Nothing Special   »   [go: up one dir, main page]

Prenatal Stress Promotes Insulin Resistance Without Inflammation or Obesity in C57BL/6J Male Mice

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Stress

The International Journal on the Biology of Stress

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ists20

Prenatal stress promotes insulin resistance


without inflammation or obesity in C57BL/6J male
mice

Sofia Quiroga, Yamila Raquel Juárez, María Paula Marcone, María Agustina
Vidal, Ana María Genaro & Adriana Laura Burgueño

To cite this article: Sofia Quiroga, Yamila Raquel Juárez, María Paula Marcone, María
Agustina Vidal, Ana María Genaro & Adriana Laura Burgueño (2021): Prenatal stress promotes
insulin resistance without inflammation or obesity in C57BL/6J male mice, Stress, DOI:
10.1080/10253890.2021.1978425

To link to this article: https://doi.org/10.1080/10253890.2021.1978425

Published online: 28 Sep 2021.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ists20
STRESS
https://doi.org/10.1080/10253890.2021.1978425

RESEARCH ARTICLE

Prenatal stress promotes insulin resistance without inflammation or obesity in


C57BL/6J male mice
Sofia Quiroga,, Yamila Raquel Juarez,, Marıa Paula Marcone, Marıa Agustina Vidal, Ana Marıa Genaro and
Adriana Laura Burguen ~o
Instituto de Investigaciones Biomedicas, Consejo Nacional de Investigaciones Cientıficas y Tecnicas (CONICET)–Pontificia Universidad Catolica
Argentina, Buenos Aires, Argentina

ABSTRACT ARTICLE HISTORY


During gestation, stress exposure increases the risk of developing cognitive and physiological altera- Received 3 May 2021
tions in either the long or short term. Among them, metabolic alterations have been described. Accepted 4 September 2021
Adipose tissue is responsible for the secretion of several factors involved in controlling body weight
KEYWORDS
and energy expenditure, the regulation of insulin sensitivity, and the development of inflammation,
Prenatal stress; gene
among others. Moreover, the liver regulates glucose homeostasis and lipid metabolism, playing an expression; liver; adipose
essential role in developing insulin resistance. In this work, we analyzed if prenatal stress leads to alter- tissue; insulin resistance;
ations in metabolism and the relationship between these alterations and gene expression in the adi- fetal programming
pose tissue and the liver. Prenatal stress-exposed animals developed disturbances in the glucose and
insulin response curve, showing in both tests higher glycemia than the control group. However, they
did not exhibit increased body weight. At the same time, in the adipose tissue, we observed an
increase in mRNA expression of Leptin and Resistin and a decrease in Adiponectin. In the liver, we
observed a lower mRNA expression of several genes involved in glucose metabolism and fatty acid oxi-
dation, such as Sirt1, Pgc1a, Ppara, among others. In both tissues, we observed a lower expression of
inflammatory genes. These results suggest that prenatal stress exposure produces insulin resistance at
both physiological and molecular levels without pro-inflammatory signaling or obesity.

Introduction offspring’s metabolic health have been reported (Cao-Lei et


al., 2020).
Maternal health during pregnancy plays a crucial role in
Adipose tissue is an endocrine organ involved in meta-
shaping the health of the offspring. The developmental ori-
bolic regulation and inflammatory events by releasing several
gins of health and disease hypothesis suggests a specific
adipokines (Greenberg and Obin, 2006). Inflammation and
challenge in early life could alter an individual’s develop- dysregulation of its endocrine function induce low-grade sys-
ment, leading to adverse metabolic outcomes later in life temic inflammation and insulin resistance (Hotamisligil et al.,
(Zambrano et al., 2016). Epidemiological studies showed an 1993). Besides, it is responsible for modulating the function-
association between intrauterine disorders and the increased ing and the metabolism of the brain, liver, muscles, and car-
incidence of type 2 diabetes, obesity (Reynolds, 2010), and diovascular system (Li et al., 2013).
hypertension in adulthood (Yu et al., 2018). The main adipokines, such as Adiponectin, Leptin, and
Nowadays, stress seems a risk we are exposed daily and it Resistin, are closely related to obesity and IR development.
is a prognostic factor for the early onset of complex and Adiponectin improves insulin sensitivity and has an anti-
common age-related diseases (Kivim€aki and Steptoe, 2018). inflammatory effect. Circulating Adiponectin levels are nega-
Stress is a threat to physiological and/or psychological tively associated with obesity, IR, type 2 diabetes, and meta-
homeostasis. Exposure to prenatal stress (PS) increases the bolic syndrome (Recinella et al., 2020). Leptin regulates food
risk of short- and long-term adverse cognitive and physio- intake and energy expenditure and upregulates inflammatory
logical health outcomes (Entringer et al., 2015). Several stud- molecules such as Tumor necrosis factor-alpha (Tnf-a) and
ies have shown that PS is associated with a significant Interleukin 6 (IL-6) associated with the development of IR
decrease in birth weight (Drago et al., 1999; Lesage et al., and type 2 diabetes (Recinella et al., 2020). At the same time,
2004). In recent years, it appears that rapid catch-up growth it has been suggested in several rodent models that Resistin
may be considered a risk factor for developing cardiovascular induces IR (Recinella et al., 2020).
disease and its associated phenotypes (Berends et al., 2013; The liver plays a central role in controlling glucose homeo-
Kelishadi et al., 2015). In rodents, different effects on the stasis, regulating several pathways of glucose metabolism

CONTACT Adriana Laura Burguen ~o alburgueno@conicet.gov.ar Instituto de Investigaciones Biomedicas, Consejo Nacional de Investigaciones Cientıficas y
Tecnicas (CONICET)–Pontificia Universidad Cat
olica Argentina, Alicia Moreau de Justo 1600, Piso 3, Buenos Aires C1107AFF, Argentina
These authors contributed equally to this work.
ß 2021 Informa UK Limited, trading as Taylor & Francis Group
2 S. QUIROGA ET AL.

(Han et al., 2016). In the liver, several critical genes in regulat- On postnatal day (PND) 5, pups were culled to six per
ing lipid and energy metabolism (such as Pgc1a, Ppara and mother, when possible. Litters with less than 6 offspring
Sirt1) were impaired in a range of metabolic diseases such as were removed from the study and data from these litters
obesity and hepatic steatosis, leading to the development of were only used to estimate the number of offspring born,
IR (Canto  and Auwerx, 2009; Yang et al., 2021; Zhou et sex and mortality ratios. After weaning (at PND 21), same-sex
al., 2018). mice were housed in groups of four per cage. For this work,
The aim of this work was to study whether PS induces we only used male mice. Only one pup from the same litter
metabolic disturbances in adulthood, such as obesity, hyper- was used per group. The remaining animals were used in
glycemia and/or IR. In addition, to determine if these distur- other experiments. Body weight was recorded weekly
bances were related to changes in the gene expression throughout the rest of the study.
profile of adipokines and inflammatory cytokines in two of
the most critical tissues in glucose metabolism, the adipose Intraperitoneal glucose tolerance test
tissue and the liver.
Animals at 24 weeks of age were fasted for 6 h (McGuinness
et al., 2009) (10:00 AM-04:00 PM), and basal blood glucose
Materials and methods levels were measured by tail nick (time point 0) using a com-
mercial glucometer (OneTouch UltraMini, LifeScan Inc,
Animals
Johnson & Johnson). Subsequently, a 2 g/kg body weight
Female and male C57BL/6J mice (twelve weeks old) bred in solution of glucose (Sigma, St. Louis, MO, USA) was adminis-
our Institute were housed on a 12-h light-dark cycle under tered intraperitoneally, and glycemia was measured at 15, 30,
controlled temperature (21 ± 2  C) with ad-libitum access to 60, and 120 min after the injection.
food and water. All animal procedures were approved by the
Institutional Animal Care and Use Committee at the
Intraperitoneal insulin sensitivity test
Biomedical Research Institute (BIOMED, N 007/2016) and
were compliant with NIH Guidelines for the Care and Use of Mice underwent an insulin sensitivity test at 25 weeks old.
Laboratory Animals. Since we have previously observed that the administration of
insulin to C57BL/6J fasting animals produces severe hypogly-
cemia and, given that this risks the animal’s life, we per-
Stress protocol formed the test without fasting.
90 days-old female mice were mated with males for two days Animals were intraperitoneally injected with 1 UI/kg body
(ratio 2:1, n ¼ 12 females per group). At gestational day (GD) weight of human recombinant insulin (Insuman R, 100UI/ml,
14, pregnant females were randomly divided into two Sanofi-Aventis Argentina SA). Blood glucose concentration
groups: (1) non-Prenatal Stress (NPS, n ¼ 10) or (2) Prenatal was measured at 0, 15, 30, 60, and 120 minutes by tail nick,
Stress (PS, n ¼ 10). The control dams (NPS) were left undis- using a commercial glucometer (OneTouch UltraMini,
LifeScan Inc, Johnson & Johnson, Malvern, PA, USA).
turbed during their pregnancy, while the experimental group
(PS) were exposed to restraint stress. In this case, restraint
stress consisted in placing the dams in a ventilated plastic Tissue collection
cylindrical tube (4 cm diameter, 10 cm long) for 2 h daily
(from 10:00 AM to 12:00 PM) from GD 14 until delivery (GD At 28 weeks of age, body weight was recorded, and animals
were anesthetized in a CO2 chamber before were euthanized
20–21), as previously described (Juarez et al., 2020). This
following retro-orbital bleeding. For biochemical analysis,
stressor was selected because the mother’s stress can indir-
plasma was collected. Retroperitoneal adipose tissue and liver
ectly affect the fetus (Mairesse et al., 2007; Ward and Weisz,
were dissected, weighed, and stored at 80  C until further
1984). This protocol has shown to significantly affect the car-
processing. To avoid any influence of hormonal fluctuations,
diovascular system (Igosheva et al., 2004), metabolism (Juarez
animals were sacrificed between 11 AM and 1 PM (ZT 4–6)
et al., 2020), and neuroendocrine stress reactivity in adult off-
(de Boer and Van der Gugten, 1987; Raghow, 2018; Tsang et
spring (Sternberg and Ridgway, 2003). The protocol used did al., 2016).
not affect the number of offspring born, the male-female
ratio, or the mortality rate (Table 1).
Plasma metabolic parameters
Table 1. Litter parameters analyzed on 10 litters of each group.
Plasma levels of total cholesterol, triglycerides, and insulin
Treatment NPS PS p
were measured. Blood samples were centrifuged at 4  C, and
N of litters 10 10
Litter size mean 6.6 ± 0.3 6.2 ± 0.4 NS plasma was stored at 80  C until processing. Total choles-
Male/Female Ratio 33/32 31/30 NS terol was quantified using Colestat enzimatico Reagent
Mortality ratio (dead/total) 1/65 1/61 NS (Wiener Lab, Rosario, Argentina) and triglycerides levels using
Litter size was analyzed using a t-test, male/female ratio, and mortality ratio TG Color Reagent (Wiener Lab, Rosario, Argentina). Plasma
with a v2 test (with Yates correction for mortality ratio). One of the litters in
the control group contained less than 6 offspring, as well as two litters in insulin concentration was determined by an enzyme-linked
the stress group; therefore, they were discarded at weaning. immunosorbent assay (Mercodia Mouse Insulin ELISA,
STRESS 3

Uppsala, Sweden) following the manufacturer’s protocol. The using the Kolmogorov-Smirnov test and Levene’s test,
detection limit was 0.2 ug/L with a coefficient of variability respectively. In case the assumptions were not met, the data
of 3%. were transformed as appropriate. Differences between groups
were analyzed using one-way ANOVA. When the time factor
was also involved, repeated-measures ANOVA was used. If
Quantitative assessment of mRNA expression by real- the transformed data did not comply with normality and/or
time polymerase chain reaction homogeneity of variance, the non-parametric Mann-Whitney
Total RNA was isolated from adipose tissue and liver using U test was used. In this case, the data were expressed as a
Transzol, according to the manufacturer’s instructions median and interquartile range. When significant interactions
(Transgenbiotech, Beijing, China). cDNA was synthesized were obtained, planned comparisons were used. Significance
using 3 lg of mRNA as a template using the Moloney Murine was determined at p < .05.
Leukemia Virus Reverse Transcriptase (M-MLV) with deficient
RNase H activity (Transgenbiotech, Beijing, China) and Results
oligo(dT) primers according to manufacturer’s protocol. PCRs
were performed in a 7500 Real-Time PCR System (Applied Prenatal stress does not change body, fat, or
Biosystems, Foster City, CA, USA) using FastStart Universal liver weight
SYBR Green Master (Rox) (Roche Applied Science, Mannheim, As shown in Figure 1(A), body weight increased during the
Germany). All reactions were run in duplicate. The cycling experiment with a significant interaction between prenatal
protocol featured an initial denaturation step of 5 min at treatment and time (F(24,360) ¼ 1.88, p ¼ .008). Using
95  C with 40 cycles of denaturation for 10 s at 95  C, fol- planned comparisons, we observed that prenatal treatment
lowed by annealing for 15 s at 58–62  C (according to each does not affect body weight (p ¼ .08). Retroperitoneal adi-
primer), and extension for 20 s at 72  C. The gene expression pose tissue weight/body weight (AT/BW, Figure 1(B)) and
levels were normalized using glyceraldehyde-3-phosphate liver weight/body weight (LW/BW, Figure 1(C)) ratios analysis
dehydrogenase (Gapdh) mRNA as an internal control. Gapdh showed no significant influence of PS (AT/BW, F(1,15) ¼
was found to be the most stable reference gene for testing 2.35 NS and LW/BW, F(1,15) ¼ 0.27 NS).
adipose tissue and liver mRNA expression among other
housekeeping genes tested before starting the experiment
(b-actin, cyclophilin B, and b2-microglobulin). Primer sequen- Prenatal stress produces insulin resistance without
ces are summarized in Table 2. changes in plasmatic insulin and triglyceridemia
In the glucose tolerance test (Figure 2(A)), it was detected a sig-
Statistical analysis nificant interaction between time and prenatal treatment
(F(4,60) ¼ 7.28 p ¼ .00008). Planned comparisons showed higher
Values were expressed as the mean ± standard error of the glucose values in PS animals at 15, 30, and 60 minutes post-glu-
mean (SEM) for each group. Statistical analyses were con- cose administration (15 min: p ¼ .02, 30 min: p ¼ .0002 and
ducted using STATISTICA 7.0 software (StatSoft, Inc., Tulsa, 60 min: p ¼ .00002). In the insulin tolerance test, it was observed
Oklahoma, USA). To analyze litter size, we used a t-test, and a significant interaction between time and prenatal treatment
for other litter parameters v2 test was used. The normality (Figure 2(B), F(3,39) ¼ 4.79 p ¼ .006). Planned comparisons
and homogeneity of variance for the dataset were tested showed higher blood glucose levels in PS animals at basal, 15-,

Table 2. List of primer sequences for quantitative assessment of mRNA expression.


Gene Sequence Amplicon length (bp) Annealing T
Sirt1 Fw: 50 -GCAGGTTGCAGGAATCCAAA-30 176 62
Rv: 50 -CTGGCTTCATGATGGCAAGTG-30
Sirt3 Fw: 50 -GCGTTGTGAAACCCGACATT-30 170 62
Rv: 50 -CAGTCGGGGCACTGATTTCT-30
Socs3 Fw: 50 -CTCTTACGACCGCTGTCTCTC-30 292 60
Rv: 50 -CATCCCGGGGAGCTAGT-30
Ppar-a Fw: 50 -TGTGGCTGGTCAAGTTCGG-30 220 58
Rv: 50 -CCAGAGCTCTCCTCACCGAT-30
Pgc1-a Fw: 50 -CGGAAATCATATCCAACCAGTACA-30 93 60
Rv: 50 -TGAGAACCGCTAGCAAGTTTG-30
Adiponectin Fw: 50 -GATGGCAGAGATGGCACTCC-30 173 60
Rv: 50 -GAGCGATACACATAAGCGGC-30
Resistin Fw: 50 -TGTCCTGCTAAGTCCTCTGC-30 256 60
Rv: 50 -CAAGACTGCTGTGCCTTCTG-30
Leptin Fw: 50 -AGCTGCAAGGTGCAAGAAGA-30 193 60
Rv: 50 -GGATACCGACGTGTGTGAAATG-30
Tnf-a Fw: 50 -GGGTGATCGGTCCCCAAAG-30 159 60
Rv: 50 -TGAGATCCATGCCGTTGGC-30
IL-1b Fw: 50 -GCCACCTTTTGACAGTGATG-30 165 58
Rv: 50 -AGTGATACTGCCTGCCTGAA-30
Gapdh Fw: 50 -CGTCCCGTAGACAAAATGGT-30 177 60
Rv: 50 -GAATTTGCCGTGAGTGGAGT-30
4 S. QUIROGA ET AL.

U Test, U ¼ 12, p ¼ .02) and Resistin (F(1,15) ¼ 12.92, p ¼ .003)


along with a reduction in Adiponectin (F(1,15) ¼ 20.62,
p ¼ .0004) due to PS exposure.
Although the role of Sirt1 in the expression of
Adiponectin is still controversial (Liu and Liu, 2009), we
decided to analyze Sirt1 mRNA expression in this model. We
found a decrease in Sirt1 mRNA expression after PS exposure
(F(1,15) ¼ 23.81 p ¼ .0002). However, we did not detect dif-
ferences in the expression of Pparc, a well-known positive
regulator of Adiponectin (Maeda et al., 2001) (data
not shown).
Due to the critical role of inflammation in the progression
of IR (Rehman and Akash, 2016), we evaluated the mRNA
expression of the two major inflammatory molecules
Interleukin-1 beta (IL-1b) and Tnf-a (Rehman and Akash,
2016). We observed that both have a lower expression in the
retroperitoneal adipose tissue of PS animals (IL-1b: Mann-
Whitney U Test U ¼ 3, p ¼ .0006 and Tnf-a: F(1,15) ¼
6.89, p ¼ .02).

Prenatal stress triggers alterations in liver gene


expression related to insulin resistance
We analyzed liver mRNA gene expression (Figure 4) of Pgc1a
since it was associated with several inflammatory and meta-
bolic diseases (Rius-Perez et al., 2020). We found a decrease
in mRNA expression of Pgc1a (F(1,15) ¼ 7.09, p ¼ .02). In add-
ition, we studied the mRNA expression of some genes related
to Pgc1a. Ppara interacts with Pgc1a promoting mitochon-
drial fatty acid oxidation (Vega et al., 2000). We found a
decrease in Ppara mRNA expression in animals under PS
(F(1,14) ¼ 4.93, p ¼ .04). As Sirt1 activates Pgc1a (Nogueiras
et al., 2012), we analyzed Sirt1 gene expression and observed
a dowregulation by PS (F(1,15) ¼ 14.78, p ¼ .002).
Furthermore, we found a positive correlation between Pgc1a
and Sirt1 mRNA expression (Spearman R ¼ 0.58, p ¼ .01, data
not shown).
Sirt3 has been shown to play an important role in pre-
venting metabolic syndrome and was downregulated in dia-
Figure 1. Body weight curve (A), relative visceral adipose tissue content (B) and betes and other metabolic diseases (Pinteric et al., 2020). In
relative liver weight (C). A. Body weight was recorded weekly from week 4 to PS, we found a down-regulation in Sirt3 (F(1,14) ¼
28. Non-prenatally stressed (NPS, dark circles) and prenatally stressed (PS, light
circles). B: visceral adipose tissue weight/body weight ratio (%) and C: liver tis- 7.87, p ¼ .01).
sue weight/body weight ratio (%). NPS (dark bars) and PS (white bars). All val- In the liver, deletion of Socs3 improves IR in mice (Torisu
ues are presented as mean ± standard error (n ¼ 8–9 mice in each group). et al., 2007). We observed a reduction in mRNA expression of
Socs3 (F(1,14) ¼ 8.00, p ¼ .01). Moreover, we found a positive
correlation between Socs3 and Sirt1 mRNA expression
30-, and 60-min post-insulin administration (basal: p ¼ .003, (Spearman R ¼ 0.63, p ¼ .006, data not shown).
15 min: p ¼ .00003, 30 min: p ¼ .0003 and 60 min: p ¼ .0001).
PS did not alter non-fasting insulin levels (Figure 2(C); F(1,8)
¼ 0.01, NS), triglyceridemia (Figure 2(D), F(1,15) ¼ 0.18 NS) or Discussion
total cholesterol (Figure 2(E); Mann-Whitney U Test, U ¼ 23, NS).
Injuries suffered during pregnancy can result in long-term
consequences for the offspring’s future health (Entringer et
Prenatal stress exposure leads to altered mRNA al., 2012). In the present work, we describe for the first time
the development of IR induced by PS exposure without the
expression of adipokines, without evidence of
presence of inflammation or obesity. C57BL/6J male mice
inflammation in retroperitoneal adipose tissue
showed alterations in the glucose tolerance test and the
The mRNA expression in retroperitoneal adipose tissue insulin sensitivity curve. In the adipose tissue, it was associ-
(Figure 3) showed increased levels of Leptin (Mann-Whitney ated with an imbalance in adipokine gene expression,
STRESS 5

Figure 2. (A) Glucose tolerance test: Blood glucose measured during the glucose tolerance curve. (B) Insulin tolerance test: Blood glucose measured during the insu-
lin sensitivity test for non-prenatally stressed (NPS, dark circles), prenatally stressed (PS, light circles). (C) Non-fasting insulin levels. (D) Triglycerides. (E) total choles-
terol. Panels A–D: values are presented as mean ± standard error (for A–B and D: n ¼ 8–9 mice in each group; for C: n ¼ 5 mice in each group), Panel E: values are
presented as median and interquartile range (n ¼ 8–9 mice in each group). NPS: non-prenatal stress (dark bars); PS: prenatal stress (white bars). p < .05, p < .01
and  p < .001.

together with a decrease in inflammatory genes. However, and the insulin measurement at sacrifice on non-fasting ani-
we observed a decline in the mRNA levels of genes related mals. We found no changes in plasmatic insulin due to PS.
to glucose metabolism and fatty acid oxidation in the liver. These results suggest IR presence, probably at an early stage,
as we do not found alterations in insulin values. Similarly,
other authors reported IR in males exposed to PS using a
Physiological parameters
sleep fragmentation stress model in C57BL/6J mice (Khalyfa
Body weight, adiposity, and liver weight showed no signifi- et al., 2015) with higher glycemia on the glucose tolerance
cant variation in animals exposed to PS (compared to non- test and insulin sensitivity curve, but with higher insulin lev-
prenatally stressed animals). Recently, we reported that PS els. Contradictory results are published: we and others have
does not increase body weight or adiposity in adult BALB/c previously reported no difference in the glucose tolerance
male mice (Juarez et al., 2020). Similar results occurred in test, insulin sensitivity curve, or plasmatic insulin levels in
male Sprague Dawley rats exposed to PS using a variable BALB/c mice (Juarez et al., 2020; Luft et al., 2021) and rats
stress paradigm (Tamashiro et al., 2009) and when PS (Panetta et al., 2017; Tamashiro et al., 2009). Even when GCs
became induced by exogenous glucocorticoid (GC) adminis- were used to developed PS, exposed Sprague Dawley males
tration (Tsai et al., 2019). On the other hand, using BALB/c exhibited increase blood glucose during the glucose toler-
mice, Luft and colleagues reported a reduction in body ance test, but insulin sensitivity curve and plasma insulin
weight in 60 days-old mice exposed to PS using a 30-minute were unaffected (Tsai et al., 2019). However, when research-
movement restriction paradigm every other day from day 8 ers used Wistar rats GCs treatment did not affect basal gly-
of gestation to delivery (Luft et al., 2021). However, sleep cemia in the offspring (Wyrwoll et al., 2008).
fragmentation-PS C57BL/6J male mice showed increased In the liver, glucocorticoids (GCs) promote gluconeogene-
body weight with higher visceral adipose tissue content, sis, while in white adipose tissue they decrease glucose
while females showed no difference (Khalyfa et al., 2015). To uptake and utilization, antagonizing insulin response. Thus,
our knowledge, there are no studies measuring liver weight high levels of GCs result in hyperglycemia and IR (Kuo et al.,
in rodents under PS. 2015). Preliminary results of this model suggest that cortico-
We found higher blood glucose levels in PS during the sterone levels were not affected by PS.
glucose tolerance test at 15-, 30- and 60-minutes post-injec- It seems that disturbances in glucose and lipid metabolism
tion. Similarly, in the insulin sensitivity curve, we observed depend on several variables such as the rodent used (rat or
higher glucose levels at basal, 15-, 30- and 60-minutes post- mice), its genetic background, type of breeding (outbreeding
injection. We performed both the insulin sensitivity curve or inbreeding), and the type of PS used.
6 S. QUIROGA ET AL.

Figure 3. Retroperitoneal adipose tissue gene expression. A: Leptin. B: Resistin. C: Adiponectin. D: Sirtuin-1. E: Interleukin 1 beta and F: Tumor necrosis factor alfa.
Panels A and E: Values are presented as median and interquartile range (n ¼ 8–9 mice in each group). Panels B-D and F: values are presented as mean ± standard
error (n ¼ 8–9 mice in each group). NPS: non-prenatal stress (dark bars); PS: prenatal stress (white bars).  p < .05,  p < .01 and  p < .001.

Adipose tissue gene expression fragmentation model in C57BL/6J mice. They showed an
Adipokines Leptin and Resistin are associated with an increase in Leptin mRNA expression and a decrease in
increase in the number and size of fat cells and, therefore, Adiponectin mRNA in males. Circulation levels of both adipo-
with an increase in body weight or fat content (Ko €nner and kines followed the same pattern (Khalyfa et al., 2015). This
€ning, 2012; Owecki et al., 2010). Both are involved in the
Bru group also reported insulin resistance in PS males (Khalyfa et
development of IR (Barnes and Miner, 2009). Adiponectin is al., 2015). On the other hand, Tsai and colleagues showed
responsible for sensitizing tissues to insulin effects. In the that rats exposed to GCs administration during gestation had
present study, we observed that Leptin and Resistin mRNA lower Leptin mRNA levels in retroperitoneal adipose tissue
expression rose in animals exposed to PS. In contrast, together with an increase in glucose concentration after a
Adiponectin mRNA expression decreased, all signals that glucose tolerance test (Tsai et al., 2019). Moreover, many
would indicate the development of IR. Similar results were authors have reported no changes in circulating Leptin or
reported by Khalyfa and colleagues using a sleep Adiponectin levels under PS in IR absence (Panetta et al.,
STRESS 7

Figure 4. Liver gene expression. A: Peroxisome proliferator-activated receptor gamma coactivator 1-alpha. B: Peroxisome Proliferator-Activated Receptor Alpha. C:
Sirtuin-1. D: Sirtuin-3. E: Suppressor of cytokine signaling 3. Panels A-E: Values are presented as mean ± standard error (n ¼ 8–9 mice in each group). NPS: non-prenatal
stress (dark bars); PS: prenatal stress (white bars).  p < .05 and  p < .01.

2017; Tamashiro et al. 2009). Traditionally, plasma leptin lev- decrease in adiponectin would be impacting the insulin sen-
els were associated with visceral adipose tissue content. sitivity of the peripheral tissues.
However, Franko et al. (2010) have reported, in a model of Sirt1, NAD þ dependent deacetylase protein, is a crucial
PS induced by dexamethasone administration, a significant factor connecting nutrient signals from the environment with
decrease in visceral adipose tissue (determined by weight) energy homeostasis (Schug and Li, 2011). Sirt1 controls
with no difference in the total adipose tissue measured by inflammatory responses, regulates adipokine secretion and
DEXA (dual emission X-ray absorption) but with an increase preserves glucose and lipid homeostasis. Moreover, Sirt1
in circulating leptin levels. These animals showed no changes increases insulin secretion by pancreatic b cells and modu-
in body weight. These findings support our observations on lates insulin signaling (Llorente et al., 2002). We found a
the presence of IR. We propose that alterations in the adipo- decrease in adipose tissue mRNA expression of Sirt1, linked
kines gene expression reflect their concentration in circula- to a reduction in Adiponectin gene expression and inflamma-
tion. Therefore the increase in leptin and resistin and the tory molecules such as Tnf-a and IL-1b. In our model, the
8 S. QUIROGA ET AL.

decrease in Adiponectin expression seems to be directly asso- expression of Socs3 rises due to increased inflammatory cyto-
ciated with the reduction in Sirt1 gene expression. Further kines such as IL-6 and Tnf-a. In the liver, Socs3 suppression
work is needed to study what occurs with Sirt1 protein in improves IR in obese and diabetic db/db mice
this model. (Sachithanandan et al., 2010). Furthermore, mice with liver-
Regarding IL-1b and Tnf-a, exogenous administration of specific deletion of Socs3 exhibit improved liver insulin sensi-
GCs during gestation leads to increased expression of both tivity. However, they also develop obesity and systemic IR
genes in adipose tissue (Mark et al., 2014). However, in our (Torisu et al., 2007). We found that PS C57BL/6J mice showed
PS model, we found no evidence of inflammation. It can be a decrease in Socs3 mRNA expression in the liver. These
attributed to the model, as this is a more physiological model would be a positive sign in terms of its role in the liver.
and therefore with a weaker response than the one observed However, in this work, the gene expression balance seems to
to the administration of exogenous GCs. We propose that be biased toward systemic IR.
the decrease in the expression of inflammatory molecules
might be due to epigenetic modifications produced by
Limitations and conclusion
exposure to PS, such as DNA hypermethylation of these
genes or a key regulator gene (such as NF-jB) leading to a This work has some limitations: First, we did not assess dams
lower expression of inflammatory molecules. These results corticosterone concentration; however, previously, it has
could be explained by an adaptive effect produced by expos- been shown that stressed mothers under restraint stress had
ure to PS; however, more research is needed to study higher corticosterone levels than non-stressed ones (D’mello
this event. and Liu, 2006; Ward and Wainwright, 1988). Second, we did
not record birth weight to avoid excessive handling of the
animals neither studied mothers’ behavior. However, in a
Liver gene expression recent meta-analysis, we showed that birthweight decreases
Sirt1 is responsible for regulating glucose metabolism, fatty significantly in animals subjected to PS and that cross-foster-
acid metabolism, and cholesterol metabolism in the liver. ing to non-stressed mothers had no effects on body weight
During fasting or caloric restriction, Sirt1 is upregulated in (Burguen ~o et al., 2020), suggesting that maternal behavior
this organ, promoting gluconeogenesis by activating Pgc1a has no influence on body weight in this model. Third, we
and Foxo1 while inhibiting Stat3 (Nogueiras et al., 2012). tested several housekeeping genes and selected the most
We found a decrease in Sirt1 mRNA expression in the liver stable, however, we only used one reference gene. Finally,
due to PS. mRNA expression of Pgc1a and Ppara (Ppara inter- we did not analyze protein expression, and conclusions have
acts with Pgc1a promoting mitochondrial fatty acid oxida- been derived solely from the mRNA analysis, something to
tion) were similarly affected. Pgc1a has a key role in multiple be modified in the following studies.
pathways related to energy metabolism control. A decrease In summary, in the present work, we used C57BL/6J mice,
in Pgc1a leads to the development of hepatic steatosis a strain widely used for metabolic research. We found no
(Leone et al., 2005). Ppara is a key regulator of systemic and changes in body weight, visceral adipose tissue, or relative
intrahepatic lipid homeostasis (Leone et al., 1999). liver weight. However, we observed the presence of IR,
These genes decrease in several animal models of high-fat shown by alterations in the glucose tolerance test and the
diet intake, diabetes, and nonalcoholic fatty liver disease insulin sensitivity curve. IR was related to an increase in
(Gencoglu, 2020; Li et al., 2020; Santos et al., 2020; Zhang mRNA expression of Leptin and Resistin with a decrease in
et al., 2020). Similarly, Brunton and colleagues showed a sig- Adiponectin, IL-1b, and Tnf-a, in the adipose tissue. In con-
nificant decrease in mRNA gene expression of Ppara in the trast, in the liver, we observed a reduction in mRNA levels of
liver of males prenatally exposed to an intruder, along with a Pgc1a, Ppara, Sirtuin-1 and 3, and Socs3. In this paper, we
trend toward a reduction in Pgc1a (Brunton et al., 2013). Luft describe, for the first time, an animal model of IR induced by
and colleagues showed an increase in the mRNA expression PS without the presence of inflammation or obesity. These
results suggest this may be an initial stage of the disease
of Ppara in the liver of PS BALB/c male mice, with a decrease
due to normal insulin values found. The alterations observed
in females (Luft et al., 2021). Although we did not observe
in the mRNA expression of both the adipose tissue and the
alterations in cholesterol or triglyceride levels, changes in the
liver support the physiological observation of the presence
expression of key genes in hepatic lipid metabolism would
of IR.
suggest that PS programs an increased susceptibility to hep-
atic fat accumulation, which could lead to the development
of steatosis. Acknowledgments
Sirt3 plays an essential role in preventing metabolic syn-
The authors would like to thank Johnson & Johnson for the kind dona-
drome, being upregulated in response to caloric restriction tion of the glucose test strips and Maria Rosa Gonzalez Murano for her
and exercise, while down-regulated with age, high-fat diet, invaluable technical assistance.
and diabetes (Pinteric et al., 2020). Here, we observed a
decrease in Sirt3 mRNA expression in young adults
(28 weeks old). Ethical standards
Suppressor of cytokine signaling proteins (Socs) are The authors assert that all procedures contributing to this work comply
induced by pro-inflammatory cytokines. In obesity, the with the ethical standards of the relevant international guides on the
STRESS 9

care and use of Laboratory animals (“Guide for the Care and Use of Neuroscience and Biobehavioral Reviews, 117, 198–210. https://doi.org/
Laboratory Animals” (NIH) (revision 2011) and to the EC Directive 86/609/ 10.1016/j.neubiorev.2017.05.016
EEC (revision 2010)) and has been approved by the institutional commit- de Boer, S. F., & Van der Gugten, J. (1987). Daily variations in plasma nor-
tee (CICUAL BIOMED Res N 007/2016). adrenaline, adrenaline and corticosterone concentrations in rats.
Physiology & Behavior, 40(3), 323–328. https://doi.org/10.1016/0031-
9384(87)90054-0
Disclosure statement D’mello, A. P., & Liu, Y. (2006). Effects of maternal immobilization stress
on birth weight and glucose homeostasis in the offspring.
No potential conflict of interest was reported by the author(s). Psychoneuroendocrinology, 31(3), 395–406. https://doi.org/10.1016/j.
psyneuen.2005.10.003
Drago, F., Di Leo, F., & Giardina, L. (1999). Prenatal stress induces body
Funding weight deficit and behavioural alterations in rats: The effect of diaze-
n pam. European Neuropsychopharmacology, 9(3), 239–245. https://doi.
This work was partially supported by Agencia Nacional de Promocio
gica [AMG, PICT 2016-2727], and Consejo Nacional de org/10.1016/S0924-977X(98)00032-7
Cientıfica Tecnolo
Entringer, S., Buss, C., Swanson, J. M., Cooper, D. M., Wing, D. A., Waffarn,
Investigaciones Cientıficas y T ecnicas [AMG, PIP 2015-2017 N
F., & Wadhwa, P. D. (2012). Fetal programming of body composition,
11220150100163]. MAV, AMG and ALB are members of Consejo Nacional
de Investigaciones Cientıficas y T
ecnicas. obesity, and metabolic function: the role of intrauterine stress and
stress biology. Journal of Nutrition and Metabolism, 2012, https://doi.
org/10.1155/2012/632548
Notes on contributors Entringer, S., Buss, C., & Wadhwa, P. D. (2015). Prenatal stress, develop-
ment, health and disease risk: A psychobiological perspective-2015
Sofia Quiroga is currently conducting her Ph.D. with a fellowship from Curt Richter Award Paper. Psychoneuroendocrinology, 62, 366–375.
Agencia Nacional de Promocio n Cientıfica y Tecnolo
gica. https://doi.org/10.1016/j.psyneuen.2015.08.019
Franko, K. L., Forhead, A. J., & Fowden, A. L. (2010). Differential effects of
Yamila Raquel Jua rez has a Ph.D. from Facultad de Ciencias Exactas y
prenatal stress and glucocorticoid administration on postnatal growth
Naturales de la Universidad de Buenos Aires. and glucose metabolism in rats. The Journal of Endocrinology, 204(3),
Marıa Paula Marcone is currently pursuing her Ph.D. with a fellowship 319–329. https://doi.org/10.1677/JOE-09-0390
from Consejo Nacional de Investigaciones Cientıficas y Gencoglu, H. (2020). Maca modulates fat and liver energy metabolism
Tecnicas (CONICET). markers insulin, IRS1, leptin, and SIRT1 in rats fed normal and high-fat
diets. Archives of Physiology and Biochemistry, 2020, 1–7. https://doi.
Marıa Agustina Vidal is a veterinarian from Facultad de Ciencias org/10.1080/13813455.2020.1821064
Veterinarias de la Universidad de Buenos Aires, she is currently a mem- Greenberg, A. S., & Obin, M. S. (2006). Obesity and the role of adipose
ber of the Support Staff for Research and Development Career tissue in inflammation and metabolism. The American Journal of
of CONICET. Clinical Nutrition, 83(2), 461S–465S. https://doi.org/10.1093/ajcn/83.2.
461S
Ana Marıa Genaro obtained her Ph.D. at Facultad de Farmacia y Han, H.-S., Kang, G., Kim, J. S., Choi, B. H., & Koo, S.-H. (2016). Regulation
Bioquımica de la Universidad de Buenos Aires. She is a member of the of glucose metabolism from a liver-centric perspective. Experimental &
Scientific and Technological Researcher Career at CONICET and is cur- Molecular Medicine, 48, e218. https://doi.org/10.1038/emm.2015.122
rently the Head of the Laboratory of Psychoneuroendocrineimmunology Hotamisligil, G. S., Shargill, N. S., & Spiegelman, B. M. (1993). Adipose
lica Argentina.
at Pontificia Universidad Cato expression of tumor necrosis factor-alpha: Direct role in obesity-linked
Adriana Laura Burguen ~o has a Ph.D. from Facultad de Ciencias Exactas y insulin resistance. Science, 259(5091), 87–91. https://doi.org/10.1126/
Naturales de la Universidad de Buenos Aires and is a member of the science.7678183
Scientific and Technological Researcher Career at CONICET. Igosheva, N., Klimova, O., Anishchenko, T., & Glover, V. (2004). Prenatal
stress alters cardiovascular responses in adult rats. The Journal of
Physiology, 557(Pt 1), 273–285. https://doi.org/10.1113/jphysiol.2003.
References 056911
Juarez, Y. R., Quiroga, S., Prochnik, A., Wald, M., Tellechea, M. L., Genaro,
Barnes, K. M., & Miner, J. L. (2009). Role of resistin in insulin sensitivity in A. M., & Burguen ~o, A. L. (2020). Influence of prenatal stress on meta-
rodents and humans. Current Protein & Peptide Science, 10(1), 96–107. bolic abnormalities induced by postnatal intake of a high-fat diet in
https://doi.org/10.2174/138920309787315239 BALB/c mice. Journal of Developmental Origins of Health and Disease,
Berends, L. M., Fernandez-Twinn, D. S., Martin-Gronert, M. S., Cripps, R. L., 2020, 1–10. https://doi.org/10.1017/S2040174420000987
& Ozanne, S. E. (2013). Catch-up growth following intra-uterine Kelishadi, R., Haghdoost, A. A., Jamshidi, F., Aliramezany, M., &
growth-restriction programmes an insulin-resistant phenotype in adi- Moosazadeh, M. (2015). Low birthweight or rapid catch-up growth:
pose tissue. International Journal of Obesity, 37(8), 1051–1057. https:// Which is more associated with cardiovascular disease and its risk fac-
doi.org/10.1038/ijo.2012.196 tors in later life? A systematic review and cryptanalysis. Paediatrics
Brunton, P. J., Sullivan, K. M., Kerrigan, D., Russell, J. A., Seckl, J. R., & and International Child Health, 35(2), 110–123. https://doi.org/10.1179/
Drake, A. J. (2013). Sex-specific effects of prenatal stress on glucose 2046905514Y.0000000136
homoeostasis and peripheral metabolism in rats. The Journal of Khalyfa, A., Carreras, A., Almendros, I., Hakim, F., & Gozal, D. (2015). Sex
Endocrinology, 217(2), 161–173. https://doi.org/10.1530/JOE-12-0540 dimorphism in late gestational sleep fragmentation and metabolic
Burguen ~o, A. L., Juarez, Y. R., Genaro, A. M., & Tellechea, M. L. (2020). dysfunction in offspring mice. Sleep, 38(4), 545–557. https://doi.org/10.
Prenatal stress and later metabolic consequences: Systematic review 5665/sleep.4568
and meta-analysis in rodents. Psychoneuroendocrinology, 113, 104560. Kivim€aki, M., & Steptoe, A. (2018). Effects of stress on the development
https://doi.org/10.1016/j.psyneuen.2019.104560 and progression of cardiovascular disease. Nature Reviews. Cardiology,
Canto, C., & Auwerx, J. (2009). PGC-1alpha, SIRT1 and AMPK, an energy 15(4), 215–229. https://doi.org/10.1038/nrcardio.2017.189
sensing network that controls energy expenditure. Current Opinion in Ko€nner, A. C., & Br€ uning, J. C. (2012). Selective insulin and leptin resist-
Lipidology, 20(2), 98–105. https://doi.org/10.1097/MOL. ance in metabolic disorders. Cell Metabolism, 16(2), 144–152. https://
0b013e328328d0a4 doi.org/10.1016/j.cmet.2012.07.004
Cao-Lei, L., de Rooij, S. R., King, S., Matthews, S. G., Metz, G. A. S., Kuo, T., McQueen, A., Chen, T.-C., & Wang, J.-C. (2015). Regulation of glu-
Roseboom, T. J., & Szyf, M. (2020). Prenatal stress and epigenetics. cose homeostasis by glucocorticoids. Advances in Experimental
10 S. QUIROGA ET AL.

Medicine and Biology, 872, 99–126. https://doi.org/10.1007/978-1-4939- independent from insulin resistance. Experimental and Clinical
2895-8 Endocrinology & Diabetes, 119(02), 117–121. https://doi.org/10.1055/s-
Leone, T. C., Lehman, J. J., Finck, B. N., Schaeffer, P. J., Wende, A. R., 0030-1263111
Boudina, S., Courtois, M., Wozniak, D. F., Sambandam, N., Bernal- Panetta, P., Berry, A., Bellisario, V., Capoccia, S., Raggi, C., Luoni, A.,
Mizrachi, C., Chen, Z., Holloszy, J. O., Medeiros, D. M., Schmidt, R. E., Longo, L., Riva, M. A., & Cirulli, F. (2017). Long-Term Sex-Dependent
Saffitz, J. E., Abel, E. D., Semenkovich, C. F., & Kelly, D. P. (2005). PGC- Vulnerability to Metabolic challenges in Prenatally Stressed Rats.
1alpha deficiency causes multi-system energy metabolic derange- Frontiers in Behavioral Neuroscience, 11, 113. https://doi.org/10.3389/
ments: Muscle dysfunction, abnormal weight control and hepatic stea- fnbeh.2017.00113
tosis. PLoS Biology, 3(4), e101. https://doi.org/10.1371/journal.pbio. Pinteric, M., Podgorski, I. I., Hadzija, M. P., Bujak, I. T., Dekanic, A., Bagaric,
0030101 R., Farkas, V., Sobocanec, S., & Balog, T. (2020). Role of Sirt3 in
Leone, T. C., Weinheimer, C. J., & Kelly, D. P. (1999). A critical role for the Differential Sex-Related Responses to a High-Fat Diet in Mice.
peroxisome proliferator-activated receptor alpha (PPARalpha) in the Antioxidants, 9(2), 174. https://doi.org/10.3390/antiox9020174
cellular fasting response: The PPARalpha-null mouse as a model of Raghow, R. (2018). Circadian rhythms of hormone secretion and obesity.
fatty acid oxidation disorders. Proceedings of the National Academy of World Journal of Diabetes, 9(11), 195–198. https://doi.org/10.4239/wjd.
Sciences of the United States of America, 96(13), 7473–7478. https://doi. v9.i11.195
org/10.1073/pnas.96.13.7473 Recinella, L., Orlando, G., Ferrante, C., Chiavaroli, A., Brunetti, L., & Leone,
Lesage, J., Del-Favero, F., Leonhardt, M., Louvart, H., Maccari, S., Vieau, D., S. (2020). Adipokines: New potential therapeutic target for obesity
& Darnaudery, M. (2004). Prenatal stress induces intrauterine growth and metabolic, rheumatic, and cardiovascular diseases. Frontiers in
restriction and programmes glucose intolerance and feeding behav- Physiology, 11, 578966. https://doi.org/10.3389/fphys.2020.578966
iour disturbances in the aged rat. The Journal of Endocrinology, 181(2), Rehman, K., & Akash, M. S. H. (2016). Mechanisms of inflammatory
291–296. https://doi.org/10.1677/joe.0.1810291 responses and development of insulin resistance: How are they inter-
Li, S., Qian, Q., Ying, N., Lai, J., Feng, L., Zheng, S., Jiang, F., Song, Q., linked? Journal of Biomedical Science, 23(1), 87. https://doi.org/10.
Chai, H., & Dou, X. (2020). Activation of the AMPK-SIRT1 pathway con- 1186/s12929-016-0303-y
tributes to protective effects of Salvianolic acid A against lipotoxicity Reynolds, R. M. (2010). Corticosteroid-mediated programming and the
in hepatocytes and NAFLD in mice. Frontiers in Pharmacology, 11, pathogenesis of obesity and diabetes. The Journal of Steroid
560905. https://doi.org/10.3389/fphar.2020.560905 Biochemistry and Molecular Biology, 122(1–3), 3–9. https://doi.org/10.
Li, Y., Ding, L., Hassan, W., Abdelkader, D., & Shang, J. (2013). Adipokines 1016/j.jsbmb.2010.01.009
and hepatic insulin resistance. Journal of Diabetes Research, 2013, Rius-Perez, S., Torres-Cuevas, I., Millan, I., Ortega, A. L., & P
erez, S. (2020).
170532. https://doi.org/10.1155/2013/170532 PGC-1a, Inflammation, and Oxidative Stress: An Integrative View in
Liu, M., & Liu, F. (2009). Transcriptional and post-translational regulation
Metabolism. Oxidative Medicine and Cellular Longevity, 2020, 1452696.
of adiponectin. The Biochemical Journal, 425(1), 41–52. https://doi.org/
https://doi.org/10.1155/2020/1452696
10.1042/BJ20091045
Sachithanandan, N., Fam, B. C., Fynch, S., Dzamko, N., Watt, M. J.,
Llorente, E., Brito, M. L., Machado, P., & Gonzalez, M. C. (2002). Effect of
Wormald, S., Honeyman, J., Galic, S., Proietto, J., Andrikopoulos, S.,
prenatal stress on the hormonal response to acute and chronic stress
Hevener, A. L., Kay, T. W. H., & Steinberg, G. R. (2010). Liver-specific
and on immune parameters in the offspring. Journal of Physiology and
suppressor of cytokine signaling-3 deletion in mice enhances hepatic
Biochemistry, 58(3), 143–149. https://doi.org/10.1007/BF03179851
insulin sensitivity and lipogenesis resulting in fatty liver and obesity.
Luft, C., Levices, I. P., Pedrazza, L., de Oliveira, J. R., & Donadio, M. V. F.
Hepatology, 52(5), 1632–1642. https://doi.org/10.1002/hep.23861
(2021). Sex-dependent metabolic effects of pregestational exercise on
Santos, F. O., Correia, B. R. O., Marinho, T. S., Barbosa-da-Silva, S.,
prenatally stressed mice. Journal of Developmental Origins of Health
Mandarim-de-Lacerda, C. A., & Souza-Mello, V. (2020). Anti-steatotic
and Disease, 12(2), 271–279. https://doi.org/10.1017/
linagliptin pleiotropic effects encompasses suppression of de novo
S2040174420000343
Maeda, N., Takahashi, M., Funahashi, T., Kihara, S., Nishizawa, H., Kishida, lipogenesis and ER stress in high-fat-fed mice. Molecular and Cellular
K., Nagaretani, H., Matsuda, M., Komuro, R., Ouchi, N., Kuriyama, H., Endocrinology, 509, 110804. https://doi.org/10.1016/j.mce.2020.110804
Hotta, K., Nakamura, T., Shimomura, I., & Matsuzawa, Y. (2001). Schug, T. T., & Li, X. (2011). Sirtuin 1 in lipid metabolism and obesity.
PPARgamma ligands increase expression and plasma concentrations Annals of Medicine, 43(3), 198–211. https://doi.org/10.3109/07853890.
of adiponectin, an adipose-derived protein. Diabetes, 50(9), 2010.547211
2094–2099. https://doi.org/10.2337/diabetes.50.9.2094 Sternberg, W. F., & Ridgway, C. G. (2003). Effects of gestational stress and
Mairesse, J., Lesage, J., Breton, C., Br eant, B., Hahn, T., Darnaud ery, M., neonatal handling on pain, analgesia, and stress behavior of adult
Dickson, S. L., Seckl, J., Blondeau, B., Vieau, D., Maccari, S., & Viltart, O. mice. Physiology & Behavior, 78(3), 375–383. https://doi.org/10.1016/
(2007). Maternal stress alters endocrine function of the feto-placental S0031-9384(03)00015-5
unit in rats. American Journal of Physiology. Endocrinology and Tamashiro, K. L. K., Terrillion, C. E., Hyun, J., Koenig, J. I., & Moran, T. H.
Metabolism, 292(6), E1526–33. https://doi.org/10.1152/ajpendo.00574. (2009). Prenatal stress or high-fat diet increases susceptibility to diet-
2006 induced obesity in rat offspring. Diabetes, 58(5), 1116–1125. https://
Mark, P. J., Wyrwoll, C. S., Zulkafli, I. S., Mori, T. A., & Waddell, B. J. (2014). doi.org/10.2337/db08-1129
Rescue of glucocorticoid-programmed adipocyte inflammation by Torisu, T., Sato, N., Yoshiga, D., Kobayashi, T., Yoshioka, T., Mori, H., Iida,
omega-3 fatty acid supplementation in the rat. Reproductive Biology M., & Yoshimura, A. (2007). The dual function of hepatic SOCS3 in
and Endocrinology, 12, 39. https://doi.org/10.1186/1477-7827-12-39 insulin resistance in vivo. Genes to Cells, 12(2), 143–154. https://doi.
McGuinness, O. P., Ayala, J. E., Laughlin, M. R., & Wasserman, D. H. (2009). org/10.1111/j.1365-2443.2007.01044.x
NIH experiment in centralized mouse phenotyping: The Vanderbilt Tsai, C.-C., Tiao, M.-M., Sheen, J.-M., Huang, L.-T., Tain, Y.-L., Lin, I.-C., Lin,
experience and recommendations for evaluating glucose homeostasis Y.-J., Lai, Y.-J., Chen, C.-C., Chang, K.-A., & Yu, H.-R. (2019). Obesity pro-
in the mouse. American Journal of Physiology. Endocrinology and grammed by prenatal dexamethasone and postnatal high-fat diet
Metabolism, 297(4), E849–55. https://doi.org/10.1152/ajpendo.90996. leads to distinct alterations in nutrition sensory signals and circadian-
2008 clock genes in visceral adipose tissue. Lipids in Health and Disease,
Nogueiras, R., Habegger, K. M., Chaudhary, N., Finan, B., Banks, A. S., 18(1), 19. https://doi.org/10.1186/s12944-019-0963-1
Dietrich, M. O., Horvath, T. L., Sinclair, D. A., Pfluger, P. T., & Tscho €p, Tsang, A. H., Astiz, M., Friedrichs, M., & Oster, H. (2016). Endocrine regula-
M. H. (2012). Sirtuin 1 and sirtuin 3: Physiological modulators of tion of circadian physiology. The Journal of Endocrinology, 230(1),
metabolism. Physiological Reviews, 92(3), 1479–1514. https://doi.org/10. R1–R11. https://doi.org/10.1530/JOE-16-0051
1152/physrev.00022.2011 Vega, R. B., Huss, J. M., & Kelly, D. P. (2000). The coactivator PGC-1 coop-
Owecki, M., Miczke, A., Nikisch, E., Pupek-Musialik, D., & Sowin ski, J. erates with peroxisome proliferator-activated receptor alpha in tran-
(2010). Serum resistin concentrations are higher in human obesity but scriptional control of nuclear genes encoding mitochondrial fatty acid
STRESS 11

oxidation enzymes. Molecular and Cellular Biology, 20(5), 1868–1876. Yu, H.-R., Tain, Y.-L., Tiao, M.-M., Chen, C.-C., Sheen, J.-M., Lin, I.-C., Li, S.-
https://doi.org/10.1128/MCB.20.5.1868-1876.2000 W., Tsai, C.-C., Lin, Y.-J., Hsieh, K.-S., & Huang, L.-T. (2018). Prenatal
Ward, G. R., & Wainwright, P. E. (1988). Reductions in maternal food and dexamethasone and postnatal high-fat diet have a synergistic effect
water intake account for prenatal stress effects on neurobehavioral of elevating blood pressure through a distinct programming mechan-
development in B6D2F2 mice. Physiology & Behavior, 44(6), 781–786. ism of systemic and adipose renin-angiotensin systems. Lipids in
https://doi.org/10.1016/0031-9384(88)90062-5 Health and Disease, 17(1), 50. https://doi.org/10.1186/s12944-018-0701-
Ward, I. L., & Weisz, J. (1984). Differential effects of maternal stress on cir- 0
culating levels of corticosterone, progesterone, and testosterone in Zambrano, E., Iban ~ez, C., Martınez-Samayoa, P. M., Lomas-Soria, C.,
Durand-Carbajal, M., & Rodrıguez-Gonzalez, G. L. (2016). Maternal
male and female rat fetuses and their mothers. Endocrinology, 114(5),
obesity: Lifelong metabolic outcomes for offspring from poor develop-
1635–1644. https://doi.org/10.1210/endo-114-5-1635
mental trajectories during the perinatal period. Archives of Medical
Wyrwoll, C. S., Mark, P. J., Mori, T. A., & Waddell, B. J. (2008).
Research, 47(1), 1–12. https://doi.org/10.1016/j.arcmed.2016.01.004
Developmental programming of adult hyperinsulinemia, increased
Zhang, Y., Zhou, B., Wen, M., Hu, M., Peng, J.-G., Wang, Y., Fan, L.-L., &
proinflammatory cytokine production, and altered skeletal muscle
Tang, L. (2020). ZG02 Improved hepatic glucose metabolism and insu-
expression of SLC2A4 (GLUT4) and uncoupling protein 3. The Journal lin sensitivity via activation of AMPK/Sirt1 signaling pathways in a
of Endocrinology, 198(3), 571–579. https://doi.org/10.1677/JOE-08- high-fat diet/streptozotocin-induced Type 2 diabetes model. Diabetes,
0210 Metabolic Syndrome and Obesity, 13, 4333–4339. https://doi.org/10.
Yang, Z., Roth, K., Agarwal, M., Liu, W., & Petriello, M. C. (2021). The tran- 2147/DMSO.S275145
scription factors CREBH, PPARa, and FOXO1 as critical hepatic media- Zhou, S., Tang, X., & Chen, H.-Z. (2018). Sirtuins and insulin resistance.
tors of diet-induced metabolic dysregulation. The Journal of Nutritional Frontiers in Endocrinology, 9, 748. https://doi.org/10.3389/fendo.2018.
Biochemistry, 95, 108633. https://doi.org/10.1016/j.jnutbio.2021.108633 00748

You might also like