ArticlePDF AvailableLiterature Review

Early AGEing and metabolic diseases: is perinatal exposure to glycotoxins programming for adult-life metabolic syndrome?

Authors:

Abstract and Figures

Perinatal early nutritional disorders are critical for the developmental origins of health and disease. Glycotoxins, or advanced glycation end-products, and their precursors such as the methylglyoxal, which are formed endogenously and commonly found in processed foods and infant formulas, may be associated with acute and long-term metabolic disorders. Besides general aspects of glycotoxins, such as their endogenous production, exogenous sources, and their role in the development of metabolic syndrome, we discuss in this review the sources of perinatal exposure to glycotoxins and their involvement in metabolic programming mechanisms. The role of perinatal glycotoxin exposure in the onset of insulin resistance, central nervous system development, cardiovascular diseases, and early aging also are discussed, as are possible interventions that may prevent or reduce such effects.
Content may be subject to copyright.
Lead Article
Early AGEing and metabolic diseases: is perinatal exposure to
glycotoxins programming for adult-life metabolic syndrome?
Fl
avio A. Francisco*, Lucas P.J. Saavedra*, Marcos D.F. Junior ,C
atia Barra, Paulo Matafome,
Paulo C.F. Mathias, and Rodrigo M. Gomes
Perinatal early nutritional disorders are critical for the developmental origins of
health and disease. Glycotoxins, or advanced glycation end-products, and their pre-
cursors such as the methylglyoxal, which are formed endogenously and commonly
found in processed foods and infant formulas, may be associated with acute and
long-term metabolic disorders. Besides general aspects of glycotoxins, such as their
endogenous production, exogenous sources, and their role in the development of
metabolic syndrome, we discuss in this review the sources of perinatal exposure to
glycotoxins and their involvement in metabolic programming mechanisms. The
role of perinatal glycotoxin exposure in the onset of insulin resistance, central ner-
vous system development, cardiovascular diseases, and early aging also are dis-
cussed, as are possible interventions that may prevent or reduce such effects.
INTRODUCTION
The developmental origins of health and disease con-
cept focuses on the potential associations between a
suboptimal fetal and/or postnatal environment and
several pathologies in the offspring, such as the meta-
bolic syndrome. Several animal models have been de-
veloped to explore the pathophysiology and
mechanisms of developmental programming of the
metabolic syndrome. Features of cardiometabolic dis-
eases have been found in the offspring of diabetic
rodents, as well as in the offspring of rodents fed a
high-fat diet or fructose-enriched diet.
15
High sugar
intake is associated with harmful effects, such as car-
diovascular diseases, obesity, insulin resistance, and
diabetes. In this way, hyperglycemia is related to in-
creased levels of advanced glycation end-products
(AGEs), and these glycotoxins are closely associated
with the development and progression of diabetes and
its complications.
611
AGEs also are involved in the
deterioration of metabolic homeostasis in obesity,
namely the development of insulin resistance–associ-
ated pathologies such as cardio- and cerebrovascular
diseases, nonalcoholic steatohepatitis, and central ner-
vous system disorders, including dementia, in adult
and pediatric patients.
1226
Vascular aging due to
AGEs exposure, or vascular AGEing, is related to oxi-
dative stress due to increased generation of reactive
species of oxygen and nitrogen,
2730
endothelial dys-
function,
3133
and changes in the extracellular matrix
32
and in inflammatory factors.
34
Infant formulas are
used worldwide as a substitute for breast milk; previ-
ous studies have reported high AGE content in breast
milk.
3538
Thus, infants’ exposure to these nutritional
Affiliation: F.A. Francisco, L.P.J. Saavedra, and P.C.F. Mathias are with the Department of Biotechnology, Genetics, and Cellular Biology,
State University of Maringa, Maringa, PR, Brazil. M.D.F. Junior and R.M. Gomes are with the Department of Physiological Sciences, Federal
University of Goi
as, Goi^
ania, GO, Brazil. C. Barra and P. Matafome are with the Institute of Physiology and Coimbra Institute of Clinical and
Biomedical Research, Faculty of Medicine, and the Center for Innovative Biotechnology and Biomedicine, University of Coimbra; and the
Clinical Academic Center of Coimbra, Coimbra, Portugal.
*These authors contributed equally.
Correspondence: R.M. Gomes, Department of Physiological Sciences, Biological Sciences Institute 2, room 101, Federal University of Goi
as,
Esperanc¸a Ave s/n, 74690-900 Goi^
ania, GO, Brazil. Email: Gomesrm@ufg.br.
Key words: advanced glycation end products (AGEs), glycotoxins, metabolic programming, metabolic syndrome, methylglyoxal.
V
CThe Author(s) 2020. Published by Oxford University Press on behalf of the International Life Sciences Institute.
All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.
doi: 10.1093/nutrit/nuaa074
Nutrition Reviews
V
R
Vol. 0(0):1–12 1
Downloaded from https://academic.oup.com/nutritionreviews/advance-article/doi/10.1093/nutrit/nuaa074/5909185 by Universidade Federal de Goi�s user on 21 September 2020
contaminants early in life may contribute to the develop-
ment of cardiometabolic disorders at adulthood.
37,39,40
In this review, we provide an overview of the cur-
rent knowledge about the contribution of perinatal
glycotoxin exposure to metabolic programming and
the development of metabolic syndrome–related pa-
thologies. Evidence of increased glycotoxin exposure
of the fetus or newborn, due to maternal or infant di-
etary AGE consumption, in addition to infant for-
mula feeding, on developmental programming of
metabolic syndrome are discussed, as are interven-
tions to prevent the consequences of perinatal expo-
sure to AGEs. Figure 1 provides an overview of main
sources and potential mechanisms of the develop-
ment of cardiometabolic disease development during
adult life, due to the exposure to glycotoxins during
perinatal life.
CLINICAL EVIDENCE OF PERINATAL PROGRAMMING
FOR ADULT-LIFE METABOLIC SYNDROME
Pregnancy is a critical period for the health of both the
fetus and the mother and is very sensitive to environ-
mental disturbances. Several studies established a rela-
tionship between disturbances in the pregnancy and
diseases in offspring throughout life.
4150
The magni-
tude of such effects depends on the stage of gestation in
which the fetus was exposed and the nature of the ag-
gressive agent.
44
It is well established that tobacco, alco-
hol, distress, nutritional unbalances, and other
metabolic disruptors affect the proper development of
the fetus during intrauterine life.
46,48,5052
One of the
most common gestational disorders is gestational diabe-
tes mellitus (GDM), which is associated with pregesta-
tional overweight and has been implicated in adverse
perinatal outcomes such as increased weight gain dur-
ing the gestational period and high sugar consump-
tion.
45,53,54
Fetal development is very susceptible to
diabetes, given that this condition can promote severe
changes in tissues and organs, with cardiovascular and
neural tube defects being the most frequent malforma-
tions.
43,46
Mothers with pregestational diabetes mellitus
(PGDM) and a poorly controlled hyperglycemia during
the first trimester have a 5% to 10% higher risk of hav-
ing newborns with a major birth defect and a 15% to
20% higher risk of spontaneous abortion.
55
On the
other hand, GDM is associated more with pregnancy
complications, such as macrosomia, and pre- and peri-
natal mortality, than with congenital anomalies.
46
The
offspring of mothers with PGDM have increased adi-
posity and overweight resulting from transplacental
passage of maternal glucose and induction of fetal
hyperinsulinemia.
46
Pregnant women with GDM have
an increased risk of delivering large-for-gestational-age
(LGA) newborns, who have an higher risk of being
obese at childhood.
43,56
Diet composition before and during pregnancy
may influence the metabolic profile of both the
mother and the newborn, and may affect the new-
born’s size at birth.
57,58
Nutritional changes may lead
to impairment of fetal growth and intrauterine
growth restriction, as well as fetal adiposity, insulin
resistance, and pancreatic b-cell dysfunction.
59
In a
case-control study, Amezcua-Prieto et al
58
suggest the
increased consumption during pregnancy of industrial
bakery products, pastries, and products containing re-
fined sugar is associated with a higher risk of having
a small-for-gestational-age (SGA) newborn. In con-
trast, higher consumption of whole-grain cereal and
bread is related to a lower risk of delivering an SGA
infant.
58
According to another cohort study, the daily
consumption of artificially sweetened beverages dur-
ing pregnancy induces a 2-fold higher risk of having
a child with overweight at the child’s first year.
60
Ornoy et al
46
showed that the offspring of mothers
with GDM have a high frequency of overweight, as
do babies who are breastfed by mothers with diabe-
tes. Palatianou et al
61
found an increased association
of the LGA condition with nondiabetic obesity com-
pared with type 2 diabetes. On the other hand, LGA
infants from mothers with diabetes (either GDM or
PGDM) are above the 90th percentile in height and
weight and have increased weight gain in the first 4
months of life.
46,62
A meta-analysis performed by
Schellong et al
63
revealed a predisposition to adult-
hood overweight in LGA newborns but not in SGA
newborns. However, both the LGA and SGA condi-
tions have a similar risk for development of adult-
hood diabetes, with the risk that following a U-
shaped and not a linear relationship.
64
Children who
are SGA born to mothers with PGDM and associated
nephropathy are more susceptible to prematurity, re-
duced growth at age 3 years and body weight and
height below the 50th percentile when compared with
children of mothers with PGDM without complica-
tions. As well, SGA individuals who gained a sub-
stantial amount of weight in early childhood
exhibited higher risk of developing hypertension and
diabetes and also higher coronary heart disease mor-
tality in adulthood compared with their age-matched
counterparts.
59
Thus, maternal obesity and type 2 diabetes affect
birth weight, and both the SGA and LGA conditions
are associated with increased risk of metabolic impair-
ment and related complications in the adult life.
Moreover, the presence of diabetic complications in the
2Nutrition Reviews
V
R
Vol. 0(0):1–12
Downloaded from https://academic.oup.com/nutritionreviews/advance-article/doi/10.1093/nutrit/nuaa074/5909185 by Universidade Federal de Goi�s user on 21 September 2020
mother is apparently related to an increased risk to the
newborn.
Metabolic effects of glycotoxins on metabolic
syndrome
One of the main glycotoxins is methylglyoxal (MG),
which may change cell behavior through modification
of biomolecules, such as proteins and DNA, and conse-
quent formation of AGEs.
18
Modification of arginine
residues by MG leads to the formation of Nd-(5-hydro-
5-methyl-4-imidazolon-2-yl)-ornithine (MG-H1) and
argpyrimidine from the imidazolones family, whereas
lysine modification leads to the formation of methyl-
glyoxal lysine dimer and (carboxyethyl)lysine.
6569
Modification of amino acid residues by MG affects in-
tracellular (i.e., transcription factors and cytoplasmic
proteins, including the proteasome and stress-response
pathways), circulating (ie, hemoglobin, albumin, or lip-
oproteins), and extracellular matrix proteins, changing
cell behavior and activating inflammatory and death
pathways.
18,7077
Indeed, MG modifies proteasome sub-
units and protein quality-control pathways (namely,
Hsc70, Hsp90, and Hsp27), causing endoplasmic reticu-
lum stress and impaired degradation of misfolded pro-
teins, in turn leading to a vicious circle of progressive
accumulation of misfolded proteins and impaired acti-
vation of detoxification systems.
7881
Besides directly
modifying protein structure through modification of
amino acid residues, MG also increases oxidative stress,
namely, the formation of superoxide anion,
8285
hydro-
gen peroxide, and peroxynitrite
82,83,86
in different types
of cells, including endothelial cells,
87
rat kidney mesan-
gial cells,
88
rat hepatocytes,
86,89
blood cells,
83,90
osteo-
blasts,
90
and in rat and mouse neurons.
9194
MG also
induces the depletion of antioxidant defenses, predis-
posing cells for oxidative damage.
82,88,9599
Given that
MG detoxification systems, namely the glyoxalase sys-
tem, are glutathione dependent, such mechanisms lead
to a self-perpetuating circle of reactive oxygen species
and AGEs formation and mitochondrial dysfunction.
97
Extracellular AGEs may change cell behavior
through activation of membrane receptors, such as
RAGE, which recognizes 2 major types of ligands: imi-
dazolones (MG-derived) and Ne-(carboxymethyl)lysine
(CML) adducts.
100
Upon activation, RAGE triggers in-
tracellular signaling pathways such as NF-jB, involved
in activation of inflammatory and proliferation or stress
signals, as well as generation of oxidative stress.
75,101106
Inhibition of RAGE or expression of soluble RAGE iso-
forms with the ability to scavenge AGEs prevented vas-
cular disease in several animal models.
101,107,108
Thus,
MG-induced changes in cell behavior involve several
mechanisms, namely the modification of biomolecules,
accumulation of misfolded proteins, activation of mem-
brane receptors, generation of oxidative stress, changes
in transcription factors, and activation of inflammatory
or stress pathways.
MG has been implicated in the development of dia-
betes complications such as retinopathy, nephropathy,
and peripheral neuropathy, given that its levels are in-
creased in patients with diabetes patients, and insulin-
independent cells like endothelial cells, podocytes, and
neurons are more susceptible to hyperglycemia-driven
MG formation.
18
Several studies have addressed the in-
volvement of MG in the mechanisms governing the de-
velopment of such pathologies, namely endothelial cell
senescence and angiogenesis impairment,
70,77,109,110
podocyte effacement and death,
111,112
glomerular fibro-
sis,
76,101,105,113
apoptosis of retinal pericytes and retinal
pigmented cells,
114117
and changes in the nociception
and pain stimuli (hyperalgesia).
118,119
Moreover, MG is
involved in the pathophysiology of cardio- and cerebro-
vascular diseases. MG causes structural changes in the
blood-brain barrier
120,121
and is involved in other neu-
rodegenerative disorders, such as increased neurotoxic-
ity,
122,123
b-amyloid protein neurotoxic effects,
124,125
and loss of dopaminergic neurons.
126128
In the cardio-
vascular system, MG impairs calcium handling between
sarcoplasmic reticulum and cytoplasm of cardiomyo-
cytes
129
and also affects survival and apoptotic pathways
during ischemia,
130,131
and angiogenic deficits.
132
Features of endothelial dysfunction, hypertension, and
atherosclerosis have also been reported, such as oxida-
tive stress and stiffness of the aorta, impaired elasticity,
acetylcholine-dependent relaxation, nitric oxide bio-
availability,
33,133136
activation of the renin-angiotensin
system,
137,138
increased glycoxidation of low-density li-
poprotein particles
139,140
and increased risk of throm-
bosis and atherosclerosis through platelet
hyperaggregation and RAGE activation.
141,142
Besides being implicated in the development of dia-
betic complications or associated diseases, MG also con-
tributes to the process of loss of metabolic homeostasis
itself, namely in the development of b-cell dysfunction
and insulin resistance. MG transiently activates insulin
secretion due to b-cell depolarization,
143
but it hampers
b-cell survival and long-term insulin synthesis and se-
cretion.
144
In insulin signaling, MG causes a redox-
independent inhibition of the insulin-receptor pathway
and GLUT4 translocation in muscle cells and 3T3 adi-
pocytes.
9,145,146
In vivo, MG caused insulin resistance in
several animal models,
144,145,147
but only when supra-
physiological doses were used.
18
Other studies did not
show MG-induced insulin resistance, which was only
observed in obese animal models.
25,148,149
Several stud-
ies have also shown AGE-induced overexpression of in-
flammatory mediators in the liver,
26,150,151
but again,
Nutrition Reviews
V
R
Vol. 0(0):1–12 3
Downloaded from https://academic.oup.com/nutritionreviews/advance-article/doi/10.1093/nutrit/nuaa074/5909185 by Universidade Federal de Goi�s user on 21 September 2020
hepatic insulin resistance was only observed in obese
animals.
26
Such results suggest that glycation may have
an impact in obesity-associated insulin resistance, possi-
bly through increased depletion of antioxidant and de-
toxifying mechanisms, but has a less dramatic effect in
lean models. In humans, elevated MG and AGE levels
have been reported in patients with diabetes and in met-
abolically unhealthy, obese patients, but no correlation
was found between AGE levels and impaired glucose
homeostasis.
152,153
Nevertheless, AGE-restricted diets
improve insulin sensitivity in normal and overweight
individuals, as well as patients with metabolic syndrome
and type 2 diabetes.
154157
Reports have shown the im-
pact of oral AGE restriction in the improvement of in-
sulin resistance, even in patients with metabolic
syndrome, which may reduce the risk of progression
from metabolically unhealthy and obese and having
metabolic syndrome to type 2 diabetes.
In summary, MG and MG-derived AGEs are in-
volved in several pathologies associated with metabolic
syndrome and diabetes, but their progressive accumula-
tion in biological systems may be also associated with
impaired lipid handling and increased susceptibility to
oxidative damage, which may contribute to the develop-
ment of insulin resistance in adipose tissue and liver in
obesity and predispose to the metabolically unhealthy,
obese phenotype. Together with increased b-cell dam-
age, such mechanisms are likely to contribute to the
progressive deterioration of metabolic homeostasis and
development of prediabetes and type 2 diabetes.
Importantly, the impact of early glycotoxin exposure
since the perinatal period is unknown, although recent
evidence suggests such exposure may increase the risk
of metabolic dysregulation and development of
diabetes-like complications in adult life.
Sources of perinatal glycotoxins exposure
In utero exposure to AGEs during embryonic
development. Similar to the other types of diabetes,
GDM-related hyperglycemia increases serum levels of
MG and AGEs, such as CML.
158,159
Increased serum
AGE levels are associated with insulin resistance, oxida-
tive stress, cardiovascular diseases, and diabetes comor-
bidities in normal individuals and pregnant
women.
33,160164
In addition to hyperglycemia, mater-
nal AGEs may also derive from dietary absorption,
given that industrialized foods are rich in AGEs
54
and
given their possible transfer to the embryo through the
placenta.
35
Accordingly, Konishi et al
165
reported the
impairment of implantation and placental growth and
function by the accumulation of AGEs through RAGE
activation, oxidative stress, low human chorionic go-
nadotropin levels, and apoptosis in human first-
trimester trophoblasts. Similarly, Hao et al
166
and
Haucke et al
167
reported the adverse effects of GDM
through raised AGEs levels during embryonic develop-
ment, which promote RAGE activation, inflammation,
and AGE accumulation in the embryo. This environ-
mental stress may collaborate to cause embryo resorp-
tion, fetus malformation, or preterm birth.
168
On the
other hand, knockout of soluble RAGE in pregnant dia-
betic rats prevents embryonic dysmorphogenesis,
169
and the administration of the soluble form of RAGE
during pregnancy reduces NF-jB activity in rat fetal
tissues.
170
Elevated sugar-sweetened soft beverages and re-
fined carbohydrates consumption during pregnancy
arestronglycorrelatedwithhighserumAGElevels,
offspring congenital heart defects, SGA newborns, and
increased risk of offspring overweight.
58,60,170,171
These data reinforce the role of AGE exposure on the
diabetic embryopathy and its implications for proper
fetus development, which are widely related to devel-
opmental origins of diseases at later stages of life.
However, data regarding the mechanisms involved in
AGEs passage through the placenta are not currently
available, to our knowledge, and studies are necessary
in this field.
Glycotoxin exposure during lactation period through
breast milk and infant formula. The lactation period is
essential to the proper development and maturation of
different organs and systems of the newborn, because
breast milk is to supply this nutritional demand. Given
the abundance of evidence regarding the importance of
breastfeeding in infant health, the World Health
Organization recommends exclusive breastfeeding until
6 months of life and complementary until age 2 years.
172
Breastfeeding prevents diseases such as diabetes, multi-
ple sclerosis, and celiac disease.
173
More than just a
source of calories, breast milk is an important source of
bioactive molecules such as antibodies, oligosacchar-
ides, and hormones, which exert beneficial effects for
the healthy development of newborns.
173,174
Insulin
may be found in breast milk and plays an important
role in the process of gut maturation, decreasing perme-
ability to macromolecules.
175
The milk composition depends on the maternal
metabolic status and there is evidence that breast milk
may also be a source of glycotoxins during lactation.
Human studies have shown that the neonatal intake of
breast milk from mothers with diabetes was related to
overweight and glucose intolerance.
176
Mericq et al
35
found a correlation between blood AGE levels of lactat-
ing mothers and their infants, raising the question of
whether maternal diet during lactation influences infant
glycoxidative stress. Even in other diseases, such as
4Nutrition Reviews
V
R
Vol. 0(0):1–12
Downloaded from https://academic.oup.com/nutritionreviews/advance-article/doi/10.1093/nutrit/nuaa074/5909185 by Universidade Federal de Goi�s user on 21 September 2020
beriberi, when an accumulation of glucose metabolites
such as MG occurs, there is an increased concentration
of these substances in breast milk.
177
Infants whose
mothers smoked during pregnancy and/or lactation
have increased accumulation of AGEs in their skin, in-
dicating that the transmission of glycotoxins from
mother to child may also occur through breast milk.
37
Altered composition of breast milk from obese dams,
caused by high-sugar consumption, programs rat off-
spring to develop obesity due to the impairment of mel-
anocortin system.
178
Other studies have demonstrated that the levels
and effects of breast-milk AGEs may also originate in
maternal diet. Cows fed a diet high in AGEs had in-
creased glycated compounds in their milk, such as MG-
H1.
179
On the other hand, a diet low in AGEs during
pregnancy and the neonatal period prevented the devel-
opment of type 1 diabetes in the offspring of NOD
mice.
38
In this regard, it was previously demonstrated
that oral administration of MG to lactating rats in-
creased the content of the glycation intermediary fruc-
tosamine in their milk, which was related to the
development of a diabetic phenotype in the offspring
during adult life.
180
Such observations are in line with
evidence that AGE levels in the blood could be derived
from the diet and not just produced endogenously. In
fact, a strong correlation between intake of AGEs and
AGE levels in the plasma has been demonstrated.
181,182
Similarly, evidence from human studies have shown
that dietary restriction of AGEs decreases their concen-
tration in plasma and their renal excretion.
155,183185
In
animals fed a
14
C-labelled, AGE-rich diet, as in humans,
10% of dietary AGEs are absorbed.
183,186
Indeed, the
glycation compound pirralyne, as well as major AGEs
such as CML, (carboxyethyl)lysine, and MG-H1, are
absorbed in the form of dipeptides via PEPT1 trans-
porter in intestinal cells.
187,188
Another source of glycotoxins during the perinatal
period are infant formulas, which commonly contain
high levels of AGEs, reaching almost a 35-fold higher
concentration of CML than breast milk of healthy
mothers.
39,189
AGEs are formed in heat-treated foods,
as a product from Maillard reaction, or nonenzymatic
browning. In fact, traditional methods of cooking that
use high temperature (100C–250C), such as frying,
baking, and grilling, contribute to a higher grade of
AGE formation, because foods rich in reducing sugars
and proteins are more prone to the formation of these
compounds.
190192
For instance, grilled beef has 5 times
higher AGE levels (5963 kU/100 g) than boiled beef
(1124 kU/100 g).
193
Also, infant formulas are rich in
sugars and proteins, and their industrial production
includes heat treatment. Hence, it was demonstrated
that hydrolysate infant formulas, rich in whey, have
higher concentrations of CML because whey proteins
are subjected to great heat treatment during
manufacturing of infant formula.
194
A positive correlation between formula-derived
AGEs, increased AGE circulating levels, and their uri-
nary excretion was found in newborns, indicating its
absorption.
189,195
In an animal model of intrauterine
growth restriction, animals fed a high-AGE formula
during suckling had CML accumulation in renal tubu-
lar cells that was associated with increased protein oxi-
dation and expression of pro-inflammatory and
apoptotic factors.
36
Similarly, intrauterine growth re-
stricted piglets fed a high-AGE formula during suckling
have increased liver oxidative stress at adulthood, due
to impaired antioxidant activity.
196
Some authors sug-
gest high consumption of glycation compounds through
infant formulas during early life may predispose to the
development of oxidative stress and diseases later in
life, such as diabetes.
35,197
It was observed that increased
maternal AGE levels were correlated with the infant
AGE levels, which may precondition the young to high
oxidative stress, inflammation, and insulin resistance.
35
A more recent investigation observed decreased insulin
sensitivity in infants fed AGE-rich formula compared
with those fed only breast milk, although the specific
AGE contribution to decreased insulin sensitivity was
not clear, because no differences were observed in
infants fed a low-AGE formula.
198
In this context, it was shown that glycation of
dairy protein by MG or glyoxal may decrease the pro-
tein digestibility by proteases, mainly due to cross-
linked AGEs.
199
On the other hand, noncross-linked
AGEs,suchasCML,(carboxyethyl)lysine,andMG-H1
are more prone to be absorbed by intestinal epithelial
cells.
200
High-molecular-weight AGEs are harder to di-
gest and absorb, so they are more able to advance in
the intestinal tract and interact with the colonic micro-
biota.
200,201
In fact, dietary AGEs may influence the
microbiota composition. In rats, dietary AGEs reduced
the diversity of microbiota, decreasing short-chain
fatty acid–producing bacteria and damaging the co-
lonic epithelial barrier.
202
Human studies also report
the interaction between dietary AGEs and changes in
gut microbiota composition, highlighting the impor-
tance of this interaction to human health.
203,204
However, little is known about the mechanisms of
AGE absorption in the neonatal gut. The newborn gut
is not totally mature, and the epithelial gut barrier of
newborns is still permeable to the passage of macro-
molecules, such as hormones, carbohydrates, and pep-
tides.
175,205
Thus, the newborn gut may be more
complacent to the passage of glycotoxins, making the
Nutrition Reviews
V
R
Vol. 0(0):1–12 5
Downloaded from https://academic.oup.com/nutritionreviews/advance-article/doi/10.1093/nutrit/nuaa074/5909185 by Universidade Federal de Goi�s user on 21 September 2020
rat pup more susceptible to the absorption and accu-
mulation of AGEs and their precursors. Newborn rats
are more susceptible to the toxic effects of orally deliv-
ered MG, because the lethal dose is almost 4 times
lower than that for an adult male rat (531 mg/kg vs
1990 mg/kg).
206
In short, exposure to increased AGE
levels by infant formulas or via breast milk are detri-
mental to health and proper development of the infant.
In general, the mechanisms of AGE absorption, diges-
tion, and interaction with the microbiota are not well
understood, and less is known about these mecha-
nisms in infancy; thus, more studies are necessary to
clarify them.
EFFECTS OF PERINATAL AGE EXPOSURE ON
PROGRAMMING OF METABOLIC SYNDROME,
CARDIOVASCULAR DISEASES AND EARLY AGING
Although several studies have reported high perinatal
exposure to AGEs during embryonic development and
lactation, little is known about their effects in metabolic
programming and in increasing the risk of development
of noncommunicable diseases in adulthood. Moreover,
the consumption of AGEs through milk or infant for-
mulas disturbs metabolic homeostasis in newborns and
is associated with pancreatic dysfunction and cardiovas-
cular and central nervous system diseases. Exposure of
lactating rats to high dietary levels of sucrose or high-
fructose corn syrup was observed to lead to increased
free fatty acid levels, adiposity, and liver fat in the off-
spring at weaning.
207
Accordingly, Csongov
aetal
208
have shown increased predisposition for weight gain
and insulin resistance in the progeny of females fed an
AGE-rich diet during pregnancy, and Francisco et al
180
have shown a similar impact of increased maternal ex-
posure to MG during lactation leading to an impaired
lipid profile and adiposity in the offspring. The authors
also reported decreased b-cell function in the off-
spring.
180
Accordingly, using type 1 diabetic NOD
mice, 2 different studies have shown the impact of peri-
natal AGE exposure on b-cell function. Peppa et al
38
have shown that low-glycotoxin fetal and neonatal envi-
ronments, through maternal AGE dietary restriction,
decreased T-cell inflammatory activity in the pancreas,
resulting in lower glycemia and increased survival.
Accordingly, Borg et al
209
have shown deteriorated b-
cell function in the progeny of NOD females exposed to
increased dietary AGE levels during pregnancy and
lactation.
The impact of perinatal AGEs exposure to other
pathologies is less studied, although a few studies have
implicated perinatal AGEing in the development of car-
diovascular diseases and central nervous system
disorders. Vascular diseases in adult life are associated
with increased glycoxidative stress, and increased pre-
natal AGE exposure also resulted in early cardiac
changes. Embryos of diabetic female rats accumulated
higher levels of CML, which was associated with lower
vascular endothelial growth factor levels.
210
As well,
AGE levels were increased in the heart of newborns of
streptozotocin-induced diabetic dams and were associ-
ated with increased oxidative stress and inflammatory
markers.
168
Recent reports have suggested impairment of the
AGE-RAGE axis in preterm birth. Chiavaroli et al
211
have shown decreased levels of soluble RAGE and en-
dogenous secretory RAGE in overweight prepubertal
children who were LGA or SGA, and these were corre-
lated with insulin resistance. In the central nervous sys-
tem, increased hippocampal RAGE expression was
observed in the offspring of streptozotocin-induced dia-
betic female rats, which was associated with increased
excitability and behavioral changes.
212
Increased glyca-
tion during gestational diabetes has been implicated in
impaired neural development, namely, in the decrease
of cortical neural precursor cells.
213
Authors have
shown that glyoxalase pathway disruption during em-
bryonic development leads to premature neurogenesis,
depletion of cortical neural precursor cells, and behav-
ioral changes, which were found in the offspring of dia-
betic murine mothers.
213
Thus, high AGE levels in mothers can predispose
their progeny to impaired metabolic homeostasis, and
recent data suggest defining cutoff values for maternal
glycated albumin levels during pregnancy to prevent
neonatal complications.
214,215
INTERVENTIONS TO PREVENT PERINATAL AGE
EXPOSURE AND METABOLIC PROGRAMMING
As previously described, exposure to glycotoxins during
perinatal life may occur in utero, because AGEs can
cross the placental barrier and impair fetal develop-
ment, activating the RAGE axis and increasing oxida-
tive stress, which may underlie the embryopathy related
to GDM. Furthermore, the exposure during lactation
may occur via breast milk, because maternal circulating
AGE levels may influence AGE concentration in the
milk. It is well established that uncontrolled glycemia in
GDM increases MG and AGE circulating levels, to
which the embryo is exposed. Thus, the first approach
to prevent MG and AGE exposure should be a proper
glycemic control. Metformin was suggested as an effi-
cient and safe drug for GDM treatment.
216
Besides im-
proving insulin sensitivity and decreasing hepatic
gluconeogenesis, metformin may directly react with
and scavenge MG, preventing the formation of MG-
6Nutrition Reviews
V
R
Vol. 0(0):1–12
Downloaded from https://academic.oup.com/nutritionreviews/advance-article/doi/10.1093/nutrit/nuaa074/5909185 by Universidade Federal de Goi�s user on 21 September 2020
derived AGEs such as MG-H1.
217,218
During lactation,
the same interventions may be administered to treat
maternal diabetes, thus preventing the transmission of
glycotoxins from mother to the infant through breast
milk.
As previously described, the diet is one of the main
sources of external glycotoxins. Because maternal AGEs
may be transmitted to the infant via placenta or breast
milk, the consumption of ultraprocessed or high-
temperature cooked food should be discouraged or con-
trolled, because they present high levels of AGEs. The
intake of fresh foods should be encouraged, such as in
natura vegetables and fruits as part of balanced diet.
Attention should be taken in the cooking process,
avoiding high temperature methods such as frying and
grilling, opting for low-temperature methods such as
boiling.
AGEs are largely found in infant formulas, contrib-
uting to increase the pool of AGEs in the infant. As rec-
ommended by the World Health Organization,
breastfeeding must be exclusive during the first
6 months of life.
172
In this sense, infant formula must be
implemented only when breast milk was not available,
thus avoiding unnecessary use. As mentioned, the in-
dustrial process to obtain whey protein leads to a higher
degree of AGE formation; therefore, the addition of
whey protein should be avoided. The use of milk from
different animals, such as goat, should be encouraged,
because their amino acidic profile is more similar to the
human milk, making the addition of whey protein un-
necessary, thus reducing the amount of AGEs in the fi-
nal product.
194
Thus, some interventions may be taken to prevent
AGE exposure during perinatal life, including proper
glycemic control in mothers with diabetes and the
adoption of a balanced diet low in ultraprocessed food.
Quitting smoking may also be an important interven-
tion, because smoking during lactation may increase
AGE levels in breast milk.
37
Infant formulas should be
prescribed with caution, and industry should be en-
couraged to develop infant formulas with low AGE
levels.
CONCLUSION
More studies are needed to understand the mechanisms
underlying the effects of perinatal, neonatal, and in-
fancy exposure to glycotoxins to prevent the metabolic
programming of diseases due to the embryo and infant
exposure to AGEs. In clinical practice, the advice to
Figure 1 Main sources and potential mechanisms by which exposure to glycotoxins during perinatal life (eg, gestation, lactation)
may potentially program cardiometabolic disease development during adult life. Abbreviations: AGE, advanced glycation end product;
MG, methylglyoxal.
Nutrition Reviews
V
R
Vol. 0(0):1–12 7
Downloaded from https://academic.oup.com/nutritionreviews/advance-article/doi/10.1093/nutrit/nuaa074/5909185 by Universidade Federal de Goi�s user on 21 September 2020
pregnant and lactating women about the importance of
the diet and glycemic control is essential. To study the
long-term effects of intrauterine and postnatal exposure
to glycotoxins in humans, a long follow-up of the off-
spring and mother is required, given that studies about
this issue are currently scarce.
Acknowledgments
Author contributions. F.A.F., P.M., and R.M.G. outlined
and drafted the manuscript. All the authors contributed
to the writing of the manuscript. P.M., P.C.F.M., and
R.M.G. supervised the work. All the authors revised and
approved the manuscript for publication.
Funding. Financial support was received from the fol-
lowing Brazilian funding agencies: Conselho Nacional
de Desenvolvimento Cient
ıfico e Tecnol
ogico and
Coordenac¸~
ao de Aperfeic¸oamento Pessoal de N
ıvel
Superior. None of the funding agencies were involved
with the conception, design, performance, or approval
of this study.
Declaration of interest. None.
REFERENCES
1. Albracht-Schulte K, Kalupahana NS, Ramalingam L, et al. Omega-3 fatty acids in
obesity and metabolic syndrome: a mechanistic update. JNutrBiochem.
2018;58:1–16.
2. Desai M, Jellyman JK, Han G, et al. Maternal obesity and high-fat diet pro-
gram offspring metabolic syndrome. Am J Obstet Gynecol. 2014;211:237.
e1–237.e13.
3. Pan Y, Kong LD. High fructose diet-induced metabolic syndrome: pathophysio-
logical mechanism and treatment by traditional Chinese medicine. Pharmacol
Res. 2018;130:438–450.
4. Sarman I. Review shows that early foetal alcohol exposure may cause adverse
effects even when the mother consumes low levels. Acta Paediatr. 2018;107:
938–941.
5. Walters KA, Bertoldo MJ, Handelsman DJ. Evidence from animal models on the
pathogenesis of PCOS. Best Pract Res Clin Endocrinol Metab. 2018;32:271–281.
6. McLellan AC, Thornalley PJ. Glyoxalase activity in human red blood cells frac-
tioned by age. Mech Ageing Dev. 1989;48:63–71.
7. McLellan AC, Thornalley PJ, Benn J, et al. Glyoxalase system in clinical diabetes
mellitus and correlation with diabetic com plications. Clin Sci. 1994;87:21– 29.
8. Oliveira LM, Lages A, Gomes RA, et al. Insulin glycation by methylglyoxal results
in native-like aggregation and inhibition of fibril formation. BMC Biochem.
2011;12:41.
9. Riboulet-Chavey A, Pierron A, Durand I, et al. Methylglyoxal impairs the insulin
signaling pathways independently of the formation of intracellular reactive oxy-
gen species. Diabetes. 2006;55:128 9–1299.
10. Thornalley PJ. The glyoxalase system: new developments towards functional
characterization of a metabolic pathway fundamental to biological life. Biochem
J. 1990;269:1–11.
11. Thornalley PJ. Modification of the glyoxalase system in human red blood cells by
glucose in vitro. Biochem J. 1988;254:751–755.
12. Baidoshvili A, Niessen HWM, Stooker W, et al. Ne-(carboxymethyl)lysine deposi-
tions in human aortic heart valves: similarities with atherosclerotic blood vessels.
Atherosclerosis. 2004;174:28 7–292.
13. Cuccurullo C, Iezzi A, Fazia ML, et al. Suppression of RAGE as a basis of
simvastatin-dependent plaque stabilization in type 2 diabetes. Arterioscler
Thromb VascBiol. 2006;26:2716–2723.
14. Hanssen NMJ, Stehouwer CDA, Schalkwijk CG. Methylglyoxal and glyoxalase I in
atherosclerosis. Biochem Soc Trans.2014;42:443–449.
15. Hanssen NMJ, Wouters K, Huijberts MS, etal. Higher levels of advanced glycation
endproducts in human carotid atherosclerotic plaques are associated with a
rupture-prone phenotype. Eur Heart J. 2014;35:1137–1146.
16. Heier M, Margeirsdottir HD, TorjesenPA, et al. The advanced glycation end prod-
uct methylglyoxal-derived hydroimidazolone-1 and early signs of atherosclerosis
in childhooddiabetes. Diabetes Vasc Dis Res. 2015;12:139–145.
17. Kume S, Takeya M, Mori T, et al. Immunohistochemical and ultrastructural detec-
tion of advanced glycation end products in atherosclerotic lesions of human
aorta with a novelspecific monoclonal antibody. Am J Pathol. 1995;14 7:654–66 7.
18. Matafome P, Rodrigues T, Sena C, et al. Methylglyoxal in metabolic disorders:
facts, myths, and promises.Med Res Rev. 2017;37:368–403.
19. Matafome P, Sena C, Seic¸a R. Methylglyoxal, obesity, and diabetes. Endocrine.
2013;43:472–484.
20. Nakamura Y, Horii Y, Nishino T, et al. Immunohistochemical localization of ad-
vanced glycosylation endproducts in coronary atheroma and cardiactissue in di-
abetes mellitus. Am J Pathol. 1993;143:1649 –1656.
21. Nakayama K, Nakayama M, Iwabuchi M, et al. Plasma a-oxoaldehyde levels in di-
abetic and nondiabetic chronic kidney disease patients. Am J Nephrol.
2008;28:871–878.
22. Odani H, Shinzato T, Matsumoto Y, et al. Increase in three a,b-dicarbonyl com-
pound levels in human uremic plasma: specific in vivo determination of inter-
mediates in advanced Maillard reaction. Biochem Biophys Res Commun.
1999;256:89–93.
23. Sell DR, Monnier VM. Molecular basis of arterial stiffening: role of glycation-a
mini-review. Gerontology. 2012;58:227–237.
24. SrikanthV, Westcott B, ForbesJ, et al. Methylglyoxal, cognitive function andcere-
bral atrophyin older people. J Gerontol A Biol Sci MedSci. 2013;6 8:68–73.
25. Rodrigues T, Matafome P, Sereno J, et al. Methylglyoxal-induced glycation
changes adipose tissue vascular architecture, flow and expansion, leading to in-
sulin resistance. Sci Rep. 2017;7:1698.
26. Neves C, Rodrigues T, Sereno J, et al. Dietary glycotoxins impair hepatic lipidemic
profile in diet-induced obese rats causing hepatic oxidative stress and insulin re-
sistance. Oxid Med Cell Longev. 2019;2019:1–14.
27. Lee HJ, Howell SK, Sanford RJ, et al. Methylglyoxal can modify GAPDH activity
and structure. Ann N Y Acad Sci. 2005;1043:135–1 45.
28. Loske C, Neumann A, Cunningham AM, et al. Cytotoxicity of advanced glycation
endproducts is mediated by oxidative stress. JNeuralTransm.
1998;105:1005–1015.
29. Rosca MG, Mustata TG, Kinter MT, et al. Glycation of mitochondrial proteins from
diabetic rat kidney is associated with excess superoxide formation. Am J Physiol
Renal Physiol. 2005;289:F420–F430.
30. Wu L, Juurlink B. Increased methylglyoxal and oxidative stress in hypertensive
ratvascularsmoothmusclecells.Hypertension. 2002;39:809–814 .
31. Brownlee M. Biochemistry and molecular cell biology of diabetic complications.
Nature. 2001;414:813–820.
32. Funk SD, Yurdagul A, Orr AW. Hyperglycemia and endothelial dysfunction in ath-
erosclerosis: lessonsfrom type 1 diabetes. Int J Vasc Med. 2012;2012:1–19.
33. Sena CM, Matafome P, Cris
ostomo J, et al. Methylglyoxal promotes oxidative
stress and endothelial dysfunction. Pharmacol Res. 2012;65:497–506.
34. Su Y, Lei X, Wu L, etal. The role of endothelial cell adhesion molecules P-selectin,
E-selectin and intercellular adhesion molecule-1 in leucocyte recruitment in-
duced by exogenous methylglyoxal. Immunology. 2012;137:65–79.
35. Mericq V, Piccardo C, Cai W, et al. Maternally transmitted and food-derived gly-
cotoxins: a factor preconditioning the young to diabetes? Diabetes Care.
2010;33:2232–2237.
36. Elmhiri G, Mahmood DFD, Niquet-Leridon C, et al. Formula-derived advanced
glycation end products are involved in the development of long-term inflamma-
tion and oxidative stress in kidney of IUGR piglets. Mol Nutr Food Res.
2015;59:939–947.
37. Federico G, Gori M, Randazzo E, et al. Skin advanced glycation end-products
evaluation in infants according to the type of feeding and mother’s smoking
habits. SAGEOpen Med. 2016; 4:205031211668 212.
38. Peppa M, He C, Hattori M, et al. Fetal or neonatal low-glycotoxin environment
prevents autoimmune diabetes in NOD mice. Diabetes. 2003;52:1441–1448.
39. Kutlu T. Dietary glycotoxins and infant formulas. Turk Pediatri Ars.
2016;51:179–185.
40. Pischetsrieder M, Henle T. Glycation products in infant formulas: chemical, ana-
lytical and physiological aspects. Amino Acids. 2012;42:1111–1118.
41. Angueira AR, Ludvik AE, Reddy TE, et al. New insights into gestational glucose
metabolism: lessons learned from 21st century approaches. Diabetes.
2015;64:327–334.
42. Arnaout R, Nah G, Marcus G, et al. Pregnancy complications and premature car-
diovascular events among 1.6 million California pregnancies. Open Heart.
2019;6:1–10.
43. Farrar D, Simmonds M, Griffin S, et al. The identification and treatment of women
with hyperglycaemia in pregnancy: an analysisof individual participant data, sys-
tematic reviews, meta-analyses and an economic evaluation. Health Technol
Assess. 2016;20:1–382.
44. Holness N. High-risk pregnancy. Nurs Clin North Am. 2018;53:241–251 .
8Nutrition Reviews
V
R
Vol. 0(0):1–12
Downloaded from https://academic.oup.com/nutritionreviews/advance-article/doi/10.1093/nutrit/nuaa074/5909185 by Universidade Federal de Goi�s user on 21 September 2020
45. Giannakou K, Evangelou E, Yiallouros P, et al. Risk factors for gestationaldiabetes:
an umbrella review of meta-analyses of observational studies. PLoS One.
2019;14:E0215372.
46. Ornoy A, Reece EA, Pavlinkova G, et al. Effect of maternal diabetes on the em-
bryo, fetus, and children: congenital anomalies, genetic and epigenetic changes
and developmental outcomes. Birth DefectRes C Embryo Today. 2015;105:53–72.
47. Papathakis PC, Singh LN, Manary MJ. How maternal malnutrition affects linear
growth and development in the offspring. Mol Cell Endocrinol. 2016;435:40–47.
48. Sinzato YK, Bevilacqua EM, Volpato GT, et al. Maternal oxidative stress, placental
morphometry, and fetal growth in diabetic rats exposed to cigarette smoke.
Reprod Sci. 2019;26:1287–1293.
49. Zeng Z, Liu F, Li S. Metabolic adaptations in pregnancy: a Review. Ann Nutr
Metab. 2017;70:59–65.
50. Silva CCV, Vehmeijer FOL, El Marroun H, et al. Maternal psychological distress
during pregnancy and childhood cardio-metabolic risk factors. Nutr Metab
Cardiovasc Dis. 2019;29:572–579.
51. Barker DJP, Osmond C, Golding J, et al. Growth in utero, blood pressure in child-
hood and adult life, and mortality from cardiovascular disease. Br Med J.
1989;298:564–567.
52. Eriksson UJ, Wentzel P. The status of diabetic embryopathy. Ups J Med Sci.
2016;121:96–112.
53. Li S, Yang H. Relationship between advanced glycation end products and gesta-
tional diabetes mellitus. JMaternNeonatalMed. 2019;32 :2783–27 89.
54. Takeuchi M, Takino JI, Furuno S, et al. Assessment of the concentrations of vari-
ous advanced glycation end-products in beverages and foods that are com-
monly consumed in Japan. PLoSOne. 2015;10:E0118652.
55. Reece EA. Diabetes-induced birth defects: what do we know? What can we do?
Curr Diab Rep. 2012;12:24–32.
56. A. Lawlor D, Fraser A, Lindsay RS, et al. Association of existing diabetes, gesta-
tional diabetes and glycosuria in pregnancy with macrosomia and offspring
body mass index, waist and fat mass in later childhood: findings from a prospec-
tive pregnancy cohort. Diabetologia. 2010;53:89–97.
57. Olmedo-Requena R, G
omez-Fern
andez J, Amezcua-Prieto C, et al. Pre-pregnancy
adherence to the Mediterranean diet and gestational diabetes mellitus: a case-
control study. Nutrients. 2019;11:1003– 1 011.
58. Amezcua-Prieto C, Mart
ınez-Galiano JM, Cano-Ib
a~
nez N, et al. Types of carbohy-
drates intake during pregnancy and frequency of a small for gestational age
newborn: a case-control study. Nutrients. 2019;11:523.
59. Vaiserman A, Lushchak O. Prenatal malnutrition-induced epigenetic dysregula-
tion as a risk factor for type 2 diabetes. Int J Genomics. 201 9;2019:1 –11.
60. Mullie P, Clarys P. Consumption of artificially sweetened beverages during preg-
nancy is associated with a twofold higher risk of infant being overweight at 1
year. Evid Based Nurs. 2017;20:11–11.
61. Palatianou ME, Simos YV, Andronikou SK, et al. Long-term metabolic effects of
high birth weight: a critical review of the literature. Horm Metab Res.
2014;46:911–920.
62. Plagemann A, Harder T, Rodekamp E, et al. Rapid neonatal weight gain increases
risk of childhood overweight in offspring of diabetic mothers. J Perinat Med.
2012;40:557–563.
63. Schellong K, Schulz S, Harder T, et al. Birth weight and long-term overweight
risk: systematic review and a meta-analysis including 643,902 persons from 66
studies and 26 countries globally. PLoS One.2012;7:E47776.
64. Harder T, Rodekamp E, Schellong K, et al. Birth weight and subsequent risk of
type 2 diabetes: a meta-analysis. Am J Epidemiol. 2007;16 5:849–857.
65. Kalapos MP. Where does plasma methylglyoxal originate from? Diabetes Res Clin
Pract. 2013;99:260 –271.
66. Poulsen MW, Hedegaard RV, Andersen JM, et al. Advanced glycation end prod-
ucts in food andtheir effects on health. Food Chem Toxicol. 2013;60:10–37.
67. Allaman I, B
elanger M, Magistretti PJ. Methylglyoxal, the dark side of glycolysis.
Front Neurosci. 9:23.
68. Falone S, D’Alessandro A, Mirabilio A, et al. Long term running biphasi-
cally improves methylglyoxal-related metabolism, redox homeostasis and
neurotrophic support within adult mouse brain cortex. PLoS One.
2012;7:E31401.
69. Masania J, Malczewska-Malec M, Razny U, et al. Dicarbonyl stress in clinical obe-
sity. Glycoconj J. 2016;33:581–589.
70. Yao D, TaguchiT, Matsumura T, et al. High glucose increasesangiopoietin-2 tran-
scription in microvascular endothelial cells through methylglyoxal modification
of mSin3A. JBiolChem.2 007;282 :31038–3 1045.
71. Carlsson H, To¨rnqvist M. Strategy for identifying unknown hemoglobin adducts
using adductome LC-MS/MS data: identification of adducts corresponding to
acrylic acid, glyoxal, methylglyoxal, and 1-octen-3-one. Food Chem Toxicol.
2016;92:94–103.
72. Bose T, Bhattacherjee A, Banerjee S, et al. Methylglyoxal-induced modifications
of hemoglobin: structural and functional characteristics. Arch Biochem Biophys.
2013;529:99–104.
73. Guerin-Dubourg A, Catan A, Bourdon E, et al. Structural modifications of human
albumin in diabetes. Diabetes Metab. 2012;38:171–178.
74. Rabbani N, Godfrey L, Xue M, et al. Glycation of LDL by methylglyoxal increases
arterial atherogenicity: a possible contributor to increased risk of cardiovascular
disease in diabetes. Diabetes. 2011;60:1 973–1980.
75. Goldin A, BeckmanJA, SchmidtAM, et al. Advanced glycation end products: spark-
ing the development of diabetic vascular injury. Circulation. 2006;114:597–605.
76. Pedchenko VK, Chetyrkin SV, Chuang P, et al. Mechanism of perturbation of
integrin-mediated cell-matrix interactions by reactive carbonyl compounds and
its implication for pathogenesis of diabetic nephropathy. Diabetes.
2005;54:2952–2960.
77. Bento CF, Fernandes R, Matafome P, et al. Methylglyoxal-induced imbalance in
the ratio of vascular endothelial growth factor to angiopoietin 2 secreted by reti-
nal pigment epithelial cells leads to endothelial dysfunction. Exp Physiol.
2010;95:955–970.
78. Bento CF, Marques F, Fernandes R, et al. Methylglyoxal alters the function and
stability of critical components of the protein quality control. Cotterill S, ed. PLoS
One. 2010;5:E13007.
79. Padival AK,Crabb JW, Nagaraj RH. Methylglyoxal modifies heat shock protein 27
in glomerularmesangial cells. FEBS Lett. 2003;551:113–118.
80. Palsamy P, Bidasee KR, Ayaki M, et al. Methylglyoxal induces endoplasmic reticu-
lum stressand DNA demethylation in the Keap1 promoter of human lens epithe-
lial cells and age-related cataracts. Free Radic Biol Med. 2014;72:134–148.
81. Nam DH, Han JH, Lee TJ, et al. CHOP deficiency prevents methylglyoxal-induced
myocyte apoptosis and cardiac dysfunction. J Mol Cell Cardiol. 2015;85:168–1 77.
82. Chang T, Wang R, Wu L. Methylglyoxal-induced nitric oxide and peroxynitrite
production in vascular smooth muscle cells. Free Radic Biol Med.
2005;38:286–293.
83. Ward RA, McLeish KR. Methylglyoxal: a stimulus to neutrophil oxygen radical
production in chronic renal failure? NephrolDial Transplant. 2004;19:170 2–1707.
84.
Skrha J, G
all J, Buchal R, et al. Glucose and its metabolites have distinct effects
on the calcium-induced mitochondrial permeability transition. Folia Biol (Czech
Republic). 2011;57:96–103.
85. Remor AP, de Matos FJ, Ghisoni K, et al. Differential effects of insulin on periph-
eral diabetes-related changes in mitochondrial bioenergetics: involvement of ad-
vanced glycosylated end products. Biochim Biophys Acta Mol Basis Dis.
2011;1812:1460–1471.
86. Kalapos MP, Littauer A,qde Groot H. Has reactive oxygen a role in methylglyoxal
toxicity?A study on cultured rathepatocytes. Arch Toxicol. 1993;67:369–372.
87. Akhand AA, Hossain K, Mitsui H, et al. Glyoxal and methylglyoxal trigger distinct
signals for MAP family kinases and caspase activation in human endothelial cells.
Free Radic BiolMed. 200 1;31:20–30 .
88. Uriuhara A, Miyata S, Liu BF, et al. Methylglyoxal induces prostaglandin E2 pro-
duction in rat mesangial cells. Kobe J Med Sci. 2008;53:305–315.
89. Seo K, Ki SH, Shin SM. Methylglyoxal induces mitochondrial dysfunction and cell
death in liver. Toxicol Res. 2014;30:193–198.
90. Suh KS, Choi EM, Rhee SY, et al. Methylglyoxal induces oxidative stress and mito-
chondrial dysfunction in osteoblastic MC3T3-E1 cells. Free Radic Res.
2014;48:206–217.
91. Di Loreto S, Caracciolo V, Colafarina S, et al. Methylglyoxal induces oxidative
stress-dependent cell injury and up-regulation of interleukin-1band nerve
growth factor in cultured hippocampal neuronal cells. Brain Res.
2004;1006:157–167.
92. Dafre AL, Goldberg J, Wang T, et al. Methylglyoxal, the foe and friend of glyoxa-
lase and Trx/TrxR systems in HT22 nerve cells. Free Radic Biol Med. 2015;89:8–19.
93. Kikuchi S, Shinpo K, Moriwaka F, et al. Neurotoxicity of methylglyoxal and 3-
deoxyglucosone on cultured cortical neurons: synergism between glycation and
oxidative stress, possibly involved in neurodegenerative diseases. JNeurosciRes.
1999;57:280–289.
94. Amicarelli F, Colafarina S, Cattani F, et al. Scavenging system efficiency is crucial
for cell resistance to ROS-mediated methylglyoxal injury. Free Radic Biol Med.
2003;35:856–871.
95. Morgan PE, Sheahan PJ, Pattison DI, et al. Methylglyoxal-induced modification of
arginine residues decreases the activity of NADPH-generating enzymes. Free
Radic Biol Med. 2013;61:229–2 42.
96. Suravajjala S, Cohenford M, Frost LR, etal. Glycation of human erythrocyte gluta-
thione peroxidase: effect on the physical and kinetic properties. Clin Chim Acta.
2013;421:170–176.
97. Pun PBL, Logan A, Darley-Usmar V, et al. A mitochondria-targeted mass spec-
trometry probe to detect glyoxals: implications for diabetes. Free Radic Biol Med.
2014;67:437–450.
98. Paget C, Lecomte M, Ruggiero D, et al. Modification ofenzymatic antioxidants in
retinal microvascular cells by glucose or advanced glycation end products. Free
Radic Biol Med. 1998;25:121–1 29.
99. Di Loreto S, Zimmitti V, Sebastiani P, et al. Methylglyoxal causes strong weaken-
ing of detoxifying capacity and apoptotic cell death in rat hippocampal neurons.
Int J Biochem Cell B iol. 2008;40:245–257.
100. Yan SF, Ramasamy R, Schmidt AM. Receptor for AGE (RAGE) and its ligands-cast
into leading roles in diabetes and the inflammatory response. J Mol Med.
2009;87:235–247.
Nutrition Reviews
V
R
Vol. 0(0):1–12 9
Downloaded from https://academic.oup.com/nutritionreviews/advance-article/doi/10.1093/nutrit/nuaa074/5909185 by Universidade Federal de Goi�s user on 21 September 2020
101. Negre-Salvayre A, Salvayre R, Aug
e N, et al. Hyperglycemia and glycation in dia-
betic complications. Antioxidants Redox Signal. 2009;11:307 1–3109.
102. Yan SF, Ramasamy R, Naka Y, etal. Glycation, Inflammation, and RAGE: a scaffold
for the macrovascular complications of diabetes and beyond. Circ Res.
2003;93:1159–1169.
103. Xue J, Rai V, Singer D, et al. Advanced glycation end product recognition by the
receptor forAGEs. Structure. 2011;19:722–732.
104. Xue J, Ray R, Singer D, et al. The receptor for advanced glycation end products
(RAGE) specifically recognizes methylglyoxal-derived AGEs. Biochemistry.
2014;53:3327–3335.
105. Liu BF, Miyata S, Hirota Y, et al. Methylglyoxal induces apoptosis through activa-
tion of p38 mitogen-activated protein kinase in rat mesangial cells. Kidney Int.
2003;63:947–957.
106. Du J, Cai S, Suzuki H, et al. Involvement of MEKK1/ERK/P21Waf1/Cip1 signal
transduction pathway in inhibition of IGF-I-mediated cell growth response by
methylglyoxal. J Cell Biochem. 2003;88:1235–1246.
107. Yao D, Brownlee M. Hyperglycemia-induced reactive oxygen species increase ex-
pression of the receptor for advanced glycation end products (RAGE) and RAGE
ligands. Diabetes. 2010;59:249 –255.
108. Ueno H, Koyama H, Shoji T, et al. Receptor for advanced glycation end-products
(RAGE) regulation of adiposity and adiponectin is associated with atherogenesis
in apoE-deficient mouse. Atherosclerosi s. 2010;211:431–436.
109. Thangarajah H, Yao D, Chang EI, et al. The molecular basis for impaired hypoxia-
induced VEGF expression in diabetic tissues. Proc Natl Acad Sci USA.
2009;106:13505–13510.
110. Berlanga J, Cibrian D, Guill
en I, et al. Methylglyoxal administration induces
diabetes-like microvascular changes and perturbs the healing process of cutane-
ous wounds. Clin Sci. 2005;109:83 –95.
111. Diez-Sampedro A, Lenz O, Fornoni A. Podocytopathy in diabetes: a metabolic
and endocrine disorder. Am J Kidney Dis. 2011;58:637 –646.
112. Giacco F, Du X, D’Agati VD, et al. Knockdown ofglyoxalase 1 mimics diabetic ne-
phropathyin nondiabetic mice. Diabetes. 2014;63:291–2 99.
113. Mostafa AA, Randell EW, Vasdev SC, et al. Plasma protein advanced glycation
end products, carboxymethyl cysteine, and carboxyethyl cysteine, are elevated
and related to nephropathy in patients with diabetes. Mol Cell Biochem.
2007;302:35–42.
114. Kim J, Kim OS, Kim CS, et al. Accumulation of argpyrimidine, a methylglyoxal-
derived advanced glycation end product, increases apoptosis of lens epithelial
cells both in vitro and in vivo. Exp Mol Med. 2012;44:167–175.
115. Kim J, Kim OS, Kim CS, et al. Cytotoxic role of methylglyoxal in rat retinal peri-
cytes: Involvement of a nuclear factor-kappaB and inducible nitric oxide synthase
pathway. Chem Biol Interact.2010;188:86–93.
116. Kim OS, Kim J, Kim CS, et al. KIOM-79 prevents methyglyoxal-induced retinal
pericyte apoptosis in vitroand in vivo. J Ethnopharmacol. 2010;129:285–29 2.
117. Kim J, Son JW, Lee JA, et al. Methylglyoxal induces apoptosis mediated by reac-
tive oxygen species in bovine retinal pericytes. J Korean Med Sci.
2004;19:95–100.
118. Bierhaus A, Fleming T, Stoyanov S, et al. Methylglyoxal modification of Na v 1.8
facilitates nociceptive neuron firingand causes hyperalgesiain diabetic neuropa-
thy. Nat Med. 2012;18:926–933.
119. Skapare E, Konrade I, Liepinsh E, et al. Association of reduced glyoxalase 1 activ-
ity and painful peripheral diabetic neuropathy in type 1 and 2 diabetes mellitus
patients. JDiabetes Complications. 2013;27:262–267.
120. Li W, Maloney RE, Aw TY. High glucose, glucose fluctuation and carbonyl stress
enhance brain microvascular endothelial barrier dysfunction: implications for di-
abetic cerebral microvasculature. Redox Biol. 2015;5:80–9 0.
121. Li W, Maloney RE, Circu ML, et al. Acute carbonyl stress induces occludin glyca-
tion and brain microvascular endothelial barrier dysfunction: role for
glutathione-dependent metabolism of methylglyoxal. Free Radic Biol Med.
2013;54:51–61.
122. Chun HJ, Lee Y, Kim AH, et al. Methylglyoxal causes cell death in neural progeni-
tor cells and impairs adult hippocampal neurogenesis. Neurotox Res.
2016;29:419–431.
123. Heimfarth L, Loureiro SO, Pierozan P, et al. Methylglyoxal-induced cytotoxicity in
neonatal rat brain: a role for oxidative stress and MAP kinases. Metab Brain Dis.
2013;28:429–438.
124. Li XH, Du LL, Cheng XS, et al. Glycation exacerbates the neuronal toxicity of b-
amyloid. Cell Death Dis. 2013;4:e673.
125. Lovestone S, Smith U. Advanced glycation end products, dementia, and diabe-
tes. Proc Natl AcadSci USA. 2014;111:4743–4744.
126. Xie B, Lin F, Peng L, et al. Methylglyoxal increases dopamine level and leads to
oxidative stress in SH-SY5Y cells. Acta Biochim Biophys Sin (Shanghai).
2014;46:950–956.
127. Xie B, Lin F, Ullah K, et al. A newly discovered neurotoxin ADTIQ associated with
hyperglycemia and Parkinson’s disease. Biochem Biophys Res Commun.
2015;459:361–366.
128. Song DW,Xin N, Xie BJ, et al. Formation of a salsolinol-like compound,the neuro-
toxin, 1-acetyl-6,7-dihydroxy-1,2,3,4-tetrahydroisoquinoline, in a cellular model
of hyperglycemia and a rat model of diabetes. Int J Mol Med. 2014;33:736–742.
129. Tian C, Alomar F, Moore CJ, et al. Reactive carbonyl species and their roles in sar-
coplasmic reticulum Ca 2þcycling defect in the diabetic heart. Heart Fail Rev.
2014;19:101–112.
130. Cris
ostomo J, Matafome P, Santos-Silva D, et al. Methylglyoxal chronic adminis-
tration promotes diabetes-like cardiac ischaemia disease in Wistar normal rats.
Nutr Metab Cardiovasc Dis. 2013;23:1223–1230.
131. Almeida F, Santos-Silva D, Rodrigues T, et al. Pyridoxamine reverts
methylglyoxal-induced impairment of survival pathways during heart ischemia.
Cardiovasc Ther. 2013;31:e79–e85.
132. Molgat ASD, Tilokee EL, Rafatian G, et al.Hyperglycemia inhibits cardiacstem cell-
mediated cardiac repair and an giogenic capacity. Circulation. 2014;13 0:S70–S76.
133. Su Y, Qadri SM, Wu L, et al. Methylglyoxal modulates endothelial nitric oxide
synthase-associated functions in EA.hy926 endothelial cells. Cardiovasc Diabetol.
2013;12:134.
134. Su Y, Qadri SM, Hossain M, et al. Uncoupling of eNOS contributes to redox-
sensitive leukocyte recruitment and microvascular leakage elicited by methyl-
glyoxal. Biochem Pharmacol. 2013;86:1 762–1774.
135. Mukohda M, Yamawaki H, Nomura H, et al. Methylglyoxal inhibits smooth mus-
cle contraction in isolatedblood vessels. J PharmacolSci. 2009;109:305–310.
136. Semba RD, Najjar SS, Sun K, Lakatta EG, et al. Serum carboxymethyl-lysine, an ad-
vanced glycation end product, is associated with increased aortic pulse wave ve-
locity in adults. Am J Hypertens. 2009;22:74–79.
137. Dhar I, Dhar A, Wu L, et al. Methylglyoxal, a reactive glucose metabolite,
increases renin angiotensin aldosterone and blood pressure in male Sprague-
Dawley rats.Am J Hypertens. 2014;27:308–316 .
138. Dhar I, Dhar A, Wu L, et al. Increased methylglyoxal formation with upregulation
of renin angiotensin system in fructose fed Sprague Dawley rats. PLoS One.
2013;8:E74212.
139. Rabbani N, Chittari MV, Bodmer CW, et al. Increased glycation and oxidative
damage to apolipoprotein B100 of LDL cholesterol in patients with type 2 diabe-
tes and effect of metformin. Diabetes. 2010;59 :1038–10 45.
140. Beisswenger PJ, Howell SK, Touchette AD, et al. Metformin reduces systemic
methylglyoxal levels in type 2 diabetes. Diabetes. 1999; 48:198–202.
141. Tikellis C, Pickering RJ, Tsorotes D, et al. Dicarbonyl stress in the absence of hy-
perglycemia increases endothelial inflammation and atherogenesis similar to
that observed in diabetes.Diabetes. 2014;63:3915–3925.
142. Hadas K, Randriamboavonjy V, Elgheznawy A, et al. Methylglyoxal induces plate-
let hyperaggregation and reduces thrombus stability by activating PKC and
inhibiting PI3K/Akt pathway. PLoS One. 2013;8:E74401.
143. Yang Y, Konduru AS, Cui N, et al. Acute exposure of methylglyoxal leads to acti-
vation of KATP channels expressed in HEK293 cells. Acta Pharmacol Sin.
2014;35:58–64.
144. Dhar A, DharI, Jiang B, et al. Chronicmethylglyoxal infusion by minipump causes
pancreatic b-cell dysfunction and induces type 2 diabetes in Sprague-Dawley
rats. Diabetes. 2011;60:899–908.
145. Jia X, Wu L. Accumulation of endogenous methylglyoxal impaired insulin signaling
in adipose tissue of fructose-fed rats. Mol Cell Biochem. 2007;306:133–139.
146. Engelbrecht B, Stratmann B, Hess C, et al. Impact of GLO1 knock down on GLUT4
trafficking and glucose uptake in L6 myoblasts. PLoS One. 2013;8:E65195.
147. Dhar A, Desai KM, Wu L. Alagebrium attenuates acute methylglyoxal-induced
glucose intolerance inSprague-Dawley rats. Br J Pharmacol. 2010;159:166–175.
148. Hofmann SM, Dong HJ, Li Z, et al. Improved insulin sensitivity is associated with
restricted intake of dietary glycoxidation products in the db/db mouse. Diabetes.
2002;51:2082–2089.
149. Rodrigues T, Matafome P, Santos-Silva D, et al. Reduction of methylglyoxal-
induced glycation by pyridoxamine improves adipose tissue microvascular
lesions. J Diabetes Res. 2013;2013:1–9 .
150. Wei Y, Wang D, Moran G, et al. Fructose-induced stress signaling in the liver
involves methylglyoxal. Nutr Metab.10:32.
151. Gaens KHJ, Niessen PMG, Rensen SS, et al. Endogenous formation of Ne-(carbox-
ymethyl)lysine is increased in fatty livers and induces inflammatory markers in
an in vitro model of hepatic steatosis. J Hepatol. 2012;56:647–655.
152. Uribarri J, Cai W, Woodward M, et al. Elevated serum advanced glycation end
products in obese indicate risk for the metabolic syndrome: a link between
healthy andunhealthy obesity? J Clin Endocrino l Metab. 2015;100:1957–196 6.
153. Kong X, zhe MM, Huang K, et al. Increased plasma levels of the methylglyoxal in
patients with newly diagnosed type 2 diabetes. J Diabetes. 2014;6:535–540.
154. UribarriJ, Cai W, Ramdas M, et al. Restriction of advanced glycation end products
improves insulin resistance in human type 2 diabetes: potential role of AGER1
and SIRT1. Diabetes Care. 2011;34:1610–1616.
155. De Courten B, De Courten MPJ, Soldatos G, et al. Diet low in advanced glycation
end products increases insulin sensitivity in healthy overweight individuals: a
double-blind, randomized, crossover trial. Am J Clin Nutr. 2016;103:1426 –1433.
156. Mac
ıas-Cervantes MH, Rodr
ıguez-Soto JMD, Uribarri J, et al. Effect of an ad-
vanced glycation end product-restricted diet and exercise on metabolic parame-
ters in adult overweight men. Nutrition. 2015;31:446–451.
157. VlassaraH, Cai W, Tripp E, et al. Oral AGE restriction ameliorates insulin resistance
in obese individuals with the metabolic syndrome: a randomised controlled trial.
Diabetologia. 2016; 59:2181–2192.
10 Nutrition Reviews
V
R
Vol. 0(0):1–12
Downloaded from https://academic.oup.com/nutritionreviews/advance-article/doi/10.1093/nutrit/nuaa074/5909185 by Universidade Federal de Goi�s user on 21 September 2020
158. Jones ML, Buhimschi IA, Zhao G, et al. Acute glucose load, inflammation, oxida-
tive stress, nonenzymatic glycation, and screening for gestational diabetes.
Reprod Sci. 2019;193371911983177.
159. Bartakova V, Kollarova R, Kuricova K, et al. Serum carboxymethyl-lysine, a domi-
nant advanced glycationend product, is increased in womenwith gestational di-
abetes mellitus. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub.
2016;160:70–75.
160. Kellow NJ,Coughlan MT. Effect of diet-derived advanced glycation end products
on inflammation. Nutr Rev. 2015;73:737–759.
161. Villegas-Rodr
ıguez ME, Uribarri J, Solorio-Meza SE, et al. The AGE-RAGE axis and
its relationship to markers of cardiovascular disease in newly diagnosed diabetic
patients. PLoS One. 2016;11:E0159175.
162.
Sebekov
a K, Brouder
Sebekov
a K. Glycated proteins in nutrition: friend or foe?
Exp Gerontol. 2019;117:76–90.
163. Guosheng L, Hongmei S, Chuan N, et al. The relationship of serum AGE levels in
diabetic mothers with adversefetal outcome. J Perinatol. 2009;29:483–488.
164. Lappas M. Activation of inflammasomes in adipose tissue of women with gesta-
tional diabetes. Mol CellEndocrinol. 2014;382:74–83.
165. Konishi H,Nakatsuka M, Chekir C, et al.Advanced glycation end products induce
secretion of chemokines and apoptosis in human first trimester trophoblasts.
Hum Reprod. 2004;19:2156– 2162.
166. Hao L, Noguchi S, Kamada Y, et al. Adverse effects of advanced glycation end
products on embryonal development. Acta MedOkayama. 2008;62:93–99.
167. Haucke E, Santos AN, Simm A, et al. Accumulation of advanced glycation end
products in the rabbit blastocyst under maternal diabetes. Reproduction.
2014;148:169–178.
168. Kawaharada R, Masuda H, Chen Z, et al. Intrauterine hyperglycemia-induced in-
flammatory signalling via the receptor for advanced glycation end products in
the cardiac muscle of the infants of diabetic mother rats. Eur J Nutr.
2018;57:2701–2712.
169. Ejdesjo¨ A, Brings S, Fleming T, et al. Receptor for advanced glycation end prod-
ucts (RAGE) knockout reduces fetal dysmorphogenesis in murine diabetic preg-
nancy. Reprod Toxicol. 2016;62:62–70.
170. Tang X, Qin Q, Xie X, et al. Protective effect of sRAGE on fetal development in
pregnant rats with gestational diabetes mellitus. Cell Biochem Biophys.
2015;71:549–556.
171. Dale MTG, Magnus P, Leirgul E, et al. Intake of sucrose-sweetenedsoft beverages
during pregnancy and risk of congenital heart defects (CHD) in offspring: a
Norwegianpregnancy cohort study.Eur J Epidemiol. 2019;34:383–396.
172. World Health Organization. Global Strategy for Infant and Young Child Feeding.
Geneva: World Health Organization; 2003.
173. Vieira Borba V, Sharif K, Shoenfeld Y. Breastfeeding and autoimmunity: program-
ing health from the beginning. Am J Reprod Immunol. 2018;79:E12778.
174. Donovan SM, Odle J. Growth factors in milk as mediators of infant development.
Annu Rev Nutr. 1994;14:147–167.
175. ShehadehN, Shamir R, Berant M, et al. Insulin in human milk and the prevention
of type 1 diabetes.Pediatr Diabetes. 2001;2:175–177.
176. Plagemann A, Harder T, Schellong K, et al. Early postnatal life as a critical time
window for determination of long-term metabolic health. Best Pract Res Clin
Endocrinol Metab. 2012;26:641–653.
177. Knowles JA. Breast milk: a source of more than nutrition for the neonate. Clin
Toxicol. 1974;7:69–82.
178. Gomes RM, Bueno FG, Schamber CR, et al. Maternal diet-induced obesity during
suckling period programs offspring obese phenotype and hypothalamic leptin/
insulin resistance. J Nutr Biochem. 2018;61:24–32 .
179. Schwarzenbolz U, HofmannT, Sparmann N, et al. Free Maillard reaction products
in milk reflect nutritional intake of glycated proteins and can be used to distin-
guish “organic” and “conventionally” produced milk. J Agric Food Chem.
2016;64:5071–5078.
180. Francisco FA, Barella LF, SilveiraS da S, et al. Methylglyoxal treatmentin lactating
mothers leads to type 2 diabetes phenotype in male rat offspring at adulthood.
Eur J Nutr. 2018;57:477–486.
181. Uribarri J, Cai W, Sandu O, et al. Diet-derived advanced glycation end products
are major contributors to the body’s AGE pool and induce inflammation in
healthy subjects. Ann N Y Acad Sci. 2005;1043:461–466.
182. Uribarri J, Cai W, Peppa M, et al. Circulating glycotoxins and dietary advanced
glycation endproducts: two links to inflammatory response, oxidative stress, and
aging. J Gerontol A Biol Sci Med Sci. 2007;62:427–433.
183. Koschinsky T, He CJ, Mitsuhashi T, et al. Orally absorbed reactive glycation prod-
ucts (glycotoxins): an environmental risk factor in diabetic nephropathy. Proc
Natl Acad Sci USA. 1997;94:6474–6479.
184. Birlouez-Aragon I, Saavedra G, Tessier FJ,et al. A diet based on high-heat-treated
foods promotes risk factors for diabetes mellitus andcardiovascular diseases. Am
JClinNutr. 2010;91:1220–1226.
185. Mark AB, Poulsen MW, Andersen S, et al. Consumptionof a diet low in advanced
glycation end products for 4 weeks improves insulin sensitivity in overweight
women. Diabetes Care. 2014;37:88–95.
186. He C, Sabol J, Mitsuhashi T, et al. Dietary glycotoxins: inhibition of reactive prod-
ucts by aminoguanidine facilitates renal clearance and reduces tissue sequestra-
tion. Diabetes. 1999;48:1308–1315 .
187. Hellwig M, Geissler S, Peto A, et al. Transport of free and peptide-bound
pyrraline at intestinal and renal epithelial cells. J Agric Food Chem.
2009;57:6474–6480.
188. Hellwig M, Geissler S, Matthes R, et al. Transport of free and peptide-bound gly-
cated amino acids: synthesis, transepithelial flux at Caco-2 cell monolayers, and
interaction with apical membrane transport proteins. ChemBioChem.
2011;12:1270–1279.
189. DittrichR, Hoffmann I, Stahl P, et al. Concentrations of Ne-carboxymethyllysine in
human breast milk, infant formulas, and urine of infants. J Agric Food Chem.
2006;54:6924–6928.
190. Han L, Li L, Li B, et al. Review of the characteristics of food-derived and endoge-
nous Ne-carboxymethyllysine. JFoodProt. 2013;76 :912–918 .
191. O’Brien J, Morrissey PA, Ames JM. Nutritional and toxicological aspects of the
Maillard browning reactionin foods. Crit Rev Food Sci Nutr. 1989;28:211–248.
192. Zamora R, Hidalgo FJ. Coordinate contribution of lipid oxidation and Maillard re-
action to the nonenzymatic food browning. Crit Rev Food Sci Nutr.
2005;45:49–59.
193. Uribarri J, Woodruff S, Goodman S, et al. Advanced glycation end products in
foods and a practical guide to their reduction in the diet. JAmDietAssoc.
2010;110:911–916.e12.
194. ProsserCG, Carpenter EA, Hodgkinson AJ. N e-carboxymethyllysine in nutritional
milk formulas for infants. FoodChem. 2019; 274:886–890.
195.
Sebekov
a K, SaavedraG, Zumpe C, et al. Plasma concentrationand urinary excre-
tion of Ne- (carboxymethyl)lysine in breast milk- and formula-fed infants. Ann N
YAcadSci. 2008;1126:177–180.
196. Firmin S, Elmhiri G, Crepin D, et al. Formula derived Maillard reaction products in
post-weaning intrauterine growth-restricted piglets induce developmental pro-
gramming of hepatic oxidative stress independently of microRNA-21 and
microRNA-155. J Dev Orig HealthDis. 2018;9:566–572.
197. Elliott RB. Diabetes - a man made disease. Med Hypotheses. 2006;67:388 –391.
198. Klenovics KS, Boor P, Somoza V, et al. Advanced glycation end products in infant
formulas do not contribute to insulin resistance associated with their consump-
tion. PLoS One. 2013;8:E53056.
199. Renzone G, Arena S, Scaloni A. Proteomic characterization of intermediate and ad-
vanced glycation end-products in commercial milk samples. JProteomics.
2015;117:12–23.
200. Zhao D, Le TT, Larsen LB, et al. Effect of glycation derived from a-dicarbonyl com-
pounds on the in vitro digestibility of b-casein and b-lactoglobulin: A model study
with glyoxal, methylglyoxal and butanedione. Food Res Int. 2017;102:313–322.
201. Snelson M, Coughlan MT. Dietary advanced glycation end products: digestion,
metabolismand modulation of gut microbial ecology. Nutrients. 2019;11:215.
202. Qu W, Yuan X, Zhao J, et al. Dietary advanced glycation end products modify gut
microbial composition and partially increase colon permeability in rats. Mol Nutr
Food Res. 2017;61:1700118.
203. Seiquer I, Rubio LA, Peinado MJ, et al. Maillard reaction products modulate gut
microbiota compositionin adolescents. Mol Nutr FoodRes. 2014;58:1552–1560.
204. Yacoub R, Nugent M, Cai W, et al. Advanced glycation end products dietary re-
striction effects on bacterial gut microbiota in peritoneal dialysis patients; a ran-
domized openlabel controlled trial.PLoS One. 2017;12:E0184789.
205. Harada E, Syuto B. Precocious cessation of intestinal macromolecular transmis-
sion and sucrase development induced by insulin in adrenalectomized suckling
rat. Comp Biochem Physiol – Part A Physiol. 1991;99:327–331.
206. Peters MA, Hudson PM, Jurgelske W. The acute toxicity of methylglyoxal in rats:
the influence of age, sex, and pregnancy.Ecotoxicol Environ Saf. 1978;2: 369–374.
207. Toop CR, Muhlhausler BS, O’Dea K, et al. Impact of perinatal exposure to sucrose
or high fructose corn syrup (HFCS-55) on adiposity andhepatic lipid composition
in rat offspring. J Physiol. 2017;595:4379–4398.
208. Csongov
a M, Gureck
aR,Koborov
a I, et al. The effects of a maternal advanced
glycation end product-rich diet on somatic features, reflex ontogeny and meta-
bolic parameters of offspring mice. Food Funct. 2018;9:3432–3446.
209. Borg DJ, Yap FYT, Keshvari S, et al. Perinatal exposure to high dietary advanced
glycation end products in transgenic NOD8.3 mice leads to pancreatic beta cell
dysfunction. Islets. 2018;10:10–24.
210. Roest PAM, Molin DGM, Schalkwijk CG, et al. Specific local cardiovascular
changes of n e-(carboxymethyl) lysine, vascular endothelial growth factor, and
smad2 in the developing embryos coincide with maternal diabetes-induced con-
genital h eart defects. Diabetes. 200 9;58:1222–1228.
211. Chiavaroli V, D’AdamoE, Giannini C, et al. Serumlevels of receptors for advanced
glycation end products in normal-weight and obese children born small and
large for ge stational age. Diabetes Care. 2012;35:1361–1363.
212. Chandna AR, Kuhlmann N, Bryce CA, et al. Chronic maternal hyperglycemia in-
duced during mid-pregnancy in rats increases RAGE expression, augments hip-
pocampal excitability, and alters behavior of the offspring. Neuroscience.
2015;303:241–260.
Nutrition Reviews
V
R
Vol. 0(0):1–12 11
Downloaded from https://academic.oup.com/nutritionreviews/advance-article/doi/10.1093/nutrit/nuaa074/5909185 by Universidade Federal de Goi�s user on 21 September 2020
213. Yang G, Cancino GI, Zahr SK, et al. A Glo1-methylglyoxal pathway that is per-
turbed in maternal diabetes regulates embryonic and adult neural stem cell
pools in murine offspring. Cell Rep. 2016;17:1 022–1036 .
214. Shimizu I, Hiramatsu Y, Omori Y, et al. Comparison of HbA1c and glycated albu-
min as a control marker for newborn complications in diabetic women in a mul-
ticentre study in Japan (Japan Glycated Albumin Study Group: Study 2). Ann Clin
Biochem. 2018;55:639–646.
215. Sugawara D, Sato H, Ichihashi K, et al. Glycated albumin level during late preg-
nancy as a predictive factor for neonatal outcomes of women with diabetes. J
Matern Neonatal Med. 2018;31:2007–2012.
216. Feng Y, Yang H. Metformin–a potentially effective drug for gestational diabetes
mellitus: a systematic review and meta-analysis. JMaternNeonatalMed.
2017;30:1874–1881.
217. Kinsky OR, Hargraves TL, Anumol T, et al. Metformin scavenges methylglyoxal to
form a novel imidazolinone metabolite in humans. Chem Res Toxicol.
2016;29:227–234.
218. Foretz M, Guigas B, Bertrand L, et al. Metformin: from mechanisms of action to
therapies. Cell Metab. 2014;20:953–966.
Downloaded from https://academic.oup.com/nutritionreviews/advance-article/doi/10.1093/nutrit/nuaa074/5909185 by Universidade Federal de Goi�s user on 21 September 2020
... A high fructose intake is related to AGE accumulation in different tissues, which leads to insulin resistance and dyslipidemia [10]. Studies have reported that perinatal exposure to AGEs during pregnancy and lactation is one of the factors causing metabolic programming, increasing the risk of developing NCDs in adulthood [11]. ...
Article
Full-text available
Obesity and the intake of high-sugar diets have dramatically increased in recent decades. However, it is still uncertain how sugar intake during the critical development phase affects the long-term health of children. In this context, the Developmental Origins of Health and Disease (DOHaD) concept established a correlation between early life environment and the development of cardiometabolic diseases in adulthood. This review summarizes the current knowledge about the consequences of sugar intake during the critical development phase for the onset of non-communicable diseases (NCDs). We found evidence that increased sugar intake during pregnancy contributes to maternal obesity and many cardiometabolic dysfunctions in the offspring. Furthermore, dietary sugar during the suckling period provokes the obese phenotype in adulthood. Finally, high-sugar diet intake during childhood induces metabolic syndrome and depressive-like behavior.
... Maternal hyperglycaemia during pregnancy has been shown to increase MG and AGEs circulation levels, which induces premature hippocampal neurogenesis and depletion of neuronal cells in the prefrontal cortex [9], due to its passage from the placenta, contributing to behavioural alterations in early adulthood [7]. Additionally, MG treatment in lactating mothers contributes to offspring adiposity and overweight, glucose intolerance and impaired beta-cell function, increasing the susceptibility to develop metabolic syndrome at adulthood [13]. ...
Article
Aim: Lactation is an important programming window for metabolic disease and neuronal alterations later in life. We aimed to study the effect of maternal glycation during lactation on offspring neurodevelopment and behaviour, assessing possible sex differences and underpinning molecular players. Methods: Female Wistar rats were treated with the Glyoxalase-1 inhibitor S-p-Bromobenzylguthione cyclopentyl diester (BBGC 5 mg/kg). A control and vehicle group treated with dimethyl sulfoxide were considered. Male and female offspring were tested at infancy for neurodevelopment hallmarks. After weaning, triglycerides and total antioxidant capacity were measured in breast milk. At adolescence, offspring were tested for locomotor ability, anxious-like behaviour, and recognition memory. Metabolic parameters were assessed, and the hippocampus and prefrontal cortex were collected for molecular analysis. Key findings: Maternal glycation reduced triglycerides and total antioxidant capacity levels in breast milk. At infancy, both male and female offspring presented an anticipation on the achievement of neurodevelopmental milestones. At adolescence, male offspring exposed to maternal glycation presented hyperlocomotion, whereas offspring of both sexes presented a risk-taking phenotype, accompanied by GABAA receptor upregulation in the hippocampus. Females also demonstrated GABAA and PSD-95 changes in prefrontal cortex. Furthermore, lower levels of GLO1 and consequently higher accumulation of AGES were also observed in both male and female offspring hippocampus. Significance: Early exposure to maternal glycation induces changes in milk composition leading to neurodevelopment changes at infancy, and sex-specific behavioural and neurometabolic changes at adolescence, further evidencing that lactation period is a critical metabolic programming window and in sculpting behaviour.
Article
Full-text available
Background & Aims High hydrostatic pressure (HHP) processing is a non-thermal method proposed as an alternative to Holder pasteurization (HoP) for the treatment of human milk. HHP preserves numerous milk bioactive components that are degraded by HoP, but no data are available for milk oligosaccharides (HMOs) or the formation of Maillard reaction products, which may be deleterious for preterm newborns. Methods We evaluated the impact of HHP processing of human milk on 22 HMOs measured by liquid chromatography with fluorescence detection and on furosine, lactuloselysine, carboxymethyllysine (CML) and carboxyethyllysine (CEL) measured by liquid chromatography with tandem mass spectrometric detection (LC-MS/MS), four established indicators of the Maillard reaction. Human raw milk was sterilized by HoP (62.5°C for 30 min) or processed by HHP (350 MPa at 38°C). Results Neither HHP nor HoP processing affected the concentration of HMOs, but HoP significantly increased furosine, lactuloselysine, CML and CEL levels in milk. Conclusions Our findings demonstrate that HPP treatment preserves HMOs and avoids formation of Maillard reaction products. Our study confirms and extends previous findings that HHP treatment of human milk provides safe milk, with fewer detrimental effects on the biochemically active milk components than HoP.
Article
Background Prenatal stress is associated with increased susceptibility to psychiatric and metabolic disorders later in life. Prenatal exposure to stress mediators may have sex-dependent effects on offspring brain and metabolic function, promoting a sex-specific vulnerability to psychopathology and metabolic alterations at adulthood. In this work, the impact of prenatal stress on glucose homeostasis and peripheral metabolism of male and female offspring was investigated in a chronic anxiety animal model. Methods Pregnant Wistar rats were injected with saline or glucocorticoid (dexamethasone: 1 mg/kg, subcutaneous) at gestational days 18 and 19. Male and female offspring weight was monitored, and anxious-like behavior and peripheral insulin sensitive tissues were analyzed at adulthood. Results At birth, females and males prenatally exposed to stress presented decreased body weight which remained low in females. At adulthood a morphological disorganization of the Langerhans islets was observed in both sexes prenatally exposed to stress, yet not changes in insulin levels were detected. Also, prenatal stress increased glucose transporter 4 (GLUT-4) levels in female and male adipose tissue, and decreased insulin receptor levels in the liver and skeleton muscle but only in females. Conclusions Exposure to stress mediators in critical periods of development negatively affects behavior and metabolism. Prenatal stress programs offspring peripheral metabolism in a sex-specific manner, emphasizing that the response to stress in critical periods of development may be sex-specific having each sex different vulnerabilities to psychiatric and metabolic disorders. Considering sex-specificities may provide critical clues for the design of preventive strategies and for early therapeutic intervention.
Article
Full-text available
Nonalcoholic fatty liver disease (NAFLD) is caused by excessive liver lipid accumulation, but insulin resistance is specifically associated with impaired lipid saturation, oxidation, and storage (esterification), besides increased de novo lipogenesis. We hypothesized that dietary glycotoxins could impair hepatic lipid metabolism in obesity contributing to lipotoxicity-driven insulin resistance and thus to the onset of nonalcoholic steatohepatitis (NASH). In diet-induced obese rats with methylglyoxal-induced glycation, magnetic resonance spectroscopy, mass spectrometry, and gas chromatography were used to assess liver composition in fatty acyl chains and phospholipids. High-fat diet-induced obesity increased liver lipid fraction and suppressed de novo lipogenesis but did not change fatty acid esterification and saturation or insulin sensitivity. Despite a similar increase in total lipid fraction when supplementing the high-fat diet with dietary glycotoxins, impairment in the suppression of de novo lipogenesis and decreased fatty acid unsaturation and esterification were observed. Moreover, glycotoxins also decreased polyunsaturated cardiolipins and caused oxidative stress, portal inflammation, and insulin resistance in high-fat diet-induced obese rats. Dietary glycated products do not change total lipid levels in the liver of obese rats but dramatically modify the lipidemic profile, leading to oxidative stress, hepatic lipotoxicity, and insulin resistance in obesity and thus contribute to the onset of NASH.
Article
Full-text available
Gestational diabetes mellitus (GDM), an important public health problem that affects mothers and offspring, is a common metabolic disorder. We evaluated the effect of the pre-pregnancy Mediterranean diet (MD) level of exposure on the odds of GDM development. A case-control study (291 GDM cases and 1175 controls without GDM) was conducted in pregnant women. Pre-pregnancy dietary intake was assessed using a validated food frequency questionnaire to calculate an MD adherence index (range score 0–9: low ≤ 2; middle 3–4; high 5–6; very high ≥ 7). Adjusted odds ratios (aOR) and their 95% confidence intervals (CI) were estimated using multivariable logistic regression models including age, BMI, family history of diabetes mellitus, previous GDM, miscarriages, and gravidity. Overall, middle-high MD adherence was 216/291 (74.2%) and very high adherence was 17/291 (5.8%) in cases. In controls the corresponding figures were 900/1175 (76.6%) and 73/1175 (6.2%), respectively. Compared to low adherence, high MD adherence was associated with GDM reduction (aOR 0.61, 95% CI 0.39,0.94; p = 0.028), and very high MD adherence was even more strongly associated (aOR 0.33, 95% CI 0.15, 0.72; p = 0.005). The protective effect of adherence to the MD prior to pregnancy should be considered as a preventive tool against the development of GDM.
Article
Full-text available
Background/Objective Gestational diabetes mellitus (GDM) is a common pregnancy complication, with complex disease mechanisms, and several risk factors may contribute to its onset. We performed an umbrella review to summarize the evidence from meta-analyses of observational studies on risk factors associated with GDM, evaluate whether there are indications of biases in this literature and identify which of the previously reported associations are supported by convincing evidence. Methods We searched PubMed and ISI Web of Science from inception to December 2018 to identify meta-analyses examining associations between putative risk factors for GDM. For each meta-analysis we estimated the summary effect size, the 95% confidence interval, the 95% prediction interval, the between-study heterogeneity, evidence of small-study effects, and evidence of excess-significance bias. Results Thirty eligible meta-analyses were identified, providing data on 61 associations. Fifty (82%) associations had nominally statistically significant findings (P<0.05), while only 15 (25%) were significant at P<10⁻⁶ under the random-effects model. Only four risk factors presented convincing evidence:, low vs. normal BMI (cohort studies), BMI ~30–35 kg/m2 vs. normal BMI, BMI >35 kg/m2 vs. normal BMI, and hypothyroidism. Conclusions The compilation of results from synthesis of observational studies suggests that increased BMI and hypothyroidism show the strongest consistent evidence for an association with GDM. Diet and lifestyle modifications in pregnancy should be tested in large randomized trials. Our findings suggest that women with known thyroid disease may be offered screening for GDM earlier in pregnancy.
Article
Full-text available
The objective of this study was to assess the relationship between consumption of different types of carbohydrates (CHO) during pregnancy and the risk of having a small for gestational age (SGA) newborn. A retrospective matched case–control design was carried out with a total of 518 mother-offspring pairs. A total of 137 validated items were included in the food frequency questionnaire (FFQ). Conditional logistic regression models were used to calculate crude odds ratios (cORs) and adjusted odds ratios (aORs) with 95% confidence intervals (CIs). Having more than 75 g/day of brown bread showed an inverse association with SGA (aOR = 0.64, CI 0.43–0.96). In contrast, an intake of industrial sweets more than once a day (aOR = 2.70, CI 1.42–5.13), or even 2–6 times a week (aOR = 1.84, CI 1.20–2.82), increased the odds of having a SGA newborn. During pregnancy, the higher the increase of wholegrain cereal and bread, the lower the possibility of having a SGA newborn, but the opposite occurred with refined sugar products—just consuming industrial bakery products or pastries twice a week increased the odds of having an SGA infant. Case–control studies cannot verify causality and only show associations, which may reflect residual confusion due to the presence of unknown factors. It is possible that a high consumption of sugary foods is a marker of a generally poor lifestyle.
Article
Full-text available
Type 2 diabetes (T2D) is commonly regarded as a disease originating from lifestyle-related factors and typically occurring after the age of 40. There is, however, consistent experimental and epidemiological data evidencing that the risk for developing T2D may largely depend on conditions early in life. In particular, intrauterine growth restriction (IUGR) induced by poor or unbalanced nutrient intake can impair fetal growth and also cause fetal adipose tissue and pancreatic β -cell dysfunction. On account of these processes, persisting adaptive changes can occur in the glucose-insulin metabolism. These changes can include reduced ability for insulin secretion and insulin resistance, and they may result in an improved capacity to store fat, thereby predisposing to the development of T2D and obesity in adulthood. Accumulating research findings indicate that epigenetic regulation of gene expression plays a critical role in linking prenatal malnutrition to the risk of later-life metabolic disorders including T2D. In animal models of IUGR, changes in both DNA methylation and expression levels of key metabolic genes were repeatedly found which persisted until adulthood. The causal link between epigenetic disturbances during development and the risk for T2D was also confirmed in several human studies. In this review, the conceptual models and empirical data are summarized and discussed regarding the contribution of epigenetic mechanisms in developmental nutritional programming of T2D.
Article
Full-text available
Background Cardiovascular complications of pregnancy present an opportunity to assess risk for subsequent cardiovascular disease. We sought to determine whether peripartum cardiomyopathy and hypertensive disorder of pregnancy subtypes predict future myocardial infarction, heart failure or stroke independent of one another and of other risks such as gestational diabetes, preterm birth and intrauterine growth restriction. Methods and results The California Healthcare Cost and Utilization Project database was used to identify all hospitalised pregnancies from 2005 to 2009, with follow-up through 2011, for a retrospective cohort study. Pregnancies, exposures, covariates and outcomes were defined by International Classification of Diseases, Ninth Revision codes. Among 1.6 million pregnancies (mean age 28 years; median follow-up time to event excluding censoring 2.7 years), 558 cases of peripartum cardiomyopathy, 123 603 hypertensive disorders of pregnancy, 107 636 cases of gestational diabetes, 116 768 preterm births and 23 504 cases of intrauterine growth restriction were observed. Using multivariable Cox proportional hazards models, peripartum cardiomyopathy was independently associated with a 39.2-fold increase in heart failure (95% CI 30.0 to 51.9), resulting in ~1 additional hospitalisation per 1000 person-years. There was a 13.0-fold increase in myocardial infarction (95% CI 4.1 to 40.9) and a 7.7-fold increase in stroke (95% CI 2.4 to 24.0). Hypertensive disorders of pregnancy were associated with 1.4-fold (95% CI 1.0 to 2.0) to 7.6-fold (95% CI 5.4 to 10.7) higher risk of myocardial infarction, heart failure and stroke, resulting in a maximum of ~1 additional event per 1000 person-years. Gestational diabetes, preterm birth and intrauterine growth restriction had more modest associations. Conclusion These findings support close monitoring of women with cardiovascular pregnancy complications for prevention of early cardiovascular events and study of mechanisms underlying their development.
Article
Full-text available
The formation of advanced glycation end products (AGEs) in foods is accelerated with heat treatment, particularly within foods that are cooked at high temperatures for long periods of time using dry heat. The modern processed diet is replete with AGEs, and excessive AGE consumption is thought to be associated with a number of negative health effects. Many dietary AGEs have high molecular weight and are not absorbed in the intestine, and instead pass through to the colon, where they are available for metabolism by the colonic bacteria. Recent studies have been conducted to explore the effects of AGEs on the composition of the gut microbiota as well as the production of beneficial microbial metabolites, in particular, short-chain fatty acids. However, there is conflicting evidence regarding the impact of dietary AGEs on gut microbiota reshaping, which may be due, in part, to the formation of alternate compounds during the thermal treatment of foods. This review summarises the current evidence regarding dietary sources of AGEs, their gastrointestinal absorption and role in gut microbiota reshaping, provides a brief overview of the health implications of dietary AGEs and highlights knowledge gaps and avenues for future study.
Article
Full-text available
Studies report increased risk of congenital heart defects (CHD) in the offspring of mothers with diabetes, where high blood glucose levels might confer the risk. We explored the association between intake of sucrose-sweetened soft beverages during pregnancy and risk of CHD. Prospective cohort data with 88,514 pregnant women participating in the Norwegian Mother and Child Cohort Study was linked with information on infant CHD diagnoses from national health registers and the Cardiovascular Diseases in Norway Project. Risk ratios were estimated by fitting generalized linear models and generalized additive models. The prevalence of children with CHD was 12/1000 in this cohort (1049/88,514). Among these, 201 had severe and 848 had non-severe CHD (patent ductus arteriosus; valvular pulmonary stenosis; ventricular septal defect; atrial septal defect). Only non-severe CHD was associated with sucrose-sweetened soft beverages. The adjusted risk ratios (aRR) for non-severe CHD was 1.30 (95% CI 1.07–1.58) for women who consumed 25–70 ml/day and 1.27 (95% CI 1.06–1.52) for women who consumed ≥ 70 ml/day when compared to those drinking ≤ 25 ml/day. Dose–response analyses revealed an association between the risk of non-severe CHD and the increasing exposure to sucrose-sweetened soft beverages, especially for septal defects with aRR = 1.26 (95% CI 1.07–1.47) per tenfold increase in daily intake dose. The findings persisted after adjustment for maternal diabetes or after excluding mothers with diabetes (n = 19). Fruit juices, cordial beverages and artificial sweeteners showed no associations with CHD. The findings suggest that sucrose-sweetened soft beverages may affect the CHD risk in offspring.
Article
Background and aims: Previous studies suggest that psychological distress during pregnancy may lead to fetal developmental adaptations, which programme cardio-metabolic disease of the offspring. We examined the associations of maternal overall psychological distress, depression and anxiety during pregnancy with cardio-metabolic risk factors in 10-year-old children and explore potential sex-specific differences. Methods and results: In a population-based prospective cohort study among 4,088 mothers and their children, information about overall psychological distress, including depression and anxiety was obtained through the Brief Symptom Inventory during pregnancy. We measured child blood pressure and heart rate and insulin, glucose, serum lipids and C-reactive protein blood concentrations at 10 years. Analyses were performed in the total group and in boys and girls separately. Psychological distress during pregnancy was associated with higher childhood heart rate among boys only (differences 0.34 (95% Confidence Interval (CI) 0.18, 0.50) standard deviation scores (SDS), 0.22 (95% CI 0.06, 0.38) SDS, 0.33 (95% CI 0.19, 0.48) SDS, for overall psychological distress, depression and anxiety, respectively). Maternal anxiety during pregnancy was associated with higher childhood triglycerides among girls (difference 0.35 (95% CI 0.17, 0.53) SDS). Maternal psychological distress was not associated with childhood blood pressure, cholesterol, insulin, glucose and C-reactive protein concentrations. Conclusions: Maternal psychological distress may influence their offspring heart rate and triglycerides concentrations. Further studies are needed to replicate these findings and assess the long-term cardio-metabolic consequences of maternal psychological distress.
Article
Aims:: To investigate if oral glucose tolerance test (OGTT) associates with changes in maternal symptoms (ie, flushing, sweating), blood nonenzymatic advanced glycation end products (AGE), acute-phase reactive inflammatory markers, and oxidative stress. Methods:: Prospective case-control study of patients screened for gestational diabetes mellitus (GDM). One hundred nonfasting, second-trimester consecutive pregnant women allocated to either 50 g OGTT or water. Five women who had a 3-hour fasting 100 g OGTT also enrolled. Maternal serum glucose, AGE, soluble receptor for AGE (sRAGE), interleukin (IL)-6, and C-reactive protein (CRP) were immunoassayed. Total radical-trapping antioxidant parameter (TRAP) estimated with antioxidant capacity-peroxyl assay. Data corrected for gestational age and maternal body mass index. Results:: During 50 g OGTT there was a decrease in systolic blood pressure not accompanied by the onset of adverse clinical symptoms. There was a decrease in serum glucose levels 1 hour after water ( P = .019) but not glucose ingestion. Serum CRP ( P = .001) but not IL-6 was increased. The AGE, sRAGE, and TRAP levels remained unchanged. Similar results were seen during 100 g OGTT, except serum glucose was significantly elevated after 1 hour. Conclusion:: Results suggest screening tools for gestational diabetes are safe and clinically well tolerated during pregnancy. Clinical Trial Registration: ClinicalTrials.gov NCT03029546.