Abstract
Obesity has become a global pandemic. The approaches researched to prevent it include decreasing energy intake and/or enhancing energy expenditure. Therefore, research on brown adipose tissue is of great importance. Brown adipose tissue is characterized by its high mitochondrial content. Mitochondrial uncoupling protein 1 (UCP1) releases energy as heat instead of chemical energy. Thermogenesis increases energy expenditure. Berberine, a phytochemical widely used in Asian countries, has positive effects on body weight control. While the precise mechanisms behind this effect remain unclear, the adenosine monophosphate-activated protein kinase (AMPK) pathway is known to play a crucial role. Berberine activates AMPK through phosphorylation, significantly impacting brown adipose tissue by enhancing lipolytic activity and increasing the expression of UCP1, peroxisome proliferator-activated receptor γ-co-activator-1α (PGC1α), and PR domain containing 16 (PRDM16). While investigating the mechanism of action of berberine, both the AMPK pathway is being examined in more detail and alternative pathways are being explored. One such pathway is growth differentiation factor 15 (GDF15), known for its appetite-suppressing effect. Berberine’s low stability and bioavailability, which are the main obstacles to its clinical use, have been improved through the development of nanotechnological methods. This review examines the potential mechanisms of berberine on browning and summarizes the methods developed to enhance its effect.
Keywords: Adipose tissue, Berberine, Browning, Obesity, Thermogenesis
Introduction
Obesity is a growing health concern worldwide, primarily driven by an imbalance between energy intake and expenditure (Mayoral et al., 2020). This condition is associated with numerous metabolic disorders, including diabetes, cardiovascular diseases, and non-alcoholic fatty liver disease. Addressing obesity requires innovative approaches to increase energy expenditure while reducing energy storage. Brown adipose tissue (BAT) and the browning of white adipose tissue have emerged as promising therapeutic targets due to their roles in thermogenesis. This process converts stored energy into heat, thereby increasing total energy expenditure (Marlatt & Ravussin, 2017).
Berberine is a natural compound extensively utilized in traditional medicine across many Asian countries, especially China (Wang et al., 2017). It is an isoquinoline alkaloid derived from several plants, such as Coptis chinensis, Berberis aquifolium, Berberis vulgaris, and Berberis aristate. Plants containing berberine have been used since ancient times. Coptis chinensis was used approximately 2,200 years ago for many health issues, particularly digestive system diseases (Song, Hao & Fan, 2020). Approximately 1,500 years ago, Hongjing Tao mentioned the antidiabetic properties of berberine plants in the book “Note of Elite Physicians” (Zhang et al., 2014a). With technological advancements, the active component in these plants was identified as berberine, and consequently, the number of studies on berberine has recently increased. Berberine is believed to have numerous effects, including anti-obesity, hypoglycemic, hypolipidemic, hypotensive, and anti-inflammatory effects (Hesari et al., 2018; Pirillo & Catapano, 2015; Yarla et al., 2016).
Berberine is regarded as a potential anti-obesity agent because of its beneficial health effects. Berberine may increase thermogenesis, positively affect carbohydrate and lipid metabolism, suppress appetite, regulate intestinal permeability and hepatic gluconeogenesis, and modulate the microbiota (Ilyas et al., 2020; Park, Jung & Shim, 2020; Rong et al., 2021; Wu et al., 2019; Zhang et al., 2020, 2014c). Taking 500 mg of berberine three times a day for 12 weeks may result in an average weight loss of approximately 2.3 kg (5 pounds) in individuals with obesity (Hu et al., 2012). Berberine’s effect on thermogenesis is one of the most researched topics in this area. Although the exact mechanism is not yet fully understood, berberine induces adipose tissue browning and thermogenesis through various pathways (Zhang et al., 2015, 2008). This browning effect is considered a key mechanism contributing to berberine’s potential role in weight loss, as it promotes increased energy expenditure and thermogenesis, addressing the energy imbalance central to obesity. Beyond its metabolic effects, berberine is relatively safe. While berberine toxicity is rarely observed in animals, human studies have reported some mild side effects, such as gastrointestinal disturbances like diarrhea or constipation (Imenshahidi & Hosseinzadeh, 2019; Zhang et al., 2010). Plants rich in berberine have been reported to be safe, showing no adverse effects on creatinine levels or liver function (Linn et al., 2012). The side effects of berberine vary depending on the route of administration, dosage, and duration of use.
This review aims to clarify the mechanisms of adipose tissue browning, which are essential for preventing obesity and its associated conditions, as well as to examine the effects of berberine on these mechanisms. There are different reviews in the literature examining the health effects of berberine. However, the number of articles presenting up-to-date data on the effects of berberine on adipose tissue browning and BAT activation is limited. Due to its low bioavailability, most studies on berberine are in vitro. Methods that could address this issue are provided in this review.
Survey methodology
In this review, articles containing the keywords “berberine” along with “brown adipose tissue,” “browning,” and “thermogenesis” in their titles or abstracts were searched in the PubMed, Science Direct, and Scopus databases. Since most of the relevant sources were recently published, no year restriction was applied. Only articles in English are considered. Research articles and reviews were included. A search using these criteria resulted in 278 research articles. After applying search filters, the titles of the resulting articles were reviewed first, followed by their abstracts. Articles with abstracts relevant to the research topic were then examined in detail. Studies that met the search criteria but were not relevant to the topic, did not provide sufficient data, or were inaccessible in full text were excluded. As a result, 10 studies relevant to the purpose of this review were included. Limiting the search to articles in English resulted in the exclusion of studies written in native languages from Asian countries, where the use of berberine is more prevalent. This can be considered a limitation of this study.
The audience it is intended for
This review may attract the attention of experts particularly interested in phytochemicals and adipose tissue, those investigating methods used to combat obesity, and those with an interest in nanotechnological approaches. With a better understanding of berberine’s effects on adipose tissue browning and the underlying mechanisms, it can be considered a potential drug for obesity prevention and/or treatment.
Berberine and its pharmacokinetic properties
Berberine (2,3-methylenedioxy-9,10-dimethoxyprotoberberine chloride) is yellow, odorless, and has a bitter taste (Feng et al., 2019). It is more soluble in organic solvents and has low water solubility. Its molecular weight is 336.36 g/mol. It can be extracted from its source plants, or it can be synthesized (Feng et al., 2019).
Absorption and bioavailability
While the health effects of berberine are intriguing, its low oral bioavailability (approximately 5%) is well-documented (Habtemariam, 2020; Wang et al., 2017). One reason for its limited bioavailability is its high binding affinity for plasma proteins (Mirhadi, Rezaee & Malaekeh-Nikouei, 2018). Therefore, research has focused on the metabolites of berberine and their health effects. Clinical evaluations have shown that intravenous administration of berberine increases its concentration in the blood (Han et al., 2021). However, this increase can dangerously lower blood pressure, potentially leading to death. Therefore, oral intake is safer than intravenous administration in clinical applications.
Distribution
The distribution of berberine varies depending on its form and route of administration. With oral administration, tissue distribution is high, while plasma concentration is relatively low (Tan et al., 2013). It particularly accumulates in the liver, adipose tissue, kidneys, and muscles. In intravenous administration, tissue distribution is faster (Liu et al., 2010). While this is desirable in acute conditions, it is not practical for chronic use. Intraperitoneal administration offers higher bioavailability compared to the oral route, but tissue distribution is slower compared to intravenous administration. Four hours after administration, berberine levels in many tissues are about 70 times higher than in plasma (Han et al., 2021). However, berberine levels remain stable in certain tissues such as the liver and muscles. Encapsulation of berberine or co-administration with P-glycoprotein inhibitors enhances its absorption and improves tissue distribution (Imenshahidi & Hosseinzadeh, 2019; Liu et al., 2010).
Metabolism
Berberine administered orally undergoes primary metabolism in the liver and intestines. The liver enzymes responsible for metabolizing berberine include cytochrome (CY) P2D6 and CYP1A2 subtypes of CYP450 (Fig. 1) (Li et al., 2011). In vivo studies showed that the primary metabolic pathways of berberine include demethylation, demethylenation, reduction, and hydroxylation (Liu et al., 2009). These processes lead to phase 1 metabolites of berberine. Phase 2 metabolites form through the conjugation of these metabolites with sulfuric acid or glucuronic acid.
Figure 1. Metabolism of berberine in the liver.
Berberine also undergoes metabolism in the intestines, where its structure and content can be altered by intestinal flora (Han et al., 2021). This alteration occurs through demethoxylation and hydrogenation pathways, involving nitroreductases produced by intestinal flora. Dihydroberberine, a form that can be absorbed in the intestines, is produced through hydrogenation. After absorption, this form oxidizes back to berberine and enters circulation (Han et al., 2021).
Berberine is metabolized into four primary metabolites: berberrubine, thalifendine, demethyleneberberine, and jatrorrhizine (Hu et al., 2018). After oral ingestion, berberine is distributed throughout the body, including the small intestine (undergoing presystemic elimination), liver (where it accumulates), kidneys, muscles, heart, and pancreas. The primary metabolic pathways include oxidative demethylation leading to the production of berberrubine, followed by glucuronidation. After intravenous administration, berberine undergoes oxidative demethylation, resulting in the production of demethyleneberberine, followed by glucuronidation of demethyleneberberine (Hu et al., 2018).
Excretion
Berberine is primarily excreted via urine, feces, and bile (Ma et al., 2013). Due to enterohepatic circulation, excretion via bile is slow. The excretion of berberine varies depending on the route of administration (Han et al., 2021). In rats, oral or gavage administration of berberine results in feces being the primary route of excretion, with the excreted form remaining unchanged as berberine (Feng et al., 2020). Excretion via urine and bile is minimal and primarily in the form of berberine metabolites. Intravenous administration of berberine shows urine as the primary excretion route (Feng et al., 2020).
Types of adipose tissue and browning
Adipose tissue consists of white adipose tissue (WAT) and BAT, composed mainly of white and brown adipocytes, respectively (Kurylowicz & Puzianowska-Kuznicka, 2020). The origins, morphologies, anatomical locations, and nearly all functions of these two types are different from each other (Table 1). White adipocytes consist of a single large lipid droplet with a non-centrally located nucleus, and very few mitochondria (Bargut et al., 2017). Brown adipocytes contain many small lipid droplets, have a centrally located nucleus and are dark in color due to the high number of mitochondria. Adipocyte precursor cells, also known as adipose stem cells, can differentiate into white, beige, or brown adipocytes (Xue et al., 2015b). The expression of myogenic factor-5 determines the difference between white and brown adipocytes. Myogenic factor-5 is associated with thermogenic activities and present in brown adipocyte precursors but not in white adipocytes (Xue et al., 2015b).
Table 1. Characteristics of different adipocytes.
White | Beige | Brown | |
---|---|---|---|
Anatomical location | Subcutaneous, visceral | White depots and supraclavicular | Interscapular, adrenal, neck |
Morphology | Unilocular | Multilocular | Multilocular |
Lipid droplets | Large | Numerous and small | Numerous and small |
Progenitor | Pdgfr-α | Pdgfr-α | Myf5+ |
Main function | Energy storage | Thermogenesis | Thermogenesis |
Mitochondrial biogenesis | Low | Medium | High |
Note:
Pdgfr-α, platelet-derived growth factor receptor alpha; Myf5+, myogenic factor 5-positive.
White adipose tissue begins to develop during the second trimester of pregnancy, and BAT starts to develop towards the end of the second trimester (Cypess, 2022). In newborns, both WAT and BAT are fully developed to perform their functions. WAT is categorized into two main types: visceral and subcutaneous and its primary function is to store energy. Energy stored in the form of triglycerides undergoes lipolysis when required, resulting in the release of fatty acids as fuel. Fifty years ago, it was thought that BAT, which was known to be present in infants, was not present in adults due to insufficient imaging techniques. With the advancement of positron emission tomographic and computed tomographic (PET/CT) imaging technique, active BAT was initially observed in the supraclavicular region of adult humans (Cypess et al., 2009). Later, the presence of BAT was also identified in the neck, axillary, abdominal, and paraspinal regions in adults (Keuper & Jastroch, 2021). There is a high concentration of mitochondria in brown adipocytes. Uncoupling protein 1 (UCP1) in the inner membrane of mitochondria is essential for browning and thermogenesis mechanisms (Kurylowicz & Puzianowska-Kuznicka, 2020). Uncoupling protein 1 releases energy as heat instead of chemical energy by uncoupling mitochondrial respiration from adenosine triphosphate (ATP) synthesis. Therefore, it facilitates thermogenesis and promotes energy expenditure. Consequently, the identification of BAT in adults represents a promising avenue for combating the obesity epidemic. Additionally, “beige/brite” adipocytes are morphologically resemble white adipocytes but exhibit brown adipocyte function under adequate stimuli (Cheng et al., 2021). Phytochemicals such as berberine, resveratrol, and curcumin, and dietary components like fish oil and retinoic acid, along with cold exposure, exercise, and β-adrenergic factors, stimulate beige adipocytes through a process known as “browning” (Cheng et al., 2021; Okla et al., 2017). As browning progresses, beige adipocytes, which have a morphology similar to white adipocytes, begin to perform functions similar to brown adipocytes. As mitochondrial biogenesis and UCP1 expression increase, energy expenditure through thermogenesis will also increase. This is very important in combating obesity, which is mainly caused by an imbalance between energy intake and expenditure. The beige adipocytes lose their brown characteristics and return to the characteristics of the white adipocytes when the stimulus is removed (Ziqubu et al., 2023). This process, called “whitening,” is considered the opposite of browning. It can be seen in obesity and during aging (Graja, Gohlke & Schulz, 2019; Ziqubu et al., 2023).
Energy expenditure and thermogenesis
Obesity typically arises when energy intake exceeds energy expenditure (Lin & Li, 2021). To prevent or treat obesity, energy intake must be reduced and/or energy expenditure must be increased. Dietary interventions are employed to reduce energy intake. To increase energy expenditure, it is crucial to understand the components of total energy expenditure. Approximately 70% comes from the resting metabolic rate, including the thermic effect of the foods, which reflects the energy used for digestion, absorption, and processing of nutrients (Tran et al., 2022). Twenty percent is attributed to energy expenditure from physical activity, divided into non-exercise activity thermogenesis and exercise-induced thermogenesis. Ten percent arises from diet-induced thermogenesis, which occurs in response to excess caloric intake. Lastly, cold-induced thermogenesis is variable and involves mechanisms like shivering thermogenesis and non-shivering thermogenesis (Saito et al., 2020). Among these components, diet-induced thermogenesis and non-shivering thermogenesis are primarily mediated by BAT (Tran et al., 2022). This is because thermogenesis is primarily associated with mitochondria and UCP1, and BAT has a high mitochondrial content (Van Thi-Tuong, Van Vu & Van Pham, 2023). An increase in BAT is expected to enhance these components and, consequently, increase overall energy expenditure.
Browning mechanisms and bat activation
There are two ways to increase thermogenesis through adipose tissue. The first is to increase browning and the second is to increase the already existing BAT activation. Browning can occur in two ways (Kurylowicz & Puzianowska-Kuznicka, 2020). The first is by differentiating from precursor/stem cells and the second is by transdifferentiating from mature adipocytes. Subcutaneous adipocytes are more likely to brown than visceral adipocytes due to their ability to differentiate (Gustafson & Smith, 2015).
Beta-adrenergic receptor activation is considered a key stimulus for browning. The receptors involved in this system may vary between species. For example, in rodents, the β-3 adrenergic receptor (β3-AR) is involved in browning, whereas in humans the β2-AR is involved (Blondin et al., 2020). Cold exposure, similar to β-adrenergic agonists, activates the sympathetic nervous system and releases norepinephrine. When the β-adrenergic receptor is stimulated, it activates cyclic adenosine monophosphate (cAMP) and protein kinase A (PKA).
Protein kinase A activates cAMP response element-binding protein (CREB), p38 mitogen-activated protein kinase (p38-MAPK), and mechanistic target of rapamycin (mTOR) phosphorylation. cAMP response element-binding protein and p38-MAPK increase the transcription of peroxisome proliferator-activated receptor γ-co-activator-1α (PGC-1α), which activates transcription factors that induce mitochondrial biogenesis (Deng et al., 2019). Mechanistic target of rapamycin is also important for mitochondrial biogenesis (Wei et al., 2015).
Sympathetic activation is central to the complex mechanisms that lead to mitochondrial biogenesis, browning, and thermogenesis. Current research highlights several stimuli that enhance these processes, including exercise, specific dietary components, and pharmacological agents. For instance, exercise stimulates the production and release of irisin, which activates the p38-MAPK and extracellular signal-regulated kinase (ERK) pathways, thereby increasing UCP1 expression (Zhang et al., 2014b). Exercise also increases the expression of fibroblast growth factor-21 (FGF21) in the liver and adipose tissue. Its increase in adipose tissue induces UCP1 expression in white adipocytes (Reilly et al., 2021).
Additionally, exercise-induced reactive oxygen species (ROS) and its effects on the nervous system also play a role in adipose tissue browning (Mu et al., 2021). Chronic administration of β-adrenergic agonists and leptin increases sympathetic innervation and stimulates thermogenesis (Jimenez et al., 2003; Wang et al., 2020).
Other factors also play a role in promoting browning. For example, the lipid-lowering agent fenofibrate increases thermogenesis by activating peroxisome proliferator-activated receptor (PPAR)-α (Rachid et al., 2015). Similarly, PPAR agonists, agents that activate the Adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathway, and substances such as nicotine (but not smoking) can stimulate browning by promoting mitochondrial biogenesis (Gaidhu et al., 2009; Yoshida et al., 1999).
Examining the expression and/or protein levels of transcription factors is one of the primary methods used to assess browning. These factors interact with each other, influencing adipogenesis and browning (Table 2). Among these markers, UCP1 is considered a definitive indicator of browning and thermogenic activity. Uncoupling protein 1 activity is regulated by free fatty acids, which enhance its activity, and purine nucleotides, which inhibit it (Macher et al., 2018). Which regulatory protein binds to the gene determines the transcriptional regulation of UCP1 (Villarroya, Peyrou & Giralt, 2017). In the absence of UCP1, lipogenesis and liver steatosis increase (Winn et al., 2017).
Table 2. Some transcription factors in browning.
Transcription factor | Role in browning/thermogenesis | Interactions |
---|---|---|
UCP1 | Key marker of browning and thermogenesis. Activates heat production in adipocytes. | Regulated by free fatty acids (activates) and purine nucleotides (inhibits). Transcription regulated by various factors (e.g., PRDM16, PPARγ) (Macher et al., 2018; Jash et al., 2019). |
PPARγ | Regulates both fat and carbohydrate metabolism. Plays a role in adipogenesis and lipid storage. Can induce browning under certain conditions. | Interacts with LXR and RIP140 to downregulate UCP1. Agonists increase insulin sensitivity and browning but can also increase adiposity (Machado et al., 2022; Wang et al., 2008). |
PGC-1α | Key factor for mitochondrial biogenesis. Stimulates thermogenesis in muscle and brown adipocytes. | Activated by β-adrenergic receptor stimulation. Regulates UCP1 and other thermogenic genes. Stimulated by exercise, cold, and pharmacological agents (Deng et al., 2019; Ishibashi & Seale, 2015). |
CIDEA | Prevents downregulation of UCP1, thus promoting browning and thermogenesis. | Inhibits LXR to prevent UCP1 downregulation (Jash et al., 2019). |
PRDM16 | Activates thermogenic genes in WAT. Essential for the browning of subcutaneous WAT. | Stimulates PGC-1α expression and is necessary for maintaining beige adipocytes. Low PRDM16 expression can reverse browning (Harms et al., 2014; Ishibashi & Seale, 2015). |
Note:
CIDEA, Cell Death-Inducing DNA Fragmentation Factor-Like Effector A; LXR, Liver X receptor; PGC-1α, Peroxisome Proliferator-Activated Receptor γ Co-Activator 1α; PPARγ, Peroxisome Proliferator-Activated Receptor γ; PRDM16, PR Domain Containing 16; RIP140, receptor-interacting protein 140; UCP1, Uncoupling Protein 1; WAT, white adipose tissue.
PPARγ-co-activator-1α is one of the most effective factors in stimulating mitochondrial biogenesis in both muscle and brown adipocytes (Deng et al., 2019). Another important browning factor is cell death-inducing DNA fragmentation factor-like effector A (CIDEA), which prevents the downregulation of UCP1 by inhibiting liver X receptors (LXRs) (Jash et al., 2019). PR domain containing 16 (PRDM16) can activate thermogenic genes in WAT (Ishibashi & Seale, 2015). It activates PGC-1α and is necessary for the browning of subcutaneous WAT. Low expression of PRDM16 can reverse browning and convert beige adipocytes back to white adipocytes (Harms et al., 2014). PR domain containing 16 is thus critical for maintaining beige adipocytes and their thermogenic activity.
Peroxisome proliferator-activated receptor-γ is another key transcription factor, influencing both fat and carbohydrate metabolism. It interacts with LXR and receptor-interacting protein 140 (RIP140) to downregulate UCP1 (Wang et al., 2008). Applying PPARγ agonists can increase insulin sensitivity and browning but may also increase visceral adiposity and unwanted body weight (Machado et al., 2022). Therefore, PPARγ plays a crucial role in both browning and whitening processes.
Cold exposure increases browning partly by enhancing noradrenergic stimulation, which increases iodothyronine deiodinase-2 (DIO2), converting thyroxin (T4) to triiodothyronine (T3) (Kurylowicz & Puzianowska-Kuznicka, 2020). Elevated T3 levels stimulate the sympathetic nervous system, thereby increasing UCP1 expression. Fibroblast growth factor-21 increases UCP1 expression by upregulating PGC-1α (Fisher et al., 2012). It also enhances browning by increasing intracellular Ca++ levels. Forkhead box C2 (FoxC2), which is expressed in adipose tissue, mediates a thermogenic effect via the PKA pathway by increasing the expression of PGC-1α and UCP1 (Kajimura, Seale & Spiegelman, 2010).
Both browning and the activation of brown adipose tissue are triggered by similar stimuli (Kurylowicz & Puzianowska-Kuznicka, 2020). The method used to determine BAT activation is 2-deoxy-2-[18F] fluoro-D-glucose ([18F]FDG)-PET/CT imaging. This imaging technique allows for the tracking of the presence and size of brown adipose tissue (van der Lans et al., 2014).
Effects of berberine on browning and bat activation
A significant portion of berberine’s effects on browning and BAT activation occurs via the AMPK pathway. Beyond this long-studied area, new pathways are being explored, with recent attention on growth differentiation factor 15 (GDF15). The effects of berberine on brown adipose tissue are shown in Fig. 2. Studies investigating the effects of berberine on browning are summarized in Table 3.
Figure 2. The effect of berberine on adipose tissue browning.
Berberine derived from plant sources phosphorylates AMPK, inducing HSL, ATGL, LPL, and CD36, and stimulates lipolysis. Through ACC activation, it prevents CPT1 inhibition. By increasing FGF21 expression, it enhances binding to the FGFR1c receptor. These interactions, which result in increased lipolysis and fatty acids, initiate a mechanism that leads to thermogenesis with UCP1. During inadequate energy intake, LKB1 activates AMPK, which phosphorylates RAPTOR and TSC2, suppressing mTOR (negative impact on BAT activation). By stimulating PGC1α, it plays a crucial role in browning. Independently of AMPK, berberine increases GDF15, promoting its binding to the GFRAL receptor. GFRAL stimulates appetite-related neuropeptides and the TGF- β R in the hypothalamus. The TGF-β R is a regulator of precursor cells that promote browning. Abbreviations: ACC, acetyl-CoA carboxylase; AMPK, AMP-activated protein kinase; ATGL, adipose triglyceride lipase; BAT, brown adipose tissue; CD36, cluster of differentiation 36; CPT1, carnitine palmitoyltransferase 1; FGF21, fibroblast growth factor 21; FGFR1c, fibroblast growth factor receptor 1c; GDF15, Growth differentiation factor 15; GFRAL, glial cell-derived neurotrophic factor family receptor alpha-like; HSL, hormone sensitive lipase; LKB1, liver kinase B1; mTOR, mechanistic target of rapamycin; LPL, lipoprotein lipase; PGC1α, PPARγ-co-activator-1α; RAPTOR, regulatory-associated protein of mTOR; TGF- β R, transforming growth factor β receptors; TSC2, tuberous sclerosis complex 2; UCP1, uncoupling protein 1. Created with BioRender.com.
Table 3. Studies investigating the effects of berberine on browning.
Dose | Duration | Suggested Pathway | Results | References | |
---|---|---|---|---|---|
3T3-L1 preadipocytes | 0.5, 1, 5, 10 µM | 7 days | cAMP/PKA | Adipogenic genes (C/EBP-α, PPARγ) ↓, CREB activity↓ | Zhang et al. (2015) |
HepG2 cells | 5, 10, 15 µM | N/A | AMPK | AMPK phosphorylation↑, ACC↑, fatty acid oxidation↑ | Brusq et al. (2006) |
Male Syrian golden hamsters | 100 mg/kg/day | 10 days | |||
Male C57BL/6J mice | 50 and 100 mg/kg/day | 14 days | GDF15 | Serum GDF15↑, GFRAL↑, appetite↓, UCP1↑ | Li et al. (2023) |
Obese C57BLKS/J-Leprdb/Leprdb male mice and wild-type mice | 5 mg/kg/day | 4 weeks | AMPK- PGC1α | Energy expenditure↑, weight gain↓, BAT activity↑, UCP1↑, PGC-1α↑, CIDEA↑ | Zhang et al. (2014c) |
Male C57BL/6J mice | 100 mg/kg/day | 10 weeks | AMPK | AMPK↑, complex I↓, AMP/ATP↑, ADP/ATP↑ | Turner et al. (2008) |
Male Wistar rats | 4 weeks | ||||
Female Sprague-Dawley rats | 380 mg/kg/day | 2 weeks | N/A | Olanzapine-induce BAT loss↓, weight gain↓, adiposity↓, AMPK↑, UCP1↑, PGC1α↑. Food intake did not change. | Hu et al. (2014) |
Male C57BL/6J mice | 25 and 100 mg/kg/day | 12 weeks | AMPK-SIRT1 | Distribution of BAT↑, thermogenesis↑, body weight↓, AMPK/SIRT1 activation↑, PPAR↑ deacetylation↑, UCP1 expression↑ | Xu et al. (2021) |
Obese C57BLKS/J-Leprdb/Leprdb male mice | 5 mg/kg/day | 26 days | AMPK | Lipogenesis (FAS, PPARγ)↓, expression of browning markers (PGC1α)↑, AMPK activation↑, body weight↓. Food intake did not change. | Lee et al. (2006) |
Wistar rats | 380 mg/kg/day | 2 weeks | |||
Female Sprague-Dawley rats | 380 mg/kg/day | 2 weeks | N/A | Blood lipid levels↓, weight loss↑ | Hu et al. (2012) |
Obese humans | 1.5 g/day | 12 weeks | |||
Male C57BL/6J mice | 1.5 mg/kg/day | 6 weeks | AMPK–PRDM16 | Both in mice and humans: Brown adipocyte differentiation↑, PRDM16 transcription↑In mice: thermogenesis↑, energy expenditure↑AMPK is essential for the browning effect of berberine. | Wu et al. (2019) |
NAFLD patients | 1.5 g/day | 1 month |
Note:
ACC, acetyl-CoA carboxylase; ADP, adenosine diphosphate; AMP, adenosine monophosphate; AMPK, AMP-activated protein kinase; ATP, adenosine triphosphate; BAT, brown adipose tissue; cAMP, cyclic adenosine monophosphate; C/EBP-α, CCAAT/enhancer-binding protein alpha; CIDEA, cell death-inducing DNA fragmentation factor-like effector A; CREB, cAMP response element-binding protein; FAS, fatty acid synthetase; GDF15, Growth differentiation factor 15; GFRAL, glial cell-derived neurotrophic factor family receptor alpha-like; PGC1α, PPARγ-co-activator-1α; PKA, protein kinase A; PPARγ, Peroxisome proliferator-activated receptor-γ; PRDM16, PR domain containing 16; SIRT1, Sirtuin 1; UCP1, uncoupling protein 1.
Adenosine monophosphate-activated protein kinase
AMP-activated protein kinase, a serine/threonine kinase, is one of the key regulators of energy metabolism (Wu & Zou, 2020). Especially in critical situations such as insufficient energy intake, it binds to adenosine diphosphate (ADP) or AMP and regulates key enzymes and proteins in carbohydrate, protein, and lipid metabolism. One of its important targets is PGC1α, which plays a role in mitochondrial homeostasis by converting type IIb muscle fibers into type I and type II fibers that contain more mitochondria (Jager et al., 2007). AMP-activated protein kinase is one of the important factors regulating mitochondrial biogenesis in adipocytes and other tissues (Yang et al., 2016). Mitochondrial biogenesis ensures the production of ATP that meets the increased energy expenditure. Through these mechanisms, AMPK ensures energy homeostasis by increasing ATP production and/or reducing its consumption (van der Vaart, Boon & Houtkooper, 2021).
The activation of AMPK is linked to browning in adipose tissue. When factors stimulating the beta-adrenergic receptor decrease, BAT activation is reduced, leading to decreased AMPK phosphorylation in BAT. AMP-activated protein kinase is influenced by triggers that activate the beta-adrenergic system, such as cold exposure (Mulligan et al., 2007). Lipases play an important role in linking AMPK to brown adipose tissue activation/browning (van der Vaart, Boon & Houtkooper, 2021). AMP-activated protein kinase increases the phosphorylation of hormone-sensitive lipase (HSL) and adipose triglyceride lipase (ATGL), regulates lipoprotein lipase (LPL) and cluster of differentiation 36 (CD36), and stimulates acetyl-CoA carboxylase (ACC) to remove the suppression of carnitine palmitoyltransferase 1 (CPT1). All these actions lead to an increase in fatty acids or their entry into the mitochondria. The fatty acids bind to UCP1, inducing thermogenesis. It also increases UCP1 expression by enhancing PPARγ deacetylation through the AMPK/Sirtuin 1 (SIRT1) pathway (Xu et al., 2021). In addition to activating UCP1, AMPK also stimulates PRDM16 and PPARγ by reducing isocitrate dehydrogenase 2 (IDH2) and α-ketoglutarate (Yang et al., 2016).
Furthermore, berberine increases FGF21 expression via the AMPK pathway, enhancing binding to the FGFR1c receptor. This stimulates both lipolysis and the activation of PGC1α and UCP1 (Fisher et al., 2012; Hirai et al., 2019). When energy intake is limited, liver kinase B1 (LKB1) is activated and upregulates AMPK (Agarwal et al., 2015). AMP-activated protein kinase phosphorylates regulatory-associated protein of mTOR (RAPTOR) and tuberous sclerosis complex 2 (TSC2), thereby suppressing mTOR. For brown adipocyte differentiation, mTOR is crucial. Therefore, it can be inferred that the activation of AMPK does not have a positive effect on BAT development. However, AMPK is important in regulating browning (Perdikari et al., 2018). This is indicated by the inability to brown in the absence of AMPK. AMP-activated protein deficiency prevents brown adipocyte maturation, with the key subunit involved being AMPK-α1 (Perdikari et al., 2017). However, α2 or combined knockdown of other subunits (β1, β2, γ1, and γ3) also prevents browning by reducing UCP1. While studies are showing that berberine is an activator of AMPK, it has been shown specifically to bind to the γ-subunit and activate the AMPK-α-ketoglutarate-PRDM16 pathway (Garcia & Shaw, 2017; Hardie, 2013; Wu et al., 2019). Other mechanisms include the activation of AMPK by berberine through inhibition of complex I, thereby increasing the AMP:ATP and ADP:ATP ratios (Turner et al., 2008).
Growth differentiation factor 15
Growth differentiation factor 15 (GDF15) is a cellular stress biomarker (Li et al., 2023). Growth differentiation factor 15 expression is negatively associated with appetite and food intake in diet-induced obese mice. Some drugs, like metformin, promote weight loss by increasing GDF15 levels as one of their mechanisms of action (Coll et al., 2020). GDF15 binding to its receptor, glial cell-derived neurotrophic factor family receptor alpha-like (GFRAL) leads to reduced appetite in the hypothalamus by inhibiting specific neuropeptides and transforming growth factor-β receptors (TGF-β-R) (Wang et al., 2021; Yang et al., 2017). Transforming growth factor β receptors are regulators of precursor cells that promote browning (Wankhade et al., 2018). In a recent in vitro study, berberine was shown to increase GDF15 expression in adipocytes (Li et al., 2023). Subsequently, when berberine was administered by gavage to obese mice, the circulating levels of GDF15 increased, appetite and food intake decreased, and as a result, the mice lost weight. Although there are studies suggesting that GDF15 can stimulate browning, thermogenesis, and energy expenditure by increasing UCP1 expression, it has also been observed that GDF15 does not affect or even downregulate UCP1 (Choi et al., 2020; Chrysovergis et al., 2014; Li et al., 2023). While GDF15 secretion from adipocytes is normally quite low, berberine intake significantly increased this secretion, particularly in BAT (Li et al., 2023; Wang et al., 2021). Although the effect of increasing GDF15 on UCP1 is not definitive, increasing BAT mass may enhance berberine-induced GDF15 secretion, thereby helping to control body weight.
Future implications
Berberine chloride and sulfate salts are more soluble, but the clinical use of the free form is limited due to its hydrophobic nature, low gastrointestinal absorption, and rapid metabolism. Different strategies have been developed to address the issue of low bioavailability in phytochemicals like berberine (Li et al., 2021; Mirhadi, Rezaee & Malaekeh-Nikouei, 2018; Qiao et al., 2018; Yu et al., 2017). These methods can be broadly categorized into three main approaches. These include:
Changing the administration method (nanotechnology methods)
Altering the chemical structure (organic acid salts of berberine)
Co-administration with P-glycoprotein inhibitors (e.g., glycine, cyclosporine A)
Among these methods, nanotechnology methods are the most used, especially in clinical studies. Nanoparticles are particles with diameters ranging from 10 to 1,000 nm. With these methods, particle size is reduced as much as possible, surface properties are optimized, and the biologically active material is released at an optimal level. This structure, also known as a nano-carrier, helps berberine reach target tissues while preserving its properties. Nano-carriers can include materials such as micelles, carbon-based compounds, liposomes, and polymers (Behl et al., 2022). Some nanoencapsulation methods used to enhance the bioavailability of berberine are shown in Fig. 3.
Figure 3. Some nanoencapsulation methods to improve berberine’s bioavailability.
Solid lipid nanoparticles
In a system developed to increase the bioavailability of berberine and extend its duration of action, berberine is transported within solid lipid nanoparticles (Xue et al., 2015a). When berberine-SLN was administered orally to db/db mice, berberine was stored in the brain, liver, and jejunum. In this system, the elimination of berberine from the body was reduced, and its circulating levels were increased. Berberine encapsulated in SLNs reduced triglyceride and alanine transaminase (ALT) concentrations in the liver, and its anti-diabetic effect was enhanced through nanoencapsulation. Additionally, the presence of berberine in the brain demonstrated that SLN-encapsulated berberine could cross the blood-brain barrier. Berberine-SLN was spherical. The efficiency of encapsulation was 58%, with a loading capacity of 4.2%. The particle size measured 76.8 nm, and the zeta potential was 7.87 mV. The bioavailability of orally administered berberine-SLN (50 mg/kg body weight) was higher compared to free berberine. In the study conducted by Xue et al. (2013), the peak plasma concentration of free berberine was reported as 11.1 ± 6.24, whereas that of berberine-SLNs was significantly higher at 44.651 ± 4.77. Similarly, the area under the curve (AUC) values were 56.5 ± 29.61 for free berberine and 113.6 ± 72.93 for berberine-SLNs, indicating a substantial improvement in bioavailability with the SLN formulation (Xue et al., 2013). Berberine chloride-loaded SLNs are used in studies aimed at preventing and treating various health issues, including cancer therapy (Wang et al., 2014; Xue et al., 2013).
Nanostructured lipid carriers
This nano-carrier method was developed to address the limitations of SLNs by replacing some of the solid lipids in the structure with liquid lipids. This modification increased the loading capacity and prevented berberine leakage during storage. Berberine-NLCs are also spherical, with an encapsulation efficiency of 88%, a particle size of 186 nm, a zeta potential of −36.86 mV, and a polydispersity index of 0.108. Berberine-NLC structures are frequently encountered in studies related to liver health, cognitive functions, and various tumors (Gendy et al., 2022; Raju et al., 2021).
Liposomes
Liposomes are nano-carriers composed of cholesterol and phospholipids. Like other lipid-based nanoencapsulation methods, they are spherical. Due to the hydrophobic and hydrophilic properties of phospholipids, liposomes are used to deliver antibacterial, antifungal, anticancer, and anti-inflammatory drugs, as well as phytochemicals (Akbarzadeh et al., 2013). The literature contains studies on the use of liposomal berberine for liver diseases, cardiovascular diseases, and certain tumors (Allijn et al., 2017; Calvo et al., 2020; Lin et al., 2013).
Micelles
Micelles are complex structures based on surfactants that use various phosphatidylcholine mixtures. They can be spherical or resemble a disk. Encapsulating berberine in anhydrous reverse micelles (ARM) increases berberine’s oral bioavailability by 2.4 times (Wang et al., 2011). Berberine-loaded micelles were found to increase berberine solubility by 800% and its absorption by 364%. Additionally, the efflux rate of berberine within the micelles decreased from 7.54 to 1.05. This indicates that the inhibition of P-glycoprotein-mediated efflux leads to an increase in the intestinal absorption of berberine (Kwon et al., 2020).
Dendrimers
Dendrimers have a branched structure and are polymeric nano-carriers with many functional groups on their surfaces (Sherje et al., 2018). Due to these characteristics, dendrimers exhibit high efficacy and bioavailability. Their unique branched structure, high solubility in water, ability to neutralize various toxins, antigens, or microorganisms, and the simplicity of their production method make them particularly useful in the field of pharmacology. Berberine encapsulated in polyamidoamine (PAMAM) dendrimers increases its permeability, enhancing its bioavailability and therapeutic effects (An et al., 2023). These dendrimers are biocompatible and safe, making them suitable for use in various medical applications (Gupta et al., 2017). They are commonly employed in cancer research, where they help improve drug delivery by targeting cancer cells more effectively and providing controlled release (Yadav, Semwal & Dewangan, 2023).
Conclusions
Berberine, a herbal compound used in Asia for centuries, has recently gained attention for its health benefits, particularly in weight management. While studies generally report positive effects, the underlying mechanisms remain unclear. Key mechanisms include AMPK pathway activation, increased browning markers like UCP1, and appetite-regulating markers such as GDF15. Given the importance of adipose tissue browning and BAT activation in preventing obesity, berberine’s potential to enhance energy expenditure is critical.
However, its therapeutic potential is limited by low stability and poor bioavailability. For berberine to exert its effects, it must achieve and maintain effective concentrations in circulation when taken orally. Nanotechnological approaches, which improve stability and bioavailability, represent a promising solution. Despite their benefits, these methods face challenges such as high production costs, scalability issues, and regulatory hurdles. Advances in manufacturing techniques and cost-reduction strategies are essential for integrating nanotechnology-based therapies into routine clinical practice.
Future research should focus on developing new methods suitable for oral administration that enhance encapsulation efficiency and loading capacity. Additional clinical trials are needed to address the low bioavailability and insufficient toxicity data, which currently prevent the U.S. Food and Drug Administration (FDA) from classifying berberine as a drug. Comprehensive studies in diverse populations are crucial to fully establish berberine’s efficacy, optimal dosage, and clinical application.
Funding Statement
The authors received no funding for this work.
Additional Information and Declarations
Competing Interests
The authors declare that they have no competing interests.
Author Contributions
Aslıhan Alpaslan Ağaçdiken conceived and designed the experiments, performed the experiments, analyzed the data, prepared figures and/or tables, authored or reviewed drafts of the article, and approved the final draft.
Zeynep Göktaş conceived and designed the experiments, performed the experiments, analyzed the data, authored or reviewed drafts of the article, and approved the final draft.
Data Availability
The following information was supplied regarding data availability:
This is a literature review.
References
- Agarwal et al. (2015).Agarwal S, Bell CM, Rothbart SB, Moran RG. AMP-activated Protein Kinase (AMPK) Control of mTORC1 Is p53- and TSC2-independent in pemetrexed-treated carcinoma cells. Journal of Biological Chemistry. 2015;290(46):27473–27486. doi: 10.1074/jbc.M115.665133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akbarzadeh et al. (2013).Akbarzadeh A, Rezaei-Sadabady R, Davaran S, Joo SW, Zarghami N, Hanifehpour Y, Samiei M, Kouhi M, Nejati-Koshki K. Liposome: classification, preparation, and applications. Nanoscale Research Letters. 2013;8:102. doi: 10.1186/1556-276X-8-102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allijn et al. (2017).Allijn IE, Czarny BMS, Wang X, Chong SY, Weiler M, da Silva AE, Metselaar JM, Lam CSP, Pastorin G, de Kleijn DPV, Storm G, Wang JW, Schiffelers RM. Liposome encapsulated berberine treatment attenuates cardiac dysfunction after myocardial infarction. Journal of Controlled Release. 2017;247:127–133. doi: 10.1016/j.jconrel.2016.12.042. [DOI] [PubMed] [Google Scholar]
- An et al. (2023).An H, Deng X, Wang F, Xu P, Wang N. Dendrimers as nanocarriers for the delivery of drugs obtained from natural products. Polymers (Basel) 2023;15(10):2292. doi: 10.3390/polym15102292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bargut et al. (2017).Bargut TCL, Souza-Mello V, Aguila MB, Mandarim-de-Lacerda CA. Browning of white adipose tissue: lessons from experimental models. Hormone Molecular Biology and Clinical Investigation. 2017;31:e0051. doi: 10.1515/hmbci-2016-0051. [DOI] [PubMed] [Google Scholar]
- Behl et al. (2022).Behl T, Singh S, Sharma N, Zahoor I, Albarrati A, Albratty M, Meraya AM, Najmi A, Bungau S. Expatiating the pharmacological and nanotechnological aspects of the alkaloidal drug berberine: current and future trends. Molecules. 2022;27(12):3705. doi: 10.3390/molecules27123705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blondin et al. (2020).Blondin DP, Nielsen S, Kuipers EN, Severinsen MC, Jensen VH, Miard S, Jespersen NZ, Kooijman S, Boon MR, Fortin M, Phoenix S, Frisch F, Guerin B, Turcotte EE, Haman F, Richard D, Picard F, Rensen PCN, Scheele C, Carpentier AC. Human brown adipocyte thermogenesis is driven by beta2-AR stimulation. Cell Metabolism. 2020;32(2):287–300. doi: 10.1016/j.cmet.2020.07.005. [DOI] [PubMed] [Google Scholar]
- Brusq et al. (2006).Brusq JM, Ancellin N, Grondin P, Guillard R, Martin S, Saintillan Y, Issandou M. Inhibition of lipid synthesis through activation of AMP kinase: an additional mechanism for the hypolipidemic effects of berberine. Journal of Lipid Research. 2006;47(6):1281–1288. doi: 10.1194/jlr.M600020-JLR200. [DOI] [PubMed] [Google Scholar]
- Calvo et al. (2020).Calvo A, Moreno E, Larrea E, Sanmartin C, Irache JM, Espuelas S. Berberine-loaded liposomes for the treatment of leishmania infantum-infected BALB/c mice. Pharmaceutics. 2020;12(9):858. doi: 10.3390/pharmaceutics12090858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng et al. (2021).Cheng L, Wang J, Dai H, Duan Y, An Y, Shi L, Lv Y, Li H, Wang C, Ma Q, Li Y, Li P, Du H, Zhao B. Brown and beige adipose tissue: a novel therapeutic strategy for obesity and type 2 diabetes mellitus. Adipocyte. 2021;10:48–65. doi: 10.1080/21623945.2020.1870060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choi et al. (2020).Choi MJ, Jung SB, Lee SE, Kang SG, Lee JH, Ryu MJ, Chung HK, Chang JY, Kim YK, Hong HJ, Kim H, Kim HJ, Lee CH, Mardinoglu A, Yi HS, Shong M. An adipocyte-specific defect in oxidative phosphorylation increases systemic energy expenditure and protects against diet-induced obesity in mouse models. Diabetologia. 2020;63(4):837–852. doi: 10.1007/s00125-019-05082-7. [DOI] [PubMed] [Google Scholar]
- Chrysovergis et al. (2014).Chrysovergis K, Wang X, Kosak J, Lee SH, Kim JS, Foley JF, Travlos G, Singh S, Baek SJ, Eling TE. NAG-1/GDF-15 prevents obesity by increasing thermogenesis, lipolysis and oxidative metabolism. International Journal of Obesity. 2014;38(12):1555–1564. doi: 10.1038/ijo.2014.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coll et al. (2020).Coll AP, Chen M, Taskar P, Rimmington D, Patel S, Tadross JA, Cimino I, Yang M, Welsh P, Virtue S, Goldspink DA, Miedzybrodzka EL, Konopka AR, Esponda RR, Huang JT, Tung YCL, Rodriguez-Cuenca S, Tomaz RA, Harding HP, Melvin A, Yeo GSH, Preiss D, Vidal-Puig A, Vallier L, Nair KS, Wareham NJ, Ron D, Gribble FM, Reimann F, Sattar N, Savage DB, Allan BB, O’Rahilly S. GDF15 mediates the effects of metformin on body weight and energy balance. Nature. 2020;578(7795):444–448. doi: 10.1038/s41586-019-1911-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cypess (2022).Cypess AM. Reassessing human adipose tissue. New England Journal of Medicine. 2022;386(8):768–779. doi: 10.1056/NEJMra2032804. [DOI] [PubMed] [Google Scholar]
- Cypess et al. (2009).Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, Kuo FC, Palmer EL, Tseng YH, Doria A, Kolodny GM, Kahn CR. Identification and importance of brown adipose tissue in adult humans. New England Journal of Medicine. 2009;360(15):1509–1517. doi: 10.1056/NEJMoa0810780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deng et al. (2019).Deng X, Zhang S, Wu J, Sun X, Shen Z, Dong J, Huang J. Promotion of mitochondrial biogenesis via activation of AMPK-PGC1a signaling pathway by Ginger (Zingiber officinale Roscoe) extract, and its major active component 6-Gingerol. Journal of Food Science. 2019;84(8):2101–2111. doi: 10.1111/1750-3841.14723. [DOI] [PubMed] [Google Scholar]
- Feng et al. (2019).Feng X, Sureda A, Jafari S, Memariani Z, Tewari D, Annunziata G, Barrea L, Hassan STS, Smejkal K, Malanik M, Sychrova A, Barreca D, Ziberna L, Mahomoodally MF, Zengin G, Xu S, Nabavi SM, Shen AZ. Berberine in cardiovascular and metabolic diseases: from mechanisms to therapeutics. Theranostics. 2019;9(7):1923–1951. doi: 10.7150/thno.30787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng et al. (2020).Feng X, Wang K, Cao S, Ding L, Qiu F. Pharmacokinetics and excretion of berberine and its nine metabolites in rats. Frontiers in Pharmacology. 2020;11:594852. doi: 10.3389/fphar.2020.594852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fisher et al. (2012).Fisher FM, Kleiner S, Douris N, Fox EC, Mepani RJ, Verdeguer F, Wu J, Kharitonenkov A, Flier JS, Maratos-Flier E, Spiegelman BM. FGF21 regulates PGC-1alpha and browning of white adipose tissues in adaptive thermogenesis. Genes & Development. 2012;26(3):271–281. doi: 10.1101/gad.177857.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaidhu et al. (2009).Gaidhu MP, Fediuc S, Anthony NM, So M, Mirpourian M, Perry RL, Ceddia RB. Prolonged AICAR-induced AMP-kinase activation promotes energy dissipation in white adipocytes: novel mechanisms integrating HSL and ATGL. Journal of Lipid Research. 2009;50(4):704–715. doi: 10.1194/jlr.M800480-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garcia & Shaw (2017).Garcia D, Shaw RJ. AMPK: mechanisms of cellular energy sensing and restoration of metabolic balance. Molecular Cell. 2017;66(6):789–800. doi: 10.1016/j.molcel.2017.05.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gendy et al. (2022).Gendy AM, Elnagar MR, Allam MM, Mousa MR, Khodir AE, El-Haddad AE, Elnahas OS, Fayez SM, El-Mancy SS. Berberine-loaded nanostructured lipid carriers mitigate warm hepatic ischemia/reperfusion-induced lesion through modulation of HMGB1/TLR4/NF-kappaB signaling and autophagy. Biomedicine & Pharmacotherapy. 2022;145:112122. doi: 10.1016/j.biopha.2021.112122. [DOI] [PubMed] [Google Scholar]
- Graja, Gohlke & Schulz (2019).Graja A, Gohlke S, Schulz TJ. Aging of brown and beige/brite adipose tissue. Handbook of Experimental Pharmacology. 2019;251:55–72. doi: 10.1007/978-3-030-10513-6. [DOI] [PubMed] [Google Scholar]
- Gupta et al. (2017).Gupta L, Sharma AK, Gothwal A, Khan MS, Khinchi MP, Qayum A, Singh SK, Gupta U. Dendrimer encapsulated and conjugated delivery of berberine: a novel approach mitigating toxicity and improving in vivo pharmacokinetics. International Journal of Pharmaceutics. 2017;528(1–2):88–99. doi: 10.1016/j.ijpharm.2017.04.073. [DOI] [PubMed] [Google Scholar]
- Gustafson & Smith (2015).Gustafson B, Smith U. Regulation of white adipogenesis and its relation to ectopic fat accumulation and cardiovascular risk. Atherosclerosis. 2015;241(1):27–35. doi: 10.1016/j.atherosclerosis.2015.04.812. [DOI] [PubMed] [Google Scholar]
- Habtemariam (2020).Habtemariam S. Berberine pharmacology and the gut microbiota: a hidden therapeutic link. Pharmacological Research. 2020;155(5):104722. doi: 10.1016/j.phrs.2020.104722. [DOI] [PubMed] [Google Scholar]
- Han et al. (2021).Han Y, Xiang Y, Shi Y, Tang X, Pan L, Gao J, Bi R, Lai X. Pharmacokinetics and pharmacological activities of berberine in diabetes mellitus treatment. Evidence-Based Complementary and Alternative Medicine. 2021;2021(1):1–15. doi: 10.1155/2021/9987097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hardie (2013).Hardie DG. AMPK: a target for drugs and natural products with effects on both diabetes and cancer. Diabetes. 2013;62(7):2164–2172. doi: 10.2337/db13-0368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harms et al. (2014).Harms MJ, Ishibashi J, Wang W, Lim HW, Goyama S, Sato T, Kurokawa M, Won KJ, Seale P. Prdm16 is required for the maintenance of brown adipocyte identity and function in adult mice. Cell Metabolism. 2014;19(4):593–604. doi: 10.1016/j.cmet.2014.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hesari et al. (2018).Hesari A, Ghasemi F, Cicero AFG, Mohajeri M, Rezaei O, Hayat SMG, Sahebkar A. Berberine: a potential adjunct for the treatment of gastrointestinal cancers? Journal of Cellular Biochemistry. 2018;119(12):9655–9663. doi: 10.1002/jcb.27392. [DOI] [PubMed] [Google Scholar]
- Hirai et al. (2019).Hirai T, Mitani Y, Kurumisawa K, Nomura K, Wang W, Nakashima KI, Inoue M. Berberine stimulates fibroblast growth factor 21 by modulating the molecular clock component brain and muscle Arnt-like 1 in brown adipose tissue. Biochemical Pharmacology. 2019;164(1):165–176. doi: 10.1016/j.bcp.2019.04.017. [DOI] [PubMed] [Google Scholar]
- Hu et al. (2012).Hu Y, Ehli EA, Kittelsrud J, Ronan PJ, Munger K, Downey T, Bohlen K, Callahan L, Munson V, Jahnke M, Marshall LL, Nelson K, Huizenga P, Hansen R, Soundy TJ, Davies GE. Lipid-lowering effect of berberine in human subjects and rats. Phytomedicine. 2012;19(10):861–867. doi: 10.1016/j.phymed.2012.05.009. [DOI] [PubMed] [Google Scholar]
- Hu et al. (2014).Hu Y, Young AJ, Ehli EA, Nowotny D, Davies PS, Droke EA, Soundy TJ, Davies GE. Metformin and berberine prevent olanzapine-induced weight gain in rats. PLOS ONE. 2014;9(3):e93310. doi: 10.1371/journal.pone.0093310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu et al. (2018).Hu X, Zhang Y, Xue Y, Zhang Z, Wang J. Berberine is a potential therapeutic agent for metabolic syndrome via brown adipose tissue activation and metabolism regulation. American Journal of Translational Research. 2018;10:3322–3329. [PMC free article] [PubMed] [Google Scholar]
- Ilyas et al. (2020).Ilyas Z, Perna S, Al-Thawadi S, Alalwan TA, Riva A, Petrangolini G, Gasparri C, Infantino V, Peroni G, Rondanelli M. The effect of Berberine on weight loss in order to prevent obesity: a systematic review. Biomedicine & Pharmacotherapy. 2020;127(7):110137. doi: 10.1016/j.biopha.2020.110137. [DOI] [PubMed] [Google Scholar]
- Imenshahidi & Hosseinzadeh (2019).Imenshahidi M, Hosseinzadeh H. Berberine and barberry (Berberis vulgaris): a clinical review. Phytotherapy Research. 2019;33(3):504–523. doi: 10.1002/ptr.6252. [DOI] [PubMed] [Google Scholar]
- Ishibashi & Seale (2015).Ishibashi J, Seale P. Functions of Prdm16 in thermogenic fat cells. Temperature (Austin) 2015;2(1):65–72. doi: 10.4161/23328940.2014.974444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jager et al. (2007).Jager S, Handschin C, St-Pierre J, Spiegelman BM. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(29):12017–12022. doi: 10.1073/pnas.0705070104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jash et al. (2019).Jash S, Banerjee S, Lee MJ, Farmer SR, Puri V. CIDEA transcriptionally regulates UCP1 for britening and thermogenesis in human fat cells. iScience. 2019;20:73–89. doi: 10.1016/j.isci.2019.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jimenez et al. (2003).Jimenez M, Barbatelli G, Allevi R, Cinti S, Seydoux J, Giacobino JP, Muzzin P, Preitner F. Beta 3-adrenoceptor knockout in C57BL/6J mice depresses the occurrence of brown adipocytes in white fat. European Journal of Biochemistry. 2003;270(4):699–705. doi: 10.1046/j.1432-1033.2003.03422.x. [DOI] [PubMed] [Google Scholar]
- Kajimura, Seale & Spiegelman (2010).Kajimura S, Seale P, Spiegelman BM. Transcriptional control of brown fat development. Cell Metabolism. 2010;11(4):257–262. doi: 10.1016/j.cmet.2010.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keuper & Jastroch (2021).Keuper M, Jastroch M. The good and the BAT of metabolic sex differences in thermogenic human adipose tissue. Molecular and Cellular Endocrinology. 2021;533(18):111337. doi: 10.1016/j.mce.2021.111337. [DOI] [PubMed] [Google Scholar]
- Kurylowicz & Puzianowska-Kuznicka (2020).Kurylowicz A, Puzianowska-Kuznicka M. Induction of adipose tissue browning as a strategy to combat obesity. International Journal of Molecular Sciences. 2020;21(17):6241. doi: 10.3390/ijms21176241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwon et al. (2020).Kwon M, Lim DY, Lee CH, Jeon JH, Choi MK, Song IS. Enhanced intestinal absorption and pharmacokinetic modulation of berberine and its metabolites through the inhibition of P-Glycoprotein and Intestinal metabolism in rats using a berberine mixed micelle formulation. Pharmaceutics. 2020;12(9):882. doi: 10.3390/pharmaceutics12090882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee et al. (2006).Lee YS, Kim WS, Kim KH, Yoon MJ, Cho HJ, Shen Y, Ye JM, Lee CH, Oh WK, Kim CT, Hohnen-Behrens C, Gosby A, Kraegen EW, James DE, Kim JB. Berberine, a natural plant product, activates AMP-activated protein kinase with beneficial metabolic effects in diabetic and insulin-resistant states. Diabetes. 2006;55(8):2256–2264. doi: 10.2337/db06-0006. [DOI] [PubMed] [Google Scholar]
- Li et al. (2023).Li C, Leng Q, Li L, Hu F, Xu Y, Gong S, Yang Y, Zhang H, Li X. Berberine ameliorates obesity by inducing GDF15 secretion by brown adipocytes. Endocrinology. 2023;164(4):253. doi: 10.1210/endocr/bqad035. [DOI] [PubMed] [Google Scholar]
- Li et al. (2011).Li Y, Ren G, Wang YX, Kong WJ, Yang P, Wang YM, Li YH, Yi H, Li ZR, Song DQ, Jiang JD. Bioactivities of berberine metabolites after transformation through CYP450 isoenzymes. Journal of Translational Medicine. 2011;9:62. doi: 10.1186/1479-5876-9-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li et al. (2021).Li DD, Yu P, Xu H, Wang ZZ, Xiao W, Zhao LG. Discovery of C-9 modified berberine derivatives as novel lipid-lowering agents. Chemical and Pharmaceutical Bulletin. 2021;69(1):59–66. doi: 10.1248/cpb.c20-00453. [DOI] [PubMed] [Google Scholar]
- Lin et al. (2013).Lin YC, Kuo JY, Hsu CC, Tsai WC, Li WC, Yu MC, Wen HW. Optimizing manufacture of liposomal berberine with evaluation of its antihepatoma effects in a murine xenograft model. International Journal of Pharmaceutics. 2013;441(1–2):381–388. doi: 10.1016/j.ijpharm.2012.11.017. [DOI] [PubMed] [Google Scholar]
- Lin & Li (2021).Lin X, Li H. Obesity: epidemiology, pathophysiology, and therapeutics. Frontiers in Endocrinology. 2021;12:706978. doi: 10.3389/fendo.2021.706978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Linn et al. (2012).Linn YC, Lu J, Lim LC, Sun H, Sun J, Zhou Y, Ng HS. Berberine-induced haemolysis revisited: safety of Rhizoma coptidis and Cortex phellodendri in chronic haematological diseases. Phytotherapy Research. 2012;26(5):682–686. doi: 10.1002/ptr.3617. [DOI] [PubMed] [Google Scholar]
- Liu et al. (2010).Liu YT, Hao HP, Xie HG, Lai L, Wang Q, Liu CX, Wang GJ. Extensive intestinal first-pass elimination and predominant hepatic distribution of berberine explain its low plasma levels in rats. Drug Metabolism and Disposition. 2010;38(10):1779–1784. doi: 10.1124/dmd.110.033936. [DOI] [PubMed] [Google Scholar]
- Liu et al. (2009).Liu Y, Hao H, Xie H, Lv H, Liu C, Wang G. Oxidative demethylenation and subsequent glucuronidation are the major metabolic pathways of berberine in rats. Journal of Pharmaceutical Sciences. 2009;98(11):4391–4401. doi: 10.1002/jps.21721. [DOI] [PubMed] [Google Scholar]
- Ma et al. (2013).Ma JY, Feng R, Tan XS, Ma C, Shou JW, Fu J, Huang M, He CY, Chen SN, Zhao ZX, He WY, Wang Y, Jiang JD. Excretion of berberine and its metabolites in oral administration in rats. Journal of Pharmaceutical Sciences. 2013;102(11):4181–4192. doi: 10.1002/jps.23718. [DOI] [PubMed] [Google Scholar]
- Machado et al. (2022).Machado SA, Pasquarelli-do-Nascimento G, da Silva DS, Farias GR, de Oliveira Santos I, Baptista LB, Magalhaes KG. Browning of the white adipose tissue regulation: new insights into nutritional and metabolic relevance in health and diseases. Nutrition & Metabolism. 2022;19:61. doi: 10.1186/s12986-022-00694-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Macher et al. (2018).Macher G, Koehler M, Rupprecht A, Kreiter J, Hinterdorfer P, Pohl EE. Inhibition of mitochondrial UCP1 and UCP3 by purine nucleotides and phosphate. Biochimica et Biophysica Acta (BBA) - Biomembranes. 2018;1860(3):664–672. doi: 10.1016/j.bbamem.2017.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marlatt & Ravussin (2017).Marlatt KL, Ravussin E. Brown adipose tissue: an update on recent findings. Current Obesity Reports. 2017;6(4):389–396. doi: 10.1007/s13679-017-0283-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mayoral et al. (2020).Mayoral LP, Andrade GM, Mayoral EP, Huerta TH, Canseco SP, Rodal Canales FJ, Cabrera-Fuentes HA, Cruz MM, Perez Santiago AD, Alpuche JJ, Zenteno E, Ruiz HM, Cruz RM, Jeronimo JH, Perez-Campos E. Obesity subtypes, related biomarkers & heterogeneity. Indian Journal of Medical Research. 2020;151(1):11–21. doi: 10.4103/ijmr.IJMR_1768_17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mirhadi, Rezaee & Malaekeh-Nikouei (2018).Mirhadi E, Rezaee M, Malaekeh-Nikouei B. Nano strategies for berberine delivery, a natural alkaloid of Berberis. Biomedicine & Pharmacotherapy. 2018;104(2):465–473. doi: 10.1016/j.biopha.2018.05.067. [DOI] [PubMed] [Google Scholar]
- Mu et al. (2021).Mu WJ, Zhu JY, Chen M, Guo L. Exercise-mediated browning of white adipose tissue: its significance, mechanism and effectiveness. International Journal of Molecular Sciences. 2021;22(21):11512. doi: 10.3390/ijms222111512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mulligan et al. (2007).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. The Journal of Physiology. 2007;580(2):677–684. doi: 10.1113/jphysiol.2007.128652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okla et al. (2017).Okla M, Kim J, Koehler K, Chung S. Dietary factors promoting brown and beige fat development and thermogenesis. Advances in Nutrition. 2017;8(3):473–483. doi: 10.3945/an.116.014332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park, Jung & Shim (2020).Park HJ, Jung E, Shim I. Berberine for appetite suppressant and prevention of obesity. BioMed Research International. 2020;2020(1):3891806. doi: 10.1155/2020/3891806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perdikari et al. (2017).Perdikari A, Kulenkampff E, Rudigier C, Neubauer H, Luippold G, Redemann N, Wolfrum C. A high-throughput, image-based screen to identify kinases involved in brown adipocyte development. Science Signaling. 2017;10(466):R473. doi: 10.1126/scisignal.aaf5357. [DOI] [PubMed] [Google Scholar]
- Perdikari et al. (2018).Perdikari A, Leparc GG, Balaz M, Pires ND, Lidell ME, Sun W, Fernandez-Albert F, Muller S, Akchiche N, Dong H, Balazova L, Opitz L, Roder E, Klein H, Stefanicka P, Varga L, Nuutila P, Virtanen KA, Niemi T, Taittonen M, Rudofsky G, Ukropec J, Enerback S, Stupka E, Neubauer H, Wolfrum C. BATLAS: deconvoluting brown adipose tissue. Cell Reports. 2018;25(3):784–797. doi: 10.1016/j.celrep.2018.09.044. [DOI] [PubMed] [Google Scholar]
- Pirillo & Catapano (2015).Pirillo A, Catapano AL. Berberine, a plant alkaloid with lipid- and glucose-lowering properties: from in vitro evidence to clinical studies. Atherosclerosis. 2015;243(2):449–461. doi: 10.1016/j.atherosclerosis.2015.09.032. [DOI] [PubMed] [Google Scholar]
- Qiao et al. (2018).Qiao X, Wang Q, Wang S, Kuang Y, Li K, Song W, Ye M. A 42-markers pharmacokinetic study reveals interactions of berberine and glycyrrhizic acid in the anti-diabetic Chinese medicine formula Gegen-Qinlian decoction. Frontiers in Pharmacology. 2018;9:622. doi: 10.3389/fphar.2018.00622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rachid et al. (2015).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. Molecular and Cellular Endocrinology. 2015;402(Suppl. 5):86–94. doi: 10.1016/j.mce.2014.12.027. [DOI] [PubMed] [Google Scholar]
- Raju et al. (2021).Raju M, Kunde SS, Auti ST, Kulkarni YA, Wairkar S. Berberine loaded nanostructured lipid carrier for Alzheimer’s disease: design, statistical optimization and enhanced in vivo performance. Life Sciences. 2021;285:119990. doi: 10.1016/j.lfs.2021.119990. [DOI] [PubMed] [Google Scholar]
- Reilly et al. (2021).Reilly SM, Abu-Odeh M, Ameka M, DeLuca JH, Naber MC, Dadpey B, Ebadat N, Gomez AV, Peng X, Poirier B, Walk E, Potthoff MJ, Saltiel AR. FGF21 is required for the metabolic benefits of IKKepsilon/TBK1 inhibition. The Journal of Clinical Investigation. 2021;131(10):e145546. doi: 10.1172/JCI145546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rong et al. (2021).Rong Q, Han B, Li Y, Yin H, Li J, Hou Y. Berberine reduces lipid accumulation by promoting fatty acid oxidation in renal tubular epithelial cells of the diabetic kidney. Frontiers in Pharmacology. 2021;12:729384. doi: 10.3389/fphar.2021.729384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saito et al. (2020).Saito M, Matsushita M, Yoneshiro T, Okamatsu-Ogura Y. Brown adipose tissue, diet-induced thermogenesis, and thermogenic food ingredients: from mice to men. Front Endocrinol (Lausanne) 2020;11:222. doi: 10.3389/fendo.2020.00222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sherje et al. (2018).Sherje AP, Jadhav M, Dravyakar BR, Kadam D. Dendrimers: a versatile nanocarrier for drug delivery and targeting. International Journal of Pharmaceutics. 2018;548(1):707–720. doi: 10.1016/j.ijpharm.2018.07.030. [DOI] [PubMed] [Google Scholar]
- Song, Hao & Fan (2020).Song D, Hao J, Fan D. Biological properties and clinical applications of berberine. Frontiers of Medicine. 2020;14(5):564–582. doi: 10.1007/s11684-019-0724-6. [DOI] [PubMed] [Google Scholar]
- Tan et al. (2013).Tan XS, Ma JY, Feng R, Ma C, Chen WJ, Sun YP, Fu J, Huang M, He CY, Shou JW, He WY, Wang Y, Jiang JD. Tissue distribution of berberine and its metabolites after oral administration in rats. PLOS ONE. 2013;8(10):e77969. doi: 10.1371/journal.pone.0077969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tran et al. (2022).Tran LT, Park S, Kim SK, Lee JS, Kim KW, Kwon O. Hypothalamic control of energy expenditure and thermogenesis. Experimental & Molecular Medicine. 2022;54(4):358–369. doi: 10.1038/s12276-022-00741-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turner et al. (2008).Turner N, Li JY, Gosby A, To SW, Cheng Z, Miyoshi H, Taketo MM, Cooney GJ, Kraegen EW, James DE, Hu LH, Li J, Ye JM. Berberine and its more biologically available derivative, dihydroberberine, inhibit mitochondrial respiratory complex I: a mechanism for the action of berberine to activate AMP-activated protein kinase and improve insulin action. Diabetes. 2008;57(5):1414–1418. doi: 10.2337/db07-1552. [DOI] [PubMed] [Google Scholar]
- van der Lans et al. (2014).van der Lans AA, Wierts R, Vosselman MJ, Schrauwen P, Brans B, van Marken Lichtenbelt WD. Cold-activated brown adipose tissue in human adults: methodological issues. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology. 2014;307(2):R103–R113. doi: 10.1152/ajpregu.00021.2014. [DOI] [PubMed] [Google Scholar]
- van der Vaart, Boon & Houtkooper (2021).van der Vaart JI, Boon MR, Houtkooper RH. The role of AMPK signaling in brown adipose tissue activation. Cells. 2021;10(5):1122. doi: 10.3390/cells10051122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Thi-Tuong, Van Vu & Van Pham (2023).Van Thi-Tuong N, Van Vu V, Van Pham P. Brown adipocyte and browning thermogenesis: metabolic crosstalk beyond mitochondrial limits and physiological impacts. Adipocyte. 2023;12:2237164. doi: 10.1080/21623945.2023.2237164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Villarroya, Peyrou & Giralt (2017).Villarroya F, Peyrou M, Giralt M. Transcriptional regulation of the uncoupling protein-1 gene. Biochimie. 2017;134(11):86–92. doi: 10.1016/j.biochi.2016.09.017. [DOI] [PubMed] [Google Scholar]
- Wang et al. (2021).Wang D, Day EA, Townsend LK, Djordjevic D, Jorgensen SB, Steinberg GR. GDF15: emerging biology and therapeutic applications for obesity and cardiometabolic disease. Nature Reviews Endocrinology. 2021;17(10):592–607. doi: 10.1038/s41574-021-00529-7. [DOI] [PubMed] [Google Scholar]
- Wang et al. (2017).Wang K, Feng X, Chai L, Cao S, Qiu F. The metabolism of berberine and its contribution to the pharmacological effects. Drug Metabolism Reviews. 2017;49(2):139–157. doi: 10.1080/03602532.2017.1306544. [DOI] [PubMed] [Google Scholar]
- Wang et al. (2014).Wang L, Li H, Wang S, Liu R, Wu Z, Wang C, Wang Y, Chen M. Enhancing the antitumor activity of berberine hydrochloride by solid lipid nanoparticle encapsulation. AAPS PharmSciTech. 2014;15(4):834–844. doi: 10.1208/s12249-014-0112-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang et al. (2020).Wang P, Loh KH, Wu M, Morgan DA, Schneeberger M, Yu X, Chi J, Kosse C, Kim D, Rahmouni K, Cohen P, Friedman J. A leptin-BDNF pathway regulating sympathetic innervation of adipose tissue. Nature. 2020;583(7818):839–844. doi: 10.1038/s41586-020-2527-y. [DOI] [PubMed] [Google Scholar]
- Wang et al. (2011).Wang T, Wang N, Song H, Xi X, Wang J, Hao A, Li T. Preparation of an anhydrous reverse micelle delivery system to enhance oral bioavailability and anti-diabetic efficacy of berberine. European Journal of Pharmaceutical Sciences. 2011;44(1–2):127–135. doi: 10.1016/j.ejps.2011.06.015. [DOI] [PubMed] [Google Scholar]
- Wang et al. (2008).Wang H, Zhang Y, Yehuda-Shnaidman E, Medvedev AV, Kumar N, Daniel KW, Robidoux J, Czech MP, Mangelsdorf DJ, Collins S. Liver X receptor alpha is a transcriptional repressor of the uncoupling protein 1 gene and the brown fat phenotype. Molecular and Cellular Biology. 2008;28(7):2187–2200. doi: 10.1128/MCB.01479-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wankhade et al. (2018).Wankhade UD, Lee JH, Dagur PK, Yadav H, Shen M, Chen W, Kulkarni AB, McCoy JP, Finkel T, Cypess AM, Rane SG. TGF-beta receptor 1 regulates progenitors that promote browning of white fat. Molecular Metabolism. 2018;16(Pt. 4):160–171. doi: 10.1016/j.molmet.2018.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei et al. (2015).Wei Y, Zhang YJ, Cai Y, Xu MH. The role of mitochondria in mTOR-regulated longevity. Biological Reviews of the Cambridge Philosophical Society. 2015;90(1):167–181. doi: 10.1111/brv.12103. [DOI] [PubMed] [Google Scholar]
- Winn et al. (2017).Winn NC, Vieira-Potter VJ, Gastecki ML, Welly RJ, Scroggins RJ, Zidon TM, Gaines TL, Woodford ML, Karasseva NG, Kanaley JA, Sacks HS, Padilla J. Loss of UCP1 exacerbates Western diet-induced glycemic dysregulation independent of changes in body weight in female mice. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology. 2017;312(1):R74–R84. doi: 10.1152/ajpregu.00425.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu et al. (2019).Wu L, Xia M, Duan Y, Zhang L, Jiang H, Hu X, Yan H, Zhang Y, Gu Y, Shi H, Li J, Gao X, Li J. Berberine promotes the recruitment and activation of brown adipose tissue in mice and humans. Cell Death & Disease. 2019;10(6):468. doi: 10.1038/s41419-019-1706-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu & Zou (2020).Wu S, Zou MH. AMPK, mitochondrial function, and cardiovascular disease. International Journal of Molecular Sciences. 2020;21(14):4987. doi: 10.3390/ijms21144987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu et al. (2021).Xu Y, Yu T, Ma G, Zheng L, Jiang X, Yang F, Wang Z, Li N, He Z, Song X, Wen D, Kong J, Yu Y, Cao L. Berberine modulates deacetylation of PPARgamma to promote adipose tissue remodeling and thermogenesis via AMPK/SIRT1 pathway. International Journal of Biological Sciences. 2021;17(12):3173–3187. doi: 10.7150/ijbs.62556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xue et al. (2015b).Xue R, Lynes MD, Dreyfuss JM, Shamsi F, Schulz TJ, Zhang H, Huang TL, Townsend KL, Li Y, Takahashi H, Weiner LS, White AP, Lynes MS, Rubin LL, Goodyear LJ, Cypess AM, Tseng YH. Clonal analyses and gene profiling identify genetic biomarkers of the thermogenic potential of human brown and white preadipocytes. Nature Medicine. 2015b;21(7):760–768. doi: 10.1038/nm.3881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xue et al. (2013).Xue M, Yang MX, Zhang W, Li XM, Gao DH, Ou ZM, Li ZP, Liu SH, Li XJ, Yang SY. Characterization, pharmacokinetics, and hypoglycemic effect of berberine loaded solid lipid nanoparticles. International Journal of Nanomedicine. 2013;8:4677–4687. doi: 10.2147/IJN.S51262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xue et al. (2015a).Xue M, Zhang L, Yang MX, Zhang W, Li XM, Ou ZM, Li ZP, Liu SH, Li XJ, Yang SY. Berberine-loaded solid lipid nanoparticles are concentrated in the liver and ameliorate hepatosteatosis in db/db mice. International Journal of Nanomedicine. 2015a;10:5049–5057. doi: 10.2147/IJN. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yadav, Semwal & Dewangan (2023).Yadav D, Semwal BC, Dewangan HK. Grafting, characterization and enhancement of therapeutic activity of berberine loaded PEGylated PAMAM dendrimer for cancerous cell. Journal of Biomaterials Science, Polymer Edition. 2023;34(8):1053–1066. doi: 10.1080/09205063.2022.2155782. [DOI] [PubMed] [Google Scholar]
- Yang et al. (2017).Yang L, Chang CC, Sun Z, Madsen D, Zhu H, Padkjaer SB, Wu X, Huang T, Hultman K, Paulsen SJ, Wang J, Bugge A, Frantzen JB, Norgaard P, Jeppesen JF, Yang Z, Secher A, Chen H, Li X, John LM, Shan B, He Z, Gao X, Su J, Hansen KT, Yang W, Jorgensen SB. GFRAL is the receptor for GDF15 and is required for the anti-obesity effects of the ligand. Nature Medicine. 2017;23(10):1158–1166. doi: 10.1038/nm.4394. [DOI] [PubMed] [Google Scholar]
- Yang et al. (2016).Yang Q, Liang X, Sun X, Zhang L, Fu X, Rogers CJ, Berim A, Zhang S, Wang S, Wang B, Foretz M, Viollet B, Gang DR, Rodgers BD, Zhu MJ, Du M. AMPK/alpha-ketoglutarate axis dynamically mediates DNA demethylation in the Prdm16 promoter and brown adipogenesis. Cell Metabolism. 2016;24(4):542–554. doi: 10.1016/j.cmet.2016.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yarla et al. (2016).Yarla NS, Bishayee A, Sethi G, Reddanna P, Kalle AM, Dhananjaya BL, Dowluru KS, Chintala R, Duddukuri GR. Targeting arachidonic acid pathway by natural products for cancer prevention and therapy. Seminars in Cancer Biology. 2016;40–41:48–81. doi: 10.1016/j.semcancer.2016.02.001. [DOI] [PubMed] [Google Scholar]
- Yoshida et al. (1999).Yoshida T, Sakane N, Umekawa T, Kogure A, Kondo M, Kumamoto K, Kawada T, Nagase I, Saito M. Nicotine induces uncoupling protein 1 in white adipose tissue of obese mice. International Journal of Obesity and Related Metabolic Disorders. 1999;23(6):570–575. doi: 10.1038/sj.ijo.0800870. [DOI] [PubMed] [Google Scholar]
- Yu et al. (2017).Yu F, Ao M, Zheng X, Li N, Xia J, Li Y, Li D, Hou Z, Qi Z, Chen XD. PEG-lipid-PLGA hybrid nanoparticles loaded with berberine-phospholipid complex to facilitate the oral delivery efficiency. Drug Delivery. 2017;24(1):825–833. doi: 10.1080/10717544.2017.1321062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang et al. (2014b).Zhang Y, Li R, Meng Y, Li S, Donelan W, Zhao Y, Qi L, Zhang M, Wang X, Cui T, Yang LJ, Tang D. Irisin stimulates browning of white adipocytes through mitogen-activated protein kinase p38 MAP kinase and ERK MAP kinase signaling. Diabetes. 2014b;63(2):514–525. doi: 10.2337/db13-1106. [DOI] [PubMed] [Google Scholar]
- Zhang et al. (2008).Zhang Y, Li X, Zou D, Liu W, Yang J, Zhu N, Huo L, Wang M, Hong J, Wu P, Ren G, Ning G. Treatment of type 2 diabetes and dyslipidemia with the natural plant alkaloid berberine. The Journal of Clinical Endocrinology & Metabolism. 2008;93(7):2559–2565. doi: 10.1210/jc.2007-2404. [DOI] [PubMed] [Google Scholar]
- Zhang et al. (2015).Zhang J, Tang H, Deng R, Wang N, Zhang Y, Wang Y, Liu Y, Li F, Wang X, Zhou L. Berberine suppresses adipocyte differentiation via decreasing CREB transcriptional activity. PLOS ONE. 2015;10(4):e0125667. doi: 10.1371/journal.pone.0125667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang et al. (2010).Zhang H, Wei J, Xue R, Wu JD, Zhao W, Wang ZZ, Wang SK, Zhou ZX, Song DQ, Wang YM, Pan HN, Kong WJ, Jiang JD. Berberine lowers blood glucose in type 2 diabetes mellitus patients through increasing insulin receptor expression. Metabolism. 2010;59(2):285–292. doi: 10.1016/j.metabol.2009.07.029. [DOI] [PubMed] [Google Scholar]
- Zhang et al. (2020).Zhang L, Wu X, Yang R, Chen F, Liao Y, Zhu Z, Wu Z, Sun X, Wang L. Effects of berberine on the gastrointestinal microbiota. Frontiers in Cellular and Infection Microbiology. 2020;10:588517. doi: 10.3389/fcimb.2020.588517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang et al. (2014a).Zhang Q, Xiao X, Li M, Li W, Yu M, Zhang H, Ping F, Wang Z, Zheng J. Berberine moderates glucose metabolism through the GnRH-GLP-1 and MAPK pathways in the intestine. BMC Complementary and Alternative Medicine. 2014a;14(1):188. doi: 10.1186/1472-6882-14-188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang et al. (2014c).Zhang Z, Zhang H, Li B, Meng X, Wang J, Zhang Y, Yao S, Ma Q, Jin L, Yang J, Wang W, Ning G. Berberine activates thermogenesis in white and brown adipose tissue. Nature Communications. 2014c;5:5493. doi: 10.1038/ncomms6493. [DOI] [PubMed] [Google Scholar]
- Ziqubu et al. (2023).Ziqubu K, Dludla PV, Mthembu SXH, Nkambule BB, Mabhida SE, Jack BU, Nyambuya TM, Mazibuko-Mbeje SE. An insight into brown/beige adipose tissue whitening, a metabolic complication of obesity with the multifactorial origin. Frontiers in Endocrinology. 2023;14:1114767. doi: 10.3389/fendo.2023.1114767. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The following information was supplied regarding data availability:
This is a literature review.