ArticlePDF Available

Abstract and Figures

Brown adipose tissue (BAT) undergoes pronounced changes after birth coincident with the loss of the BAT-specific uncoupling protein (UCP)1 and rapid fat growth. The extent to which this adaptation may vary between anatomical locations remains unknown, or whether the process is sensitive to maternal dietary supplementation. We, therefore, conducted a data mining based study on the major fat depots (i.e. epicardial, perirenal, sternal (which possess UCP1 at 7 days), subcutaneous and omental) (that do not possess UCP1) of young sheep during the first month of life. Initially we determined what effect adding 3% canola oil to the maternal diet has on mitochondrial protein abundance in those depots which possessed UCP1. This demonstrated that maternal dietary supplementation delayed the loss of mitochondrial proteins, with the amount of cytochrome C actually being increased. Using machine learning algorithms followed by weighted gene co-expression network analysis, we demonstrated that each depot could be segregated into a unique and concise set of modules containing co-expressed genes involved in adipose function. Finally using lipidomic analysis following the maternal dietary intervention, we confirmed the perirenal depot to be most responsive. These insights point at new research avenues for examining interventions to modulate fat development in early life.
This content is subject to copyright. Terms and conditions apply.
1
SCIENTIfIC REPORTS | (2018) 8:9628 | DOI:10.1038/s41598-018-27376-3
www.nature.com/scientificreports
Transcriptional analysis of adipose
tissue during development reveals
depot-specic responsiveness to
maternal dietary supplementation
Hernan P. Fainberg1, Mark Birtwistle
1, Reham Alagal1,5, Ahmad Alhaddad1, Mark Pope1,
Graeme Davies1, Rachel Woods
1, Marcos Castellanos3, Sean T. May
3, Catharine A. Ortori4,
David A. Barrett
4, Viv Perry6, Frank Wiens7, Bernd Stahl7, Eline van der Beek7,8,
Harold Sacks9, Helen Budge1 & Michael E. Symonds1,2
Brown adipose tissue (BAT) undergoes pronounced changes after birth coincident with the loss of the
BAT-specic uncoupling protein (UCP)1 and rapid fat growth. The extent to which this adaptation may
vary between anatomical locations remains unknown, or whether the process is sensitive to maternal
dietary supplementation. We, therefore, conducted a data mining based study on the major fat depots
(i.e. epicardial, perirenal, sternal (which possess UCP1 at 7 days), subcutaneous and omental) (that do
not possess UCP1) of young sheep during the rst month of life. Initially we determined what eect
adding 3% canola oil to the maternal diet has on mitochondrial protein abundance in those depots
which possessed UCP1. This demonstrated that maternal dietary supplementation delayed the loss
of mitochondrial proteins, with the amount of cytochrome C actually being increased. Using machine
learning algorithms followed by weighted gene co-expression network analysis, we demonstrated
that each depot could be segregated into a unique and concise set of modules containing co-expressed
genes involved in adipose function. Finally using lipidomic analysis following the maternal dietary
intervention, we conrmed the perirenal depot to be most responsive. These insights point at new
research avenues for examining interventions to modulate fat development in early life.
e association between excessive fat storage (i.e. obesity) and increased risk of metabolic disease, which leads
to a reduction in life quality and expectancy, is well documented1. To identify modiable factors that drive
unhealthy fat deposition it could be informative to better understand the pronounced developmental changes in
adipose tissue that occur soon aer birth2. is age period is characterised by the rapid activation of nonshivering
thermogenesis in brown adipose, an energy-using process involving tissue-specic uncoupling protein (UCP)13,
which helps the newborn to achieve a physiological body temperature. Subsequently, brown fat is gradually lost
and replaced with white adipose tissue4, which contains cells that store energy for later usage as cellular fuel.
Over the last decade, high-throughput genome-wide association studies (GWAS), together with gene expression
proling, epigenetic and integrative genomic analysis, have all contributed to a better understanding of adipose
1Division of Child Health, Obstetrics & Gynaecology, The University of Nottingham, Nottingham, United Kingdom.
2Nottingham Digestive Disease Centre and Biomedical Research Centre, School of Medicine, Queen’s Medical
Centre, The University of Nottingham, Nottingham, United Kingdom. 3Nottingham Arabidopsis Stock Centre,
School of Biosciences, The University of Nottingham, Nottingham, United Kingdom. 4Centre for Analytical
Bioscience, School of Pharmacy, The University of Nottingham, Nottingham, United Kingdom. 5Princess Nourah
Bint Abdulrahman University, Department of Nutrition and food science, College of Home Economics, Riyadh, BOX:
84428, Saudi Arabia. 6Robinson Research Institute, Medical School, University of Adelaide, Adelaide, Australia.
7Nutricia Research, Utrecht, The Netherlands. 8Department of Pediatrics, University Medical Centre Groningen,
University of Groningen, Groningen, The Netherlands. 9VA Endocrinology and Diabetes Division, VA Greater Los
Angeles Healthcare System, and Department of Medicine, David Geen School of Medicine, University of California
Los Angeles, California, USA. Correspondence and requests for materials should be addressed to M.E.S. (email:
michael.symonds@nottingham.ac.uk)
Received: 2 November 2017
Accepted: 30 May 2018
Published: xx xx xxxx
OPEN
Content courtesy of Springer Nature, terms of use apply. Rights reserved
www.nature.com/scientificreports/
2
SCIENTIfIC REPORTS | (2018) 8:9628 | DOI:10.1038/s41598-018-27376-3
tissue biology and its crucial role in the metabolic syndrome5,6. Scientic break-through, however, may have been
hampered by wrongly assuming that adipose tissue at various anatomical sites is regulated in an identical manner
for coping with metabolic challenges during current and later life2. To test for site-specic regulation and develop-
mental plasticity of adipose tissue we used sheep, based on the rapid transformation from brown to white adipose
tissue characteristics over the rst month of life3.
e function and development of adipose tissue depots are oen studied in isolation7, so, potential dierences
between depots and inuence of adjacent organs are not well established3,6,7. With recent advances in computer
power and functional annotation of transcriptome data, primary genes can be identied based on their pattern of
expression across the genome5,6. Furthermore, genes with similar co-expression patterns innately cluster together
and/or form distinct modules representing pathways involved in the regulation of interdependent biological
functions8. Dierences in the changes in the transcriptome with age between similar tissues may reect variations
in cell type, but also in their function, transcriptional regulation, and responsiveness to external cues8. In the pres-
ent study, we employed a machine learning (ML) algorithm followed by a weighted gene co-expression network
analysis in order to nd biologically meaningful associations in microarray datasets from the ve major fat depots
in sheep at 7 (when brown characteristics can dominate) and 28 (when brown fat is scarce) days of age. ese
measures enabled us to elucidate the distribution of cellular plasticity in response to a nutritional intervention
between depots. e maternal diet was modied to cause a shi in the fatty acid (FA) composition of maternal
milk achieved through feeding the mothers a supplement of canola oil. e inclusion of canola in dairy ruminants
feed has been reported to reduce the omega-6/omega-3 FA ratio and conjugated linoleic acid (CLA) in milk, both
which are properties that have been found to up-regulate UCP genes in adipose tissue9.
Methods
Animal Model. All of the procedures were performed with full institutional ethical approval from the
University of Nottingham as designated under the United Kingdom Animals (Scientic Procedures) Act, 1986.
All laboratory procedures were carried out at e University of Nottingham under the United Kingdom code of
laboratory practice (COSHH: SI NO 1657, 1988). For this study, thirteen twin-bearing (non-identical) Bluefaced
Leicester cross Swaledale ewes were randomly assigned immediately aer giving birth to receive their a standard
diet of roughage and concentrate throughout lactation (control, n = 5) or received the same diet supplemented
with 3% canola oil (i.e. 45 g in 1500 g of concentrate) (n = 8). All mothers delivered spontaneously at term (~147
d). One (sex-matched) twin from each mother was randomly assigned to be humanely euthanased at 7 days and
adipose tissues sampled from the epicardial, perirenal, sternal, subcutaneous and omental depots. Tissues were
quickly dissected and weighed before being sectioned and snap frozen in liquid nitrogen for storage at 80 °C.
Additional representative sections were xed in 10% v/v formalin and embedded in paran wax for histolog-
ical analysis. Each remaining twin was reared naturally with their mother until 28 days of age when they were
humanely euthanased, and adipose tissue sampled. Samples of the mother’s milk were also taken manually from
each mother on days 7 and 28 at ~08.00 h prior to euthanasia of the ospring. e milk was transferred into two
sterile 15 ml tubes (Greiner Bio-One, Gloucester, UK) and stored at 80 °C until being shipped on dry ice for anal-
ysis of milk FA composition.
Immunohistochemistry. Tissue sections were prepared as previously published10 and stained using hae-
matoxylin and eosin and for UCP1. At least 20 slides per animal alongside a negative control were labelled with a
random identier, loaded into the Leica BondMax IHC slide processor (Leica Microsystem), and run on an auto-
mated soware program (Vision Biosystems Bond version 3.4A) using bond polymer rene detection reagents
(Leica Microsystem) and a 1:500 dilution of primary rabbit polyclonal antibody to UCP1 (ab10983, Abcam).
Western blotting. The relative abundance of UCP1, voltage dependent anion channel 1 (VDAC) and
cytochrome c were determined in the perineal and sternal adipose tissue samples as previously described11. is
analysis was not performed on epicardial fat as insucient sample was available. All data was corrected against
the density of staining for total protein. Each antibody gave a signal at the correct molecular and the specicity of
binding for each anti-body was conrmed using non-immune rabbit serum.
RNA isolation, quantication, and quality control. For RNA isolation from each fat depot from 5 ani-
mals within each nutritional group, and a 100 mg was used from samples collected at 7, and 1000 mg from taken at
28 days of age, respectively. ese were mixed with 2 ml of TRI reagent (Sigma-Aldrich). Total RNA was extracted
using the RNeasy Plus kit (Qiagen) according to the manufacturer’s instructions and its quantity measured
with a NanoDrop ND-1000 Spectrophotometer (ermo Scientic). Optical density ratios (260/280 nm) were
>1.9 for all samples. Total RNA quality was assayed by the Agilent BioAnalyzer RNA 6000 Nano Kit (Agilent
Technologies) and only used if distinct ribosomal peaks measured (i.e. RIN > 7).
Transcriptome proling with Aymetrix GeneChip. Representative samples of mRNA were labelled
and hybridized onto Human Genome U133A plus 2 arrays according to manufacturers recommendations using
the GeneChip 3 IVT Express kit (Aymetrix). e Aymetrix Human U133 + 2 gene chip array can be used to
study ovine tissues11 and detection was performed using a GeneChip Scanner 3000 7G.
Gene expression array analysis. Normalization and network analyses were undertaken using free and
open source packages from the R project (http://cran.r-project.org/) unless otherwise stated. We used the open
source Bioconductor community (http://www.bioconductor.org/) with the function “gcrma” embedded in
the “ay” package for pre-processing data, including background correction, normalization, and probe match
verication. For statistical analysis of gene expression, we used the “limma” library, which enabled us to per-
form empirical Bayesian statistical modelling between the 5 adipose tissue depots and the eect of the maternal
Content courtesy of Springer Nature, terms of use apply. Rights reserved
www.nature.com/scientificreports/
3
SCIENTIfIC REPORTS | (2018) 8:9628 | DOI:10.1038/s41598-018-27376-3
nutritional intervention. For all other statistical analysis, unless otherwise stated, we applied the FDR approach
and considered q 0.05 as signicant12. e R-package “gplots” was used to assess fold changes, and make heat
maps, and expression plots.
Accession numbers. All original microarray data were deposited in the NCBI’s Gene Expression Omnibus
(accession GSE115799).
Selection of informative genes using the RF-PSOL algorithm. e ML algorithm was prepared
using the R package ml-DNA, of which the PSOL algorithm was used to correctly identify ~95–98% of “inform-
ative” genes13. It employed a “training dataset” comprising the top 100 genes selected through empirical Bayesian
statistical modelling between each dierent tissue at each age. Each result from the classier or the prediction
accuracy of the RF was tested using the 5-fold cross-validation method14. e validity for each selection cycle
of interactions was assessed by values of the area under the curve (AUC) received from operating characteristic
analysis or ROC (i.e. the two-dimensional plot between the false (x axis) versus the true positives rate (y axis)
at all possible thresholds). e AUC values ranged from 0 to 1, with a higher AUC indicating better prediction
accuracy for the random forest model.
Gene Co-expression Network Construction. Datasets from both age groups (i.e. 25 samples, compris-
ing 5 dierent adipose tissues depots from 5 animals) were constructed separately using a standard workow
as recommended by weighted correlation network analysis (WGCNA)15. We used a signed weighted correla-
tion network for both age groups and the resulting Pearson correlation matrix was transformed into a matrix of
connection strengths (e.g., an adjacency matrix) using a power of 19. For the dynamic tree-cutting algorithms,
a merging function was set at 0.25, which identied 11 modules at 7 days and 14 modules at 28 days of age. To
better describe molecular outgoing events in each age group dataset, our analysis was restricted to genes selected
as informative by the ML-based ltering process13.
e function ‘module Preservation’ added into the WGCNA R package provides a reliable module preser-
vation statistics base on the generation of 200 random permutations between two independent datasets. ese
permutations allow the calculation of a series of network Z-scores for statistical properties such as network con-
nectivity and density. By averaging these Z-scores in a single Z-score or Z-summary, this value can be used as an
indication of relationship preservation between genes in two independent networks. For example, a Z-summary
for a module that scores >10 could be interpreted as strongly preserved (i.e. no changes in the topology), or
scores between 2 and 10 are considered to be moderately preserved, or <2 indicates that the relationship between
the genes are not preserved15.
Gene ontogeny analysis. Functional annotation was performed with the WEB-based GEneSeTAnaLysis
Toolkit (or WebGestalt) and all genes within each module were analysed. e GO terms with a FDR < 0.05 and
enrich >5 genes per classication16.
Determination of milk fat content and composition. Milk fat concentration was determined by meth-
ods following Bligh & Dyer17. High-resolution capillary gas-liquid chromatography was used to determine the
composition of short- to long chain FAs in milk fat as previously described18. A total of 46 FAs were analyzed this
way and included saturated FAs: C4:0, C6:0, C8:0, C10:0, C11:0, C12:0, C13:0, C13ai, C14:0, C14ai, C15:0, C15ai,
C16:0, C16ai, C17:0, C18:0, C18i, C20:0, C20i, C21:0, C22:0, C23:0, monounsaturated FAs: C14:1n-5, C15:1n-5,
C16:1n-7, polysaturated FAs, omega (n)-3: C18:3n3, C18:4n-3, C20:3n3, C20:5n-3, C22:5n3, C22:6n3. Omega
(n)-6: C18:2n6tr, C18:2n6, C18:3n-6, C20:2n-6 C20:3n6, C20:4n6, C22:2n-6, C22:4n-6, C22:5n-6 and omega
(n)-9: C16:1n-9/7t, C18:1n-9, C20:1n-9, C22:1n-9, C24:1n-9. Results are expressed as % milk fat. All data were
evaluated using the “Limma” and “gplots” in a similar fashion as described above for with the microarray analysis.
Determination of adipose tissue lipid composition. Adipose tissue samples (100–200 mg) were
ground in Retsch ball mill for 3 min using 6 mm stainless steel balls, all pre-chilled to 18 °C. Ice cold chloroform:
methanol (2:1) was added (0.5 mL) and the slurry was agitated at room temperature for 20 min, and centrifuged
for 10 min at 10,000 × g at 4 °C. e lower layer was removed and dried in a centrifugal evaporator. e sam-
ples were reconstituted in 100 µL chloroform:methanol 1:2 centrifuged, then decanted into amber glass LC vials
with inserts and stored at 80 °C until LC-MS analysis. Lipidomic QC samples were created from pooling equal
volumes of all sample extracts. e extracted lipids were injected (5 µL) onto an Agilent Poroshell 120 SB-C18
50 × 2.1 mm (2.7 µm particle size) with guard, held at 45 °C, and eluted using 0.1% aqueous ammonium acetate
(A), to 0.1% aqueous ammonium acetate /acetonitrile/isopropanol gradient (1:1:8) (B) using gradient elution. A
ermoScientic Accela modular HPLC system (Hemel Hempstead, UK) was used at a ow rate of 0.45 mL/min.
Ions in the range m/z 100 to 1900 were detected using an Exactive series mass spectrometer (ermoScientic
Hemel Hempstead, UK) in electrospray mode with +/ve switching at a resolution setting of 25000. Data was
normalised to the total ion count, pre-processed and exported to Excel for further processing using Progenesis QI
soware (Progenesis, Newcastle on Tyne, UK). A product ion prediction tool from Lipidmaps (Lipid MS Predict)
was use to assist spectral interpretation. e identities of selected isobaric lipid species were subsequently eluci-
dated by generating MS/MS spectra (nominal mass) using the same LC method with a ermo Scientic LTQ
Velos ion trap mass spectrometer using equivalent electrospray source settings and with a collision energy of 4019.
Results
Changes in the cellular landscape of brown and white adipose tissue during the rst four weeks
of postnatal life. To establish which fat depots could be classied as being brown and whether the rate of
loss of brown adipocytes occurs at similar rates in those depots, we rst examined the distribution of UCP1 by
Content courtesy of Springer Nature, terms of use apply. Rights reserved
www.nature.com/scientificreports/
4
SCIENTIfIC REPORTS | (2018) 8:9628 | DOI:10.1038/s41598-018-27376-3
immunohistochemistry. At 7 days of age, UCP1 was more abundant in perirenal and sternal than in epicardial
adipose tissue (Fig.1A) and was not present in subcutaneous or omental fat (not shown). UCP1 abundance was
substantially reduced by 28 days of age, although more UCP1 signal remained in the epicardial compared to other
depots. In addition, the rate of loss of UCP1 appeared to be delayed by the addition CLA to the mother’s diet.
We then further determined whether this response was specic to UCP1 using western blotting, in those brown
fat depots of which we had sucient tissue i.e. perirenal and sternal (Fig.1B). is demonstrated that the rate of
loss of both UCP1 and VDAC were delayed in both depots at 7 and 28 days of age (Fig.1B), whilst the amount of
cytochrome C was enhanced.
In order to identify changes in gene regulation with age, we performed a transcriptomic comparison between
all 5 depots. At 7 days of age, 839 genes were signicantly upregulated between depots (false discovery rate
(FDR) 0.05; Supplement: Dataset1) and the greatest dierence was between epicardial and omental fat (429
signicant genes, FDR 0.05; Fig.2A). Only perirenal adipose tissue, however, exhibited a consistent upregu-
lation of UCP1 and other thermogenesis-related genes compared with the omental and subcutaneous adipose
depots, which were populated primarily by white adipocytes (Supplement: Dataset1). ese results indicate local
dierences in transcriptional regulation, which is indicative of substantial changes in cell population between fat
depots during postnatal life. is is in accord with their divergent developmental ontogeny between depots. In
this regard epicardial, perirenal and sternal are all present in the fetus20, whereas omental and to a lesser extent
subcutaneous fat only appears aer birth21.
Next, we performed a clustering analysis of the transcriptomic data at 7 days of age. Unsupervised hierarchical
clustering and principal component analysis (PCA) revealed that each depot forming a distinct cluster (Fig.2),
with the rst two components explaining 80.2% of the total variance in the data set. is suggests a strong t
between the computational model and the existence of intrinsic dierences in gene expression between depots.
PCA cluster analysis further showed that the pro-thermogenic perirenal and sternal fat depots clustered together
as did subcutaneous and omental depots (white fat depots) whereas epicardial adipose tissue was separate from
other depots (Fig.2C).
We repeated these statistical analyses at 28 days of age to identify the primary changes in gene expression,
and identied 2059 dierentially expressed genes between depots (FDR 0.05; Supplement: Dataset2). At this
time point, sternal adipose tissue showed a more pronounced downregulation of the transcriptional architecture
compared with the other fat depots (Fig.2B). Similar to our observation at 7 days of age, each depot had a distinc-
tive pattern of gene expression. Genes within the perirenal depot clustered together with those from omental fat
and close to those from the subcutaneous depot, but away from the epicardial and sternal depots. Overall these
samples formed three stable clusters and the rst two components of the PCA analysis accounted for 73.1% of the
variance observed in gene expression between depots (Fig.2D). In summary, clustering analyses demonstrated
that anatomical location determines transcriptome dierences which are modied with age. e sternal and per-
irenal depots exhibited the greatest transcriptomic remodeling, reecting changes in their biological/metabolic
functions with age4.
Figure 1. (A) Representative immunohistochemical detection of uncoupling protein (UCP)1 from sternal,
perirenal and epicardial, sampled from 7 and 28 day old sheep. Rectangular outlines indicate clusters of
uncoupling protein 1 (UCP) positive cells found in the epicardial adipose tissue at 28 days (scale bar = 50 μm;
Magnication 40x) and (B) mean mitochondrial protein abundance as determined by western blotting
in adipose tissue sampled from 7 and 28 day old ospring born to mothers fed a control diet (n = 5) or
supplemented with 3% canola oil (n = 8). Values are means with their standard errors signicant dierences
between dietary groups at the same age denoted by *p < 0.05; **p < 0.01. UCP, Voltage dependent anion
channel 1, VDAC.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
www.nature.com/scientificreports/
5
SCIENTIfIC REPORTS | (2018) 8:9628 | DOI:10.1038/s41598-018-27376-3
Use of ML and gene network analyses for identifying the transcriptional architecture changes
between adipose tissue depots with age. To determine the magnitude of potential changes in gene
function between depots during early life, we combined computer-assisted learning algorithms with weighted
gene co-expression network analysis. Through a reiterative process of error minimisation and supervised
learning algorithms (i.e. random forest (RF)), the optimal gene expression pattern for each adipose depot was
established14. Tissue gene expression datasets were submitted for each age group, the RF approach enabled iden-
tication beyond a 98% certainty of the informative genes with an overall accuracy of 95–100%.
Figure 2. Comparison in gene expression of ve adipose tissue depots at (A) 7 and (B) 28 days of age. Heat
map and unsupervised hierarchical clustering dendrograms are shown for the top 100 dierentially-expressed
gene transcript comparisons identied by microarray analysis (average linkage, Euclidean distance metric) as
selected by eBayes moderated t-statistics (FDR < 0.05). Gene expression was transformed to a Z-score, and blue
represents a relative is a decrease and red an increase in gene expression between each depot at the same age.
Principal component analysis (PCA) of gene expression data from ve dierent adipose tissue depots from each
animal was performed using 10055 data sets that passed the variance test QC at (C) 7 and (D) 28 days of age.
Each sample is represented by a sphere (7 days) or rectangle (28 days) and color-coded to indicate the age and
tissue to which it belongs.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
www.nature.com/scientificreports/
6
SCIENTIfIC REPORTS | (2018) 8:9628 | DOI:10.1038/s41598-018-27376-3
e number of informative genes varied with age indicating dierent response patterns of the transcriptomes
between depots, with 2274 and 3339 genes identied as informative at 7 and 28 days of age, respectively. Next, we
used both data sets of informative genes to generate two independent weighted co-expressed networks for each
age. We identied 11 distinct modules (designated as C1.1–11; Fig.3A and Supplement: Dataset3) at 7 days of
age and 14 modules of co-expressed genes at 28 days (designated as C2.1–14; Fig.3B and Supplement: Dataset4).
Further analyses demonstrated that most gene modules corresponded to dierent transcriptional or metabolic
functions performed in each depot, whereas the intensity of these events varied with age.
Functional enrichment of gene clusters obtained at 7 days of age. e gene network analyses
demonstrated that the sternal and perirenal depots shared common transcriptional functionalities, although they
were more pronounced in perirenal fat (Fig.4A and B). Many of these modules (i.e. C1.1, C1.2 and C1.3) con-
tained genes involved in regulating mitochondrial biogenesis and aerobic respiration. e C1.3 and C1.2 modules
showed several gene ontology enriched terms related to oxidation-reduction processes and mechanisms of mito-
chondrial regulation of mRNA translation, respectively (Supplement: Dataset5). However, the separation of both
co-expressed modules suggests the potential existence of multiple transcriptional loops regulating mitochondrial
genes5,22. ese transcriptional separations could potentially allow brown adipocytes to regulate the expression of
mitochondrial genes independent of UCP1 expression23.
e majority of gene modules aligning within epicardial adipose tissue coincided with specic stages of
changes in transcriptional cardiomyocyte cell dierentiation (Fig.5A)24. For example, the C1.4 module showed
hallmarks of mRNA and DNA processes associated with the maintenance of stem cell pluripotency24. Module
C1.6 had a large transcriptional signature usually found in mature cardiomyocytes, suggesting that these tran-
scripts reect an advanced stage in cell dierentiation25. All these ndings are in agreement with the known
pluripotency of adipose tissue-derived stem cells towards dierentiation into cardiac myocyte-like cells or brown
adipocytes26.
Modules, C1.7 and C1.8, both describe transcription events occurring mainly within subcutaneous adipose
tissue (Fig.4A), and contain genes functionally linked to myocyte precursors26. ese are considered to give
rise to brown adipocytes and a subset of white adipocytes populating subcutaneous fat (Supplement: Dataset5).
Figure 3. Co-expression dendrogram analysis from the ve adipose tissue depots sampled at either (A) 7 or (B)
28 days of age. In each dendrogram, the rst row is subdivided into co-expressed modules founded in each age
group. Rows 2 to 6 show the dierential expression relationships between module genes and the adipose depot.
e relationship of each gene with the assigned module is colour coded from blue (negative co-expression) to
red (positive co-expression).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
www.nature.com/scientificreports/
7
SCIENTIfIC REPORTS | (2018) 8:9628 | DOI:10.1038/s41598-018-27376-3
Finally, we identied modules C1.9, C1.10 and C1.11 that were enriched within omental fat and contained genes
linked to carbohydrate metabolism, cytoskeleton composition and tissue remodeling mediated through activa-
tion of the immune system (Supplement: Dataset5)27.
Changes in transcriptional control and its consequences for adipocyte function with age. To
address whether the dierences in transcriptome regulation identied between depots at 7 days of age had any
phenotypical consequences, we generated a second gene co-expression network at 28 days of age. As observed
from the PCA analysis, the perirenal depot underwent the greatest shi in gene expression. Four co-expression
modules with selective enrichment in that depot were detected, of which the C2.7 module was also enriched in
omental fat (Fig.4C,D). is common module indicates a major metabolic change as perirenal adipose tissue
adapts from a heat production depot populated by brown adipocytes to a white fat depot, storing energy as
lipid4,22. is module contained many genes associated with FA metabolism, including GHR, ABHD5, DECR1 and
PPARG that also regulates adipocyte dierentiation (see Supplement: Datasets4 and 6)28. In addition, this mod-
ule revealed that genes associated with white adipose tissue expansion were co-expressed, including angiogenic
genes (HOXA5, HIF1A) and pre-adipocyte precursor genes (HOXB6, HOXB8, HOXB5; Supplement: Datasets4
and 6)29,30. Other genes enriched in perirenal fat were those in module C2.5, and included transcripts associated
with activation of inammatory responses and endoplasmic reticulum, indicating cellular stress (Supplement:
Dataset6)27. e last two modules aligned within this depot, C2.9 and C2.8, had large signatures associated with
cell division and mRNA transcriptional regulation, suggesting an increase in cell diversication or dierentiation
(Fig.4C and Supplement: Dataset6)3,31,32. Conversely, in sternal fat, these cellular events appeared to be repressed
(Supplement: Dataset2), and the only cluster positively aligned was C2.1, which was enriched with genes indi-
cating chemical or hormonal responsiveness and those leading to tissue angiogenesis (Fig.5B and Supplement:
Dataset6)30,33. As observed at 7 days, the epicardial adipose tissue also exhibited modules enriched with genes
with a large genetic signature associated with cardiomyocyte cell dierentiation such as module C2.2 (Fig.5B)26.
Modules C2.10 and C2.11 showed signicant relationships in omental adipose tissue (Fig.4C,D)32, and exhib-
ited over-representation of transcripts linked to the balance between anabolic and catabolic pathways occurring
in mature adipocytes (Supplement: Dataset6)34. Finally, in subcutaneous adipose tissue, most genes were assigned
to modules C2.12, C2.13 and C2.14 and these were associated with cell dierentiation, growth and remodelling30.
Changes in gene networks represent functional adaptations of dierent adipose depots during
development. To explore the biological relevance of each module in more detail, we applied a statistical
approach based on a series of random permutations between datasets. is enabled us to nd evidence of similar-
ities and dierences in the network topology at both ages. We found that the majority of genetic interactions per-
sisted at both ages (Fig.5A,B). At 7 days, 86.4% of all the genes were allocated to a conserved module, including
module C1.2 which was enriched with mitochondrial genes such UCP1 (Fig.5A). Gene ontology analysis of the
Figure 4. Summary of adipose tissue depot- and age-specic functional organisation of modules within each
gene network. ey were related individually by their rst principal component, referred to as the module
eigengene (ME). Each dendrogram illustrates the modules of co-expressed genes and their positive alignment
within the ME at (A) 7 and (C) 28 days of age. e height (X-axis) indicates the magnitude of correlation
expressed as Euclidean distances. Heat maps represent the correlation (and corresponding p-values) between
co-expressed modules for each fat depot at (B) 7 and (D) 28 days of age. e colour scheme, from blue to red,
indicates the magnitude of correlation, from low to high. Regional-specic modules identied as being highly
correlated (i.e. over-expressed) for each adipose depot are shown in the columns.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
www.nature.com/scientificreports/
8
SCIENTIfIC REPORTS | (2018) 8:9628 | DOI:10.1038/s41598-018-27376-3
four non-conserved modules showed that they all shared functional similarities associated with early stages of cell
dierentiation and adipose tissue remodelling30. At 28 days, 71.4% of all the genes were allocated to a conserved
module within the network. e gene ontogeny enrichment analysis from these conserved modules revealed
metabolic and transcriptomic processes associated with mature adipocyte function which mainly involve lipid
metabolism, immune responses and tissue remodeling (Fig.5B).
Functional assessment of changes in transcriptional architecture. In order to evaluate the potential
impact of the transcriptional dierences between adipose depots, we performed lipidomic and gene expression
analyses on perirenal and sternal fat at 28 days of age in ospring of mothers who consumed a diet supplemented
with 3% canola oil. By this point of lactation milk from supplemented mothers had developed an altered FA
prole without changes in the total fat content. Besides, the maternal nutritional supplementation had no eect
on growth or body weight and of the ospring. Features of the FA prole that had changed in relation to the
control group according to predictions included the statistically signicantly lowered proportion of linoleic acid
and a lowered omega-6/omega-3 FA ratio (Supplement: Dataset7 and Supplement: Fig.1). e supplemented
milk exhibited similar omega 3 FAs but a lower arachidonic (C20:4n6) and γ-linolenic FA (C18:3n6) content.
Figure 5. Summary of cross-adipose tissue depot module preservation with age. e test uses a Z score
summary of dierent network properties to determine gene connectivity at (A) 7 and (B) 28 days of age. Each
row represents a module and each column a unique feature of each module including positive alignment with
each ME and the number of genes per module. A Z summary value >2 represents a moderately preserved
module, and a value >10 provides strong evidence of module preservation.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
www.nature.com/scientificreports/
9
SCIENTIfIC REPORTS | (2018) 8:9628 | DOI:10.1038/s41598-018-27376-3
Dietary supplementation produced a localised change in the lipidome of perirenal fat (Supplement: Dataset8
and Supplement: Fig.2). Overall, the intervention led to dierences in the relative abundance of 28 day perirenal
adipose tissue spectrometry lipid masses. Amongst those that we were able to identify, we observed decreased
proportions of phosphatidylcholines (PC; a major constituent of cell membranes) and long chain FAs (carbon
chain length >20). In contrast, in sternal adipose tissue, there were no dierences in lipid composition between
the intervention and control groups at 28 days of age. Overall, microarray analyses of both fat depots revealed that
only the genome of perirenal fat responded to the nutritional intervention. Data mining (Supplement: Dataset9)
results thus showed a signicant increase in the expression of only 4 genes. Interestingly, when comparing the
most dierentially regulated genes, eects of the intervention on the down regulation of NR3C1 were highlighted.
is gene encodes for the glucocorticoid receptor and is directly associated with inammatory responses, cellular
proliferation, lipid metabolism, cell dierentiation and more importantly the modulation of thermogenesis in
brown fat35,36. Potentially, due to the relatively short duration of intervention and/or by modest amount of canola
oil added to the diet, we could not observe substantial changes in UCP1. Taken together, these results demon-
strate that the cellular architecture of perirenal fat is unique among fat depots in that it can undergo pronounced
remodeling and is sensitive even to a modest nutritional stimulus.
Discussion
We have shown profound dierences in gene expression proles between the major fat depots in sheep through
early postnatal life, coincident with the transition from brown to white fat depots3. e developmental changes
markedly diered between depots despite them showing a similar macroscopic morphology at each developmen-
tal age. For example, at 28 days when in sheep fat is considered primarily white3, each adipose depot kept a dis-
tinct expression prole. is appeared to determine its capacity to respond to modication of the composition of
the mother’s milk. Adipose tissue has been considered a metabolic organ with important functions beyond lipid
storage but the extent to which this varies between depots especially during development is largely unexplored.
Our new data support the concept that adipose tissue functions not as one metabolic organ, but as several auto-
nomic organs which appear to have distinct functions32.
By using a computer-assisted supervised learning algorithm, we demonstrate that during postnatal develop-
ment each fat depot contains a transcriptome which forms dynamic networks with unique sets of genes8. Over
time these gene networks can undergo profound reorganisation by accommodating novel members and/or losing
some of their original components8. Whilst ontogenic plasticity can be driven entirely by intrinsic factors, sur-
vival value in uctuating environments can be enhanced by responsiveness to extrinsic factors3. However, both
types of plasticity are prone to error and maladaptation which can ultimately lead to obesity, increasingly threat-
ening metabolic health1. Understanding how environmental factors, particularly during early life, interfere with
pathways of energy utilisation or storage is one of the most important intermediate goals in obesity prevention.
By examining the postnatal development of adipose tissue through gene network analysis, we have been able to
construct novel biological interpretations8 specic to each fat depot over the period when any large mammal
needs to respond to environmental, nutritional and physiological challenges3. We, therefore, explored dynamic
changes in gene regulation and identied the main regulatory relationships. ese have crucial regulatory roles so
each separate adipose tissue depot can dierentiate and adapt, potentially enabling the dierent genes involved to
modulate metabolic homeostasis32. Despite recent eorts to elucidate the cellular and transcriptome composition
of dierent fat depots4,23, the inuence of genetic, endocrine or environmental factors on fat development remains
largely unknown. However, in our study we observed that the co-expression modules within networks show a
depot-specic pattern enriched with genes performing specic functions.
Studies on adipose tissue function during early postnatal life have mostly focused on explaining the loss
of genes associated with cellular thermogenesis, especially UCP13. Our comparison between the three depots
enriched with brown adipocytes suggests the existence of dierent networks in the regulation of mitochondrial
activity5. e expression of mRNA is regulated by a balance between transcription and mRNA degradation, and
the C1.2 module captures this complexity in the control of UCP137. We found transcription factors that stimulate
a cell’s transition from myoblastic precursors to brown fat cells, including C/EBPβ and EP30022,38. Other mem-
bers of this module were EIF4B, EIF4G3, EIF3D and EIF3G, known transcriptional factors that downregulate
mRNA transcription of UCP1 in response to raised temperature39. We also found a large number of ribosomal
proteins co-expressed with UCP1 that are similarly regulated, including RPS5 and RPS9. ese comprise part of
the original mitochondrial protein assembly machinery40. UCP1 is also regulated by AU-rich elements, which
are mRNA binding proteins37. In humans, it has been estimated that less than 8% of genes are regulated in this
manner, raising the possibility that mRNA binding proteins such as ZFP36L1 and DHX32 co-expressed with
UCP1 could potentially degrade this gene37,41. ZFP36L1 specically binds at its 3-UTR mRNA site and recruits
the Cnot7-Tob-BRF1 axis, resulting in mRNA destabilisation41. ese genes are co-regulated with multiple tran-
scripts involved in energy metabolism and mRNA transcription allocated to the C1.1 and C1.3 modules, which
are both related to the functional changes observed in perirenal fat up to 28 days of age.
Another novel nding is the relationship between epicardial adipose tissue and genes associated with cardi-
omyocyte cell dierentiation24. Cardiomyocytes rapidly dierentiate and proliferate during fetal life, but exit the
cell cycle soon aer birth, limiting the ability of the heart to restore function aer any signicant injury26. ere
are reservoirs of multipotent stem cells in most fat depots which can dierentiate in vitro into brown adipocytes
and also, as observed, into cardiomyocytes25. As shown in Fig.5, the highest biologically expressed genes were
those of cardiac myocytes in epicardial fat cells in module C1.6 at 7 and module C2.2 at 28 days, respectively. One
pathway to brown adipocyte dierentiation is through chronic adrenergic stress, as observed in rodents aer
prolonged cold exposure39 and in patients with severe skin burn injuries42. We have previously observed that
epicardial fat maintains a signicant number of brown adipocytes, in children with congenital heart disease43. It
is, therefore, possible that the fate of these multipotent stem cells, or their ability to dierentiate into other cell
Content courtesy of Springer Nature, terms of use apply. Rights reserved
www.nature.com/scientificreports/
10
SCIENTIfIC REPORTS | (2018) 8:9628 | DOI:10.1038/s41598-018-27376-3
types, could be regulated by specic growth factors supporting normal physiological development or enabling
them to respond to disease24,43.
Our study conrms that the perirenal and sternal depots follow dierent development patterns20. Although
UCP1 expression ceases in both depots with age, only perirenal fat exhibits the major hallmarks of white adipose
tissue development7. is insight was further substantiated by the observation that, when ospring consumed
milk from mothers that had received a 3% supplement of canola oil, only the cellular lipidome of the perire-
nal adipose tissue was modied. is was accompanied with the retention of UCP1 up to 28 days of age, that
is likely to be mediated by dierences in milk lipid proles found between control and intervention groups.
Supplementation of canola oil in the maternal diet did not have a direct eect on omega 3, but limited the accre-
tion of two omega 6 FAs, γ-linolenic acid (C18:3n6) and more signicantly of arachidonic acid (C20:4n6). is
essential FA is metabolized by a transcellular process using cyclooxygenases to induce prostaglandin synthe-
sis, thus triggering a pro-inammatory response44. e changes in arachidonic acid in perirenal adipose tissue
may explain in part the dierences in membrane architecture and the up-regulation of genes related to inam-
mation between groups. Downstream, we observed that these changes in FA milk proles were accompanied
with reduced NR3C1 gene expression in perirenal fat. is is the transcript of the type 2 glucocorticoid receptor
mRNA, which is important in regulating the actions of glucocorticoids in most tissues3. Furthermore, glucocorti-
coids are not only necessary for adipocyte dierentiation they also modulate thermogenesis in a species and depot
specic manner35. Taken together, our observations suggest that dierences in the tissue microenvironment, pos-
sibly dictated by nearby endocrine organs such as the adrenals, determine changes in the metabolic/phenotypic
characteristics of existing fat cells6,32.
In conclusion, adipose tissue depots dier dramatically in terms of their gene expression signature, dieren-
tiation ability, cellular composition, and capacity to respond to local environmental stimuli45. Perirenal adipose
tissue shows the greatest propensity to dierentiate and respond to an external stimulus. In contrast, fat depots
such as sternal and epicardial do not exhibit an adipogenic prole and would therefore, complete their normal
programmed development. e data presented here suggest that microarray gene expression in combination with
advanced data analytic tools provide a robust and accurate approach for producing adipose depot-specic gene
signatures. Moreover, this approach could enhance our ability to identify and manipulate specic characteristics
of adipose tissue in dierent anatomical locations.
References
1. Gregg, E. W. & Shaw, J. E. Global Health Eects of Overweight and Obesity. N Engl J Med 377, 80–81 (2017).
2. Spalding, . L. et al. D ynamics of fat cell turnover in humans. Nature 453, 783–787 (2008).
3. Symonds, M. E., Pope, M. & Budge, H. e Ontogeny of Brown Adipose Tissue. Annu ev Nutr 35, 295–320 (2015).
4. ocstroh, D. et al. Direct evidence of brown adipocytes in dierent fat depots in children. Plos One 10, e0117841 (2015).
5. Claussnitzer, M. et al. FTO Obesity Variant Circuitry and Adipocyte Browning in Humans. N Engl J Med 373, 895–907 (2015).
6. Lotta, L. A. et al. Integrative genomic analysis implicates limited peripheral adipose storage capacity in the pathogenesis of human
insulin resistance. Nat Genet 49, 17–26 (2017).
7. Gesta, S., Tseng, Y. H. & ahn, C. . Developmental origin of fat: tracing obesity to its source. Cell 131, 242–256 (2007).
8. Barabasi, A. L. & Oltvai, Z. N. Networ biology: understanding the cell’s functional organization. Nat ev Genet 5, 101–113 (2004).
9. Welter, . C. et al . Canola Oil in Lactating Dairy Cow Diets educes Mil Saturated Fatty Acids and Improves Its Omega-3 and Oleic
Fatty Acid Content. Plos One 11, e0151876 (2016).
10. Pope, M., Budge, H. & Symonds, M. E. e developmental transition of ovine adipose tissue through early life. Acta Physiol (Oxf)
210, 20–30 (2014).
11. Mostyn, A. et al. Ontogeny and nutritional manipulation of mitochondrial protein abundance in adipose tissue and the lungs of
postnatal sheep. Br J Nutr 90, 323–328 (2003).
12. Wettenhall, J. M. & Smyth, G. . limmaGUI: a graphical user interface for linear modeling of microarray data. Bioinformatics 20,
3705–3706 (2004).
13. Ma, C., Xin, M., Feldmann, . A. & Wang, X. Machine learning-based dierential networ analysis: a study of stress-responsive
transcriptomes in Arabidopsis. Plant Cell 26, 520–537 (2014).
14. Breiman, L. andom Forests. Machine Learning 45, 27 (2001).
15. Oldham, M. C., Horvath, S. & Geschwind, D. H. Conservation and evolution of gene coexpression networs in human and
chimpanzee brains. Proc Natl Acad Sci USA 103, 17973–17978 (2006).
16. Wang, J., Duncan, D., Shi, Z. & Zhang, B. WEB-based GEne SeT AnaLysis Toolit (WebGestalt): update 2013. Nucleic Acids es 41,
W77–83 (2013).
17. Bligh, E. G. & Dyer, W. J. A rapid method of total lipid extraction and purication. Can J Biochem Physiol 37, 911–917 (1959).
18. ovacs, A., Fune, S., Marosvolgyi, T., Burus, I. & Decsi, T. Fatty acids in early human mil aer preterm and full-term delivery. J
Pediatr Gastroenterol Nutr 41, 454–459 (2005).
19. avipati, S., Baldwin, D. ., Barr, H. L., Fogarty, A. W. & Barrett, D. A. Plasma lipid biomarer signatures in squamous carcinoma
and adenocarcinoma lung cancer patients. Metabolomics 11, 1600–1611 (2015).
20. Henry, B. A. et al. Ontogeny and ermogenic ole for Sternal Fat in Female Sheep. Endocrinology 158, 2212–2225 (2017).
21. Saroha, V. et al. Tissue cell stress response to obesity and its interaction with late gestation diet. eprod Fertil Dev 30, 430–411 (2017).
22. ajimura, S. et al. Initiation of myoblast to brown fat switch by a PDM16-C/EBP-beta transcriptional complex. Nature 460,
1154–1158 (2009).
23. Lidell, M. E. et al. Evidence for two types of brown adipose tissue in humans. Nat Med 19, 631–634 (2013).
24. Liu, Q. et al. Epicardium-to-fat transition in injured heart. Cell es 24, 1367–1369 (2014).
25. Boheler, . . et al. Dierentiation of pluripotent embryonic stem cells into cardiomyocytes. Circ es 91, 189–201 (2002).
26. Yamada, Y., Wang, X. D., Yooyama, S., Fuuda, N. & Taaura, N. Cardiac progenitor cells in brown adipose tissue repaired
damaged myocardium. Biochem Biophys es Commun 342, 662–670 (2006).
27. Lu, C., umar, P. A., Fan, Y., Sperling, M. A. & Menon, . . A novel effect of growth hormone on macrophage modulates
macrophage-dependent adipocyte dierentiation. Endocrinology 151, 2189–2199 (2010).
28.  ajaumari, S. et al. EBF2 determines and maintains brown adipocyte identity. C ell Metab 17, 562–574 (2013).
29. Cantile, M., Procino, A., D’Armiento, M., Cindolo, L. & Cillo, C. HOX gene networ is involved in the transcriptional regulation of
in vivo human adipogenesis. J Cell Physiol 194, 225–236 (2003).
30. Par, Y. . et al. Hypoxia-inducible factor-2alpha-dependent hypoxic induction of Wnt10b expression in adipogenic cells. J Biol
Chem 288, 26311–26322 (2013).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
www.nature.com/scientificreports/
11
SCIENTIfIC REPORTS | (2018) 8:9628 | DOI:10.1038/s41598-018-27376-3
31. Lee, Y. H., Petova, A. P. & Granneman, J. G. Identication of an adipogenic niche for adipose tissue remodeling and restoration. Cell
Metab 18, 355–367 (2013).
32. Macotela, Y. et al. Intrinsic dierences in adipocyte precursor cells from dierent white fat depots. Diabetes 61, 1691–1699 (2012).
33. Sharey, D. et al. Impact of early onset obesity and hypertension on the unfolded protein response in renal tissues of juvenile sheep.
Hypertension 53, 925–931 (2009).
34. Pietilainen, . H. et al. Association of lipidome remodeling in the adipocyte membrane with acquired obesity in humans. Plos Biol
9, e1000623 (2011).
35. Lu, N. Z. et al. International Union of Pharmacology. LXV. e pharmacology and classication of the nuclear receptor superfamily:
Glucocorticoid, mineralocorticoid, progesterone, and androgen receptors. Pharmacol ev 58, 782–797 (2006).
36. amage, L. E. et al. Glucocorticoids Acutely Increase Brown Adipose Tissue Activity in Humans, evealing Species-Specic
Dierences in UCP-1 egulation. Cell Metab 24, 130–141 (2016).
37. Taahashi, A. et al. Post-transcriptional Stabilization of Ucp1 mNA Protects Mice from Diet-Induced Obesity. Cell ep 13,
2756–2767 (2015).
38. Vargas, D. et al. egulation of human subcutaneous adipocyte dierentiation by EID1. J Mol Endocrinol 56, 113–122 (2016).
39. van Breuelen, F., Sonenberg, N. & Martin, S. L. Seasonal and state-dependent changes of eIF4E and 4E-BP1 during mammalian
hibernation: implications for the control of translation during torpor. Am J Physiol egul Integr Comp Physiol 287, 349–353 (2004).
40. Maier, U. G. et al. Massively convergent evolution for ribosomal protein gene content in plastid and mitochondrial genomes.
Genome Biol Evol 5, 2318–2329 (2013).
41. Adachi, S. et al. ZFP36L1 and ZFP36L2 control LDL mNA stability via the E-S pathway. Nucleic Acids es 42, 10037–10049
(2014).
42. Sidossis, L. S. et al. Browning of Subcutaneous White Adipose Tissue in Humans aer Severe Adrenergic Stress. Cell Metab 22,
219–227 (2015).
43. Ojha, S. et al. Gene pathway development in human epicardial adipose tissue during early life. JCI Insight 1, e87460 (2016).
44. Smith, W. L. & Song, I. e enzymology of prostaglandin endoperoxide H synthases-1 and -2. Prostaglandins Other Lipid Mediat
68–69, 115–128 (2002).
45. Lee, Y. H., im, S. N., won, H. J. & Granneman, J. G. Metabolic heterogeneity of activated beige/brite adipocytes in inguinal
adipose tissue. Sci ep 7, 39794 (2017).
Acknowledgements
This work was supported by the Biotechnology and Biological Sciences Research Council [grant number
FS/15/4/31184, BB/I016015/1], and e Cardiometabolic Disease Research Foundation (Los Angeles, USA).
Author Contributions
M.E.S. and F.W. designed the study. M.E.S., M.B., G.D. and V.P. conducted the in vivo studies. H.P.F., A.A., C.M.
and M.S.T. conducted the microarray analysis. R.A. conducted the histological and western blot analysis. M.P.
and R.W. conducted the additional laboratory analysis. C.A.O. and D.B. conducted the lipidomic analysis. H.B.,
H.S., B.S. and E.v.d.B. further developed the study. H.P.F. and M.E.S. led the manuscript writing and development.
H.P.F. prepared all the tables and gures. All authors edited, contributed and reviewed the nal manuscript.
Additional Information
Supplementary information accompanies this paper at https://doi.org/10.1038/s41598-018-27376-3.
Competing Interests: e authors declare no competing interests.
Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Cre-
ative Commons license, and indicate if changes were made. e images or other third party material in this
article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons license and your intended use is not per-
mitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the
copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
© e Author(s) 2018
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Nei neonati, ha proprietà e funzioni simili al tessuto adiposo bruno, con una limitata flessibilità e reattività ai fattori esterni. Con l'invecchiamento, gli adipociti epicardici diventano più sensibili a fattori ambientali, metabolici ed emodinamici, che modificano gradualmente la funzione di EAT dalla termogenesi all'immagazzinamento di energia [9]. Tali cambiamenti si riflettono anche a livello strutturale: la proporzione di adipociti bruni diminuisce a favore di un maggior numero di adipociti bianchi uniloculari [8]. ...
... Tali cambiamenti si riflettono anche a livello strutturale: la proporzione di adipociti bruni diminuisce a favore di un maggior numero di adipociti bianchi uniloculari [8]. Anche in condizioni ischemiche croniche l'attività termogenica di EAT è ridotta [9]. Tuttavia, il tessuto può essere indotto a ripristinare la sua funzione originale e esercitare gli effetti benefici sul tessuto miocardico [9]. ...
... Anche in condizioni ischemiche croniche l'attività termogenica di EAT è ridotta [9]. Tuttavia, il tessuto può essere indotto a ripristinare la sua funzione originale e esercitare gli effetti benefici sul tessuto miocardico [9]. ...
Article
Full-text available
Sommario Il tessuto adiposo epicardico (EAT) è un fattore di rischio cardiovascolare in quanto promuove la progressione della fibrillazione atriale, della malattia coronarica e dell’insufficienza cardiaca. EAT si caratterizza per rapido metabolismo, misurabilità clinica e facile modificabilità e rappresenta un bersaglio terapeutico peculiare per farmaci innovativi, quali gli agonisti del recettore del peptide glucagone-simile 1 e gli inibitori del co-trasportatore sodio-glucosio 2, che appaiono salutari dal punto di vista cardiometabolico ben oltre i loro effetti sul glucosio e sul peso corporeo (Materiale Supplementare).
... To counteract this, an increased expression in pro-inflammatory cytokine genes occurs. In addition to these molecular changes, the sheer quantity of brown adipocytes also decreases in favor of white adipocytes [10][11][12]. Countering the gene expression that reduces BAT-like function of the EAT is a potential therapeutic target for patients with ischemic cardiovascular disease [13]. ...
Article
Full-text available
Epicardial adipose tissue is a unique visceral adipose tissue depot that plays a crucial role in myocardial metabolism. Epicardial adipose tissue is a major source of energy and free fatty acids for the adjacent myocardium. However, under pathological conditions, epicardial fat can affect the heart through the excessive and abnormal influx of lipids. The cardio-lipotoxicity of the epicardial adipose tissue is complex and involves different pathways, such as increased inflammation, the infiltration of lipid intermediates such as diacylglycerol and ceramides, mitochondrial dysfunction, and oxidative stress, ultimately leading to cardiomyocyte dysfunction and coronary artery ischemia. These changes can contribute to the pathogenesis of various cardio-metabolic diseases including atrial fibrillation, coronary artery disease, heart failure, and obstructive sleep apnea. Hence, the role of the cardio-lipotoxicity of epicardial fat and its clinical implications are discussed in this review.
... Chronic ischemic conditions, particularly in advanced CVDs, further diminish the brown fat-like activity within EAT [58]. Patients with advanced CAD displayed a downregulation of genes associated with adipocyte browning and thermogenic activation in EAT, coupled with a reciprocal increase in the expression of genes encoding pro-inflammatory cytokines. ...
Article
Full-text available
Implications of epicardial adipose tissue (EAT) on the development of coronary artery disease (CAD) have garnered recent attention. Located between the myocardium and visceral pericardium, EAT possesses unique morphological and physiological contiguity to the heart. The transcriptome and secretome of EAT differ from that of other fat stores in the body. Physiologically, EAT protects the adjacent myocardium through its brown-fat-like thermogenic function and rapid fatty acid oxidation. However, EAT releases pro-inflammatory mediators acting on the myocardium and coronary vessels, thus contributing to the development and progression of cardiovascular diseases (CVD). Furthermore, EAT-derived mesenchymal stromal cells indicate promising regenerative capabilities that offer novel opportunities in cell-based cardiac regeneration. This review aims to provide a comprehensive understanding and unraveling of EAT mechanisms implicated in regulating cardiac function and regeneration under pathological conditions. A holistic understanding of the multifaceted nature of EAT is essential to the future development of pharmacological and therapeutic interventions for the management of CVD.
... It has peculiar characteristics that differentiate it from the other types of visceral adipose tissue; EAT shares properties with both white and brown adipose tissue, so the term "beige" adipose tissue has been proposed to appropriately define it [1]. EAT plays several roles in the regulation of cardiac function and has a profound and complex interaction with the heart: it represents a storage of energy resources for the cardiac cells (in the older age), and it has thermogenesis properties (in the youngest) [2]; through endocrine and paracrine interactions, it mediates inflammatory response and modulates the immune system [3,4]; finally, ganglionated plexi, key structures of the autonomic nervous system of the heart, are located in the EAT. ...
Article
Full-text available
The relationship between epicardial adipose tissue (EAT) and atrial fibrillation (AF) has gained interest in recent years. The previous literature on the topic presents great heterogeneity, focusing especially on computed tomography imaging. The aim of the present study is to determine whether an increased volume of left atrial (LA) EAT evaluated at routine pre-procedural cardiac magnetic resonance imaging (MRI) relates to AF recurrences after catheter ablation. A total of 50 patients undergoing AF cryoballoon ablation and pre-procedural cardiac MRI allowing quantification of LA EAT were enrolled. In one patient, the segmentation of LA EAT could not be achieved. After a median follow-up of 16.0 months, AF recurrences occurred in 17 patients (34%). The absolute volume of EAT was not different in patients with and without AF recurrences (10.35 mL vs. 10.29 mL; p-value = 0.963), whereas the volume of EAT indexed on the LA volume (EATi) was lower, albeit non-statistically significant, in patients free from arrhythmias (12.77% vs. 14.06%; p-value = 0.467). The receiver operating characteristic curve testing the ability of LA EATi to predict AF recurrence after catheter ablation showed sub-optimal performance (AUC: 0.588). The finest identified cut-off of LA EATi was 10.65%, achieving a sensitivity of 0.5, a specificity of 0.82, a positive predictive value of 0.59 and a negative predictive value of 0.76. Patients with values of LA EATi lower than 10.65% showed greater survival, free from arrhythmias, than patients with values above this cut-off (84% vs. 48%; p-value = 0.04). In conclusion, EAT volume indexed on the LA volume evaluated at cardiac MRI emerges as a possible independent predictor of arrhythmia recurrence after AF cryoballoon ablation. Nevertheless, prospective studies are needed to confirm this finding and eventually sustain routine EAT evaluation in the management of patients undergoing AF catheter ablation.
... Coronary artery disease (CAD) is a dynamic process involving the accumulation of atherosclerotic plaques and functional alterations in coronary circulation. Adverse cardiometabolic conditions, such as chronic and long-term ischemic circumstances typically seen in CAD, can lead to a shift in EAT phenotype and biology, resulting in a pro-inflammatory profile [9]. ...
Article
Full-text available
Background: this study aimed to assess the complex relationship between EAT thickness, as measured with echocardiography, and the severity of coronary artery disease (CAD). We investigated whether individuals with higher EAT thickness underwent coronary revascularization. Subsequently, we conducted a three-year follow-up to explore any potential modifications in EAT depots post-angioplasty. Methods: we conducted a prospective and retrospective cross-sectional observational study involving 150 patients consecutively referred for acute coronary syndrome, including ST-elevation myocardial infarction (STEMI), non-ST elevation myocardial infarction (NSTEMI), and unstable angina. Upon admission (T0), all patients underwent coronary angiography to assess the number of pathologic coronary vessels. Percutaneous transluminal coronary angioplasty (PTCA) was performed based on angiogram results if indicated. The sample was categorized into two groups: non-revascularized (no-PTCA) and revascularized (PTCA). Transthoracic echocardiograms to measure epicardial fat thickness were conducted at admission (T0) and after a 3-year follow-up (T1). Results and conclusions: findings revealed a positive correlation between EAT thickness and the severity of coronary artery disease (CAD), with patients undergoing PTCA showing decreased EAT thickness after three years. Echocardiography demonstrated reliability in assessing EAT, offering potential for risk stratification. The study introduces a cut-off value of 0.65 cm as a diagnostic tool for cardiovascular risk. Incorporating EAT measurements into clinical practice may lead to more precise risk stratification and tailored treatment strategies, ultimately reducing the burden of cardiovascular disease.
... Attention to the role of epicardial adipose tissue (EAT) in the development of atrial fibrillation (AF) has increased in recent years. EAT plays several roles in the regulation of cardiac function and has a profound and complex interaction with the heart: it represents a storage of energy resources for the cardiac cells (in the older age), and it has thermogenesis properties (in the youngest) [Fainberg 2018]; through endocrine and paracrine interactions it mediates inflammatory response and modulates the immune system [Iacobellis 2004,Iacobellis 2009]; finally, ganglionated plexi, key structures of the autonomic nervous system of the heart, are located in the EAT. It has peculiar characteristics that differentiate it from the other types of visceral adipose tissue; EAT shares properties with both white and brown adipose tissue, so that the term "beige" adipose tissue has Disclaimer/Publisher's Note: The statements, opinions, and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). ...
Preprint
Full-text available
The relationship between epicardial adipose tissue (EAT) and atrial fibrillation (AF) has gained interest in recent years. Previous literature on the topic presents great heterogeneity, focusing especially on computed tomog-raphy imaging. Aim of the present study is to determine whether an increased volume of left atrial (LA) EAT evaluated at routine pre-procedural cardiac magnetic resonance imaging (MRI) relates to AF recurrences after catheter ablation. 50 patients undergoing AF cryoballoon ablation and pre-procedural cardiac MRI allowing quantification of LA EAT were enrolled. In one patient segmentation of LA EAT could not be achieved. After a median follow-up of 16.0 months, AF recurrences occurred in 17 patients (34%). Absolute volume of EAT was not different in patients with and without AF recurrences (10.35 ml vs. 10.29 ml; p-value=0.963), whereas volume of EAT indexed on the LA (EATi) was lower, albeit non statistically significant, in patients free from arrhythmias (12.77% vs. 14.06%; p-value=0.467). Receiver operating characteristic curve testing the ability of EATi to predict AF recurrence after catheter ablation showed sub-optimal performance (AUC: 0.588). The finest identified cut-off of EATi was 10.65%, achieving a sensitivity of 0.5, a specificity of 0.82, a positive predictive value of 0.59 and a negative predictive value of 0.76. Patients with values of EATi lower than 10.65% showed greater survival free from arrhythmias than patients with values above this cut-off (84% vs. 48%; p-value=0.04). In conclusion, indexed LA EAT volume evaluated at cardiac MRI independently relates to arrhythmia recurrence after AF cryoballoon ablation.
Preprint
Full-text available
Background The influence of epicardial adipose tissue (EAT) on cardiovascular health appears to be modulated by age, metabolic status, and underlying cardiac pathology.The relationship between EAT and pericardial adipose tissue (PAT) remains unclear.The impact of epicardial and pericardial adipose tissue (EPAT) on human health remains unclear. This study aimed to elucidate the causal relationships between EPAT and various health outcomes using large-scale genetic data. Methods We conducted phenome-wide association studies (PheWAS) using data from FinnGen (n = 412,181) and UK Biobank (n > 500,000) to identify EPAT-associated traits. Two-sample Mendelian randomization (MR) analyses were performed to assess causal relationships between EPAT and identified outcomes. Mediation analyses explored potential pathways through which EPAT exerts its effects. Results PheWAS revealed 171 and 181 EPAT-associated traits in FinnGen and UK Biobank, respectively, including cardiovascular, metabolic, psychiatric, and respiratory diseases.EPAT showed protective causal relationships with type 2 diabetes (OR 0.91, 95% CI 0.86 to 0.96, P = 0.0009), high cholesterol (OR 0.88, 95% CI 0.78 to 0.99, P = 0.04), adult-onset asthma (OR 9.49×10⁻⁴¹, 95% CI 8.98×10⁻⁴⁷ to 1.00×10⁻³⁴, P = 9.14×10⁻³⁹), and bipolar disorder (OR 3.61×10⁻⁴¹, 95% CI 2.91×10⁻⁴⁷ to 4.47×10⁻³⁵, P = 1.09×10⁻³⁸). EPAT was also associated with increased testosterone levels (β = 0.25, 95% CI 0.04 to 0.46, P = 0.02) and enhanced right ventricular ejection fraction (β = 7.26, 95% CI 1.34 to 13.18, P = 0.02). Mediation analyses revealed that these effects were partially mediated by various factors, including plasma proteins (e.g., LRRN1 for type 2 diabetes), sex hormone-binding globulin (for high cholesterol), insulin-like growth factor 1 (for testosterone levels), specific immune cells (for asthma), and cerebrospinal fluid metabolites (for bipolar disorder). Conclusion This study reveals a complex and multifaceted role of EPAT in human health, extending beyond its established role in cardiovascular disease. Our findings indicate that EPAT could be a promising therapeutic target for multiple diseases. Potential drug development strategies include reversing harmful EPAT to a beneficial state or maintaining its beneficial properties long-term.
Article
Objective Epicardial adipose tissue (EAT) quantity is associated with poor cardiovascular outcomes. However, the quality of EAT may be of incremental prognostic value. Cardiac magnetic resonance (CMR) is the gold standard for tissue characterization but has never been applied for EAT quality assessment. We aimed to investigate EAT quality measured on CMR T1 mapping as a predictor of poor outcomes in an all‐comer cohort. Methods We investigated the association of EAT area and EAT T1 times (EAT‐T1) with a composite endpoint of nonfatal myocardial infarction, heart failure hospitalization, and all‐cause death. Results A total of 966 participants were included (47.2% female; mean age: 58.4 years) in this prospective observational CMR registry. Mean EAT area and EAT‐T1 were 7.3 cm ² and 268 ms, respectively. On linear regression, EAT‐T1 was not associated with markers of obesity, dyslipidemia, or comorbidities such as diabetes ( p > 0.05 for all). During a follow‐up of 57.7 months, a total of 280 (29.0%) events occurred. EAT‐T1 was independently associated (adjusted hazard ratio per SD: 1.202; 95% CI: 1.022–1.413; p = 0.026) with the composite endpoint when adjusted for established clinical risk. Conclusions EAT quality (as assessed via CMR T1 times), but not EAT quantity, is independently associated with a composite endpoint of nonfatal myocardial infarction, heart failure hospitalization, and all‐cause death. image
Article
Epicardial adipose tissue (EAT) is located between the heart muscle and visceral pericardium, where it has direct contact with coronary blood vessels. Elevated thickness of this tissue can induce local inflammation affecting the myocardium and the underlying coronary arteries, contributing to various cardiovascular diseases such as coronary artery disease, atrial fibrillation, or heart failure with preserved ejection fraction. Recent studies have identified EAT thickness as a simple and reliable biomarker for certain cardiovascular outcomes. Examples include the presence of atherosclerosis, incident cardiovascular disease (CVD) in individuals with type 2 diabetes mellitus (T2DM), and the prevalence of atrial fibrillation. Furthermore, EAT measurements can help to identify patients with a higher risk of developing metabolic syndrome. Since the EAT thickness can be easily measured using echocardiography, such examinations could serve as a useful and cost-effective preventive tool for assessing cardiovascular health. This review also summarizes therapeutical interventions aimed at reducing EAT. Reducing EAT thickness has been shown to be possible through pharmacological, surgical, or lifestyle-change interventions. Pharmaceutical therapies, including thiazolidinediones, glucagon-like peptide 1-receptor agonists, sodium-glucose cotransporter 2 inhibitors, dipeptidyl peptidase-4 inhibitors, and statins, have been shown to influence EAT thickness. Additionally, EAT thickness can also be managed more invasively through bariatric surgery, or noninvasively through lifestyle changes to diet and exercise routines.
Article
Full-text available
Brown adipose tissue acting through a unique uncoupling protein (UCP1) has a critical role in preventing hypothermia in new-born sheep but is then considered to rapidly disappear during postnatal life. The extent to which the anatomical location of fat influences postnatal development and thermogenic function, particularly following feeding, in adulthood, are not known and were both examined in our study. Changes in gene expression of functionally important pathways (i.e. thermogenesis, development, adipogenesis and metabolism) were compared between sternal and retroperitoneal fat depots together with a representative skeletal muscle over the first month of postnatal life, coincident with the loss of brown fat and accumulation of white fat. In adult sheep, implanted temperature probes were used to characterise the thermogenic response of fat and muscle to feeding and the effects of reduced or increased adiposity. UCP1 was more abundant within sternal than retroperitoneal fat and was only retained in the sternal depot of adults. Distinct differences in the abundance of gene pathway markers were apparent between tissues, with sternal fat exhibiting some similarities with muscle that were not apparent in the retroperitoneal depot. In adults, the post-prandial rise in temperature was greater and more prolonged in sternal than retroperitoneal fat and muscle, a difference that was maintained with altered adiposity. In conclusion, sternal adipose tissue retains UCP1 into adulthood when it shows a greater thermogenic response to feeding than muscle and retroperitoneal fat. Sternal fat may be more amenable to targeted interventions that promote thermogenesis in large mammals.
Article
Full-text available
Sustained β3 adrenergic receptor (ADRB3) activation simultaneously upregulates fatty acid synthesis and oxidation in mouse brown, beige, and white adipose tissues; however, the cellular basis of this dual regulation is not known. Treatment of mice with the ADRB3 agonist CL316,243 (CL) increased expression of fatty acid synthase (FASN) and medium chain acyl-CoA dehydrogenase (MCAD) protein within the same cells in classic brown and white adipose tissues. Surprisingly, in inguinal adipose tissue, CL-upregulated FASN and MCAD in distinct cell populations: high MCAD expression occurred in multilocular adipocytes that co-expressed UCP1+, whereas high FASN expression occurred in paucilocular adipocytes lacking detectable UCP1. Genetic tracing with UCP1-cre, however, indicated nearly half of adipocytes with a history of UCP1 expression expressed high levels of FASN without current expression of UCP1. Global transcriptomic analysis of FACS-isolated adipocytes confirmed the presence of distinct anabolic and catabolic phenotypes, and identified differential expression of transcriptional pathways known to regulate lipid synthesis and oxidation. Surprisingly, paternally-expressed genes of the non-classical gene imprinted network were strikingly enriched in anabolic phenotypes, suggesting possible involvement in maintaining the balance of metabolic phenotypes. The results indicate that metabolic heterogeneity is a distinct property of activated beige/brite adipocytes that might be under epigenetic control.
Article
Full-text available
Insulin resistance is a key mediator of obesity-related cardiometabolic disease, yet the mechanisms underlying this link remain obscure. Using an integrative genomic approach, we identify 53 genomic regions associated with insulin resistance phenotypes (higher fasting insulin levels adjusted for BMI, lower HDL cholesterol levels and higher triglyceride levels) and provide evidence that their link with higher cardiometabolic risk is underpinned by an association with lower adipose mass in peripheral compartments. Using these 53 loci, we show a polygenic contribution to familial partial lipodystrophy type 1, a severe form of insulin resistance, and highlight shared molecular mechanisms in common/mild and rare/severe insulin resistance. Population-level genetic analyses combined with experiments in cellular models implicate CCDC92, DNAH10 and L3MBTL3 as previously unrecognized molecules influencing adipocyte differentiation. Our findings support the notion that limited storage capacity of peripheral adipose tissue is an important etiological component in insulin-resistant cardiometabolic disease and highlight genes and mechanisms underpinning this link.
Article
Full-text available
Studies in rodents and newborn humans demonstrate the influence of brown adipose tissue (BAT) in temperature control and energy balance and a critical role in the regulation of body weight. Here, we obtained samples of epicardial adipose tissue (EAT) from neonates, infants, and children in order to evaluate changes in their transcriptional landscape by applying a systems biology approach. Surprisingly, these analyses revealed that the transition to infancy is a critical stage for changes in the morphology of EAT and is reflected in unique gene expression patterns of a substantial proportion of thermogenic gene transcripts (~10%). Our results also indicated that the pattern of gene expression represents a distinct developmental stage, even after the rebound in abundance of thermogenic genes in later childhood. Using weighted gene coexpression network analyses, we found precise anthropometric-specific correlations with changes in gene expression and the decline of thermogenic capacity within EAT. In addition, these results indicate a sequential order of transcriptional events affecting cellular pathways, which could potentially explain the variation in the amount, or activity, of BAT in adulthood. Together, these results provide a resource to elucidate gene regulatory mechanisms underlying the progressive development of BAT during early life.
Article
Full-text available
The discovery of brown adipose tissue (BAT) in adult humans presents a new therapeutic target for metabolic disease; however, little is known about the regulation of human BAT. Chronic glucocorticoid excess causes obesity in humans, and glucocorticoids suppress BAT activation in rodents. We tested whether glucocorticoids regulate BAT activity in humans. In vivo, the glucocorticoid prednisolone acutely increased 18fluorodeoxyglucose uptake by BAT (measured using PET/CT) in lean healthy men during mild cold exposure (16°C–17°C). In addition, prednisolone increased supraclavicular skin temperature (measured using infrared thermography) and energy expenditure during cold, but not warm, exposure in lean subjects. In vitro, glucocorticoids increased isoprenaline-stimulated respiration and UCP-1 in human primary brown adipocytes, but substantially decreased isoprenaline-stimulated respiration and UCP-1 in primary murine brown and beige adipocytes. The highly species-specific regulation of BAT function by glucocorticoids may have important implications for the translation of novel treatments to activate BAT to improve metabolic health.
Article
Full-text available
To produce milk that is healthier for human consumption, the present study evaluated the effect of including canola oil in the diet of dairy cows on milk production and composition as well as the nutritional quality of this milk fat. Eighteen Holstein cows with an average daily milk yield of 22 (± 4) kg/d in the middle stage of lactation were used. The cows were distributed in 6 contemporary 3x3 Latin squares consisting of 3 periods and 3 treatments: control diet (without oil), 3% inclusion of canola oil in the diet and 6% inclusion of canola oil in the diet (dry matter basis). The inclusion of 6% canola oil in the diet of lactating cows linearly reduced the milk yield by 2.51 kg/d, short-chain fatty acids (FA) by 41.42%, medium chain FA by 27.32%, saturated FA by 20.24%, saturated/unsaturated FA ratio by 39.20%, omega-6/omega-3 ratio by 39.45%, and atherogenicity index by 48.36% compared with the control treatment. Moreover, with the 6% inclusion of canola oil in the diet of cows, there was an increase in the concentration of long chain FA by 45.91%, unsaturated FA by 34.08%, monounsaturated FA by 40.37%, polyunsaturated FA by 17.88%, milk concentration of omega-3 by 115%, rumenic acid (CLA) by 16.50%, oleic acid by 44.87% and h/H milk index by 94.44% compared with the control treatment. Thus, the inclusion of canola oil in the diet of lactating dairy cows makes the milk fatty acid profile nutritionally healthier for the human diet; however, the lactating performance of dairy cows is reduce.
Article
Full-text available
Uncoupling protein 1 (Ucp1) contributes to thermogenesis, and its expression is regulated at the transcriptional level. Here, we show that Ucp1 expression is also regulated post-transcriptionally. In inguinal white adipose tissue (iWAT) of mice fed a high-fat diet (HFD), Ucp1 level decreases concomitantly with increases in Cnot7 and its interacting partner Tob. HFD-fed mice lacking Cnot7 and Tob express elevated levels of Ucp1 mRNA in iWAT and are resistant to diet-induced obesity. Ucp1 mRNA has an elongated poly(A) tail and persists in iWAT of Cnot7(-/-) and/or Tob(-/-) mice on a HFD. Ucp1 3'-UTR-containing mRNA is more stable in cells expressing mutant Tob that is unable to bind Cnot7 than in WT Tob-expressing cells. Tob interacts with BRF1, which binds to an AU-rich element in the Ucp1 3'-UTR. BRF1 knockdown partially restores the stability of Ucp1 3'-UTR-containing mRNA. Thus, the Cnot7-Tob-BRF1 axis inhibits Ucp1 expression and contributes to obesity.
Article
Intra-uterine growth restriction in late pregnancy can contribute to adverse long term metabolic health in the offspring. We utilised an animal (sheep) model of maternal dietary manipulation in late pregnancy, combined with exposure of the offspring to a low activity, obesogenic environment after weaning, to characterise the effects on glucose homeostasis. Dizygotic twin-pregnant sheep were either fed to 60% of requirements (nutrient restriction (R)) or fed ad libitum (~ 140% of requirements (A)) from 110 days gestation until term (~147d). After weaning (~3 months of age), their offspring were kept in either a standard (in order to remain lean) or low activity, obesogenic environment. R mothers gained less weight and produced smaller offspring. As adults, obese offspring were heavier and fatter with reduced glucose tolerance, irrespective of maternal diet. Molecular markers of stress and autophagy in liver and adipose tissue were increased with obesity, with gene expression of hepatic Grp78 and of omental Atf6, Grp78 and Edem1 only being increased in R offspring. In conclusion, the adverse effect of juvenile onset obesity on insulin responsive tissues can be amplified by previous exposure to a suboptimal nutritional environment in utero, thereby contributing to earlier onset of insulin resistance.