Abstract
The epidemic of overweight and obesity underlies many common metabolic diseases. Approaches aimed to reduce energy intake and/or stimulate energy expenditure represent potential strategies to control weight gain. Adipose tissue is a major energy balancing organ. It can be classified as white adipose tissue (WAT) and brown adipose tissue (BAT). While WAT stores excess metabolic energy, BAT dissipates it as heat via adaptive thermogenesis. WAT also participates in thermogenesis by providing thermogenic fuels and by directly generating heat after browning. Browned WAT resembles BAT morphologically and metabolically and is classified as beige fat. Like BAT, beige fat can produce heat. Human adults have BAT-like or beige fat. Recruitment and activation of this fat type have the potential to increase energy expenditure, thereby countering against obesity and its metabolic complications. Given this, agents capable of inducing WAT browning have recently attracted broad attention from biomedical, nutritional and pharmaceutical societies. In this review, we summarize natural bioactive compounds that have been shown to promote beige adipocyte recruitment and activation in animals and cultured cells. We also discuss potential molecular mechanisms for each compound to induce adipose browning and metabolic benefits.
Keywords: adipose browning, food supplements, obesity, thermogenesis
INTRODUCTION
Obesity is a major public health burden in developed societies. Currently it affects more than two billion people (1). Metabolic complications of obesity include, but not limited to, dyslipidemia, insulin resistance, type 2 diabetes, fatty liver, cardiovascular disease, and cancer (2). Overall, obesity is a result of increased energy intake and/or reduced energy expenditure. While agents reducing energy intake and nutrient absorption or food intake may reduce weight gain (3), those enhancing energy dissipation may also be beneficial. A major pathway in dissipating metabolic energy is the nonshivering adaptive thermogenesis (4). In rodents, anatomically identifiable brown adipose tissue (BAT) depots are responsible for heat production through the adaptive thermogenesis. Human infants also possess visible BAT in the interscapular region. However, this BAT is lost during growth. Over a long time, scientists believed that adult humans did not have BAT. This belief was overturned in 2009 when three independent groups reported the existence of brown fat-like adipose tissues in human adults, particularly in those after cold exposure (5–7). These milestone human studies revitalized research in brown fat biology and the role of BAT thermogenesis in whole-body energy balance. BAT is composed of brown adipocytes and many other cell types. Brown adipocytes are responsible for heat production in the BAT, though other cell types play important regulatory roles. A classical brown adipocyte is characterized by multilocular lipid droplets, abundant mitochondria, and expression of uncoupling protein-1 (UCP-1). UCP-1 protein localizes at the inner mitochondrial membrane and short-circuits the mitochondrial proton gradient to produce heat via oxidative phosphorylation of metabolic fuels such as fatty acids (8). It has long been known that multilocular lipid droplet-containing adipocytes exist in the classical white fat pads in humans and rodents (9). These cells are now known as brite or beige adipocytes because they express UCP-1 and are thermogenic (10). Importantly, they can be induced or recruited to the white adipose tissue (WAT) in response to environmental and pharmacological stimuli such as cold exposure and adrenergic activators (9–11). The process of inducing beige adipocyte appearance in the WAT is called WAT browning (12). Animal and human studies have shown that increased WAT browning is associated with resistance to diet-induced obesity and metabolic disorders, and with leanness in humans (13). Therefore, targeting thermogenic adipose tissues may counter obesity and its associated metabolic sequelae.
Adipose browning is regulated by a complex interplay of endogenous and environmental factors (14, 15). A hallmark of white fat browning is the expression and activation of UCP-1, though UCP-1-independent adipose browning mechanisms also exist (16–19). UCP-1 expression is regulated predominantly at the transcriptional level (4). Peroxisome proliferator-activated receptor γ (PPARγ) coactivator-1α (PGC-1α), a master regulator of mitochondrial biogenesis and oxidative metabolism, can induce the expression of UCP-1 and other thermogenic genes in adipocytes (13, 20). It does so by complexing with PPARγ and a transcriptional coregulator PR domain zinc finger protein 16 (PRDM16) (21, 22). PRDM16 is essential in maintaining brown fat characteristics and its deficiency induces expression of muscle genes in brown preadipocytes and brown fat pads (22). PRDM16 can interact with multiple DNA-binding transcriptional factors, including PPARα, PPARγ, and several CCAAT/enhancer-binding protein family members, and this interaction is capable of inducing PGC-1α expression (23, 24). In addition to PRDM16, PGC-1α, and PPARs, other signaling pathways also induce UCP-1 expression and adipose browning. For example, BAT activation is often associated with increased phosphorylation of 5’-AMP-activated protein kinase (AMPK) under in vitro and in vivo conditions (25, 26), and prolonged treatment with the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide enhances WAT browning in mice (27). The crosstalk between the intracellular Ca2+-activated calcium/calmodulin-dependent protein kinase II-AMPK signaling and Sirtuin-1 (SIRT1), a sensor of cellular metabolism and energy utilization, mediates adipose browning downstream of capsaicin-induced activation of transient receptor potential cation channel subfamily V member 1 (TRPV1) (28). Fibroblast growth factor 21 (FGF21) is capable of inducing white fat browning (29), though not essential for nonshivering thermogenesis (30). Other growth factors, such as several bone morphogenetic proteins (BMPs), also promote brown phenotypes in adipose tissues (31–35). Pharmacological approaches targeting the aforementioned pathways induce adipose browning (28, 36–40). Nonetheless, PRDM16 expression and UCP-1 induction are most consistent with the presence of beige/brite adipocytes within white fat pads (41, 42), highlighting important roles of PRDM16 and other UCP-1-inducing factors in driving thermogenic or browning phenotypes in adipose tissues.
Natural plant-derived compounds have been the backbone of many synthetic drugs that are used today. Approximately 50% of approved drugs in the last 3 decades are directly or indirectly from natural compounds (43). Emerging evidence suggests that some food compounds, phytochemicals in particular, may be promising candidates for obesity management, a proportion of which may counter obesity by promoting adipose browning (44). Several excellent review articles have summarized some bioactive nutritional compounds that are capable of increasing energy expenditure through brown and white fat thermogenesis, such as those by Concha et al. (45), Kang et al. (46), and Zhang et al. (47). Each of these reviews was focused on certain categories of nutritional agents or bioactive compounds that activate BAT and/or WAT. In this review, we provide an update to include more natural compounds, such as polyphenols, terpenoids, and alkaloids. We focus on those capable of inducing white adipose browning in cultured adipocytes and whole animals, discuss whether the compounds’ browning effect is linked to their metabolic benefits, and highlight the potential mechanisms by which the compounds induce adipose browning. The review also identifies current challenges and opportunities for translational research to evaluate the role of phytochemicals in adipose browning and metabolic health.
NATURAL BIOACTIVE COMPOUNDS INVOLVED IN ADIPOSE BROWNING
Polyphenols
Resveratrol
Resveratrol (3, 5, 4′ trihydroxystilbene) (RSV) is a polyphenolic compound enriched in Polygonum cuspidatum. A small amount of RSV is also found in red wine, peanuts, grapes, berries, red cabbage, and spinach (48, 49). In the past 15 years or so, RSV is one of the most widely studied polyphenols due to its metabolic and health benefits, including antioxidative, anti-inflammatory, and anti-cancer effects (50–55). Studies have shown that RSV induces metabolic benefits by targeting multiple pathways, including adipogenesis, fatty acid oxidation, lipolysis, and apoptosis in adipocytes (50, 51, 56–59).
It has been shown that RSV protects rodents against high fat-induced obesity and metabolic abnormalities by promoting mitochondrial oxidative capacity of BAT, skeletal muscle and liver via the AMPK-SIRT1-PGC-1α pathway (50, 51, 60). In 2012, Park and associates reported that RSV attenuates aging-related metabolic disorders by inhibiting cAMP-degrading phosphodiesterases (PDEs) and activating the cAMP effector Epac1 (61). This study identified RSV as a nonselective PDE inhibitor and provided a unifying upstream mechanism underlying the activation of the AMPK-SIRT1-PGC-1α pathway by RSV. A few recent studies suggest that RSV supplementation may reduce weight gain and improve metabolic health by inducing energy expenditure through activation of thermogenesis and WAT browning (57, 62, 63). In a study when 5-month-old CD1 female mice were challenged for 4 weeks with a high fat diet (HFD) supplemented with or without 0.1% RSV, the mice treated with RSV versus those without RSV treatment display reduced weight gain and increased browning of the inguinal subcutaneous WAT (iWAT) as evidenced by increased multilocular lipid droplets-containing adipocytes, UCP-1 expression, and fatty acid oxidation (57). This study further showed that RSV at 10 μM increases mRNA levels of genes related to adipocyte browning, including PRDM16, UCP-1, Cidea, ELOVL Fatty Acid Elongase 3 (Elovl3), PGC-1α, CD137, T-Box Transcription Factor 1 (Tbx1), and transmembrane protein 26 (TMEM26) in differentiated stromal vascular cells (SVCs) isolated from the iWAT of mice (57) (Table I). This effect was shown to be entirely AMPKα-dependent, demonstrating a critical role of AMPK in mediating RSV-induced adipose browning (57), which is consistent with an early study showing that RSV does not exert metabolic benefits in AMPK-α1 or α2-deficient mice (52) and that RSV can activate AMPK (50). In another study, Hui et al. reported that type 2 diabetic male db/db mice fed a chow diet supplemented with 0.4% RSV for 10 weeks exhibit improved glucose homeostasis, which is associated with enhanced BAT activation and WAT browning (62) (Table I). In this study, the mice treated with RSV were found to have altered plasma and fecal bile acid compositions that were enriched of lithocholic acid (LCA). LCA is a potent endogenous ligand for Takeda G protein coupled receptor 5, a membrane-bound bile acid receptor known to mediate bile acid-induced activation of BAT thermogenesis (64, 65). Bile acids are produced in the liver and subject to extensive modifications by the gut microflora (66–68). Likewise, RSV is metabolized in the liver and gut (69). In the gut, intestinal bacteria are involved in the metabolism of RSV (70). Therefore, RSV has the potential to alter the gut microbiome. Indeed, studies have shown that RSV treatments alter the gut microbiota and promote adipose browning that depends, at least in part, on the gut microbiome in db/db or high fat-fed mice (62, 63) (Table I). RSV not only improves metabolic health in adult rodents. A recent study showed that the supplement of RSV together with nicotinamide riboside in early postnatal life attenuates fat-induced metabolic abnormalities and promotes WAT browning in adult male, but not female mice (71).
Table I.
Dietary compounds shown to stimulate WAT browning in animals
Bioactive Compounds |
Animals & treatments (dosage, duration) | Effects on WAT gene expression | Metabolic phenotypes | Ref |
---|---|---|---|---|
Polyphenols | ||||
RSV | HFD-fed CD1 female mice (0.1% in diet, 4 wks) | ↑ UCP-1, PRDM16, Cidea, Elovl3, PGC-1α, CD137, Tbx1, and TMEM26 in SVCs of iWAT |
|
Wang et al. (57) |
db/db mice (0.4% in chow diet, 10 wks) | ↑ UCP-1, Cidea, PGC-1α, PPARα, PPARγ, and PRDM16 in iWAT |
|
Hui et al. (62) | |
HFD-fed C57BL/6 mice (0.4% in diet or received feces from HFD-RVS-fed mice, 4 wks) | ↑ UCP-1, PPARγ, PGC-1α, and SIRT1 in eWAT and iWAT |
|
Liao et al. (63) | |
HFD-fed FVB/N mice (400mg/kg/day in diet, 8 wks) | ↑ UCP-1, PRDM16, PGC-1α, and SIRT1 in subcutaneous WAT |
|
Andrade et al. (75) | |
Quercetin | HFD-fed C57BL/6 J mice (0.1 % in diet, 12 wks) | ↑ UCP-1 and Elovl3 in iWAT |
|
Kuipers et al. (85) |
HFD-fed C57BL/6 mice (0.5% in diet, 8 wks) | ↑ UCP-1, PRDM16, Cidea, and PGC-1α in subcutaneous WAT and retroperitoneal WAT |
|
Lee et al. (86) | |
GTC | HFD-fed SD rats (100 mg/kg/day, oral gavage, 30 or 45 days) | ↑ UCP-1 (visceral WAT and subcutaneous WAT) |
|
Yan et al. (91) |
Epicatechin | HFD-fed Wistar rats (1 mg/kg daily, 2 wks) | ↑ UCP-1, PGC-1α, SIRT1, and Dio2 in abdominal WAT |
|
Gutiérrez-Salmeán et al. (92) |
Curcumin | C57BL/6 mice (50 mg/kg/day, oral gavage, 50 days) | ↑ UCP-1, PGC-1α, PRDM16, Dio2, PPARα, Cidea, Elovl3, Nrf1, mtTfα, and ATPsyn in iWAT |
|
Wang et al. (98) |
ArtC | C57BL/6 J mice (10 mg/kg in diet, 4 wks) | ↑ UCP-1 and PRDM16 in iWAT |
|
Nishikawa et al. (106) |
Rutin | HFD-fed C57BL/6 J mice or db/db mice (1 mg/ml in drinking water, 10 wks) | ↑ UCP-1, PGC-1α, PGC-1β, CPT1α, PPARα, CD137, and TBX in iWAT |
|
Yuan et al. (112) |
Luteolin | HFD-fed C57BL/6 mice (0.01% in diet, 12 wks) | ↑ UCP-1, PGC-1α, Elovl3, SIRT1, and Cited1 in iWAT |
|
Zhang et al. (118) |
Myricetin | db/db mice (400 mg/kg/day, oral gavage, 14 wks) | ↑ SIRT1 and PGC-1α in iWAT |
|
Hu et al. (127) |
Sudachitin | HFD-fed C57BL/6 J mice (5 mg/kg/day, oral gavage, 12 wks) | ↑ UCP-1 and UCP-3 in iWAT |
|
Tsutsumi et al. (129) |
C3G | db/db mice (1 mg/ml in drinking water, 16 wks) | ↑ UCP-1, Cidea, PGC-1α, PGC-1β, CPT1α, PPARα, TMTM26, CD 137, and Tbx1 in subcutaneous WAT |
|
You et al. (135) |
Terpenoids | ||||
Phytol | C57BL/6 J mice (500 mg/kg by gavage every other day, 7 wks) | ↑ UCP-1, PRDM16, PGC-1α, PDH, and Cyto C in iWAT |
|
Zhang et al. (141) |
Fucoxanthin | Wistar rats (2% in diet, 4 wks) | ↑ UCP-1 in abdominal WAT |
|
Maeda et al. (147) |
Gypenoside | HFD-fed C57BL/6 J mice (100 or 300 mg/kg in diet, 12 wks) | ↑ UCP-1, PGC-1α, PRDM16, AMPKα, Cidea, Cox7α, and Elovl3 in iWAT |
|
Liu et al. (155) |
Menthol | HFD-fed C57BL/6 mice (1 % in diet, 12 wks) | ↑ UCP-1, PGC-1α, PRDM16, β3-AR, and TRPM8 in iWAT and eWAT |
|
Jiang et al. (166) |
Celastrol | HFD-fed C57BL/6 mice (3 mg/kg/day in diet, 3 wks) | ↑ PRDM16, Cidea, Elovl3, TMEM26, CD137, SP100, Tbx1, Scl27a1, and Hsp70 in iWAT |
|
Ma et al. (172) |
HFD-fed C57BL/6 mice (100 mg/kg, i.p., 6 days) | ↑ UCP-1 in iWAT |
|
Pfuhlmann et al. (173) | |
Alkaloids | ||||
Capsaicin | HFD-fed TRPV1 KO and control mice (0.01% in diet, 12 wks) | ↑ UCP-1, PPARγ, BMP8β, PGC-1α, SIRT1/PPARγ/PRDM16 in iWAT and eWAT of WT mice |
|
Baskaran et al. (28) |
LACA mice (2 mg/kg, oral gavage, alternate days, 12 wks) | ↑ UCP-1, PGC-1α, BDNF and Cideain iWAT |
|
Baboota et al. (182) | |
Berberine | C57BL/6 mice (5 mg/kg/day, i.p., 4 wks) | ↑ UCP-1, Dio2, and PRDM16 in WAT |
|
Sun et al. (188) |
db/db mice (5 mg/kg/day, i.p., 4 wks) | ↑ UCP-1, PGC-1α, Isdp5, Cidea, and COX8b in iWAT |
|
Zhang et al. (193) |
This study suggests that early use of RSV may exert long-term metabolic benefits by programming the body to a different metabolic state epigenetically. Indeed, the same group reported that RSV or nicotinamide treatment modifies methylation marks in PRDM16 and Slc27α1, two genes related to WAT browning, in the iWAT of mice (72). They also showed that RSV and nicotinamide directly induce the DNA methylation machinery to display more browning features in 3 T3-L1 adipocytes. Recently, Liu et al. (73) reported that RSV enhances browning of white adipocytes in vitro possibly by mediating the activation of mammalian target of rapamycin (mTOR). They showed that brown adipocyte-specific markers are decreased after treatment with rapamycin, an inhibitor of mTOR, in 3 T3-L1 adipocytes. On the other hand, treatment with MHY1485, an activator of mTOR, recapitulates the effect of RSV on browning markers in these cells. Another study suggest that RSV may alter lipid and oxidative metabolism in a SIRT1-dependent mechanism in 3 T3-L1 adipocytes (74). In this study, RSV treatment reduces triglyceride (TG) accumulation in 3 T3-L1 cells, and this reduction is abolished by inhibiting SIRT1 and PGC-1α. The study also showed that RSV treatment induces expression of PPARγ, PGC-1α, and carnitine palmitoyltransferase-1α (CPT-1α) in 3 T3-L1 adipocytes, all of which are abolished by SIRT1 inhibition (74). Interestingly, in a study evaluating the effect of RSV on expression of genes involved in thermogenesis, Andrade et al. showed that the HFD-fed male FVB/N mice treated with RSV (400 mg/kg/day) for 8 weeks, or obese human volunteers treated with trans-RSV (500 mg/day) for 4 weeks, displayed improved glycemic and lipid profiles along with increased mRNA levels of UCP1, PRDM16, PGC1α, SIRT1 and FNDC5 in the subcutaneous adipose tissue (75).
Quercetin
Quercetin (3,3′,4′,5,7-pentahydroxyflavone) is found in onions, apples, broccoli, and berries, and is one of the major flavonoids present in human diets (76). It has been reported that quercetin improves dyslipidemia and insulin sensitivity, reduces hypertension and BW gain, and at a high dose, lowers adipose and aortic inflammation in Zucker rats (77). In the HFD-fed mice, quercetin also improves insulin sensitivity and reduces BW gain and adipose inflammation (78, 79). Dong et al. showed quercetin attenuates mast cell and macrophage infiltration into epididymal WAT (eWAT) through the AMPKα-SIRT1 pathway in the HFD-fed mice (79). Kobori et al. (78) found that quercetin suppresses adipose infiltration of macrophages and lymphocytes in mice fed a Western diet high in fat. Obesity is often associated with a state of low-grade chronic adipose inflammation, which facilitates the development of insulin resistance and type 2 diabetes (80–83). Quercetin-induced improvement in insulin sensitivity may be linked to its suppression of inflammation in addition to the reduced weight gain.
Quercetin does not seem to reduce HFD-induced weight gain in mice by reducing food intake (79, 84, 85). Increased energy expenditure may play a part because quercetin-treated mice show increased UCP-1 expression in the BAT (79) as well as elevated mitochondrial oxidative phosphorylation in the WAT (78). Consistently, quercetin and quercetin-rich red onion extract have been demonstrated to increase energy expenditure in mice fed a HFD (84). In this study, authors further showed that both quercetin and the quercetin-rich red onion extract improve skeletal muscle mitochondrial number and function, though only quercetin but not the red onion extract increases physical activity. A couple of recent studies have shown that quercetin induces expression of genes related to adipose browning in the WAT of HFD-fed mice (Table I), though not affecting weight gain and energy expenditure (85, 86). Quercetin-induced adipose browning increases adipose uptake of TG-derived fatty acids, which may explain why quercetin lowers blood TG levels (85). In differentiated 3 T3-L1 adipocytes, quercetin at 100 μM increases expression of brown adipocyte-specific genes and CPT1α (Table II), which is associated with decreased lipid accumulation and increased multilocular lipid droplets in these adipocytes (86). The quercetin-induced browning of 3 T3-L1 adipocytes may be mediated in part by the activation of AMPK since treating these cells with 5 μM dorsomorphin, an AMPK inhibitor, attenuates quercetin-induced increases in mRNA levels of PRMD16, PGC-1α, CPT1α (a downstream target of PGC-1α), and SIRT1, though not UCP-1 (86) (Table II).
Table II.
Dietary compounds shown to induce browning in cultured adipocytes
Bioactive Compounds |
Cells (treatment dosage) | Effects on browning-related genes | Ref. |
---|---|---|---|
Polyphenols | |||
RSV | 3T3-L1 adipocyte (20 μM) | ↑ UCP1, PPARγ, and PGC-1α | Liu et al. (73) |
3T3-L1 adipocyte (50 μM) | ↑ SIRT1, PPARγ, CPT1α, and PGC-1α | Imamura et al. (74) | |
Quercetin | 3T3-L1 adipocyte (100 μM) | ↑ CPT1α, SIRT1, and PGC-1α | Lee et al. (86) |
Curcumin | Primary white adipocytes (2 μM) | ↑ UCP-1, PPARα, and PGC-1α | Song et al. (100) |
3T3-L1 and primary white adipocytes (20 μM) | ↑ UCP-1, PRDM16, and PGC-1α | Lone et al. (101) | |
ArtC | C3H10T1/2 cells (10 μM) | ↑ UCP-1, Cidea, Cox8b, Elovl3, PPARγ, and PRDM16 | Nishikawa et al. (106) |
Rutin | C3H10T1/2 cells (10 μM) | ↑ UCP-1, PRDM16, and PGC-1α | Yuan et al. (112) |
Myricetin | C3H10T1/2 cells (10 μM) | ↑ UCP-1 and PRDM16 | Hu et al. (127) |
Genistein | 3T3-L1 adipocyte (100 μM) | ↑ UCP-1 and CD137 | Aziz et al. (214) |
Butein | C3H10T1/2 cells (20 μM) | ↑ UCP-1 and PRDM4 | Song et al. (215) |
Chrysin | 3T3-L1 adipocyte (50 μM) | ↑ UCP-1, Cidea, FGF21, PGC-1α, PRDM16, Tbx1, TMEM26, etc. | Choi et al. (216) |
C3G | 3T3-L1 adipocyte (100 μM) | ↑ UCP-1, Tbx1, and CITED1 | Matsukawa et al. (217) |
Terpenoids | |||
Phytol | 3T3-L1 adipocyte (100 μM) | ↑ UCP-1, PRDM16, PGC-1α, Cidea, CD137, TMEM26, etc. | Zhang et al. (141) |
Ginsenoside Rb1 | 3T3-L1 adipocyte (10 μM) | ↑ UCP-1, PGC-1α, and PRDM16 | Mu et al. (156) |
Thymol | 3T3-L1 adipocyte (20 μM) | ↑ UCP-1, PPARγ, PPARδ, pAMPK, pACC, HSL, CPT1α, PGC-1α, etc. | Choi et al. (218) |
Alkaloids | |||
Capsaicin | 3T3-L1 adipocyte (1 μM) | ↑ UCP-1, PGC-1α, SIRT1, PRDM16, Dio2, Cidea, and PPARα | Baboota et al. (182) |
It is currently unknown why quercetin reduces BW gain in obese rodents in some but not all studies, despite similar doses of quercetin are used. This discrepancy may be addressed by using a standardized treatment regimen under identical experimental conditions including animal housing environment and diet sources.
Green Tea Catechins
Green tea catechins (GTC) are polyphenolic compounds present in the unfermented dried leaves of the plant Camellia sinensis. Epigallocatechin-3-gallate is the most abundant catechin of green tea, representing 50–80% of the total catechin content, and also considered to be the most bioactive component of green tea (87). Other minor catechins include epicatechin-3-gallate, epigallocatechin, epicatechin, and catechin (87). Green tea has anti-inflammatory, anti-atherosclerotic, and anti-cancer properties (88). During recent years, a growing number of studies have confirmed the beneficial effects of green tea on obesity, glucose homeostasis, and fat mass gain in HFD-induced obese rodent models (89, 90).
A potential mechanism by which GTCs induce anti-obesity effects may relate to GTCs’ effects on energy metabolism and adipose browning. For example, oral gavage of GTC at 100 mg/kg/day for 30 or 45 days significantly reduces BW gain, liver weight, and blood TG levels in male Sprague-Dawley rats fed a HFD (91). The authors suggested that GTCs exert their anti-obesity effects, in part, by modulating PPAR signaling pathways. They found that GTCs increase PPARγ protein levels in the subcutaneous WAT, decrease PPARγ protein levels in the visceral WAT, and increase PPARδ protein levels in both BAT and WAT along with increased mRNA expression of genes involved in fatty acid oxidation such as PPARδ target genes including alternative oxidase, CPT1α, and UCP-1 in the BAT. The expression of UCP-1 mRNA is also increased in the subcutaneous WAT and visceral WAT in the chow or HFD-fed rats treated with GTCs, suggesting enhanced browning of these adipose tissues upon GTC treatment (Table I). In another study, (−)-Epicatechin (EPI), a component of green tea catechins (92), seems to increase adipose and skeletal muscle expression of UCP-1 and iodothyronine deiodinase 2 (Dio2), which are two adipose browning markers (Table I). In this study, male Wistar rats were fed a HFD for 5 weeks, followed by a 2-week EPI administration by gavage (1 mg/kg/day) under the same dietary condition. It was found that EPI treatment reduces BW gain, hyperglycemia, and blood TG levels without affecting food intake or nutrient absorption. Because protein expression levels of UCP-1, PGC-1α, SIRT1, and Dio2 are increased in the abdominal adipose tissue, the authors suggested that increased energy expenditure may have contributed to the decreased weight gain in the EPI-treated animals (Table I). A concern for this study is the basal level of UCP-1, which is not normally expressed at a significant level in the WAT and skeletal muscle. Authors detected abundant UCP-1 protein expression in these tissues under the basal condition. It is unclear if this is specific to male Wistar rats and/or the experimental environment.
In accordance with animal studies, Nirengi et al. (93) found that ingestion of a catechin-rich beverage at 540 mg catechin/day for 12 weeks significantly increased BAT density (indicative of BAT activation) and decreased extramyocellular lipid content in healthy young women. It would be interesting to determine whether this drink promotes white fat browning.
Curcumin
Curcumin, a bioactive polyphenol component derived from the turmeric rhizomes, has been investigated in prevention of obesity, insulin resistance, diabetes and dyslipidemia in mice, rabbits and rats (94–96). Curcumin may exert its metabolic benefits by regulating lipid metabolism, increasing antioxidant enzyme expression, reducing inflammatory cytokine production, and activating fatty acid oxidation (97).
Wang et al. (98) showed that curcumin administration at 50 or 100 mg/kg/day for 50 days reduces BW gain and fat mass without affecting food intake in male C57BL/6 mice fed a chow diet. These fat and BW gain changes are associated with higher body temperatures during 6 h of cold exposure (98). Consistent with body temperature changes, the animals display increased expression of many brown fat-specific genes in the iWAT, including mRNA and protein for UCP-1 and mRNAs for PGC-1α, PRDM16, Dio2, PPARα, Cidea, Elovl3, nuclear respiratory factor 1, mitochondrial transcription factor A (Tfam), and ATP synthase (Table I). The study further showed that curcumin-treated mice have elevated blood norepinephrine (NE) concentrations and increased mRNA for β−3 adrenergic receptor (β3-AR) in the iWAT. This led the authors to suggest that curcumin induces adipose browning via the NE-β3-AR pathway. Typically, it is the locally released NE from the sympathetic nerves innervating adipose tissues, not the circulating NE, that serves as the major mediator of adipose thermogenesis during cold exposure (4, 99). Increased circulating NE levels after curcumin treatment imply a role of curcumin in stimulating NE release from adrenal glands. It would be interesting to examine whether some of the cardio-metabolic effects of curcumin are mediated by adrenal NE and epinephrine. Another study by Song et al. (100) reported that male C57BL/6 mice fed a HFD containing 1% curcumin for 18 weeks have increased energy expenditure and body temperature in response to cold exposure, which is associated with increased expression of UCP-1, PRDM16 and PPARα in the BAT but not WAT. In rat primary adipocytes, these researchers observed that the increased mRNA expression of UCP-1 cannot be completely blocked by a PPARα or PPARγ antagonist, suggesting that curcumin may increase energy expenditure partially via PPARα/PPARγ-independent mechanisms (Table II). In 3 T3-L1 and primary white adipocytes, Lone et al. (101) observed that curcumin induces brown fat phenotypes via the AMPK pathway. Curcumin treatments (1, 10, or 20 μM) increase expression of UCP-1 and other brown adipocyte-specific markers (PGC-1α and PPARγ) in a dose-dependent manner in these cells. Inhibition of AMPK by dorsomorphin abolishes expression of UCP-1, PRDM16 and PGC-1α while the AMPK activator AICAR does the opposite (Table II).
Artepillin C
Artepillin C (3,5-diprenyl-4-hydroxycinnamic acid) (ArtC) is a major component of Brazilian green propolis that is collected from the plant Baccharis dracunculiforia (102). Several studies have documented potential roles of ArtC in oxidative stress prevention, immunoregulation, chemoprevention, and cancer treatment (103–105). Nishikawa et al. (106) showed that ArtC treatment for 4 weeks at 10 mg/kg/day induces appearance of brown-like adipocytes and expression of UCP-1 and PRDM16 proteins in the iWAT of male C57BL/6 J mice (Table I). Unlike curcumin, ArtC-induced browning of WAT is not associated with significant changes in plasma NE concentrations and adipose β3-AR mRNA levels (106). ArtC seems to promote adipose browning in a cell autonomous manner because ArtC at 10 μM induces expression of mRNAs for UCP-1, Cidea, cytochrome c oxidase subunit 8B (Cox8b), and Elovl3 in murine C3H10T1/2 cells and primary iWAT-derived adipocytes (Table II), and this induction largely depends on PPARγ activation and PRDM16 protein stability.
Rutin
Rutin (3,3,4,5,7-pentahydroxyflavone-3-rhamnoglucoside) is a polyphenolic bioflavonoid from various plants and fruits, such as buckwheat, apricots, oranges, lemons, grapes, limes, berries, and peaches (107, 108). Pharmacological studies have shown that rutin has antioxidative, anti-inflammatory, and antidiabetic properties, suggesting that rutin may have therapeutic potentials in many diseases (109–111). Yuan et al. (112) reported that in both genetically obese db/db male mice and the HFD-fed C57BL/6 J male mice, rutin treatment in the drinking water (1 mg/ml) for 10 weeks attenuates weight gain and adiposity, increases energy expenditure, and improves glucose homeostasis. This occurs without changes in food intake, water intake, and physical activity. Interestingly, rutin-treated animals versus controls display a significantly higher mean core body temperature during cold (4°C) exposure for 4 h. The expression of mRNAs for several BAT-specific genes, including UCP-1, PGC-1α, PGC-1β, CPT1α, and PPARα, as well as beige cell markers, such as CD137 and TBX, are significantly increased in the subcutaneous WAT after rutin treatment (Table I). In this study, rutin (10 μM) significantly increases expression of UCP-1 and other thermogenic genes, such as PRDM16 and PGC-1α in C3H10T1/2 cells (Table II). Mechanistic studies suggest that rutin directly binds and stabilizes SIRT1, leading to hypoacetylation of PGC-1α protein and Tfam transactivation. Activation of this SIRT1-PGC-1α-Tfam pathway in heat-generating adipocytes may contribute to rutin-induced adipose browning and energy expenditure.
Luteolin
Luteolin (3′,4′,5,7-tetrahydroxyflavone) is a flavone present in fruits, vegetables, and several medicinal herbs such as peppermint, thyme, and parsley (113). Luteolin has antioxidant, anti-inflammatory, antimicrobial, and anticancer effects (113). Previous animal studies have shown that luteolin protects against high glucose-induced endothelial dysfunction as well as diet-induced hepatic steatosis and insulin resistance (114–117). Zhang et al. (118) found that male C57BL/6 mice fed a HFD supplemented with 0.01% luteolin for 12 weeks are protected against HFD-induced weight gain, body fat accumulation, and insulin resistance. These animals show no changes in energy intake and body temperature, but increases in energy expenditure, respiratory exchange ratio, and expression of thermogenic genes in BAT and subcutaneous WAT tissues (118) (Table I). Compared to the mice fed the HFD only, the luteolin-treated mice have more brown-like adipocytes and UCP-1 expression in the subcutaneous WAT. In addition, the authors observed that luteolin treatment significantly increases expression of many thermogenic genes in differentiated primary brown and subcutaneous adipocytes, which is associated with increased phosphorylation of both AMPKα and acetyl-CoA carboxylase. The AMPK inhibitor Compound C (20 μM) can reverse the effects of luteolin on thermogenic gene expression, suggesting that AMPK may mediate some of luteolin’s metabolic benefits.
Myricetin
Myricetin (3,5,7,3′,4′,5′-hexahydroxyflavone cannabiscetin) is a natural flavonoid found in wine, onions, bayberries, grapes, and tea, among others (119, 120). It has been reported that myricetin possesses various pharmacological actions, including antioxidative, anti-inflammatory, and anticancer activities (121–123). In rats, myricetin can improve glucose utilization, increase insulin sensitivity, and reduce hyperglycemia associated with diet feeding and diabetes (124–126). Similar effects were also observed in mice. Hu et al. (127) reported that diabetic db/db mice orally gavaged with myricetin at 400 mg/kg/day in distilled water for 14 weeks starting at 4 weeks of age slows down weight and body fat gain and improves plasma lipid profiles and insulin sensitivity. This is associated with increased thermogenic protein expression and mitochondrial biogenesis in both BAT and iWAT, resulting in augmented energy expenditure and cold resistance (Table I). During differentiation of murine C3H10T1/2 mesenchymal stem cells to brown adipocytes, myricetin treatment at 10 μM increases mRNA expression and secretion of adiponectin (127) (Table II). Myricetin treatment also increases adiponectin mRNA levels in BAT, eWAT, and iWAT as well as plasma concentrations of adiponectin in the db/db mice. This finding led the authors to suggest that myricetin may exert its metabolic benefits by directly regulating adiponectin expression in adipocytes.
Sudachitin
Sudachitin (5,7,4′-trihydroxy-6,8,3-trimethoxyflavone) is a polymethoxyflavone found in the peel of Citrus sudachin. In addition to its anti-inflammatory effect (128), sudachitin has been shown to possess anti-obesity action (129). Using male C57BL/6 J mice fed a HFD and diabetic db/db mice fed a normal diet, Tsutsumi et al. showed that sudachitin treatment at 5 mg/kg/day for 12 weeks prevents HFD-induced weight gain, body fat deposition and adipocyte hypertrophy without altering food intake. (129). In addition, sudachitin treatment improves insulin sensitivity in the HFD-fed C57BL/6 J mice and in the chow-fed db/db mice. Interestingly, sudachitin supplementation to the HFD significantly increases UCP-1 mRNA levels in the subcutaneous WAT in addition to increasing mitochondrial biogenesis and fatty acid oxidation in the skeletal muscle, implying that sudachitin may protect overnutrition-associated metabolic disorders in part by promoting WAT browning (Table I).
Cyanidin-3-O-β-Glucoside
Cyanidin-3-O-β-glucoside (C21H21O11+) (C3G) is a component in blueberries and one of the most widely distributed anthocyanins in fruits and vegetables (130, 131). C3G seems to have potent anti-obesity and anti-diabetes properties (132–134). You et al. (135) reported that obese male db/db mice treated with C3G in drinking water (1 mg/mL) for 16 weeks display reduced BW gain, hepatic steatosis, serum lipids, and insulin resistance. The animals also have increased energy expenditure, cold tolerance, and BAT activity. These effects of C3G are not associated with significant differences in food or water intake, energy absorption, or physical activity, but with enhanced white fat browning as evidenced by increased expression of genes related to thermogenesis, fatty acid oxidation and beigeing, including UCP-1, Cidea, PGC-1α, PGC-1β, CPT1α, PPARα, TMEM26, CD137, and Tbx1, in the subcutaneous WAT (Table I). C3G treatment also increases mitochondrial number and function in both BAT and subcutaneous WAT, suggest that C3G may induce adipose browning by promoting mitochondrial biogenesis.
Terpenoids
Phytol
Phytol (3,7,11,15-tetramethylhexaded-2-en-1-ol) is a plastidial isoprenoid found in plant foods such as fruits, vegetables, and grains. It is abundant in nature as a part of the chlorophyll molecule (136, 137). Animal studies suggest that phytol may have anti-inflammatory and anti-oxidative effects (138). A study reported that phytol alters fatty acid metabolism and decreases plasma fatty acid concentrations by activating retinoid-X-receptor and PPARs in mice (139, 140). Zhang et al. (141) reported that 7 weeks of phytol treatment at 500 mg/kg every other day by oral gavage reduces weight gain without affecting the food intake in male C57BL/6 J mice fed a HFD. This treatment induces iWAT browning as evidenced by increased multilocular adipocytes and thermogenic gene expression in this fat depot (Table I). The results from in vitro cell culture studies also support the role of phytol in stimulating adipocyte browning. Phytol at 100 μM stimulates browning or the formation of brown-like adipocytes in the differentiated 3 T3-L1 cells, which is associated with activation of UCP-1 gene promoter activity and upregulated expression of brown and beige adipocyte marker genes, including PRDM16, PGC-1α, Cidea, Elovl3, CD137 and TMEM26 (141) (Table II). In this study, phytol was also shown to activate AMPKα in adipose tissues and cultured 3 T3-L1 adipocytes. Using the AMPK inhibitor Compound C (100 μM), the authors demonstrated that the AMPKα signaling pathway is required for phytol to stimulate brown adipogenic differentiation.
Fucoxanthin
Fucoxanthin (C42H58O6) is a marine carotenoid. It can be found in edible brown seaweeds, such as Undaria pinnatifida (Wakame), Saccharina japonica (Makonbu), and Sargassum fulvellum (Hondawara) (142). Fucoxanthin has antioxidant, anti-inflammatory, and anticancer activities (143, 144). It has attracted wide attention because of its anti-obesity and anti-diabetic effects (145, 146). Maeda et al. (147) observed that male Wistar rats and obese female KK-Ay mice fed 2% Undaria lipids have reduced weight gain and fat weight, but no changes in daily food intake compared to the control diets. KK-Ay mice fed the diet containing 2% Undaria lipids show increased BAT weight than those on the control diets. Interestingly, fucoxanthin-containing diets significantly increase UCP-1 protein and mRNA expression in the WAT composed of perirenal, gonadal, retroperitoneal and mesenteric adipose tissues, though not in the BAT (147) (Table I). It would be interesting to determine fat depot-specific effect of fucoxanthin given the distinct function of each fat depot in energy metabolism. Nonetheless, this finding suggests that WAT browning may play a role in the anti-obesity activity of seaweed carotenoid fucoxanthin in obese mice.
In contrast with the animal studies, Rebello et al. (148) reported that human adipocytes treated with fucoxanthin or fucoxanthinol at different concentrations (1 μM, 0.1 μM, 0.01 μM, 1 nM, 0.1 nM) showed no changes in the oxygen consumption rate (OCR) or expression levels of mRNAs for UCP1, CPT-1β, and GLUT4. In addition, no significant changes were observed for the mRNA levels of PGC-1α, PPARα, PPARγ, PDK4, FAS, and the lipolytic enzymes. The authors concluded that neither fucoxanthin nor its metabolite fucoxanthinol stimulates human white adipocyte browning at the dose that is expected to exist in the human plasma without being toxic to the cells. Further studies are needed to address the discrepancy in the role of fucoxanthin in WAT browning between animals and humans.
Gypenosides
Gypenosides (C47H76O18) are triterpenoid saponins isolated from the Gynostemma pentaphyllum plant. It may have potential benefits in inflammation, cardiovascular diseases, cancer, diabetes, and obesity-associated metabolic disorders (149–154). Liu et al. (155) showed that treatment with gypenosides at 300 mg/kg BW/day for 12 weeks reduces weight gain, fat weight, and insulin resistance in the HFD-fed male C57BL/6 J mice. These effects are associated with increased protein and mRNA expression of UCP-1, PGC-1α, and PRDM16 as well as increased mRNAs for Cidea, Cox7α, and Elovl3 in the BAT and iWAT (Table I), suggesting an adipose browning-inducing role of gypenosides in mice. In addition, this study showed that gypenosides substantially alter the gut microbiome composition with a significant increase in Akkermansia muciniphila (A. muciniphila). It was proposed that gypenoside may exert metabolic benefits in part by stimulating adipose browning and regulating the gut microbiota. Mu et al. (156) reported that ginsenoside Rb1 (10 μM) induces brown-specific genes partially through induction of PPARγ in 3 T3-L1 mature adipocytes and this effect can be inhibited by the PPARγ antagonist GW9662, suggesting that ginsenoside Rb1 may induce adipose browning at least in part by activating PPARγ (Table II).
Menthol
Menthol (C10H20O) is rich in cornmint or peppermint oil. It is added to foods, mouthwashes, toothpastes, drugs, among others as a cooling and flavoring agent. The cooling effect of menthol is attributable to its action as an agonist of transient receptor potential melastatin 8 (TRPM8), an ion channel that detects cold stimuli in the thermosensory system (157–159). Mice deficient in the menthol receptor TRPM8 display severely attenuated sensation of cold temperature (158, 160–162). Recent studies suggest that menthol may be used as an anti-obesity compound due to its thermogenic property (163, 164). Ma et al. (163) observed that menthol increases UCP-1 levels in murine primary brown adipocytes in a TRPM8-dependent manner and critically dependent on the Ca2+-dependent PKA phosphorylation. They also observed that dietary supplementation of menthol (0.5%, w/w) for 28 weeks increases the locomotor activities and core temperatures in the HFD-fed wild-type, but not TRPM8-deficient mice (163). Rossato et al. (165) showed that activation of TRPM8 by menthol dose-dependently increases UCP-1 expression in human white adipocytes, leading to increased heat production, though no changes in expression of the genes regulating mitochondrial biogenesis (165). In addition, Jiang et al. (166) showed that menthol (100 μM) increases expression of thermogenic genes such as UCP-1 and PGC-1α in cultured mouse white adipocytes, which can be inhibited by BAPTA-AM (10 μM), a membrane-permeable calcium chelator, or KT5720 (10 μM), a specific protein kinase A (PKA) inhibitor. This study suggests that menthol may induce UCP-1 and PGC-1α expression by increasing intracellular Ca2+ concentration and PKA phosphorylation in white adipocytes. Consistently, dietary menthol treatment at 1% (w/w) for 12 weeks attenuates HFD-induced weight gain in male C57BL/6 mice (166). The reduction in weight gain is accompanied by enhanced browning and reduced adipocyte size in the subcutaneous WAT, and by improved glucose tolerance and insulin sensitivity (166). Expression levels of thermogenic genes (UCP-1, PGC-1α, PRDM16, β3-AR) and TRPM8 are significantly increased in the subcutaneous WAT and eWAT of these menthol-treated mice (Table I). The authors concluded that TRPM8 may be involved in menthol-induced WAT browning by increasing expression of genes related to thermogenesis and energy metabolism (166).
Celastrol
Celastrol (C29H38O4) is a major active component in Chinese herbal medicine Tripterygium wilfordii (‘Thunder of God Vine’) (167, 168). It has diverse biological properties, such as anti-cancer, anti-obesity, anti-diabetic, and neuroprotective (169). Celastrol’s potent anti-obesity effect has been reproduced in several studies, though the underlying mechanism has yet to be settled (170–173). One study showed that celastrol suppresses food intake and increases energy expenditure by improving leptin sensitivity, leading up to 45% weight loss in hyperleptinemic diet-induced obese mice (170). Another study showed that celastrol treatment at 3 mg/kg/day for 3 weeks reduces weight gain, fat mass, hepatic steatosis and insulin resistance without affecting food intake, locomotor activity, or respiratory exchange ratio (RER) in HFD-fed mice (172). In this study, celastrol treatment increases brown (PRDM16, UCP-1, Cidea, and Elovl3) and beige (TMEM26, CD137, SP100, Tbx1, Scl27a1, and Hsp70) selective genes in the iWAT (Table I). The induction of these thermogenic genes requires heat shock factor 1 (HSF1) and PGC-1α because it is blunted in cells derived from the iWAT of HSF1-or PGC-1α-null mice (172). However, Pfuhlmann et al. (173) showed that celastrol decreases BW independently from UCP-1 and has no effect on energy expenditure. They observed that celastrol treatment at 100 mg/kg/day for 6 days increases expression of UCP-1 in the BAT and iWAT of HFD-fed mice (Table I), but transcription of PGC-1α and key thermogenic genes (e.g., PRDM16 and Cidea) is unaffected in these tissues, and there are comparable BW loss and hypophagia between UCP-1 KO and WT mice treated with celastrol. This observation implies that UCP-1-mediated thermogenesis is not a major driver for the BW-lowering effect of celastrol. Although celastrol-induced weight loss may not depend on UCP-1, one cannot exclude its dependence on thermogenic activation because UCP1-independent thermogenic programs exist in adipose and other tissues (18, 19, 174, 175). Celastrol was shown to activate HFS1 and PGC-1α-dependent thermogenic program (172). Nur77 was identified as a critical intracellular target for celastrol, and the binding of celastrol to Nur77 promotes Nur7 translocation from the nucleus to mitochondria, helping clearance of inflamed mitochondria to suppress inflammation (171). These studies indicate that celastrol may have a profound impact on mitochondrial function and dynamics, thereby critically influencing energy metabolism.
Alkaloids
Capsaicin
Capsaicin (C18H27NO3) is an alkaloid compound found in pepper, which belongs to the genus Capsicum (176). It has antioxidant and anti-cancer properties (177, 178). In addition, capsaicin has been shown to have anti-obesity effects by mechanisms that include promotion of adipose lipolysis and energy expenditure (179, 180). Oral administration of capsaicin at 2 mg/kg/day was shown to alter gut microbial compositions, increase hypothalamic satiety, and induce adipose browning in male Swiss albino mice fed a HFD (181). The browning effect is supported by increased expression of genes related to thermogenesis and mitochondrial biogenesis including UCP-1, PGC-1α, PPARα, Cidea, and brain-derived neurotrophic factor (BDNF) in the BAT and subcutaneous WAT (Table I). Because the pair feeding was used in the study, the authors suggested that the induction of adipose browning may contribute in part to the reduced weight gain in the capsaicin-treated animals. In differentiated 3 T3-L1 adipocytes, Baboota et al. (182) reported that low dose (1 μM) capsaicin induces browning-specific gene expression, an effect similar to that induced by resiniferatoxin (RTX), an agonist of TRPV1. The genes induced include PPARγ and its downstream targets as well as brown fat cell markers UCP-1, PGC-1α, PRDM16, Dio2, PPARα, and FOXC2 (Table II). The study also showed that expression of UCP-1, PGC-1α, BDNF, and Cidea are significantly upregulated in the subcutaneous WAT of male Wistar rats upon oral administration of capsaicin at 2 mg/kg BW on alternate days for 3 months (Table I). Baskaran et al. (28) analyzed the effects of dietary capsaicin (0.01%) on the HFD-induced obesity in male WT and TRPV1 KO mice. They found that dietary capsaicin suppresses HFD-induced weight gain in WT, but not TRPV1 KO mice by stimulating heat production and physical activity (28). Expression of PGC-1α, SIRT1, and PRDM16 are increased in the eWAT and iWAT of WT but not TRPV1 KO mice (Table I). In addition, there is an induction of UCP-1 in the eWAT and an increase of UCP-1 and BMP8β in the iWAT in the capsaicin-treated animals. The authors suggested that capsaicin may stimulate SIRT1-dependent deacetylation of PPARγ and PRDM16, thereby facilitating PPARγ and PRDM16 interaction to induce adipose browning via a TRPV1-dependent mechanism.
Berberine
Berberine (C20H18NO4+), a naturally occurring alkaloid, is found in certain species of flowering plants like Berberidaccae, Coptis rhizomes and Hydrastis Canadensis that are used in traditional Chinese medicine (183). Many studies have shown that berberine has a variety of benefits in health, such as anti-inflammatory, anti-cancer, hypolipidemic, hypoglycemic, and insulin-sensitizing effects (184, 185). Multiple mechanisms may have been implicated in berberine’s health and metabolic benefits, such as activation of AMPK and SIRT1 (186–188), up-regulation of LDL receptor (189), and modulation of gut hormones and microbiota (190–192).
Studies have suggested a role of berberine in adipose browning. Sun et al. (188) reported that the beneficial effects of berberine (5 mg/kg/day, i.p., 4 weeks) on weight gain and hepatic steatosis in high-fat high-fructose diet-fed C57BL/6 mice require hepatic SIRT1 because liver-specific SIRT1 deletion abolishes these benefits. Berberine treatment increases FGF21 production from liver and primary hepatocytes and induces thermogenic gene expression in white and brown adipose tissues in mice (188). These effects also require SIRT1 and are associated with increased whole-body energy expenditure. Consistently, Zhang et al. (193) reported that male db/db mice treated with berberine (5 mg/kg/day, i.p.) have increased energy expenditure, reduced weight gain, and improved insulin sensitivity without changes in physical activity. The expression of UCP-1 and other classical BAT marker genes, such as Cidea and cytochrome c oxidase subunit 8b, are induced in the BAT and iWAT in these mice. Cell culture studies suggest that berberine upregulates UCP-1 transcription by activating the AMPK-PGC-1α pathway (193). In addition to activating AMPK/PGC-1α, long-term treatment with berberine increases expression of tyrosine hydroxylase in both BAT and iWAT in WT and db/db mice, suggesting that berberine may augment the sympathetic outflow to adipose tissues, which may contribute in part to berberine’s adipose browning effect. Interestingly, berberine has been shown to promote recruitment and activation of brown fat-like tissues in mildly overweight subjects with non-alcoholic fatty liver disease (194). This study also showed that berberine promotes brown adipogenic program in chow-fed mice, HFD-fed mice, and cultured mouse and human preadipocytes. Moreover, the study demonstrated that the berberine-induced adipose browning and thermogenic program depend on adipose AMPK because berberine has no such effects in adipose AMPKα1 and AMPKα2 KO mice. These findings collectively highlight an important role of the AMPK-dependent adipose browning in mediating berberine’s metabolic benefits.
In a human study, Wu et al. (194) examined the effect of berberine intervention to combat overweight. They found that mildly overweight individuals with NAFLD treated with berberine (500 mg, 3 times per day) for 1 month have increased BAT volume and activity. It is currently unknown whether berberine promotes WAT browning in humans.
CONCLUDING REMARKS
In this review, we focus on natural bioactive compounds as potential activators of WAT browning or beiging in preclinical animal models and cell culture systems. Current findings from animal and cell culture studies suggest that many compounds may possess such an activity. Several mechanisms have been suggested to mediate bioactive compounds-associated adipose browning, such as activation of AMPK, SIRT1, and PGC-1α pathways (Fig. 1). These pathways are well known to regulate energy metabolism and promote metabolic benefits. Humans have brown fat-like or beige adipose tissues. Since these thermogenic tissues dissipate metabolic energy as heat, their recruitment and activation by bioactive compounds may help combat obesity and related metabolic diseases in humans. Generally, naturally occurring compounds hold a special interest due to their better acceptability than pharmacological drugs and invasive procedures of obesity and metabolic disorders. Unfortunately, most bioactive compounds and their metabolites have low bioavailability (195–205), which limits their bioefficacy in animals and humans. For example, resveratrol, the most widely studied nonflavonoid polyphenol, has shown many metabolic benefits in in vitro and in vivo preclinical studies, but the low bioavailability of resveratrol and its metabolites limits their applications in humans (70, 206–209). Similarly, many flavonoids have poor bioavailability despite their potential health benefits (210, 211). Substantial efforts have been made to improve the bioavailability of resveratrol and some flavonoids and the outcomes are encouraging (212, 213). Similar efforts should be made to improve the bioavailability of each natural bioactive compound because it can proceed to large scale clinical studies, which are scarce, particularly those focusing on adipose browning and associated metabolic outcomes.
Fig. 1.
A schematic of potential mechanisms for natural agent-induced adipose browning. Many natural bioactive compounds appear to induce UCP-1-mediated adipose browning via several signaling pathways known to promote UCP-1 transcription and mitochondrial oxidation in WAT. The signaling pathways involved in the up-regulation of UCP-1 and its transcriptional regulators are shown. Some bioactive compounds appear to induce adipose browning by activating the AMPK/SIRT1/PGC-1α pathway directly or indirectly. Activation of AMPK can promote mitochondrial bioenergetics by turning on PGC-1α transcription, directly phosphorylating PGC-1α, or activating PGC-1α in a SIRT1-dependent manner. SIRT1 activates PGC-1α through deacetylation. The transcription coregulator PRDM16 can interact with PGC-1α and PPARα/γ to promote UCP-1 expression. Some bioactive compounds likely induce adipose browning by stimulating TRPM8, β3-AR receptor, or TGR5, which can lead to the activation of multiple PKA-dependent signaling pathways, including those through p38 MARP, CREB, and T3. Ac, acetylation; AC, adenylyl cyclase; AMPK, 5’ AMP-activated protein kinase; ArtC, artepillin C; ATF2, activating transcription factor 2; β3-AR, beta-3 adrenergic receptor; C3G, cyanidin-3-O-β-glucoside; CREB, cAMP-response element binding protein; Dio2, iodothyronine deiodinase 2; EPI, (−)-epicatechin; LCA, lithocholic acid; P, phosphorylation; PGC-1α, peroxisome proliferator-activated receptor γ (PPARγ) co-activator-1α; PKA, protein kinase A; PKC, protein kinase C; PRDM16, PR domain zinc finger protein 16; RSV, resveratrol; SIRT1, sirtuin-1; TGR5, Takeda G protein-coupled receptor 5; TR, thyroid hormone receptor; TRPM8, transient receptor potential cation channel subfamily M member 8; TRPV1, transient receptor potential cation channel subfamily V member 1; UCP-1, uncoupling protein-1.
In addition to an urgent need to solve the pool bioavailability problem of most bioactive food compounds, other issues are also noted in the literature. 1) the majority of the studies use male mice or rats, making the results largely limited to one gender; 2) the physiological relevance of the doses applied in the studies needs to be carefully tested to avoid unwanted side or toxic effects of a compound; 3) lack of direct assessment of the effect of adipose browning on metabolic benefits such as weight loss and insulin sensitivity; 4) lack of studies with bioactive browning agents during aging; 5) scarcity of solid evidence supporting the direct involvement of a pathway in the mediation of browning effects; and 6) targeted tissue delivery and bioavailability, and importantly toxicity or safety of a compound need to be established before performing comprehensive, large-scale and randomized clinical trials in humans. Use of natural products is a promising route for obesity management. With more research, specific natural compounds may emerge as effective and safe agents for treatment and prevention of obesity and associated metabolic sequelae.
Acknowledgments and Disclosures.
This work was supported in part by Award Numbers DK111052 (L.Y.) and DK116496 (L.Y.) from the National Institute of Diabetes and Digestive and Kidney Diseases and Award Number 1-18-IBS-346 (L.Y.) from American Diabetes Association. The authors report no conflicts of interest.
ABBREVIATIONS
- AMPK
5’ AMP-activated protein kinase
- ArtC
Artepillin C
- BAT
Brown adipose tissue
- BDNF
Brain-derived neurotrophic factor
- BMPs
Bone morphogenetic proteins
- C3G
Cyanidin-3-O-β-glucoside
- CPT1α
Carnitine Palmitoyltransferase 1α
- Dio2
Iodothyronine Deiodinase 2
- Elovl3
ELOVL Fatty Acid Elongase 3
- EPI
(−)-Epicatechin
- eWAT
Epididymal white adipose tissue
- FGF21
Fibroblast growth factor 21
- GTC
Green tea catechins
- HFD
High-fat diet
- iWAT
Inguinal subcutaneous WAT
- LCA
Lithocholic acid
- mTOR
Mammalian target of rapamycin
- NE
Norepinephrine
- PDEs
Phosphodiesterases
- PGC-1α
PPARγ co-activator-1α
- PKA
Protein kinase A
- PPARγ
Peroxisome proliferator-activated receptor γ
- PRDM16
PR domain zinc finger protein 16
- RSV
Resveratrol
- SIRT1
Sirtuin-1
- SVCs
Stromal vascular cells
- Tbx1
T-Box Transcription Factor 1
- Tfam
Mitochondrial transcription factor A
- TG
Triglyceride
- TMEM26
Transmembrane protein 26
- TRPM8
Transient Receptor Potential Cation Channel Subfamily M Member 8
- TRPV1
Transient Receptor Potential Cation Channel Subfamily V Member 1
- UCP-1
Uncoupling protein-1
- WAT
White adipose tissue
- β3-AR
Beta-3 adrenergic receptor
REFERENCES
- 1.Caballero B Humans against Obesity: Who Will Win? Adv Nutr. 2019;10(suppl_1):S4–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Apovian CM, Aronne LJ, Bessesen DH, McDonnell ME, Murad MH, Pagotto U, et al. Pharmacological management of obesity: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2015;100(2):342–62. [DOI] [PubMed] [Google Scholar]
- 3.Saltiel AR. New therapeutic approaches for the treatment of obesity. Sci Transl Med. 2016;8(323):323rv322. [DOI] [PubMed] [Google Scholar]
- 4.Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004;84(1):277–359. [DOI] [PubMed] [Google Scholar]
- 5.van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, Drossaerts JM, Kemerink GJ, Bouvy ND, et al. Cold-activated brown adipose tissue in healthy men. N Engl J Med. 2009;360(15):1500–8. [DOI] [PubMed] [Google Scholar]
- 6.Virtanen KA, Lidell ME, Orava J, Heglind M, Westergren R, Niemi T, et al. Functional brown adipose tissue in healthy adults. N Engl J Med. 2009;360(15):1518–25. [DOI] [PubMed] [Google Scholar]
- 7.Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, et al. Identification and importance of brown adipose tissue in adult humans. N Engl J Med. 2009;360(15):1509–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Denjean F, Lachuer J, Geloen A, Cohen-Adad F, Moulin C, Barre H, et al. Differential regulation of uncoupling protein-1, −2 and −3 gene expression by sympathetic innervation in brown adipose tissue of thermoneutral or cold-exposed rats. FEBS Lett. 1999;444(2–3):181–5. [DOI] [PubMed] [Google Scholar]
- 9.Ikeda K, Maretich P, Kajimura S. The common and distinct features of Brown and Beige adipocytes. Trends Endocrinol Metab. 2018;29(3):191–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Shabalina IG, Petrovic N, de Jong JM, Kalinovich AV, Cannon B, Nedergaard J. UCP1 in brite/beige adipose tissue mitochondria is functionally thermogenic. Cell Rep. 2013;5(5):1196–203. [DOI] [PubMed] [Google Scholar]
- 11.Fenzl A, Kiefer FW. Brown adipose tissue and thermogenesis. Horm Mol Biol Clin Investig. 2014;19(1):25–37. [DOI] [PubMed] [Google Scholar]
- 12.Wu J, Bostrom P, Sparks LM, Ye L, Choi JH, Giang AH, et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell. 2012;150(2):366–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Harms M, Seale P. Brown and beige fat: development, function and therapeutic potential. Nat Med. 2013;19(10):1252–63. [DOI] [PubMed] [Google Scholar]
- 14.Bartelt A, Heeren J. Adipose tissue browning and metabolic health. Nat Rev Endocrinol. 2014;10(1):24–36. [DOI] [PubMed] [Google Scholar]
- 15.Sidossis L, Kajimura S. Brown and beige fat in humans: thermogenic adipocytes that control energy and glucose homeostasis. J Clin Invest. 2015;125(2):478–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Liu X, Rossmeisl M, McClaine J, Riachi M, Harper ME, Kozak LP. Paradoxical resistance to diet-induced obesity in UCP1-deficient mice. J Clin Invest. 2003;111(3):399–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bertholet AM, Kazak L, Chouchani ET, Bogaczynska MG, Paranjpe I, Wainwright GL, et al. Mitochondrial patch clamp of beige adipocytes reveals UCP1-positive and UCP1-negative cells both exhibiting futile Creatine cycling. Cell Metab. 2017;25(4):811–22 e814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kazak L, Chouchani ET, Jedrychowski MP, Erickson BK, Shinoda K, Cohen P, et al. A creatine-driven substrate cycle enhances energy expenditure and thermogenesis in beige fat. Cell. 2015;163(3):643–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ikeda K, Kang Q, Yoneshiro T, Camporez JP, Maki H, Homma M, et al. UCP1-independent signaling involving SERCA2b-mediated calcium cycling regulates beige fat thermogenesis and systemic glucose homeostasis. Nat Med. 2017;23(12):1454–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Cao W, Daniel KW, Robidoux J, Puigserver P, Medvedev AV, Bai X, et al. p38 mitogen-activated protein kinase is the central regulator of cyclic AMP-dependent transcription of the brown fat uncoupling protein 1 gene. Mol Cell Biol. 2004;24(7):3057–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Seale P, Conroe HM, Estall J, Kajimura S, Frontini A, Ishibashi J, et al. Prdm16 determines the thermogenic program of subcutaneous white adipose tissue in mice. J Clin Invest. 2011;121(1):96–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Seale P, Bjork B, Yang W, Kajimura S, Chin S, Kuang S, et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature. 2008;454(7207):961–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kajimura S, Seale P, Kubota K, Lunsford E, Frangioni JV, Gygi SP, et al. Initiation of myoblast to brown fat switch by a PRDM16-C/EBP-beta transcriptional complex. Nature. 2009;460(7259):1154–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Seale P, Kajimura S, Yang W, Chin S, Rohas LM, Uldry M, et al. Transcriptional control of brown fat determination by PRDM16. Cell Metab. 2007;6(1):38–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Mulligan JD, Gonzalez AA, Stewart AM, Carey HV, Saupe KW. Upregulation of AMPK during cold exposure occurs via distinct mechanisms in brown and white adipose tissue of the mouse. J Physiol. 2007;580(Pt. 2):677–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hutchinson DS, Chernogubova E, Dallner OS, Cannon B, Bengtsson T. Beta-adrenoceptors, but not alpha-adrenoceptors, stimulate AMP-activated protein kinase in brown adipocytes independently of uncoupling protein-1. Diabetologia. 2005;48(11): 2386–95. [DOI] [PubMed] [Google Scholar]
- 27.Vila-Bedmar R, Lorenzo M, Fernandez-Veledo S. Adenosine 5′-monophosphate-activated protein kinase-mammalian target of rapamycin cross talk regulates brown adipocyte differentiation. Endocrinology. 2010;151(3):980–92. [DOI] [PubMed] [Google Scholar]
- 28.Baskaran P, Krishnan V, Ren J, Thyagarajan B. Capsaicin induces browning of white adipose tissue and counters obesity by activating TRPV1 channel-dependent mechanisms. Br J Pharmacol. 2016;173(15):2369–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Fisher FM, Kleiner S, Douris N, Fox EC, Mepani RJ, Verdeguer F, et al. FGF21 regulates PGC-1alpha and browning of white adipose tissues in adaptive thermogenesis. Genes Dev. 2012;26(3):271–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Keipert S, Kutschke M, Ost M, Schwarzmayr T, van Schothorst EM, Lamp D, et al. Long-term cold adaptation does not require FGF21 or UCP1. Cell Metab. 2017;26(2):437–46 e435. [DOI] [PubMed] [Google Scholar]
- 31.Whittle AJ, Carobbio S, Martins L, Slawik M, Hondares E, Vazquez MJ, et al. BMP8B increases brown adipose tissue thermogenesis through both central and peripheral actions. Cell. 2012;149(4):871–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kim S, Choe S, Lee DK. BMP-9 enhances fibroblast growth factor 21 expression and suppresses obesity. Biochim Biophys Acta. 2016;1862(7):1237–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Tseng YH, Kokkotou E, Schulz TJ, Huang TL, Winnay JN, Taniguchi CM, et al. New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature. 2008;454(7207):1000–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Schulz TJ, Huang P, Huang TL, Xue R, McDougall LE, Townsend KL, et al. Brown-fat paucity due to impaired BMP signalling induces compensatory browning of white fat. Nature. 2013;495(7441):379–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kuo MM, Kim S, Tseng CY, Jeon YH, Choe S, Lee DK. BMP-9 as a potent brown adipogenic inducer with anti-obesity capacity. Biomaterials. 2014;35(10):3172–9. [DOI] [PubMed] [Google Scholar]
- 36.Qiang L, Wang L, Kon N, Zhao W, Lee S, Zhang Y, et al. Brown remodeling of white adipose tissue by SirT1-dependent deacetylation of Ppargamma. Cell. 2012;150(3):620–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Rachid TL, Penna-de-Carvalho A, Bringhenti I, Aguila MB, Mandarim-de-Lacerda CA, Souza-Mello V. Fenofibrate (PPARalpha agonist) induces beige cell formation in subcutaneous white adipose tissue from diet-induced male obese mice. Mol Cell Endocrinol. 2015;402:86–94. [DOI] [PubMed] [Google Scholar]
- 38.Liu D, Bordicchia M, Zhang C, Fang H, Wei W, Li JL, et al. Activation of mTORC1 is essential for beta-adrenergic stimulation of adipose browning. J Clin Invest. 2016;126(5):1704–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.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(1). [DOI] [PubMed] [Google Scholar]
- 40.Schlessinger K, Li W, Tan Y, Liu F, Souza SC, Tozzo E, et al. Gene expression in WAT from healthy humans and monkeys correlates with FGF21-induced browning of WAT in mice. Obesity (Silver Spring). 2015;23(9):1818–29. [DOI] [PubMed] [Google Scholar]
- 41.Spiegelman BM. Banting lecture 2012: regulation of adipogenesis: toward new therapeutics for metabolic disease. Diabetes. 2013;62(6):1774–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wu J, Cohen P, Spiegelman BM. Adaptive thermogenesis in adipocytes: is beige the new brown? Genes Dev. 2013;27(3):234–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Veeresham C Natural products derived from plants as a source of drugs. J Adv Pharm Technol Res. 2012;3(4):200–1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Mele L, Bidault G, Mena P, Crozier A, Brighenti F, Vidal-Puig A, et al. Dietary (poly)phenols, Brown adipose tissue activation, and energy expenditure: a narrative review. Adv Nutr. 2017;8(5):694–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Concha F, Prado G, Quezada J, Ramirez A, Bravo N, Flores C, et al. Nutritional and non-nutritional agents that stimulate white adipose tissue browning. Rev Endocr Metab Disord. 2019;20(2): 161–71. [DOI] [PubMed] [Google Scholar]
- 46.Kang HW, Lee SG, Otieno D, Ha K. Flavonoids, Potential Bioactive Compounds, and Non-Shivering Thermogenesis. Nutrients. 2018;10(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zhang X, Li X, Fang H, Guo F, Li F, Chen A, et al. Flavonoids as inducers of white adipose tissue browning and thermogenesis: signalling pathways and molecular triggers. Nutr Metab (Lond). 2019;16:47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Del Rio D, Rodriguez-Mateos A, Spencer JP, Tognolini M, Borges G, Crozier A. Dietary (poly)phenolics in human health: structures, bioavailability, and evidence of protective effects against chronic diseases. Antioxid Redox Signal. 2013;18(14):1818–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Silvester AJ, Aseer KR, Yun JW. Dietary polyphenols and their roles in fat browning. J Nutr Biochem. 2019;64:1–12. [DOI] [PubMed] [Google Scholar]
- 50.Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006;444(7117):337–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Lerin C, Daussin F, et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell. 2006;127(6):1109–22. [DOI] [PubMed] [Google Scholar]
- 52.Um JH, Park SJ, Kang H, Yang S, Foretz M, McBurney MW, et al. AMP-activated protein kinase-deficient mice are resistant to the metabolic effects of resveratrol. Diabetes. 2010;59(3):554–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Bhatt JK, Thomas S, Nanjan MJ. Resveratrol supplementation improves glycemic control in type 2 diabetes mellitus. Nutr Res. 2012;32(7):537–41. [DOI] [PubMed] [Google Scholar]
- 54.Timmers S, Konings E, Bilet L, Houtkooper RH, van de Weijer T, Goossens GH, et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab. 2011;14(5):612–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Price NL, Gomes AP, Ling AJ, Duarte FV, Martin-Montalvo A, North BJ, et al. SIRT1 is required for AMPK activation and the beneficial effects of resveratrol on mitochondrial function. Cell Metab. 2012;15(5):675–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Mitterberger MC, Zwerschke W. Mechanisms of resveratrol-induced inhibition of clonal expansion and terminal adipogenic differentiation in 3T3-L1 preadipocytes. J Gerontol A Biol Sci Med Sci. 2013;68(11):1356–76. [DOI] [PubMed] [Google Scholar]
- 57.Wang S, Liang X, Yang Q, Fu X, Rogers CJ, Zhu M, et al. Resveratrol induces brown-like adipocyte formation in white fat through activation of AMP-activated protein kinase (AMPK) alpha1. Int J Obes. 2015;39(6):967–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lasa A, Schweiger M, Kotzbeck P, Churruca I, Simon E, Zechner R, et al. Resveratrol regulates lipolysis via adipose triglyceride lipase. J Nutr Biochem. 2012;23(4):379–84. [DOI] [PubMed] [Google Scholar]
- 59.Rayalam S, Yang JY, Ambati S, Della-Fera MA, Baile CA. Resveratrol induces apoptosis and inhibits adipogenesis in 3T3-L1 adipocytes. Phytother Res. 2008;22(10):1367–71. [DOI] [PubMed] [Google Scholar]
- 60.Alberdi G, Rodriguez VM, Miranda J, Macarulla MT, Churruca I, Portillo MP. Thermogenesis is involved in the body-fat lowering effects of resveratrol in rats. Food Chem. 2013;141(2):1530–5. [DOI] [PubMed] [Google Scholar]
- 61.Park SJ, Ahmad F, Philp A, Baar K, Williams T, Luo H, et al. Resveratrol ameliorates aging-related metabolic phenotypes by inhibiting cAMP phosphodiesterases. Cell. 2012;148(3):421–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hui S, Liu Y, Huang L, Zheng L, Zhou M, Lang H, et al. Resveratrol enhances brown adipose tissue activity and white adipose tissue browning in part by regulating bile acid metabolism via gut microbiota remodeling. Int J Obes. 2020;44(8):1678–90. [DOI] [PubMed] [Google Scholar]
- 63.Liao W, Yin X, Li Q, Zhang H, Liu Z, Zheng X, Zheng L, Feng X. Resveratrol-Induced White Adipose Tissue Browning in Obese Mice by Remodeling Fecal Microbiota. Molecules. 2018;23(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Velazquez-Villegas LA, Perino A, Lemos V, Zietak M, Nomura M, Pols TWH, et al. TGR5 signalling promotes mitochondrial fission and beige remodelling of white adipose tissue. Nat Commun. 2018;9(1):245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Watanabe M, Houten SM, Mataki C, Christoffolete MA, Kim BW, Sato H, et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature. 2006;439(7075):484–9. [DOI] [PubMed] [Google Scholar]
- 66.Ridlon JM, Kang DJ, Hylemon PB, Bajaj JS. Bile acids and the gut microbiome. Curr Opin Gastroenterol. 2014;30(3):332–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Chiang JYL. The gut’s feeling on bile acid signaling in NAFLD. Hepatobiliary Surg Nutr. 2018;7(2):151–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Russell DW, Setchell KD. Bile acid biosynthesis. Biochemistry. 1992;31(20):4737–49. [DOI] [PubMed] [Google Scholar]
- 69.Wenzel E, Somoza V. Metabolism and bioavailability of trans-resveratrol. Mol Nutr Food Res. 2005;49(5):472–81. [DOI] [PubMed] [Google Scholar]
- 70.Wang P, Sang S. Metabolism and pharmacokinetics of resveratrol and pterostilbene. Biofactors. 2018;44(1):16–25. [DOI] [PubMed] [Google Scholar]
- 71.Serrano A, Asnani-Kishnani M, Rodriguez AM, Palou A, Ribot J, Bonet ML. Programming of the beige phenotype in white adipose tissue of adult mice by mild resveratrol and nicotinamide riboside supplementations in early postnatal life. Mol Nutr Food Res. 2018;62(21):e1800463. [DOI] [PubMed] [Google Scholar]
- 72.Serrano A, Asnani-Kishnani M, Couturier C, Astier J, Palou A, Landrier JF, Ribot J, Bonet ML. DNA Methylation Changes are Associated with the Programming of White Adipose Tissue Browning Features by Resveratrol and Nicotinamide Riboside Neonatal Supplementations in Mice. Nutrients. 2020;12(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Liu Z, Liao W, Yin X, Zheng X, Li Q, Zhang H, et al. Resveratrol-induced brown fat-like phenotype in 3T3-L1 adipocytes partly via mTOR pathway. Food Nutr Res. 2020;64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Imamura H, Nagayama D, Ishihara N, Tanaka S, Watanabe R, Watanabe Y, et al. Resveratrol attenuates triglyceride accumulation associated with upregulation of Sirt1 and lipoprotein lipase in 3T3-L1 adipocytes. Mol Genet Metab Rep. 2017;12:44–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Andrade JMO, Barcala-Jorge AS, Batista-Jorge GC, Paraiso AF, Freitas KM, Lelis DF, et al. Effect of resveratrol on expression of genes involved thermogenesis in mice and humans. Biomed Pharmacother. 2019;112:108634. [DOI] [PubMed] [Google Scholar]
- 76.Nijveldt RJ, van Nood E, van Hoorn DE, Boelens PG, van Norren K, van Leeuwen PA. Flavonoids: a review of probable mechanisms of action and potential applications. Am J Clin Nutr. 2001;74(4):418–25. [DOI] [PubMed] [Google Scholar]
- 77.Rivera L, Moron R, Sanchez M, Zarzuelo A, Galisteo M. Quercetin ameliorates metabolic syndrome and improves the inflammatory status in obese Zucker rats. Obesity (Silver Spring). 2008;16(9):2081–7. [DOI] [PubMed] [Google Scholar]
- 78.Kobori M, Takahashi Y, Sakurai M, Akimoto Y, Tsushida T, Oike H, et al. Quercetin suppresses immune cell accumulation and improves mitochondrial gene expression in adipose tissue of diet-induced obese mice. Mol Nutr Food Res. 2016;60(2):300–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Dong J, Zhang X, Zhang L, Bian HX, Xu N, Bao B, et al. Quercetin reduces obesity-associated ATM infiltration and inflammation in mice: a mechanism including AMPKalpha1/SIRT1. J Lipid Res. 2014;55(3):363–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112(12):1821–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112(12):1796–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860–7. [DOI] [PubMed] [Google Scholar]
- 83.Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006;444(7121):840–6. [DOI] [PubMed] [Google Scholar]
- 84.Henagan TM, Cefalu WT, Ribnicky DM, Noland RC, Dunville K, Campbell WW, et al. In vivo effects of dietary quercetin and quercetin-rich red onion extract on skeletal muscle mitochondria, metabolism, and insulin sensitivity. Genes Nutr. 2015;10(1):451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kuipers EN, Dam ADV, Held NM, Mol IM, Houtkooper RH, Rensen PCN, Boon MR. Quercetin Lowers Plasma Triglycerides Accompanied by White Adipose Tissue Browning in Diet-Induced Obese Mice. Int J Mol Sci. 2018;19(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lee SG, Parks JS, Kang HW. Quercetin, a functional compound of onion peel, remodels white adipocytes to brown-like adipocytes. J Nutr Biochem. 2017;42:62–71. [DOI] [PubMed] [Google Scholar]
- 87.Khan N, Mukhtar H. Tea Polyphenols in Promotion of Human Health. Nutrients. 2018;11(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Yang CS, Chen G, Wu Q. Recent scientific studies of a traditional chinese medicine, tea, on prevention of chronic diseases. J Tradit Complement Med. 2014;4(1):17–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ueda-Wakagi M, Nagayasu H, Yamashita Y, Ashida AH. Green Tea Ameliorates Hyperglycemia by Promoting the Translocation of Glucose Transporter 4 in the Skeletal Muscle of Diabetic Rodents. Int J Mol Sci. 2019;20(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Wang LC, Pan TM, Tsai TY. Lactic acid bacteria-fermented product of green tea and Houttuynia cordata leaves exerts anti-adipogenic and anti-obesity effects. J Food Drug Anal. 2018;26(3):973–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Yan J, Zhao Y, Zhao B. Green tea catechins prevent obesity through modulation of peroxisome proliferator-activated receptors. Sci China Life Sci. 2013;56(9):804–10. [DOI] [PubMed] [Google Scholar]
- 92.Gutierrez-Salmean G, Ortiz-Vilchis P, Vacaseydel CM, Garduno-Siciliano L, Chamorro-Cevallos G, Meaney E, et al. Effects of (−)-epicatechin on a diet-induced rat model of cardio-metabolic risk factors. Eur J Pharmacol. 2014;728:24–30. [DOI] [PubMed] [Google Scholar]
- 93.Nirengi S, Amagasa S, Homma T, Yoneshiro T, Matsumiya S, Kurosawa Y, et al. Daily ingestion of catechin-rich beverage increases brown adipose tissue density and decreases extramyocellular lipids in healthy young women. Springerplus. 2016;5(1):1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Weisberg SP, Leibel R, Tortoriello DV. Dietary curcumin significantly improves obesity-associated inflammation and diabetes in mouse models of diabesity. Endocrinology. 2008;149(7):3549–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Quiles JL, Aguilera C, Mesa MD, Ramirez-Tortosa MC, Baro L, Gil A. An ethanolic-aqueous extract of Curcuma longa decreases the susceptibility of liver microsomes and mitochondria to lipid peroxidation in atherosclerotic rabbits. Biofactors. 1998;8(1–2): 51–7. [DOI] [PubMed] [Google Scholar]
- 96.Rao DS, Sekhara NC, Satyanarayana MN, Srinivasan M. Effect of curcumin on serum and liver cholesterol levels in the rat. J Nutr. 1970;100(11):1307–15. [DOI] [PubMed] [Google Scholar]
- 97.Alappat L, Awad AB. Curcumin and obesity: evidence and mechanisms. Nutr Rev. 2010;68(12):729–38. [DOI] [PubMed] [Google Scholar]
- 98.Wang S, Wang X, Ye Z, Xu C, Zhang M, Ruan B, et al. Curcumin promotes browning of white adipose tissue in a norepinephrine-dependent way. Biochem Biophys Res Commun. 2015;466(2):247–53. [DOI] [PubMed] [Google Scholar]
- 99.Nedergaard J, Cannon B. The browning of white adipose tissue: some burning issues. Cell Metab. 2014;20(3):396–407. [DOI] [PubMed] [Google Scholar]
- 100.Song Z, Revelo X, Shao W, Tian L, Zeng K, Lei H, et al. Dietary curcumin intervention targets mouse white adipose tissue inflammation and Brown adipose tissue UCP1 expression. Obesity (Silver Spring). 2018;26(3):547–58. [DOI] [PubMed] [Google Scholar]
- 101.Lone J, Choi JH, Kim SW, Yun JW. Curcumin induces brown fat-like phenotype in 3T3-L1 and primary white adipocytes. J Nutr Biochem. 2016;27:193–202. [DOI] [PubMed] [Google Scholar]
- 102.Matsuda AH, de Almeida-Muradian LB. Validated method for the quantification of artepillin-C in Brazilian propolis. Phytochem Anal. 2008;19(2):179–83. [DOI] [PubMed] [Google Scholar]
- 103.Simoes LM, Gregorio LE, Da Silva Filho AA, de Souza ML, Azzolini AE, Bastos JK, et al. Effect of Brazilian green propolis on the production of reactive oxygen species by stimulated neutrophils. J Ethnopharmacol. 2004;94(1):59–65. [DOI] [PubMed] [Google Scholar]
- 104.Cheung KW, Sze DM, Chan WK, Deng RX, Tu W, Chan GC. Brazilian green propolis and its constituent, Artepillin C inhibits allogeneic activated human CD4 T cells expansion and activation. J Ethnopharmacol. 2011;138(2):463–71. [DOI] [PubMed] [Google Scholar]
- 105.Szliszka E, Helewski KJ, Mizgala E, Krol W. The dietary flavonol fisetin enhances the apoptosis-inducing potential of TRAIL in prostate cancer cells. Int J Oncol. 2011;39(4):771–9. [DOI] [PubMed] [Google Scholar]
- 106.Nishikawa S, Aoyama H, Kamiya M, Higuchi J, Kato A, Soga M, et al. Artepillin C, a typical Brazilian Propolis-derived component, induces Brown-like adipocyte formation in C3H10T1/2 cells, primary inguinal white adipose tissue-derived adipocytes, and mice. PLoS One. 2016;11(9):e0162512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Kreft S, Knapp M, Kreft I. Extraction of rutin from buckwheat (Fagopyrum esculentumMoench) seeds and determination by capillary electrophoresis. J Agric Food Chem. 1999;47(11):4649–52. [DOI] [PubMed] [Google Scholar]
- 108.Huang WY, Zhang HC, Liu WX, Li CY. Survey of antioxidant capacity and phenolic composition of blueberry, blackberry, and strawberry in Nanjing. J Zhejiang Univ Sci B. 2012;13(2):94–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Korkmaz A, Kolankaya D. Protective effect of rutin on the ischemia/reperfusion induced damage in rat kidney. J Surg Res. 2010;164(2):309–15. [DOI] [PubMed] [Google Scholar]
- 110.Stanley Mainzen Prince P, Kamalakkannan N. Rutin improves glucose homeostasis in streptozotocin diabetic tissues by altering glycolytic and gluconeogenic enzymes. J Biochem Mol Toxicol. 2006;20(2):96–102. [DOI] [PubMed] [Google Scholar]
- 111.Gao M, Ma Y, Liu D. Rutin suppresses palmitic acids-triggered inflammation in macrophages and blocks high fat diet-induced obesity and fatty liver in mice. Pharm Res. 2013;30(11):2940–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Yuan X, Wei G, You Y, Huang Y, Lee HJ, Dong M, et al. Rutin ameliorates obesity through brown fat activation. FASEB J. 2017;31(1):333–45. [DOI] [PubMed] [Google Scholar]
- 113.Lopez-Lazaro M Distribution and biological activities of the flavonoid luteolin. Mini Rev Med Chem. 2009;9(1):31–59. [DOI] [PubMed] [Google Scholar]
- 114.Qian LB, Wang HP, Chen Y, Chen FX, Ma YY, Bruce IC, et al. Luteolin reduces high glucose-mediated impairment of endothelium-dependent relaxation in rat aorta by reducing oxidative stress. Pharmacol Res. 2010;61(4):281–7. [DOI] [PubMed] [Google Scholar]
- 115.El-Bassossy HM, Abo-Warda SM, Fahmy A. Chrysin and luteolin attenuate diabetes-induced impairment in endothelial-dependent relaxation: effect on lipid profile. AGEs and NO generation Phytother Res. 2013;27(11):1678–84. [DOI] [PubMed] [Google Scholar]
- 116.Xu N, Zhang L, Dong J, Zhang X, Chen YG, Bao B, et al. Low-dose diet supplement of a natural flavonoid, luteolin, ameliorates diet-induced obesity and insulin resistance in mice. Mol Nutr Food Res. 2014;58(6):1258–68. [DOI] [PubMed] [Google Scholar]
- 117.Kwon EY, Jung UJ, Park T, Yun JW, Choi MS. Luteolin attenuates hepatic steatosis and insulin resistance through the interplay between the liver and adipose tissue in mice with diet-induced obesity. Diabetes. 2015;64(5):1658–69. [DOI] [PubMed] [Google Scholar]
- 118.Zhang X, Zhang QX, Wang X, Zhang L, Qu W, Bao B, et al. Dietary luteolin activates browning and thermogenesis in mice through an AMPK/PGC1alpha pathway-mediated mechanism. Int J Obes. 2016;40(12):1841–9. [DOI] [PubMed] [Google Scholar]
- 119.Harnly JM, Doherty RF, Beecher GR, Holden JM, Haytowitz DB, Bhagwat S, et al. Flavonoid content of U.S. fruits, vegetables, and nuts. J Agric Food Chem. 2006;54(26):9966–77. [DOI] [PubMed] [Google Scholar]
- 120.Chen W, Li Y, Li J, Han Q, Ye L, Li A. Myricetin affords protection against peroxynitrite-mediated DNA damage and hydroxyl radical formation. Food Chem Toxicol. 2011;49(9):2439–44. [DOI] [PubMed] [Google Scholar]
- 121.Lu J, Papp LV, Fang J, Rodriguez-Nieto S, Zhivotovsky B, Holmgren A. Inhibition of mammalian thioredoxin reductase by some flavonoids: implications for myricetin and quercetin anticancer activity. Cancer Res. 2006;66(8):4410–8. [DOI] [PubMed] [Google Scholar]
- 122.Chen W, Feng L, Shen Y, Su H, Li Y, Zhuang J, et al. Myricitrin inhibits acrylamide-mediated cytotoxicity in human Caco-2 cells by preventing oxidative stress. Biomed Res Int. 2013;2013:724183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Chen W, Zhuang J, Li Y, Shen Y, Zheng X. Myricitrin protects against peroxynitrite-mediated DNA damage and cytotoxicity in astrocytes. Food Chem. 2013;141(2):927–33. [DOI] [PubMed] [Google Scholar]
- 124.Ong KC, Khoo HE. Effects of myricetin on glycemia and glycogen metabolism in diabetic rats. Life Sci. 2000;67(14):1695–705. [DOI] [PubMed] [Google Scholar]
- 125.Liu IM, Liou SS, Lan TW, Hsu FL, Cheng JT. Myricetin as the active principle of Abelmoschus moschatus to lower plasma glucose in streptozotocin-induced diabetic rats. Planta Med. 2005;71(7):617–21. [DOI] [PubMed] [Google Scholar]
- 126.Liu IM, Tzeng TF, Liou SS, Lan TW. Myricetin, a naturally occurring flavonol, ameliorates insulin resistance induced by a high-fructose diet in rats. Life Sci. 2007;81(21–22):1479–88. [DOI] [PubMed] [Google Scholar]
- 127.Hu T, Yuan X, Wei G, Luo H, Lee HJ, Jin W. Myricetin-induced brown adipose tissue activation prevents obesity and insulin resistance in db/db mice. Eur J Nutr. 2018;57(1):391–403. [DOI] [PubMed] [Google Scholar]
- 128.Yuasa K, Tada K, Harita G, Fujimoto T, Tsukayama M, Tsuji A. Sudachitin, a polymethoxyflavone from Citrus sudachi, suppresses lipopolysaccharide-induced inflammatory responses in mouse macrophage-like RAW264 cells. Biosci Biotechnol Biochem. 2012;76(3):598–600. [DOI] [PubMed] [Google Scholar]
- 129.Tsutsumi R, Yoshida T, Nii Y, Okahisa N, Iwata S, Tsukayama M, et al. Sudachitin, a polymethoxylated flavone, improves glucose and lipid metabolism by increasing mitochondrial biogenesis in skeletal muscle. Nutr Metab (Lond). 2014;11:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Olivas-Aguirre FJ, Rodrigo-Garcia J, Martinez-Ruiz ND, Cardenas-Robles AI, Mendoza-Diaz SO, Alvarez-Parrilla E, Gonzalez-Aguilar GA, de la Rosa LA, Ramos-Jimenez A, Wall-Medrano A. Cyanidin-3-O-glucoside: Physical-Chemistry, Foodomics and Health Effects. Molecules. 2016;21(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Wang Y, Zhao L, Wang D, Huo Y, Ji B. Anthocyanin-rich extracts from blackberry, wild blueberry, strawberry, and chokeberry: antioxidant activity and inhibitory effect on oleic acid-induced hepatic steatosis in vitro. J Sci Food Agric. 2016;96(7): 2494–503. [DOI] [PubMed] [Google Scholar]
- 132.Guo H, Xia M, Zou T, Ling W, Zhong R, Zhang W. Cyanidin 3-glucoside attenuates obesity-associated insulin resistance and hepatic steatosis in high-fat diet-fed and db/db mice via the transcription factor FoxO1. J Nutr Biochem. 2012;23(4):349–60. [DOI] [PubMed] [Google Scholar]
- 133.Liu Y, Li D, Zhang Y, Sun R, Xia M. Anthocyanin increases adiponectin secretion and protects against diabetes-related endothelial dysfunction. Am J Physiol Endocrinol Metab. 2014;306(8):E975–88. [DOI] [PubMed] [Google Scholar]
- 134.Sasaki R, Nishimura N, Hoshino H, Isa Y, Kadowaki M, Ichi T, et al. Cyanidin 3-glucoside ameliorates hyperglycemia and insulin sensitivity due to downregulation of retinol binding protein 4 expression in diabetic mice. Biochem Pharmacol. 2007;74(11):1619–27. [DOI] [PubMed] [Google Scholar]
- 135.You Y, Yuan X, Liu X, Liang C, Meng M, Huang Y, Han X, Guo J, Guo Y, Ren C, Zhang Q, Sun X, Ma T, Liu G, Jin W, Huang W, Zhan J. Cyanidin-3-glucoside increases whole body energy metabolism by upregulating brown adipose tissue mitochondrial function. Mol Nutr Food Res. 2017;61(11). [DOI] [PubMed] [Google Scholar]
- 136.Swiezewska E, Danikiewicz W. Polyisoprenoids: structure, biosynthesis and function. Prog Lipid Res. 2005;44(4):235–58. [DOI] [PubMed] [Google Scholar]
- 137.van den Brink DM, Wanders RJ. Phytanic acid: production from phytol, its breakdown and role in human disease. Cell Mol Life Sci. 2006;63(15):1752–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Santos CC, Salvadori MS, Mota VG, Costa LM, de Almeida AA, de Oliveira GA, et al. Antinociceptive and antioxidant activities of phytol in vivo and in vitro models. Neurosci J. 2013;2013:949452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Elmazar MM, El-Abhar HS, Schaalan MF, Farag NA. Phytol/Phytanic acid and insulin resistance: potential role of phytanic acid proven by docking simulation and modulation of biochemical alterations. PLoS One. 2013;8(1):e45638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Goto T, Takahashi N, Kato S, Egawa K, Ebisu S, Moriyama T, et al. Phytol directly activates peroxisome proliferator-activated receptor alpha (PPARalpha) and regulates gene expression involved in lipid metabolism in PPARalpha-expressing HepG2 hepatocytes. Biochem Biophys Res Commun. 2005;337(2):440–5. [DOI] [PubMed] [Google Scholar]
- 141.Zhang F, Ai W, Hu X, Meng Y, Yuan C, Su H, et al. Phytol stimulates the browning of white adipocytes through the activation of AMP-activated protein kinase (AMPK) alpha in mice fed high-fat diet. Food Funct. 2018;9(4):2043–50. [DOI] [PubMed] [Google Scholar]
- 142.Maeda H, Hosokawa M, Sashima T, Miyashita K. Dietary combination of fucoxanthin and fish oil attenuates the weight gain of white adipose tissue and decreases blood glucose in obese/diabetic KK-ay mice. J Agric Food Chem. 2007;55(19):7701–6. [DOI] [PubMed] [Google Scholar]
- 143.Sangeetha RK, Bhaskar N, Baskaran V. Comparative effects of beta-carotene and fucoxanthin on retinol deficiency induced oxidative stress in rats. Mol Cell Biochem. 2009;331(1–2):59–67. [DOI] [PubMed] [Google Scholar]
- 144.Lee SJ, Bai SK, Lee KS, Namkoong S, Na HJ, Ha KS, et al. Astaxanthin inhibits nitric oxide production and inflammatory gene expression by suppressing I(kappa)B kinase-dependent NF-kappaB activation. Mol Cells. 2003;16(1):97–105. [PubMed] [Google Scholar]
- 145.Maeda H Nutraceutical effects of fucoxanthin for obesity and diabetes therapy: a review. J Oleo Sci. 2015;64(2):125–32. [DOI] [PubMed] [Google Scholar]
- 146.Gammone MA, D’Orazio N. Anti-obesity activity of the marine carotenoid fucoxanthin. Mar Drugs. 2015;13(4):2196–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Maeda H, Hosokawa M, Sashima T, Funayama K, Miyashita K. Fucoxanthin from edible seaweed, Undaria pinnatifida, shows antiobesity effect through UCP1 expression in white adipose tissues. Biochem Biophys Res Commun. 2005;332(2):392–7. [DOI] [PubMed] [Google Scholar]
- 148.Rebello CJ, Greenway FL, Johnson WD, Ribnicky D, Poulev A, Stadler K, et al. Fucoxanthin and its metabolite Fucoxanthinol do not induce Browning in human adipocytes. J Agric Food Chem. 2017;65(50):10915–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Yang F, Shi H, Zhang X, Yu LL. Two novel anti-inflammatory 21-nordammarane saponins from tetraploid Jiaogulan (Gynostemma pentaphyllum). J Agric Food Chem. 2013;61(51):12646–52. [DOI] [PubMed] [Google Scholar]
- 150.Yang F, Shi H, Zhang X, Yang H, Zhou Q, Yu LL. Two new saponins from tetraploid jiaogulan (Gynostemma pentaphyllum), and their anti-inflammatory and alpha-glucosidase inhibitory activities. Food Chem. 2013;141(4):3606–13. [DOI] [PubMed] [Google Scholar]
- 151.Megalli S, Davies NM, Roufogalis BD. Anti-hyperlipidemic and hypoglycemic effects of Gynostemma pentaphyllum in the Zucker fatty rat. J Pharm Pharm Sci. 2006;9(3):281–91. [PubMed] [Google Scholar]
- 152.Hu Y, Ip FC, Fu G, Pang H, Ye W, Ip NY. Dammarane saponins from Gynostemma pentaphyllum. Phytochemistry. 2010;71(10):1149–57. [DOI] [PubMed] [Google Scholar]
- 153.Yeo J, Kang YJ, Jeon SM, Jung UJ, Lee MK, Song H, et al. Potential hypoglycemic effect of an ethanol extract of Gynostemma pentaphyllum in C57BL/KsJ-db/db mice. J Med Food. 2008;11(4):709–16. [DOI] [PubMed] [Google Scholar]
- 154.Gauhar R, Hwang SL, Jeong SS, Kim JE, Song H, Park DC, et al. Heat-processed Gynostemma pentaphyllum extract improves obesity in Ob/Ob mice by activating AMP-activated protein kinase. Biotechnol Lett. 2012;34(9):1607–16. [DOI] [PubMed] [Google Scholar]
- 155.Liu J, Li Y, Yang P, Wan J, Chang Q, Wang TTY, et al. Gypenosides reduced the risk of overweight and insulin resistance in C57BL/6J mice through modulating adipose thermogenesis and gut microbiota. J Agric Food Chem. 2017;65(42):9237–46. [DOI] [PubMed] [Google Scholar]
- 156.Mu Q, Fang X, Li X, Zhao D, Mo F, Jiang G, et al. Ginsenoside Rb1 promotes browning through regulation of PPARgamma in 3T3-L1 adipocytes. Biochem Biophys Res Commun. 2015;466(3):530–5. [DOI] [PubMed] [Google Scholar]
- 157.Almeida MC, Hew-Butler T, Soriano RN, Rao S, Wang W, Wang J, et al. Pharmacological blockade of the cold receptor TRPM8 attenuates autonomic and behavioral cold defenses and decreases deep body temperature. J Neurosci. 2012;32(6):2086–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Bautista DM, Siemens J, Glazer JM, Tsuruda PR, Basbaum AI, Stucky CL, et al. The menthol receptor TRPM8 is the principal detector of environmental cold. Nature. 2007;448(7150):204–8. [DOI] [PubMed] [Google Scholar]
- 159.McKemy DD, Neuhausser WM, Julius D. Identification of a cold receptor reveals a general role for TRP channels in thermosensation. Nature. 2002;416(6876):52–8. [DOI] [PubMed] [Google Scholar]
- 160.Colburn RW, Lubin ML, Stone DJ Jr, Wang Y, Lawrence D, D’Andrea MR, et al. Attenuated cold sensitivity in TRPM8 null mice. Neuron. 2007;54(3):379–86. [DOI] [PubMed] [Google Scholar]
- 161.Dhaka A, Murray AN, Mathur J, Earley TJ, Petrus MJ, Patapoutian A. TRPM8 is required for cold sensation in mice. Neuron. 2007;54(3):371–8. [DOI] [PubMed] [Google Scholar]
- 162.Voets T, Owsianik G, Janssens A, Talavera K, Nilius B. TRPM8 voltage sensor mutants reveal a mechanism for integrating thermal and chemical stimuli. Nat Chem Biol. 2007;3(3):174–82. [DOI] [PubMed] [Google Scholar]
- 163.Ma S, Yu H, Zhao Z, Luo Z, Chen J, Ni Y, et al. Activation of the cold-sensing TRPM8 channel triggers UCP1-dependent thermogenesis and prevents obesity. J Mol Cell Biol. 2012;4(2):88–96. [DOI] [PubMed] [Google Scholar]
- 164.Sakellariou P, Valente A, Carrillo AE, Metsios GS, Nadolnik L, Jamurtas AZ, et al. Chronic l-menthol-induced browning of white adipose tissue hypothesis: a putative therapeutic regime for comating obesity and improving metabolic health. Med Hypotheses. 2016;93:21–6. [DOI] [PubMed] [Google Scholar]
- 165.Rossato M, Granzotto M, Macchi V, Porzionato A, Petrelli L, Calcagno A, et al. Human white adipocytes express the cold receptor TRPM8 which activation induces UCP1 expression, mitochondrial activation and heat production. Mol Cell Endocrinol. 2014;383(1–2):137–46. [DOI] [PubMed] [Google Scholar]
- 166.Jiang C, Zhai M, Yan D, Li D, Li C, Zhang Y, et al. Dietary menthol-induced TRPM8 activation enhances WAT “browning” and ameliorates diet-induced obesity. Oncotarget. 2017;8(43):75114–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Kannaiyan R, Shanmugam MK, Sethi G. Molecular targets of celastrol derived from thunder of god vine: potential role in the treatment of inflammatory disorders and cancer. Cancer Lett. 2011;303(1):9–20. [DOI] [PubMed] [Google Scholar]
- 168.Salminen A, Lehtonen M, Paimela T, Kaarniranta K. Celastrol: molecular targets of thunder god vine. Biochem Biophys Res Commun. 2010;394(3):439–42. [DOI] [PubMed] [Google Scholar]
- 169.Ng SW, Chan Y, Chellappan DK, Madheswaran T, Zeeshan F, Chan YL, et al. Molecular modulators of celastrol as the keystones for its diverse pharmacological activities. Biomed Pharmacother. 2019;109:1785–92. [DOI] [PubMed] [Google Scholar]
- 170.Liu J, Lee J, Salazar Hernandez MA, Mazitschek R, Ozcan U. Treatment of obesity with celastrol. Cell. 2015;161(5):999–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Hu M, Luo Q, Alitongbieke G, Chong S, Xu C, Xie L, et al. Celastrol-induced Nur77 interaction with TRAF2 alleviates inflammation by promoting mitochondrial ubiquitination and autophagy. Mol Cell. 2017;66(1):141–53 e146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Ma X, Xu L, Alberobello AT, Gavrilova O, Bagattin A, Skarulis M, et al. Celastrol protects against obesity and metabolic dysfunction through activation of a HSF1-PGC1alpha transcriptional Axis. Cell Metab. 2015;22(4):695–708. [DOI] [PubMed] [Google Scholar]
- 173.Pfuhlmann K, Schriever SC, Baumann P, Kabra DG, Harrison L, Mazibuko-Mbeje SE, et al. Celastrol-induced weight loss is driven by Hypophagia and independent from UCP1. Diabetes. 2018;67(11):2456–65. [DOI] [PubMed] [Google Scholar]
- 174.Enerback S, Jacobsson A, Simpson EM, Guerra C, Yamashita H, Harper ME, et al. Mice lacking mitochondrial uncoupling protein are cold-sensitive but not obese. Nature. 1997;387(6628):90–4. [DOI] [PubMed] [Google Scholar]
- 175.Chouchani ET, Kazak L, Spiegelman BM. New advances in adaptive thermogenesis: UCP1 and beyond. Cell Metab. 2019;29(1):27–37. [DOI] [PubMed] [Google Scholar]
- 176.Al Othman ZA, Ahmed YB, Habila MA, Ghafar AA. Determination of capsaicin and dihydrocapsaicin in Capsicum fruit samples using high performance liquid chromatography. Molecules. 2011;16(10):8919–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Dairam A, Fogel R, Daya S, Limson JL. Antioxidant and iron-binding properties of curcumin, capsaicin, and S-allylcysteine reduce oxidative stress in rat brain homogenate. J Agric Food Chem. 2008;56(9):3350–6. [DOI] [PubMed] [Google Scholar]
- 178.Zheng L, Chen J, Ma Z, Liu W, Yang F, Yang Z, et al. Capsaicin enhances anti-proliferation efficacy of pirarubicin via activating TRPV1 and inhibiting PCNA nuclear translocation in 5637 cells. Mol Med Rep. 2016;13(1):881–7. [DOI] [PubMed] [Google Scholar]
- 179.Narang N, Jiraungkoorskul W, Jamrus P. Current understanding of Antiobesity property of capsaicin. Pharmacogn Rev. 2017;11(21):23–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Janssens PL, Hursel R, Martens EA, Westerterp-Plantenga MS. Acute effects of capsaicin on energy expenditure and fat oxidation in negative energy balance. PLoS One. 2013;8(7):e67786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Baboota RK, Murtaza N, Jagtap S, Singh DP, Karmase A, Kaur J, et al. Capsaicin-induced transcriptional changes in hypothalamus and alterations in gut microbial count in high fat diet fed mice. J Nutr Biochem. 2014;25(9):893–902. [DOI] [PubMed] [Google Scholar]
- 182.Baboota RK, Singh DP, Sarma SM, Kaur J, Sandhir R, Boparai RK, et al. Capsaicin induces “brite” phenotype in differentiating 3T3-L1 preadipocytes. PLoS One. 2014;9(7):e103093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Chow YL, Sogame M, Sato F. 13-Methylberberine, a berberine analogue with stronger anti-adipogenic effects on mouse 3T3-L1 cells. Sci Rep. 2016;6:38129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Zhang WL, Zhu L, Jiang JG. Active ingredients from natural botanicals in the treatment of obesity. Obes Rev. 2014;15(12):957–67. [DOI] [PubMed] [Google Scholar]
- 185.Zhang Y, Li X, Zou D, Liu W, Yang J, Zhu N, et al. Treatment of type 2 diabetes and dyslipidemia with the natural plant alkaloid berberine. J Clin Endocrinol Metab. 2008;93(7):2559–65. [DOI] [PubMed] [Google Scholar]
- 186.Li Y, Wang P, Zhuang Y, Lin H, Li Y, Liu L, et al. Activation of AMPK by berberine promotes adiponectin multimerization in 3T3-L1 adipocytes. FEBS Lett. 2011;585(12):1735–40. [DOI] [PubMed] [Google Scholar]
- 187.Lee YS, Kim WS, Kim KH, Yoon MJ, Cho HJ, Shen Y, et al. Berberine, a natural plant product, activates AMP-activated protein kinase with beneficial metabolic effects in diabetic and insulin-resistant states. Diabetes. 2006;55(8):2256–64. [DOI] [PubMed] [Google Scholar]
- 188.Sun Y, Xia M, Yan H, Han Y, Zhang F, Hu Z, et al. Berberine attenuates hepatic steatosis and enhances energy expenditure in mice by inducing autophagy and fibroblast growth factor 21. Br J Pharmacol. 2018;175(2):374–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Kong W, Wei J, Abidi P, Lin M, Inaba S, Li C, et al. Berberine is a novel cholesterol-lowering drug working through a unique mechanism distinct from statins. Nat Med. 2004;10(12):1344–51. [DOI] [PubMed] [Google Scholar]
- 190.Hu Y, Davies GE. Berberine inhibits adipogenesis in high-fat diet-induced obesity mice. Fitoterapia. 2010;81(5):358–66. [DOI] [PubMed] [Google Scholar]
- 191.Wang L, Ye X, Hua Y, Song Y. Berberine alleviates adipose tissue fibrosis by inducing AMP-activated kinase signaling in high-fat diet-induced obese mice. Biomed Pharmacother. 2018;105:121–9. [DOI] [PubMed] [Google Scholar]
- 192.Xu JH, Liu XZ, Pan W, Zou DJ. Berberine protects against diet-induced obesity through regulating metabolic endotoxemia and gut hormone levels. Mol Med Rep. 2017;15(5):2765–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Zhang Z, Zhang H, Li B, Meng X, Wang J, Zhang Y, et al.Berberine activates thermogenesis in white and brown adipose tissue. Nat Commun. 2014;5:5493. [DOI] [PubMed] [Google Scholar]
- 194.Wu L, Xia M, Duan Y, Zhang L, Jiang H, Hu X, et al. Berberine promotes the recruitment and activation of brown adipose tissue in mice and humans. Cell Death Dis. 2019;10(6):468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Rein MJ, Renouf M, Cruz-Hernandez C, Actis-Goretta L, Thakkar SK, da Silva Pinto M. Bioavailability of bioactive food compounds: a challenging journey to bioefficacy. Br J Clin Pharmacol. 2013;75(3):588–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Oracz J, Nebesny E, Zyzelewicz D, Budryn G, Luzak B. Bioavailability and metabolism of selected cocoa bioactive compounds: a comprehensive review. Crit Rev Food Sci Nutr. 2020;60(12):1947–85. [DOI] [PubMed] [Google Scholar]
- 197.Teng H, Chen L. Polyphenols and bioavailability: an update. Crit Rev Food Sci Nutr. 2019;59(13):2040–51. [DOI] [PubMed] [Google Scholar]
- 198.Dima C, Assadpour E, Dima S, Jafari SM. Bioavailability and bioaccessibility of food bioactive compounds; overview and assessment by in vitro methods. Compr Rev Food Sci Food Saf. 2020;19(6):2862–84. [DOI] [PubMed] [Google Scholar]
- 199.Jarosova V, Vesely O, Doskocil I, Tomisova K, Marsik P, Jaimes JD, Smejkal K, Kloucek P, Havlik J. Metabolism of cis- and trans-Resveratrol and Dihydroresveratrol in an Intestinal Epithelial Model. Nutrients. 2020;12(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Luca SV, Macovei I, Bujor A, Miron A, Skalicka-Wozniak K, Aprotosoaie AC, et al. Bioactivity of dietary polyphenols: the role of metabolites. Crit Rev Food Sci Nutr. 2020;60(4):626–59. [DOI] [PubMed] [Google Scholar]
- 201.Erlund I, Freese R, Marniemi J, Hakala P, Alfthan G. Bioavailability of quercetin from berries and the diet. Nutr Cancer. 2006;54(1):13–7. [DOI] [PubMed] [Google Scholar]
- 202.Cai ZY, Li XM, Liang JP, Xiang LP, Wang KR, Shi YL, Yang R, Shi M, Ye JH, Lu JL, Zheng XQ, Liang YR. Bioavailability of Tea Catechins and Its Improvement. Molecules. 2018;23(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Chen DJ, Hu HG, Xing SF, Liu HM, Piao XL. Metabolite profiling of gypenoside LVI in rat after oral and intravenous administration. Arch Pharm Res. 2015;38(6):1157–67. [DOI] [PubMed] [Google Scholar]
- 204.Chen W, Miao YQ, Fan DJ, Yang SS, Lin X, Meng LK, et al. Bioavailability study of berberine and the enhancing effects of TPGS on intestinal absorption in rats. AAPS PharmSciTech. 2011;12(2):705–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Liu YT, Hao HP, Xie HG, Lai L, Wang Q, Liu CX, et al. Extensive intestinal first-pass elimination and predominant hepatic distribution of berberine explain its low plasma levels in rats. Drug Metab Dispos. 2010;38(10):1779–84. [DOI] [PubMed] [Google Scholar]
- 206.Springer M, Moco S. Resveratrol and Its Human Metabolites-Effects on Metabolic Health and Obesity. Nutrients. 2019;11(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Patel KR, Andreadi C, Britton RG, Horner-Glister E, Karmokar A, Sale S, Brown VA, Brenner DE, Singh R, Steward WP, Gescher AJ, Brown K. Sulfate metabolites provide an intracellular pool for resveratrol generation and induce autophagy with senescence. Sci Transl Med. 2013;5(205):205ra133. [DOI] [PubMed] [Google Scholar]
- 208.Gambini J, Ingles M, Olaso G, Lopez-Grueso R, Bonet-Costa V, Gimeno-Mallench L, et al. Properties of resveratrol: in vitro and in vivo studies about metabolism, bioavailability, and biological effects in animal models and humans. Oxidative Med Cell Longev. 2015;2015:837042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Walle T Bioavailability of resveratrol. Ann N Y Acad Sci. 2011;1215:9–15. [DOI] [PubMed] [Google Scholar]
- 210.Hostetler GL, Ralston RA, Schwartz SJ. Flavones: food sources, bioavailability, metabolism, and bioactivity. Adv Nutr. 2017;8(3): 423–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Gonzales GB, Smagghe G, Grootaert C, Zotti M, Raes K, Van amp J. Flavonoid interactions during digestion, absorption, distribution and metabolism: a sequential structure-activity/property relationship-based approach in the study of bioavailability and bioactivity. Drug Metab Rev. 2015;47(2):175–90. [DOI] [PubMed] [Google Scholar]
- 212.Chimento A, De Amicis F, Sirianni R, Sinicropi MS, Puoci F, Casaburi I, Saturnino C, Pezzi V. Progress to Improve Oral Bioavailability and Beneficial Effects of Resveratrol. Int J Mol Sci. 2019;20(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Thilakarathna SH, Rupasinghe HP. Flavonoid bioavailability and attempts for bioavailability enhancement. Nutrients. 2013;5(9): 3367–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Aziz SA, Wakeling LA, Miwa S, Alberdi G, Hesketh JE, Ford D. Metabolic programming of a beige adipocyte phenotype by genistein. Mol Nutr Food Res. 2017;61(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Song NJ, Choi S, Rajbhandari P, Chang SH, Kim S, Vergnes L, et al. Prdm4 induction by the small molecule butein promotes white adipose tissue browning. Nat Chem Biol. 2016;12(7):479–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Choi JH, Yun JW. Chrysin induces brown fat-like phenotype and enhances lipid metabolism in 3T3-L1 adipocytes. Nutrition. 2016;32(9):1002–10. [DOI] [PubMed] [Google Scholar]
- 217.Matsukawa T, Villareal MO, Motojima H, Isoda H. Increasing cAMP levels of preadipocytes by cyanidin-3-glucoside treatment induces the formation of beige phenotypes in 3T3-L1 adipocytes. J Nutr Biochem. 2017;40:77–85. [DOI] [PubMed] [Google Scholar]
- 218.Choi JH, Kim SW, Yu R, Yun JW. Monoterpene phenolic compound thymol promotes browning of 3T3-L1 adipocytes. Eur J Nutr. 2017;56(7):2329–41. [DOI] [PubMed] [Google Scholar]