Science 28Feb2020

(lily) #1

RESEARCH ARTICLE



MICROBIOTA


Maternal gut microbiota in pregnancy influences


offspring metabolic phenotype in mice


Ikuo Kimura1,2†, Junki Miyamoto1,2, Ryuji Ohue-Kitano1,2, Keita Watanabe^1 , Takahiro Yamada^3 ,
Masayoshi Onuki^3 , Ryo Aoki4,5, Yosuke Isobe^6 , Daiji Kashihara^7 , Daisuke Inoue^7 , Akihiko Inaba^8 ,
Yuta Takamura^9 , Satsuki Taira^1 , Shunsuke Kumaki^8 , Masaki Watanabe^9 , Masato Ito^3 ,
Fumiyuki Nakagawa10,11, Junichiro Irie2,12, Hiroki Kakuta^9 , Masakazu Shinohara^13 , Ken Iwatsuki^8 ,
Gozoh Tsujimoto^7 , Hiroaki Ohno2,14, Makoto Arita6,15,16, Hiroshi Itoh2,12, Koji Hase3,17†


Antibiotics and dietary habits can affect the gut microbial community, thus influencing disease
susceptibility. Although the effect of microbiota on the postnatal environment has been well
documented, much less is known regarding the impact of gut microbiota at the embryonic stage. Here
we show that maternal microbiota shapes the metabolic system of offspring in mice. During pregnancy,
short-chain fatty acids produced by the maternal microbiota dictate the differentiation of neural,
intestinal, and pancreatic cells through embryonic GPR41 and GPR43. This developmental process
helps maintain postnatal energy homeostasis, as evidenced by the fact that offspring from germ-free
mothers are highly susceptible to metabolic syndrome, even when reared under conventional conditions.
Thus, our findings elaborate on a link between the maternal gut environment and the developmental
origin of metabolic syndrome.


T


he gut microbiota substantially contrib-
utes to energy extraction from indigestible
polysaccharides, such as oligosaccharides
and soluble dietary fibers, and influences
host energy homeostasis during infancy
and adulthood ( 1 – 6 ). Therefore, maintenance


of the symbiotic microbial community is of
paramount importance for host health. Changes
in microbial composition may cause dysbiosis,
leading to disease-prone phenotypes in the
host. Dysbiosis has been implicated in a grow-
ing number of systemic disorders, including
metabolic syndrome ( 7 – 9 ). Obesity is a major
risk factor for metabolicsyndrome, predispos-
ing patients to cardiovascular disease and type
2 diabetes. In addition to genetic and epige-
netic factors, changes in the gut microbiota
have been implicated in the development of
obesity, in combination with dietary factors.
Gut microbiota–derived metabolites represented
by short-chain fatty acids (SCFAs; e.g., acetate,
propionate, and butyrate) ( 9 – 14 ) not only fuel
host cells but also serve as signaling molecules
between the gut microbiota and extraintesti-
nal organs.
We have previously shown that SCFAs sup-
press insulin signaling in adipocytes and ulti-
mately inhibit fat deposition via adipose GPR43
( 15 ). Furthermore, GPR41 stimulation by pro-
pionate potently activates sympathetic neurons
to regulate energy expenditure ( 16 ). A recent
study further showed that feeding pregnant
mice a high-fiber or acetate-supplemented diet
decreases offspring susceptibility to allergic air-
way disease ( 17 ).Thesefindingsraisethepos-
sibility that maternal SCFAs play a key role in
the regulation of disease susceptibility during
postnatal life in the context of the developmen-
tal origins of health and disease theory ( 18 ).
However, the underlying mechanisms and
the biological importance of the maternal-
embryonic cross-talk via microbial metabo-
lites remain obscure.

In this study, we explored the impact of the
maternal gut microbiota on energy homeostasis
in offspring in a mouse model. We observed
that the offspring of germ-free (GF) mothers are
more prone to obesity and glucose intolerance
than those of specific pathogen–free (SPF)
mothers. Maternal microbiota–derived SCFAs
translocated to the embryos to facilitate de-
velopment of the sympathetic nervous system
and regulation of insulin levels via GPR41 and
GPR43 signaling. Thus, during pregnancy, the
gut microbiota provides an environmental cue
that fine-tunes energy homeostasis in offspring.

Offspring of GF mothers and
obesity development
To investigate the impact of the maternal gut
microbiota during pregnancy on offspring, preg-
nant mice were bred under SPF and GF con-
ditions. On day 18.5 of gestation, pregnant GF
mice received a fecal microbiota transplant from
SPF mice of the corresponding strain to prevent
overgrowth of unfavorable microbes. Newborn
animals were raised by foster mothers under
conventional conditions to align growth envi-
ronments after birth. After weaning, the male
mice were fed a high-fat diet (HFD) to induce
obesity (Fig. 1A). Although the postnatal body
weight of newborns from GF ICR mothers
was less than that of offspring from SPF ICR
mothers (fig. S1A), the offspring developed
marked obesity upon HFD consumption dur-
ing growth (Fig. 1A). In addition, the weight of
perirenal or subcutaneous white adipose tissue
(WAT) and the liver was significantly higher
in offspring derived from GF mothers (GF off-
spring)thaninthosefromSPFmothers(SPF
offspring) at 16 weeks (Fig. 1A), which is in
accordance with increases in WAT adipocyte
size and hepatic triglycerides (TGs) (fig. S1, B
and C). Concomitantly, plasma glucose, TGs,
non-esterified fatty acids (NEFAs), and total
cholesterol levels were significantly elevated
in GF offspring (Fig. 1B). Body temperature
and heart rate were significantly reduced (Fig.
1C), whereas plasma insulin levels and pan-
creatic islet sizes were significantly higher in
GF offspring than in SPF offspring (fig. S1D).
Moreover, the GF offspring exhibited elevated
food intake (fig. S1E), with reduced plasma
levels of the gut hormone peptide YY (PYY)
and glucagon-like peptide-1 (GLP-1) (Fig. 1D)
as well as reduced energy expenditure (Fig. 1E).
These results indicate that the GF offspring
exhibited an obese phenotype upon HFD feeding.
In support of this interpretation, HFD-induced
glucose intolerance and insulin resistance were
significantly accelerated in GF offspring (Fig.
1F), indicating impaired insulin sensitivity. No-
tably, female GF offspring also exhibited similar
phenotypes (fig. S2, A to H).
Mammalian neonates are initially exposed
to the vaginal microbiota, which substantially
contributes to the establishment of the gut

RESEARCH


Kimuraet al.,Science 367 , eaaw8429 (2020) 28 February 2020 1of12


(^1) Department of Applied Biological Science, Graduate School of
Agriculture, Tokyo University of Agriculture and Technology,
Fuchu-shi, Tokyo 183-8509, Japan.^2 AMED-CREST, Japan
Agency for Medical Research and Development, Chiyoda-ku,
Tokyo 100-0004, Japan.^3 Division of Biochemistry, Faculty of
Pharmacy and Graduate School of Pharmaceutical Science,
Keio University, Tokyo 105-8512, Japan.^4 Division of
Gastroenterology and Hepatology, Department of Internal
Medicine, Keio University School of Medicine, Tokyo 160-8582,
Japan.^5 Institute of Health Sciences, Ezaki Glico Co., Ltd.,
Osaka 555-8502, Japan.^6 Laboratory for Metabolomics, RIKEN
Center for Integrative Medical Sciences, Kanagawa 230-0045,
Japan.^7 Department of Genomic Drug Discovery Science,
Graduate School of Pharmaceutical Sciences, Kyoto University,
Kyoto 606-8501, Japan.^8 Department of Nutritional Science
and Food Safety, Tokyo University of Agriculture, Tokyo 156-
8502, Japan.^9 Division of Pharmaceutical Sciences, Okayama
University Graduate School of Medicine, Dentistry and
Pharmaceutical Sciences, Okayama 700-8530, Japan.
(^10) Department of Medicine, Shiga University of Medical Science,
Shiga 520-2192, Japan.^11 Nishiwaki Laboratory, CMIC Pharma
Science Co., Ltd., Hyogo 677-0032, Japan.^12 Department of
Endocrinology, Metabolism and Nephrology, School of
Medicine, Keio University, Tokyo 160-8582, Japan.^13 Division of
Epidemiology, Kobe University Graduate School of Medicine,
Kobe 650-0017, Japan.^14 Department of Bioorganic Medicinal
Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto
University, Kyoto 606-8501, Japan.^15 Division of Physiological
Chemistry and Metabolism, Keio University Faculty of
Pharmacy, Tokyo, 105-0011, Japan.^16 Cellular and Molecular
Epigenetics Laboratory, Graduate School of Medical Life
Science, Yokohama City University, Kanagawa 230-0045,
Japan.^17 International Research and Development Center for
Mucosal Vaccines, The Institute of Medical Science, The
University of Tokyo (IMSUT), Tokyo 108-8639, Japan.
*These authors contributed equally to this work.
†Corresponding author. Email: [email protected] (I.K.);
[email protected] (K.H.)

Free download pdf