You are on page 1of 19

Review Article

Metabolic syndrome and related conditions


pISSN: 2287-4208 / eISSN: 2287-4690
World J Mens Health Published online Jan 19, 2023
https://doi.org/10.5534/wjmh.220224

Thermogenic Brown Fat in Humans: Implications


in Energy Homeostasis, Obesity and Metabolic
Disorders
Masayuki Saito , Yuko Okamatsu-Ogura
Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan

In mammals including humans, there are two types of adipose tissue, white and brown adipose tissues (BATs). White adipose
tissue is the primary site of energy storage, while BAT is a specialized tissue for non-shivering thermogenesis to dissipate en-
ergy as heat. Although BAT research has long been limited mostly in small rodents, the rediscovery of metabolically active
BAT in adult humans has dramatically promoted the translational studies on BAT in health and diseases. It is now established
that BAT, through its thermogenic and energy dissipating activities, plays a role in the regulation of body temperature, whole-
body energy expenditure, and body fatness. Moreover, increasing evidence has demonstrated that BAT secretes various para-
crine and endocrine factors, which influence other peripheral tissues and control systemic metabolic homeostasis, suggesting
BAT as a metabolic regulator, other than for thermogenesis. In fact, clinical studies have revealed an association of BAT not
only with metabolic disorders such as insulin resistance, diabetes, dyslipidemia, and fatty liver, but also with cardiovascular
diseases including hypertension and atherosclerosis. Thus, BAT is an intriguing tissue combating obesity and related metabol-
ic diseases. In this review, we summarize current knowledge on human BAT, focusing its patho-physiological roles in energy
homeostasis, obesity and related metabolic disorders. The effects of aging and sex on BAT are also discussed.

Keywords: Aging; Brown adipose tissue; Energy expenditure; Metabolic syndrome; Obesity; Sex

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0)
which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

INTRODUCTION of triglyceride (TG) in adipose tissues because of a pro-


longed positive energy balance. In mammals including
We have been facing a worldwide pandemic of obe- humans, there are two types of adipose tissue, white
sity, which is closely associated with not only muscu- and brown adipose tissues (BATs). The two tissues are
loskeletal disorders but also common diseases such as similar in their major population of adipocyte having
diabetes mellitus, dyslipidemias, fatty liver, hyperten- intracellular lipid droplets, but are quite different in
sion, and arteriosclerosis. Moreover, obesity predisposes the physiological functions. White adipose tissue (WAT)
to adverse outcomes in some diseases, as in the ex- is the primary site of energy storage, while BAT is
ample of the increased mortality in patients with CO- a specialized tissue for non-shivering thermogenesis
VID-19. Obesity is the state of excessive accumulation (NST) to dissipate energy as heat.

Received: Oct 12, 2022 Accepted: Nov 8, 2022 Published online Jan 19, 2023
Correspondence to: Masayuki Saito https://orcid.org/0000-0002-3058-3003
Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Kita-ku, Kita18 Nishi-9, Sapporo 060-0818, Japan.
Tel: +81-11-706-5205, Fax: +81-11-757-0703, E-mail: saito@vetmed.hokudai.ac.jp

Copyright © 2023 Korean Society for Sexual Medicine and Andrology


https://doi.org/10.5534/wjmh.220224

Although BAT research has long been limited mostly BAT-dependent CIT, together with skeletal muscle
in small rodents, the rediscovery of metabolically active shivering, plays a role in the maintenance of body
BAT using fluorodeoxyglucose (FDG)-positron emission temperature under cold conditions. In fact, BAT ther-
tomography (PET) and computed tomography (CT) in mogenesis is activated by acute cold exposure, and mice
adult humans [1-4] has dramatically accelerated the deficient of uncoupling protein 1 (UCP1), a key mito-
translational studies on BAT in health and diseases. It chondrial molecule for BAT thermogenesis, are cold-in-
is now established that BAT, through its thermogenic tolerant and cannot maintain their body temperature
and energy dissipating activities, plays a role in the after acute cold exposure [10,11]. Similarly, in humans,
regulation of body temperature, energy expenditure cold exposure activates BAT as reflected as increased
(EE), and body fatness [5,6]. Moreover, over a past de- uptake of FDG and fatty acid derivatives on PET im-
cade, increasing evidence has demonstrated that BAT aging (Fig. 1A) [1,2,4,12-16]. Moreover, the cold-induced
cross talks with some peripheral tissues and controls metabolic activation of BAT correlates positively with
their functions, systemic homeostasis of energy and CIT (Fig. 1B, 1C) [12,17,18], confirming a significant role
metabolic substrates, suggesting BAT as a metabolic of BAT for CIT in humans. Now, cold-induced FDG up-
regulator beyond thermogenesis [7-9]. In this review, we take is used as a surrogate of activity and amount of
summarize current knowledge on human BAT, with BAT in humans.
reference to its patho-physiological roles in energy ho- EE above the basal metabolic rate in response to
meostasis, obesity and metabolic diseases. meal intake is referred to as the “specific dynamic ac-
tion of food” or “diet-induced thermogenesis (DIT)”,
BROWN FAT AS A SITE OF ENERGY which is another component of NST [19,20]. As men-
EXPENDITURE BY NON-SHIVERING tioned above, cold exposure is the most potent and
THERMOGENESIS physiological stimulus to activate BAT, but BAT-de-
pendent CIT would be very low in our daily life under
In general, daily energy consumption is roughly di- well-controlled conditions with the presence of clothing
vided into three components: basal/resting EE, exercise/ and heating systems. In contrast, DIT is observed after
physical activity-associated EE, and NST. A represen- every meal intake independently of environmental
tative example of NST is cold-induced thermogenesis temperature, and estimated as about 10% of ingested
(CIT), an increased EE seen after cold exposure. It is energy of food. DIT is usually divided into two compo-
now well-established that BAT is a site of CIT, and nents: obligatory and facultative thermogenesis. Oblig-

A 18
F-FDG-PET/CT

BAT-negative BAT-positive
Fig. 1. Brown adipose tissue (BAT), en-
ergy expenditure (EE), and non-shivering
B C D
Basal EE CIT DIT thermogenesis. (A) BAT detected by 18F-
Energy expenditure (kcal/d)

Energy expenditure (kcal/d)

Energy expenditure (kcal/d)

1,600 800 ** FDG-PET/CT after acute cold exposure.


300 15 *
(B) Basal EE under a warm condition.
1,500 600 (C) Cold-induced thermogenesis (CIT)
200 10 after acute cold exposure. (D) Diet-
%

1,400 400
induced thermogenesis (DIT) after meal
100 5 intake. *p<0.05, **p<0.01. Adapted
1,300 200
from Yoneshiro et al (Obeisty [Silver
Spring] 2011;19:13-6) [12] and con-
0 0 0 0
+ + + structed from Hibi et al (Int J Obes [Lond]
BAT BAT BAT 2016;40:1655-61) [29].

2 www.wjmh.org
Masayuki Saito and Yuko Okamatsu-Ogura: Thermogenic Brown Fat in Humans

atory thermogenesis refers to the obligatory response mogenesis and mitochondrial substrate oxidation [27].
including digestion, absorption, and storage of ingested Din et al [28] demonstrated that oxygen consumption
nutrients, whereas facultative thermogenesis refers to and blood flow in BAT rose immediately after meal
the additional responses to obligatory thermogenesis intake to an extent comparable to those observed after
and may be closely related to BAT activation. Activa- cold exposure. To confirm the role of BAT in DIT, we
tion of BAT after food intake has been proved in small measured whole-body EE continuously for 24 hours
rodents. For example, a single meal ingestion produced in healthy humans using a human calorimeter [29,30].
a rapid increase in tissue respiration [21], glucose uti- When the participants were divided into BAT-positive
lization and fatty acid synthesis in intact BAT of rats, and -negative groups according to the result of FDG-
but to a much lower extent in surgically denervated PET/CT examination, there was no significant differ-
BAT [22]. Moreover, EE after food intake is lower in ence in body composition and basal EE between the
UCP1-deficient mice than in wild-type mice [23] two groups. However, EE after meals was significantly
In humans, the possible contribution of BAT thermo- higher in the BAT-positive group (9.7% of the total
genesis to DIT was suggested by Nagai et al [24], who energy intake) than in the BAT-negative group (6.5%)
showed that single nucleotide polymorphism (SNP) in (Fig. 1D), suggesting that about 3% is BAT-dependent.
the UCP1 gene is associated with reduced DIT. Redis- All these results indicate that BAT contributes to DIT,
covery of BAT in adult humans has prompted further at least in part, in humans.
studies to test whether BAT thermogenesis is activated
after single meals. Some studies using FDG-PET/CT re- MECHANISMS OF BROWN-FAT
vealed a reduced or unexpectedly low FDG uptake into ASSOCIATED NON-SHIVERING
BAT after meal intake [25,26]. Although these results THERMOGENESIS
seem conflicting with the idea of postprandial activa-
tion of BAT thermogenesis, they can be explained by The mechanism of cold-induced activation of BAT
increased insulin-stimulated FDG uptake into skeletal has extensively been investigated in vivo and in vitro,
muscle, which reduces FDG bioavailability for BAT, and is schematically depicted in Fig. 2. When animals
which in turn leads to underestimation of BAT activ- are exposed to cold temperatures, cold is perceived by
ity [26]. temperature sensors, transient receptor potential (TRP)
This limitation of FDG-PET/CT is overcome by mea- channels, which are membrane proteins that transmit
suring oxygen uptake using 15O[O2]-PET and blood flow information about changes in the environment such as
using 15O[H2O]-PET, which are direct indicators of ther- temperature, touch, pain, osmolarity, and naturally oc-

Diet-induced Cold-induced
thermogenesis thermogenesis

Glossopharyngeal/ Somato-
facial N sensory N
Taste TRP
receptors
Food Cold

Sympathetic N
Vagus N
NA
Fig. 2. Neuro-endocrine mechanisms of
AR
CCK cold- and diet-induced brown fat ther-
Secretin SCTR mogenesis. βAR: β-adrenergic receptor,
CCK: cholecystokinin, SCTR: secretin
Bile acids TGR5 UCP1 receptor, N: nerve, NA: noradrenaline,
TGR5: G-protein-coupled bile acid-
activated receptor, TRP: transient recep-
Brown/beige Heat tor potential channel, UCP1: uncoupling
adipocyte protein 1.

www.wjmh.org 3
https://doi.org/10.5534/wjmh.220224

curring substances. Cold-activated TRP on sensory neu- expression of the secretin receptor in murine brown
rons on the body surface transmit information to the adipocytes, and demonstrated that secretin activates
brain and increase the activity of sympathetic nerves UCP1- and secretin receptor-dependent thermogenesis
(SNs). Noradrenaline (NA) released from SN endings in vitro and in vivo. They also confirmed in humans
stimulates brown adipocytes via the β-adrenergic recep- that the increment of plasma secretin levels induced
tor (βAR) and triggers intracellular events including by a single meal positively correlated with BAT activ-
hydrolysis of TG, oxidation of resulting fatty acids, and ity, and that secretin infusion increased FDG uptake
activates UCP1. in BAT. In addition to secretin, other gut hormones
When animals are exposed to cold temperatures for a such as cholecystokinin and glucagon-like peptide 1
long time, they adapt to their surroundings by increas- were suggested to triggers BAT thermogenesis in small
ing the number of brown adipocytes and the amount of rodents through the vagal afferent and sympathetic
UCP1 through the proliferation of interstitial preadi- nervous system [46,47]. A stimulatory role of the va-
pocytes and matured adipocytes [31,32]. In addition to gal afferent in BAT thermogenesis was also reported
BAT hyperplasia, prolonged cold exposure gives rise to in humans [48]. Another humoral factor may be bile
an apparent induction of UCP1-positive adipocytes in acids, which are secreted into the intestinal lumen in
WAT. This type of adipocytes, termed “beige” or “brite” response to meal intake and modified by gut flora. Bile
cells, is developmentally distinct from “classical” brown acids are now recognized as a metabolic regulator, af-
adipocytes [33,34], and has the thermogenic potential in fecting multiple functions, in addition to lipid-digestive
response to βAR stimulation [35,36]. Thus, chronic cold functions, to regulate energy metabolism, as well as
exposure results in increased EE through the persis- glucose and lipid metabolism [49]. In fact, bile acids
tent activation and recruitment of classical brown adi- were reported to activate BAT thermogenesis in mice
pocytes and beige cells, and the consequent “browning” and humans [50-52]. Thus, it is conceivable that these
of WAT and body fat reduction. As the UCP1-positive multiple gut derived factors, together with the SN
human adipose depot consists of a mixture of brown system, participate in BAT-associated DIT, but further
and beige adipocytes [37-39], here we refer to it collec- studies are needed to uncover the detailed neuroendo-
tively as BAT and thermogenic adipocytes. crine mechanisms of DIT in humans.
Several mechanisms/factors have been proposed to
participate in postprandial BAT activation. Based on BROWN FAT AS A REGULATOR OF
the principal role of the SN-βAR axis for CIT, it is con- ENERGY BALANCE AND BODY FAT
ceivable that this axis is also a key mechanism in BAT-
associated DIT (Fig. 2). In fact, in both experimental Consistent with the significant role of BAT in NST
animals and humans, the plasma levels of NA and tis- and short-term regulation of EE, there is substantial
sue NA turnover are low during fasting but increases evidence for BAT as a long-term regulator of EE and
immediately after food intake [40-43]. We [22] found in body fatness: almost all obese model animals express
rats that postprandial metabolic activation of BAT was lower levels of UCP1 in BAT, while mice over-express-
diminished either after surgical severing of SNs enter- ing UCP1 are leaner [53,54]. Mice lacking UCP1 get
ing BAT or by giving a meal through a gastric tube. obese when they are kept at thermoneutral tempera-
LeBlanc et al [40] and Diamond et al [44] showed in tures [55] or on a high-fat diet [56].
humans and dogs that responses in oxygen consump- In humans, studies on SNP in some BAT-related
tion, and plasma levels of NA and insulin shortly after genes have suggested a significant contribution of
food intake were substantially reduced when food was BAT thermogenesis to regulation of energy balance
administered through a stomach tube. These results and body fatness. For example, Trp64Arg mutation
collectively suggest a significant role of sympathetic in the β3AR gene and A3826G mutation in the UCP1
activation triggered by oropharyngeal taste sensation gene are associated with higher body fat content, lower
in BAT-associated DIT. metabolic rate, and smaller weight loss via treatment
Food intake evokes a rapid release of various gas- with low-calorie diets [57-60]. Consistently, retrospective
trointestinal hormones, some of which are likely fac- readings of FDG-PET/CT in thousands of patients have
tors participating in DIT. Li et al [45] found abundant revealed that BAT prevalence is lower in patients with

4 www.wjmh.org
Masayuki Saito and Yuko Okamatsu-Ogura: Thermogenic Brown Fat in Humans

higher BMI [3,61-64]. Prospective studies in healthy also been reported in various clinical studies [3,61,62].
participants also demonstrated that BAT-negative sub- The aging process produces increased fat mass and
jects are higher in their adiposity-related parameters decreased lean mass, therefore, it is possible that age-
than BAT-positive subjects (Fig. 3A) [1,2,65-67]. Thus, related accumulation of body fat is associated with de-
the inverse relationship between BAT and body fat- creased BAT activity. This is supported by the findings
ness is well confirmed (Fig. 3B). The apparent associa- that the adiposity-related parameters increased with
tion between BAT prevalence and adiposity, however, age in the BAT-negative group, while they remained
is to be carefully evaluated, because these are consider- unchanged from the twenties to forties in the BAT-
ably influenced by age. In fact, the mean age is lower positive group (Fig. 3D) [67]. BAT-associated NST is
in the BAT-positive participant group than the BAT- rather small (<100 kcal/day) in our daily life (Fig. 1C,
negative group (Fig. 3A). Detailed analysis revealed 1D), but it is maximally 36,500 kcal/year, equivalent to
that the prevalence of cold-activated BAT is more than 4 kg fat, which may be enough to explain the age-relat-
50% in young subjects of the twenties, decreased with ed accumulation of body fat. Such age-related changes
age, and in less than 10% of the fifties and sixties (Fig. in BAT will be discussed again in a latter section.
3C) [67]. A strong impact of age on BAT prevalence has

A Age (y) Age (y) B


40 20 0 0 20 40
Male Female
Age 100

Visceral fat (cm )


2
BMI 80

Total fat
60

Visceral fat BAT+


BAT
40
200 150 100 50 0 0 50 100 150 200 -0.5 0 0.5 1.0 1.5
2 2
Fat area (cm ) Fat area (cm ) BAT activity (relative)

C D E
Baby Young Old
60 120 * Energy
100 expenditure
BAT prevalence (%)

Visceral fat (cm )


Visceral fat (cm )

BAT
2

Lean
40
75 80

Inactivation Reactivation
20 of BAT of BAT
50 40 BAT+
2

Energy Obese
expenditure
0 25 0
20 30 40 50 >60 20 30 >40
29 39 49 59 29 39
Age (y) Age (y)
Fig. 3. Age-related changes in brown fat and body fatness. The prevalence and activity of brown adipose tissue (BAT) was assessed by FDG-PET/
CT combined with acute cold exposure. (A) Mean age and obesity-related parameters in subjects with detectable BAT (BAT+) and those without it
(BAT-). (B) Inverse correlation between BAT activity and visceral fat area. (C) Effects of age on BAT prevalence and visceral fat area. (D) Age-related
accumulation of visceral fat in subjects with detectable BAT (BAT+) and those without it (BAT-). (E) The thermogenic activity of BAT is high during
neonatal periods, but decreases with age in some individuals who get obese. In contrast, other individuals who keep BAT during their adulthood
do not get obese, suggesting that BAT is protective against age-related accumulation of body fat, and that its reactivation/recruitment is effective
for combating obesity. *p<0.05, **p<0.01. Constructed from Saito et al (Diabetes 2009;58:1526-31) [1] and Yoneshiro et al (Obesity [Silver Spring]
2011;19:1755-60) [67].

www.wjmh.org 5
https://doi.org/10.5534/wjmh.220224

ACTIVATION AND RECRUITMENT OF man BAT, it would seem difficult to increase exposure
BROWN FAT AS AN ANTI-OBESITY to cold in daily life. As shown in Fig. 2, cold stimulus
REGIMEN is received by TRP. Among the members of the TRP
family, TRPM8 and TRPA1 are the most likely recep-
The finding that BAT is protective against body fat tor candidates sensitive to low temperatures [74]. The
accumulation has encouraged the search how to acti- mean activation temperatures of TRPA1 and TRPM8
vate or recruit BAT (Fig. 3E). This is particularly in- are around 20°C, being comparable with those ap-
triguing because people with lower or undetectable BAT plied in human studies to activate BAT. Accordingly,
activities are more obese and to be treated. As noted chemical activation of these receptors would mimic
in previous sections, cold is the most physiological and the effects of cold exposure. Actually, there are various
powerful stimulus for activation and recruitment of ingredients in food acting as agonists for these TRPs
BAT. Cold exposure elicits increased proliferation and [74], a representative of which is menthol, a cooling and
differentiation of classical brown adipocyte but also flavor compound in mint, acting on TRPM8. TRPA1 is
a remarkable induction of beige adipocytes in WAT. activated by allyl- and benzyl-isothiocyanates, pungent
Some genes expressed selectively in mouse beige cells elements in mustard and Wasabi (Japanese horserad-
are also highly expressed in human supraclavicular fat ish). Among the TRP agonists, the most extensively
deposits identified as BAT by FDG-PET/CT [37,68,69]. studied is capsaicin, a pungent principle of chili pep-
Lee et al [70] reported that preadipocytes isolated from per, which is a potent agonist for a nociceptive receptor
human supraclavicular fat were capable of differenti- TRPV1. Both animal and human studies have dem-
ating into UCP1-positive adipocytes in vitro. Moreover, onstrated that capsaicin and its nonpungent analogue
we found that BAT activity in humans is remarkably (capsinoids) increase BAT thermogenesis through the
increased during winter in individuals who showed activation of TRPV1 and the SN system, and decrease
undetectable activities in summer [1]. All these facts body fat [71,75-78]. Thus, these food ingredients activat-
suggest that human BAT is largely composed of beige ing TRPs may be promising as an anti-obesity regimen
cells and inducible in response to appropriate stimula- easily applicable in daily life [79,80].
tion. In fact, when men with undetectable or low BAT Pharmacological activation of BAT thermogenesis
activity were kept in a cold environment for 2–6 hours targeting β3AR expressed abundantly in adipocytes has
every day for several weeks, their BAT activity was long been expected as an anti-obesity regimen, while the
significantly increased while body fat was decreased β3AR agonists so fat developed showed, more or less,
[71-73] (Fig. 4A, 4B). More importantly, the change in undesirable cardiovascular side effects in humans [81,82].
BAT activity was negatively correlated with those in Among them, mirabegron may be promising, because it
body fat content [71] (Fig. 4C). These results indicate induces WAT browning with minimal side effects. Clini-
that human BAT can be induced and/or recruited, and cally, mirabegron is widely used for the treatment of
is involved in reducing body fat. overactive bladder, because it relaxes the smooth muscle
Although daily cold exposure thus can recruit hu- of the bladder where β3AR is expressed [83]. Cypess et

A B C
6 0.4 3.0
*
2.0 R= 0.703
0.0 Fig. 4. Recruitment of brown fat and
BAT activity (SUV)

p=0.023
1.0 reduction of body fat by repeated cold
Body fat (kg)

Body fat (kg)

4
0.4 0.0 exposure. Participants were exposed to
either a cold (Cold) or warm (Control)
1.0
0.8 condition for 2 hours every day for 6
2
2.0 weeks. (A) Brown adipose tissue (BAT)
1.2 activity assessed by FDG-PET/CT. (B)
3.0
* Changes in body fat. (C) Correlation be-
0 1.6 4.0 tween the changes in body fat and BAT
0 6 Cold Control 2 0 2 4 6 activity. Adapted from Yoneshiro et al (J
Cold exposure (wk) BAT (SUV) Clin Invest 2013;123:3404-8) [71].

6 www.wjmh.org
Masayuki Saito and Yuko Okamatsu-Ogura: Thermogenic Brown Fat in Humans

al [84] reported acute mirabegron treatment increases higher BAT activities, BAT being an independent
FDG uptake into BAT, in parallel with an increase in determinant of these parameters (Fig. 5A). These re-
EE, heart rate, and blood pressure, at a high dose (200 sults seem compatible with retrospective analyses of
mg/kg), but only slightly at clinically approved doses (50 patient data showing blood glucose as a determinant
mg/kg). Following studies, however, demonstrated that of BAT prevalence [3,62-64]. Chondronikola et al [90]
chronic treatment with 50 mg/kg mirabegron induces reported that mild cold exposure increased whole-body
UCP1 in WAT and improved glucose homeostasis [85-87]. glucose disposal, plasma glucose oxidation, and insulin
It is to be noted in these studies that chronic mirabe- sensitivity in men with significant amounts of BAT,
gron treatment resulted in undetectable change in body but not in those without detectable BAT. Improved
fatness, but improved glucose tolerance and insulin sen- insulin sensitivity in parallel with BAT recruitment
sitivity, suggesting beneficial effects of beige adipocytes after cold acclimation was also shown in healthy men
on systemic metabolism as discussed in the next section. [91,92] and in patients with type 2 diabetes mellitus
[93]. In connection with the beneficial effects of BAT
BROWN FAT REGULATES on insulin sensitivity, interesting is that BAT actively
SYSTEMIC METABOLISM BEYOND metabolizes branched chain amino acids (BCAA) to
THERMOGENESIS lower their plasma levels in humans [94]. Since the ac-
cumulation of intracellular BCAA is known to inhibit
Increasing evidence has suggest that BAT has an insulin signaling through mTOR activation, impaired
impact on systemic metabolism, independent of that insulin sensitivity often seen in obesity and aging may
on the regulation of EE and body fatness. For instance, be attributable to increased circulating BCAA result-
mouse studies have shown that transplantation of ing from reduced BAT activity [95].
BAT or brown adipocytes results in improved glucose Very recently, Seki et al [96] reported that the BAT-
tolerance and increased insulin sensitivity [88,89]. In associated changes in systemic glucose metabolism re-
humans, we [66] found in healthy adults that blood sult in a marked suppression of tumor. Exposure of tu-
glucose and HbA1c levels are lower in individuals with mor-bearing mice to cold conditions inhibits the growth

A B

Glucose T2DM

HbA1c Dyslipidemia

Insulin AF

HOMA-R CAD

HDL-Cho CVD

LDL-Cho CHF

TG Hypertension

0.5 1.0 1.5 2.0 0.5 1.0 1.5


Odds ratio Odds ratio
Fig. 5. Impact of brown fat on blood parameter and cardiometabolic diseases. (A) Blood parameters in healthy subjects. HDL-Cho: high-density
lipoprotein cholesterol, LDL-Cho: low-density lipoprotein cholesterol, TG: triglyceride. Constructed from Matsushita et al (Int J Obes [Lond]
2014;38:812-7) [66]. (B) Cardiometabolic diseases. AF: atrial fibrillation/atrial flutter, CAD: coronary artery disease, CHF: congestive heart failure,
CVD: cerebrovascular disease, T2DM: type 2 diabetes. Closed circles show a significant association with BAT independent of body adiposity, age,
and sex. Adapted from Becher et al (Nat Med 2021;27:58-65) [64].

www.wjmh.org 7
https://doi.org/10.5534/wjmh.220224

of various types of solid tumor. The cold-induced tumor cardiovascular diseases [100]. Becher et al [64] catego-
suppression is ablated by surgical removal of BAT and rized 53,475 patients by presence or absence of BAT,
feeding on a high-glucose diet, and in UCP1-deficient and found improved profiles of blood glucose, TG, and
mice. Mechanistically, cold-induced BAT activation HDL-cholesterol in individuals with BAT, as already
results in increased systemic glucose metabolism and reported in healthy humans. Notably, BAT indepen-
decreased blood glucose, which impedes the glycolysis- dently correlated with lower rates of type 2 diabetes,
based metabolism in cancer cells. A pilot study in a dyslipidemia, hypertension, coronary artery disease,
patient with Hodgkin’s lymphoma showed a markedly and congestive heart failure (Fig. 5B). These results are
reduced FDG uptake into the tumor tissue after mild well consistent with previously reported association
cold exposure, but more human studies are needed to between BAT and diabetes [62] and also with a 5-year
strengthen its clinical relevance. follow-up study [101] showing that BAT activity cor-
BAT has also significant effects on systemic lipid related with lower carotid intima-media thickness and
metabolism. The main energy source of BAT thermo- higher carotid elasticity. An association of BAT with
genesis is fatty acids derived from intracellular TG hepatic steatosis was also reported in humans [102-104].
and incorporated from circulation. BAT activation by
cold exposure markedly accelerates clearance of plasma BROWN FAT-DERIVED ENDOCRINE
TG-rich lipoproteins due to increased uptake into BAT, FACTORS AFFECTING SYSTEMIC
and corrects hyperlipidemia [97]. Similar effects were METABOLISM
also found by pharmacologic activation of BAT with a
ß3AR agonist, which improves dyslipidemia, and more As mentioned above, even small amounts of BAT
importantly, protects hyperlipidemic model mice from influence the broad range of systemic metabolisms.
atherosclerosis [98]. Activation of BAT thermogenesis Mechanistically, some of the effects may be attrib-
increases high-density lipoprotein (HDL) levels, pro- uted to the relatively high metabolic activity of BAT
moting HDL cholesterol turnover [86,99]. All these facts itself, as in the case of the lowering effect on TG-rich
suggest a direct contribution of BAT to the regulation lipoprotein and BCAA; however, other effects seem dif-
of blood lipoprotein metabolism. ficult to be explained solely by its own activity. In this
The effects of BAT on systemic metabolism would be connection, it is interesting that BAT secretes various
closely related to the etiology of various metabolic and molecules into extracellular fluid, collectively called as

Fatty acid uptake Thermogenesis

Exercise capacity Sympathetic


nerve growth
12,13-diHOME

Muscle Myostatin NGF Brown fat

VEGF Angiogenesis
Lipogenesis NRG4

Fgf21 Exosomal Beige adipocyte


expression miR-99b FGF21 induction
Fig. 6. Endocrine actions of brown
IL-6 fat-derived factors, BATkines: 12,13-
Inflammation Maresin 2 diHOME,12,13-dihydroxyoctadecaenoic
Energy expenditure
acid, FA: fatty acid, FGF21: fibroblast
growth factor 21, IL-6: interleukin-6, miR:
Stress-induced Insulin sensitivity
gluconeogenesis Whole body microRNA, NGF: nerve growth factor,
Liver
NRG: neuregulin 4, VEGF: vascular endo-
thelial growth factor.

8 www.wjmh.org
Masayuki Saito and Yuko Okamatsu-Ogura: Thermogenic Brown Fat in Humans

batokine (BATkine), which may mediate the effects of reported that mice with adipose-tissue-specific deletion
BAT on other tissues [8,105] (Fig. 6). of the miRNA-processing enzyme exhibit a substantial
Activated BAT secretes some growth factors, such as decrease in the levels of circulating exosomal miRNAs,
vascular endothelial growth factor and nerve growth including miRNA-99b, and that the transplantation of
factors, which promote angiogenesis and SN growth BAT restores the circulating miRNA level, improves
associated with tissue hyperplasia [106-108]. In addition glucose tolerance, and suppresses hepatic FGF21 ex-
to these paracrine factors, BAT secretes a wide variety pression. These results suggest that exosomal miRNA-
of polypeptides into circulating blood. Among them, 99b is secreted from BAT into blood circulation, acts
interleukin-6 (IL-6) may be a likely BATkine to regu- on the liver, and suppresses hepatic FGF21 expression.
late systemic glucose metabolism. In the study of BAT It was also reported that circulating levels of miR-92a-
transplantation, Stanford et al [109] found that the im- 3p and miRNA-122 are inversely correlated with BAT
proved metabolic profile was lost when the BAT used activity in humans [121,122].
for transplantation was obtained from IL-6-deficient Some lipid molecules are also suggested as active
mice. Qing et al [110] reported that acute stress induces BATkines. Oxylipins, molecules derived from the oxi-
IL-6 secretion from BAT, which is the required instruc- dation of polyunsaturated fatty acids, are attracted in-
tive signal for mediating hyperglycemia through he- terest because of their action as intercellular signaling
patic gluconeogenesis. molecules and involvement in the regulation of many
Another candidate of BATkines is fibroblast growth cell and tissue responses. Particularly interesting is a
factor 21 (FGF21). FGF21 is released abundantly from linoleic acid derivative 12,13-dihydroxyoctadecaenoic
the liver, but it is also released from activated BAT acid (12,13-diHOME) secreted from BAT upon cold ex-
and contributes to increased circulating levels [111-113]. posure. 12,13-diHOME activates BAT in autocrine man-
FGF21 is known as an important regulator of systemic ner to enhance thermogenesis, resulting in decreased
metabolism and whole-body energy balance; it increas- levels of serum TGs [123]. Moreover, 12,13-diHOME
es energy expenditure by inducing BAT thermogenesis secretion from BAT is enhanced by exercise and in-
and WAT browning, while it suppresses sugar intake creases fatty acid uptake in muscle [124]. Very recently,
by actin on the ventromedial hypothalamus. FGF21 Sugimoto et al [125] reported that cold- and β3AR ag-
stimulates glucose utilization in WAT, hepatic fatty onist-activated BAT produces maresin 2, a member of
acid oxidation, and peripheral lipoprotein catabolism the anti-inflammatory pro-resolving mediators synthe-
[114,115]. Ruan et al also [116] reported that brown sized from docosahexaenoic acid, and reduces inflam-
adipocyte-specific FGF21 knockout impaired the effects mation in obesity in part by targeting macrophages in
of adenosine A2A receptor agonism in attenuating the liver. A role of BAT metabolism of succinate, an
hypertensive cardiac remodeling, suggesting an endo- intermediate of the tricarboxylic cycle, has also been
crine role of BAT in controlling hypertensive cardiac suggested in liver inflammation. Cold exposure produc-
remodeling though the release of FGF21. Despite many es substantial and selective accumulation of succinate
studies in mice, however, it remains to be investigated in BAT, which is sufficient to elevate UCP1-dependent
whether FGF21 secreted from BAT, compared with thermogenesis and sequester elevated circulating succi-
that from the liver, plays key physiological roles in hu- nate [126,127]. In contrast, without UCP1, BAT exhibits
mans. a diminished capacity to clear succinate from the cir-
In addition, many polypeptides such as neureglin4 culation, and elevated extracellular succinate in liver
(NRG4) and myostatin were identified as BATkines tissue that drives inflammation through the action on
[117,118], but a comprehensive overview is beyond the liver resident stellate cells and macrophages [128]. As
scope of this article and found in other literatures such, BAT may regulate liver immune cell infiltration
[8,105]. and pathology through succinate metabolism.
MicroRNAs (miRNAs) may serve as signals between In addition, several small molecules were identified
BAT and other tissues. miRNAs produced in adipo- as BATkine candidates: for example, 3-methyl-2-oxova-
cytes have a role in the differentiation and function of leric acid, 5-oxoproline and β-hydroxyisobutyric acid
adipocyte itself and are also secreted through exosomes synthesized in and released from brown adipocytes
and taken-up into other cells [119]. Thomou et al [120] induce thermogenic gene expression in white adipo-

www.wjmh.org 9
https://doi.org/10.5534/wjmh.220224

cytes and energy metabolism in skeletal myocytes [129]. BAT to sympathetic stimulation. Consistent with this
Thus, BATkines are likely to mediate the beneficial idea, there is a report demonstrating decreased β3AR
effects of activated BAT on other tissues/organs, but mRNA expression and impaired cellular responses to
their pathophysiological roles in humans are largely adrenergic stimulation in BAT of aged mice [145]. This
elusive to date. seems well compatible with the observations in hu-
mans that SN activity increases with age, while ther-
AGE-RELATED CHANGES IN BROWN mogenic response to cold decreases [146]. We demon-
FAT FUNCTIONS strated a significant impact of SNP in the β3AR gene
on the age-related decline in human BAT [60]. Bahler
As mentioned above, BAT is a promising tissue et al [147] reported, however, using imaging technics
combating obesity and cardiometabolic diseases. It is with a 123I-labelled NA analogue and 18F-FDG, that both
also true, however, that the amount and activity of sympathetic drive and BAT activity are lower in older
human BAT decline remarkably with age, while the men than in younger men. Thus, further studies are
prevalence of these diseases increases. Accordingly, it needed to draw out the precise mechanism underlying
is important to understand the mechanisms/factors of the age-related decline in BAT in terms of SN activity.
age-related decline of BAT and how to re-activate and Another likely mechanism is related to the produc-
recruit BAT in elderly [130]. tion of pro-inflammatory mediators by macrophages
Ageing is associated with a decline of both classical infiltrating in adipose tissue [148]. It is known that the
and beige adipocytes, reflected by increased intracel- number of M1, but not M2 macrophage, in adipose tis-
lular lipid accumulation, reduced mitochondrial and sue increases in obesity. While anti-inflammatory resi-
UCP1 content, and reduced ability of beige adipocyte dent M2 macrophages are stimulatory to beige adipo-
induction [131,132]. There is a heterogenous population cyte induction [149], pro-inflammatory M1 macrophages
of beige adipocyte progenitor cell, which is maintained impair brown adipocyte activity and beige adipogenesis
during postnatal life by its ability to proliferate and [150-152]. Pro-inflammatory cytokines such as tumor
differentiate in response to beiging signals [133]. Aging necrosis factor a and interleukin-1β induce a decreased
is associated with not only an impaired ability of beige viability of brown adipocytes accompanied by a mas-
adipocyte progenitor cell to proliferate, which leads to sive reactive oxygen species production and down-
a marked decrease in the progenitor cell number, but regulation of thermogenic gene expression [153], and
also a decreased differentiating ability of the progeni- induce apoptosis of brown adipocytes [154]. It is thus
tor cell [134-136]. likely that increased infiltration of M1 macrophage
The age-related change of BAT is regulated by mul- in WAT facilitates the attenuated browning of WAT,
tiple intrinsic factors, including a longevity gene Sir- suggesting the suppression of M1 macrophage activity
tuin 1 [137], CD81 [138], and the FSTL1 gene encoding in WAT would be effective to induce beige adipocytes,
the follistatin-like one glycoprotein [139]. Mitochondrial thereby preventing against obesity and related meta-
dysfunctions are also likely to link intrinsically to age- bolic disorders.
related decline of BAT thermogenic function [140]. Ta-
jima et al [141] reported that mitochondrial lipoylation SEX DIFFERENCES IN BROWN FAT
is reduced in aged BAT, but its enhancement by a-
lipoic acid supplementation restores BAT function, Ageing is associated with several endocrine changes,
thereby preventing age-associated obesity and glucose including diminished gonadal function. Animal stud-
intolerance. ies have demonstrated sex differences in BAT: for ex-
Among various neuro-endocrine mechanisms/factors, ample, compared to BAT of male rats, that of females
the SN- bAR system may be critical. In aged animals, shows higher mitochondrial density and cristae height,
sympathetic signaling to BAT is greater both at ther- higher UCP1 levels, and higher sensitivity to adrener-
moneutrality and during cold exposure [142,143], where- gic stimulation [155]. Higher ability of WAT browning
as BAT cell proliferation induced by cold exposure is was also reported in female mice [156]. A comprehen-
substantially attenuated [144]. These facts imply that sive gene expression analysis revealed higher UCP1
ageing is associated with the decreased sensitivity of expression in WAT of female mice [157]. Consistently,

10 www.wjmh.org
Masayuki Saito and Yuko Okamatsu-Ogura: Thermogenic Brown Fat in Humans

human studies have demonstrated sex difference in receptor were shown to display a reduced expression of
CIT and BAT throughout the life span [158,159]. In thermogenic genes in BAT and get obese [184]. Thus,
children, the activity and mass of BAT change slightly the in vivo effects of androgens appear rather compli-
with age along with sexual maturation, being higher in cated, but a part of them may be explained by locally
prepubertal girls than in boys [160]. In adults, although increased estrogen level due to intratissue aromatizai-
the prevalence of BAT detected by FDG-PET scans tion of androgens in WAT and the brain [185]. In fact,
markedly declines with age, its sex difference persists, the testosterone-induced reduction of WAT mass in
being higher in women than in men [3,61-63,161,162]. hypogonadal male mice requires ER in the brain [186].
Moreover, higher UCP1 expression and browning po- Further studies are needed to uncover the mechanisms
tential of WAT in women were reported [163,164]. It is and factors responsible to the sex differences in BAT.
to be careful, however, that the sex difference in BAT
detected by FDG-PET may not always reflect the dif- CONCLUSION AND FUTURE
ference in the amount and/or intrinsic activity of BAT. PERSPECTIVES
In almost all clinical studies in patients so far reported,
FDG-PET was carried out at room temperatures with- As discussed throughout the previous sections, BAT
out maximizing BAT activity by cold exposure. Con- participates in the regulation of whole-body EE, sys-
sidering sexual dimorphism of thermic, metabolic and temic metabolism, and cardiovascular functions in
cardiovascular responses to cold exposure [165], as well humans. Accordingly, this specific tissue is now rec-
as cold sensation [166], it is possible that the above- ognized as an intriguing therapeutic target of obesity
mentioned sex difference in human BAT may be due and metabolic disorders such as diabetes mellitus,
to the difference in the sensitivity to cold stimulus. In dyslipidemia, and related cardiovascular diseases. In
fact, several prospective studies in healthy participants fact, several drugs and food ingredients targeting BAT
revealed no significant sex difference in the prevalence have been tested for pharmacological and nutritional
and activity of BAT estimated under cold conditions therapy of obesity and metabolic syndrome [79,81,187].
[15,167,168], despite of higher CIT in women. Although not discussed in this article, possible contri-
The sex dimorphism suggests the direct and indirect bution of BAT to some beneficial effects of exercise is
effects of gonadal hormones on BAT [169]. Estrogens also suggested, particularly in context with the inter-
are the most likely hormone to enhance the activity action between skeletal muscle and beige adipocytes
and differentiation of BAT [170]. The stimulatory ef- [188,189].
fects of estrogens are mediated both directly through Despite these advances in BAT researches, there are
the action on nuclear receptors (ER) expressed in some unsolved but critical problems in humans, includ-
brown adipocytes [171] and indirectly through the ing the genetic and environmental factors responsible
brain-SN system [172]. In contrast to estrogens, andro- for a remarkable individual difference in the amount
gens were reported to inhibit differentiation and UCP1 and activity of BAT, and the patho-physiological rel-
expression in brown adipocytes in vitro [173,174]. In evance of UCP1-independent thermogenic mechanisms
vivo studies reported that surgical removal of testis re- of beige adipocytes [190]. Particularly important is the
sults in increased UCP1 expression in BAT and WAT method used in assessing human BAT. To date, FDG-
[175,176]. These results seem consistent with those in PET is a standard tool [191]; however, this method has
human studies that hyperandrogenism in women as serious limitations, including the enormous cost of
in polycystic ovary syndrome (PCOS) is associated devices, radiation exposure, and acute cold exposure,
with lower BAT activity and obesity [177,178]. Reduced which make repeated measurements difficult and an
UCP1 expression was also found in androgen-induced impediment in basic and clinical studies. Moreover, the
PCOS model animals [179,180]. However, in vivo ef- uptake of FDG into BAT is not always associated with
fects of androgens so far reported are controversial. its thermogenic activity. There is therefore an urgent
For example, several studies have shown no effects of need to establish less invasive and simpler methods for
dihydroxytestosterone treatment on UCP1 expression quantitative assessment of human BAT [192-195]. This
in BAT of orchiectomized mice or female mice [181- would promote prospective human studies, including
183]. Moreover, mice deficient of the nuclear androgen longitudinal observations and the development of prac-

www.wjmh.org 11
https://doi.org/10.5534/wjmh.220224

tical, easy, and effective regimens that can activate H, Harper ME, et al. Mice lacking mitochondrial uncoupling
and recruit BAT. protein are cold-sensitive but not obese. Nature 1997;387:90-
4.
Conflict of Interest 11. Rowland LA, Bal NC, Kozak LP, Periasamy M. Uncoupling
protein 1 and sarcolipin are required to maintain optimal
The authors have nothing to disclose. thermogenesis, and loss of both systems compromises sur-
vival of mice under cold stress. J Biol Chem 2015;290:12282-
Funding 9.
12. Yoneshiro T, Aita S, Matsushita M, Kameya T, Nakada K,
None. Kawai Y, et al. Brown adipose tissue, whole-body energy ex-
penditure, and thermogenesis in healthy adult men. Obesity
Author contribution (Silver Spring) 2011;19:13-6.
13. Orava J, Nuutila P, Lidell ME, Oikonen V, Noponen T, Vil-
Writing-original draft: MS, Writing-review & editing: YOO. janen T, et al. Different metabolic responses of human brown
adipose tissue to activation by cold and insulin. Cell Metab

REFERENCES 2011;14:272-9.
14. Ouellet V, Labbé SM, Blondin DP, Phoenix S, Guérin B, Ha-
1. Saito M, Okamatsu-Ogura Y, Matsushita M, Watanabe K, man F, et al. Brown adipose tissue oxidative metabolism con-
Yoneshiro T, Nio-Kobayashi J, et al. High incidence of meta- tributes to energy expenditure during acute cold exposure in
bolically active brown adipose tissue in healthy adult humans: humans. J Clin Invest 2012;122:545-52.
effects of cold exposure and adiposity. Diabetes 2009;58:1526- 15. Chen KY, Brychta RJ, Linderman JD, Smith S, Courville A,
31. Dieckmann W, et al. Brown fat activation mediates cold-
2. van Marken Lichtenbelt WD, Vanhommerig JW, Smulders induced thermogenesis in adult humans in response to a mild
NM, Drossaerts JM, Kemerink GJ, Bouvy ND, et al. Cold- decrease in ambient temperature. J Clin Endocrinol Metab
activated brown adipose tissue in healthy men. N Engl J Med 2013;98:E1218-23.
2009;360:1500-8. 16. Blondin DP, Labbé SM, Phoenix S, Guérin B, Turcotte ÉE,
3. Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Gold- Richard D, et al. Contributions of white and brown adipose
fine AB, et al. Identification and importance of brown adipose tissues and skeletal muscles to acute cold-induced metabolic
tissue in adult humans. N Engl J Med 2009;360:1509-17. responses in healthy men. J Physiol 2015;593:701-14.
4. Virtanen KA, Lidell ME, Orava J, Heglind M, Westergren R, 17. Huo C, Song Z, Yin J, Zhu Y, Miao X, Qian H, et al. Effect
Niemi T, et al. Functional brown adipose tissue in healthy of acute cold exposure on energy metabolism and activity
adults. N Engl J Med 2009;360:1518-25. Erratum in: N Engl J of brown adipose tissue in humans: a systematic review and
Med 2009;361:1123. meta-analysis. Front Physiol 2022;13:917084.
5. Saito M. Brown adipose tissue as a regulator of energy expen- 18. Saito M, Yoneshiro T, Matsushita M. Activation and recruit-
diture and body fat in humans. Diabetes Metab J 2013;37:22- ment of brown adipose tissue by cold exposure and food
9. ingredients in humans. Best Pract Res Clin Endocrinol Metab
6. Carpentier AC, Blondin DP, Virtanen KA, Richard D, Haman 2016;30:537-47.
F, Turcotte ÉE. Brown adipose tissue energy metabolism in 19. Westerterp KR. Diet induced thermogenesis. Nutr Metab
humans. Front Endocrinol (Lausanne) 2018;9:447. (Lond) 2004;1:5.
7. Kajimura S, Spiegelman BM, Seale P. Brown and beige fat: 20. Quatela A, Callister R, Patterson A, MacDonald-Wicks L. The
physiological roles beyond heat generation. Cell Metab energy content and composition of meals consumed after an
2015;22:546-59. overnight fast and their effects on diet induced thermogen-
8. Scheele C, Wolfrum C. Brown adipose crosstalk in tissue esis: a systematic review, meta-analyses and meta-regressions.
plasticity and human metabolism. Endocr Rev 2020;41:53-65. Nutrients 2016;8:670.
9. Yuko OO, Saito M. Brown fat as a regulator of systemic 21. Glick Z, Teague RJ, Bray GA. Brown adipose tissue: thermic
metabolism beyond thermogenesis. Diabetes Metab J response increased by a single low protein, high carbohydrate
2021;45:840-52. meal. Science 1981;213:1125-7.
10. Enerbäck S, Jacobsson A, Simpson EM, Guerra C, Yamashita 22. Saito M, Minokoshi Y, Shimazu T. Metabolic and sympathetic

12 www.wjmh.org
Masayuki Saito and Yuko Okamatsu-Ogura: Thermogenic Brown Fat in Humans

nerve activities of brown adipose tissue in tube-fed rats. Am J 36. Shabalina IG, Petrovic N, de Jong JM, Kalinovich AV, Cannon
Physiol 1989;257(3 Pt 1):E374-8. B, Nedergaard J. UCP1 in brite/beige adipose tissue mito-
23. von Essen G, Lindsund E, Cannon B, Nedergaard J. Adap- chondria is functionally thermogenic. Cell Rep 2013;5:1196-
tive facultative diet-induced thermogenesis in wild-type but 203.
not in UCP1-ablated mice. Am J Physiol Endocrinol Metab 37. Sharp LZ, Shinoda K, Ohno H, Scheel DW, Tomoda E, Ruiz
2017;313:E515-27. L, et al. Human BAT possesses molecular signatures that re-
24. Nagai N, Sakane N, Ueno LM, Hamada T, Moritani T. The semble beige/brite cells. PLoS One 2012;7:e49452.
-3826 A-->G variant of the uncoupling protein-1 gene di- 38. Lidell ME, Betz MJ, Dahlqvist Leinhard O, Heglind M, Eland-
minishes postprandial thermogenesis after a high fat meal in er L, Slawik M, et al. Evidence for two types of brown adipose
healthy boys. J Clin Endocrinol Metab 2003;88:5661-7. tissue in humans. Nat Med 2013;19:631-4.
25. Vrieze A, Schopman JE, Admiraal WM, Soeters MR, Nieu- 39. Cypess AM, White AP, Vernochet C, Schulz TJ, Xue R, Sass
wdorp M, Verberne HJ, et al. Fasting and postprandial ac- CA, et al. Anatomical localization, gene expression profiling
tivity of brown adipose tissue in healthy men. J Nucl Med and functional characterization of adult human neck brown
2012;53:1407-10. fat. Nat Med 2013;19:635-9.
26. Vosselman MJ, Brans B, van der Lans AA, Wierts R, van Baak 40. LeBlanc J, Cabanac M, Samson P. Reduced postprandial heat
MA, Mottaghy FM, et al. Brown adipose tissue activity after a production with gavage as compared with meal feeding in
high-calorie meal in humans. Am J Clin Nutr 2013;98:57-64. human subjects. Am J Physiol 1984;246(1 Pt 1):E95-101.
27. Muzik O, Mangner TJ, Leonard WR, Kumar A, Janisse J, 41. Glick Z, Raum WJ. Norepinephrine turnover in brown
Granneman JG. 15O PET measurement of blood flow and adipose tissue is stimulated by a single meal. Am J Physiol
oxygen consumption in cold-activated human brown fat. J 1986;251(1 Pt 2):R13-7.
Nucl Med 2013;54:523-31. 42. Schwartz RS, Jaeger LF, Silberstein S, Veith RC. Sympathetic
28. Din MU, Saari T, Raiko J, Kudomi N, Maurer SF, Lahesmaa M, nervous system activity and the thermic effect of feeding in
et al. Postprandial oxidative metabolism of human brown fat man. Int J Obes 1987;11:141-9.
indicates thermogenesis. Cell Metab 2018;28:207-216.e3. 43. van Baak MA. Meal-induced activation of the sympathetic
29. Hibi M, Oishi S, Matsushita M, Yoneshiro T, Yamaguchi T, nervous system and its cardiovascular and thermogenic ef-
Usui C, et al. Brown adipose tissue is involved in diet-induced fects in man. Physiol Behav 2008;94:178-86.
thermogenesis and whole-body fat utilization in healthy hu- 44. Diamond P, Brondel L, LeBlanc J. Palatability and postprandi-
mans. Int J Obes (Lond) 2016;40:1655-61. al thermogenesis in dogs. Am J Physiol 1985;248(1 Pt 1):E75-
30. Matsushita M, Nirengi S, Hibi M, Wakabayashi H, Lee SI, Do- 9.
michi M, et al. Diurnal variations of brown fat thermogenesis 45. Li Y, Schnabl K, Gabler SM, Willershäuser M, Reber J, Karlas
and fat oxidation in humans. Int J Obes (Lond) 2021;45:2499- A, et al. Secretin-activated brown fat mediates prandial ther-
505. mogenesis to induce satiation. Cell 2018;175:1561-74.e12.
31. Bukowiecki LJ, Géloën A, Collet AJ. Proliferation and dif- 46. Yamazaki T, Morimoto-Kobayashi Y, Koizumi K, Takahashi
ferentiation of brown adipocytes from interstitial cells during C, Nakajima S, Kitao S, et al. Secretion of a gastrointestinal
cold acclimation. Am J Physiol 1986;250(6 Pt 1):C880-7. hormone, cholecystokinin, by hop-derived bitter components
32. Okamatsu-Ogura Y, Fukano K, Tsubota A, Nio-Kobayashi J, activates sympathetic nerves in brown adipose tissue. J Nutr
Nakamura K, Morimatsu M, et al. Cell-cycle arrest in mature Biochem 2019;64:80-7.
adipocytes impairs BAT development but not WAT brown- 47. Beiroa D, Imbernon M, Gallego R, Senra A, Herranz D, Vil-
ing, and reduces adaptive thermogenesis in mice. Sci Rep larroya F, et al. GLP-1 agonism stimulates brown adipose
2017;7:6648. tissue thermogenesis and browning through hypothalamic
33. Harms M, Seale P. Brown and beige fat: development, func- AMPK. Diabetes 2014;63:3346-58.
tion and therapeutic potential. Nat Med 2013;19:1252-63. 48. Vijgen GH, Bouvy ND, Leenen L, Rijkers K, Cornips E, Ma-
34. Kajimura S, Saito M. A new era in brown adipose tissue biol- joie M, et al. Vagus nerve stimulation increases energy expen-
ogy: molecular control of brown fat development and energy diture: relation to brown adipose tissue activity. PLoS One
homeostasis. Annu Rev Physiol 2014;76:225-49. 2013;8:e77221.
35. Okamatsu-Ogura Y, Fukano K, Tsubota A, Uozumi A, Terao 49. Vítek L, Haluzík M. The role of bile acids in metabolic regula-
A, Kimura K, et al. Thermogenic ability of uncoupling protein tion. J Endocrinol 2016;228:R85-96.
1 in beige adipocytes in mice. PLoS One 2013;8:e84229. 50. Watanabe M, Houten SM, Mataki C, Christoffolete MA, Kim

www.wjmh.org 13
https://doi.org/10.5534/wjmh.220224

BW, Sato H, et al. Bile acids induce energy expenditure by S, et al. Prevalence, mass, and glucose-uptake activity of 18F-
promoting intracellular thyroid hormone activation. Nature FDG-detected brown adipose tissue in humans living in a
2006;439:484-9. temperate zone of Italy. PLoS One 2013;8:e63391.
51. Broeders EP, Nascimento EB, Havekes B, Brans B, Roumans 64. Becher T, Palanisamy S, Kramer DJ, Eljalby M, Marx SJ, Wib-
KH, Tailleux A, et al. The bile acid chenodeoxycholic acid mer AG, et al. Brown adipose tissue is associated with cardio-
increases human brown adipose tissue activity. Cell Metab metabolic health. Nat Med 2021;27:58-65.
2015;22:418-26. 65. Zhang Q, Ye H, Miao Q, Zhang Z, Wang Y, Zhu X, et al. Dif-
52. Velazquez-Villegas LA, Perino A, Lemos V, Zietak M, Nomu- ferences in the metabolic status of healthy adults with and
ra M, Pols TWH, et al. TGR5 signalling promotes mitochon- without active brown adipose tissue. Wien Klin Wochenschr
drial fission and beige remodelling of white adipose tissue. 2013;125:687-95.
Nat Commun 2018;9:245. 66. Matsushita M, Yoneshiro T, Aita S, Kameya T, Sugie H, Saito
53. Cannon B, Nedergaard J. Brown adipose tissue: function and M. Impact of brown adipose tissue on body fatness and
physiological significance. Physiol Rev 2004;84:277-359. glucose metabolism in healthy humans. Int J Obes (Lond)
54. Lowell BB, Spiegelman BM. Towards a molecular understand- 2014;38:812-7.
ing of adaptive thermogenesis. Nature 2000;404:652-60. 67. Yoneshiro T, Aita S, Matsushita M, Okamatsu-Ogura Y, Ka-
55. Feldmann HM, Golozoubova V, Cannon B, Nedergaard J. meya T, Kawai Y, et al. Age-related decrease in cold-activated
UCP1 ablation induces obesity and abolishes diet-induced brown adipose tissue and accumulation of body fat in healthy
thermogenesis in mice exempt from thermal stress by living humans. Obesity (Silver Spring) 2011;19:1755-60.
at thermoneutrality. Cell Metab 2009;9:203-9. 68. Wu J, Boström P, Sparks LM, Ye L, Choi JH, Giang AH, et al.
56. Kontani Y, Wang Y, Kimura K, Inokuma KI, Saito M, Suzuki- Beige adipocytes are a distinct type of thermogenic fat cell in
Miura T, et al. UCP1 deficiency increases susceptibility to mouse and human. Cell 2012;150:366-76.
diet-induced obesity with age. Aging Cell 2005;4:147-55. 69. Shinoda K, Luijten IH, Hasegawa Y, Hong H, Sonne SB,
57. Oppert JM, Vohl MC, Chagnon M, Dionne FT, Cassard- Kim M, et al. Genetic and functional characterization of
Doulcier AM, Ricquier D, et al. DNA polymorphism in the clonally derived adult human brown adipocytes. Nat Med
uncoupling protein (UCP) gene and human body fat. Int J 2015;21:389-94.
Obes Relat Metab Disord 1994;18:526-31. 70. Lee P, Swarbrick MM, Zhao JT, Ho KK. Inducible brown adi-
58. Valve R, Heikkinen S, Rissanen A, Laakso M, Uusitupa M. pogenesis of supraclavicular fat in adult humans. Endocrinol-
Synergistic effect of polymorphisms in uncoupling protein 1 ogy 2011;152:3597-602.
and beta3-adrenergic receptor genes on basal metabolic rate 71. Yoneshiro T, Aita S, Matsushita M, Kayahara T, Kameya T,
in obese Finns. Diabetologia 1998;41:357-61. Kawai Y, et al. Recruited brown adipose tissue as an antiobe-
59. Kogure A, Yoshida T, Sakane N, Umekawa T, Takakura Y, sity agent in humans. J Clin Invest 2013;123:3404-8.
Kondo M. Synergic effect of polymorphisms in uncoupling 72. van der Lans AA, Hoeks J, Brans B, Vijgen GH, Visser MG,
protein 1 and beta3-adrenergic receptor genes on weight loss Vosselman MJ, et al. Cold acclimation recruits human brown
in obese Japanese. Diabetologia 1998;41:1399. fat and increases nonshivering thermogenesis. J Clin Invest
60. Yoneshiro T, Ogawa T, Okamoto N, Matsushita M, Aita S, 2013;123:3395-403.
Kameya T, et al. Impact of UCP1 and β3AR gene polymor- 73. Blondin DP, Labbé SM, Tingelstad HC, Noll C, Kunach M,
phisms on age-related changes in brown adipose tissue and Phoenix S, et al. Increased brown adipose tissue oxidative
adiposity in humans. Int J Obes (Lond) 2013;37:993-8. capacity in cold-acclimated humans. J Clin Endocrinol Metab
61. Pfannenberg C, Werner MK, Ripkens S, Stef I, Deckert A, 2014;99:E438-46.
Schmadl M, et al. Impact of age on the relationships of brown 74. Caterina MJ. Transient receptor potential ion channels as
adipose tissue with sex and adiposity in humans. Diabetes participants in thermosensation and thermoregulation. Am J
2010;59:1789-93. Physiol Regul Integr Comp Physiol 2007;292:R64-76.
62. Ouellet V, Routhier-Labadie A, Bellemare W, Lakhal-Chaieb 75. Whiting S, Derbyshire E, Tiwari BK. Capsaicinoids and caps-
L, Turcotte E, Carpentier AC, et al. Outdoor temperature, age, inoids. A potential role for weight management? A systematic
sex, body mass index, and diabetic status determine the prev- review of the evidence. Appetite 2012;59:341-8.
alence, mass, and glucose-uptake activity of 18F-FDG-detect- 76. Yoneshiro T, Aita S, Kawai Y, Iwanaga T, Saito M. Nonpun-
ed BAT in humans. J Clin Endocrinol Metab 2011;96:192-9. gent capsaicin analogs (capsinoids) increase energy expen-
63. Persichetti A, Sciuto R, Rea S, Basciani S, Lubrano C, Mariani diture through the activation of brown adipose tissue in hu-

14 www.wjmh.org
Masayuki Saito and Yuko Okamatsu-Ogura: Thermogenic Brown Fat in Humans

mans. Am J Clin Nutr 2012;95:845-50. brown adipose tissue transplantation. Mol Aspects Med
77. Sun L, Camps SG, Goh HJ, Govindharajulu P, Schaeffer- 2019;68:74-81.
koetter JD, Townsend DW, et al. Capsinoids activate brown 89. Payab M, Abedi M, Foroughi Heravani N, Hadavandkhani
adipose tissue (BAT) with increased energy expenditure as- M, Arabi M, Tayanloo-Beik A, et al. Brown adipose tissue
sociated with subthreshold 18-fluorine fluorodeoxyglucose transplantation as a novel alternative to obesity treatment: a
uptake in BAT-positive humans confirmed by positron emis- systematic review. Int J Obes (Lond) 2021;45:109-21.
sion tomography scan. Am J Clin Nutr 2018;107:62-70. 90. Chondronikola M, Volpi E, Børsheim E, Porter C, Annamalai
78. Nirengi S, Homma T, Inoue N, Sato H, Yoneshiro T, Matsu- P, Enerbäck S, et al. Brown adipose tissue improves whole-
shita M, et al. Assessment of human brown adipose tissue body glucose homeostasis and insulin sensitivity in humans.
density during daily ingestion of thermogenic capsinoids us- Diabetes 2014;63:4089-99.
ing near-infrared time-resolved spectroscopy. J Biomed Opt 91. Lee P, Smith S, Linderman J, Courville AB, Brychta RJ, Di-
2016;21:091305. eckmann W, et al. Temperature-acclimated brown adipose
79. Yoneshiro T, Matsushita M, Saito M. Translational aspects of tissue modulates insulin sensitivity in humans. Diabetes
brown fat activation by food-derived stimulants. Handb Exp 2014;63:3686-98.
Pharmacol 2019;251:359-79. 92. Iwen KA, Backhaus J, Cassens M, Waltl M, Hedesan OC,
80. Saito M, Matsushita M, Yoneshiro T, Okamatsu-Ogura Y. Merkel M, et al. Cold-induced brown adipose tissue activity
Brown adipose tissue, diet-induced thermogenesis, and ther- alters plasma fatty acids and improves glucose metabolism in
mogenic food ingredients: from mice to men. Front Endocri- men. J Clin Endocrinol Metab 2017;102:4226-34.
nol (Lausanne) 2020;11:222.   93. Hanssen MJ, Hoeks J, Brans B, van der Lans AA, Schaart G,
81. Arch JR. The discovery of drugs for obesity, the metabolic van den Driessche JJ, et al. Short-term cold acclimation im-
effects of leptin and variable receptor pharmacology: perspec- proves insulin sensitivity in patients with type 2 diabetes mel-
tives from beta3-adrenoceptor agonists. Naunyn Schmiede- litus. Nat Med 2015;21:863-5.
bergs Arch Pharmacol 2008;378:225-40. 94. Yoneshiro T, Wang Q, Tajima K, Matsushita M, Maki H, Iga-
82. Collins S. β-Adrenergic receptors and adipose tissue metabo- rashi K, et al. BCAA catabolism in brown fat controls energy
lism: evolution of an old story. Annu Rev Physiol 2022;84:1- homeostasis through SLC25A44. Nature 2019;572:614-9.
16. 95. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien
83. Nitti VW, Chapple CR, Walters C, Blauwet MB, Herschorn S, LF, et al. A branched-chain amino acid-related metabolic sig-
Milsom I, et al. Safety and tolerability of the β3 -adrenoceptor nature that differentiates obese and lean humans and contrib-
agonist mirabegron, for the treatment of overactive bladder: utes to insulin resistance. Cell Metab 2009;9:311-26. Erratum
results of a prospective pooled analysis of three 12-week ran- in: Cell Metab 2009;9:565-6.
domised phase III trials and of a 1-year randomised phase III 96. Seki T, Yang Y, Sun X, Lim S, Xie S, Guo Z, et al. Brown-fat-
trial. Int J Clin Pract 2014;68:972-85. mediated tumour suppression by cold-altered global metabo-
84. Cypess AM, Weiner LS, Roberts-Toler C, Franquet Elía E, lism. Nature 2022;608:421-8.
Kessler SH, Kahn PA, et al. Activation of human brown adi- 97. Bartelt A, Bruns OT, Reimer R, Hohenberg H, Ittrich H, Peld-
pose tissue by a β3-adrenergic receptor agonist. Cell Metab schus K, et al. Brown adipose tissue activity controls triglycer-
2015;21:33-8. ide clearance. Nat Med 2011;17:200-5.
85. Finlin BS, Memetimin H, Confides AL, Kasza I, Zhu B, Vek- 98. Berbée JF, Boon MR, Khedoe PP, Bartelt A, Schlein C, Worth-
aria HJ, et al. Human adipose beiging in response to cold and mann A, et al. Brown fat activation reduces hypercholester-
mirabegron. JCI Insight 2018;3:e121510. olaemia and protects from atherosclerosis development. Nat
86. O'Mara AE, Johnson JW, Linderman JD, Brychta RJ, Mc- Commun 2015;6:6356.
Gehee S, Fletcher LA, et al. Chronic mirabegron treatment 99. Bartelt A, John C, Schaltenberg N, Berbée JFP, Worthmann A,
increases human brown fat, HDL cholesterol, and insulin sen- Cherradi ML, et al. Thermogenic adipocytes promote HDL
sitivity. J Clin Invest 2020;130:2209-19. turnover and reverse cholesterol transport. Nat Commun
87. Finlin BS, Memetimin H, Zhu B, Confides AL, Vekaria HJ, El 2017;8:15010.
Khouli RH, et al. The β3-adrenergic receptor agonist mirabe- 100. Ying Z, Tramper N, Zhou E, Boon MR, Rensen PCN, Kooij-
gron improves glucose homeostasis in obese humans. J Clin man S. Role of thermogenic adipose tissue in lipid metabo-
Invest 2020;130:2319-31. lism and atherosclerotic cardiovascular disease: lessons
88. White JD, Dewal RS, Stanford KI. The beneficial effects of from studies in mice and humans. Cardiovasc Res 2022. doi:

www.wjmh.org 15
https://doi.org/10.5534/wjmh.220224

10.1093/cvr/cvac131 [Epub]. 113. Hanssen MJ, Broeders E, Samms RJ, Vosselman MJ, van der
101. Raiko J, Orava J, Savisto N, Virtanen KA. High brown fat Lans AA, Cheng CC, et al. Serum FGF21 levels are associ-
activity correlates with cardiovascular risk factor levels cross- ated with brown adipose tissue activity in humans. Sci Rep
sectionally and subclinical atherosclerosis at 5-year follow-up. 2015;5:10275.
Arterioscler Thromb Vasc Biol 2020;40:1289-95. 114. Fisher FM, Maratos-Flier E. Understanding the physiology of
102. Wibmer AG, Becher T, Eljalby M, Crane A, Andrieu PC, Jiang FGF21. Annu Rev Physiol 2016;78:223-41.
CS, et al. Brown adipose tissue is associated with healthier 115. Szczepańska E, Gietka-Czernel M. FGF21: a novel regulator
body fat distribution and metabolic benefits independent of of glucose and lipid metabolism and whole-body energy bal-
regional adiposity. Cell Rep Med 2021;2:100332. ance. Horm Metab Res 2022;54:203-11.
103. Ahmed BA, Ong FJ, Barra NG, Blondin DP, Gunn E, Ores- 116. Ruan CC, Kong LR, Chen XH, Ma Y, Pan XX, Zhang ZB, et
kovich SM, et al. Lower brown adipose tissue activity is as- al. A2A receptor activation attenuates hypertensive cardiac
sociated with non-alcoholic fatty liver disease but not changes remodeling via promoting brown adipose tissue-derived
in the gut microbiota. Cell Rep Med 2021;2:100397. FGF21. Cell Metab 2018;28:476-89.e5. Erratum in: Cell Metab
104. Acosta FM, Sanchez-Delgado G, Martinez-Tellez B, Osuna- 2020;32:689.
Prieto FJ, Mendez-Gutierrez A, Aguilera CM, et al. A larger 117. Wang GX, Zhao XY, Meng ZX, Kern M, Dietrich A, Chen Z,
brown fat volume and lower radiodensity are related to a et al. The brown fat-enriched secreted factor Nrg4 preserves
greater cardiometabolic risk, especially in young men. Eur J metabolic homeostasis through attenuation of hepatic lipo-
Endocrinol 2022;187:171-83. genesis. Nat Med 2014;20:1436-43.
105. Villarroya J, Cereijo R, Gavaldà-Navarro A, Peyrou M, Giralt 118. Kong X, Yao T, Zhou P, Kazak L, Tenen D, Lyubetskaya A, et
M, Villarroya F. New insights into the secretory functions of al. Brown adipose tissue controls skeletal muscle function via
brown adipose tissue. J Endocrinol 2019;243:R19-27. the secretion of myostatin. Cell Metab 2018;28:631-43.e3.
106. Asano A, Morimatsu M, Nikami H, Yoshida T, Saito M. 119. Lee MW, Lee M, Oh KJ. Adipose tissue-derived signatures for
Adrenergic activation of vascular endothelial growth factor obesity and type 2 diabetes: adipokines, batokines and mi-
mRNA expression in rat brown adipose tissue: implication in croRNAs. J Clin Med 2019;8:854.
cold-induced angiogenesis. Biochem J 1997;328(Pt 1):179-83. 120. Thomou T, Mori MA, Dreyfuss JM, Konishi M, Sakaguchi M,
107. Néchad M, Ruka E, Thibault J. Production of nerve growth Wolfrum C, et al. Adipose-derived circulating miRNAs regu-
factor by brown fat in culture: relation with the in vivo devel- late gene expression in other tissues. Nature 2017;542:450-5.
opmental stage of the tissue. Comp Biochem Physiol Comp Erratum in: Nature 2017;545:252.
Physiol 1994;107:381-8. 121. Chen Y, Buyel JJ, Hanssen MJ, Siegel F, Pan R, Naumann J, et
108. Rosell M, Kaforou M, Frontini A, Okolo A, Chan YW, Niko- al. Exosomal microRNA miR-92a concentration in serum re-
lopoulou E, et al. Brown and white adipose tissues: intrinsic flects human brown fat activity. Nat Commun 2016;7:11420.
differences in gene expression and response to cold exposure 122. Okamatsu-Ogura Y, Matsushita M, Bariuan JV, Nagaya K,
in mice. Am J Physiol Endocrinol Metab 2014;306:E945-64. Tsubota A, Saito M. Association of circulating exosomal
109. Stanford KI, Middelbeek RJ, Townsend KL, An D, Nygaard miR-122 levels with BAT activity in healthy humans. Sci Rep
EB, Hitchcox KM, et al. Brown adipose tissue regulates 2019;9:13243.
glucose homeostasis and insulin sensitivity. J Clin Invest 123. Lynes MD, Leiria LO, Lundh M, Bartelt A, Shamsi F, Huang
2013;123:215-23. TL, et al. The cold-induced lipokine 12,13-diHOME pro-
110. Qing H, Desrouleaux R, Israni-Winger K, Mineur YS, Fogel- motes fatty acid transport into brown adipose tissue. Nat Med
man N, Zhang C, et al. Origin and function of stress-induced 2017;23:631-7. Erratum in: Nat Med 2017;23:1384.
IL-6 in murine models. Cell 2020;182:1660. Erratum for: Cell 124. Stanford KI, Lynes MD, Takahashi H, Baer LA, Arts PJ,
2020;182:372-87.e14. May FJ, et al. 12,13-diHOME: an exercise-induced lipokine
111. Hondares E, Iglesias R, Giralt A, Gonzalez FJ, Giralt M, that increases skeletal muscle fatty acid uptake. Cell Metab
Mampel T, et al. Thermogenic activation induces FGF21 2018;27:1111-20.e3. Erratum in: Cell Metab 2018;27:1357.
expression and release in brown adipose tissue. J Biol Chem 125. Sugimoto S, Mena HA, Sansbury BE, Kobayashi S, Tsuji T,
2011;286:12983-90. Wang CH, et al. Brown adipose tissue-derived MaR2 contrib-
112. Lee P, Linderman JD, Smith S, Brychta RJ, Wang J, Idelson C, utes to cold-induced resolution of inflammation. Nat Metab
et al. Irisin and FGF21 are cold-induced endocrine activators 2022;4:775-90.
of brown fat function in humans. Cell Metab 2014;19:302-9. 126. Mills EL, Pierce KA, Jedrychowski MP, Garrity R, Winther S,

16 www.wjmh.org
Masayuki Saito and Yuko Okamatsu-Ogura: Thermogenic Brown Fat in Humans

Vidoni S, et al. Accumulation of succinate controls activation 141. Tajima K, Ikeda K, Chang HY, Chang CH, Yoneshiro T, Oguri
of adipose tissue thermogenesis. Nature 2018;560:102-6. Y, et al. Mitochondrial lipoylation integrates age-associated
127. Okamatsu-Ogura Y, Kuroda M, Tsutsumi R, Tsubota A, Saito decline in brown fat thermogenesis. Nat Metab 2019;1:886-
M, Kimura K, et al. UCP1-dependent and UCP1-independent 98.
metabolic changes induced by acute cold exposure in brown 142. Kawate R, Talan MI, Engel BT. Aged C57BL/6J mice respond
adipose tissue of mice. Metabolism 2020;113:154396. to cold with increased sympathetic nervous activity in inter-
128. Mills EL, Harmon C, Jedrychowski MP, Xiao H, Garrity R, scapular brown adipose tissue. J Gerontol 1993;48:B180-3.
Tran NV, et al. UCP1 governs liver extracellular succinate and 143. McDonald RB, Hamilton JS, Horwitz BA. Influence of age
inflammatory pathogenesis. Nat Metab 2021;3:604-17. and gender on brown adipose tissue norepinephrine turnover.
129. Whitehead A, Krause FN, Moran A, MacCannell ADV, Scragg Proc Soc Exp Biol Med 1993;204:117-21.
JL, McNally BD, et al. Brown and beige adipose tissue regulate 144. Florez-Duquet M, Horwitz BA, McDonald RB. Cellular pro-
systemic metabolism through a metabolite interorgan signal- liferation and UCP content in brown adipose tissue of cold-
ing axis. Nat Commun 2021;12:1905. exposed aging Fischer 344 rats. Am J Physiol 1998;274:R196-
130. Mattson MP. Perspective: does brown fat protect against dis- 203.
eases of aging? Ageing Res Rev 2010;9:69-76. 145. Scarpace PJ, Mooradian AD, Morley JE. Age-associated de-
131. Rogers NH, Landa A, Park S, Smith RG. Aging leads to a pro- crease in beta-adrenergic receptors and adenylate cyclase ac-
grammed loss of brown adipocytes in murine subcutaneous tivity in rat brown adipose tissue. J Gerontol 1988;43:B65-70.
white adipose tissue. Aging Cell 2012;11:1074-83. 146. Florez-Duquet M, McDonald RB. Cold-induced thermoregu-
132. Graja A, Schulz TJ. Mechanisms of aging-related impairment lation and biological aging. Physiol Rev 1998;78:339-58.
of brown adipocyte development and function. Gerontology 147. Bahler L, Verberne HJ, Admiraal WM, Stok WJ, Soeters MR,
2015;61:211-7. Hoekstra JB, et al. Differences in sympathetic nervous stimu-
133. Cohen P, Kajimura S. The cellular and functional complexity lation of brown adipose tissue between the young and old,
of thermogenic fat. Nat Rev Mol Cell Biol 2021;22:393-409. and the lean and obese. J Nucl Med 2016;57:372-7.
134. Zoico E, Rubele S, De Caro A, Nori N, Mazzali G, Fantin F, et 148. Kawai T, Autieri MV, Scalia R. Adipose tissue inflamma-
al. Brown and beige adipose tissue and aging. Front Endocri- tion and metabolic dysfunction in obesity. Am J Physiol Cell
nol (Lausanne) 2019;10:368. Physiol 2021;320:C375-91.
135. Silva GDN, Amato AA. Thermogenic adipose tissue ag- 149. Nguyen KD, Qiu Y, Cui X, Goh YP, Mwangi J, David T, et al.
ing: mechanisms and implications. Front Cell Dev Biol Alternatively activated macrophages produce catecholamines
2022;10:955612. to sustain adaptive thermogenesis. Nature 2011;480:104-8.
136. Shin W, Okamatsu-Ogura Y, Machida K, Tsubota A, Nio- 150. Sakamoto T, Takahashi N, Sawaragi Y, Naknukool S, Yu R,
Kobayashi J, Kimura K. Impaired adrenergic agonist-depen- Goto T, et al. Inflammation induced by RAW macrophages
dent beige adipocyte induction in aged mice. Obesity (Silver suppresses UCP1 mRNA induction via ERK activation in
Spring) 2017;25:417-23. 10T1/2 adipocytes. Am J Physiol Cell Physiol 2013;304:C729-
137. Khanh VC, Zulkifli AF, Tokunaga C, Yamashita T, Hiramatsu 38.
Y, Ohneda O. Aging impairs beige adipocyte differentiation of 151. Sakamoto T, Nitta T, Maruno K, Yeh YS, Kuwata H, Tomita
mesenchymal stem cells via the reduced expression of Sirtuin K, et al. Macrophage infiltration into obese adipose tissues
1. Biochem Biophys Res Commun 2018;500:682-90. suppresses the induction of UCP1 level in mice. Am J Physiol
138. Oguri Y, Shinoda K, Kim H, Alba DL, Bolus WR, Wang Q, et Endocrinol Metab 2016;310:E676-87.
al. CD81 controls beige fat progenitor cell growth and energy 152. Villarroya F, Cereijo R, Gavaldà-Navarro A, Villarroya J, Gi-
balance via FAK signaling. Cell 2020;182:563-77.e20. ralt M. Inflammation of brown/beige adipose tissues in obe-
139. Huang Z, Zhang Z, Moazzami Z, Heck R, Hu P, Nanda H, sity and metabolic disease. J Intern Med 2018;284:492-504.
et al. Brown adipose tissue involution associated with pro- 153. Rebiger L, Lenzen S, Mehmeti I. Susceptibility of brown adi-
gressive restriction in progenitor competence. Cell Rep pocytes to pro-inflammatory cytokine toxicity and reactive
2022;39:110575. oxygen species. Biosci Rep 2016;36:e00306.
140. Amorim JA, Coppotelli G, Rolo AP, Palmeira CM, Ross 154. Nisoli E, Briscini L, Giordano A, Tonello C, Wiesbrock SM,
JM, Sinclair DA. Mitochondrial and metabolic dysfunc- Uysal KT, et al. Tumor necrosis factor alpha mediates apopto-
tion in ageing and age-related diseases. Nat Rev Endocrinol sis of brown adipocytes and defective brown adipocyte func-
2022;18:243-58. tion in obesity. Proc Natl Acad Sci U S A 2000;97:8033-8.

www.wjmh.org 17
https://doi.org/10.5534/wjmh.220224

155. Rodriguez-Cuenca S, Pujol E, Justo R, Frontera M, Oliver 2021;534:111365.


J, Gianotti M, et al. Sex-dependent thermogenesis, differ- 169. Quarta C, Mazza R, Pasquali R, Pagotto U. Role of sex hor-
ences in mitochondrial morphology and function, and mones in modulation of brown adipose tissue activity. J Mol
adrenergic response in brown adipose tissue. J Biol Chem Endocrinol 2012;49:R1-7.
2002;277:42958-63. 170. González-García I, Tena-Sempere M, López M. Estradiol
156. Kim SN, Jung YS, Kwon HJ, Seong JK, Granneman JG, Lee regulation of brown adipose tissue thermogenesis. Adv Exp
YH. Sex differences in sympathetic innervation and browning Med Biol 2017;1043:315-35.
of white adipose tissue of mice. Biol Sex Differ 2016;7:67. 171. Gantner ML, Hazen BC, Eury E, Brown EL, Kralli A. Com-
157. Norheim F, Hasin-Brumshtein Y, Vergnes L, Chella Krishnan plementary roles of estrogen-related receptors in brown adi-
K, Pan C, Seldin MM, et al. Gene-by-sex interactions in mi- pocyte thermogenic function. Endocrinology 2016;157:4770-
tochondrial functions and cardio-metabolic traits. Cell Metab 81.
2019;29:932-49.e4. 172. Xu Y, López M. Central regulation of energy metabolism by
158. Keuper M, Jastroch M. The good and the BAT of metabolic estrogens. Mol Metab 2018;15:104-15.
sex differences in thermogenic human adipose tissue. Mol 173. Monjo M, Rodríguez AM, Palou A, Roca P. Direct effects of
Cell Endocrinol 2021;533:111337. testosterone, 17 beta-estradiol, and progesterone on adrener-
159. Gómez-García I, Trepiana J, Fernández-Quintela A, Giralt gic regulation in cultured brown adipocytes: potential mecha-
M, Portillo MP. Sexual dimorphism in brown adipose tissue nism for gender-dependent thermogenesis. Endocrinology
activation and white adipose tissue browning. Int J Mol Sci 2003;144:4923-30.
2022;23:8250. 174. Rodríguez-Cuenca S, Monjo M, Gianotti M, Proenza AM,
160. Gilsanz V, Smith ML, Goodarzian F, Kim M, Wren TA, Hu Roca P. Expression of mitochondrial biogenesis-signaling fac-
HH. Changes in brown adipose tissue in boys and girls dur- tors in brown adipocytes is influenced specifically by 17beta-
ing childhood and puberty. J Pediatr 2012;160:604-9.e1. estradiol, testosterone, and progesterone. Am J Physiol Endo-
161. Cohade C, Mourtzikos KA, Wahl RL. "USA-fat": prevalence is crinol Metab 2007;292:E340-6.
related to ambient outdoor temperature-evaluation with 18F- 175. Hashimoto O, Noda T, Morita A, Morita M, Ohtsuki H, Sugi-
FDG PET/CT. J Nucl Med 2003;44:1267-70. yama M, et al. Castration induced browning in subcutane-
162. Cronin CG, Prakash P, Daniels GH, Boland GW, Kalra MK, ous white adipose tissue in male mice. Biochem Biophys Res
Halpern EF, et al. Brown fat at PET/CT: correlation with pa- Commun 2016;478:1746-50.
tient characteristics. Radiology 2012;263:836-42. 176. Harnichar AE, Zubiría MG, Giordano AP, Miguel I, Rey
163. Nookaew I, Svensson PA, Jacobson P, Jernås M, Taube M, MA, Spinedi E, et al. Inhibitory effect of androgens on white
Larsson I, et al. Adipose tissue resting energy expenditure adipose tissue thermogenic capacity. Mol Cell Endocrinol
and expression of genes involved in mitochondrial function 2022;543:111542.
are higher in women than in men. J Clin Endocrinol Metab 177. Shorakae S, Jona E, de Courten B, Lambert GW, Lambert EA,
2013;98:E370-8. Phillips SE, et al. Brown adipose tissue thermogenesis in poly-
164. van den Beukel JC, Grefhorst A, Hoogduijn MJ, Steenbergen J, cystic ovary syndrome. Clin Endocrinol (Oxf) 2019;90:425-
Mastroberardino PG, Dor FJ, et al. Women have more poten- 32.
tial to induce browning of perirenal adipose tissue than men. 178. Oliveira FR, Mamede M, Bizzi MF, Rocha ALL, Ferreira CN,
Obesity (Silver Spring) 2015;23:1671-9. Gomes KB, et al. Brown adipose tissue activity is reduced in
165. Graham TE. Thermal, metabolic, and cardiovascular changes women with polycystic ovary syndrome. Eur J Endocrinol
in men and women during cold stress. Med Sci Sports Exerc 2019;181:473-80.
1988;20(5 Suppl):S185-92. 179. Benrick A, Chanclón B, Micallef P, Wu Y, Hadi L, Shelton JM,
166. Karjalainen S. Thermal comfort and gender: a literature re- et al. Adiponectin protects against development of metabolic
view. Indoor Air 2012;22:96-109. disturbances in a PCOS mouse model. Proc Natl Acad Sci U S
167. Fletcher LA, Kim K, Leitner BP, Cassimatis TM, O'Mara AE, A 2017;114:E7187-96.
Johnson JW, et al. Sexual dimorphisms in adult human brown 180. Yuan X, Hu T, Zhao H, Huang Y, Ye R, Lin J, et al. Brown
adipose tissue. Obesity (Silver Spring) 2020;28:241-6. adipose tissue transplantation ameliorates polycystic ovary
168. Herz CT, Kulterer OC, Prager M, Marculescu R, Langer FB, syndrome. Proc Natl Acad Sci U S A 2016;113:2708-13.
Prager G, et al. Sex differences in brown adipose tissue activ- 181. Movérare-Skrtic S, Venken K, Andersson N, Lindberg MK,
ity and cold-induced thermogenesis. Mol Cell Endocrinol Svensson J, Swanson C, et al. Dihydrotestosterone treatment

18 www.wjmh.org
Masayuki Saito and Yuko Okamatsu-Ogura: Thermogenic Brown Fat in Humans

results in obesity and altered lipid metabolism in orchidecto- 189. Vidal P, Stanford KI. Exercise-induced adaptations to adi-
mized mice. Obesity (Silver Spring) 2006;14:662-72. pose tissue thermogenesis. Front Endocrinol (Lausanne)
182. Nohara K, Laque A, Allard C, Münzberg H, Mauvais-Jarvis 2020;11:270.
F. Central mechanisms of adiposity in adult female mice with 190. Ikeda K, Yamada T. UCP1 dependent and independent ther-
androgen excess. Obesity (Silver Spring) 2014;22:1477-84. mogenesis in brown and beige adipocytes. Front Endocrinol
183. Spaanderman DCE, Nixon M, Buurstede JC, Sips HC, Schilp- (Lausanne) 2020;11:498.
eroort M, Kuipers EN, et al. Androgens modulate glucocorti- 191. Chen KY, Cypess AM, Laughlin MR, Haft CR, Hu HH, Bre-
coid receptor activity in adipose tissue and liver. J Endocrinol della MA, et al. Brown adipose reporting criteria in imaging
2018. doi: 10.1530/JOE-18-0503 [Epub]. studies (BARCIST 1.0): recommendations for standard-
184. Fan W, Yanase T, Nomura M, Okabe T, Goto K, Sato T, et al. ized FDG-PET/CT experiments in humans. Cell Metab
Androgen receptor null male mice develop late-onset obesity 2016;24:210-22.
caused by decreased energy expenditure and lipolytic activ- 192. Chondronikola M, Beeman SC, Wahl RL. Non-invasive meth-
ity but show normal insulin sensitivity with high adiponectin ods for the assessment of brown adipose tissue in humans. J
secretion. Diabetes 2005;54:1000-8. Physiol 2018;596:363-78.
185. Stocco C. Tissue physiology and pathology of aromatase. Ste- 193. Law J, Morris DE, Izzi-Engbeaya C, Salem V, Coello C, Rob-
roids 2012;77:27-35. inson L, et al. Thermal imaging is a noninvasive alternative to
186. Kim NR, David K, Corbeels K, Khalil R, Antonio L, Schol- PET/CT for measurement of brown adipose tissue activity in
laert D, et al. Testosterone reduces body fat in male mice by humans. J Nucl Med 2018;59:516-22.
stimulation of physical activity via extrahypothalamic ERα 194. Nirengi S, Wakabayashi H, Matsushita M, Domichi M, Suzuki
signaling. Endocrinology 2021;162:bqab045. S, Sukino S, et al. An optimal condition for the evaluation of
187. Giordano A, Frontini A, Cinti S. Convertible visceral fat as human brown adipose tissue by infrared thermography. PLoS
a therapeutic target to curb obesity. Nat Rev Drug Discov One 2019;14:e0220574.
2016;15:405-24. 195. Hamaoka T, Nirengi S, Fuse S, Amagasa S, Kime R, Kuroiwa
188. Phillips KJ. Beige fat, adaptive thermogenesis, and its regu- M, et al. Near-infrared time-resolved spectroscopy for assess-
lation by exercise and thyroid hormone. Biology (Basel) ing brown adipose tissue density in humans: a review. Front
2019;8:57. Endocrinol (Lausanne) 2020;11:261.

www.wjmh.org 19

You might also like