
Van den Veyver, I.B. (2002). Genetic Effects of
Methylation Diets. Annu. Rev. Nutr, 22, 255-82.
doi: 10.1146/annurev.nutr.22.010402.102932
Vergères, G. (2010). From molecular nutrition
to nutritional systems biology. In A. G. Hasl-
berger (Ed.), Epigenetics and human health:
Linking hereditary, environmental and nutri-
tional aspects (pp. 127-139). Weinheim, Ger-
many: Wiley-VCH Verlag GmbH & Co. KgaA.
Herausgeber: Univ.-Prof. Dr. Kurt Widhalm, Öster-
reichisches Akademisches Institut für Ernährungs-
medizin, Alser Straße 14/1/4a, 1090 Wien;
Medizinische Universität Wien, Univ.-Klinik für
Kinder- und Jugendheilkunde, Währinger Gürtel
18-20, 1090 Wien
Chefredaktion: Dr. Karin Gruber, Tel. +43 1 512 44
86-23, Fax +43 1 512 44 86-24, E-Mail k.gruber@
aerzteverlagshaus.at
Redaktionsteam Wissenschaft:
Andrea Hofbauer, MSc., MBA, Präsidentin des Ver-
bandes der Diaetologen Österreichs; Univ.-Prof. Dr.
Bernhard Ludvik, Medizinische Universität Wien,
Univ.-Klinik für Innere Medizin III; Univ.-Prof. Dr.
Wolfgang Marktl, Akademie für Ganzheitsmedizin,
Wien; Univ.-Prof. Dr. Kurt Widhalm
Wissenschaftlicher Beirat: Univ.-Prof. Dr. Doris
Impressum
Balogh, Innsbruck; Univ.-Prof. Dr. Wilfred Druml,
Wien; Univ.-Prof. Dr. Robert Gasser, Graz; Univ.-
Prof. Dr. Alain Golay, Genf; Univ.-Prof. Dr. J.
Michael Hackl, Innsbruck; Univ.-Prof. Dr. Fritz Hop-
pichler, Salzburg; Univ.-Prof. Dr. Klaus-Dieter Jany,
Karlsruhe; Univ.-Prof. Dr. Michael Kunze, Wien;
Univ.-Prof. Dr. Monika Lechleitner, Innsbruck;
Univ.-Prof. Dr. Herbert Lochs, Innsbruck; Univ.-
Prof. Dr. Erich Roth, Wien; Univ.-Prof. Dr. Thomas
Stulnig, Wien; Univ.-Prof. Dr. Guntram Scherntha-
ner, Wien; Univ.-Prof. Dr. Hermann Toplak, Graz;
Univ.-Prof. Dr. Michael Trauner, Wien; Univ.-Doz.
Dr. Nicole Unger-Manhart, Wien; Dr. Volker Veitl,
Salzburg; Univ.-Prof. Dr. Thomas C. Wascher, Graz/
Wien
Anzeigenberatung: Agentur inSELL, Tel. 0676/525
21 54, E-Mail jemanzeigen@aerzteverlagshaus.at
Grafik & Layout: 1000ideen.at, DI Gerald Kasca,
Michael Zimmer, Humboldtstraße 21, 8010 Graz,
Tel +43 316 72 26 83, E-Mail office@1000ideen.at,
Homepage www.1000ideen.at;
Herstellung: Universitätsdruckerei Klampfer GmbH
Abonnement € 36,-, im Ausland zuzüglich Porto-
kosten
10.800 Druckauflage lt. ÖAK 1. Halbjahr 2011
Die in der Rubrik Sonder-
berichte und Promotion
erscheinenden Beiträge
sind bezahlt und berichten
über spezielle Produkte und
Methoden.
LCPUFAs IN SCHWANGER-
SCHAFT UND STILLZEIT
– Auswirkungen auf Kind und BMI
Publiziert & Kommentiert Muhlhausler B.S., Gibson R.A.,
Makrides M. Eect of long-
chain polyunsaturated fatty
acid supplementation during
pregnancy or lactation on
infant and child body com-
position: a systematic review.
American Journal of Clinical
Nutrition 2010: 92:857-863
EINFÜHRUNG
Langkettige, mehrfach ungesättigte Fett-
säuren (LCPUFAs) können die Zelldiffe-
renzierung und Fettspeicherung beim Er-
wachsenen hemmen. Es ergibt sich daraus
die Hypothese, dass eine LCPUFAs-Sup-
plementierung bei der schwangeren Mut-
ter die Fettmasse des Kindes im späteren
Leben reduzieren kann. Es gibt bereits ei-
nige randomisierte Studien, die den Effekt
von Omega-3-LCPUFAs-Supplementen
während Schwangerschaft und Stillzeit
auf die Körperzusammensetzung im
Säuglings- und Kindesalter untersuchten.
Bei den Studien zeigten sich sehr deutliche
Unterschiede bezüglich Studienqualität
und Studiendesign. Auch die Ergebnisse
waren sehr unterschiedlich. Dieser Review
soll auf die mangelhafte und fehlende Da-
tenlage in diesem Zusammenhang hin-
weisen. Kindliche Adipositas ist ein glo-
bales Problem. Zahlreiche Faktoren haben
einen zentralen Einfluss auf die Entwick-
lung. Vor allem das Ernährungsverhalten
der Mutter während der letzten Wochen
der Schwangerschaft sowie die Ernährung
des Säuglings nach der Geburt können die
Entstehung von Übergewicht und Adipo-
sitas signifikant beeinflussen. So spielt vor
allem die Zusammensetzung der Fettsäu-
ren in der Ernährung der Frau während
Schwangerschaft und Stillzeit für
die Körperzusammensetzung
des Säuglings eine entscheidende Rolle. In
dem vorliegenden Review wird der aktu-
elle Stand der Literatur zum Thema LCPU-
FAs-Supplemente während Schwanger-
schaft und Stillzeit und ihr Einfluss auf die
spätere Entwicklung und Körperzusam-
mensetzung des Säuglings besprochen.
METHODEN
In diversen Datenbanken wurde nach Hu-
manstudien zu diesem Thema gesucht.
Unter den passenden Artikeln wurden
sieben relevante Studien ausgewählt.
Wichtige Auswahlkriterien waren dabei:
maternale LCPUFAs-Supplementierung
für mindestens zwei Wochen, sowie Mes-
sungen von BMI und Prozent an Körper-
fett. Die Studien wurden aussortiert und
es blieben drei Studien übrig, die in die-
sem Review aufgenommen wurden.
RESULTATE
Es wurden drei Studien zusammengefasst.
In der ersten Studie wurde Olivenöl sup-
plementiert, in der zweiten Studie Mais-
keimöl und in der dritten Studie wurden
probiotische Supplemente und Vitamin-
tabletten mit LCPUFAs verabreicht. Die
LCPUFAs-Dosen lagen zwischen 0,2
und 1,18g pro Tag. Der Zeitpunkt der
Intervention lag zwischen der 18. und
21. Schwangerschaftswoche bis
zum dritten Lebensmonat.
Als Outcome wurden BMI,
BMI Z-Score und Haut-
faltendicke bestimmt.
Die Messungen er-
Dieser Review untersucht die Effekte
einer hohen Aufnahme von LCPUFAs-
Supplementen während Schwanger-
schaft und Stillzeit auf die Körperzusam-
mensetzung des Säuglings und Kindes.
Er weist auf einen Mangel an Studien
hin, die sich mit diesem Thema ausei-
nandersetzen. Es konnten nur drei Hu-
manstudien gefunden werden, die den
Auswahlkriterien entsprachen. Innerhalb
dieser drei Studien bestehen deutliche
Unterschiede bezüglich Studiendesign,
Studienqualität, Dauer und Zeitpunkt
der Intervention, sowie Outcome-Mes-
sungen. So waren auch die Ergebnisse
sehr unterschiedlich. Es kam zu einem
positiven, einem negativen und einem
neutralen Ergebnis. Derzeit ist es nicht
möglich, definitive Aussagen zu treffen
und die Empfehlung einer LCPUFAs-
Supplementierung während Schwan-
gerschaft und Stillzeit kann momentan
nicht ausgesprochen werden. Es besteht
ein deutlicher Mangel an gehaltvollen
Daten aus Humanstudien. Um das
bestehende Loch an aussagekräftigen
Informationen zu füllen, sind weitere
Langzeitstudien notwendig, und diese
sollten besser aufgebaut werden.
Conclusio
*Mag. Karin Gatternig, Univ.-Prof. Dr. Kurt
Widhalm, Österreichisches Akademisches
Institut für Ernährungsmedizin, Alserstraße
14/4a, 1090 Wien, E-Mail office@oeaie.org
folgten jeweils im 21. Lebensmonat, mit
2,5 Jahren und im 7. Lebensjahr. In al-
len drei Studien war die Drop-Out-Rate
sehr hoch. In der ersten Studie war der
BMI der Kinder im 21. Lebensmonat bei
gesteigerter Aufnahme von LCPUFAs
deutlich reduziert. In der zweiten Studie
zeigte sich kein Effekt durch die mater-
nale LCPUFAs-Supplementierung. Die
dritte Studie zeigte sogar ein negatives
Ergebnis, da eine maternale LCPUFAs-
Supplementierung während der Stillzeit
eine Erhöhung des BMI und Bauchum-
fangs mit 2,5 Jahren bedeutete.
LCPUFAs: Derzeit
noch keine defi-
nitiven Aussagen
möglich.
© Foto: Fotol ia/Anatoliy Samar a
30 JEM Dezember 2011
Bericht & Report
31 JEM Dezember 2011
Publiziert & Kommentiert
... In addition, several gene variants and other nutrients are interactive factors. Indeed, the new epigenetic evidence and the research on the functional role of polymorphism in the aetiology of NTDs and other adverse conditions in older people suggest that there is a need for a more personalised approach to preventive nutrition [39]. ...
Article
Full-text available
The term epigenetics describes mecha- nisms inducing changes in gene expres- sion or phenotype not caused by altera- tions in the underlying DNA sequence. Compared to the genome, which is al- most identical in different cell types and consistent throughout life, the epige- nome is varying between different cell types as well as over the course of a life- time (1). Twin studies demonstrated, that identical twin pairs, being epigenetically indistinguishable at early life exhibit stri- king differences in epigenetic patterns, as over time they accumulated signifi cant differences in global levels of epigenetic marks (2). It is becoming increasingly evident that distinct nutritional stimuli are able to alter the DNA confi guration at critical ontoge- nic stages as well as that these alterna- tions persist in gene expression. Occur- ring in the gametes these changes may be heritable and they are affected from
Article
Full-text available
Interactions between adaptative and selective processes are illustrated in the model of recursive causality as defined in Rupert Riedl's systems theory of evolution. One of the main features of this theory also termed as theory of evolving complexity is the centrality of the notion of 'recursive' or 'feedback' causality - 'the idea that every biological effect in living systems, in some way, feeds back to its own cause'. Our hypothesis is that "recursive" or "feedback" causality provides a model for explaining the consequences of interacting genetic and epigenetic mechanisms which are known to play a key role in development of cancer. Epigenetics includes any process that alters gene activity without changes of the DNA sequence. The most important epigenetic mechanisms are DNA-methylation and chromatin remodeling. Hypomethylation of so-called oncogenes and hypermethylation of tumor suppressor genes appear to be critical determinants of cancer. Folic acid, vitamin B12 and other nutrients influence the function of enzymes that participate in various methylation processes by affecting the supply of methyl groups into a variety of molecules which may be directly or indirectly associated with cancerogenesis. We present an example from our own studies by showing that vitamin D3 has the potential to de-methylate the osteocalcin-promoter in MG63 osteosarcoma cells. Consequently, a stimulation of osteocalcin synthesis can be observed. The above mentioned enzymes also play a role in development and differentiation of cells and organisms and thus illustrate the close association between evolutionary and developmental mechanisms. This enabled new ways to understand the interaction between the genome and environment and may improve biomedical concepts including environmental health aspects where epigenetic and genetic modifications are closely associated. Recent observations showed that methylated nucleotides in the gene promoter may serve as a target for solar UV-induced mutations of the p53 tumor suppressor gene. This illustrates the close interaction of genetic and epigenetic mechanisms in cancerogenesis resulting from changes in transcriptional regulation and its contribution to a phenotype at the micro- or macroevolutionary level. Above-mentioned interactions of genetic and epigenetic mechanisms in oncogenesis defy explanation by plain linear causality, things like the continuing adaptability of complex systems. They can be explained by the concept of recursive causality and has introduced molecular biology into the realm of cognition science and systems theory: based on the notion of so-called feedback- or recursive causality a model for epigenetic mechanisms with relevance for oncology and biomedicine is provided.
Article
Full-text available
Methylenetetrahydrofolate reductase (MTHFR) is a key enzyme in the metabolism of folate, whose role in gastric carcinogenesis is controversial. The authors performed a meta-analysis and individual data pooled analysis of case-control studies that examined the association between C677T and A1298C polymorphisms (the former being associated with low folate serum levels) and gastric cancer (meta-analyses: 16 studies, 2,727 cases and 4,640 controls for C677T and seven studies, 1,223 cases and 2,015 controls for A1298C; pooled analyses: nine studies, 1,540 cases and 2,577 controls for C677T and five studies, 1,146 cases and 1,549 controls for A1298C). An increased risk was found for MTHFR 677 TT in the meta-analysis (odds ratio (OR) = 1.52, 95% confidence interval (CI): 1.31, 1.77) and pooled analysis (OR = 1.49, 95% CI: 1.14, 1.95). No association resulted for MTHFR 1298 CC (meta-OR = 0.94, 95% CI: 0.65, 1.35; pooled OR = 0.90, 95% CI: 0.69, 1.34). Results from the pooled analysis of four studies on C677T stratified according to folate levels showed an increased risk for individuals with low (OR = 2.05, 95% CI: 1.13, 3.72) versus high (OR = 0.95, 95% CI: 0.54, 1.67) folate levels. Overall, these findings support the hypothesis that folate plays a role in gastric carcinogenesis.
Article
With parallel advances in genomics and in nutrition research, a new hybrid science has emerged at their intersection, often referred to as nutritional genomics which includes nutrigenomics and nutrigenetics. While the operational definitions will continue to evolve as nutritional genomics matures as a new field of inquiry, a central tenet will likely be on ways in which the human genome interacts with nutritional exposures. This emerging form of science has considerable implications for research in biomedicine and for public health. The progress in nutrigenomics research will likely increase our understanding of chronic disease etiology and the relationship between nutrients and common complex diseases. The potential for nutritional genomics in chronic disease prevention is also of great interest given that diet is a modifiable risk factor and because of the marked interindividual variability in response to the diet. The inclusion of genetic information as part of an overall strategy to improve the population health will be critical as more genomic data accumulate in nutrition science and used to develop recommendations for specific dietary requirements based on individual genetic make-up. In addition, use of genetic information from companies offering at-home direct-to-consumer nutrigenomics tests may have significant consequences for the population health and how we perceive and relate to food and other members of the society depending on human genetic variation. These implications of nutrigenomics and nutrigenetics such as the translation of research into public health practice will be discussed. Another dimension of interest is the need for well-trained health professionals. To this end, registered dietitians are essential to the efforts for incorporation of genomics into public health. Finally, future perspectives of nutrigenomics and nutrigenetics will be examined, with a view to how best to integrate the nascent field of nutrigenomics with established public health research and practices.
Article
The tumor suppressor genes p15(INK)⁴(b) and p16(INK)⁴(a) as well as the estrogen receptor-α (ESR1) gene are abnormally methylated and expressed in colon cancer. The cancer-preventative abilities of several bioactive food components have been linked to their estrogenic and epigenetic activities. The effect of folic acid, zebularine, resveratrol, genistein and epigallocatechin-3-gallate (EGCG) on tumor cell growth, promoter methylation of ESR1, p15(INK)⁴(b) and p16(INK)⁴(a) and gene expression of ESR1 and ESR2 was analyzed in Caco-2 cells. Gene expression was measured using real-time PCR, and promoter CpG methylation was assessed using bisulfite conversion and methylation-specific PCR. After exposure to a high concentration of folic acid (20 μmol/l), enhanced cancer cell growth and concomitant increased methylation of the ESR1 (3.6-fold), p16(INK)⁴(a) and p15(INK)⁴(b) promoters was observed. A lower concentration of folic acid (2 μmol/l) decreased cell growth. The phytoestrogens genistein and resveratrol enhanced expression of ESR1 (genistein 200 μmol/l: 2.1-fold; resveratrol 50 μmol/l: 6.3-fold) and ESR2 (2.6- and 3.6-fold, respectively). Genistein and resveratrol treatment increased promoter methylation of ESR1 (genistein 200 μmol/l: 2.9-fold; resveratrol 50 μmol/l: 1.4-fold). For p16(INK)⁴(a), increased methylation was found after exposure to 10 μmol/l resveratrol, but for p15(INK)⁴(b), decreased methylation was found. Both components showed growth-inhibitory activities. For EGCG, growth inhibition at 100 μmol/l and suppressed promoter methylation of tumor suppressor genes (p16(INK)⁴(a): 0.9-fold; p15(INK)⁴(b): 0.6-fold) was seen. Our results show that these food compounds regulate ESR and tumor suppressor gene expression by multiple mechanisms including epigenetic processes. An improved understanding of these epigenetic effects could therefore support specific dietary concepts of epigenetic cancer prevention and intervention.
Article
The Institute of Medicine recently convened a workshop to review the state of the various domains of nutritional genomics research and policy and to provide guidance for further development and translation of this knowledge into nutrition practice and policy. Nutritional genomics holds the promise to revolutionize both clinical and public health nutrition practice and facilitate the establishment of (a) genome-informed nutrient and food-based dietary guidelines for disease prevention and healthful aging, (b) individualized medical nutrition therapy for disease management, and (c) better targeted public health nutrition interventions (including micronutrient fortification and supplementation) that maximize benefit and minimize adverse outcomes within genetically diverse human populations. As the field of nutritional genomics matures, which will include filling fundamental gaps in knowledge of nutrient-genome interactions in health and disease and demonstrating the potential benefits of customizing nutrition prescriptions based on genetics, registered dietitians will be faced with the opportunity of making genetically driven dietary recommendations aimed at improving human health.
Article
The data generated from the human genome project offers unprecedented opportunities to elucidate the etiology of chronic diseases and developmental anomalies that arise from deleterious genome-diet interactions. Folate metabolism is an attractive system to explore such relationships. Folate is necessary for the synthesis of purine and thymidine deoxyribonucleotides and S-adenosylmethionine, a cofactor required for DNA methylation. Impaired folate metabolism results from primary folate deficiency, alcohol, gastrointestinal disorders that result in malabsorption, single nucleotide polymorphisms, increased folate catabolism and secondary nutrient deficiencies in vitamin B-6, vitamin B-12 and iron arising from a variety of pathologies. Any of these conditions singly or in combination influence DNA synthesis, DNA integrity, allelic-specific gene expression, chromatin structure and DNA mutation rates. Biochemical manifestations of impaired folate metabolism include increased uracil uptake into DNA, altered DNA methylation status and elevated homocysteine and S-adenosylhomocysteine in serum and tissues. These biochemical changes are associated with risk for cancer, cardiovascular disease, neural tube defects and some neuropathies and anemia, although direct causative mechanisms have not been established in all cases. Interactions between folate and the genome are reciprocal; polymorphisms in key genes influence folate nutritional requirements, indicating that dietary folate adequacy likely exerts selective pressure and thereby influences genetic variation. Other studies indicate that exposure to excess folate, perhaps at levels that occur at the upper end of the intake distribution curve, may have unintended consequences in promoting embryo viability. Therefore individualizing folic acid dietary recommendations necessitates a detailed understanding of all genetic and physiological variables that influence the interaction of folate with the genome and their relationship to the disease process.
Article
Randomized controlled trials have proven that periconceptional folic acid intake reduces the risk of neural tube defects (NTDs). This lead to different public health policies: fortification of foods in many countries and supplementation in some others. We concentrate here on pro's and con's of fortification policies. Meanwhile, new beneficial but also potential adverse effects are being hypothesized. Highest level evidence is available for the protective effect of folic acid on NTDs. Lower level evidence suggests other protective effects, but also some potential adverse effects, such as masking Vitamin B-12 deficiency, increasing twinning rates and an 'acceleration phenomenon' in pre-existing malignant neoplasms. While observational studies show lower cancer rates associated with increased folate intake, some case reports and animal experiments suggest opposite effects. Thus, public health policy makers are facing the question of balancing beneficial and potential adverse effects repeatedly. We propose that the scientific debate no longer focuses on NTDs alone, but that a comprehensive evaluation be undertaken by a public health authority with experience in complex meta-analyses and technology assessment.
Article
Dietary factors have a profound effect on many aspects of health including ageing and do so, at least partly, through interactions with the genome which result in altered gene expression. The application of high throughput genomics technologies in nutritional research (nutrigenomics) offers a new approach to understanding the molecular mechanisms by which nutrition affects ageing. To date, transcriptional profiling techniques have been applied in studies of the mode of action of energy (caloric) restriction. Two further areas which appear to be particularly promising are (i) nutritional modulation of DNA damage and repair and (ii) nutritional modulation of epigenetic markings. Epigenetic-mediated changes in gene expression in response to dietary and other lifestyle exposures appear to be a major molecular mechanism linking environmental factors with the genome with consequences for cell function and health throughout the life course.
ESCO report prepared by the EFSA Scientific Cooperation Working Group on Analysis of Risks and Benefits of Fortification of Food with Folic Acid
Folic Acid Food Fortification-Its History, Effect, Concerns, and Future Directions. Nutrients, 3(3), 370-384. doi:10.3390/nu3030370. EFSA (2009). ESCO report prepared by the EFSA Scientific Cooperation Working Group on Analysis of Risks and Benefits of Fortification of Food with Folic Acid. Retrieved from http:// www.efsa.europa.eu/en/scdocs/doc/3e.pdf Eurocat (2009). Prevention of Neural Tube Defects by Periconceptional Folic Acid Supplementation in Europe (Updated version December 2009). Retrieved from http://www. eurocat-network.eu/content/Special-ReportNTD-3rdEd-Part-I.pdf