Download as pdf or txt
Download as pdf or txt
You are on page 1of 39

Brown fat-activating lipokine 12,13-diHOME in human milk is associated with infant adiposity

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


Danielle Wolfs, M.P.H.1; Matthew D. Lynes, Ph.D.1; Yu-Hua Tseng, Ph.D.1; Stephanie Pierce, M.D.2;
Valerie Bussberg, B.S.c.3, Abena Darkwah, B.S.c.3, Vladimir Tolstikov, Ph.D.3, Niven R. Narain, Ph.D.3,
Michael C. Rudolph, Ph.D.5, Michael A. Kiebish, Ph.D.3, Ellen W. Demerath, Ph.D.4; David A. Fields,
Ph.D.5; Elvira Isganaitis, M.D., M.P.H.1,6

1. Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA

t
2. Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center,

ip
Oklahoma City, OK

3. BERG, Framingham, MA

cr
4. Division of Epidemiology and Community Health, University of Minnesota School of Public Health,

us
Minneapolis, MN

5. Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK


an
6. Department of Pediatrics, Harvard Medical School, Boston, MA
M

Corresponding author:

Elvira Isganaitis, M.D., M.P.H.


d

Assistant Investigator and Pediatric Endocrinologist


e

Joslin Diabetes Center


pt

One Joslin Place, room 655A


ce

Boston, MA 02215

Tel: 617-732-2603
Ac

Fax: 617-309-2593

Elvira.Isganaitis@joslin.harvard.edu

Clinical trial registry #: www.clinicaltrials.gov, NCT02535637

© The Author(s) 2020. Published by Oxford University Press on behalf of the Endocrine
Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com
Disclosure summary: We would like to thank those who made this study possible. DAF and EWD
are supported by the Eunice Kennedy Shriver National Institute of Child Health & Human
Development of the National Institutes of Health under award number R01HD080444. YT receives

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


funding from United States Army Medical Research grant W81XWH-17-1-0428 and National
Institutes of Health (NIH) grants: R01DK077097 and R01DK102898. SP receives funding from the
University of Oklahoma College of Medicine Alumni Association. DAF, DW, SP, and EI receive funding
from the Harold Hamm Diabetes Center Team Science Grant. MDL was supported by NIH grant
K01DK111714. DW, MDL, EI, and YT by the DRC award (P30DK036836 –PI: George King). The authors
report no conflicts of interest related to the study. All authors read and approved the final
manuscript.

t
ip
cr
us
an
M
e d
pt
ce
Ac
Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020
ABSTRACT

Context: Little is known about the specific breastmilk components responsible for protective effects

t
on infant obesity. Whether 12,13-diHOME, an oxidized linoleic acid metabolite and activator of

ip
brown fat metabolism, is present in human milk, or linked to infant adiposity, is unknown.

cr
Objective: To examine associations between concentrations of 12,13-diHOME in human milk and

infant adiposity.

us
an
Design: Prospective cohort study between 2015-2019, following participants from birth to 6-months.

Setting: Academic medical centers.


M

Participants: Volunteer sample of 58 exclusively breastfeeding mother-infant pairs; exclusion criteria


d

included smoking, gestational diabetes, and health conditions with the potential to influence
e

maternal or infant weight gain.


pt

Main outcome measures: Infant anthropometric measures including weight, length, BMI, and body
ce

composition at birth and at 1, 3 and 6 months postpartum.


Ac

Results: We report for the first time that 12,13-diHOME is present in human milk. Higher milk 12,13-

diHOME was associated with increased weight-for-length (WFL) Z-score at birth (=0.5742,

p=0.0008), lower infant fat mass at 1-month (p=0.021), and reduced gain in body mass index (BMI)

Z-score from 0 to 6-months (=-0.3997, p=0.025). We observed similar associations between infant

adiposity and milk abundance of related oxidized linoleic acid metabolites 12,13-epOME and 9,10-

diHOME, and metabolites linked to thermogenesis including succinate and lyso-phosphatidylglycerol


18:0. Milk abundance of 12,13-diHOME was not associated with maternal BMI, but was positively

associated with maternal height, milk glucose concentration, and was significantly increased after a

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


bout of moderate exercise.

Conclusions: We report novel associations between milk abundance of 12,13-diHOME and adiposity

during infancy.

t
Keywords: breastmilk, thermogenic metabolites, infant adiposity, brown fat activators, 12,13-

ip
diHOME, infant metabolism

cr
us
an
M
e d
pt
ce
Ac
INTRODUCTION

Despite efforts to promote breastfeeding as a way to combat childhood obesity, relatively

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


little is still known about specific components in breastmilk responsible for its protective effects. A

recent analysis uncovered a role for alkylglycerol lipids in human milk as regulators of mitochondrial

metabolism and adipocyte “beige-ing” (1), suggesting that breastfeeding may activate adipocyte

thermogenesis in the neonate. Whether this effect is limited to alkylglycerols, and whether

t
additional signaling lipids and metabolites in human milk might also modulate adipocyte metabolism

ip
or activate brown fat, remains unknown.

cr
Human milk is rich in lipids, and several signaling lipids, or “lipokines”, have been reported to

us
act like hormones to regulate inflammation, insulin sensitivity, and systemic metabolism (2,3). One

recently identified lipokine, 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME), an oxidized


an
derivative of linoleic acid, was shown to regulate brown adipose tissue fuel uptake and
M

thermogenesis in mice and humans (4). The term “thermogenic metabolite” has been used to

describe the brown-fat activating potential of 12,13-diHOME and other metabolites (e.g. succinate,
d

BAIBA, 12-HETE, 12-HEPE, uridine, etc.). Plasma levels of 12,13-diHOME are increased in response to
e

cold exposure and acute exercise (4-7) and inversely related to body mass index (BMI), plasma
pt

triglycerides, insulin, and markers of liver function (4,8). Moreover, 12,13-diHOME in newborn feces
ce

has been linked to higher risk of asthma and atopic diseases (9). However, this lipid has not

previously been identified in human milk.


Ac

With this analysis, we tested whether 12,13-diHOME is present in human milk, and

examined its associations with infant weight gain, BMI, and body composition during the early

postnatal period. We additionally examined associations between milk abundance of other

thermogenic metabolites and infant adiposity. We report that human milk concentration of 12,13-

diHOME, related oxylipids including 12,13-epOME and 9,10-epOME, the Krebs cycle metabolite

succinate, and lyso-phosphatidylglycerol 18:0 (Lyso-PG 18:0), are inversely associated with infant
adiposity. Our data add to the emerging evidence that human milk contains lipids and metabolites

that can modulate adipocyte metabolism (1) and provide further support for the possibility that

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


differences in milk composition could be functionally related to early life obesity risk.

METHODS

Mother-infant participants and study procedures.

t
ip
This prospective cohort study enrolled 58 mother-infant pairs at Oklahoma University Health

cr
Sciences Center and University of Minnesota (CTI: NCT02535637). Eligibility requirements included

us
intention to breastfeed exclusively from birth to 6-months and a singleton, term pregnancy resulting

in a healthy infant. Exclusion criteria included maternal smoking, gestational or pre-gestational


an
diabetes, and any health conditions with the potential to influence maternal weight gain during

pregnancy or infant growth, including but not limited to: hypertension, pre-eclampsia, use of oral
M

glucocorticoids, fetal congenital anomalies, etc. Medical records and patient questionnaires provided
d

demographic and clinical information. The mother-infant pairs presented to the study site for weight
e

measurements and to provide milk samples at 1, 3, and 6-months (± 5 days) post-delivery. Milk
pt

samples were collected between 08:00 and 10:00 hours, at least 1.5 hours after the last infant

feeding, and with the mother fasted for at least 1 hour. The infant’s weight was measured by a high
ce

sensitivity scale (Seca 728) and the mothers then breastfed from both breasts (for a full and complete
Ac

milk expression). Upon completion of all testing protocols, the mothers provided a single breast

expression sample by breast pump from whichever side they felt could provide the fullest expression.

The breast milk was collected in 150 mL BPA-free polypropylene vessels, aliquoted, and kept at -80°C

until the time of analysis. The University of Oklahoma Health Sciences Institutional Review Board

approved all the procedures and protocols detailed in this study.


Acute exercise bout.

To test whether milk abundance of 12,13-diHOME is influenced by maternal exercise, we

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


performed a pilot study of acute exercise in a distinct volunteer sample of sixteen breastfeeding

mother-infant pairs recruited separately at Oklahoma University Health Sciences Center between

2018-2020. The exercise bout was done at 1 month postpartum, and consisted of the mother

walking for three bouts of 10 minutes (with 2-5 minutes rest between each bout) on a treadmill at

t
65-75% of heart rate reserve per the Karvonen Formula: [Target heart rate = (max heart rate –

ip
resting heart rate) x intensity + resting heart rate]. Speed/grade was adjusted to maintain the target

cr
intensity. Milk samples were collected at baseline and 90 minutes after exercise.

us
an
Infant body composition analysis.

A digital scale (Seca 728) and infantometer (Seca 416) were used to measure the infant’s
M

weight and length (crown-to-heel) at 1, 3, and 6 months. Air-displacement plethysmography (ADP,


d

COSMED) was performed on infants at their 1-month appointment. At 6-months, a dual-energy x-ray
e

absorptiometry (DXA, Lunar scanner, GE Healthcare) device was used to measure infant body
pt

composition (% fat, total fat and fat-free mass). Skin fold thicknesses were measured at the

abdomen, subscapular, thigh and triceps in duplicate using Harpenden skinfold calipers by the same
ce

technician (DAF). Further, arm and waist circumferences were taken in duplicate with a tension tape
Ac

measure. The same investigator performed the DXA scans and positioned the infants to minimize

variability.
Assay Methodology

Lipidomic Analysis and 12,13-diHOME Quantification

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


Concentrations of 12,13-diHOME and other lipokines were measured through liquid

chromatography-tandem mass spectrometry (LC-MS/MS). Breast milk samples were thawed at room

temperature and immediately placed on ice. Aliquots of 200 µL were taken and added to 600 µL of

methanol for a protein crash. Ten microliters of a mixture of 5 deuterated internal standards were

t
spiked into the samples, vortexed and stored at -20⁰C overnight. The deuterated standard mixture

ip
contains d4-9-HODE, d4-9,10-diHOME, d8-5(S)-HETE, d4-LTB4, and d4-PGE2 at a concentration of 100

cr
ng/mL. Breast milk samples were then subjected to a solid phase extraction. C18 cartridges at

us
500mg/6mL (Biotage, Uppsala, Sweden) were conditioned with 10 mL of methanol followed by 10

mL of water. The samples were centrifuged at 14000 g, and the pH of the supernatant of the
an
samples was adjusted by adding pH 3.5 water before loading the samples onto the C18 cartridges.
M

The cartridges were washed with 5 mL water followed by 5 mL hexanes. Fractions were dried down

under a stream of N2 gas, and reconstituted in 50 µL methanol:water (1:1, by vol). Samples were
d

centrifuged at 20000 g and the supernatant was transferred to LC-MS vials for analysis.
e

Electrospray ionization (ESI) LC-MS/MS was performed on a TripleTOF 6600 (Sciex,


pt

Framingham, MA, USA) coupled to an Eksigent Ekspert MicroLC 200 (Sciex, Framingham, MA, USA)
ce

system with a Synergi Fusion-RP capillary C18 column (150 x 0.5mm, 4 µm; Phenomenex) with a

column oven heated to 40⁰C. Eleven µL of sample was injected at a flow rate of 20 µL/min and were
Ac

separated by reverse phase chromatography with mobile phases A (100% H2O, 0.1% acetic acid) and

B (100% MeOH, 0.1% acetic acid). The gradient started at 60% B for 0.5 minutes, increased to 80% B

by 5 minutes, increased to 95% B by 9 minutes, held at 95% B for 1 minute, and decreased to 60% B

by 12 minutes to equilibrate. Samples were only acquired in negative mode due to the chemical

structure of the targeted lipids. The ESI DuoSpray Ion Source parameter settings were set to: ion

source gas 1 (GS1) at 15, ion source gas 2 (GS2) at 20, curtain gas (CUR) at 20, temperature at 400⁰C,
and ion spray voltage floating (ISVF) at -4500 V. The high-resolution multiple reaction monitoring

(HR-MRM) acquisition method consisted of a TOF MS experiment looped with 31 MS/MS high

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


sensitivity product ion experiments with 50 ms accumulation time for each scan. The mass

spectrometer acquisition time was set to 10 minutes with a total of 353 cycles per experiment. The

declustering potential (DP) was set to -100 and the collision energy (CE) was set to -30 eV with a

collision energy spread (CES) of 10 eV. Mass shift was controlled by calibrant solution delivered

every 10 samples at 500 µL/min by the atmospheric-pressure chemical ionization (APCI) probe. The

t
ip
LC-MS/MS data was acquired by Analyst 1.7 software (Sciex, Framingham, MA, USA) and processed

cr
with MultiQuant 3.0 (Sciex, Framingham, MA, USA) using a list of 110 targeted oxidized lipid species.

us
An external standard calibration curve with eight points between 0.5 pg/µL to 1000 pg/µL

was used for absolute quantification of 12,13-diHOME. Relative quantification was calculated for all
an
other species based on the MS2 peak area ratio of the lipid species compared to its internal

standard.
M
d

Metabolomics Analysis
e

Samples for metabolomics analysis were prepared as previously described (10-14). Breast
pt

milk was thawed on ice, vortexed and further aliquoted. 200µL of breast milk was incubated with
ce

extraction solvent in a 1:2 sample volume to solvent volume ratio at -20°C for 30 minutes.

Metabolite extraction was achieved using a mixture of isopropanol, acetonitrile, and water at a ratio
Ac

of 3:3:2 v/v. Extracts were divided in to three parts: 75 uL for gas chromatography combined with

time-of-flight high-resolution mass spectrometry, 150 uL for reversed-phase liquid chromatography

coupled with high-resolution mass spectrometry, and 150 uL for hydrophilic interaction

chromatography with liquid chromatography and tandem mass-spectrometry, and analyzed as

previously described (14). We used the NEXERA XR UPLC system (Shimadzu, Columbia, MD, USA),

coupled with the Triple Quad 5500 System (AB Sciex, Framingham, MA, USA) to perform hydrophilic
interaction liquid chromatography analysis, NEXERA XR UPLC system (Shimadzu, Columbia, MD,

USA), coupled with the Triple TOF 6600 System (AB Sciex, Framingham, MA, USA) to perform

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


reversed-phase liquid chromatography analysis, and Agilent 7890B gas chromatograph (Agilent, Palo

Alto, CA, USA) interfaced to a Time-of-Flight Pegasus HT Mass Spectrometer (Leco, St. Joseph, MI,

USA). The GC system was fitted with a Gerstel temperature-programmed injector, cooled injection

system (model CIS 4). An automated liner exchange (ALEX) (Gerstel, Muhlheim an der Ruhr,

Germany) was used to eliminate cross-contamination from the sample matrix that was occurring

t
ip
between sample runs. Quality control was performed using metabolite standards mixture and

cr
pooled samples applying methodology preciously described (15-18). A quality control sample

containing a standard mixture of amino and organic acids purchased from Sigma-Aldrich as certified

us
reference material, was injected daily to perform analytical system suitability test and monitor
an
recorded signals day to day reproducibility as it was described (10-14). A pooled quality control

sample was obtained by taking an aliquot of the same volume of all samples from the study and
M

injected daily with a batch of analyzed samples to determine the optimal dilution of the batch

samples and validate metabolite identification and peak integration. Collected raw data was
d

manually inspected, merged, imputed and normalized by the sample median. Metabolite
e

identification was performed using in house authentic standards analysis. Metabolite annotation
pt

was used utilizing recorded retention time and retention indexes, recorded MSn and HRAMSn data
ce

matching with METLIN, NIST MS, Wiley Registry of Mass Spectral Data, HMDB, MassBank of North

America, MassBank Europe, Golm Metabolome Database, SCIEX Accurate Mass Metabolite Spectral
Ac

Library, MzCloud, and IDEOM databases.


Statistical Methods

Of the 58 mother-infant pairs participating in this study, 57 were included in the final

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


analysis. One pair was excluded due to implausible measurements and missing data. Tables 1 and 2

include details on the variables of interest included in the analysis. Excessive weight gain was

calculated based on the current Institute of Medicine recommendations on weight gain during

pregnancy based on pre-gestational BMI (19). Infant birth weight was classified as low birth weight

(LBW, <2,500 g), normal birth weight (2,500-4,000 g), or macrosomia (>4,000 g). We calculated

t
BMI Z-score using the zscorer program in RStudio based on World Health Organization (WHO) BMI

ip
for age reference ranges. Z-scores for weight-for-length (WFL), weight-for-age (WFA), and length-

cr
for-age (LFA) were calculated based on WHO 2006 growth charts.

Exploratory analyses by tertile


us
an
Given that there were no prior reports on 12,13-diHOME and related metabolites in breast

milk in relation to biologic outcomes, we examined relationships between tertiles of 12,13-diHOME


M

and demographic and clinical characteristics. We report the actual concentrations of 12,13-diHOME

in ng/mL for the tertiles and log transformed the area ratio for the statistical analyses. Our decision to
d

include specific infant and maternal characteristics in the modeling was based on prior literature of
e

human milk composition and infant weight status (20). We adjusted for maternal characteristics
pt

including pre-gravid BMI, gestational weight gain (GWG), and parity, and for infant sex and
ce

gestational age. Simple and multiple linear regressions were performed to examine the relationship

between infant BMI Z-scores at birth, modeling variables, and the log of the metabolites. We
Ac

additionally examined associations with the following metabolites and lipids in breast milk: AMP,

BAIBA, 12-HEPE, 12-HETE, uridine, Lyso-PG 18:0 and succinate. These metabolites and lipids

were selected a priori using a candidate approach based on recent publications documenting their

effects on adipocyte metabolism and/or thermogenesis (2,6,8,21-23). Finally, we also performed

principal component and partial least square analyses using MetaboAnalyst to examine associations

between infant adiposity and the breast milk lipidome (including 94 signaling lipids and 669 structural

lipids). Infants (n=57) were grouped into tertiles of adiposity at 1 month, with adiposity classified as
0 = tertile 1 (3.4-14.29 percent body fat), 1 = tertile 2 (14.3 – 19.09 percent), and 2 = tertile 3 (19.1 –

29 percent). Principal component analysis was also used to examine differences between the lipidome

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


of breast milk and two brands of commercial infant formula.

Analyses were performed using RStudio with a significance level of  = 0.05. P-values for the

ANOVA, Chi-Square, or Kruskal-Wallis tests for differences between the tertiles were included in

Tables 1 and 2. Results are presented as mean and standard deviation, statistical estimates, or

number and proportion.

t
ip
cr
RESULTS

us
Demographic and clinical characteristics according to milk 12,13-diHOME
an
We first examined maternal and infant characteristics according to abundance of 12,13-

diHOME in milk at 1-month postpartum. Milk 12,13-diHOME concentrations ranged from 0.30 ng/mL
M

to 53.72 ng/mL; tertiles 1, 2, and 3 included concentrations from 0.30 ng/mL to 3.50 ng/mL, 3.50

ng/mL to 7.38 ng/mL, and 7.38 ng/mL to 53.72 ng/mL, respectively. Milk 12,13-diHOME was
d

positively correlated with maternal height; the highest tertile for 12,13-diHOME abundance had the
e
pt

highest mean maternal height (166.6 cm in tertile 3, vs. 161.5 cm in tertile 1, p=0.017). There were

significant differences in milk glucose between the top and bottom tertiles of 12,13-diHOME (tertile
ce

3: 36.2 mg/dl vs. tertile 1: 26.18 mg/dl, p=0.009), which was not seen for other milk analytes
Ac

including leptin, insulin, Il-6, or CRP (Supplement Table 1, Supplement Figure 1A) (24). 12,13-

diHOME was not, however, associated with maternal age, parity, weight, pre-pregnancy BMI, or

gestational weight gain (Table 1). We also did not observe any seasonal variation in milk 12,13-

diHOME levels, nor did the levels differ between the study the Oklahoma and Minnesota study sites

(data not shown).


Abundance of 12,13-diHOME in breast milk was associated with several infant

anthropometric measures (Table 2). Higher milk 12,13-diHOME was associated with higher BMI Z-

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


score at birth (tertile 3: 0.32 vs. tertile 1: -0.60, p=0.006, Figure 1A) and WFL Z-score at birth (tertile

3: WFL Z-score 0.14 vs. tertile 1: -1.08, p=0.005). Milk abundance of 12,13-diHOME was not

associated with infant gestational age, birth weight or length, or sex. By 1-month postpartum,

higher milk abundance of 12,13-diHOME was associated with lower subcutaneous fat in the infant.

Triceps skin fold thickness (p=0.0192), thigh skin fold thickness (p=0.0147), and arm circumference

t
ip
(p=0.0401) showed significant differences between the tertiles with the same inverse pattern: as

cr
12,13-diHOME abundance increased (per tertile) the subcutaneous adiposity measures decreased

accordingly. Similarly, whole-body adiposity at 1-month postpartum (assessed by air displacement

us
plethysmography) tended to be reduced in the highest tertile of 12,13-diHOME (tertile 3: 15.32 %fat
an
vs. tertile 1: 18.61 %fat, p=0.077, Figure 1B). 12,13-diHOME was one of the few signaling lipids that

tracked with tertile of infant adiposity; principal components analysis of the 94 signaling lipids and
M

669 structural lipids did not reveal strong clustering (Supplement Figure 2) (24). Significant

differences in infant whole-body adiposity between the tertiles emerged after adjustments for infant
d

sex, gestational age, maternal pregravid BMI, gestational weight gain and parity (adjusted fat mass
e

in tertile 3: 14.52 %fat vs. tertile 1: 18.59 %fat, p=0.021).


pt

We also detected 12,13-diHOME in two brands of cow’s milk based commercial infant
ce

formula. Levels of 12,13-diHOME in these formulas was comparable to the upper tertile of the
Ac

concentrations found in human breast milk; principal component analysis suggests that the lipidome

of breast milk is distinct from that of infant formula (Supplement Figure 3) (24). We did not,

however, evaluate whether 12,13-diHOME levels in commercial formula was associated with infant

growth as breastfeeding mother and infant dyads were the primary focus of our study.
Human milk 12,13-diHOME is associated with WFL Z-score at birth independent of maternal and

infant characteristics

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


Abundance of 12,13-diHOME in milk at 1-month was positively associated with WFL Z-score

at birth (= 0.5742, p=0.0008). We next tested whether there was an independent association

between milk 12,13-diHOME abundance and WFL Z-score at birth, adjusting for maternal and infant

covariates. Milk 12,13-diHOME remained significantly associated with WFL Z-score at birth after

t
adjustment for maternal parity, pregravid BMI, and GWG (= 0.559, p=0.0018). The association was

ip
attenuated but remained significant upon further adjustment for infant sex, gestational age, and age

cr
in days at the 1-month visit (= 0.440, p= 0.0249). Thus, milk abundance of 12,13-diHOME is

us
independently associated with infant WFL Z-score at birth.
an
Milk 12,13-diHOME is associated with change in infant BMI and weight-for-length (WFL) from 0-6
M

months

We next asked whether abundance of 12,13-diHOME in milk was associated with gain in
d

adiposity, assessed by BMI and WFL Z-scores, during the early postnatal period. We observed a
e

significant negative association between log[12,13-diHOME] in milk and change in BMI Z-score over
pt

the first 6 months (= -0.400, p=0.0252, Figure 1C). Similarly, we observed a significant negative
ce

association between log[12,13-diHOME] in milk and change in WFL Z-score over the first 6-months

(= -0.534, p=0.0112, Figure 1F). This effect remained significant after adjustment for maternal
Ac

parity, pregravid BMI, and gestational weight gain (= -0.554, p=0.0131). The association between

milk 12,13-diHOME and change in WFL Z-score from 0-6 months also remained statistically

significant after additional adjustment for infant sex, gestational age, and age at the 1-month visit

(= -0.522, p=0.0374). However, further adjustment for 1-month milk glucose concentration

attenuated the effect (= -0.439, p=0.116).


Ephx1-4 metabolites are associated with infant weight status and adiposity

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


We next examined whether related metabolites were similarly associated with infant weight

status and adiposity. The enzymes Ephx1-4 synthesize the isomeric diols 12,13-diHOME and 9,10-

diHOME from linoleic acid via 12,13-epOME or 9,10-epOME by cytochrome P450 (Figure 2A) (4).

Thus, we examined associations between infant weight status and milk abundance of 12,13-epOME

t
and 9,10-diHOME. Both metabolites have significant, positive associations with the infant’s BMI Z-

ip
score at birth (Figure 2B, E). Log[12,13-diHOME] has the largest positive correlation with BMI Z-

cr
scores at birth (r=0.392, p=0.004), followed by log[9,10-diHOME] (r=0.376, p=0.007) and log[12,13-

us
epOME] (r=0.309, p=0.026). Log[9,10-diHOME] was positively correlated with 1-month breast milk

glucose concentration (Supplement Figure 1B) (24).


an
We next examined associations between 12,13-diHOME and related metabolites in milk at 1-
M

month postpartum with early infancy obesity. The significant inverse correlation between change in

WFL Z-score from 0-6 months and the metabolites was also seen with log[9,10-diHOME] (r= -0.307,
d

p=0.029, Figure 2D), but was weaker with log[12,13-epOME] (r= -0.261, p=0.064, Figure 2G).
e

Trajectories for the WFL Z-score measures at birth, 1 month, 3 months, and 6 months, by 12,13-
pt

diHOME tertile were created, along with weight for age Z-score and length for age Z-score (Figure
ce

1D-F). Similar growth trajectories were seen for 9,10-diHOME and 12,13-epOME (Figure 2C, F).
Ac

Associations of thermogenic metabolites in milk and infant weight status

Given these associations between milk abundance of 12,13-diHOME, related metabolites,

and infant BMI, we examined whether other signaling lipids and metabolites that have recently been

linked to brown adipose tissue activation (AMP, BAIBA, 12-HEPE, 12-HETE, uridine, Lyso-PG 18:0)

were associated with weight status in infancy (2,6,20-22). We analyzed the unadjusted correlation

between these metabolites and 1-month body fat percentage, of which only Lyso-PG 18:0 was
significantly associated. Log[Lyso-PG 18:0] was negatively associated with 1-month body fat

percentage (r= -0.311, p= 0.021, Supplement Figure 4A) (24). We next analyzed the unadjusted

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


linear association between these metabolites and 6-month BMI Z-score, of which only succinate was

significantly associated. We saw a negative linear association between succinate and 6-month BMI Z-

score (= -5.25, r= -0.301, p=0.028, Supplement Figure 4B) (24). We applied the same modeling as in

the 12,13-diHOME analysis to succinate and the significant association was retained when adjusting

for maternal characteristics (p=0.0416), but attenuated on adjustment for both infant and maternal

t
ip
characteristics (p=0.102). When we examined associations between the overall breast milk

cr
metabolome at 1-month postpartum and infant adiposity (% fat mass, assessed by ADP) in the

current study, we found that 26 milk metabolites were associated with infant adiposity at a nominal

us
P<0.05. We found that similar metabolite classes were affected as in our prior analyses (25).
an
Reproducing our prior findings, we again observed a positive correlation between breast milk purine

nucleotides (e.g. 1-methyladenosine, 7-methylguanine) and infant adiposity, which is intriguing in


M

light of data implicating purinergic receptors in the regulation of adipogenesis (Supplement Table 2)

(24, 26).
e d
pt

Association between 12,13-diHOME concentrations and acute exercise


ce

Plasma 12,13-diHOME levels differ between sedentary and active individuals, and have been

reported to be increased by acute exercise (4,5,7). We therefore tested, in a separate sample of 16


Ac

women for proof of concept, whether an acute bout of moderate exercise in the postpartum period

(4 weeks postpartum) may influence milk 12,13-diHOME abundance. Of the 16 participants, 8 were

normal weight and 8 were obese. We observed an increase in 12,13-diHOME breastmilk

concentrations in most subjects at 90-minutes post-acute exercise (Figure 3). On average, the total

sample saw a 1.39-fold increase in 12,13-diHOME concentrations at 90-minutes post-acute exercise


(paired T-test p = 0.0076), with obese women having a 1.73-fold increase and normal women having

a 1.50-fold increase.

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


DISCUSSION

In this analysis, we identify novel associations between levels of 12,13-diHOME and related

thermogenic metabolites in human breastmilk, and weight status and adiposity in infancy. We found

that 12,13-diHOME in milk was positively associated with BMI Z-score at birth, negatively associated

t
with fat mass and skinfold thicknesses at 1-month, and negatively associated with change in BMI

ip
from 0-6 months. Similar associations were seen for other metabolites in the same biosynthetic

cr
pathway, including 9,10-diHOME and 12,13-epOME. Maternal acute exercise may regulate 12,13-

us
diHOME abundance in milk, as evidenced by a significant increase in concentration of this lipid after

a bout of moderate exercise at 1 month postpartum. Moreover, we also observed an association


an
between milk levels of succinate, which has recently been linked to brown adipose tissue activation
M

in humans, and lower infant BMI at 6-months. We similarly observed an inverse association between

milk Lyso-PG 18:0 and infant body fat at 1-month. Together, our data suggest that milk levels of
d

brown fat activating metabolites may play a role in weight gain and adiposity in infancy.
e

Our analysis adds to the growing evidence that differences in human milk composition are
pt

associated with infant weight gain. Breastmilk is not only a source of nutrition, but also contains
ce

numerous non-nutritive bioactive compounds – including hormones, cytokines, nucleotides, cells,

microbiota, and exosomes – that may influence signaling and developmental processes in the infant
Ac

and may contribute to obesity risk. Breastfeeding provides many health benefits for mother and

child, but there is some controversy regarding effects on childhood obesity risk (27-29). In a meta-

analysis of formula versus breast feeding, a small protective effect was found on adult BMI (30).

However, in many studies the associations between breastfeeding and obesity in children are

weakened when adjusting for maternal education, socioeconomic status, obesity, and smoking (31).

Moreover, a randomized control trial of a breastfeeding promotion intervention in Belarus found a


slightly increased adolescent BMI in the intervention group (32). These inconsistencies could be

explained by the variation of certain constituents in milk composition between individuals.

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


Consistent with this possibility, untargeted metabolomics analyses in breastmilk have recently

identified associations among maternal obesity, longitudinal changes in the milk metabolome, and

fat accrual during infancy (25,33). We previously reported that abundance of human milk

nucleotides and oligosaccharides is related to maternal and infant BMI and infant fat mass, and that

milk insulin and leptin are inversely associated with infant weight gain (25,34). Based on the current

t
ip
analysis, we propose that breastmilk 12,13-diHOME, Lyso-PG 18:0, and succinate may similarly have

cr
protective effects on obesity in offspring. Recent analyses by other groups have also described

associations between breast milk content of three dihomo-γ-linolenic acid (DGLA)-derived oxylipins

us
(15S-HpEDE and two deoxy-dimethyl-PGE2) and faster growth of preterm infants (35), suggesting an
an
important influence of the breast milk lipidome on postnatal growth trajectories.

We are not aware of prior reports of 12,13-diHOME in human milk, nor of its association
M

with infant weight gain or adiposity. Prior studies have shown that 12,13-diHOME is increased in

plasma following cold exposure and acute exercise, and that 12,13-diHOME increases fatty acid
e d

uptake via FATP1 and CD36 in brown adipocytes (4). Beyond 12,13-diHOME and other metabolites,
pt

additional stimuli such as cold exposure, norepinephrine, FGF21, and cytokines have been described

to induce “beige-ing”, or the acquisition of a brown-like phenotype (i.e., upregulation of


ce

thermogenesis, mitochondrial biogenesis) by white adipocytes in adults. By contrast, infants are


Ac

born with a predominance of beige-like cells in their adipose depots, which are progressively

replaced with white adipocytes (36); the extent to which beige adipocytes are replaced with white

adipocytes is associated with childhood obesity risk (37). However, it is unclear to what extent 12,13-

diHOME may play a role in adipose tissue remodeling in infancy. This signaling lipid has, however,

been reported in infant feces, where it is produced by the gut microbiota, decreases numbers of T-

regulatory cells and may increase risk of asthma and atopic diseases (9). (We did not have access to

information about asthma diagnoses in the infants in our study.) Based on these data, it is unclear
whether observed associations between higher 12,13-diHOME and lower BMI gain might reflect

increased inflammation/atopy versus changes in adipocyte function. Some emerging data suggest

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


that thermogenesis and inflammation may be closely interrelated processes: for example, short-

term cold exposure in adults induces CD4 T-cell differentiation into anti-inflammatory T-regulator

cells (38) and cytokines such as TGF-beta and IL-6 may activate adipocyte thermogenesis (39, 40).

On the other hand, pro-inflammatory cytokine secretion and macrophage infiltration may also

impair the development of beige adipocytes (41) and increased expression of immune mediators

t
ip
(e.g. IL-1b, TNF-a, and MCP-1) in brown fat inhibits UCP1 expression and activity (42-44).

cr
It is unclear to what extent 12,13-diHOME levels in breast milk reflect the infant’s exposure

us
to this lipid in early life. Beyond breast milk and the gut microbiome, there are several other

potential sources of exposure to 12,13-diHOME in the infant, including cow’s milk (45), infant
an
formulas (Supplement Figure 3) (24), and endogenous synthesis by leukocytes and other tissues that

express Ephx 1-4. Vernix caseosa, the creamy protective film that covers the newborn’s skin at birth,
M

has also been reported to contain 12,13-diHOME and other oxylipins: levels of 12,13-diHOME (and

related oxylipins 12,13-epOME and 9,10-epOME) in vernix were associated with maternal
e d

“anthroposophic” lifestyle (e.g. vegetarian or organic diet, home birth) (4,46). Future studies
pt

examining associations between infant adiposity and 12,13-diHOME in milk, in maternal and infant

plasma, and in tissues, will be essential.


ce

Although we identified associations between 12,13-diHOME in milk and infant adiposity, we


Ac

did not directly test whether this oxylipin play a causal role in adipogenesis or adipose tissue

development. There are no prior reports on 12,13-diHOME’s role in adipocyte differentiation.

However, the related oxylipin 9,10-diHOME has been shown to activate PPAR-gamma, the

transcriptional “master regulator” of adipogenesis, and to promote accumulation of lipid droplets in

a mesenchymal cell line (U-33/γ2 cells), while inhibiting osteoblast differentiation, suggesting that

lipid metabolites in this pathway could play a direct in adipocyte differentiation (47). Based on the
observation that 12,13-diHOME increased fatty acid uptake and upregulated the fatty transporters

FATP1 and CD36 to the cell membrane in brown adipocytes (4), one might speculate that 12,13-

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


diHOME could similarly promote fatty acid uptake and lipid accumulation during the final stages of

adipogenesis. On the other hand, expression and activity of Ephx2 or soluble epoxide hydrolase

(sEH), one of the enzymes that can synthesize 12,13-diHOME, is upregulated during adipogenesis

(48, 49), and inhibition of sEH using a chemical inhibitor (AUDA, 12-(3-adamantan-1-yl-ureido)-

dodecanoic acid) was reported to decrease adipogenesis in mesenchymal stem cells (50). Moreover,

t
ip
inhibition of sEH using AUDA or siRNA in mature white adipocytes resulted in a decrease in

cr
adipocyte size, increased adiponectin expression, and upregulation of mitochondrial and

thermogenic genes, consistent with a shift to a “beige-like” phenotype (51). Thus, it remains unclear

us
whether exposure to 12,13-diHOME or other products of epoxide hydrolase would have a net
an
positive or negative effect on adipogenesis in infancy. Additional experiments using animal models

and/or cell lines will be essential to test what role 12,13-diHOME may play in adipocyte
M

differentiation.

It is also unclear based on our data to what extent differences in maternal adipose tissue
e d

metabolism may account for the observed association between 12,13-diHOME in milk and infant
pt

adiposity. Plasma levels of 12,13-diHOME are thought to be increased with greater adipose tissue

lipolysis, as they have been reported correlate closely with plasma non-esterified fatty acid levels
ce

(NEFA), and with plasma beta-hydroxybutyrate, reflecting greater availability of NEFA for hepatic
Ac

fatty acid oxidation (8). Our observation that a bout of moderate exercise results in increased 12,13-

diHOME in milk would support this possibility, as exercise is a stimulus for lipolysis (52). Thus, the

association between higher milk 12,13-diHOME and lower infant adiposity could reflect increased

maternal lipolysis and an inherited increased tendency for lipolysis in the infant. Alternatively, given

the observation that plasma 12,13-diHOME levels are higher in active vs. sedentary individuals (5),

another possibility is that maternal fitness level differs between women with high and low milk

12,13-diHOME levels, and that maternal cardiovascular fitness might underlie the association
between milk 12,13-diHOME and infant adiposity. Indeed, a recent study reported that maternal

activity level, assessed by weekly step counts, was associated with breastmilk abundance of a

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


specific human milk oligosaccharide (3’-sialyllactose) that conferred protection from high-fat diet

induced obesity in offspring mice (53). However, causal relationships between maternal exercise or

lipolysis, breastmilk 12,13-diHOME, and infant metabolic health, will need to be established in future

studies using in vitro systems or experimental models.

t
Another key finding from this analysis was a novel association between higher milk succinate

ip
levels and lower infant BMI at age 6-months. Succinate was reported to be altered in milk from

cr
mothers with inflammatory bowel disease (versus healthy controls) (54). Levels of succinate in

us
neonatal fecal samples were also reported to be modulated by milk fat globule membrane (MFGM)

(55) and by sialylated milk oligosaccharides (56). Increased fecal succinate levels induced TH2
an
immune responses in intestinal cells and was associated with changes in bone formation (56).
M

Some strengths of this analysis are that this is the first study to do a comprehensive analysis

of 12,13-diHOME in milk, in association with multiple infant adiposity measurements over time.
d

Moreover, we were able to assess other milk components in the same biosynthetic pathway, as well
e

as additional metabolites recently linked to brown adipose tissue function. We also identify a
pt

potential role of acute exercise as a regulator of 12,13-diHOME abundance in milk. We acknowledge


ce

several limitations in this analysis. First, we did not determine whether 12,13-diHOME directly

affects infant adipogenesis, nor did we examine potential influences of maternal adipocyte
Ac

metabolism on milk composition. Second, our sample size was relatively small, limiting the power.

Our findings will thus need to be replicated in independent populations. Third, our analyses are

cross-sectional and cannot be used to infer causality. Longitudinal studies would be important to

determine whether breastmilk 12,13-diHOME or other lipokines may program contribute to future

childhood obesity, metabolic syndrome, neurologic outcomes (57), or atopy. Fourth, we did not

assess levels of additional breastmilk constituents that may influence brown adipose tissue function,
such as alkylglycerol lipids (1) and hormones including FGF21 and irisin (58). Finally, we did not

measure the plasma levels of the metabolites that were measured in breast milk, and thus cannot

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


infer whether these substances might be preferentially secreted into milk, or whether they simply

reflect levels in the mother’s circulation. These will be important areas for future studies.

In conclusion, 12,13-diHOME and its metabolites show significant associations with infant

adiposity. Higher 12,13-diHOME concentration in milk was associated with higher BMI Z-score at

t
birth, but lower subcutaneous and overall adiposity at 1 month, and lower gains in WFL and BMI Z-

ip
scores from 0-6 months. Milk 12,13-diHOME was significantly increased following a maternal

cr
exercise bout. Further research is needed to determine whether milk 12,13-diHOME may play a

us
causative role in infant weight gain or adipocyte metabolism.
an
M
e d
pt
ce
Ac
Acknowledgements

The author’s responsibilities were as follows – DAF and EWD: designed the study and led the

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


clinical assessments; EI: designed the analysis, interpreted the data, and wrote the manuscript; DW:

analyzed the data, created the figures and table, and assisted in writing the manuscript; MDL:

interpreted data, advised on study design, and assisted with preparation of figures; YT: interpreted

data and advised on study design; MR and SP: assisted with the analysis of maternal exercise; VB,

t
AD, VT, NN, and MAK: performed metabolomics and lipidomics assays.

ip
cr
Data Availability Statement

us
Some data generated or analyzed during this study are included in this published article, and
an
in Supplemental Figures and Tables that can be accessed through the data repositories listed in

References. The full datasets generated during the current study are not publicly available, but are
M

available from the corresponding author on reasonable request.


e d
pt
ce
Ac
Reference List

1. Yu H, Dilbaz S, Coßmann J, et al. Breast milk alkylglycerols sustain beige adipocytes through

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


adipose tissue macrophages. J Clin Invest. 2019;129(6):2485-2499. doi:10.1172/JCI125646

2. Leiria LO, Wang C-H, Lynes MD, et al. 12-Lipoxygenase Regulates Cold Adaptation and
Glucose Metabolism by Producing the Omega-3 Lipid 12-HEPE from Brown Fat. Cell Metab.
2019;30(4):768-783.e7. doi:10.1016/j.cmet.2019.07.001

3. Cao H, Gerhold K, Mayers JR, Wiest MM, Watkins SM, Hotamisligil GS. Identification of a
Lipokine, a Lipid Hormone Linking Adipose Tissue to Systemic Metabolism. Cell. 2008;134(6):933-
944. doi:10.1016/j.cell.2008.07.048

t
ip
4. Lynes MD, Leiria LO, Lundh M, et al. The cold-induced lipokine 12,13-diHOME promotes fatty
acid transport into brown adipose tissue. Nat Med. 2017;23(5):631-637. doi:10.1038/nm.4297

cr
5. Stanford KI, Lynes MD, Takahashi H, et al. 12,13-diHOME: An Exercise-Induced Lipokine that
Increases Skeletal Muscle Fatty Acid Uptake. Cell Metab. 2018;27(5):1111-1120.e3.
doi:10.1016/j.cmet.2018.03.020

6. us
Kulterer OC, Niederstaetter L, Herz CT, et al. The Presence of Active Brown Adipose Tissue
an
Determines Cold-Induced Energy Expenditure and Oxylipin Profiles in Humans. J Clin Endocrinol
Metab. 2020;105(7):2203-2216. doi:10.1210/clinem/dgaa183
M

7. Nayor M, Shah RV, Miller PE, et al. Metabolic Architecture of Acute Exercise Response in
Middle-Aged Adults in the Community. Circulation. 2020 Sep 15.
d

8. Vasan SK, Noordam R, Gowri MS, Neville MJ, Karpe F, Christodoulides C. The proposed systemic
e

thermogenic metabolites succinate and 12,13-diHOME are inversely associated with adiposity and
pt

related metabolic traits: evidence from a large human cross-sectional study. Diabetologia.
2019;62(11):2079-2087. doi:10.1007/s00125-019-4947-5
ce

9. Levan SR, Stamnes KA, Lin DL, et al. Elevated faecal 12,13-diHOME concentration in
neonates at high risk for asthma is produced by gut bacteria and impedes immune tolerance. Nat
Microbiol. 2019;4(11):1851-1861. doi:10.1038/s41564-019-0498-2
Ac

10. Tolstikov V, Nikolayev A, Dong S, Zhao G, Kuo M-S. Metabolomics Analysis of Metabolic
Effects of Nicotinamide Phosphoribosyltransferase (NAMPT) Inhibition on Human Cancer Cells.
Campos-Olivas R, ed. PLoS ONE. 2014;9(12):e114019. doi:10.1371/journal.pone.0114019

11. Gacias M, Gaspari S, Santos P-MG, et al. Microbiota-driven transcriptional changes in


prefrontal cortex override genetic differences in social behavior. eLife. 2016;5:e13442.
doi:10.7554/eLife.13442

12. Jeremy Drolet, Vladimir Tolstikov, Brian Williams, et al. Integrated Metabolomics
Assessment of Human Dried Blood Spots and Urine Strips. Metabolites. 2017;7(3):35.
doi:10.3390/metabo7030035
13. Baskin AS, Linderman JD, Brychta RJ, et al. Regulation of Human Adipose Tissue Activation,
Gallbladder Size, and Bile Acid Metabolism by a β3-Adrenergic Receptor Agonist. Diabetes.
2018;67(10):2113-2125. doi:10.2337/db18-0462

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


14. Kiebish MA, Cullen J, Mishra P, et al. Multi-omic serum biomarkers for prognosis of disease
progression in prostate cancer. J Transl Med. 2020;18(1):10. doi:10.1186/s12967-019-02185-y

15. The Human Serum Metabolome (HUSERMET) Consortium, Dunn WB, Broadhurst D, et al.
Procedures for large-scale metabolic profiling of serum and plasma using gas chromatography and
liquid chromatography coupled to mass spectrometry. Nat Protoc. 2011;6(7):1060-1083.
doi:10.1038/nprot.2011.335

16. Bajad S, Shulaev V. LC-MS-Based Metabolomics. In: Metz TO, ed. Metabolic Profiling. Vol

t
ip
708. Methods in Molecular Biology. Humana Press; 2011:213-228. doi:10.1007/978-1-61737-985-
7_13

cr
17. Yuan M, Breitkopf SB, Yang X, Asara JM. A positive/negative ion–switching, targeted mass
spectrometry–based metabolomics platform for bodily fluids, cells, and fresh and fixed tissue. Nat

us
Protoc. 2012;7(5):872-881. doi:10.1038/nprot.2012.024

18. Want EJ, Masson P, Michopoulos F, et al. Global metabolic profiling of animal and human
an
tissues via UPLC-MS. Nat Protoc. 2013;8(1):17-32. doi:10.1038/nprot.2012.135

19. Weight Gain During Pregnancy. Am Coll Obstet Gynecol. 2013;(Committee Opinion No.
548):121:210-2.
M

20. Alderete TL, Autran C, Brekke BE, et al. Associations between human milk oligosaccharides
and infant body composition in the first 6 mo of life. Am J Clin Nutr. 2015;102(6):1381-1388.
d

doi:10.3945/ajcn.115.115451
e

21. Sanchez-Delgado G, Martinez-Tellez B, Olza J, Aguilera CM, Gil Á, Ruiz JR. Role of Exercise in
the Activation of Brown Adipose Tissue. Ann Nutr Metab. 2015;67(1):21-32. doi:10.1159/000437173
pt

22. Deng Y, Wang ZV, Gordillo R, et al. An adipo-biliary-uridine axis that regulates energy
ce

homeostasis. Science. 2017;355(6330):eaaf5375. doi:10.1126/science.aaf5375

23. Lynes MD, Shamsi F, Sustarsic EG, et al. Cold-Activated Lipid Dynamics in Adipose Tissue
Ac

Highlights a Role for Cardiolipin in Thermogenic Metabolism. Cell Rep. 2018;24(3):781-790.


doi:10.1016/j.celrep.2018.06.073

24. Brown fat-activating lipokine 12,13-diHOME in human milk is associated with infant adiposity;
supplemental tables and figures. Figshare website. September 29, 2020.
https://figshare.com/s/375e0dd631b5953bf667

25. Isganaitis E, Venditti S, Matthews TJ, Lerin C, Demerath EW, Fields DA. Maternal obesity and
the human milk metabolome: associations with infant body composition and postnatal weight gain.
Am J Clin Nutr. 2019;110(1):111-120. doi:10.1093/ajcn/nqy334
26. Tozzi M, Novak I. Purinergic Receptors in Adipose Tissue As Potential Targets in Metabolic
Disorders. Front Pharmacol. 2017 Nov 24;8:878. doi: 10.3389/fphar.2017.00878. PMID:
29249968; PMCID: PMC5715378.

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


27. Fischer J, Koch L, Emmerling C, et al. Inactivation of the Fto gene protects from obesity. Nature.
2009;458(7240):894-898. doi:10.1038/nature07848

28. Zielke LG, Bortfeldt RH, Reissmann M, Tetens J, Thaller G, Brockmann GA. Impact of
Variation at the FTO Locus on Milk Fat Yield in Holstein Dairy Cattle. Grant SFA, ed. PLoS ONE.
2013;8(5):e63406. doi:10.1371/journal.pone.0063406

t
29. Ibeagha-Awemu EM, Peters SO, Akwanji KA, Imumorin IG, Zhao X. High density genome wide

ip
genotyping-by-sequencing and association identifies common and low frequency SNPs, and novel
candidate genes influencing cow milk traits. Sci Rep. 2016;6(1):31109. doi:10.1038/srep31109

cr
30. Haran V, Lenka N. Deciphering the Epitranscriptomic Signatures in Cell Fate Determination

us
and Development. Stem Cell Rev Rep. 2019;15(4):474-496. doi:10.1007/s12015-019-09894-3

31. Hales CM, Fryar CD, Carroll MD, Freedman DS, Ogden CL. Trends in Obesity and Severe
Obesity Prevalence in US Youth and Adults by Sex and Age, 2007-2008 to 2015-2016. JAMA.
an
2018;319(16):1723. doi:10.1001/jama.2018.3060

32. Martin RM, Kramer MS, Patel R, et al. Effects of Promoting Long-term, Exclusive
M

Breastfeeding on Adolescent Adiposity, Blood Pressure, and Growth Trajectories: A Secondary


Analysis of a Randomized Clinical Trial. JAMA Pediatr. 2017;171(7):e170698.
doi:10.1001/jamapediatrics.2017.0698
d

33. Saben JL, Sims CR, Piccolo BD, Andres A. Maternal adiposity alters the human milk
e

metabolome: associations between nonglucose monosaccharides and infant adiposity. Am J Clin


Nutr. Published online August 25, 2020:nqaa216. doi:10.1093/ajcn/nqaa216
pt

34. Fields DA, Demerath EW. Relationship of insulin, glucose, leptin, IL-6 and TNF-α in human
ce

breast milk with infant growth and body composition: Analytes in human breast-milk. Pediatr Obes.
2012;7(4):304-312. doi:10.1111/j.2047-6310.2012.00059.x
Ac

35. Alexandre-Gouabau M-C, Moyon T, Cariou V, et al. Breast Milk Lipidome Is Associated with
Early Growth Trajectory in Preterm Infants. Nutrients. 2018;10(2):164. doi:10.3390/nu10020164

36. Ikeda K, Maretich P, Kajimura S. The Common and Distinct Features of Brown and Beige
Adipocytes. Trends Endocrinol Metab. 2018;29(3):191-200. doi:10.1016/j.tem.2018.01.001

37. Rockstroh D, Landgraf K, Wagner IV, et al. Direct Evidence of Brown Adipocytes in Different
Fat Depots in Children. Pfeifer A, ed. PLOS ONE. 2015;10(2):e0117841.
doi:10.1371/journal.pone.0117841

38. Becker M, Serr I, Salb VK, et al. Short-term cold exposure supports human Treg induction in
vivo. Mol Metab. 2019;28:73-82. doi:10.1016/j.molmet.2019.08.002
39. Mishra D, Richard JE, Maric I, et al. Parabrachial Interleukin-6 Reduces Body Weight and
Food Intake and Increases Thermogenesis to Regulate Energy Metabolism. Cell Rep.
2019;26(11):3011-3026.e5. doi:10.1016/j.celrep.2019.02.044

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


40. Babaei R, Schuster M, Meln I, et al. Jak-TGFβ cross-talk links transient adipose tissue
inflammation to beige adipogenesis. Sci Signal. 2018;11(527):eaai7838.
doi:10.1126/scisignal.aai7838

41. Estève D, Boulet N, Volat F, et al. Human White and Brite Adipogenesis is Supported by
MSCA1 and is Impaired by Immune Cells: MSCA1 is a Player of White/Brite Adipogenesis. STEM
CELLS. 2015;33(4):1277-1291. doi:10.1002/stem.1916

42. Alcalá M, Calderon-Dominguez M, Bustos E, et al. Increased inflammation, oxidative stress

t
ip
and mitochondrial respiration in brown adipose tissue from obese mice. Sci Rep. 2017;7(1):16082.
doi:10.1038/s41598-017-16463-6

cr
43. Martins FF, Bargut TCL, Aguila MB, Mandarim-de-Lacerda CA. Thermogenesis, fatty acid
synthesis with oxidation, and inflammation in the brown adipose tissue of ob/ob (−/−) mice. Ann

us
Anat - Anat Anz. 2017;210:44-51. doi:10.1016/j.aanat.2016.11.013

44. Goto T, Naknukool S, Yoshitake R, et al. Proinflammatory cytokine interleukin-1β suppresses


an
cold-induced thermogenesis in adipocytes. Cytokine. 2016;77:107-114.
doi:10.1016/j.cyto.2015.11.001

45. Kuhn MJ, Mavangira V, Gandy JC, Zhang C, Jones AD, Sordillo LM. Differences in the Oxylipid
M

Profiles of Bovine Milk and Plasma at Different Stages of Lactation. J Agric Food Chem.
2017;65(24):4980-4988. doi:10.1021/acs.jafc.7b01602
d

46. Checa A, Holm T, Sjödin MOD, et al. Lipid mediator profile in vernix caseosa reflects skin
barrier development. Sci Rep. 2015;5(1):15740. doi:10.1038/srep15740
e

47. Lecka-Czernik B, Moerman EJ, Grant DF, Lehmann JM, Manolagas SC, Jilka RL. Divergent
pt

Effects of Selective Peroxisome Proliferator-Activated Receptor-γ2 Ligands on Adipocyte Versus


Osteoblast Differentiation. Endocrinology. 2002;143(6):2376-2384. doi:10.1210/endo.143.6.8834
ce

48. De Taeye BM, Morisseau C, Coyle J, et al. Expression and Regulation of Soluble Epoxide
Hydrolase in Adipose Tissue. Obesity. 2010;18(3):489-498. doi:10.1038/oby.2009.227
Ac

49. Zha W, Edin ML, Vendrov KC, et al. Functional characterization of cytochrome P450-derived
epoxyeicosatrienoic acids in adipogenesis and obesity. J Lipid Res. 2014;55(10):2124-2136.
doi:10.1194/jlr.M053199

50. Kim DH, Vanella L, Inoue K, et al. Epoxyeicosatrienoic Acid Agonist Regulates Human
Mesenchymal Stem Cell–Derived Adipocytes Through Activation of HO-1-pAKT Signaling and a
Decrease in PPARγ. Stem Cells Dev. 2010;19(12):1863-1873. doi:10.1089/scd.2010.0098

51. Liu L, Puri N, Raffaele M, et al. Ablation of soluble epoxide hydrolase reprogram white fat to
beige-like fat through an increase in mitochondrial integrity, HO-1-adiponectin in vitro and in vivo.
Prostaglandins Other Lipid Mediat. 2018;138:1-8. doi:10.1016/j.prostaglandins.2018.07.004
52. Laurens C, Parmar A, Murphy E, Carper D, et al. Growth and differentiation factor 15 is
secreted by skeletal muscle during exercise and promotes lipolysis in humans. JCI Insight. 2020 Mar
26;5(6):e131870.)

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


53. Harris JE, Pinckard KM, Wright KR et al. Exercise-induced 3′-sialyllactose in breast milk is a
critical mediator to improve metabolic health and cardiac function in mouse offspring. Nat
Metab 2, 678–687 (2020).

54. Meng X, Dunsmore G, Koleva P, et al. The Profile of Human Milk Metabolome, Cytokines, and
Antibodies in Inflammatory Bowel Diseases Versus Healthy Mothers, and Potential Impact on the
Newborn. J Crohns Colitis. 2019;13(4):431-441. doi:10.1093/ecco-jcc/jjy186

55. He X, Parenti M, Grip T, et al. Fecal microbiome and metabolome of infants fed bovine MFGM

t
ip
supplemented formula or standard formula with breast-fed infants as reference: a randomized
controlled trial. Sci Rep. 2019;9(1):11589. doi:10.1038/s41598-019-47953-4

cr
56. Cowardin CA, Ahern PP, Kung VL, et al. Mechanisms by which sialylated milk
oligosaccharides impact bone biology in a gnotobiotic mouse model of infant undernutrition. Proc

us
Natl Acad Sci. Published online May 28, 2019:201821770. doi:10.1073/pnas.1821770116

57. Frondas-Chauty A, Simon L, Flamant C, Hanf M, Darmaun D, Rozé J-C. Deficit of Fat Free
an
Mass in Very Preterm Infants at Discharge is Associated with Neurological Impairment at Age 2
Years. J Pediatr. 2018;196:301-304. doi:10.1016/j.jpeds.2017.12.017

58. Lee P, Linderman JD, Smith S, et al. Irisin and FGF21 Are Cold-Induced Endocrine Activators
M

of Brown Fat Function in Humans. Cell Metab. 2014;19(2):302-309. doi:10.1016/j.cmet.2013.12.017


e d
pt
ce
Ac
Table 1. Maternal and Infant Characteristics.

Maternal and infant characteristics by 12,13-diHOME tertile. P-values refer to ANOVA (continuous

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


variables) or Chi-Square tests (categorical variables) for differences between the tertiles. Mean ± SD

for continuous variables or n (%) for categorical.

Overall N Tertile 1 Tertile 2 Tertile 3 P-Value

Maternal Age, yr 30.39 ± 4.36 56 29.84 ± 4.60 31.50 ± 4.77 30.17 ± 3.59 0.808

t
ip
Maternal Height, 164.69 ± 161.46 ± 166.62 ±

cr
57 165.80 ± 7.50 0.017
cm 6.67 5.58 5.95

Maternal

kg
Weight, 71.07

13.82
±
57
66.69

11.87
±

us
75.08 ± 14.80
69.36

11.00
±
0.516
an
Maternal Pregravid
M

26.21 ± 4.73 57 25.55 ± 4.25 27.29 ± 4.77 25.12 ± 4.42 0.298


2a
BMI, kg/m
d

Maternal GWG, kg 12.65 ± 7.13 57 11.81 ± 4.24 12.68 ± 9.52 13.47 ± 7.15 0.489
e

Excessive Weight
pt

- 21 8 (38.1) 5 (23.8) 8 (38.1)


Gain, kg
ce

Maternal Change in
Ac

weight from Birth -8.57 ± 6.02 55 -9.50 ± 3.48 -8.26 ± 9.44 -7.89 ± 3.40 0.278

to 1moa

Maternal Parity: - 57 - - - 0.652

0 30 (52.6) 10 (52.6) 9 (47.4) 10 (55.5)


1 19 (33.3) 7 (36.8) 6 (31.6) 6 (33.3)

2 7 (12.3) 2 (10.6) 4 (21.0) 1 (5.6)

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


3 1 (1.8) 0 (0.0) 0 (0.0) 1 (5.6)

Household Income - 43 - - - 0.260

1 4 (9.3) 3 (16.7) 0 (0.0) 1 (8.3)

t
ip
2 8 (18.6) 4 (22.2) 2 (15.4) 2 (16.7)

cr
3 13 (30.2) 7 (38.9) 4 (30.8) 2 (16.7)

4 11 (25.6) 1 (5.5)

us
6 (46.1) 4 (33.3)
an
5 7 (16.3) 3 (16.7) 1 (7.7) 3 (25.0)

Maternal Education - 43 - - - 0.667


M

2 4 (9.3) 3 (16.7) 1 (7.6) 0 (0.0)


d

3 5 (11.6) 2 (11.1) 2 (15.4) 1 (8.3)


e
pt

4 18 (41.9) 8 (44.4) 5 (38.5) 5 (41.7)


ce

5 13 (30.2) 3 (16.7) 5 (38.5) 5 (41.7)


Ac

6 3 (7.0) 2 (11.1) 0 (0.0) 1 (8.3)

Infant Sex: - 56 - - - 0.401

Female 24 (0.43) 9 (47.4) 6 (31.6) 9 (52.9)

Male 32 (0.57) 10 (52.6) 13 (68.4) 8 (47.1)


Birth Weight, ga 3577 ± 423 57 3435 ± 423 3667 ± 427 3653 ± 398 0.214

LGA 9 (0.16) 2 (0.04) 4 (0.07) 3 (0.05)

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


Gestational Age, 39.74 ± 1.23 57 39.51 ± 1.42 39.56 ± 1.28 40.20 ± 0.90 0.220

wka

Birth Length, cma 51.52 ± 2.11 54 51.61 ± 2.19 51.94 ± 2.12 51.12 ± 2.06 0.593

t
Footnotes:

ip
a
Kruskal-Wallis Test

cr
us
an
M
e d
pt
ce
Ac
Table 2. Infant Body Composition.

Infant body composition by 12,13-diHOME tertile. P-values refer to ANOVA (continuous variables)

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


or Chi-Square tests (categorical variables) for differences between the tertiles. Mean ± SD for

continuous variables or n (%) for categorical.

Overall N Tertile 1 Tertile 2 Tertile 3 P-Value

t
ip
-0.60 ± 0.01 ± 0.32 ±
Birth BMI Z-Score -0.09 ± 0.99 57 0.006

cr
1.18 0.81 0.81

us
-0.04 ± 0.21 ±
1mo BMI Z-Score 0.21 ± 1.08 56 0.59 ± 0.97 0.256
1.06 1.04
an
0.16 ± 0.31 ±
6mo BMI Z-Score 0.26 ± 0.94 54 0.37 ± 0.84 0.827
M

0.95 1.08

-1.08 ± -0.47 ± 0.14 ±


d

Birth WFL Z-Score -0.46 ± 1.32 54 0.005


1.52 1.06 1.15
e
pt

-0.14 ± 0.14 ±
1mo WFL Z-Score 0.16 ± 1.13 57 0.63 ± 0.99 0.164
ce

0.99 1.20

-0.15 ± -0.02 ±
Ac

3mo WFL Z-Scorea -0.04 ± 0.98 56 0.10 ± 0.91 0.672


0.85 1.21

0.25 ± 0.44 ±
6mo WFL Z-Score 0.36 ± 0.93 55 0.46 ± 0.83 0.929
0.93 1.06

1mo ADP Fat % 16.74 ± 56 18.61 ± 16.49 ± 15.32 ± 0.077


5.62 4.83 5.76 5.98

0.77 ± 0.71 ±

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


1mo ADP Fat Mass (kg) 0.78 ± 0.32 56 0.89 ± 0.31 0.079
0.31 0.32

1mo ADP Fat Free Mass 3.80 ± 3.83 ±


3.81 ± 0.42 56 3.83 ± 0.46 0.993
(kg) 0.43 0.38

34.38 ± 34.24 ± 34.77 ± 34.39 ±

t
6mo DXA Fat % 54 0.886

ip
3.32 3.06 3.27 3.66

cr
2.86 ± 2.82 ±
6mo DXA Fat Mass 2.86 ± 0.53 54 2.93 ±0.55 0.549

us
0.47 0.61

5.33 ± 5.32 ±
an
6mo DXA Fat Free Mass 5.41 ± 0.53 54 5.56 ± 0.46 0.173
0.38 0.71
M

Triceps Skin Fold - - - - - -


d

6.36 ± 6.33 ±
1 month 7.08 ± 2.28 44 8.16 ± 2.45 0.019
e

2.28 1.43
pt

7.96 ± 8.52 ±
ce

3 month 8.08 ± 2.39 55 7.77 ± 2.35 0.354


2.32 2.57
Ac

10.11 ± 9.45 ±
6 month 9.59 ± 2.72 55 9.17 ± 3.41 0.76
2.38 2.31

Thigh Skin Fold

11.69 ± 12.90 ± 11.79 ± 9.90 ±


1 month 44 0.015
3.41 3.27 4.06 2.08
3 month 16.71 ± 17.03 ± 17.20 ± 15.86 ±
55 0.403
4.14 4.21 4.03 4.30

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


6 month 20.50 ± 20.94 ± 20.60 ± 19.94 ±
53 0.551
4.85 5.02 5.21 4.49

Abdominal Skin Fold - - - - - -

1 month 6.79 ± 6.67 ±

t
7.13 ± 2.01 44 7.71 ± 2.20 0.139

ip
2.25 1.28

cr
3 month 8.94 ± 9.45 ±
9.19 ± 2.81 55 9.19 ± 2.78 0.783

us
2.72 3.06

9.57 ± 9.27 ±
an
6 month
9.56 ± 2.57 55 9.86 ± 2.83 0.497
2.61 2.37
M

Abdominal Circumference - - - - - -
d

1 month 39.11 ± 39.83 ± 39.36 ± 37.90 ±


43 0.061
e

2.75 2.69 3.26 1.93


pt

3 month 41.95 ± 42.45 ± 41.81 ± 41.60 ±


ce

55 0.426
3.16 3.21 3.55 2.75
Ac

6 month 43.51 ± 44.34 ± 43.32 ± 42.88 ±


55 0.143
2.97 3.22 2.72 2.95

Arm Circumference - - - - - -

1 month 12.16 ± 12.54 ± 12.17 ± 11.65 ±


43 0.040
1.18 1.22 1.39 0.69
Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020
0.255

0.086

t
ip
±

cr
± 13.60

± 14.58
1.68

1.48

us
± 13.50

± 14.72
1.21

1.26

an
14.15

15.31
1.37

0.97

M
55

55

d
±

e
pt
13.74

14.87
1.43

1.27
ce
Kruskal-Wallis test
Ac
Footnotes:
3 month

6 month

a
Figure Legends

Figure 1. Associations of milk 12,13-diHOME and anthropometry measures in infancy. A: Linear

Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020


Association between Log(12,13-diHOME) in milk and BMI Z-score at birth, P=0.005. B: Infant

adiposity (% fat assessed by ADP) at 1 month by milk 12,13-diHOME tertile. P = 0.077; P(adjusted) =

0.021 (covariates: infant sex, gestational age, maternal pregravid BMI, gestational weight gain and

parity) C: Linear Association between Milk log(12,13-diHOME) and change in infant BMI Z-score

t
from 0 to 6 months, P=0.025. D: Change in infant weight-for-age (WFA) Z-score according to tertile

ip
of milk 12,13-diHOME. E: Change in infant length-for-age (LFA) Z-score according to tertile of milk

cr
12,13-diHOME. F: Change in infant weight-for-length (WFL) Z-score according to tertile of milk

us
12,13-diHOME. * denotes P<0.05, ** denotes P<0.01.
an
Figure 2. Associations of milk Ephx1-4 metabolites and anthropometry measures in infancy. A:
M

Biosynthetic pathway for 12,13-diHOME. B: Linear Association between Log(9,10-diHOME) in milk

and infant BMI Z-score at birth, P=0.007. C: Change in WFL Z-score from 0-6 months according to
d

tertile of log(9,10-diHOME) in milk. D: Linear Association between Milk log(9,10-diHOME) and


e

change in WFL Z-score from 0 to 6 months, P=0.029. E: Linear Association between Log(12,13-
pt

epOME) in milk and infant BMI Z-score at birth, P=0.026. F: Change in WFL Z-score from 0-6 months
ce

according to tertile of log(12,13-epOME) in milk. G: Linear Association between Milk log(12,13-

epOME) and change in WFL Z-score from 0 to 6 months, P=0.064. * denotes P<0.05, ** denotes
Ac

P<0.01.

Figure 3. Breast milk 12,13-diHOME concentration rises after a moderate exercise bout. Change in

12,13-diHOME concentrations at baseline and 90 minutes after a bout of moderate exercise at 1 mo.

post-partum. N=16.**P = 0.0076, 2-sided paired T-test, pre- vs. post-exercise.


Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020
ip
cr
Figure 1

us
an
M
ed
pt
ce
Ac
Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020
ip
cr
Figure 2

us
an
M
ed
pt
ce
Ac
Downloaded from https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgaa799/5950351 by Auckland University of Technology user on 03 November 2020
ip
cr
Figure 3

us
an
M
ed
pt
ce
Ac

You might also like