Uploaded by troolja@yahoo.com

Methylation Diet&Lifestyle Kara Fitzerald, Romilly Hodgers

Methylation Diet and Lifestyle
Dr. Fitzgerald received her doctorate of naturopathic medicine from National College of Natural
Medicine in Portland, Oregon. She completed the first CNME-accredited post-doctorate
position in nutritional biochemistry and laboratory science at Metametrix (now Genova) Clinical
Laboratory under the direction of Richard Lord, Ph.D. Her residency was completed at
Progressive Medical Center, a large, integrative medical practice in Atlanta, Georgia. Dr.
Fitzgerald is lead author and editor of Case Studies in Integrative and Functional Medicine, a
contributing author to Laboratory Evaluations for Integrative and Functional Medicine and the
Institute for Functional Medicine’s updated Textbook for Functional Medicine. She has been published in numerous peerreviewed journals. Dr. Fitzgerald is on faculty at the Institute for Functional Medicine, and is an Institute for Functional
Medicine Certified Practitioner. She is a clinician researcher for The Institute for Therapeutic Discovery. Dr. Fitzgerald
regularly lectures internationally for several organizations and is in private practice in Sandy Hook, Connecticut.
Romilly Hodges completed her Master’s degree in Human Nutrition at the University of Bridgeport,
CT. She is a Certified Nutrition Specialist through the Board for the Certification of Nutrition
Specialists, and completed her practice hours under the supervision of Dr. Deanna Minich, PhD
and Dr. Kara Fitzgerald, ND. She has published in the Journal of Nutrition and Metabolism on
food-based modulators of detoxification enzymes, and has been a contributing author to Sinatra &
Houston, Nutritional and Integrative Strategies for Cardiovascular Medicine. She has been
teaching assistant to Dr. Minich for the Certified Food and Spirit Practitioner Program and the Food and Spirit Advanced
Detoxification Module. She is the staff nutritionist at the office of Dr. Kara Fitzgerald where she advises and supports
patients in the implementation of complex, multi-layered dietary and nutritional protocols that are uniquely personalized to
each individual’s needs.
© 2016 Kara Fitzgerald, ND
www.drkarafitzgerald.com
2
Methylation Diet and Lifestyle
All contents copyright © 2016 by Kara Fitzgerald, ND. All rights reserved. No part of this document or the related documents may be reproduced or
transmitted in any form, by any means (electronic, photocopying, recording, or otherwise) without the prior written permission of the publisher.
Disclaimer: No publication can be assumed to encompass the full scope of information that an individual practitioner brings to his or her practice and,
therefore, this book is not intended to be used as a clinical manual recommending specific treatments or tests for individual patients. It is intended for
use as an educational tool, to broaden the knowledge and perspective of the practitioner. It is the responsibility of the healthcare practitioner to make
his or her own determination of the usefulness and applicability of any information contained therein. If medical advice is required, the services of a
competent professional should be sought. The final decision to engage in any medical treatment should be made jointly by the patient and his or her
healthcare practitioner. Neither authors assume any liability for any errors or omissions or for any injury and/or damage to persons or property arising
from the use of information to be found in this publication.
© 2016 Kara Fitzgerald, ND
www.drkarafitzgerald.com
3
Methylation Diet and Lifestyle
TABLE OF CONTENTS
1 Abbreviations ..........................................................................................................................................................6
2 Preface: The Need For Conservative Yet Effective Methylation Support .......................................................8
3 Introduction ...........................................................................................................................................................11
4 An Introduction to Methylation ..........................................................................................................................14
4.1
The Biochemistry of Methylation ..........................................................................................................14
4.2
Endogenous Methylation Uses ..............................................................................................................15
4.3
DNA Methylation Plasticity ..................................................................................................................17
5 The Clinical Problem ............................................................................................................................................19
5.1
Methylation Deficits................................................................................................................................19
5.2
Folic Acid Concerns ................................................................................................................................22
5.3
Methyl Donor Intolerance ......................................................................................................................23
5.4
Methylation Gone Awry ........................................................................................................................25
5.5
Reason to Tread Carefully......................................................................................................................32
6 Assessment of Methylation Status ......................................................................................................................33
6.1
Genetic Profiling ......................................................................................................................................33
6.2
Nutrient status .........................................................................................................................................37
6.3
Methylation Metabolites ........................................................................................................................41
6.4
Nutrient Assimilation Capability..........................................................................................................43
6.5
Inflammation ...........................................................................................................................................43
6.6
Oxidative Stress .......................................................................................................................................44
7 The Methylation Diet and Lifestyle ....................................................................................................................46
7.1
Food-Based Nutrients .............................................................................................................................47
7.2
Support for Nutrient Assimilation ........................................................................................................51
7.3
Microbiome ..............................................................................................................................................52
7.4
Inflammation ...........................................................................................................................................53
7.5
Oxidative Stress .......................................................................................................................................54
7.6
Detoxification ...........................................................................................................................................55
7.7
Stress Management .................................................................................................................................57
7.8
Exercise .....................................................................................................................................................58
7.9
Nutraceutical Interactions ......................................................................................................................60
© 2016 Kara Fitzgerald, ND
www.drkarafitzgerald.com
4
Methylation Diet and Lifestyle
7.10
Medication Interactions ..........................................................................................................................61
8 The Methylation Diet Food Plan .........................................................................................................................63
9 Menu Plans.............................................................................................................................................................71
10
9.1
Sample Menu 1: Gluten-Free, Dairy-Free ............................................................................................71
9.2
Sample Menu 2: Paleo.............................................................................................................................72
9.4
Menu Nutrient Analyses ........................................................................................................................73
RECIPES ............................................................................................................................................................74
Breakfast ................................................................................................................................................................75
Lunch .....................................................................................................................................................................85
Dinner ....................................................................................................................................................................96
Side Dishes ..........................................................................................................................................................106
Snacks...................................................................................................................................................................114
Smoothies & Juices .............................................................................................................................................121
11
Appendix A: Checklist for Methylation Assessment ................................................................................128
12
Appendix B: Checklist for Methylation Interventions ..............................................................................129
13
References .......................................................................................................................................................130
© 2016 Kara Fitzgerald, ND
www.drkarafitzgerald.com
5
Methylation Diet and Lifestyle
1 ABBREVIATIONS
AHYC
AGE
AIP
ALU
ATP
ADD
ADHD
Bcl-2
BHMT
BH2
BH4
BMI
BMPRII
CBS
CDC
CIDP
COMT
CoQ10
CpG
CRP
DFE
DHA
DHF
DHFR
DNA
DNMT
EGCG
Fli-1
FODMAPs
FR
GI
HPA axis
HVA
IgE
IL
Adenosyl homocysteinase
Advanced glycation end-product
Autoimmune pancreatitis
Arthrobacter luteus
Adenoise triphosphate
Attention deficit disorder
Attention deficit hyperactivity disorder
B-cell lympoma 2
Betaine homocysteine methyltransferase
Dihydrobiopterin
Tetrahydrobiopterin
Body mass index
Bone morphogenetic protein receptor 2
Cystathionine beta-synthase
Centers for Disease Control and Prevention
Chronic inflammatory demyelinating polyneuropathy
Catechol-O-methyltransferase
Ubiquinone or coenzyme Q10
C: Cytosine triphosphate deoxynucleotide p: phosphodeister, G:
guanine triphosphate deoxynucleotide
C-reactive protein
Dietary folate equivalents
Docosahexaenoic acid
Dihydrofolate
Dihydrofolate reductase
Deoxyribonucleic acid
Deoxyribonucleic methyltransferase
Epigallocatechin 3-gallate
Friend leukemia integration 1 transcription
Fermentable Oligo –Di- Monosaccharides and Polyols
Folate receptor
Gastrointestinal
Hypothalamic-pituitary-adrenal axis
Homovanillate
Immunoglobulin E
Interleukin
© 2016 Kara Fitzgerald, ND
www.drkarafitzgerald.com
6
Methylation Diet and Lifestyle
KLF5
L-Dopa
MBP
5-mC
5-hmC
MAO
MAT
MDL
MS
MST1
MTFHR
MTHF
MTRR
MTR
MTases
NK
NRF2
NFkB
NHANES
NLRP3
8-OHdG
PABA
POP
RBC
REST
RNA
SNP
SAH
SAHH
SAMe
SIBO
SAHH
TET
TNF
UMFA
VMA
Kruppel-like factor 5
Levodopa or L-3,4-dihydroxyphenylalanine
Mega base pair
5-methylcystosine
5-hydroxymethylcystosine
Monoamine oxidase
Methionine adenosyltransferase
Methylation Diet and Lifestyle
Methionine synthase
Macrophage stimulating 1
Methylene tetrahydrofolate reductase
Methyltetrahydrofolate
Methyl tetrahydrofolate-homocysteine methyltransferase
reductase
Methyl tetrahydrofolate-homocysteine methyltransferase
SAMe dependent methyltransferases
Natural killer
Nuclear factor erythroid 2 [NF-E2]-related factor 2
Nuclear factor kappa-B
National health and nutritional examination survey
Nod-like receptor protein 3
8-hydroxy-2’-deoxyguanosine
Para-aminobenzoic acid
Persistent organic pollutants
Red blood cell
RE1-silencing transcription factor
Ribonucleic acid
Single nucleotide polymorphism
S-adenosyl homocysteine
S- adenosyl homocysteine hydrolase
S-adenosyl methionine
Small intestine bacterial overgrowth
S-adenosyl-L-homocysteine hydrolase
Ten-eleven translocation
Tumor necrosis factor
Unmetabolized folic acid, sometimes referred to as folic acid,
synthetic folate or oxidized folate
Vanilmandelic acid
© 2016 Kara Fitzgerald, ND
www.drkarafitzgerald.com
7
Methylation Diet and Lifestyle
2 PREFACE: THE NEED FOR CONSERVATIVE YET EFFECTIVE
METHYLATION SUPPORT
From the desk of Dr. Kara Fitzgerald, ND
With the mapping of the human genome in 2001, the era of Systems Medicine has burst forth.
At about 24,000 genes, the code for our lives was smaller than expected (you’ve no doubt heard
the humorous comparison: “less than a grape, but more than a chicken”). Significant attention is
now being paid to the regulatory aspects of the genome, and research on the heritable
epigenome in particular is galloping ahead. At this writing, a PubMed search on
“methylation”—a cornerstone epigenetic and metabolic process—yields more than 85,000 hits.
Many genetic methylation research abstracts are summarized with the preface statement, “For
the first time ever, we’ve shown that…”
With rapidly emerging understandings, and ready access to genetic testing, it is indeed a wildly
exciting time to be practicing Functional Medicine, the clinical application of Systems Medicine.
Many of our patients now arrive at our offices with some knowledge of their genetics, and most
often with regard to their single nucleotide polymorphisms (SNPs) relating to methylation.
Many assumptions are made around the phenotypic expression of these single nucleotide
variations. Last week, a new patient sat before me, stating with grim determinism that her
heterozygous mutation in MTHFR A1298C SNP made it impossible for her body to detoxify
efficiently.
To those of us looking for biochemical “lesions” in methylation enzymes using a combination of
genetic testing and the currently available metabolic biomarkers they appear to be common...
Sometimes correcting identified imbalances can be highly clinically significant, other times
(arguably more often than not) shifting biochemical methylation activity with high-dose methyl
donors such as folate and B12 doesn’t appear to have a huge, immediate clinical outcome. But
we know that methylation is incredibly fundamental and operates in many “behind the
scenes” activities not limited to detoxification, neurotransmitter production and of course the
all-important black box of epigenetic regulation. We’ve seen the powerful impacts of folic acid
fortification in the reduction of neural tube defects, where augmenting methylation activity is a
piece of the folic acid success story.
The spark of inspiration behind creating the Methylation Diet and Lifestyle (MDL) program
was wanting to “do right by my patients” by embracing both the importance of healthy
methylation balance while recognizing the limitations of our current understanding. What
we don’t know in the biological sciences always far outweighs what we do know, and this is
especially true in our understanding of methylation. Imbalanced methylation of the epigenome
(comprising both hyper- and hypomethylation primarily of gene promoter regions) is an
© 2016 Kara Fitzgerald, ND
www.drkarafitzgerald.com
8
Methylation Diet and Lifestyle
emerging area of investigation implicated in many disease processes, ranging from aging and
allergies to neurodegenerative processes and cancer. Regulation of the epigenome is a highly
complex process; we cannot state with certainty what the impact is of high-dose, long term
methylation interventions.
Thus, for the vast majority of our patients we need to consider whether high-dose, long-term
supplementation really is the right approach. There is a dearth of research in this area, with
the exception of folic acid, where, as we discuss below, reasonable concerns exist. There are also
those patients who do not tolerate methyl donor supplementation, suspected to be chiefly due
to either poor clearance of epinephrine and other biogenic amines, or “ramped up”
detoxification activity.
While homage to Dr. Bruce Ames and his remarkable work around increasing enzyme kinetics
using high dose cofactor supplementation is due, and understanding that this approach may
indeed be appropriate for methylation in the short-term, our emerging understanding on the
epigenome suggests that more “up-stream” and nuanced interventions are in order when
possible. Food-based folates, for instance, have only been shown to be protective. Numerous
additional phytochemicals, not directly tied to methylation, appear to favorably modulate
global epigenetic and biochemical methylation activity. And reducing methyl donor depletion
by minimizing toxic exposures, nourishing the microbiome, augmenting the stress response and
far more, is safe and impactful. By removing the obstacles and drains on methylation while
providing broad spectrum nutrient ingredients for balanced activity, we are allowing
physiological wisdom to manage the process.
The Methylation Diet and Lifestyle would not have been brought into being in my clinical
practice or in this eBook without the years of frank discussions I’ve had with leading expert
Michael Stone, MD on current research and patient care approaches1. Likewise with my Journal
Club comrades, where each month for over the last three years we meet and “tussle with” new,
often complex concepts in the biological sciences. A heart-felt “shout out” goes to my
nutritionist extraordinaire, Romilly Hodges, who has worked tirelessly with me to capture the
bulk of our thoughts in the below text.
This eBook serves as a guide to understand the current methylation issues and challenges facing
practitioners, the potential concerns with supplementation alone, how to assess methylation
Drs. Leslie and Michael Stone, along with their daughter, nutritionist Emily Rydbom, are doing
remarkable work in the field of methylation assessment and support during preconception, pregnancy
and the postnatal period at their clinic in Ashland, Oregon. Find them at www.ashlandmd.com and
www.growbabyhealth.com.
1
© 2016 Kara Fitzgerald, ND
www.drkarafitzgerald.com
9
Methylation Diet and Lifestyle
status, and how to incorporate an MDL program into your protocols. Not only are there easyto-apply menus, recipes and nutrient guidelines but important lifestyle factors are reviewed
as well.
How should we determine when to use the MDL? Well, it may be the optimal way to offer
methylation support right off the bat, especially for those who cannot tolerate
supplementation. It is also a sensible and safe long-term strategy for most patients who have
achieved balance through a short-term course of higher-dose methyl donors. Importantly, the
full MDL program also supports detoxification, stress reduction, microbiome and hormone
balance, and can be readily modified to incorporate other programs that we routinely prescribe
such as elimination diets, grain and lectin-free plans, low FODMAP diets and general gut
restoration programs. Highly restricted plans, such as the calorie restricted ketogenic diet,
sometimes used in cancer and epilepsy, can incorporate aspects of the methylation diet with
additional nutraceutical support. Virtually any dietary program can work with the MDL as a
foundation.
Acknowledgements
We would like to acknowledge P. Michael Stone, MD, MS,
Institute for Functional Medicine, for his groundbreaking work in
the area of methylation.
We would also like to thank Lara Zakaria MS for her
contributions to the menus, recipes and figures, and Brigid Krane
(www.brigidkrane.com) for the cover and logo design.
© 2016 Kara Fitzgerald, ND
www.drkarafitzgerald.com
10
Methylation Diet and Lifestyle
3 INTRODUCTION
It has become increasingly common for functional medicine practitioners to recommend highdose supplementation with methyl donors, such as 5-methyltetrahydrofolate (5-MTHF) and
methylcobalamin, in certain patients. For example in those with genetic polymorphisms that
may impair the functionality of relevant enzymes, most commonly methylenetetrahydrofolate
reductase (MTHFR), or in those with out-of-range methylation-related biomarkers such as in
hyperhomocysteinemia. Deficiency in methyl donors is a fairly frequent finding in laboratory
analyses, depending on your population, and can relate directly to clinical symptoms; for
example B12 deficiency-associated neuropathy, which is relatively common. Some practitioners
may even look to supporting methylation as a means to improve inherited or environmentally
acquired epigenetic programming.
Of course, improving global methylation status and averting the pathways of disease and
dysfunction associated with a potential deficit in methylation activity is a commendable goal.
However, as with other biochemical processes, methylation activity exists ideally in a state of
active balance, or homeodynamics. Imbalance in these mechanisms can lead to dysfunction and
disease. So while we can be confident that ensuring the adequacy of available methyl donors for
use in the body is important, we have to question whether “pushing” reaction rates using
supraphysiological doses is always safe. Rather than bluntly forcing reactions forward, perhaps
our ultimate goal should instead be enabling the body to do the right thing at the right time.
There are a number of potential issues with long-term high-dose methyl-donor
supplementation:
The impact of genetic alterations is unclear. Aside from the altered activity of MTHFR C677T
and A1298C single nucleotide polymorphisms (SNPs) which has indeed been studied to some
degree, the discovery of other SNPs remains a qualitative rather than quantitative indicator of
enzyme functionality. The overall effect of these SNPs on methylation depends on the activity
of many enzymes working together in the context of one’s internal and external environment.
This fact underlies the many variable results found in genome-wide association studies. As a
result, it isn’t readily possible at this time to determine exactly how much of an impact any one
of those alterations, even MTHFR C677T, has on overall methylation status [1].
The correct supplementation dose is as yet unknown, and may vary between individuals. No
studies have clarified yet what the right dosage or duration of methyl donor supplementation is
needed to rebalance biochemical or epigenetic methylation status Some side effects of high-dose
5-methyltetrahydrofolate supplementation have been reported in clinical practice, including
worsening of symptoms and anxiety. Consequently it may be safer in the long term to stay
within physiologic levels [2].
© 2016 Kara Fitzgerald, ND
www.drkarafitzgerald.com
11
Methylation Diet and Lifestyle
Hypermethylation states may occur and may also be detrimental: The scientific literature
contains many examples of region-specific DNA hypermethylation associated with adverse
outcomes, including cancer, immune dysfunction and Downs Syndrome. Interestingly, both
DNA hyper- and hypomethylation states can be identified in states of methyl donor deficiency
and repletion. Certainly folic acid, in either deficiency or excess, has been associated with
increased rates of cancer and immune hypersensitivity, but since the mechanism is not fully
understood, should we not also be cautious with high levels of methylated folate? The
mechanisms controlling DNA methylation and demethylation are incredibly complex, yet we
know that pushing reaction rates through supraphysiological dosing of nutrient cofactors is
possible [3]. The bottom line is that we don’t know what effect long-term, high-dose methylfactor supplementation has on DNA methylation.
Methylation status depends on many dietary and lifestyle inputs: A complex interaction of
dietary and lifestyle factors (including medications, stress, sleep, exercise and toxin exposure)
plays a role in the methylation end-result. Singular interventions with high dose nutrient
supplementation miss this intricate web of inputs and may lack long-term effectiveness, or may
not achieve desired results.
Dietary and lifestyle interventions may be the best, and safest, long term option for most
individuals with suspected methylation imbalances. This may be particularly true for certain
vulnerable populations such as individuals with active cancers. Aging is known to be associated
with diminished methylation activity, so there is a good rationale for using the MDL as an antiaging tool. Careful methylation evaluation and treatment is essential during preconception,
pregnancy and the postnatal period2. A good evaluation coupled with the MDL program and
appropriate nutraceutical support is a useful starting place.
A dietary and lifestyle approach can also be used as a long term follow-up plan to those
requiring early high-dose nutraceutical support. Food sources of methylation nutrients can be
abundant, and with proper planning, dietary modifications have been found to favorably
influence methylation activity [4], [5].
In this eBook, you’ll find a comprehensive dietary and lifestyle approach to support
methylation, aligned with a functional medicine approach and including important methylation
nutrients, specific foods to include, foods to avoid, two 7-day menu plans (with calculated
2
Drs. Leslie and Michael Stone, along with their daughter, nutritionist Emily Rydbom, are doing
remarkable work in the field of methylation assessment and support during preconception, pregnancy
and the postnatal period at their clinic in Ashland, Oregon. Find them at www.ashlandmd.com and
www.growbabyhealth.com.
© 2016 Kara Fitzgerald, ND
www.drkarafitzgerald.com
12
Methylation Diet and Lifestyle
levels of methylation nutrients), recipes and lifestyle factors. You’ll also learn the basic
biochemistry of methylation, the many roles of methylation in the body, how to assess patient
methylation status, the beneficial and potentially detrimental clinical impacts of supplemental
methyl donor interventions, and the potential issues with too little, or too much methylation
activity.
© 2016 Kara Fitzgerald, ND
www.drkarafitzgerald.com
13
Methylation Diet and Lifestyle
13 REFERENCES
[1]
M. A. Hall, “Beyond genome-wide association studies (GWAS): Emerging methods for investigating complex
associations for common traits,” THE PENNSYLVANIA STATE UNIVERSITY, 2015.
[2]
K. Smith, “DNA Methylation and the Global Genome,” Townsend Lett., vol. June, pp. 57–61, 2015.
[3]
B. N. Ames, I. Elson-Schwab, and E. A. Silver, “High-dose vitamin therapy stimulates variant enzymes with
decreased coenzyme binding affinity (increased K(m)): relevance to genetic disease and polymorphisms.,”
Am. J. Clin. Nutr., vol. 75, no. 4, pp. 616–58, 2002.
[4]
K. V. Donkena, C. Y. F. Young, and D. J. Tindall, “Oxidative stress and DNA methylation in prostate cancer.,”
Obstet. Gynecol. Int., vol. 2010, p. 302051, Jan. 2010.
[5]
T. M. Hardy and T. O. Tollefsbol, “Epigenetic diet: impact on the epigenome and cancer.,” Epigenomics, vol. 3,
no. 4, pp. 503–18, Aug. 2011.
[6]
P. M. Stone, “Functional Medicine: Laboratory Evaluation of Methylation Defects.,” 2014.
[7]
C. M. Pfeiffer, M. R. Sternberg, Z. Fazili, E. A. Yetley, D. A. Lacher, R. L. Bailey, and C. L. Johnson,
“Unmetabolized folic acid is detected in nearly all serum samples from US children, adolescents, and
adults.,” J. Nutr., vol. 145, no. 3, pp. 520–31, Mar. 2015.
[8]
S. W. Bailey and J. E. Ayling, “The extremely slow and variable activity of dihydrofolate reductase in human
liver and its implications for high folic acid intake.,” Proc. Natl. Acad. Sci. U. S. A., vol. 106, no. 36, pp. 15424–
9, Sep. 2009.
[9]
R. Obeid, W. Holzgreve, and K. Pietrzik, “Is 5-methyltetrahydrofolate an alternative to folic acid for the
prevention of neural tube defects?,” J. Perinat. Med., vol. 41, no. 5, pp. 469–83, Sep. 2013.
[10]
V. T. Ramaekers, B. Thöny, J. M. Sequeira, M. Ansseau, P. Philippe, F. Boemer, V. Bours, and E. V. Quadros,
“Folinic acid treatment for schizophrenia associated with folate receptor autoantibodies,” Mol. Genet. Metab.,
vol. 113, no. 4, pp. 307–314, 2014.
[11]
A. Imbard, J.-F. Benoist, and H. J. Blom, “Neural tube defects, folic acid and methylation.,” Int. J. Environ. Res.
Public Health, vol. 10, no. 9, pp. 4352–89, 2013.
[12]
I. P. Pogribny, L. Muskhelishvili, B. J. Miller, and S. J. James, “Presence and consequence of uracil in
preneoplastic DNA from folate/methyl-deficient rats.,” Carcinogenesis, vol. 18, no. 11, pp. 2071–6, Nov. 1997.
[13]
Geneva: World Health Organization, “Guideline: Optimal Serum and Red Blood Cell Folate Concentrations
in Women of Reproductive Age for Prevention of Neural Tube Defects,” 2015.
[14]
C. A. M. Atta, K. M. Fiest, A. D. Frolkis, N. Jette, T. Pringsheim, C. St Germaine-Smith, T. Rajapakse, G. G.
Kaplan, and A. Metcalfe, “Global Birth Prevalence of Spina Bifida by Folic Acid Fortification Status: A
Systematic Review and Meta-Analysis.,” Am. J. Public Health, vol. 106, no. 1, p. 159, Jan. 2016.
[15]
H. Meng, Y. Cao, J. Qin, X. Song, Q. Zhang, Y. Shi, and L. Cao, “DNA Methylation, Its Mediators and
Genome Integrity.,” Int. J. Biol. Sci., vol. 11, no. 5, pp. 604–617, Jan. 2015.
[16]
A. Karpf (ed), Epigenetic Alterations in Oncogenesis. 2013.
[17]
M. Yang and J. Y. Park, “DNA methylation in promoter region as biomarkers in prostate cancer.,” Methods
Mol. Biol., vol. 863, pp. 67–109, Jan. 2012.
© 2016 Dr. K Fitzgerald, ND
www.drkarafitzgerald.com
130
Methylation Diet and Lifestyle
[18]
M. S. Trivedi and R. C. Deth, “Role of a redox-based methylation switch in mRNA life cycle (pre- and posttranscriptional maturation) and protein turnover: implications in neurological disorders.,” Front. Neurosci.,
vol. 6, p. 92, Jan. 2012.
[19]
C. B. Wilson, K. W. Makar, M. Shnyreva, and D. R. Fitzpatrick, “DNA methylation and the expanding
epigenetics of T cell lineage commitment.,” Semin. Immunol., vol. 17, no. 2, pp. 105–19, Apr. 2005.
[20]
Y. Guo, S. Yu, C. Zhang, and A.-N. Tony Kong, “Epigenetic regulation of Keap1-Nrf2 signaling.,” Free Radic.
Biol. Med., Jun. 2015.
[21]
J. D. Yager, “Mechanisms of estrogen carcinogenesis: The role of E2/E1-quinone metabolites suggests new
approaches to preventive intervention - A review.,” Steroids, vol. 99, no. Pt A, pp. 56–60, Jul. 2015.
[22]
R. Lord and J. Bralley, Laboratory Evaluations for Integrative and Functional Medicine, Revised 2n. Duluth, GA:
Metametrix Institute, 2012.
[23]
R. Surtees, J. Leonard, and S. Austin, “Association of demyelination with deficiency of cerebrospinal-fluid Sadenosylmethionine in inborn errors of methyl-transfer pathway.,” Lancet (London, England), vol. 338, no.
8782–8783, pp. 1550–4, Jan. 1991.
[24]
R. A. Waterland, D. C. Dolinoy, J.-R. Lin, C. A. Smith, X. Shi, and K. G. Tahiliani, “Maternal methyl
supplements increase offspring DNA methylation at Axin Fused.,” Genesis, vol. 44, no. 9, pp. 401–6, Sep.
2006.
[25]
R. Barres and J. R. Zierath, “DNA methylation in metabolic disorders.,” Am. J. Clin. Nutr., vol. 93, no. 4, p.
897S–900, Apr. 2011.
[26]
D. C. Dolinoy, R. Das, J. R. Weidman, and R. L. Jirtle, “Metastable epialleles, imprinting, and the fetal origins
of adult diseases.,” Pediatr. Res., vol. 61, no. 5 Pt 2, p. 30R–37R, 2007.
[27]
R. A. Waterland, “Assessing the effects of high methionine intake on DNA methylation.,” J. Nutr., vol. 136,
no. 6 Suppl, p. 1706S–1710S, Jun. 2006.
[28]
O. S. Anderson, K. E. Sant, and D. C. Dolinoy, “Nutrition and epigenetics: an interplay of dietary methyl
donors, one-carbon metabolism and DNA methylation.,” J. Nutr. Biochem., vol. 23, no. 8, pp. 853–9, 2012.
[29]
Y. Luo, X. Lu, and H. Xie, “Dynamic Alu methylation during normal development, aging, and
tumorigenesis.,” Biomed Res. Int., vol. 2014, p. 784706, 2014.
[30]
D. Ingrosso, A. Cimmino, A. F. Perna, L. Masella, N. G. De Santo, M. L. De Bonis, M. Vacca, M. D’Esposito,
M. D’Urso, P. Galletti, and V. Zappia, “Folate treatment and unbalanced methylation and changes of allelic
expression induced by hyperhomocysteinaemia in patients with uraemia,” Lancet, vol. 361, no. 9370, pp.
1693–1699, 2003.
[31]
I. C. G. Weaver, “Reversal of Maternal Programming of Stress Responses in Adult Offspring through Methyl
Supplementation: Altering Epigenetic Marking Later in Life,” J. Neurosci., vol. 25, no. 47, pp. 11045–11054,
2005.
[32]
D. Cai, Y. Jia, J. Lu, M. Yuan, S. Sui, H. Song, and R. Zhao, “Maternal dietary betaine supplementation
modifies hepatic expression of cholesterol metabolic genes via epigenetic mechanisms in newborn piglets,”
Br. J. Nutr., vol. 112, no. 09, pp. 1459–1468, Sep. 2014.
[33]
V. Bollati, A. Baccarelli, L. Hou, M. Bonzini, S. Fustinoni, D. Cavallo, H.-M. Byun, J. Jiang, B. Marinelli, A. C.
Pesatori, P. A. Bertazzi, and A. S. Yang, “Changes in DNA methylation patterns in subjects exposed to lowdose benzene.,” Cancer Res., vol. 67, no. 3, pp. 876–80, Feb. 2007.
[34]
B. Stefanska, H. Karlic, F. Varga, K. Fabianowska-Majewska, and A. Haslberger, “Epigenetic mechanisms in
anti-cancer actions of bioactive food components – the implications in cancer prevention,” Br. J. Pharmacol.,
vol. 167, pp. 279–297, 2012.
© 2016 Dr. K Fitzgerald, ND
www.drkarafitzgerald.com
131
Methylation Diet and Lifestyle
[35]
A. M. Cordero, K. S. Crider, L. M. Rogers, M. J. Cannon, and R. J. Berry, “Optimal serum and red blood cell
folate concentrations in women of reproductive age for prevention of neural tube defects: World Health
Organization guidelines.,” MMWR. Morb. Mortal. Wkly. Rep., vol. 64, no. 15, pp. 421–3, Apr. 2015.
[36]
A. Greißel, M. Culmes, R. Napieralski, E. Wagner, H. Gebhard, M. Schmitt, A. Zimmermann, H.-H. Eckstein,
A. Zernecke, and J. Pelisek, “Alternation of histone and DNA methylation in human atherosclerotic carotid
plaques.,” Thromb. Haemost., vol. 114, no. 2, May 2015.
[37]
M. K. Skinner, “Environmental epigenomics and disease susceptibility.,” EMBO Rep., vol. 12, no. 7, pp. 620–
622, 2011.
[38]
Y.-L. Wu, C.-Y. Hu, S.-S. Lu, F.-F. Gong, F. Feng, Z.-Z. Qian, X.-X. Ding, H.-Y. Yang, and Y.-H. Sun,
“Association between methylenetetrahydrofolate reductase (MTHFR) C677T/A1298C polymorphisms and
essential hypertension: a systematic review and meta-analysis.,” Metabolism., vol. 63, no. 12, pp. 1503–11,
2014.
[39]
K. Szarc vel Szic, M. N. Ndlovu, G. Haegeman, and W. Vanden Berghe, “Nature or nurture: let food be your
epigenetic medicine in chronic inflammatory disorders.,” Biochem. Pharmacol., vol. 80, no. 12, pp. 1816–32,
Dec. 2010.
[40]
NIH, “Folate Dietary Supplement Fact Sheet,” 2012.
[41]
L. C. Hu XW, Qin SM, Li D, Hu LF, “Elevated homocysteine levels in levodopa-treated idiopathic
Parkinson’s disease: a meta-analysis. - PubMed - NCBI,” Acta Neurol Scand, 2013. [Online]. Available:
http://www.ncbi.nlm.nih.gov/pubmed/23432663. [Accessed: 21-Dec-2015].
[42]
S. Gilbody, S. Lewis, and T. Lightfoot, “Methylenetetrahydrofolate Reductase (MTHFR) Genetic
Polymorphisms and Psychiatric Disorders: A HuGE Review,” Am. J. Epidemiol., vol. 165, no. 1, pp. 1–13, 2006.
[43]
A. D. Smith, Y.-I. Kim, and H. Refsum, “Is folic acid good for everyone?,” Am. J. Clin. Nutr., vol. 87, no. 3, pp.
517–33, Mar. 2008.
[44]
P. Frosst, H. J. Blom, R. Milos, P. Goyette, C. A. Sheppard, R. G. Matthews, G. J. Boers, M. den Heijer, L. A.
Kluijtmans, and L. P. van den Heuvel, “A candidate genetic risk factor for vascular disease: a common
mutation in methylenetetrahydrofolate reductase.,” Nat. Genet., vol. 10, no. 1, pp. 111–3, May 1995.
[45]
N. M. van der Put, F. Gabreëls, E. M. Stevens, J. A. Smeitink, F. J. Trijbels, T. K. Eskes, L. P. van den Heuvel,
and H. J. Blom, “A second common mutation in the methylenetetrahydrofolate reductase gene: an additional
risk factor for neural-tube defects?,” Am. J. Hum. Genet., vol. 62, no. 5, pp. 1044–51, 1998.
[46]
B. L. Tsang, O. J. Devine, A. M. Cordero, C. M. Marchetta, J. Mulinare, P. Mersereau, J. Guo, Y. P. Qi, R. J.
Berry, J. Rosenthal, K. S. Crider, and H. C. Hamner, “Assessing the association between the
methylenetetrahydrofolate reductase (MTHFR) 677C>T polymorphism and blood folate concentrations: a
systematic review and meta-analysis of trials and observational studies.,” Am. J. Clin. Nutr., vol. 101, no. 6,
pp. 1286–94, Jun. 2015.
[47]
R. L. Bailey, K. W. Dodd, J. J. Gahche, J. T. Dwyer, M. A. McDowell, E. A. Yetley, C. A. Sempos, V. L. Burt, K.
L. Radimer, and M. F. Picciano, “Total folate and folic acid intake from foods and dietary supplements in the
United States: 2003-2006.,” Am. J. Clin. Nutr., vol. 91, no. 1, pp. 231–7, Jan. 2010.
[48]
R. L. Bailey, M. A. McDowell, K. W. Dodd, J. J. Gahche, J. T. Dwyer, and M. F. Picciano, “Total folate and folic
acid intakes from foods and dietary supplements of US children aged 1-13 y.,” Am. J. Clin. Nutr., vol. 92, no.
2, pp. 353–8, Aug. 2010.
[49]
J. J. Otten, J. P. Hellwig, and D. Linda, Dietary DRI Reference Intakes. 2006.
[50]
K. E. Christensen, L. G. Mikael, K.-Y. Leung, N. Lévesque, L. Deng, Q. Wu, O. V Malysheva, A. Best, M. A.
Caudill, N. D. E. Greene, and R. Rozen, “High folic acid consumption leads to pseudo-MTHFR deficiency,
© 2016 Dr. K Fitzgerald, ND
www.drkarafitzgerald.com
132
Methylation Diet and Lifestyle
altered lipid metabolism, and liver injury in mice.,” Am. J. Clin. Nutr., vol. 101, no. 3, pp. 646–58, Mar. 2015.
[51]
M. Aarabi, M. C. San Gabriel, D. Chan, N. A. Behan, M. Caron, T. Pastinen, G. Bourque, A. J. MacFarlane, A.
Zini, and J. Trasler, “High dose folic acid supplementation alters the human sperm methylome and is
influenced by the MTHFR C677T polymorphism.,” Hum. Mol. Genet., Aug. 2015.
[52]
M.-H. Tsai, W.-C. Chen, S.-L. Yu, C.-C. Chen, T.-M. Jao, C.-Y. Huang, S.-T. Tzeng, S.-J. Yen, and Y.-C. Yang,
“DNA Hypermethylation of SHISA3 in Colorectal Cancer: An Independent Predictor of Poor Prognosis.,”
Ann. Surg. Oncol., May 2015.
[53]
F. Coppedè, “Epigenetic biomarkers of colorectal cancer: Focus on DNA methylation.,” Cancer Lett., vol. 342,
no. 2, pp. 238–47, Jan. 2014.
[54]
E. A. Williams, “Folate, colorectal cancer and the involvement of DNA methylation.,” Proc. Nutr. Soc., vol. 71,
no. 4, pp. 592–7, Nov. 2012.
[55]
J.-H. Choi, Z. Yates, M. Veysey, Y.-R. Heo, and M. Lucock, “Contemporary issues surrounding folic Acid
fortification initiatives.,” Prev. Nutr. food Sci., vol. 19, no. 4, pp. 247–60, Dec. 2014.
[56]
E. Cho, X. Zhang, M. K. Townsend, J. Selhub, L. Paul, B. Rosner, C. S. Fuchs, W. C. Willett, and E. L.
Giovannucci, “Unmetabolized Folic Acid in Prediagnostic Plasma and the Risk of Colorectal Cancer.,” J. Natl.
Cancer Inst., vol. 107, no. 12, Dec. 2015.
[57]
M. K. Y. Siu, D. S. H. Kong, H. Y. Chan, E. S. Y. Wong, P. P. C. Ip, L. Jiang, H. Y. S. Ngan, X.-F. Le, and A. N.
Y. Cheung, “Paradoxical impact of two folate receptors, FRα and RFC, in ovarian cancer: effect on cell
proliferation, invasion and clinical outcome.,” PLoS One, vol. 7, no. 11, p. e47201, Jan. 2012.
[58]
I. B. Vergote, C. Marth, and R. L. Coleman, “Role of the folate receptor in ovarian cancer treatment: evidence,
mechanism, and clinical implications.,” Cancer Metastasis Rev., vol. 34, no. 1, pp. 41–52, Mar. 2015.
[59]
X. Hong and X. Wang, “Epigenetics and development of food allergy (FA) in early childhood.,” Curr. Allergy
Asthma Rep., vol. 14, no. 9, p. 460, 2014.
[60]
J. E. Sordillo, N. E. Lange, L. Tarantini, V. Bollati, D. Sparrow, P. Vokonas, J. Schwartz, A. Baccarelli, and A.
A. Litonjua, “Allergen Sensitization is Associated with DNA Methylation in Older Men,” Int Arch Allergy
Immunol, vol. 161, no. 1, pp. 37–43, 2014.
[61]
J. A. Dunstan, C. West, S. McCarthy, J. Metcalfe, S. Meldrum, W. H. Oddy, M. K. Tulic, N. D’Vaz, and S. L.
Prescott, “The relationship between maternal folate status in pregnancy, cord blood folate levels, and allergic
outcomes in early childhood.,” Allergy, vol. 67, no. 1, pp. 50–7, Jan. 2012.
[62]
J. W. Hollingsworth, S. Maruoka, K. Boon, S. Garantziotis, Z. Li, J. Tomfohr, N. Bailey, E. N. Potts, G.
Whitehead, D. M. Brass, and D. A. Schwartz, “In utero supplementation with methyl donors enhances
allergic airway disease in mice.,” J. Clin. Invest., vol. 118, no. 10, pp. 3462–9, Oct. 2008.
[63]
T. D. Schaible, R. A. Harris, S. E. Dowd, C. W. Smith, and R. Kellermayer, “Maternal methyl-donor
supplementation induces prolonged murine offspring colitis susceptibility in association with mucosal
epigenetic and microbiomic changes.,” Hum. Mol. Genet., vol. 20, no. 9, pp. 1687–96, May 2011.
[64]
C. S. Yajnik, S. S. Deshpande, A. A. Jackson, H. Refsum, S. Rao, D. J. Fisher, D. S. Bhat, S. S. Naik, K. J. Coyaji,
C. V Joglekar, N. Joshi, H. G. Lubree, V. U. Deshpande, S. S. Rege, and C. H. D. Fall, “Vitamin B12 and folate
concentrations during pregnancy and insulin resistance in the offspring: the Pune Maternal Nutrition
Study.,” Diabetologia, vol. 51, no. 1, pp. 29–38, Jan. 2008.
[65]
C. S. Yajnik and U. S. Deshmukh, “Maternal nutrition, intrauterine programming and consequential risks in
the offspring.,” Rev. Endocr. Metab. Disord., vol. 9, no. 3, pp. 203–11, Sep. 2008.
[66]
C. Kappen, “Modeling anterior development in mice: diet as modulator of risk for neural tube defects.,” Am.
J. Med. Genet. C. Semin. Med. Genet., vol. 163C, no. 4, pp. 333–56, Nov. 2013.
© 2016 Dr. K Fitzgerald, ND
www.drkarafitzgerald.com
133
Methylation Diet and Lifestyle
[67]
B. Kyte, E. Ifebi, S. Shrestha, S. Charles, F. Liu, and J. Zhang, “High red blood cell folate is associated with an
increased risk of death among adults with diabetes, a 15-year follow-up of a national cohort,” Nutr. Metab.
Cardiovasc. Dis., vol. 25, no. 11, pp. 997–1006, 2015.
[68]
K. E. Varley, J. Gertz, K. M. Bowling, S. L. Parker, T. E. Reddy, F. Pauli-Behn, M. K. Cross, B. A. Williams, J.
A. Stamatoyannopoulos, G. E. Crawford, D. M. Absher, B. J. Wold, and R. M. Myers, “Dynamic DNA
methylation across diverse human cell lines and tissues.,” Genome Res., vol. 23, no. 3, pp. 555–67, Mar. 2013.
[69]
R. C. Heine-Bröring, R. M. Winkels, J. M. S. Renkema, L. Kragt, A.-C. B. van Orten-Luiten, E. F. Tigchelaar, D.
S. M. Chan, T. Norat, and E. Kampman, “Dietary supplement use and colorectal cancer risk: a systematic
review and meta-analyses of prospective cohort studies.,” Int. J. Cancer, vol. 136, no. 10, pp. 2388–401, May
2015.
[70]
B. F. Cole, J. A. Baron, R. S. Sandler, R. W. Haile, D. J. Ahnen, R. S. Bresalier, G. McKeown-Eyssen, R. W.
Summers, R. I. Rothstein, C. A. Burke, D. C. Snover, T. R. Church, J. I. Allen, D. J. Robertson, G. J. Beck, J. H.
Bond, T. Byers, J. S. Mandel, L. A. Mott, L. H. Pearson, E. L. Barry, J. R. Rees, N. Marcon, F. Saibil, P. M.
Ueland, and E. R. Greenberg, “Folic acid for the prevention of colorectal adenomas: a randomized clinical
trial.,” JAMA, vol. 297, no. 21, pp. 2351–9, Jul. 2007.
[71]
Y.-I. Kim, “Does a high folate intake increase the risk of breast cancer?,” Nutr. Rev., vol. 64, no. 10 Pt 1, pp.
468–475, 2006.
[72]
P. Chen, C. Li, X. Li, J. Li, R. Chu, and H. Wang, “Higher dietary folate intake reduces the breast cancer risk: a
systematic review and meta-analysis.,” Br. J. Cancer, vol. 110, no. 9, pp. 2327–38, May 2014.
[73]
J. de Batlle, P. Ferrari, V. Chajes, J. Y. Park, N. Slimani, F. McKenzie, K. Overvad, N. Roswall, A. Tjønneland,
M. C. Boutron-Ruault, F. Clavel-Chapelon, G. Fagherazzi, V. Katzke, R. Kaaks, M. M. Bergmann, A.
Trichopoulou, P. Lagiou, D. Trichopoulos, D. Palli, S. Sieri, S. Panico, R. Tumino, P. Vineis, H. B. Bueno-deMesquita, P. H. Peeters, A. Hjartåker, D. Engeset, E. Weiderpass, S. Sánchez, N. Travier, M. J. Sánchez, P.
Amiano, M. D. Chirlaque, A. Barricarte Gurrea, K. T. Khaw, T. J. Key, K. E. Bradbury, U. Ericson, E.
Sonestedt, B. Van Guelpen, J. Schneede, E. Riboli, and I. Romieu, “Dietary folate intake and breast cancer risk:
European prospective investigation into cancer and nutrition.,” J. Natl. Cancer Inst., vol. 107, no. 1, p. 367, Jan.
2015.
[74]
U. Ericson, S. Borgquist, M. I. L. Ivarsson, E. Sonestedt, B. Gullberg, J. Carlson, H. Olsson, K. Jirström, and E.
Wirfält, “Plasma folate concentrations are positively associated with risk of estrogen receptor beta negative
breast cancer in a Swedish nested case control study.,” J. Nutr., vol. 140, no. 9, pp. 1661–1668, 2010.
[75]
U. C. Ericson, M. I. L. Ivarsson, E. Sonestedt, B. Gullberg, J. Carlson, H. Olsson, and E. Wirfält, “Increased
breast cancer risk at high plasma folate concentrations among women with the MTHFR 677T allele.,” Am. J.
Clin. Nutr., vol. 90, no. 5, pp. 1380–9, 2009.
[76]
F.-F. Chiang, S.-C. Huang, H.-M. Wang, F.-P. Chen, and Y.-C. Huang, “High serum folate might have a
potential dual effect on risk of colorectal cancer.,” Clin. Nutr., vol. 34, no. 5, pp. 986–90, Nov. 2015.
[77]
A. Carrer and K. E. Wellen, “Metabolism and epigenetics: a link cancer cells exploit.,” Curr. Opin. Biotechnol.,
vol. 34, pp. 23–29, 2014.
[78]
Y. Luo, Y. Wang, Y. Shu, Q. Lu, and R. Xiao, “Epigenetic mechanisms: An emerging role in pathogenesis and
its therapeutic potential in systemic sclerosis.,” Int. J. Biochem. Cell Biol., Jun. 2015.
[79]
T. Fukuhara, T. Tomiyama, K. Yasuda, Y. Ueda, Y. Ozaki, Y. Son, S. Nomura, K. Uchida, K. Okazaki, and T.
Kinashi, “Hypermethylation of MST1 in IgG4-related autoimmune pancreatitis and rheumatoid arthritis.,”
Biochem. Biophys. Res. Commun., vol. 463, no. 4, pp. 968–74, Aug. 2015.
[80]
K. Nakano, J. W. Whitaker, D. L. Boyle, W. Wang, and G. S. Firestein, “DNA methylome signature in
rheumatoid arthritis.,” Ann. Rheum. Dis., vol. 72, no. 1, pp. 110–7, Jan. 2013.
© 2016 Dr. K Fitzgerald, ND
www.drkarafitzgerald.com
134
Methylation Diet and Lifestyle
[81]
D. Martino, T. Dang, A. Sexton-Oates, S. Prescott, M. L. K. Tang, S. Dharmage, L. Gurrin, J. Koplin, A.-L.
Ponsonby, K. J. Allen, and R. Saffery, “Blood DNA methylation biomarkers predict clinical reactivity in foodsensitized infants.,” J. Allergy Clin. Immunol., vol. 135, no. 5, pp. 1319–1328.e12, 2015.
[82]
S. Jin, Y. K. Lee, Y. C. Lim, Z. Zheng, X. M. Lin, D. P. Y. Ng, J. D. Holbrook, H. Y. Law, K. Y. C. Kwek, G. S. H.
Yeo, and C. Ding, “Global DNA hypermethylation in down syndrome placenta.,” PLoS Genet., vol. 9, no. 6, p.
e1003515, Jun. 2013.
[83]
S. Jin, Y. K. Lee, Y. C. Lim, Z. Zheng, X. M. Lin, D. P. Y. Ng, J. D. Holbrook, H. Y. Law, K. Y. C. Kwek, G. S. H.
Yeo, and C. Ding, “Global DNA Hypermethylation in Down Syndrome Placenta,” PLoS Genet., vol. 9, no. 6,
2013.
[84]
K. S. Crider, T. P. Yang, R. J. Berry, and L. B. Bailey, “Folate and DNA methylation: a review of molecular
mechanisms and the evidence for folate’s role.,” Adv. Nutr., vol. 3, no. 1, pp. 21–38, Jan. 2012.
[85]
D. Pu, Y. Shen, and J. Wu, “Association between MTHFR gene polymorphisms and the risk of autism
spectrum disorders: a meta-analysis.,” Autism Res., vol. 6, no. 5, pp. 384–92, Oct. 2013.
[86]
R. J. Schmidt, R. L. Hansen, J. Hartiala, H. Allayee, L. C. Schmidt, D. J. Tancredi, F. Tassone, and I. HertzPicciotto, “Prenatal vitamins, one-carbon metabolism gene variants, and risk for autism.,” Epidemiology, vol.
22, no. 4, pp. 476–85, 2011.
[87]
J. Chen, B. K. Lipska, N. Halim, Q. D. Ma, M. Matsumoto, S. Melhem, B. S. Kolachana, T. M. Hyde, M. M.
Herman, J. Apud, M. F. Egan, J. E. Kleinman, and D. R. Weinberger, “Functional analysis of genetic variation
in catechol-O-methyltransferase (COMT): effects on mRNA, protein, and enzyme activity in postmortem
human brain.,” Am. J. Hum. Genet., vol. 75, no. 5, pp. 807–21, Nov. 2004.
[88]
M. Bortolato and J. C. Shih, “Behavioral outcomes of monoamine oxidase deficiency: preclinical and clinical
evidence.,” Int. Rev. Neurobiol., vol. 100, pp. 13–42, Jan. 2011.
[89]
M. Varela-Rey, A. Woodhoo, M.-L. Martinez-Chantar, J. M. Mato, and S. C. Lu, “Alcohol, DNA methylation,
and cancer.,” Alcohol Res., vol. 35, no. 1, pp. 25–35, Jan. 2013.
[90]
T. Huang, M. L. Wahlqvist, and D. Li, “Effect of n-3 polyunsaturated fatty acid on gene expression of the
critical enzymes involved in homocysteine metabolism.,” Nutr. J., vol. 11, p. 6, Jan. 2012.
[91]
Centers for Disease Control and Prevention, “Key Findings: Blood Folate Concentrations and Risk of Neural
Tube Defect-Affected Pregnancies: Where Does the United States Stand?,” National Center on Birth Defects and
Developmental Disabilities, 2015. [Online]. Available:
http://www.cdc.gov/ncbddd/birthdefectscount/features/blood-folate-ntd-risk-us.html. [Accessed: 22-Dec2015].
[92]
M. A. Valenzuela, “Helicobacter pylori -induced inflammation and epigenetic changes during gastric
carcinogenesis,” World J. Gastroenterol., vol. 21, no. 45, p. 12742, 2015.
[93]
J. V. Fernandes, R. N. O. Cobucci, C. A. N. Jatobá, T. A. A. de Medeiros Fernandes, J. W. V. de Azevedo, and
J. M. G. de Araújo, “The Role of the Mediators of Inflammation in Cancer Development,” Pathol. Oncol. Res.,
pp. 527–534, 2015.
[94]
D. R. Hodge, B. Peng, J. C. Cherry, E. M. Hurt, S. D. Fox, J. A. Kelley, D. J. Munroe, and W. L. Farrar,
“Interleukin 6 supports the maintenance of p53 tumor suppressor gene promoter methylation.,” Cancer Res.,
vol. 65, no. 11, pp. 4673–82, Jun. 2005.
[95]
A. P. Sharples, I. Polydorou, D. C. Hughes, D. J. Owens, T. M. Hughes, and C. E. Stewart, “Skeletal muscle
cells possess a ‘memory’ of acute early life TNF-α exposure: role of epigenetic adaptation,” Biogerontology,
2015.
[96]
H. Kasai, K. Kawai, and Y. Li, “Free radical-mediated cytosine C-5 methylation triggers epigenetic changes
© 2016 Dr. K Fitzgerald, ND
www.drkarafitzgerald.com
135
Methylation Diet and Lifestyle
during carcinogenesis,” BioMol Concepts, vol. 4, no. 3, pp. 213–220, 2013.
[97]
N. Esser, S. Legrand-Poels, J. Piette, A. J. Scheen, and N. Paquot, “Inflammation as a link between obesity,
metabolic syndrome and type 2 diabetes.,” Diabetes Res. Clin. Pract., vol. 105, no. 2, pp. 141–50, Aug. 2014.
[98]
G. Verdile, K. N. Keane, V. F. Cruzat, S. Medic, M. Sabale, J. Rowles, N. Wijesekara, R. N. Martins, P. E.
Fraser, and P. Newsholme, “Inflammation and Oxidative Stress: The Molecular Connectivity between Insulin
Resistance, Obesity, and Alzheimer’s Disease.,” Mediators Inflamm., vol. 2015, p. 105828, Jan. 2015.
[99]
E. Nilsson, P. A. Jansson, A. Perfilyev, P. Volkov, M. Pedersen, M. K. Svensson, P. Poulsen, R. Ribel-Madsen,
N. L. Pedersen, P. Almgren, J. Fadista, T. Rönn, B. Klarlund Pedersen, C. Scheele, A. Vaag, and C. Ling,
“Altered DNA methylation and differential expression of genes influencing metabolism and inflammation in
adipose tissue from subjects with type 2 diabetes.,” Diabetes, vol. 63, no. 9, pp. 2962–76, Sep. 2014.
[100]
S. Li, L. A. Papale, Q. Zhang, A. Madrid, L. Chen, P. Chopra, S. Keleş, P. Jin, and R. S. Alisch, “Genome-wide
alterations in hippocampal 5-hydroxymethylcytosine links plasticity genes to acute stress,” Neurobiol. Dis.,
2015.
[101]
K. Szarc vel Szic, K. Declerck, M. Vidaković, and W. Vanden Berghe, “From inflammaging to healthy aging
by dietary lifestyle choices: is epigenetics the key to personalized nutrition?,” Clin. Epigenetics, vol. 7, no. 1, p.
33, 2015.
[102]
A. J. A. Wright, M. J. King, C. A. Wolfe, H. J. Powers, and P. M. Finglas, “Comparison of (6 S)-5methyltetrahydrofolic acid v. folic acid as the reference folate in longer-term human dietary intervention
studies assessing the relative bioavailability of natural food folates: comparative changes in folate status
following a 16-we,” Br. J. Nutr., vol. 103, no. 5, pp. 724–9, Mar. 2010.
[103]
USDA, “USDA National Nutrient Database for Standard Reference, Release 27 (revised).” US Department of
Agriculture, Agricultural Research Service, Nutrient Data Laboratory., 2015.
[104]
K. Y. Patterson, S. A. Bhagwat, J. R. Williams, J. C. Howe, and J. M. Holden, “USDA Database for the Choline
Content of Common Foods; Release Two,” Beltsville, MD, 2008.
[105]
S. Sharma and A. Litonjua, “Asthma, allergy, and responses to methyl donor supplements and nutrients.,” J.
Allergy Clin. Immunol., vol. 133, no. 5, pp. 1246–54, 2014.
[106]
M. Masters and R. A. McCance, “The sulphur content of foods.,” Biochem. J., vol. 33, no. 8, pp. 1304–12, Aug.
1939.
[107]
D.-H. Yu, M. Gadkari, Q. Zhou, S. Yu, N. Gao, Y. Guan, D. Schady, T. N. Roshan, M.-H. Chen, E. Laritsky, Z.
Ge, H. Wang, R. Chen, C. Westwater, L. Bry, R. A. Waterland, C. Moriarty, C. Hwang, A. G. Swennes, S. R.
Moore, and L. Shen, “Postnatal epigenetic regulation of intestinal stem cells requires DNA methylation and is
guided by the microbiome,” Genome Biol., vol. 16, no. 1, p. 211, 2015.
[108]
B. Paul, S. Barnes, W. Demark-wahnefried, C. Morrow, and C. Salvador, “Influences of diet and the gut
microbiome on epigenetic modulation in cancer and other diseases,” Clin. Epigenetics, pp. 1–11, 2015.
[109]
M. Nieuwdorp, P. W. Gilijamse, N. Pai, and L. M. Kaplan, “Role of the Microbiome in Energy Regulation and
Metabolism,” Gastroenterology, vol. 146, no. 6, pp. 1525–1533, 2014.
[110]
H. Kumar, R. Lund, A. Laiho, K. Lundelin, R. E. Ley, E. Isolauri, and S. Salminen, “Gut microbiota as an
epigenetic regulator: pilot study based on whole-genome methylation analysis.,” MBio, vol. 5, no. 6, Jan. 2014.
[111]
M. Million, J.-C. Lagier, D. Yahav, and M. Paul, “Gut bacterial microbiota and obesity.,” Clin. Microbiol.
Infect., vol. 19, no. 4, pp. 305–13, Apr. 2013.
[112]
M. Rossi, A. Amaretti, and S. Raimondi, “Folate Production by Probiotic Bacteria,” Nutrients, vol. 3, no. 12,
pp. 118–134, 2011.
© 2016 Dr. K Fitzgerald, ND
www.drkarafitzgerald.com
136
Methylation Diet and Lifestyle
[113]
M. R. D’Aimmo, M. Modesto, P. Mattarelli, B. Biavati, and T. Andlid, “Biosynthesis and cellular content of
folate in bifidobacteria across host species with different diets.,” Anaerobe, vol. 30, pp. 169–77, Dec. 2014.
[114]
A. Pompei, L. Cordisco, A. Amaretti, S. Zanoni, S. Raimondi, D. Matteuzzi, and M. Rossi, “Administration of
folate-producing bifidobacteria enhances folate status in Wistar rats.,” J. Nutr., vol. 137, no. 12, pp. 2742–2746,
2007.
[115]
N. Milman, “Intestinal absorption of folic acid - new physiologic & molecular aspects.,” Indian J. Med. Res.,
vol. 136, no. 5, pp. 725–8, Nov. 2012.
[116]
K. Taki, F. Takayama, and T. Niwa, “Beneficial effects of Bifidobacteria in a gastroresistant seamless capsule
on hyperhomocysteinemia in hemodialysis patients.,” J. Ren. Nutr., vol. 15, no. 1, pp. 77–80, Jan. 2005.
[117]
E. M. M. Quigley and A. Abu-Shanab, “Small intestinal bacterial overgrowth.,” Infect. Dis. Clin. North Am.,
vol. 24, no. 4, pp. 943–59, viii–ix, Dec. 2010.
[118]
G. Escobedo, E. López-Ortiz, and I. Torres-Castro, “Gut microbiota as a key player in triggering obesity,
systemic inflammation and insulin resistance.,” Rev. Investig. clínica; organo del Hosp. Enfermedades la Nutr.,
vol. 66, no. 5, pp. 450–9, Jan. .
[119]
L. Monnier, E. Mas, C. Ginet, F. Michel, L. Villon, J.-P. Cristol, and C. Colette, “Activation of oxidative stress
by acute glucose fluctuations compared with sustained chronic hyperglycemia in patients with type 2
diabetes.,” JAMA, vol. 295, no. 14, pp. 1681–7, Apr. 2006.
[120]
I. T. Johnson and N. J. Belshaw, “Environment, diet and CpG island methylation: Epigenetic signals in
gastrointestinal neoplasia,” Food Chem. Toxicol., vol. 46, no. 4, pp. 1346–1359, 2008.
[121]
D. B. Panagiotakos, C. Pitsavos, M. Yannakoulia, C. Chrysohoou, and C. Stefanadis, “The implication of
obesity and central fat on markers of chronic inflammation: The ATTICA study.,” Atherosclerosis, vol. 183, no.
2, pp. 308–15, Dec. 2005.
[122]
Y. Belkaid, “Role of the Microbiota in Immunity and inflammation,” vol. 18, no. 9, pp. 1199–1216, 2013.
[123]
C. A. Calandrini, A. C. Ribeiro, A. C. Gonnelli, C. Ota-Tsuzuki, L. P. Rangel, E. Saba-Chujfi, and M. P. A.
Mayer, “Microbial composition of atherosclerotic plaques.,” Oral Dis., vol. 20, no. 3, pp. e128–34, Apr. 2014.
[124]
K. Matsusaka, S. Funata, M. Fukayama, and A. Kaneda, “DNA methylation in gastric cancer, related to
Helicobacter pylori and Epstein-Barr virus.,” World J. Gastroenterol., vol. 20, no. 14, pp. 3916–26, Apr. 2014.
[125]
R. Valenta, H. Hochwallner, B. Linhart, and S. Pahr, “Food allergies—the Basics,” Gastroenterology, vol. 148,
no. 6, pp. 1120–31.e4, Feb. 2015.
[126]
P. H. Black, “Stress and the inflammatory response: a review of neurogenic inflammation.,” Brain. Behav.
Immun., vol. 16, no. 6, pp. 622–53, Dec. 2002.
[127]
L. Pruimboom, C. L. Raison, and F. A. J. Muskiet, “Physical Activity Protects the Human Brain against
Metabolic Stress Induced by a Postprandial and Chronic Inflammation.,” Behav. Neurol., vol. 2015, p. 569869,
Jan. 2015.
[128]
A. Baccarelli and V. Bollati, “Epigenetics and environmental chemicals.,” Curr. Opin. Pediatr., vol. 21, no. 2,
pp. 243–51, Apr. 2009.
[129]
A. Baccarelli, R. O. Wright, V. Bollati, L. Tarantini, A. A. Litonjua, H. H. Suh, A. Zanobetti, D. Sparrow, P. S.
Vokonas, and J. Schwartz, “Rapid DNA methylation changes after exposure to traffic particles.,” Am. J.
Respir. Crit. Care Med., vol. 179, no. 7, pp. 572–8, Apr. 2009.
[130]
M. Manikkam, C. Guerrero-Bosagna, R. Tracey, M. M. Haque, and M. K. Skinner, “Transgenerational actions
of environmental compounds on reproductive disease and identification of epigenetic biomarkers of
ancestral exposures.,” PLoS One, vol. 7, no. 2, p. e31901, Jan. 2012.
© 2016 Dr. K Fitzgerald, ND
www.drkarafitzgerald.com
137
Methylation Diet and Lifestyle
[131]
J. A. Rusiecki, A. Baccarelli, V. Bollati, L. Tarantini, L. E. Moore, and E. C. Bonefeld-Jorgensen, “Global DNA
hypomethylation is associated with high serum-persistent organic pollutants in Greenlandic Inuit.,” Environ.
Health Perspect., vol. 116, no. 11, pp. 1547–52, Nov. 2008.
[132]
W. F. O. Marasas, R. T. Riley, K. A. Hendricks, V. L. Stevens, T. W. Sadler, J. Gelineau-van Waes, S. A.
Missmer, J. Cabrera, O. Torres, W. C. A. Gelderblom, J. Allegood, C. Martínez, J. Maddox, J. D. Miller, L.
Starr, M. C. Sullards, A. V. Roman, K. A. Voss, E. Wang, and A. H. Merrill, “Fumonisins disrupt sphingolipid
metabolism, folate transport, and neural tube development in embryo culture and in vivo: a potential risk
factor for human neural tube defects among populations consuming fumonisin-contaminated maize.,” J.
Nutr., vol. 134, no. 4, pp. 711–6, Apr. 2004.
[133]
A. Cardenas, D. C. Koestler, E. A. Houseman, B. P. Jackson, M. L. Kile, M. R. Karagas, and C. J. Marsit,
“Differential DNA methylation in umbilical cord blood of infants exposed to mercury and arsenic in utero.,”
Epigenetics, vol. 10, no. 6, pp. 508–15, Jan. 2015.
[134]
V. Nicolia, M. Lucarelli, and A. Fuso, “Environment, epigenetics and neurodegeneration: Focus on nutrition
in Alzheimer’s disease,” Exp. Gerontol., vol. 68, pp. 8–12, 2015.
[135]
H. Palma-Gudiel, A. Córdova-Palomera, J. C. Leza, and L. Fañanás, “Glucocorticoid receptor gene (NR3C1)
methylation processes as mediators of early adversity in stress-related disorders causality: A critical review.,”
Neurosci. Biobehav. Rev., vol. 55, pp. 520–535, Jun. 2015.
[136]
E. Vangeel, F. Van Den Eede, T. Hompes, B. Izzi, J. Del Favero, G. Moorkens, D. Lambrechts, K. Freson, and
S. Claes, “Chronic Fatigue Syndrome and DNA Hypomethylation of the Glucocorticoid Receptor Gene
Promoter 1F Region: Associations With HPA Axis Hypofunction and Childhood Trauma.,” Psychosom. Med.,
vol. 77, no. 8, pp. 853–62, Oct. 2015.
[137]
H. Harb and H. Renz, “Update on epigenetics in allergic disease,” J. Allergy Clin. Immunol., vol. 135, no. 1, pp.
15–24, 2015.
[138]
L. Cao-Lei, K. N. Dancause, G. Elgbeili, R. Massart, M. Szyf, A. Liu, D. P. Laplante, and S. King, “DNA
methylation mediates the impact of exposure to prenatal maternal stress on BMI and central adiposity in
children at age 13½ years: Project Ice Storm.,” Epigenetics, Jun. 2015.
[139]
A. R. Tyrka, L. H. Price, C. Marsit, O. C. Walters, and L. L. Carpenter, “Childhood adversity and epigenetic
modulation of the leukocyte glucocorticoid receptor: preliminary findings in healthy adults.,” PLoS One, vol.
7, no. 1, p. e30148, 2012.
[140]
S. Horsburgh, P. Robson-Ansley, R. Adams, and C. Smith, “Exercise and inflammation-related epigenetic
modifications: focus on DNA methylation.,” Exerc. Immunol. Rev., vol. 21, pp. 26–41, Jan. 2015.
[141]
A. de S. e Silva and M. P. G. da Mota, “Effects of physical activity and training programs on plasma
homocysteine levels: a systematic review.,” Amino Acids, vol. 46, no. 8, pp. 1795–804, Aug. 2014.
[142]
J. C. Neuman, K. A. Albright, and K. L. Schalinske, “Exercise prevents hyperhomocysteinemia in a dietary
folate-restricted mouse model.,” Nutr. Res., vol. 33, no. 6, pp. 487–93, Jun. 2013.
[143]
D. König, E. Bissé, P. Deibert, H.-M. Müller, H. Wieland, and A. Berg, “Influence of training volume and
acute physical exercise on the homocysteine levels in endurance-trained men: interactions with plasma folate
and vitamin B12.,” Ann. Nutr. Metab., vol. 47, no. 3–4, pp. 114–8, Jan. 2003.
[144]
M. Herrmann, H. Schorr, R. Obeid, J. Scharhag, A. Urhausen, W. Kindermann, and W. Herrmann,
“Homocysteine increases during endurance exercise.,” Clin. Chem. Lab. Med., vol. 41, no. 11, pp. 1518–24,
Nov. 2003.
[145]
B. K. Pedersen and B. Saltin, “Exercise as medicine - evidence for prescribing exercise as therapy in 26
different chronic diseases.,” Scand. J. Med. Sci. Sports, vol. 25 Suppl 3, pp. 1–72, Dec. 2015.
© 2016 Dr. K Fitzgerald, ND
www.drkarafitzgerald.com
138
Methylation Diet and Lifestyle
[146]
Q.-H. Gong, J.-F. Kang, Y.-Y. Ying, H. Li, X.-H. Zhang, Y.-H. Wu, and G.-Z. Xu, “Lifestyle Interventions for
Adults with Impaired Glucose Tolerance: A Systematic Review and Meta-Analysis of the Effects on Glycemic
Control,” Intern. Med., vol. 54, no. 3, pp. 303–310, 2015.
[147]
A. Yavari, M. Javadi, P. Mirmiran, and Z. Bahadoran, “Exercise-induced oxidative stress and dietary
antioxidants.,” Asian J. Sports Med., vol. 6, no. 1, p. e24898, Mar. 2015.
[148]
D. Camiletti-Moirón, V. A. Aparicio, P. Aranda, and Z. Radak, “Does exercise reduce brain oxidative stress?
A systematic review.,” Scand. J. Med. Sci. Sports, vol. 23, no. 4, pp. e202–12, Aug. 2013.
[149]
A. Pingitore, G. P. P. Lima, F. Mastorci, A. Quinones, G. Iervasi, and C. Vassalle, “Exercise and oxidative
stress: Potential effects of antioxidant dietary strategies in sports,” Nutrition, vol. 31, no. 7–8, pp. 916–922,
2015.
[150]
S. Voisin, N. Eynon, X. Yan, and D. J. Bishop, “Exercise training and DNA methylation in humans,” Acta
Physiol., vol. 213, no. 1, pp. 39–59, 2015.
[151]
A. J. White, D. P. Sandler, S. C. E. Bolick, Z. Xu, J. A. Taylor, and L. A. DeRoo, “Recreational and household
physical activity at different time points and DNA global methylation.,” Eur. J. Cancer, vol. 49, no. 9, pp.
2199–206, Jun. 2013.
[152]
H. Ren, V. Collins, S. J. Clarke, J.-S. Han, P. Lam, F. Clay, L. M. Williamson, and K. H. Andy Choo,
“Epigenetic changes in response to tai chi practice: a pilot investigation of DNA methylation marks.,” Evid.
Based. Complement. Alternat. Med., vol. 2012, p. 841810, 2012.
[153]
J. Uribarri, S. Woodruff, S. Goodman, W. Cai, X. Chen, R. Pyzik, A. Yong, G. E. Striker, and H. Vlassara,
“Advanced glycation end products in foods and a practical guide to their reduction in the diet.,” J. Am. Diet.
Assoc., vol. 110, no. 6, pp. 911–16.e12, Jun. 2010.
[154]
A. R. Hillman, R. V Vince, L. Taylor, L. McNaughton, N. Mitchell, and J. Siegler, “Exercise-induced
dehydration with and without environmental heat stress results in increased oxidative stress.,” Appl. Physiol.
Nutr. Metab., vol. 36, no. 5, pp. 698–706, Oct. 2011.
[155]
Y. Li and T. O. Tollefsbol, “Impact on DNA methylation in cancer prevention and therapy by bioactive
dietary components.,” Curr. Med. Chem., vol. 17, no. 20, pp. 2141–51, Jan. 2010.
[156]
S. I. Khan, P. Aumsuwan, I. a. Khan, L. a. Walker, and A. K. Dasmahapatra, “Epigenetic events associated
with breast cancer and their prevention by dietary components targeting the epigenome,” Chem. Res. Toxicol.,
vol. 25, no. 1, pp. 61–73, 2012.
[157]
S. Ribarič, “Diet and aging.,” Oxid. Med. Cell. Longev., vol. 2012, p. 741468, Jan. 2012.
[158]
W. G. Kaelin and S. L. McKnight, “Influence of metabolism on epigenetics and disease.,” Cell, vol. 153, no. 1,
pp. 56–69, Mar. 2013.
[159]
S. Lim, A. S. Chesser, J. C. Grima, P. M. Rappold, D. Blum, S. Przedborski, and K. Tieu, “D-βhydroxybutyrate is protective in mouse models of Huntington’s disease.,” PLoS One, vol. 6, no. 9, p. e24620,
Jan. 2011.
[160]
Y.-H. Youm, K. Y. Nguyen, R. W. Grant, E. L. Goldberg, M. Bodogai, D. Kim, D. D’Agostino, N. Planavsky,
C. Lupfer, T. D. Kanneganti, S. Kang, T. L. Horvath, T. M. Fahmy, P. A. Crawford, A. Biragyn, E. Alnemri,
and V. D. Dixit, “The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated
inflammatory disease.,” Nat. Med., vol. 21, no. 3, pp. 263–9, Feb. 2015.
[161]
R. G. Levy, P. N. Cooper, and P. Giri, “Ketogenic diet and other dietary treatments for epilepsy.,” Cochrane
database Syst. Rev., vol. 3, p. CD001903, Jan. 2012.
[162]
A. F. Branco, A. Ferreira, R. F. Simões, S. Magalhães-Novais, C. Zehowski, E. Cope, A. M. Silva, D. Pereira, V.
A. Sardão, and T. Cunha-Oliveira, “Ketogenic diets: from cancer to mitochondrial diseases and beyond.,”
© 2016 Dr. K Fitzgerald, ND
www.drkarafitzgerald.com
139
Methylation Diet and Lifestyle
Eur. J. Clin. Invest., vol. 46, no. 3, pp. 285–98, Mar. 2016.
[163]
P. Pissios, S. Hong, A. R. Kennedy, D. Prasad, F.-F. Liu, and E. Maratos-Flier, “Methionine and choline
regulate the metabolic phenotype of a ketogenic diet.,” Mol. Metab., vol. 2, no. 3, pp. 306–13, Jan. 2013.
[164]
A. A. Llanos, R. G. Dumitrescu, T. M. Brasky, Z. Liu, J. B. Mason, C. Marian, K. H. Makambi, S. L. Spear, B. V.
S. Kallakury, J. L. Freudenheim, and P. G. Shields, “Relationships among folate, alcohol consumption, gene
variants in one-carbon metabolism and p16INK4a methylation and expression in healthy breast tissues.,”
Carcinogenesis, vol. 36, no. 1, pp. 60–7, Jan. 2015.
[165]
S. H. Kenyon, A. Nicolaou, and W. A. Gibbons, “The effect of ethanol and its metabolites upon methionine
synthase activity in vitro.,” Alcohol, vol. 15, no. 4, pp. 305–9, May 1998.
[166]
Y. Li, M. Daniel, and T. O. Tollefsbol, “Epigenetic regulation of caloric restriction in aging.,” BMC Med., vol.
9, no. 1, p. 98, Jan. 2011.
[167]
R. E. Hodges and D. M. Minich, “Modulation of Metabolic Detoxification Pathways Using Foods and FoodDerived Components: A Scientific Review with Clinical Application.,” J. Nutr. Metab., vol. 2015, p. 760689,
Jan. 2015.
© 2016 Dr. K Fitzgerald, ND
www.drkarafitzgerald.com
140