Download PDF
Review  |  Open Access  |  27 Apr 2024

The role of brown adipose tissue in mediating healthful longevity

Views: 139 |  Downloads: 12 |  Cited:   0
J Cardiovasc Aging 2024;4:17.
10.20517/jca.2024.01 |  © The Author(s) 2024.
Author Information
Article Notes
Cite This Article

Abstract

There are two major subtypes of adipose tissue, i.e., white adipose tissue (WAT) and brown adipose tissue (BAT). It has been known for a long time that WAT mediates obesity and impairs healthful longevity. More recently, interest has focused on BAT, which, unlike WAT, actually augments healthful aging. The goal of this review is to examine the role of BAT in mediating healthful longevity. A major role for BAT and its related beige adipose tissue is thermogenesis, as a mechanism to maintain body temperature by producing heat through uncoupling protein 1 (UCP1) or through UCP1-independent thermogenic pathways. Our hypothesis is that healthful longevity is, in part, mediated by BAT. BAT protects against the major causes of impaired healthful longevity, i.e., obesity, diabetes, cardiovascular disorders, cancer, Alzheimer’s disease, reduced exercise tolerance, and impaired blood flow. Several genetically engineered mouse models have shown that BAT enhances healthful aging and that their BAT is more potent than wild-type (WT) BAT. For example, when BAT, which increases longevity and exercise performance in mice with disruption of the regulator of G protein signaling 14 (RGS14), is transplanted to WT mice, their exercise capacity is enhanced at 3 days after BAT transplantation, whereas BAT transplantation from WT to WT mice also resulted in increased exercise performance, but only at 8 weeks after transplantation. In view of the ability of BAT to mediate healthful longevity, it is likely that a pharmaceutical analog of BAT will become a novel therapeutic modality.

Keywords

Brown adipose tissue, healthful longevity, exercise, obesity, glucose and insulin protection, regulator of G protein signaling 14

INTRODUCTION

The goal of this review is to examine the role of brown adipose tissue (BAT) in mediating healthful longevity. This topic is of increasing importance as lifespan continues to increase, but is associated with adverse effects of diseases of aging, that not only decrease lifespan, but more importantly, decrease healthful lifespan. Our hypothesis is that the increased healthful lifespan is due in part to BAT. Much of the data supporting this hypothesis are derived from studying mouse models of healthful aging, a key feature of the current review.

However, in the past few years, the increase in longevity and healthful longevity has not been sustained and life expectancy has actually fallen in the U.S., which was triggered by an unprecedented rise in mortality associated with the COVID-19 pandemic, opioid overdose epidemic, and suicide[1]. According to a recent CDC report, life expectancy for the total U.S. population declined from 78.8 years in 2019 to 77.3 years in 2020, then further declined to 76.1 years in 2021, and then bounced back to 77.5 years in 2022[2]. This increase from 2022 does not fully offset the loss of 2.4 years of life expectancy between 2019 and 2021 that mostly resulted from increases in excess deaths due to the COVID-19 pandemic, opioid overdoses, and suicide[1]. For example, the number of drug overdose deaths increased by more than 16% from 2020 to 2021. From 2020 to 2021, opioid-involved death rates increased by over 15% and synthetic opioid-involved death rates (excluding methadone) increased by over 22%[3]. The current lifespan of 77.5 years is considerably longer than the lifespan prior to the year 2000. It has roughly doubled since 1860 and increased by a third since 1940[4].

White adipose tissue vs. brown adipose tissue

There are two major subtypes of adipose tissue, i.e., white adipose tissue (WAT) and brown adipose tissue (BAT). Whereas WAT plays a role in increasing obesity and impairing healthful longevity, BAT has the opposite effect. In addition to WAT and BAT, beige adipocytes are present in WAT and have functions similar to those of both WAT and BAT. It has been recognized for a long time that a major role for BAT and its related beige adipose tissue is thermogenesis as a mechanism to maintain body temperature by producing heat through uncoupling protein 1 (UCP1), which dissociates oxidative phosphorylation from ATP production, resulting in the release of heat[5-7]. Changes in BAT with aging reduce its effects on thermogenesis[8]. In addition, UCP1-independent thermogenic pathways have been found in BAT, beige adipocytes, and muscles[9]. These pathways include (1) creatine-substrate cycling in thermogenic adipocytes[10-12]; (2) Sacro-endoplasmic reticulum ATPase (SERCA)/Sarcolipin uncouple ATP hydrolysis from SERCA Ca2+ transport in BAT and muscle[13-16]; and (3) SERCA2b-mediated Ca2+ cycling in beige adipocytes[17]. BAT functions as a metabolic sink by oxidizing glucose and lipids, which produces heat. This has resulted in interest in BAT being a therapeutic modality to protect against obesity and diabetes. More recently, BAT has been shown to mediate longevity, but more importantly, healthful longevity.

Adipose tissue plays a vital role in regulating energy, and its activity relies on hormonal and nutritional signals that determine whether fat cells store excess nutrients as intracellular lipids or release stored energy as heat[18]. Changes associated with aging can affect the normal physiology of adipose tissues and alter their modulatory activity on energy regulation[19].

The major WAT depots in the body are found in the visceral cavity (vWAT) and subcutaneous cavity (scWAT). Compared with BAT and Beige adipocytes, WAT has less fatty acid oxidation, mitochondria, respiratory chain, and peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) levels[20]. Redistribution of WAT mass with advancing age is displayed by increased visceral adipose tissue in trunk and abdomen and reduced subcutaneous adipose tissue from limbs[21,22]. With aging, adipose tissue is characterized by dysregulated immune cells, preadipocytes, and senescent cells[21,22].

BAT consists of small, multilocular adipocytes (each cell has many small lipid droplets) and is responsible for dissipating energy through uncoupled respiration to produce heat[19,23]. Brown adipocytes are smaller in size than white adipocytes, with lipid droplets surrounding the nucleus. Brown adipocytes have mitochondria dispersed between the droplets, which give these cells a brown appearance. The cytoplasm also contains Golgi apparatus and only a small amount of ribosomes and endoplasmic reticulum. BAT is mainly located in the interscapular space of mice, and in humans, it is found in the interscapular, supraclavicular, suprarenal, and para-aortic spaces[24-27]. Browning of WAT is referred to as “beiging” with the cells obtaining a brown fat-like morphology and function[28].

Beige adipocytes are a distinct type of brown-like thermogenic adipocytes with multilocular morphology. They exist mainly in subcutaneous fat, but a small portion can also be found in visceral fat. Beige cells are generated through WAT browning, resulting in augmented non-shivering thermogenesis and metabolic capacity[29,30]. Beiging occurs predominantly in scWAT[31]. This process involves the upregulation of UCP1, a molecule that uncouples the respiratory chain from ATP synthesis, producing heat[32,33]. β-3 adrenergic receptors (β-3 ARs) mediate WAT browning during cold exposure[29,34-36]. Although β-1 ARs are expressed in BAT, they are not usually coupled to major functions in mature brown adipocytes[37]. β-1 ARs were reported to mediate preadipocyte recruitment, instead of WAT browning[29]. Although β-2 ARs mRNA and protein can be detected in BAT, they are most likely localized to the vascular system[38,39]. A recent report shows that β-2 ARs can activate human BAT[40]. In contrast to cold exposure, studies on local hyperthermia also revealed WAT browning[41-43]. Physical exercise copes with increased levels of specific molecules, including β-aminoisobutyric acid and irisin, which induce adipose tissue browning[44,45]. Calorie restriction and intermittent fasting also increase WAT browning and metabolic efficiency[46-48], and the intestinal microbiome regulates WAT browning, as it modulates bile acid levels, which are important for thermogenic activation[49]. Beige adipocytes are functionally related to brown adipocytes, which dissipate energy in the form of heat. The induction of beige adipocytes in human WAT depots is postulated to improve glucose, lipid metabolism, and obesity. In addition to thermogenesis and energy expenditure, like BAT, beige adipose tissue is also associated with improved glucose and lipid homeostasis and improved insulin sensitivity[50,51]. Specific cellular mechanisms have been identified in BAT, which regulate BAT’s role in metabolism, e.g., disruption of IL-6 diminishes BAT’s role in glucose homeostasis and insulin protection[52] and adipose-specific ablation of desnutrin/ATGL reduces BAT’s action by converting it to a WAT-like tissue[53]. A recent prospective clinical trial observed that in pre-diabetic humans, GLP1 analog treatment acutely induces IL-6 production by monocytes and IL-6 in the systemic circulation[54]. The same study showed that metronomic treatment with a GLP1 analog, liraglutide, leads to thermogenic adipose tissue activation in mice[54].

Secreting factors derived from BAT - brown adipokines or batokines

BAT and beige adipocytes have been identified as having a secretory role by releasing multiple autocrine and paracrine factors, to control the expansion and activity of BAT and the extent of WAT browning[55,56]. Additionally, endocrine factors derived from BAT can target peripheral tissues, such as WAT, liver, heart, pancreas, skeletal muscle, and bone. These factors derived from BAT are called brown adipokines or batokines[55,56]. Several batokines and their roles are identified, such as fibroblast growth factor 21 (FGF21)[57-60], Interleukin 6 (IL-6)[52,61], neuregulin 4 (NRG4)[62], insulin-like growth factor 1 (IGF-1)[63,64], CXCL14[65], 12,13-diHOME[66-68], Myostatin[69], GDF-15[70], and microRNAs (e.g., miR-99b)[71].

FGF21 was reported to regulate glucose uptake in BAT and browning of WAT[58], blood pressure regulation[59], and hypertensive cardiac remodeling[60]. IL-6 was reported to be associated with metabolic improvements[52] and hepatic gluconeogenesis enhancement[61]. A recent review of NRG4 has summarized its role in the regulation of energy homeostasis and glucolipid metabolism[62]. BAT-released IGF was reported to normalize glucose levels and reverse diabetes symptoms in a type 1 diabetes model[63]. Both CXCL14[65] and GDF-15[70] were reported to have effects on macrophages. 12,13-diHOME is associated with cardiac function[66], fatty-acid transportation[67], and increased fatty-acid uptake in skeletal muscle induced by exercise[68]. Elevated myostatin is associated with reduced exercise capacity, which was observed in the BAT-specific interferon regulator factor-4 (IRF4) KO mice[72]. In addition, miR-99b was reported to regulate hepatic production of FGF21[71].

BAT in humans from young to old

In infants, there is a high prevalence of BAT, but adults have less BAT, which is localized in a specific region of the body. Aherne and Hull state that in newborns, “many smaller masses of brown adipose tissue are present around the muscles and blood vessels of the neck. The main mass follows the course of the internal jugular vein and common carotid artery”[73].

In some studies, BMI and body fat percentage had a negative association with BAT prevalence, while resting metabolic rate had a strong positive correlation[26,74,75]. Cypess et al. found the prevalence of detectable BAT was higher in women (7.5%) than in men (3.1%)[25]. Out of those with detectable BAT, 48.1% of individuals were less than 50 years old, 34% were 50-64 years old, and 17.9% were over 64 years old[25]. BAT activity was observed in 23 of the 24 subjects during cold exposure but not under thermoneutral conditions[26]. The activity was significantly lower in the overweight or obese subjects than in the lean subjects[26]. The prevalence of detectable BAT was 36% in women (9 out of 25 individuals) and 32% in men (10 out of 31 individuals) with 2-h cold exposure[76]. Cold-activated BAT was detected in 125 (48%) out of 260 subjects at a median age of 26 (20-72 years). Out of those with detectable BAT, 26% of females (20 out of 76) and 54% of males (100 out of 184) exhibited detectable BAT. Compared with subjects without detectable BAT, those with detectable BAT were younger and showed lower adiposity-related parameters such as BMI, body fat mass, and abdominal fat area[77]. The loss of BAT regeneration with aging promotes the development of obesity and metabolic disorders of aging[78].

BAT mediating longevity

Studies have indicated that aging reduces BAT activity, leading to thermal dysregulation and energy imbalance[21,79,80]. However, the effects of age on BAT mass have been inconsistent[81-83]. Some studies have reported that aging increases the amount of BAT[81,82], whereas one study reported no change in BAT mass in rodents[83]. In addition, beige adipocyte formation declines with aging, which may be caused by changes in the adipose tissue microenvironment[21,22].

Several animal studies of aging have observed an association between BAT and aging, with increased age of median survival ranging from 13 to 68% compared to their wild type (WT) [Table 1], including Ames Dwarf mice[84,85], Growth Hormone Receptor/Binding Protein (GHR/BP) knockout (KO) mice[53,86], Phosphatase and Tensin Homolog transgenic (Ptentg) mice[87], Regulator of G Protein Signaling 14 (RGS14) KO mice[23], and Forkhead Box Protein A3 (Foxa3) KO mice[88]. Most of these studies have observed a correlation between aging and aspects of BAT, rather than a distinct examination of the extent to which surgical elimination of BAT affects lifespan. However, the model of disruption of the Regulator of G Protein Signaling 14 (RGS14), i.e., RGS14 KO, in mice is a model of extended longevity. When their BAT is transplanted to their WT, the RGS14 KO mouse without its BAT is no longer a model of longevity and the WT with the transplanted BAT becomes a model of longevity [Figure 1].

The role of brown adipose tissue in mediating healthful longevity

Figure 1. RGS14 KO Model of Longevity. (A) Kaplan-Meier survival curves for RGS14 KO and WT mice showed significantly augmented survival in RGS14 KO mice. (B and C) Median survival value and maximum lifespan were significantly greater in RGS14 KO mice than in WT mice for both males and females. In addition, medium and maximum lifespan were greater in female RGS14 KO mice than in male RGS14 KO mice. (D) Furthermore, 24-month-old RGS14 KO mice did not show the aging phenotype normally present in WT mice of similar age, including body atrophy, loss of hair, and greying of fur color. In support of the key role of BAT in aging, old WT RGS14 KO BAT recipient mice, which had BAT transplanted at 3-4 months of age, had the appearance of healthful aging similar to the old RGS14 KO mice. A representative example of each is shown in (D). For median lifespan analysis, Mood’s median test was used to determine differences in median lifespan. A Student’s t-test was used to test differences in maximum lifespan. Reprinted from Ref.[23].

Table 1

BAT models of longevity

ModelReferencesSexAge of median survival (Days)
MutantControl (WT)% increase
Ames dwarf[53]M1,07672349
F1,20671868
GHR/BP KO[79]M97562955
F1,03174938
Ptentg[55]M88078013
F91579616
RGS14KO[14]M84072017
F93072029
Foxa3 KO[56]M1,10085029

BAT mediating healthful longevity

More recently, interest has extended to BAT’s role in mediating healthful aging, primarily from data in genetically altered mouse models. Longevity mouse models with enhanced BAT activity / function are noted in Table 1. More importantly, these models mediate BAT’s role in healthful longevity, e.g., protection against obesity[89,90], diabetes[89,90], cardiovascular disorders[91-93], cancer[94-98], Alzheimer’s Disease[99], stroke[100,101], exercise intolerance[72], and reduced blood flow[72,102,103], all of which reduce healthful aging [Table 2]. Ames dwarf mice are protected against diabetes, CV stress, cancer, and Alzheimer’s Disease[84,104-108]. GHR/BP KO mice exhibit improved exercise capacity and are protected against obesity, hypertension, cancer, and Alzheimer’s Disease[86,109-112]. Ptentg mice are protected against obesity and diabetes, cancer, and Alzheimer’s Disease[87,113,114]. Interestingly, whereas Pten TG is a model of healthful longevity, cardiac-specific Pten KO has also been reported to have cardiac stress protection[115,116]. RGS14 KO mice exhibit improved exercise capacity[72], along with protection against obesity[23] and glucose and insulin intolerance[117], myocardial ischemia[118], and hypertension[119]. Foxa3 KO is reported to protect against obesity and diabetes and cancer[88,120] [Figure 2].

The role of brown adipose tissue in mediating healthful longevity

Figure 2. Mechanisms Promoting Healthful Aging By BAT. BAT leads to healthful aging by promoting thermogenesis, energy expenditure, insulin sensitivity, glucose uptake, oxidative stress resistance, mitochondrial function, and blood flow/angiogenesis. This results in protection against obesity, diabetes, exercise intolerance, cardiovascular disease, cancer, and Alzheimer’s disease.

Table 2

BAT models of healthy longevity

Mouse modelReferencesAspects of healthful longevity
Improved exerciseObesity protectionCV stress/coronary protectionHypertension protectionGlucose, insulin tolerance & diabetes protectionCancer protectionAlzheimer’s disease protection
WT BAT transplantation[39,70,91,92]****
BAT-specific p85α KO[93]**
Nrg4 TG[94-98]****
Ames dwarf[52,73-77]****
GHR/BP KO[54,78-80]****
Ptentg[55,81,83]*****
RGS14KO[14,70]*****
Foxa3 KO[56,90]***

In addition, there are models with enhanced BAT function or extra BAT amount by BAT transplantation that exhibit aspects of healthful longevity. WT mice receiving BAT from another WT BAT mouse exhibit improved exercise capacity, as well as protection against obesity and diabetes and cancer[52,72,121,122]. BAT-specific p85αKO mice also protect against obesity and diabetes[123]. Adipose-Specific Neuregulin 4 Transgenic (Nrg4tg) mice protect against obesity and diabetes, cardiovascular stress, and cancer[124-128]. It has also been suggested that low levels of BAT in humans are associated with obesity and glucose intolerance, whereas those with higher BAT levels maintain lower body weights and more healthful aging[129].

BAT and obesity

By 2015, 108 million children and 604 million adults worldwide were obese, contributing to 2.4 million deaths globally[130]. The activation of BAT leads to increased energy expenditure through the uncoupling of mitochondrial respiration, generating heat, and utilizing glucose and fatty acids in the process, thereby protecting against obesity[122,131]. Thus, BAT plays a crucial role in averting obesity by serving as a metabolic regulator that actively disperses energy as heat. Unlike WAT, which stores surplus energy as fat, BAT distinguishes itself through its distinctive capacity for non-shivering thermogenesis[132]. The activation of BAT is enhanced in response to cold exposure, as sympathetic nervous system stimulation releases norepinephrine, promoting the thermogenic function of BAT[133]. BAT significantly expresses UCP1 and β3-adrenoceptors, which mediate the sympathetic drive to mobilize and upregulate UCP1 to promote a large amount of energy loss in the form of heat energy[134]. By burning calories to produce heat, BAT contributes significantly to overall energy expenditure, thereby preventing the accumulation of excess fat and mitigating the risk of obesity. Apart from cold-induced thermogenesis, there is a proposed role for BAT thermogenesis in mediating diet-induced thermogenesis. A high-fat diet is linked to an increase in thermogenic capacity, elevated BAT mass, and higher levels of UCP1. Conversely, ablation of UCP1 results in reduced thermogenic capacity and increased susceptibility to diet-induced obesity[132], indicating an intricate interaction between mechanisms regulating energy balance and those controlling BAT. Recently, BAT transplantation has gained heightened attention in exploring the relationship between BAT and obesity. Studies have demonstrated that BAT transplantation enhances the utilization of stored energy, leading to a reduction in both body weight and body fat[52,135]. Beyond its role in energy metabolism, BAT emerges as a significant regulator of lipid metabolism, evidenced by a decrease in circulating triglycerides and an improvement in cholesterol profiles[52,135]. Overall, BAT serves as a dynamic metabolic force, actively burning calories to generate heat, thereby playing a crucial role in protecting against obesity by facilitating energy expenditure and metabolic well-being. Strategies to harness and activate BAT tissue hold promise for the development of anti-obesity interventions.

BAT and diabetes

As noted in the previous section, due to its role in increasing energy expenditure and promoting glucose and fatty acid uptake[122,131], BAT is recognized as an important tissue to combat the development of glucose intolerance and insulin resistance[122,131] and protect against the pre-diabetic state[77], obesity and allied metabolic disorders[86]. Ptentg mice, a longevity mouse model with increased BAT activity, demonstrated increases in energy expenditure and improved glucose homeostasis[87]. The RGS14 KO mice also show protection against glucose intolerance and insulin resistance[117]. A recent human study reported that reduced BAT mass is associated with an increased incidence of type 2 diabetes and cardiovascular disease[91].

Additionally, studies showed that increasing BAT mass by transplantation improves glucose metabolism and insulin sensitivity in mice[52,136-138]. BAT transplantation has been shown to protect against both type 1 diabetes by improving glycemia with increased IGF-1[63,139] and type 2 diabetes by improving glucose tolerance with increased IL-6[52] or adiponectin[135]. Ames dwarf mice, another aging model, also reported increased BAT activity along with greater oxygen consumption and energy expenditure[84,140], whereas surgical removal of BAT in this model resulted in a decrease in insulin sensitivity[84].

BAT and cardiovascular disorders

The role of BAT in protecting against cardiovascular diseases is a major component of its ability to induce healthful aging. Several potential mechanisms mediate the protective effects of BAT on the cardiovascular system. As noted in the BAT and Obesity section, the activation of BAT leads to increased energy expenditure through the uncoupling of mitochondrial respiration, generating heat, and utilizing glucose and fatty acids in the process. BAT activation has also been associated with improved insulin sensitivity, reduced triglyceride levels, and favorable changes in lipid profiles, all of which contribute to cardiovascular protection[52,141].

(1) BAT and Myocardial Ischemia

BAT has garnered significant attention in recent years due to its role in preventing myocardial ischemia, offering a novel perspective on cardiovascular health[91-93]. One study indicated that transplantation of CD29+ BAT-derived cells into the infarct border zone of acute myocardial infarction in rats resulted in reduced infarction area and improved left ventricular function[142]. The authors suggested that BAT-derived cells are useful for a new strategy in cardiomyocyte regeneration. Another study with BAT-derived stem cells also demonstrated smaller infarct size in rats with myocardial ischemia after 4 weeks of permanent coronary artery occlusion, with increased vessel density in the peri-infarct zone[143]. Recent studies also demonstrated that BAT dysfunction is associated with increased left ventricular mass and larger myocardial infarct size[122,144] , which also supports the role of BAT in protecting against myocardial ischemia. Similarly, the RGS14 KO mice have been shown to demonstrate myocardial ischemic protection[118], with an important mechanism of that model’s ability to increase blood flow by angiogenesis[72]. A human study found that greater BAT activity was associated with reduced myocardial ischemia and protection against cardiac arrest and myocardial infarction[145]. Further exploration of BAT's role in ischemic protection may open new avenues for innovative preventive and therapeutic strategies in the treatment of cardiovascular diseases.

(2) BAT and Heart Failure

Several reports have linked BAT and heart failure. In a murine model of heart failure with preserved ejection fraction (HFpEF), BAT function was reduced[146]. Tahara et al. reported the results of a 23-year-old female patient with heart failure who had low body temperature and suggested insufficient BAT-induced thermogenesis in this patient[147]. A more recent study reported that thoracic aortic constriction-induced heart failure reduced the thermogenic capacity of BAT in mice, leading to a significant reduction in body temperature with cold exposure[148], while increased BAT function improved cardiac function in mice with thoracic aortic constriction[148]. Conversely, thoracic aortic constriction reduced systolic function in a mouse model of genetic BAT dysfunction, resulting in reduced survival after thoracic aortic constriction[148]. Similarly, another study suggested that BAT is activated in a model of catecholamine-induced cardiomyopathy, resulting in cardioprotection and protection against pathological left ventricle remodeling[149].

(3) BAT and Hypertension

The prior literature on BAT and hypertension is controversial. One study showed that a transgenic model of reduced obesity and ablation of BAT (UCP - diphtheria toxin Achain (UCP-DTA)) mice[150] is associated with systemic hypertension[151]. In contrast, another showed that an angiotensin type II agonist induces hypertension and enhances the browning of WAT[152]. Furthermore, research showed that adenosine A2A receptor KO (A2ARKO) mouse is a hypertension model[153] with interscapular BAT (iBAT) dysfunction[60]. The RGS14 KO mouse model is also protected against hypertension[119], with an important mechanism involving its ability to increase blood flow through angiogenesis[72]. Additionally, perivascular fat and its browning have been reported to play a role in the development of hypertension[154,155], including regulation of vascular contractility[156] and vasodilation[157]. A recent study found that browning of perivascular adipose tissue prevents vascular dysfunction and reduces angiotensin II-induced hypertension in mice[158]. It has been suggested that BAT-secreted factors (batokines) contribute to the regulation of blood pressure[159]. For example, BAT secretes increased FGF21 after stimulation, and administration of FGF21 lowers blood pressure[59]. However, higher serum FGF21 levels are associated with higher blood pressure in humans[160,161]. Treatment with another batokine, IGF-1, lowers blood pressure by stimulating nitric oxide production from vascular endothelium and smooth muscle cells[162,163]. In addition, another study showed that BAT resulted in reduced vascular contractility through the Nox4-derived H2O2 pathway[164].

In humans, thermogenic brown and beige adipose tissue are considered to have protective effects on the vasculature, as individuals with detectable thermogenic adipose tissue have reduced risk for hypertension and coronary artery disease, relative to individuals without thermogenic adipose tissue[91].

BAT and blood flow and angiogenesis

Compared to WAT, active BAT is highly vascularized with abundant mitochondria that produce heat through uncoupled respiration[165]. It has been shown that by stimulating angiogenesis and the conversion of WAT to brown-like adipocytes, weight gain in obese mice can be inhibited[166]. The thermogenesis function of BAT relies on blood flow to be supplied with nutrients and oxygen and for the distribution of the generated heat to the rest of the body. VEGF is an important angiogenic factor regulating angiogenesis, arteriogenesis, and blood flow. A direct connection between VEGF and BAT has already been established, as VEGF is known to play a direct and positive role in the activation of BAT[167]. VEGF also acts in an endocrine and paracrine manner in BAT by stimulating the proliferation of vascular endothelial cells[168]. Our recent study found that RGS14 KO mice have increased VEGF expression in the skeletal muscle and BAT[72]. Removing BAT from RGS14 KO mice resulted in the loss of a significant increase in hindlimb perfusion, while the addition of RGS14 KO BAT to WT mice led to increases in perfusion[72]. This addition also led to changes in the vasculature, with RGS14 KO BAT recipients exhibiting increases in capillary and arteriole density[72]. Other studies have shown that the activation of BAT is accompanied by the proliferation of blood vessels[102], transplanted BAT from C57B/L6 mice becomes re-vascularized[52], and receiving BAT from Fat-1 transgenic mice upregulates VEGF levels in endogenous BAT[103]. BAT transplantation effectively reverses skin sclerosis in mice through mechanisms involving inflammation reduction and promotion of angiogenesis[169]. In contrast, a recent study found the opposite, i.e., that the transplanted BAT derived from C57B/L6 mice did not improve blood flow or VEGF levels in HFD-fed mice[170]. Genetic deletion, as well as pharmacological inhibition of endothelial VEGFR1, increased adipose angiogenesis and browning of subcutaneous adipose tissue, leading to elevated thermogenesis[171].

BAT and stroke

Based on the role of BAT in energy metabolism, it has been suggested that the inhibition of BAT thermogenesis could facilitate the induction of therapeutic hypothermia for fever reduction or improve outcomes in stroke through a lowering of metabolic oxygen demand[172]. Some studies reported the potential role of BAT in a cerebral ischemic rat model. O’Shaughnessy et al. reported that BAT activity correlated with resting oxygen consumption in the cerebral ischemic group[100]. Another study found that hypobaric hypoxia preconditioning significantly attenuated the increases in cellular ischemia and injury indicators in the hypothalamus, along with reduced BAT weight[101].

BAT and exercise

Enhanced exercise capacity is not only a feature of healthful aging, but also is a therapy for aging patients and patients with cardiovascular disease. Exercise is a healthy way to reduce body weight by activating the sympathetic nervous system, accelerating the decomposition of fat, and promoting the utilization and consumption of energy in skeletal muscle[173-175]. During aging, it is known that progressive loss of exercise capacity relates to loss of skeletal muscle mass and tissue function[176]. Decreased muscle mitochondrial function contributes to the loss of skeletal muscle function during aging[177-181]. Regular exercise or exercise training protects against decreased muscle function during aging[182,183], frailty status[184,185], and neurodegeneration[186,187].

Numerous studies have suggested that exercise may play a role in regulating BAT activation. Exercise boosts the expression of UCP1 and genes associated with mitochondria biogenesis, thereby improving BAT's heat production capacity[6]. For instance, swim training in rodents over six to eight weeks increased UCP1 protein levels in BAT[188,189]. Similarly, treadmill exercise in rodents for 6-8 weeks increased BAT activity and cytochrome oxidase activity, oxygen consumption rates, and BAT-specific gene markers, e.g., UCP1, FGF21, and PGC1α[190,191]. However, conflicting findings also exist, with some studies suggesting that exercise may reduce the thermogenic effect of BAT. In rats, six to eight weeks of moderate-intensity treadmill exercise led to decreased UCP1 expression in BAT and a reduction in total BAT mass[192,193]. Human studies also showed inconsistent results regarding the role of exercise on BAT modulation, with some indicating that high-intensity physical activities can increase BAT density[194], while others report that exercise decreases glucose uptake in BAT[195-197]. While most of these studies have shown that exercise increases BAT, relatively few have shown that BAT increases exercise performance.

One example of a genetic model demonstrating that BAT can enhance exercise performance is that of RGS14 KO mice, a healthful lifespan model, mediated by increased BAT[23]. One mechanism mediating the increase in healthful lifespan is enhanced exercise capacity, a feature of the RGS14 KO mouse[72] [Figure 3]. RGS14 KO mice demonstrated 160% ± 9% increased maximal running distance and 154% ± 6% increased work to exhaustion, compared to WT mice. Similarly, RGS14 KO BAT transplanted to WT mice demonstrated a 151% ± 5% increased maximal running distance and 158% ± 7% increased work to exhaustion, which corresponded to the enhanced exercise capacity of RGS14 KO mice. The enhanced exercise capacity observed in WT mice with RGS14 KO BAT transplants was observed at three days after BAT transplantation, whereas BAT transplantation from WT to WT mice also resulted in increased exercise performance, but not at 3 days, but only at 8 weeks after transplantation [Figure 4][72].

The role of brown adipose tissue in mediating healthful longevity

Figure 3. Mechanisms mediating enhanced exercise capacity in RGS14 KO and its uniquely powerful BAT. Multiple mechanisms mediated the enhanced exercise capacity in RGS14 KO mice. The most important mechanism is BAT, which mediates SIRT3, MnSOD, MEK/ERK, and VEGF pathways. These mechanisms regulate exercise capacity by improving mitochondrial function, providing protection against oxidative stress, and improving blood flow/angiogenesis. Reprinted from Ref.[72].

The role of brown adipose tissue in mediating healthful longevity

Figure 4. Increased BAT Cell Numbers and Increased Exercise Capacity in RGS14 KO Mice. RGS14 KO mice exhibited smaller brown adipocytes (A and B), and an increased number of brown adipocytes (A and C) than WT control mice. RGS14 KO mice ran longer distances (D) with increased work to exhaustion (E) compared to WT littermates. BAT transplantation from RGS14 KO mice to WT mice led to a reversal of phenotype, such that RGS14 KO BAT recipients exhibited improved running distance (F) and greater work to exhaustion (G) compared to RGS14 KO BAT donors, at 3 days after RGS14 KO BAT transplantation. In contrast, there was no improvement in running distance and work to exhaustion at 3 days after transplantation of BAT from C57BL6/J WT mice to other C57BL6/J WT mice (H and I). It required 8 weeks to achieve enhanced running distance and work to exhaustion in C57BI/6J WT mice with BAT transplantation from other C57BL6/J WT mice (J and K). Reprinted from Ref.[72].

The BAT-induced enhanced exercise capacity was mediated by (1) mitochondrial biogenesis and SIRT3; and (2) antioxidant defense and the MEK/ERK pathway, and increased hind limb perfusion [Figure 5]. Thus, BAT from WT or from RGS14 KO mice mediates enhanced exercise capacity, but the BAT from RGS14 KO mice was more powerful than from WT [Figure 4].

The role of brown adipose tissue in mediating healthful longevity

Figure 5. Enhanced Exercise by RGS14 KO Mice Is Mediated by BAT and Angiogenesis /Arteriogenesis Increasing HindLimb Blood Flow. Non-linear contrast imaging was used to measure hindlimb blood flow. The average data are presented as % of WT perfusion, which is represented as 100% (A). Hindlimb blood flow was higher in RGS14 KO mice compared to WT mice, and higher in WT mice that received RGS14 KO BAT, at 3 days after transplantation (A and B), while RGS14 KO BAT donors lost their enhanced hindlimb perfusion, with results similar to WT mice (A). With treatment of the SIRT3 inhibitor, 3-YTP, both RGS14 KO mice and RGS14 KO BAT recipients lost their enhanced hindlimb perfusion (A). Angiogenesis (reflected by capillary density) and arteriogenesis (reflected by arteriole density) were both increased in skeletal muscle of RGS14 KO mice (B and C) and RGS14 KO BAT recipients (D and E), which correlated with increased VEGF in skeletal muscle (F) and BAT (G). Reprinted from Ref.[72].

SIRT3, a mitochondrial sirtuin deacetylase, regulates the expression of many BAT mitochondrial proteins including UCP1[198], and is also upregulated with exercise in animal models[199]. We have previously shown that the SIRT3 is upregulated in the BAT and skeletal muscle of RGS14 KO mice along with enhanced mitochondrial biogenesis[72], and therefore, SIRT3 is considered an important regulator of exercise capacity in this model. These changes correlated with exercise capacity, such that RGS14 KO × SIRT3 KO mice did not show the enhanced exercise capacity of RGS14 KO mice[72]. The role of SIRT3 was necessary for the enhanced exercise capacity seen in the RGS14 KO mouse and WT mice with BAT transplantation, since the enhancement of exercise capacity upon RGS14 KO BAT transplantation to WT mice was not observed when BAT was transplanted from RGS14 KO × SIRT3 KO mice, even 6 months after transplantation[72]. MnSOD is a primary mitochondrial ROS scavenging enzyme, which can be activated by SIRT3[200]. SIRT3 and MnSOD (SOD2) have been linked to improved exercise, showing both that SIRT3 can improve exercise performance and conversely that exercise can lead to increased SIRT3[199,201,202]. Other studies have also shown that SIRT3 maintains BAT morphology and function and protects against obesity and age-related metabolic diseases[203]. The RGS14 KO mouse exhibits increased MnSOD activity, which also contributes to its enhanced exercise capacity, as confirmed by partial genetic ablation of MnSOD, which abolished the enhanced exercise capacity[72].

RGS14 itself is also known to directly affect ERK signaling[204], which is involved in angiogenesis/arteriogenesis[205,206]. The enhanced exercise capacity of RGS14 KO mice is also regulated by the MEK/ERK pathway, as this enhancement of exercise capacity was abolished by treatment with a MEK inhibitor, U0126[72]. Another powerful mediator of exercise performance is blood flow. RGS14 KO mice exhibit enhanced hindlimb blood flow, which is accompanied by increases in angiogenesis and arteriogenesis in the hindlimb vasculature, leading to increased capillary and arteriole density[72] [Figure 5]. RGS14 KO mice have increased VEGF expression, a key angiogenic factor, in the skeletal muscle and BAT[72]. Moreover, VEGF plays a positive role in the activation and expansion of BAT[167]. VEGF also acts in an endocrine and paracrine manner in BAT by stimulating the proliferation of vascular endothelial cells[168]. Removing BAT from RGS14 KO mice resulted in the loss of a significant increase in hindlimb perfusion, while the addition of RGS14 KO BAT to WT mice led to increases in hind limb perfusion, along with RGS14 KO BAT recipients exhibiting increases in capillary and arteriole density[72].

In contrast to the RGS14 KO data showing a positive action of BAT in mediating the enhanced exercise capacity, another study suggested that BAT from IRF4 KO mice might contribute negatively to skeletal muscle performance[69]. In that study, BAT from IRF4 KO mice produced and secreted myostatin[72], which negatively regulates skeletal muscle cell differentiation[69].

BAT and cancer

Cancer is the leading cause of mortality in more than 100 countries worldwide[207]. The multifaceted involvement of BAT in the realm of cancer is becoming increasingly apparent. While BAT has conventionally been recognized for its contributions to thermogenesis and energy expenditure, it has now emerged as a factor influencing the development and progression of cancer. Most studies on the relationship between BAT and cancer have shown an increased prevalence of BAT activity in cancer patients or animals[94-98]. BAT mass or size was smaller in the cancer groups mainly due to the weight loss as a response to anorexia during the development of cachexia[94]. In addition, inhibition of WAT browning ameliorates the severity of cancer-associated cachexia with skin tumors[208]. Another study identified a significant association between the expression of UCP1 and improved overall survival in a cohort of patients with colorectal cancer[209]. Seki et al. observed significant tumor suppression in immunocompetent mice subcutaneously implanted with colorectal cancer cells, when the mice were housed in a 4 °C environment compared to those in 30 °C[210]. This environmental difference led to an upregulation of BAT and an impressive 80% reduction in tumor growth by day 20 post-tumor inoculation, underscoring the potential involvement of BAT in colorectal cancer progression[210]. An investigation into the connection between hepatocellular cancer and BAT in a mouse model demonstrated that the removal of BAT resulted in increased tumor growth[211]. This was accompanied by a more pronounced increase in liver weight and serum triacylglycerol levels.

However, not all studies have found that BAT protects against cancer. One study found no difference in the prevalence of activated BAT in cancer patients vs. that in healthy control subjects[212]. Other studies showed that BAT exerts an adverse effect on cancer[96,213,214]. In addition, an accelerated tumor growth rate has been shown with BAT or WAT[213]. BAT also plays a major role in breast cancer as well. One study revealed a high expression of markers associated with BAT and beige adipocytes in breast cancer xenografts, suggesting that thermal characteristics might play a pivotal role in the progression of breast cancer[98]. Consistent with this discovery, a retrospective analysis of data from 96 breast cancer patients who underwent FDG PET/CT scans for routine staging uncovered a three-fold higher BAT activity in breast cancer patients compared to controls with other types of cancers[95]. Cancer-associated cachexia, characterized by weakness, fat loss, and muscle wasting, is the primary contributor to complications in individuals with malignancies, resulting in diminished quality of life and unfavorable outcomes[215,216]. Investigations examining the role of BAT in cancer cachexia have yielded inconsistent findings. Some studies suggest that thermogenic fat plays a role in cancer cachexia due to its pivotal functions in heat production and energy balance[217,218]. Conversely, recent retrospective analyses of cancer patients propose that BAT is not associated with cancer-associated cachexia and does not exacerbate overall survival outcomes in individuals with cachexia[219,220] .

BAT and Alzheimer’s disease

There is evidence that the age-associated thermoregulatory deficit induces diverse metabolic changes associated with Alzheimer’s Disease development. BAT has been involved in various functions that prevent Alzheimer’s Disease, such as regulating energy metabolism, secreting hormones, improving insulin sensitivity, and increasing glucose utilization[99]. BAT decreased significantly in an 18-month-old Alzheimer’s Disease mouse model, suggesting a potential role for BAT in protecting against Alzheimer’s disease[221].

Characteristics of Alzheimer’s Disease are the accumulation of neurofibrillary tangles, amyloid plaques, neuropil threads, and dystrophic neurites containing hyperphosphorylated tau[222-224]. Hypothermia is one of the notable causes of tau hyperphosphorylation. Previous studies reveal that each degree Celsius below normothermic conditions induced an 80% rise in tau phosphorylation at the pThr212 and pSer396/pSer404 epitopes[225,226]. Decreased BAT function during aging may explain the thermoregulatory deficits in the elderly and the underlying mechanisms of Alzheimer’s Disease[121].

Activation of BAT leads to an increased release of FGF21. FGF21, in turn, reduces brain oxidative stress and neuroinflammation by enhancing antioxidant activity and diminishing proinflammatory cytokines such as TNF-α and IL-6, critical factors in Alzheimer’s Disease pathogenesis[227]. Hormones produced by BAT, including adiponectin and leptin, contribute to neuroprotective effects by inhibiting proinflammatory cytokines and suppressing Aβ production, thereby attenuating tau phosphorylation[228,229], providing additional evidence of BAT's involvement in Alzheimer’s Disease.

Diabetes/insulin resistance is another risk factor for the incidence of Alzheimer’s Disease. Impaired brain insulin signaling can instigate neurocognitive diseases, and impaired glucose uptake is among the hallmark deficits in the Alzheimer’s Disease brain[230,231]. Taken together, these studies indicate that BAT could be a novel target in Alzheimer’s Disease therapy.

CONCLUSIONS

It has been recognized for a long time that obesity mediated by WAT is a major cause of reducing both longevity and healthful longevity. More recently, there has been interest in another type of adipose tissue, BAT, with a mechanism of thermogenesis, which helps maintain body temperature. Most prior studies on BAT have focused on its ability to reduce obesity and protect against diabetes. More recently, the focus has shifted to a role for BAT in mediating other aspects of healthful longevity [Figure 2]. Several mouse models derived from genetic mutations have increased BAT and have been shown to mediate healthful longevity [Tables 1 and 2]. One of these newer models, i.e., RGS14 KO, has a BAT that is more powerful than BAT in WT controls and is involved in mediating the extension of healthful longevity, and not only protects against obesity, cardiovascular disease, glucose intolerance, and Alzheimer’s disease, but also exhibits improved exercise performance and angiogenesis. In view of the ability of BAT to mediate healthful longevity, it is likely that a pharmaceutical analog of BAT will become a novel therapeutic modality.

DECLARATIONS

Authors’ contributions

Design of Review: Vatner DE, Vatner SF

Writing of the Manuscript: Zhang J, Kibret BG, Vatner DE, Vatner SF

Availability of data and materials

Not applicable.

Financial support and sponsorship

This study was supported by the National Institutes of Health grant R21AG075656 (to Vatner SF).

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2024.

REFERENCES

1. Curtin SC, Garnett MF, Ahmad FB. Provisional estimates of suicide by demographic characteristics: United States, 2022. Available from: https://stacks.cdc.gov/view/cdc/135466 [Last accessed on 24 Apr 2024].

2. Elizabeth A, Kochanek KD, Xu J, Tejada-Vera B. Provisional life expectancy estimates for 2022. Available from: https://stacks.cdc.gov/view/cdc/135467 [Last accessed on 24 Apr 2024].

3. Spencer MR, Miniño MA, Warner M. Drug overdose deaths in the United States, 2001-2021. Available from: https://stacks.cdc.gov/view/cdc/122556 [Last accessed on 24 Apr 2024].

4. Statista. Life expectancy (from birth) in the United States, from 1860 to 2020*. Available from: https://www.statista.com/statistics/1040079/life-expectancy-united-states-all-time/ [Last accessed on 24 Apr 2024].

5. Rosen ED, Spiegelman BM. What we talk about when we talk about fat. Cell 2014;156:20-44.

6. Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev 2004;84:277-359.

7. Himms-Hagen J. Brown adipose tissue thermogenesis and obesity. Prog Lipid Res 1989;28:67-115.

8. Gohlke S, Zagoriy V, Cuadros Inostroza A, et al. Identification of functional lipid metabolism biomarkers of brown adipose tissue aging. Mol Metab 2019;24:1-17.

9. Ikeda K, Yamada T. UCP1 dependent and independent thermogenesis in brown and beige adipocytes. Front Endocrinol 2020;11:498.

10. Kazak L, Chouchani ET, Jedrychowski MP, et al. A creatine-driven substrate cycle enhances energy expenditure and thermogenesis in beige fat. Cell 2015;163:643-55.

11. Kazak L, Chouchani ET, Lu GZ, et al. Genetic depletion of adipocyte creatine metabolism inhibits diet-induced thermogenesis and drives obesity. Cell Metab 2017;26:693.

12. Yamashita H, Ohira Y, Wakatsuki T, et al. Increased growth of brown adipose tissue but its reduced thermogenic activity in creatine-depleted rats fed beta-guanidinopropionic acid. Biochim Biophys Acta 1995;1230:69-73.

13. Pant M, Bal NC, Periasamy M. Sarcolipin: a key thermogenic and metabolic regulator in skeletal muscle. Trends Endocrinol Metab 2016;27:881-92.

14. Sahoo SK, Shaikh SA, Sopariwala DH, Bal NC, Periasamy M. Sarcolipin protein interaction with sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) is distinct from phospholamban protein, and only sarcolipin can promote uncoupling of the SERCA pump. J Biol Chem 2013;288:6881-9.

15. Smith WS, Broadbridge R, East JM, Lee AG. Sarcolipin uncouples hydrolysis of ATP from accumulation of Ca2+ by the Ca2+-ATPase of skeletal-muscle sarcoplasmic reticulum. Biochem J 2002;361:277-86.

16. Meis L. Brown adipose tissue Ca2+-ATPase: uncoupled ATP hydrolysis and thermogenic activity. J Biol Chem 2003;278:41856-61.

17. Ikeda K, Kang Q, Yoneshiro T, et al. UCP1-independent signaling involving SERCA2b-mediated calcium cycling regulates beige fat thermogenesis and systemic glucose homeostasis. Nat Med 2017;23:1454-65.

18. Manteiga S, Choi K, Jayaraman A, Lee K. Systems biology of adipose tissue metabolism: regulation of growth, signaling and inflammation. Wiley Interdiscip Rev Syst Biol Med 2013;5:425-47.

19. Li X, Frazier JA, Spahiu E, McPherson M, Miller RA. Muscle-dependent regulation of adipose tissue function in long-lived growth hormone-mutant mice. Aging 2020;12:8766-89.

20. Lizcano F. The beige adipocyte as a therapy for metabolic diseases. Int J Mol Sci 2019;20:5058.

21. Zoico E, Rubele S, De Caro A, et al. Brown and beige adipose tissue and aging. Front Endocrinol 2019;10:368.

22. Ou MY, Zhang H, Tan PC, Zhou SB, Li QF. Adipose tissue aging: mechanisms and therapeutic implications. Cell Death Dis 2022;13:300.

23. Vatner DE, Zhang J, Oydanich M, et al. Enhanced longevity and metabolism by brown adipose tissue with disruption of the regulator of G protein signaling 14. Aging Cell 2018;17:e12751.

24. Cinti S. Anatomy and physiology of the nutritional system. Mol Aspects Med 2019;68:101-7.

25. Cypess AM, Lehman S, Williams G, et al. Identification and importance of brown adipose tissue in adult humans. N Engl J Med 2009;360:1509-17.

26. van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, et al. Cold-activated brown adipose tissue in healthy men. N Engl J Med 2009;360:1500-8.

27. Virtanen KA, Lidell ME, Orava J, et al. Functional brown adipose tissue in healthy adults. N Engl J Med 2009;360:1518-25.

28. Cinti S. The adipose organ at a glance. Dis Model Mech 2012;5:588-94.

29. Barbatelli G, Murano I, Madsen L, et al. The emergence of cold-induced brown adipocytes in mouse white fat depots is determined predominantly by white to brown adipocyte transdifferentiation. Am J Physiol Endocrinol Metab 2010;298:E1244-53.

30. Machado SA, Pasquarelli-do-Nascimento G, da Silva DS, et al. Browning of the white adipose tissue regulation: new insights into nutritional and metabolic relevance in health and diseases. Nutr Metab 2022;19:61.

31. Stanford KI, Middelbeek RJ, Townsend KL, et al. A novel role for subcutaneous adipose tissue in exercise-induced improvements in glucose homeostasis. Diabetes 2015;64:2002-14.

32. Barbera MJ, Schluter A, Pedraza N, Iglesias R, Villarroya F, Giralt M. Peroxisome proliferator-activated receptor alpha activates transcription of the brown fat uncoupling protein-1 gene. A link between regulation of the thermogenic and lipid oxidation pathways in the brown fat cell. J Biol Chem 2001;276:1486-93.

33. Villarroya F, Peyrou M, Giralt M. Transcriptional regulation of the uncoupling protein-1 gene. Biochimie 2017;134:86-92.

34. Bargut TCL, Souza-Mello V, Aguila MB, Mandarim-de-Lacerda CA. Browning of white adipose tissue: lessons from experimental models. Horm Mol Biol Clin Investig 2017;31.

35. Grujic D, Susulic VS, Harper ME, et al. Beta3-adrenergic receptors on white and brown adipocytes mediate beta3-selective agonist-induced effects on energy expenditure, insulin secretion, and food intake. A study using transgenic and gene knockout mice. J Biol Chem 1997;272:17686-93.

36. Lim S, Park J, Um JY. Ginsenoside Rb1 induces beta 3 adrenergic receptor-dependent lipolysis and thermogenesis in 3T3-L1 adipocytes and db/db mice. Front Pharmacol 2019;10:1154.

37. Evans BA, Merlin J, Bengtsson T, Hutchinson DS. Adrenoceptors in white, brown, and brite adipocytes. Br J Pharmacol 2019;176:2416-32.

38. Chernogubova E, Hutchinson DS, Nedergaard J, Bengtsson T. Alpha1- and beta1-adrenoceptor signaling fully compensates for beta3-adrenoceptor deficiency in brown adipocyte norepinephrine-stimulated glucose uptake. Endocrinology 2005;146:2271-84.

39. Bengtsson T, Cannon B, Nedergaard J. Differential adrenergic regulation of the gene expression of the β-adrenoceptor subtypes β1, β2 and β3 in brown adipocytes. Biochem J 2000;347:643-51.

40. Straat ME, Hoekx CA, van Velden FHP, et al. Stimulation of the beta-2-adrenergic receptor with salbutamol activates human brown adipose tissue. Cell Rep Med 2023;4:100942.

41. Li Y, Wang D, Ping X, et al. Local hyperthermia therapy induces browning of white fat and treats obesity. Cell 2022;185:949-66.e19.

42. Patsouris D, Qi P, Abdullahi A, et al. Burn induces browning of the subcutaneous white adipose tissue in mice and humans. Cell Rep 2015;13:1538-44.

43. Vinaik R, Barayan D, Abdullahi A, Jeschke MG. NLRP3 inflammasome mediates white adipose tissue browning after burn. Am J Physiol Endocrinol Metab 2019;317:E751-9.

44. Boström P, Wu J, Jedrychowski MP, et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 2012;481:463-8.

45. Roberts LD, Boström P, O’Sullivan JF, et al. β-Aminoisobutyric acid induces browning of white fat and hepatic β-oxidation and is inversely correlated with cardiometabolic risk factors. Cell Metab 2014;19:96-108.

46. Fabbiano S, Suárez-Zamorano N, Rigo D, et al. Caloric restriction leads to browning of white adipose tissue through type 2 immune signaling. Cell Metab 2016;24:434-46.

47. Li G, Xie C, Lu S, et al. Intermittent fasting promotes white adipose browning and decreases obesity by shaping the gut microbiota. Cell Metab 2017;26:672-85.e4.

48. Liu B, Page AJ, Hutchison AT, Wittert GA, Heilbronn LK. Intermittent fasting increases energy expenditure and promotes adipose tissue browning in mice. Nutrition 2019;66:38-43.

49. Suárez-Zamorano N, Fabbiano S, Chevalier C, et al. Microbiota depletion promotes browning of white adipose tissue and reduces obesity. Nat Med 2015;21:1497-501.

50. Cohen P, Levy JD, Zhang Y, et al. Ablation of PRDM16 and beige adipose causes metabolic dysfunction and a subcutaneous to visceral fat switch. Cell 2014;156:304-16.

51. Min SY, Kady J, Nam M, et al. Human ‘brite/beige’ adipocytes develop from capillary networks, and their implantation improves metabolic homeostasis in mice. Nat Med 2016;22:312-8.

52. Stanford KI, Middelbeek RJ, Townsend KL, et al. Brown adipose tissue regulates glucose homeostasis and insulin sensitivity. J Clin Invest 2013;123:215-23.

53. Ahmadian M, Abbott MJ, Tang T, et al. Desnutrin/ATGL is regulated by AMPK and is required for a brown adipose phenotype. Cell Metab 2011;13:739-48.

54. Gutierrez AD, Gao Z, Hamidi V, et al. Anti-diabetic effects of GLP1 analogs are mediated by thermogenic interleukin-6 signaling in adipocytes. Cell Rep Med 2022;3:100813.

55. Villarroya F, Cereijo R, Villarroya J, Giralt M. Brown adipose tissue as a secretory organ. Nat Rev Endocrinol 2017;13:26-35.

56. Yang FT, Stanford KI. Batokines: mediators of inter-tissue communication (a mini-review). Curr Obes Rep 2022;11:1-9.

57. Hondares E, Iglesias R, Giralt A, et al. Thermogenic activation induces FGF21 expression and release in brown adipose tissue. J Biol Chem 2011;286:12983-90.

58. Emanuelli B, Vienberg SG, Smyth G, et al. Interplay between FGF21 and insulin action in the liver regulates metabolism. J Clin Invest 2014;124:515-27.

59. He JL, Zhao M, Xia JJ, et al. FGF21 ameliorates the neurocontrol of blood pressure in the high fructose-drinking rats. Sci Rep 2016;6:29582.

60. Ruan CC, Kong LR, Chen XH, et al. A(2A) receptor activation attenuates hypertensive cardiac remodeling via promoting brown adipose tissue-derived FGF21. Cell Metab 2020;32:689.

61. Qing H, Desrouleaux R, Israni-Winger K, et al. Origin and function of stress-induced IL-6 in murine models. Cell 2020;182:372-87.e14.

62. Liu Y, Chen M. Neuregulin 4 as a novel adipokine in energy metabolism. Front Physiol 2022;13:1106380.

63. Gunawardana SC, Piston DW. Reversal of type 1 diabetes in mice by brown adipose tissue transplant. Diabetes 2012;61:674-82.

64. Villarroya J, Cereijo R, Villarroya F. An endocrine role for brown adipose tissue? Am J Physiol Endocrinol Metab 2013;305:E567-72.

65. Cereijo R, Gavaldà-Navarro A, Cairó M, et al. CXCL14, a brown adipokine that mediates brown-fat-to-macrophage communication in thermogenic adaptation. Cell Metab 2018;28:750-63.e6.

66. Pinckard KM, Shettigar VK, Wright KR, et al. A novel endocrine role for the BAT-released lipokine 12,13-diHOME to mediate cardiac function. Circulation 2021;143:145-59.

67. Lynes MD, Leiria LO, Lundh M, et al. The cold-induced lipokine 12,13-diHOME promotes fatty acid transport into brown adipose tissue. Nat Med 2017;23:631-7.

68. Stanford KI, Lynes MD, Takahashi H, et al. 12,13-diHOME: an exercise-induced lipokine that increases skeletal muscle fatty acid uptake. Cell Metab 2018;27:1111-20.e3.

69. Kong X, Yao T, Zhou P, et al. Brown adipose tissue controls skeletal muscle function via the secretion of myostatin. Cell Metab 2018;28:631-43.e3.

70. Campderrós L, Moure R, Cairó M, et al. Brown adipocytes secrete GDF15 in response to thermogenic activation. Obesity 2019;27:1606-16.

71. Thomou T, Mori MA, Dreyfuss JM, et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature 2017;542:450-5.

72. Vatner DE, Oydanich M, Zhang J, Campbell SC, Vatner SF. Exercise enhancement by RGS14 disruption is mediated by brown adipose tissue. Aging Cell 2023;22:e13791.

73. Aherne W, Hull D. Brown adipose tissue and heat production in the newborn infant. J Pathol Bacteriol 1966;91:223-34.

74. Yoneshiro T, Aita S, Matsushita M, et al. Brown adipose tissue, whole-body energy expenditure, and thermogenesis in healthy adult men. Obesity 2011;19:13-6.

75. Leitner BP, Huang S, Brychta RJ, et al. Mapping of human brown adipose tissue in lean and obese young men. Proc Natl Acad Sci USA 2017;114:8649-54.

76. Saito M, Okamatsu-Ogura Y, Matsushita M, et al. High incidence of metabolically active brown adipose tissue in healthy adult humans: effects of cold exposure and adiposity. Diabetes 2009;58:1526-31.

77. Matsushita M, Yoneshiro T, Aita S, Kameya T, Sugie H, Saito M. Impact of brown adipose tissue on body fatness and glucose metabolism in healthy humans. Int J Obes 2014;38:812-7.

78. Graja A, Schulz TJ. Mechanisms of aging-related impairment of brown adipocyte development and function. Gerontology 2015;61:211-7.

79. Berry DC, Jiang Y, Arpke RW, et al. Cellular aging contributes to failure of cold-induced beige adipocyte formation in old mice and humans. Cell Metab 2017;25:166-81.

80. Lee P, Swarbrick MM, Ho KK. Brown adipose tissue in adult humans: a metabolic renaissance. Endocr Rev 2013;34:413-38.

81. Félix-Soriano E, Sáinz N, Gil-Iturbe E, et al. Changes in brown adipose tissue lipid mediator signatures with aging, obesity, and DHA supplementation in female mice. FASEB J 2021;35:e21592.

82. Gonçalves LF, Machado TQ, Castro-Pinheiro C, de Souza NG, Oliveira KJ, Fernandes-Santos C. Ageing is associated with brown adipose tissue remodelling and loss of white fat browning in female C57BL/6 mice. Int J Exp Pathol 2017;98:100-8.

83. Sellayah D, Sikder D. Orexin restores aging-related brown adipose tissue dysfunction in male mice. Endocrinology 2014;155:485-501.

84. Darcy J, McFadden S, Fang Y, et al. Brown adipose tissue function is enhanced in long-lived, male ames dwarf mice. Endocrinology 2016;157:4744-53.

85. Brown-Borg HM, Borg KE, Meliska CJ, Bartke A. Dwarf mice and the ageing process. Nature 1996;384:33.

86. Li Y, Knapp JR, Kopchick JJ. Enlargement of interscapular brown adipose tissue in growth hormone antagonist transgenic and in growth hormone receptor gene-disrupted dwarf mice. Exp Biol Med 2003;228:207-15.

87. Ortega-Molina A, Efeyan A, Lopez-Guadamillas E, et al. Pten positively regulates brown adipose function, energy expenditure, and longevity. Cell Metab 2012;15:382-94.

88. Ma X, Xu L, Gavrilova O, Mueller E. Role of forkhead box protein A3 in age-associated metabolic decline. Proc Natl Acad Sci USA 2014;111:14289-94.

89. Saito M, Okamatsu-Ogura Y. Thermogenic brown fat in humans: implications in energy homeostasis, obesity and metabolic disorders. World J Mens Health 2023;41:489-507.

90. Yuko OO, Saito M. Brown fat as a regulator of systemic metabolism beyond thermogenesis. Diabetes Metab J 2021;45:840-52.

91. Becher T, Palanisamy S, Kramer DJ, et al. Brown adipose tissue is associated with cardiometabolic health. Nat Med 2021;27:58-65.

92. Pereira RO, McFarlane SI. The role of brown adipose tissue in cardiovascular disease protection: current evidence and future directions. Int J Clin Res Trials 2019;4:136.

93. Chen HJ, Meng T, Gao PJ, Ruan CC. The role of brown adipose tissue dysfunction in the development of cardiovascular disease. Front Endocrinol 2021;12:652246.

94. Tsoli M, Moore M, Burg D, et al. Activation of thermogenesis in brown adipose tissue and dysregulated lipid metabolism associated with cancer cachexia in mice. Cancer Res 2012;72:4372-82.

95. Cao Q, Hersl J, La H, et al. A pilot study of FDG PET/CT detects a link between brown adipose tissue and breast cancer. BMC Cancer 2014;14:126.

96. Huang YC, Chen TB, Hsu CC, et al. The relationship between brown adipose tissue activity and neoplastic status: an 18F-FDG PET/CT study in the tropics. Lipids Health Dis 2011;10:238.

97. Rousseau C, Bourbouloux E, Campion L, et al. Brown fat in breast cancer patients: analysis of serial 18F-FDG PET/CT scans. Eur J Nucl Med Mol Imaging 2006;33:785-91.

98. Singh R, Parveen M, Basgen JM, et al. Increased expression of beige/brown adipose markers from host and breast cancer cells influence xenograft formation in mice. Mol Cancer Res 2016;14:78-92.

99. Tayanloo-Beik A, Nikkhah A, Alaei S, et al. Brown adipose tissue and Alzheimer’S disease. Metab Brain Dis 2023;38:91-107.

100. O’Shaughnessy CT, Rothwell NJ, Shrewsbury-Gee J. Sympathetically mediated hypermetabolic response to cerebral ischemia in the rat. Can J Physiol Pharmacol 1990;68:1334-7.

101. Chao CM, Chen CL, Niu KC, et al. Hypobaric hypoxia preconditioning protects against hypothalamic neuron apoptosis in heat-exposed rats by reversing hypothalamic overexpression of matrix metalloproteinase-9 and ischemia. Int J Med Sci 2020;17:2622-34.

102. Gong B, Tang J, Jiang X, et al. In situ fluorescence-photoacoustic measurement of the changes of brown adipose tissue in mice under hindlimb unloading. J Appl Physiol 2023;135:251-9.

103. Tsuji T, Bussberg V, MacDonald AM, Narain NR, Kiebish MA, Tseng YH. Transplantation of brown adipose tissue with the ability of converting omega-6 to omega-3 polyunsaturated fatty acids counteracts high-fat-induced metabolic abnormalities in mice. Int J Mol Sci 2022;23:5321.

104. Dominici FP, Hauck S, Argentino DP, Bartke A, Turyn D. Increased insulin sensitivity and upregulation of insulin receptor, insulin receptor substrate (IRS)-1 and IRS-2 in liver of Ames dwarf mice. J Endocrinol 2002;173:81-94.

105. Bokov AF, Lindsey ML, Khodr C, Sabia MR, Richardson A. Long-lived ames dwarf mice are resistant to chemical stressors. J Gerontol A Biol Sci Med Sci 2009;64:819-27.

106. Ikeno Y, Bronson RT, Hubbard GB, Lee S, Bartke A. Delayed occurrence of fatal neoplastic diseases in ames dwarf mice: correlation to extended longevity. J Gerontol A Biol Sci Med Sci 2003;58:291-6.

107. Schrag M, Sharma S, Brown-Borg H, Ghribi O. Hippocampus of ames dwarf mice is resistant to beta-amyloid-induced tau hyperphosphorylation and changes in apoptosis-regulatory protein levels. Hippocampus 2008;18:239-44.

108. Puig KL, Kulas JA, Franklin W, et al. The ames dwarf mutation attenuates Alzheimer’s disease phenotype of APP/PS1 mice. Neurobiol Aging 2016;40:22-40.

109. Arum O, Rickman DJ, Kopchick JJ, Bartke A. The slow-aging growth hormone receptor/binding protein gene-disrupted (GHR-KO) mouse is protected from aging-resultant neuromusculoskeletal frailty. Age 2014;36:117-27.

110. Coschigano KT, Clemmons D, Bellush LL, Kopchick JJ. Assessment of growth parameters and life span of GHR/BP gene-disrupted mice. Endocrinology 2000;141:2608-13.

111. Ikeno Y, Hubbard GB, Lee S, et al. Reduced incidence and delayed occurrence of fatal neoplastic diseases in growth hormone receptor/binding protein knockout mice. J Gerontol A Biol Sci Med Sci 2009;64:522-9.

112. Lu S, Strand KA, Mutryn MF, et al. PTEN (Phosphatase and Tensin Homolog) Protects Against Ang II (Angiotensin II)-induced pathological vascular fibrosis and remodeling-brief report. Arterioscler Thromb Vasc Biol 2020;40:394-403.

113. Garcia-Cao I, Song MS, Hobbs RM, et al. Systemic elevation of PTEN induces a tumor-suppressive metabolic state. Cell 2012;149:49-62.

114. Knafo S, Sánchez-Puelles C, Palomer E, et al. PTEN recruitment controls synaptic and cognitive function in Alzheimer’s models. Nat Neurosci 2016;19:443-53.

115. Crackower MA, Oudit GY, Kozieradzki I, et al. Regulation of myocardial contractility and cell size by distinct PI3K-PTEN signaling pathways. Cell 2002;110:737-49.

116. Liang T, Gao F, Jiang J, et al. Loss of phosphatase and tensin homolog promotes cardiomyocyte proliferation and cardiac repair after myocardial infarction. Circulation 2020;142:2196-9.

117. Oydanich M, Zhang J, Vatner DE, Vatner SF. Two mechanisms mediating enhanced glucose tolerance with regulator of GS protein 14 disruption; increased exercise capacity and increased brown adipose tissue. Diabetes 2018;67:757-P.

118. Zhang J, Vatner DE, Vatner SF. Abstract 18750: increased brown adipose tissue as a novel mechanism mediating cardioprotection. Circulation 2016;134:A18750. Available from: https://www.ahajournals.org/doi/10.1161/circ.134.suppl_1.18750 [Last accessed on 24 Apr 2024].

119. Zhang J, Guers JJ, Oydanich M, Vatner DE, Vatner SF. Abstract P427: inhibition of the regulator of G protein signaling 14, a novel anti-hypertensive mechanism. Hypertension 2017;70:AP427.

120. Chen B, Yu J, Lu L, et al. Upregulated forkhead-box A3 elevates the expression of forkhead-box A1 and forkhead-box A2 to promote metastasis in esophageal cancer. Oncol Lett 2019;17:4351-60.

121. Darcy J, Tseng YH. ComBATing aging-does increased brown adipose tissue activity confer longevity? Geroscience 2019;41:285-96.

122. Peres Valgas da Silva C, Shettigar VK, Baer LA, et al. Brown adipose tissue prevents glucose intolerance and cardiac remodeling in high-fat-fed mice after a mild myocardial infarction. Int J Obes 2022;46:350-8.

123. Gomez-Hernandez A, Lopez-Pastor AR, Rubio-Longas C, et al. Specific knockout of p85alpha in brown adipose tissue induces resistance to high-fat diet-induced obesity and its metabolic complications in male mice. Mol Metab 2020;31:1-13.

124. Chen Z, Wang GX, Ma SL, et al. Nrg4 promotes fuel oxidation and a healthy adipokine profile to ameliorate diet-induced metabolic disorders. Mol Metab 2017;6:863-72.

125. Shi L, Li Y, Xu X, et al. Brown adipose tissue-derived Nrg4 alleviates endothelial inflammation and atherosclerosis in male mice. Nat Metab 2022;4:1573-90.

126. Wang GX, Zhao XY, Meng ZX, et al. The brown fat-enriched secreted factor Nrg4 preserves metabolic homeostasis through attenuation of hepatic lipogenesis. Nat Med 2014;20:1436-43.

127. Wang H, Wang L, Hu F, et al. Neuregulin-4 attenuates diabetic cardiomyopathy by regulating autophagy via the AMPK/mTOR signalling pathway. Cardiovasc Diabetol 2022;21:205.

128. Zhang P, Chen Z, Kuang H, et al. Neuregulin 4 suppresses NASH-HCC development by restraining tumor-prone liver microenvironment. Cell Metab 2022;34:1359-76.e7.

129. Mattson MP. Perspective: does brown fat protect against diseases of aging? Ageing Res Rev 2010;9:69-76.

130. Afshin A, Forouzanfar MH, Reitsma MB, et al. GBD 2015 Obesity Collaborators. Health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med 2017;377:13-27.

131. Nedergaard J, Bengtsson T, Cannon B. Unexpected evidence for active brown adipose tissue in adult humans. Am J Physiol Endocrinol Metab 2007;293:E444-52.

132. Feldmann HM, Golozoubova V, Cannon B, Nedergaard J. UCP1 ablation induces obesity and abolishes diet-induced thermogenesis in mice exempt from thermal stress by living at thermoneutrality. Cell Metab 2009;9:203-9.

133. Smith RE, Roberts JC. Thermogenesis of brown adipose tissue in cold-acclimated rats. Am J Physiol 1964;206:143-8.

134. Seale P, Kajimura S, Yang W, et al. Transcriptional control of brown fat determination by PRDM16. Cell Metab 2007;6:38-54.

135. Liu X, Wang S, You Y, et al. Brown adipose tissue transplantation reverses obesity in Ob/Ob mice. Endocrinology 2015;156:2461-9.

136. White JD, Dewal RS, Stanford KI. The beneficial effects of brown adipose tissue transplantation. Mol Aspects Med 2019;68:74-81.

137. Liu X, Zheng Z, Zhu X, et al. Brown adipose tissue transplantation improves whole-body energy metabolism. Cell Res 2013;23:851-4.

138. Zhu Z, Spicer EG, Gavini CK, Goudjo-Ako AJ, Novak CM, Shi H. Enhanced sympathetic activity in mice with brown adipose tissue transplantation (transBATation). Physiol Behav 2014;125:21-9.

139. Gunawardana SC, Piston DW. Insulin-independent reversal of type 1 diabetes in nonobese diabetic mice with brown adipose tissue transplant. Am J Physiol Endocrinol Metab 2015;308:E1043-55.

140. Darcy J, McFadden S, Fang Y, et al. Increased environmental temperature normalizes energy metabolism outputs between normal and ames dwarf mice. Aging 2018;10:2709-22.

141. Shankar K, Kumar D, Gupta S, et al. Role of brown adipose tissue in modulating adipose tissue inflammation and insulin resistance in high-fat diet fed mice. Eur J Pharmacol 2019;854:354-64.

142. Yamada Y, Wang XD, Yokoyama S, Fukuda N, Takakura N. Cardiac progenitor cells in brown adipose tissue repaired damaged myocardium. Biochem Biophys Res Commun 2006;342:662-70.

143. Wang H, Shi J, Wang Y, et al. Promotion of cardiac differentiation of brown adipose derived stem cells by chitosan hydrogel for repair after myocardial infarction. Biomaterials 2014;35:3986-98.

144. Martí-Pàmies Í, Thoonen R, Morley M, et al. Brown adipose tissue and BMP3b decrease injury in cardiac ischemia-reperfusion. Circ Res 2023;133:353-65.

145. Takx RA, Ishai A, Truong QA, MacNabb MH, Scherrer-Crosbie M, Tawakol A. Supraclavicular brown adipose tissue 18F-FDG uptake and cardiovascular disease. J Nucl Med 2016;57:1221-5.

146. Valero-Muñoz M, Li S, Wilson RM, et al. Heart failure with preserved ejection fraction induces beiging in adipose tissue. Circ Heart Fail 2016;9:e002724.

147. Tahara A, Tahara N, Maeda-Ogata S, et al. Brown adipose tissue activation in severe heart failure. Eur Heart J 2020;41:2415.

148. Yoshida Y, Shimizu I, Shimada A, et al. Brown adipose tissue dysfunction promotes heart failure via a trimethylamine N-oxide-dependent mechanism. Sci Rep 2022;12:14883.

149. Thoonen R, Ernande L, Cheng J, et al. Functional brown adipose tissue limits cardiomyocyte injury and adverse remodeling in catecholamine-induced cardiomyopathy. J Mol Cell Cardiol 2015;84:202-11.

150. Lowell BB, S-Susulic V, Hamann A, et al. Development of obesity in transgenic mice after genetic ablation of brown adipose tissue. Nature 1993;366:740-2.

151. Cittadini A, Mantzoros CS, Hampton TG, et al. Cardiovascular abnormalities in transgenic mice with reduced brown fat: an animal model of human obesity. Circulation 1999;100:2177-83.

152. Than A, Xu S, Li R, Leow MK, Sun L, Chen P. Angiotensin type 2 receptor activation promotes browning of white adipose tissue and brown adipogenesis. Signal Transduct Target Ther 2017;2:17022.

153. Ledent C, Vaugeois JM, Schiffmann SN, et al. Aggressiveness, hypoalgesia and high blood pressure in mice lacking the adenosine A2a receptor. Nature 1997;388:674-8.

154. Huang Cao ZF, Stoffel E, Cohen P. Role of perivascular adipose tissue in vascular physiology and pathology. Hypertension 2017;69:770-7.

155. Kim HW, Belin de Chantemèle EJ, Weintraub NL. Perivascular adipocytes in vascular disease. Arterioscler Thromb Vasc Biol 2019;39:2220-7.

156. Lu C, Su LY, Lee RM, Gao YJ. Alterations in perivascular adipose tissue structure and function in hypertension. Eur J Pharmacol 2011;656:68-73.

157. Kong LR, Zhou YP, Chen DR, Ruan CC, Gao PJ. Decrease of perivascular adipose tissue browning is associated with vascular dysfunction in spontaneous hypertensive rats during aging. Front Physiol 2018;9:400.

158. Persson P, Marchetti M, Friederich-Persson M. Browning of perivascular adipose tissue prevents vascular dysfunction and reduces hypertension in angiotensin II-infused mice. Am J Physiol Regul Integr Comp Physiol 2023;325:R290-8.

159. Das E, Moon JH, Lee JH, Thakkar N, Pausova Z, Sung HK. Adipose tissue and modulation of hypertension. Curr Hypertens Rep 2018;20:96.

160. Yang SJ, Hong HC, Choi HY, et al. Effects of a three-month combined exercise programme on fibroblast growth factor 21 and fetuin-A levels and arterial stiffness in obese women. Clin Endocrinol 2011;75:464-9.

161. Semba RD, Crasto C, Strait J, Sun K, Schaumberg DA, Ferrucci L. Elevated serum fibroblast growth factor 21 is associated with hypertension in community-dwelling adults. J Hum Hypertens 2013;27:397-9.

162. Walsh MF, Barazi M, Pete G, Muniyappa R, Dunbar JC, Sowers JR. Insulin-like growth factor I diminishes in vivo and in vitro vascular contractility: role of vascular nitric oxide. Endocrinology 1996;137:1798-803.

163. Marczin N, Papapetropoulos A, Catravas JD. Tyrosine kinase inhibitors suppress endotoxin- and IL-1 beta-induced NO synthesis in aortic smooth muscle cells. Am J Physiol 1993;265:H1014-8.

164. Friederich-Persson M, Nguyen Dinh Cat A, Persson P, Montezano AC, Touyz RM. Brown adipose tissue regulates small artery function through NADPH oxidase 4-derived hydrogen peroxide and redox-sensitive protein kinase G-1α. Arterioscler Thromb Vasc Biol 2017;37:455-65.

165. Xue Y, Petrovic N, Cao R, et al. Hypoxia-independent angiogenesis in adipose tissues during cold acclimation. Cell Metab 2009;9:99-109.

166. Xue Y, Xu X, Zhang XQ, Farokhzad OC, Langer R. Preventing diet-induced obesity in mice by adipose tissue transformation and angiogenesis using targeted nanoparticles. Proc Natl Acad Sci USA 2016;113:5552-7.

167. Sun K, Kusminski CM, Luby-Phelps K, et al. Brown adipose tissue derived VEGF-a modulates cold tolerance and energy expenditure. Mol Metab 2014;3:474-83.

168. Bagchi M, Kim LA, Boucher J, Walshe TE, Kahn CR, D’Amore PA. Vascular endothelial growth factor is important for brown adipose tissue development and maintenance. FASEB J 2013;27:3257-71.

169. Zhang Q, Liang Z, Zhang Y, et al. Brown adipose tissue transplantation improves skin fibrosis in localized scleroderma. FASEB J 2023;37:e23315.

170. Chen L, Wang L, Li Y, et al. Transplantation of Normal adipose tissue improves blood flow and reduces inflammation in high fat fed mice with hindlimb ischemia. Front Physiol 2018;9:197.

171. Seki T, Hosaka K, Fischer C, et al. Ablation of endothelial VEGFR1 improves metabolic dysfunction by inducing adipose tissue browning. J Exp Med 2018;215:611-26.

172. Tupone D, Madden CJ, Morrison SF. Autonomic regulation of brown adipose tissue thermogenesis in health and disease: potential clinical applications for altering BAT thermogenesis. Front Neurosci 2014;8:14.

173. Kopelman PG. Obesity as a medical problem. Nature 2000;404:635-43.

174. Han JC, Lawlor DA, Kimm SY. Childhood obesity. Lancet 2010;375:1737-48.

175. Martin AR, Chung S, Koehler K. Is exercise a match for cold exposure? Common molecular framework for adipose tissue browning. Int J Sports Med 2020;41:427-42.

176. Roubenoff R. Sarcopenia and its implications for the elderly. Eur J Clin Nutr 2000;54 Suppl 3:S40-7.

177. Gonzalez-Freire M, de Cabo R, Bernier M, et al. Reconsidering the role of mitochondria in aging. J Gerontol A Biol Sci Med Sci 2015;70:1334-42.

178. Joseph AM, Adhihetty PJ, Buford TW, et al. The impact of aging on mitochondrial function and biogenesis pathways in skeletal muscle of sedentary high- and low-functioning elderly individuals. Aging Cell 2012;11:801-9.

179. Short KR, Bigelow ML, Kahl J, et al. Decline in skeletal muscle mitochondrial function with aging in humans. Proc Natl Acad Sci USA 2005;102:5618-23.

180. Coen PM, Jubrias SA, Distefano G, et al. Skeletal muscle mitochondrial energetics are associated with maximal aerobic capacity and walking speed in older adults. J Gerontol A Biol Sci Med Sci 2013;68:447-55.

181. Conley KE, Jubrias SA, Cress ME, Esselman P. Exercise efficiency is reduced by mitochondrial uncoupling in the elderly. Exp Physiol 2013;98:768-77.

182. Garatachea N, Pareja-Galeano H, Sanchis-Gomar F, et al. Exercise attenuates the major hallmarks of aging. Rejuvenation Res 2015;18:57-89.

183. Liu CJ, Latham NK. Progressive resistance strength training for improving physical function in older adults. Cochrane Database Syst Rev 2009;2009:CD002759.

184. Serra-Rexach JA, Bustamante-Ara N, Hierro Villarán M, et al. Short-term, light- to moderate-intensity exercise training improves leg muscle strength in the oldest old: a randomized controlled trial. J Am Geriatr Soc 2011;59:594-602.

185. Sullivan DH, Roberson PK, Smith ES, Price JA, Bopp MM. Effects of muscle strength training and megestrol acetate on strength, muscle mass, and function in frail older people. J Am Geriatr Soc 2007;55:20-8.

186. Ellis T, Motl RW. Physical activity behavior change in persons with neurologic disorders: overview and examples from Parkinson disease and multiple sclerosis. J Neurol Phys Ther 2013;37:85-90.

187. Meeusen R. Exercise, nutrition and the brain. Sports Med 2014;44 Suppl 1:S47-56.

188. Kajimura S, Spiegelman BM, Seale P. Brown and beige fat: physiological roles beyond heat generation. Cell Metab 2015;22:546-59.

189. Oh-ishi S, Kizaki T, Toshinai K, et al. Swimming training improves brown-adipose-tissue activity in young and old mice. Mech Ageing Dev 1996;89:67-78.

190. Yoshioka K, Yoshida T, Wakabayashi Y, Nishioka H, Kondo M. Effects of exercise training on brown adipose tissue thermogenesis in ovariectomized obese rats. Endocrinol Jpn 1989;36:403-8.

191. Slusher AL, Whitehurst M, Zoeller RF, Mock JT, Maharaj M, Huang CJ. Attenuated fibroblast growth factor 21 response to acute aerobic exercise in obese individuals. Nutr Metab Cardiovasc Dis 2015;25:839-45.

192. Wu MV, Bikopoulos G, Hung S, Ceddia RB. Thermogenic capacity is antagonistically regulated in classical brown and white subcutaneous fat depots by high fat diet and endurance training in rats: impact on whole-body energy expenditure. J Biol Chem 2014;289:34129-40.

193. De Matteis R, Lucertini F, Guescini M, et al. Exercise as a new physiological stimulus for brown adipose tissue activity. Nutr Metab Cardiovasc Dis 2013;23:582-90.

194. Tanaka R, Fuse S, Kuroiwa M, et al. Vigorous-intensity physical activities are associated with high brown adipose tissue density in humans. Int J Environ Res Public Health 2020;17:2796.

195. Motiani P, Virtanen KA, Motiani KK, et al. Decreased insulin-stimulated brown adipose tissue glucose uptake after short-term exercise training in healthy middle-aged men. Diabetes Obes Metab 2017;19:1379-88.

196. Singhal V, Maffazioli GD, Ackerman KE, et al. Effect of chronic athletic activity on brown fat in young women. PLoS One 2016;11:e0156353.

197. Vosselman MJ, Hoeks J, Brans B, et al. Low brown adipose tissue activity in endurance-trained compared with lean sedentary men. Int J Obes 2015;39:1696-702.

198. Sebaa R, Johnson J, Pileggi C, et al. SIRT3 controls brown fat thermogenesis by deacetylation regulation of pathways upstream of UCP1. Mol Metab 2019;25:35-49.

199. Cheng A, Yang Y, Zhou Y, et al. Mitochondrial SIRT3 mediates adaptive responses of neurons to exercise and metabolic and excitatory challenges. Cell Metab 2016;23:128-42.

200. Qiu X, Brown K, Hirschey MD, Verdin E, Chen D. Calorie restriction reduces oxidative stress by SIRT3-mediated SOD2 activation. Cell Metab 2010;12:662-7.

201. Cho SY, Chung YS, Yoon HK, Roh HT. Impact of exercise intensity on systemic oxidative stress, inflammatory responses, and sirtuin levels in healthy male volunteers. Int J Environ Res Public Health 2022;19:11292.

202. Zhou L, Pinho R, Gu Y, Radak Z. The role of SIRT3 in exercise and aging. Cells 2022;11:2596.

203. Gao P, Jiang Y, Wu H, et al. Inhibition of mitochondrial calcium overload by SIRT3 prevents obesity- or age-related whitening of brown adipose tissue. Diabetes 2020;69:165-80.

204. Vellano CP, Brown NE, Blumer JB, Hepler JR. Assembly and function of the regulator of G protein signaling 14 (RGS14)·H-Ras signaling complex in live cells are regulated by Gαi1 and Gαi-linked G protein-coupled receptors. J Biol Chem 2013;288:3620-31.

205. Deng Y, Larrivée B, Zhuang ZW, et al. Endothelial RAF1/ERK activation regulates arterial morphogenesis. Blood 2013;121:3988-96.

206. Chim SM, Kuek V, Chow ST, et al. EGFL7 is expressed in bone microenvironment and promotes angiogenesis via ERK, STAT3, and integrin signaling cascades. J Cell Physiol 2015;230:82-94.

207. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209-49.

208. Petruzzelli M, Schweiger M, Schreiber R, et al. A switch from white to brown fat increases energy expenditure in cancer-associated cachexia. Cell Metab 2014;20:433-47.

209. Alnabulsi A, Cash B, Hu Y, Silina L, Alnabulsi A, Murray GI. The expression of brown fat-associated proteins in colorectal cancer and the relationship of uncoupling protein 1 with prognosis. Int J Cancer 2019;145:1138-47.

210. Seki T, Yang Y, Sun X, et al. Brown-fat-mediated tumour suppression by cold-altered global metabolism. Nature 2022;608:421-8.

211. Liu D, Li Y, Shang Y, Wang W, Chen SZ. Effect of brown adipose tissue/cells on the growth of mouse hepatocellular carcinoma in vitro and in vivo. Oncol Lett 2019;17:3203-10.

212. Lee P, Greenfield JR, Ho KK, Fulham MJ. A critical appraisal of the prevalence and metabolic significance of brown adipose tissue in adult humans. Am J Physiol Endocrinol Metab 2010;299:E601-6.

213. Lim S, Hosaka K, Nakamura M, Cao Y. Co-option of pre-existing vascular beds in adipose tissue controls tumor growth rates and angiogenesis. Oncotarget 2016;7:38282-91.

214. Shellock FG, Riedinger MS, Fishbein MC. Brown adipose tissue in cancer patients: possible cause of cancer-induced cachexia. J Cancer Res Clin Oncol 1986;111:82-5.

215. Baracos VE, Martin L, Korc M, Guttridge DC, Fearon KCH. Cancer-associated cachexia. Nat Rev Dis Primers 2018;4:17105.

216. Dolly A, Dumas JF, Servais S. Cancer cachexia and skeletal muscle atrophy in clinical studies: what do we really know? J Cachexia Sarcopenia Muscle 2020;11:1413-28.

217. Kir S, Spiegelman BM. Cachexia & brown fat: a burning issue in cancer. Trends Cancer 2016;2:461-3.

218. Dong M, Lin J, Lim W, Jin W, Lee HJ. Role of brown adipose tissue in metabolic syndrome, aging, and cancer cachexia. Front Med 2018;12:130-8.

219. Becker AS, Zellweger C, Bacanovic S, et al. Brown fat does not cause cachexia in cancer patients: a large retrospective longitudinal FDG-PET/CT cohort study. PLoS One 2020;15:e0239990.

220. Eljalby M, Huang X, Becher T, et al. Brown adipose tissue is not associated with cachexia or increased mortality in a retrospective study of patients with cancer. Am J Physiol Endocrinol Metab 2023;324:E144-53.

221. Coleman RA, Liang C, Patel R, Ali S, Mukherjee J. Brain and brown adipose tissue metabolism in transgenic Tg2576 mice models of alzheimer disease assessed using 18F-FDG PET imaging. Mol Imaging 2017;16:1536012117704557.

222. Crews L, Masliah E. Molecular mechanisms of neurodegeneration in Alzheimer’s disease. Hum Mol Genet 2010;19:R12-20.

223. Iqbal K, Grundke-Iqbal I. Neurofibrillary pathology leads to synaptic loss and not the other way around in Alzheimer disease. J Alzheimers Dis 2002;4:235-8.

224. Mandelkow EM, Mandelkow E. Tau in Alzheimer’s disease. Trends Cell Biol 1998;8:425-7.

225. Almeida MC, Carrettiero DC. Chapter 44 - hypothermia as a risk factor for Alzheimer disease. Handb Clin Neurol 2018;157:727-35.

226. Pražienková V, Schirmer C, Holubová M, et al. Lipidized prolactin-releasing peptide agonist attenuates hypothermia-induced tau hyperphosphorylation in neurons. J Alzheimers Dis 2019;67:1187-200.

227. Sa-Nguanmoo P, Tanajak P, Kerdphoo S, et al. FGF21 improves cognition by restored synaptic plasticity, dendritic spine density, brain mitochondrial function and cell apoptosis in obese-insulin resistant male rats. Horm Behav 2016;85:86-95.

228. Choi HM, Doss HM, Kim KS. Multifaceted physiological roles of adiponectin in inflammation and diseases. Int J Mol Sci 2020;21:1219.

229. Forny-Germano L, De Felice FG, Vieira MNDN. The role of leptin and adiponectin in obesity-associated cognitive decline and Alzheimer’s disease. Front Neurosci 2018;12:1027.

230. Kshirsagar V, Thingore C, Juvekar A. Insulin resistance: a connecting link between Alzheimer’s disease and metabolic disorder. Metab Brain Dis 2021;36:67-83.

231. Sędzikowska A, Szablewski L. Insulin and insulin resistance in Alzheimer’s disease. Int J Mol Sci 2021;22:9987.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Zhang J, Kibret BG, Vatner DE, Vatner SF. The role of brown adipose tissue in mediating healthful longevity. J Cardiovasc Aging 2024;4:17. http://dx.doi.org/10.20517/jca.2024.01

AMA Style

Zhang J, Kibret BG, Vatner DE, Vatner SF. The role of brown adipose tissue in mediating healthful longevity. The Journal of Cardiovascular Aging. 2024; 4(2): 17. http://dx.doi.org/10.20517/jca.2024.01

Chicago/Turabian Style

Zhang, Jie, Berhanu Geresu Kibret, Dorothy E. Vatner, Stephen F. Vatner. 2024. "The role of brown adipose tissue in mediating healthful longevity" The Journal of Cardiovascular Aging. 4, no.2: 17. http://dx.doi.org/10.20517/jca.2024.01

ACS Style

Zhang, J.; Kibret BG.; Vatner DE.; Vatner SF. The role of brown adipose tissue in mediating healthful longevity. J. Cardiovasc. Aging. 2024, 4, 17. http://dx.doi.org/10.20517/jca.2024.01

About This Article

© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
139
Downloads
12
Citations
0
Comments
0
3

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 1 clicks
Like This Article 3 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
The Journal of Cardiovascular Aging

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/