Next Article in Journal
Understanding the Pre-Clinical Stages of Parkinson’s Disease: Where Are We in Clinical and Research Settings?
Previous Article in Journal
Strategic Advances in Targeted Delivery Carriers for Therapeutic Cancer Vaccines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Adipo-Modulation by Turmeric Bioactive Phenolic Components: From Curcuma Plant to Effects

by
Cristina Doriana Marina
1,2,†,
Daniela Puscasiu
3,†,
Corina Flangea
1,4,*,
Tania Vlad
2,3,*,
Adinela Cimporescu
4,
Roxana Popescu
3,4,
Aurica Elisabeta Moatar
5 and
Daliborca Cristina Vlad
1,4
1
Department of Biochemistry and Pharmacology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
2
Doctoral School, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
3
Department of Cell and Molecular Biology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
4
Toxicology and Molecular Biology Department, “Pius Brinzeu” County Emergency Hospital, Liviu Rebreanu Blvd 156, 300723 Timisoara, Romania
5
Clinic of Internal Medicine-Cardiology, Klinikum Freudenstadt, 72250 Freudenstadt, Germany
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(14), 6880; https://doi.org/10.3390/ijms26146880
Submission received: 19 June 2025 / Revised: 8 July 2025 / Accepted: 11 July 2025 / Published: 17 July 2025

Abstract

Obesity is not only an aesthetic problem but also an important comorbidity in metabolic syndrome and other types of pathologies. Currently discussed adjuvants are turmeric and curcumin, used as food supplements. Starting from synthesis in turmeric plant up to the use of turmeric as a spice, a significant amount of turmeric and its derivatives are lost during the processing procedure. In oral administration, the reduced bioavailability of these compounds must be taken into account, an aspect that can be improved by using different combinations and dosages. As for their pharmacodynamic effects, through its antioxidant and anti-inflammatory properties, curcumin improves mitochondrial function and promotes the browning of white adipose tissue. Another mechanism of action of curcumin in weight loss is enzymatic modulation, leading to a decrease in the activity of key enzymes involved in lipogenesis and an increase in the activity of lipolytic enzymes. These properties are enhanced by the synergistic action of the other polyphenols present in turmeric, especially calebin A, p-coumaric acid, caffeic acid and ferulic acid. Summarizing these effects, curcumin is a promising food supplement, opening new directions for further research to discover possibilities to improve or even eliminate the calamity of obesity that is currently wreaking havoc.

Graphical Abstract

1. Introduction

Curcuma Longa, especially its product turmeric, is one of the best-known medicinal plants, frequently used in gastronomy, belonging to the antioxidant-rich ginger family (Zingiberaceae). Turmeric has been used since ancient times as an important ingredient in Ayurvedic medicine and traditional Chinese medicine [1]. The curcuma plant was brought to Europe by Arabian merchants and was even included in the travel journal of the famous explorer Marco Polo [2]. In recent decades, this substance, commonly referred to as the “golden spice”, continues to be used as a culinary ingredient, but modern technology has allowed turmeric to be applied in fields like nutrition and healthcare [3].
Even if turmeric contains a variety of bioactive constituents, current scientific interest in its therapeutic properties focuses on polyphenolic compounds, among which curcumin is the principal active compound [4]. Curcumin has been shown to have a wide spectrum of bioactive and pharmacodynamic properties including antioxidant, immunomodulatory, anti-inflammatory, antibacterial, cardioprotective, hepatoprotective, nephroprotective, neuroprotective, anti-rheumatic, anti-cancer and anti-aging effects [5]. Moreover, curcumin supplementation was found to improve obesity-related parameters, including fasting blood glucose levels and lipids, in metabolic diseases such as obesity, fatty liver and metabolic syndrome [6]. Additionally, this bioactive polyphenol compound has been explored as an adjuvant therapy for polycystic ovary syndrome because of its anti-inflammatory effects, ovulation-inducing properties and ability to improve insulin sensitivity [7]. Furthermore, curcumin has demonstrated significant potential in type 2 diabetes due to its hypoglycemic effect by delaying the absorption of carbohydrates and increasing glucose catabolism [8]. Although preclinical research has shown the therapeutic potential of Curcuma Longa, human clinical trials are crucial to ensure its good applicability in the medical field (Table 1).
The administration of curcumin is generally free of adverse effects. Over the last few years, increased attention has been given to the possibility that turmeric, frequently consumed as a food or dietary supplement, may cause adverse effects. According to recent studies, there is evidence that combining curcuma longa with black pepper, which improves the former’s bioavailability, can lead to liver injury [17,18]. In addition, there is some research in the literature indicating that turmeric can produce allergic reactions such as contact urticaria and contact dermatitis [2,19]. Moreover, excessive intake of turmeric supplements could enhance the risk of developing kidney stones, which is a major concern for susceptible patients [2]. Curcumin has been associated with a significant risk of bleeding among patients who are on warfarin and anticoagulant medications [20]. Other common adverse effects triggered by the use of turmeric supplements include nausea, diarrhea and flatulence [21,22]. Despite these aspects, most of the data from the scientific literature recognize turmeric as being well tolerated, and according to the Food and Drug Administration (FDA), it has a good safety profile [23,24,25,26].
Obesity has become a leading public health problem that affects various demographic groups, regardless of age, ethnic background and economic status [27]. Obesity represents a chronic multifactorial disease induced by excessive accumulation of adipose tissue. This condition, a major component of metabolic syndrome, has been associated with excessive reactive oxygen species (ROS) levels and elevated levels of inflammatory biomarkers, such as interleukins (IL-6, IL-18), tumor necrosis factor-alpha (TNF-α), C-reactive protein, leptin, resistin and monocyte chemoattractant protein-1 (MCP-1) [28,29]. The obesity rate has increased exponentially over the last 50 years, reaching global pandemic proportions [30]. According to the World Health Organization (WHO), it is expected that by 2025, approximately 167 million people, including both adults and children, will experience health problems caused by obesity [31]. The rapidly increasing prevalence of obesity is attracting attention due to its association with multiple comorbidities such as cardiovascular disease, renal failure, several types of cancer, Alzheimer’s disease and diabetes mellitus [32]. Equally importantly, obesity has an undeniable social and economic impact, estimated to be around USD 2.0 trillion and putting a substantial strain on healthcare systems worldwide [33]. Therefore, a comprehensive approach to obesity treatment is needed, among which the use of dietary phytochemicals (for instance, Curcuma Longa) has gained a notable attention.
In this review, we bring to the light obvious molecular and biochemical processes regarding the real involvement of biologically active phenols of turmeric in lipogenesis–lipolysis changes. This aspect is related to the transformations of these biologically active compounds, starting from their synthesis in the Curcuma plant, moving through their absorption and metabolism in the body, their actions on key enzymes and related signaling pathways, as well as the transformations that they undergo throughout this process.

2. From Nature to the Appearance of Biologically Active Compounds: Biosynthesis of Curcumin and Other Related Phenols in Curcuma

To understand a molecule’s structure–effect relationship, especially when it comes to natural compounds, one must look at its overall and natural synthesis phase. This is important because, unlike synthetic products where the exact amount of substance(s) administered is known, in products extracted from biological matrices, the production is influenced by environmental conditions, soil, and precipitation, but also by other species in the vicinity.
The biosynthesis of curcumin and its derivative compounds is carried out via the polyketide pathway, but also via the phenyl-propanoid pathway, without being able to make an exact delimitation, as founded in other plant species such as Cannabis sativa, where, each synthesis pathway yields distinct compounds [34]. The synthesis starts from phenylalanine with the transformation to p-coumaric acid passing through the intermediate step of cinnamic acid. The two steps are carried out by phenylalanine-ammonia lyase and cinnamate-4-hydroxylase. p-coumaric acid can be activated directly to p-coumaryl-CoA or it can go through intermediate steps to caffeic acid, ferulic acid and then activation to feruloyl-CoA. As a side reaction, caffeic acid can be activated to caffeoyl-CoA and then converted to feruloyl-CoA. All activations are catalyzed by 4-coumarate-CoA ligase [35,36,37] (Figure 1a). In the second step, after the precursors formation, they will condense with malonyl-CoA resulting in two diketide compounds, p-coumaroyl-CoA-diketide and feruloyl-CoA-diketide. In the next step, both precursors condense with two diketides obtaining enol forms of Bis-demethoxycurcumin, Demethoxycurcumin, known as curcuminoids, and Curcumin. Following tautomerization, enol forms of Curcumin and derivatives will be transformed into keto forms [36,37,38,39] (Figure 1b).
Trace amounts of curcumin will be transformed into Calebin A in turmeric rhizomes [40]. Calebin A, a stable compound, is actually a ferulate ester obtained from a Baeyer–Villiger reaction, and not from an esterification reaction. Baeyer–Villiger-type monooxygenases catalyze insertion of oxygen into a C-C chain in the vicinity of keto groups. This reaction is carried out in the presence of microorganisms such as Ovatospora brasiliensis, which possess this enzyme [41] (Figure 2). Although it is found in small quantities, some studies attribute antiproliferative, antioxidant and anti-inflammatory properties to it [42,43] probably due to its polyphenol-type structure and also in the context of the global potentiation of all polyphenols, where the main effects are attributed to overall effect of polyphenols (e.g., honey) [44,45,46]. Thus, all intermediate products carrying the phenolic OH group may contribute to the antioxidant properties of turmeric and turmeric extract.

3. From Exogenous Intake to Pharmacokinetics: Pharmacokinetics of Curcumin and Phenolic Derivatives in the Body

A lot is known about turmeric, curcumin, and curcuminoids, but an important aspect would be the amount of the Curcuma plant compounds that actually reaches the tissues.
In most cases, the product intended for consumption, whether it is used as a spice or as a food supplement, uses turmeric. Turmeric is the powder obtained from curcuma rhizomes after the dehydration and drying process (traditionally under sunlight). Dehydration, in general, is a method of preservation and prevention of spoilage [47,48]. Moreover, with drying, the yellow-orange color also intensifies due to curcuminoid pigments. A study showed that sun drying degrades 72% of curcumin and curcuminoids, mainly due to the effect of light and UV radiation, while freeze drying and hot-air drying show a degradation of 55% and 61%, respectively [47]. When covered with a polycarbonate cover at 40 °C, it was observed that it has a protective effect against light and UV radiation, with the lowest degradation of curcumin and curcuminoids [49].
The chemical structure of curcumin and its derivatives results in low water solubility and reduced digestive tract absorption. They are also unstable in aqueous solution and degrade rapidly at physiological pH by autoxidation to bicyclopentadine, vinylether and siroepoxide [50]. At alkaline pH, they decompose to vanillin, ferulic acid, ferulic aldehyde, feruloyl-methane, and trans-6-(4′-hydroxy-3′-methoxyphenyl)-2,4 dioxo-5-hexanal [50,51]. Bioavailability is also reduced by the first hepatic passage. The half-life has been estimated at 10 min. Furthermore, the hepatic metabolism of glucuronide and sulfoconjugates is quick, and they are quickly removed by biliary and renal systems without accumulating in any organ [51,52]. Despite the fact that some authors do not consider bioavailability to be an important factor for therapeutic efficacy [53], there are already a variety of ways to increase bioavailability. Some studies focus on improving bioavailability when turmeric is used as a spice, while others concentrate on the bioavailability offered by pharmaceutical formulations when it is used as a food supplement.
The therapeutic effects of curcumin are promising, but there is an essential factor that must be considered: it has limited bioavailability due to its lipophilic molecule, fast metabolism and rapid systemic clearance [15,54]. In recent years, scientists have tried to find ways to improve the bioavailability of curcumin in order to benefit from its medical properties. One of the most used combinations is the association with piperine (a compound from black pepper), which increase the absorption of curcumin and greatly improve its bioavailability, but requires strict monitoring because it also increases the absorption of other drugs and reduces hepatic metabolism as well [2,15].
Curcumin nanomedicine formulations, including nanoemulsions, solid lipid nanoparticles, nanocomposite, liposomes, micelles and mixed micelles, hydrogels, polymeric nanoparticles and other, have been intensively investigated [55]. Clinical research has demonstrated that the application of nanotechnology is extremely effective as a drug delivery system, eliminating natural obstacles, ensuring better absorption and distribution of curcumin, increasing the circulation time, improving permeability and its resistance to the metabolic degradation [56]. Another efficient strategy to enhance the water solubility of curcumin is the creation of a supramolecular cyclodextrin-curcumin complex [57]. Furthermore, the association with lycopene amplifies the antioxidant capacity, and also co-administration with glycyrrhetinic acid could have antiproliferative effects on liver cancer cells and induce apoptosis [2]. Among these strategies, we can mention the use of metal ions such as Zn2+, Cu2+, Mg2+, and Se2+, which are able to form complexes with curcumin in order to improve its pharmacokinetic properties [2].
Thus, it can be seen that, after the ingestion of natural curcumins such as those found in turmeric, the amount available for pharmacological effects is extremely small. In addition to losses during the processing of turmeric to spice form, there is reduced absorption and low persistence in the body. Improving the natural bioavailability of curcumins by combining them with other spices does not bring a significant contribution. In order to achieve a desirable effect, administration in the form of pharmaceutical formulations is the only option.

4. What Remains to Be Done from Pharmacokinetics to Pharmacodynamics: Studies That Explain the Adipo-Modulation Action of Curcumin and Phenolic Derivatives

The anti-obesity effect has been studied over time, revealing various mechanisms that could be explained. However, the net efficacy of curcumin and its derivatives in the treatment of obesity has been questioned in some studies [58,59]. However, there are numerous hypotheses that confirm the anti-adipose effect of curcumin: anti-inflammatory and antioxidant effect, enzymatic modulator in lipid metabolism, interference with certain cellular signaling pathways in order to accumulate adipose tissue and, last but not least, stimulation of consumption by influencing thermogenesis.

4.1. Modulation of Adipogenesis Through Antioxidant and Anti-Inflammatory Capacity

It is known that the accumulation of ROS reagents causes alterations of the structure of lipids, proteins, and DNA, but also of cellular organisms, especially mitochondria, which are the source of ROS. In fact, it is considered that the major source of ROS production is mitochondria and enzymes of the type NADP-oxidase that transform NADH+ H+ and NADPH+ H+ in NAD+ and NADP+, respectively [60], which will then be converted to reduced forms through nicotinamide nucleotide transhydrogenase (NNT) in mitochondria [61]. In obese people, mitochondrial dysfunction is associated with adipogenesis, reducing the oxidation of free fatty acids and activating biosynthesis of fatty acids with lipid accumulation [29]. An excess of NADP-oxidase becomes dysfunctional because it leads to excessive formation of ROS initiating intense lipid peroxidation in obese individuals [62]. On the other hand, obese individuals show a low antioxidant capacity in the sense of reduced activity of superoxide dismutase (SOD), glutathione peroxidase (GPX), catalase and NNT [63]. It has been observed that curcumin can scavenge superoxide radicals, NO and H2O2 preventing lipid peroxidation due to increased activity of SOD and catalase [64]. In a study conducted on male albino rats fed with a high-fat diet, it was observed that the addition of curcumin to the diet caused an increase in the activity of SOD, GPX, catalase and the amount of reduced glutathione (GSH), as well as a decrease in hepatic myeloperoxidase (MPO), considered a risk marker for obesity [65]. Furthermore, it was demonstrated that curcumin is able to improve mitochondrial respiratory function by upregulating peroxisome-activating receptor γ (PPAR-γ), a nuclear receptor involved in adipogenic differentiation [66]. In this way, it seems that the improvement of mitochondrial function would play an essential role in the conversion of white adipose tissue (WAT) into beige adipose tissue through browning or even into brown adipose tissue (BAT) where, also at the mitochondrial level, uncoupling protein 1 (UCP-1) is stimulated by curcumin, especially in BAT [67,68,69].
Upregulation of PPAR-γ by curcumin in the context of inflammation and ROS generation causes a heterodimerization with retinoid X receptor (RXR), the molecular assembly formed will bind to peroxisome proliferation response element (PPRE) followed by induction of gene transcription and suppression of proinflammatory cytokine release [70,71]. Like any protein structure, PPAR-γ undergoes posttranslational modifications of which the most important is acetylation. PPAR-γ acetylation in BAT causes mitochondrial dysfunction, decreased UCP-1 expression and whitening. Intense acetylation is associated with obesity and aging [72]. In the browning phenomenon, PPAR-γ deacetylation by sirtuin-1 (SIRT-1) is observed [73], curcumin stimulating SIRT-1 thus promoting PPAR-γ deacetylation [72,74]. Furthermore, SIRT-1 deacetylates and activates peroxisome proliferator-activated receptor G coactivator 1-α (PGC-1α) and promotes mitochondrial biogenesis [71], and also reduces the translocation of dynamin-related protein 1 (DRP-1) from cytoplasm to the mitochondria, reducing mitochondrial fragmentation and preserving mitochondrial integrity [75] (Figure 3).
In addition, where there is oxidative stress, there is also inflammation because these two events are associated—ROS produces inflammation and inflammation releases ROS. Inflammation produced by ROS activates the production of nuclear factor-kB (NF-kB) and adipokines such as IL-13, IL-10, IL-4, transforming growth factor β (TGF-β) and also proinflammatory factors such as TNF-α and IL-6 [58,76]. Curcumin’s inhibition of NF-kB results in the depression of these mediators responsible for initiating and maintaining the inflammatory process [77] (Figure 4), an effect also demonstrated in THP-1 cell cultures and foam cell models [78]. In 3T3-L1 adipocytes stimulated with the bacterial endotoxin lipopolysaccharide to induce inflammatory phenomena, curcumin supplementation reduced IL-6 expression, the authors suggesting that the effect would be produced by upregulation of the mammalian target of rapamycin (mTOR) pathway [79]. In addition, curcumin suppresses macrophage inhibitory protein and MCP-1, the major factor for macrophage chemotaxis [80]. Thus, curcumin plays a major role in decreasing WAT infiltration by macrophages, reducing the main factor involved in the release of adipokines [81] and initiation of BAT apoptosis [82].
Microbial infections are also accompanied by inflammatory phenomena. A number of studies demonstrate the antimicrobial effect of turmeric and curcumins. Among microbial species, an antibacterial effect has been observed on some Gram-positive strains, for example, Staphylococcus aureus [83,84,85], Enterococcus fecalis [85], Gram-negative ones such as E. coli [83,85], Helicobacter pylori [83], Proteus mirabilis, Pseudomonas aeruginosa [85] acid-fast, and Mycobacterium tuberculosis [83], and also on Candida species [83,86]. Furthermore, there are studies that have investigated the relationship between curcumin and gut microbiota (GM). Curcumin is degraded in the digestive tract by local bacterial flora into tetrahydrocurcumin, dihydroferulic acid, dihydrocurcumin, and hexahydrocurcumin [1], but also exerts a protective effect on these microorganisms, e.g., Bacteroidaceae, Rikenellaceae [87,88], Bifidobacterium, and Lactobacillus [88]. Thus, curcumin and curcuminoids could indirectly promote weight loss, since GM can regulate monosaccharide absorption from the digestive tract and modulate hepatic lipogenesis [88,89]. It has been shown that HFD depresses beneficial GM and stimulates hepatic steatosis [89]. In a study conducted on B6.V-Lep ob/obJRj mice treated with 0.3% curcumin, it was demonstrated that turmeric promotes the multiplication of Bifidobacterium spp., Lactobacillus and contributes to alleviation in hypertriglyceridemia after 5 weeks [90]. In addition, among metabolites, tetrahydrocurcumin has an important therapeutic role by improving hepatic steatosis in obese C57/BL/6 mice [91].
In this regard, turmeric may reduce oxidative stress and inflammation through the modulation of local pathways that increases the ability of adipose tissue to remove free radicals but also has a noteworthy potential to promote WAT browning. Moreover, through its protective effect on GM, it makes an additional contribution to the improvement of hyperlipidemia, obesity and metabolic syndrome. Starting from this idea, a perspective can be opened to explore the synthesis of pharmaceutical compounds that would be capable to achieve this transformation in people with a tendency towards overweight to accomplish the prophylaxis of obesity and associated consequences.

4.2. Effect on Lipid Metabolism

The accumulation of adipose tissue along with the damage caused by ROS, especially at the mitochondrial level, favors lipogenesis and reduction in β-oxidation. It has been demonstrated that administration of curcumin as a dietary supplement can decrease the plasma concentration of LDL cholesterol and TGL with the reduction in abdominal fat. The observed results are attributed to decrease in fatty acid synthase (FAS) activity and increase in expression of PPAR-α, and carnitine palmitoyltransferase-1 (CPT-1) [64]. The increase in CPT-1 activity will improve the entry of fatty acids into mitochondria while PPAR-α will promote β-oxidation of fatty acids and cholesterol catabolism [92]. In fact, acetyl-CoA is the precursor molecule of both cholesterol and fatty acids that will later be esterified to TGL. Lipid synthesis is also modulated by 5′-AMP-activated protein kinase (AMPK) which reduces fatty acid synthesis by interfering with the activity of FAS, sterol regulating element binding proteins (SREBPs) and 3-hydroxy-3-methylglutaryl-CoA reductase, suppressing the production of cholesterol and fatty acids [58,93,94,95,96]. It is currently considered that AMPK is the control center of lipid metabolism that determines a decrease in acetyl-CoA carboxylase (ACC) activity through its phosphorylation, and reduces conversion of acetyl-CoA to malonyl-CoA, while increase CPT-1 activity [93,94,95,96,97]. When curcumin was administered to Wistar rats, its renal protective effect was observed regarding the development of dyslipidemia through the activation effect on PPAR-α and CPT-1, restoring β-oxidation and also reduces hepatic and renal lipid synthesis [98]. In this regard, following curcumin administration, several studies have reported decreases in plasma total cholesterol, TGL, LDL-cholesterol, VLDL-cholesterol [99,100,101] along with cholesterol efflux from adipocytes [102]. These beneficial effects on the lipid profile were observed at doses of 1200 mg/day, administered for 12 months [101,103].
Changes in plasma concentrations of TGL, total cholesterol and cholesterol fractions also have a positive effect on slowing down/improving atherosclerosis process by reducing the formation of oxidized LDL and also by reducing the formation of foam cells [104,105].
Therefore, doses of 1200 mg/day of curcumin or higher, administered as a food supplement, in addition to enhance lipolysis, reduce lipogenesis and body mass, also intervene in optimizing lipid profile, with a significant potential in improving atherosclerotic phenomena.

4.3. Interferences During Adipogenesis

The plasticity, or more correctly, elasticity (quickly returns to its previous state as soon as the modifying factor has ceased) of adipose tissue makes it responsive to numerous influences, changes being reversible under the action of external factors or medications.
Adult adipose tissue is divided into WAT and BAT. BAT is involved in thermogenesis, containing a large number of mitochondria, while WAT is the main form of energy storage and has few mitochondria [106]. WAT can be visceral or subcutaneous: in the visceral type, the expansion is mainly by hyperplasia, while in the subcutaneous type, hypertrophy predominates [107]. An intermediate category is represented by beige adipose tissue (BeAT) with a higher number of mitochondria and a higher thermogenic activity compared to WAT [107,108]. The transformation of WAT into BAT, initially acquiring the appearance of BeAT, a process called browning, contributes to weight loss and improvement of overweight and obesity. The browning process is accompanied by an increase in the number of multilocular lipid droplets and an increase in UCP-1 expression [109,110].
Adipogenesis is generally controlled by PPARs, CCAAT/enhancer-binding proteins (C/EBPs), SREBPs, and by lipogenic enzymes ACC and FAS [111,112]. Among these factors, the main regulator of adipocyte differentiation is PPAR-γ, which occurs in two forms: PPAR-γ1 expressed in all tissues except muscle and PPAR-γ2 expressed only in adipose tissue and the intestine [71,113]. The most adipogenic form is PPAR-γ, the form induced by HFD: weight gain appears to be caused by PPAR-γ under conditions of excess energy [73]. The activation of PPAR-γ in adipocytes determines lipogenesis with increased expression of C/EBP-α [114] as well as metabolic phenomena such as the release of free fatty acids (FFA) from lipoproteins, intracellular transport of FFA, esterification with glycerol and formation of TGL [114,115,116]. Reductions in PPAR-γ and C/EBP-α expressions by curcumin contribute to the reduction in TGL accumulation in adipocytes and a decrease in plasma concentrations of cholesterol and TGL [112] (Figure 5). Turmeric extract applied to 3T3-L1a cells showed a reduction in the activity of ACC, FAS enzymes as well as downregulation of PPAR-γ and C/EBP-α expressions [111]. Another study on the same type of cells demonstrates the triggering of preadipocyte apoptosis produced by curcumin at high doses of over 30 µM on both extrinsic and intrinsic pathways by activating caspases 8, 3, 9 while at low doses of 15 µM, the expression of PPAR-γ and C/EBP-α was inhibited. The effect was evident after incubation for 24 h, being maximal at 72 h and 48 h, respectively [117].
It is known that adipose tissue exhibits a rebound phenomenon in weight gain after various restrictive diets [118,119,120], as well as a significant capacity for its expansion and proliferation [121,122]. Transition from in vitro experiments to in vivo use in humans would involve chronic administration of turmeric/curcumin in doses that are constantly requires a sustaining high tissue concentrations. Furthermore, continuous administration over long periods of time is necessary to maintain the achieved results, taking into account that adipose tissue exhibits a rebound of weight gain when the inhibitory factor has been removed.

4.4. The Role of Curcumins in Increasing Energy Utilization and Stimulating Thermogenesis

Thermogenesis is actually the main phenomenon by which weight loss is achieved due to increased energy expenditure. Thermogenic adipose tissue is represented by BAT and BeAT, which have a large number of mitochondria and an increased expression of UCP-1 that uncouples ATP production and generates heat [123,124]. Another agent that induces browning is PGC-1α, and the absence of PGC-1α expression stops the transition from WAT to BeAT [125]. Upregulation of PGC-1α defines activation of PPARs [126]. It has been observed that administration of curcumin at a dose of 100 mg/kg body weight to different types of mice that received a high-fat diet (HFD) improves the browning process of WAT, and thermogenesis reduces the size of adipocytes and increases the expression of PGC-1α, PPAR-γ and UCP-1 [127,128]. Another study conducted on Sprague Dawley rats where curcumin was added as a supplement to the daily diet revealed a lower body weight, less adipose tissue and increased energy expenditure. Increased expression of UCP-1 was observed in adipocytes in these rats, and compared to the group without curcumin supplementation, it was observed that UCP-1 (being a protein associated with an increased thermogenesis) also represents an indicator of WAT browning and BAT activation [129].
Browning WAT and activation of thermogenesis are important phenomena triggered by curcumin, especially in patients who cannot improve their physical activity and as an adjunct in weight loss diets. Although there are studies on laboratory animals, the importance of the impact of these phenomena produced by curcumin on humans leaves room for further research and improvements.

5. Minor Amounts–Major Importance: The Role of Calebin A and Intermediates in the Global Effect of Turmeric

Minor components and intermediate compounds can be found in turmeric and may contribute to the overall pharmacological effect. Among these, importance is given to Calebin A as well as the intermediates coumaric acid, caffeic acid and ferulic acid.
Calebin A, a non-toxic compound, can suppress adipogenesis by downregulating PPAR-γ, and C/EBP in adipocytes with inhibition of adipocyte differentiation. Calebin A is also able to induce lipolysis and inhibit lipogenesis by decreasing FAS activity [130]. Regarding its action at the intestinal level, it behaves as a dipeptidyl peptidase IV inhibitor with an effect on the incretin system, running as an effective antidiabetic in type 2 diabetes mellitus but also in the treatment of obesity [131]. C57BL6/6J mice fed with a HFD supplemented with Calebin A showed a reduction in glycemia, adipose tissue and an enhancement of thermogenesis [132]. As an anti-inflammatory effect, Calebin A suppresses production of pro-inflammatory cytokines TNF-α, NF-kB [133] as well as decreases the enzymatic activity of cyclooxygenase and lipoxygenase [134].
p-Coumaric acid, a phenol identified in Curcuma longa rhizome [135] and the major secondary metabolite in leaves and stems of Curcuma longa, found in an amount of 0.431 mg/g [136], is an intermediate compound that contributes to the anti-obesity effect. In C57BL/6J mice fed with a HFD to which p-coumaric acid was added at a dose of 10 mg/kg body weight/day for 16 days, a reduction in WAT and an increase in fatty acid β-oxidation were observed, as well as an improvement in the general lipid profile [137]. Unlike curcumin and Calebin A, p-coumaric acid, caffeic acid and ferulic acid are readily absorbed in the rat intestine via a monocarboxylic acid transporter [138]. If Curcuma longa is fermented, a large amount of caffeic acid is found along with curcuminoids. The fermentation mixture product showed an inhibitory effect on SREBP and ACC as well as a reduction in PPAR-α and CPT-1 activity [139]. In addition, caffeic acid can diminish intracellular lipid accumulation [140]. Ferulic acid can reduce adipocyte size, body weight, and visceral fat accumulation [141]. Ferulic acid supplementation in HFD-fed mice attenuated hypercholesterolemia by increasing cholesterol-7α-hydroxylase expression and enhancing digestive bile acid clearance [142]. Ferulic acid administration at 5 g/kg for 8 weeks reduced body weight in these mice and improved lipid profiles [143]. Similar results were observed at 2 g/kg of ferulic acid supplementation in mice fed with the same diet, with anti-obesity effects alongside antioxidant and anti-inflammatory properties [144].
In addition to curcumin, curcuminoids and intermediates shown in Figure 1 and Figure 2 (p-coumarinic acid, caffeic acid, ferulic acid, calebin), there are a number of products considered minor but with an important contribution to the overall bioactivity of turmeric, in general [145,146,147,148]. These components are summarized in Table 2.
It seems that in natural products, the pharmacodynamic effect cannot be attributed to just one compound, even if it is predominant. It can be observed that the effects of the other components are similar, probably the overall pharmacodynamic response is the result of the synergistic action between the major and minor components.

6. Discussions and Perspectives on the Real Weight Loss Effect

Although in vitro and in vivo studies on laboratory animals show an effect of reducing adiposity and weight loss, curcumin raises questions about its effectiveness in humans.
In a systematic review where a meta-analysis of the literature data was performed and 193 studies were evaluated, of which only 2 remained eligible, it was found that curcumin can be an adjuvant in weight loss only if it is administered in pharmaceutical forms that improve bioavailability [165]. In another review, 1163 articles were analyzed, of which only 18 articles relevant to the authors remained, which included a total of 1604 people. Here, curcumin produced a decrease in body mass index, weight, waist circumference, leptin and an increase in adiponectin was noted, but without any impact on hip circumference [166]. Anthropometric indices showed changes only if 1500 mg curcumin/day was administered but not visible if doses of 1000 mg/day or less were used. In contrast, phytosomal curcumin form used in doses of 200–400 mg/day for 2–4 months showed significant reductions in anthropometric indices [167]. Administration of 1 g turmeric + 1 g black pepper/day together with each breakfast, reduced postprandial blood glucose values and increased the feeling of satiety in the 10 men and 10 women studied [168]. Combination of 500 mg curcumin + 30 mg zinc/day for 90 days administered to 20 patients as tablets, in a dosage form with increased bioavailability, produced an improvement in lipid profile in terms of decreasing TGL, LDL, and increased HDL level, but without influencing total cholesterol values, feeling of hunger, waist and hip circumferences [169].
Despite a large number of research and review articles on the use of dietary supplements in general, their administration is not accompanied by increased efficacy, persistent effects over time and clear results demonstrating significant weight loss. Batsis J.A. et al. [170] underline this in a systematic review, even though, it is well known, dietary supplements, especially antioxidants, have their well-defined role as adjuvants being included in some therapeutic strategies even in some types of cancer [171,172,173,174]. Other limitations of the literature study that addresses research on human subjects consist in the transposition of theoretical knowledge and in vivo experiments on laboratory animals to human applications, where the phenomena observed in controlled laboratory experimental conditions are irrelevant in the population studied. In general, studies conducted on human subjects were carried out for limited periods of time under different types of standardized diets throughout the research period in order to have a reference comparison. In real life, these diets are not adopted for a long time, appetite being variable depending on the season, different physiological or pathological states as well as people financial situation, which nowadays primarily determines the quality of the food consumed. Even if high doses demonstrate significant therapeutic efficacy and, in some cases, modest efficacy, curcumin should not be considered a “magic pill” that causes major weight loss or prevents weight gain in case of an increased and unbalanced food intake. Also, after stopping the administration of curcumin preparations, the gains obtained are probably lost in a variable period of time, taking into account the well-known rebound effect of weight loss.
These promising results obtained by using curcumin in weight loss therapy represent only the beginning of research in the field. Based on the findings presented above, as well as the partial elucidation of some molecular mechanisms, the possibility of further exploration of multiple variants and combinations that would fulfill the desire of most people to be and to stay young, attractive and slim can be opened.

7. Conclusions

Obesity is a major problem nowadays that causes a multitude of challenges and sometimes therapeutic failures. Among the natural alternatives is the use of turmeric or curcumin as a food supplement. Turmeric polyphenols, especially curcumin and its derivatives, have shown to be candidates to be taken into account in terms of their use as adjuvants in anti-obesity treatments. On the one hand, they mediate mitochondrial protection, the place of cellular respiration and β-oxidation of fatty acids, due to their antioxidant and anti-inflammatory properties. Improving this aspect, curcumin can trigger the browning process of WAT. Along with the activation of BAT and BeAT, the thermogenesis process is improved, and displays an important role in weight loss. On the other hand, curcumin has the ability to modify the activity of some enzymes involved in the lipogenesis-lipolysis process with a shift of the balance towards lipolysis. Although at first glance the effects on PPAR-γ are contradictory, as evidenced by the research presented, the action of curcumin is context-dependent, the complete intimate mechanism is not yet being elucidated. Nevertheless, the final product will always lead to improved mitochondrial function, lipolysis and promotion of weight loss. In humans, studies conducted have not accurately reflected the results observed in vitro or using lab animals. This is probably due to the complexity of the enzyme systems and cellular regulatory and signaling pathways, and also to the limited time of the research. Consequently, it will be necessary to identify conditions and formulations that will allow laboratory to find greater similarities in human subjects. However, the results are promising, even if sometimes controversial, but they generate challenges for further research.

Author Contributions

Conceptualization, C.D.M., C.F., T.V. and D.C.V.; methodology, A.C. and R.P.; writing, C.D.M. and D.P.; writing—review, D.C.V. and A.E.M.; visualization, A.E.M. and C.F.; scheme drawing, C.F., A.C. and R.P.; supervision, T.V. and C.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We would like to acknowledge Victor Babes University of Medicine and Pharmacy Timisoara for their support in covering the cost of publication for this review paper.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
FDAFood and Drug Administration
ROSreactive oxygen species
ILinterleukin
TNF-αtumor necrosis factor α
MCP-1monocyte chemoattractant protein-1
WHOWorld Health Organization
CoAcoenzyme A
NNTnicotinamide nucleotide transhydrogenase
SODsuperoxide dismutase
GPXgluthatione peroxidase
GSHreduced glutathione
MPOmyeloperoxidase
PPARperoxisome-activating receptor
WATwhite adipose tissue
BATbrown adipose tissue
BeATbeige adipose tissue
UCP-1uncoupling protein-1
NF-kBnuclear factor-kB
TGF-βtransforming growth factor β
mTORmammalian target of rapamycin
TGLtriglyceride
FASfatty acid synthase
AMPK5′AMP-activated proteinkinase
SREBPsterol regulating element binding proteins
ACCacetyl-CoA carboxylase
C/EBPCCAAT/enhancer-binding proteins
PGC-1αperoxisome proliferator-activated receptor G coactivator 1-α
HFDhigh fat diet
GMgut microbiota
DRP-1dynamin-related protein 1
SIRT-1sirtuin 1
RXRretinoid X receptor
PPREproliferation response element

References

  1. Servida, S.; Piontini, A.; Gori, F.; Tomaino, L.; Moroncini, G.; De Gennaro Colonna, V.; La Vecchia, C.; Vigna, L. Curcumin and gut microbiota: A narrative overview with focus on glycemic control. Int. J. Mol. Sci. 2024, 25, 7710. [Google Scholar] [CrossRef] [PubMed]
  2. Liu, S.; Liu, J.; He, L.; Liu, L.; Cheng, B.; Zhou, F.; Cao, D.; He, Y. A comprehensive review on the benefits and problems of curcumin with respect to human health. Molecules 2022, 27, 4400. [Google Scholar] [CrossRef] [PubMed]
  3. Sharifi-Rad, J.; Rayess, Y.E.; Rizk, A.A.; Sadaka, C.; Zgheib, R.; Zam, W.; Sestito, S.; Rapposelli, S.; Neffe-Skocińska, K.; Zielińska, D.; et al. Turmeric and its major compound curcumin on health: Bioactive effects and safety profiles for food, pharmaceutical, biotechnological and medicinal applications. Front. Pharmacol. 2020, 11, 01021. [Google Scholar] [CrossRef] [PubMed]
  4. Panknin, T.M.; Howe, C.L.; Hauer, M.; Bucchireddigari, B.; Rossi, A.M.; Funk, J.L. Curcumin supplementation and human disease: A scoping review of clinical trials. Int. J. Mol. Sci. 2023, 24, 4476. [Google Scholar] [CrossRef] [PubMed]
  5. Izadi, M.; Sadri, N.; Abdi, A.; Zadeh, M.M.R.; Jalaei, D.; Ghazimoradi, M.M.; Shouri, S.; Tahmasebi, S. Longevity and anti-aging effects of curcumin supplementation. GeroScience 2024, 46, 2933–2950. [Google Scholar] [CrossRef] [PubMed]
  6. Qiu, L.; Gao, C.; Wang, H.; Ren, Y.; Li, J.; Li, M.; Du, X.; Li, W.; Zhang, J. Effects of dietary polyphenol curcumin supplementation on metabolic, inflammatory, and oxidative stress indices in patients with metabolic syndrome: A systematic review and meta-analysis of randomized controlled trials. Front. Endocrinol. 2023, 14, 1216708. [Google Scholar] [CrossRef] [PubMed]
  7. Mallya, P.; Lewis, S.A. Curcumin and its formulations for the treatment of polycystic ovary syndrome: Current insights and future prospects. J. Ovarian Res. 2025, 18, 78. [Google Scholar] [CrossRef] [PubMed]
  8. Alhar, M.S.O.; El-Sofany, W.I.; AlRashidi, A.A.; Hamden, K. Protective effects of isolated curcumin from curcuma longa on key enzymes involved in the insulin signaling pathway and digestive and metabolic enzymes associated with obesity, type 2 diabetes, and hypertension. J. Diab. Res. 2025, 2025, 8050374. [Google Scholar] [CrossRef] [PubMed]
  9. Uchio, R.; Kawasaki, K.; Okuda-Hanafusa, C.; Saji, R.; Muroyama, K.; Murosaki, S.; Yamamoto, Y.; Hirose, Y. Curcuma longa extract improves serum inflammatory markers and mental health in healthy participants who are overweight: A randomized, double-blind, placebo-controlled trial. Nutr. J. 2021, 20, 91. [Google Scholar] [CrossRef] [PubMed]
  10. Calderón-Pérez, L.; Llauradó, E.; Companys, J.; Pla-Pagà, L.; Boqué, N.; Puiggrós, F.; Valls, R.M.; Pedret, A.; Llabrés, J.M.; Arola, L.; et al. Acute Effects of Turmeric Extracts on Knee Joint Pain: A Pilot, Randomized Controlled Trial. J. Med. Food 2021, 24, 436–440. [Google Scholar] [CrossRef] [PubMed]
  11. Yaikwawong, M.; Jansarikit, L.; Jirawatnotai, S.; Chuengsamarn, S. Curcumin extract improves beta cell functions in obese patients with type 2 diabetes: A randomized controlled trial. Nutr. J. 2024, 23, 119. [Google Scholar] [CrossRef] [PubMed]
  12. Gomes, T.L.N.; Zenha, R.S.S.; Antunes, A.H.; Faria, F.R.; Rezende, K.R.; de Souza, E.L.; Mota, J.F. Evaluation of the Impact of Different Doses of Curcuma longa L. on Antioxidant Capacity: A Randomized, Double-Blind, Crossover Pilot Trial. BioMed Res. Int. 2021, 2021, 3532864. [Google Scholar] [CrossRef] [PubMed]
  13. Ahmadi, R.; Salari, S.; Sharifi, M.D.; Reihani, H.; Rostamiani, M.B.; Behmadi, M.; Taherzadeh, Z.; Eslami, S.; Rezayat, S.M.; Jaafari, M.R.; et al. Oral nano-curcumin formulation efficacy in the management of mild to moderate outpatient COVID-19: A randomized triple-blind placebo-controlled clinical trial. Food Sci. Nutr. 2021, 9, 4068–4075. [Google Scholar] [CrossRef] [PubMed]
  14. Heshmati, J.; Moini, A.; Sepidarkish, M.; Morvaridzadeh, M.; Salehi, M.; Palmowski, A.; Mojtahedi, M.F.; Shidfar, F. Effects of curcumin supplementation on blood glucose, insulin resistance and androgens in patients with polycystic ovary syndrome: A randomized double-blind placebo-controlled clinical trial. Phytomedicine 2021, 80, 153395. [Google Scholar] [CrossRef] [PubMed]
  15. Singhal, S.; Hasan, N.; Nirmal, K.; Chawla, R.; Chawla, S.; Kalra, B.S.; Dhal, A. Bioavailable turmeric extract for knee osteoarthritis: A randomized, non-inferiority trial versus paracetamol. Trials 2021, 22, 105. [Google Scholar] [CrossRef] [PubMed]
  16. Jarhahzadeh, M.; Alavinejad, P.; Farsi, F.; Husain, D.; Rezazadeh, A. The effect of turmeric on lipid profile, malondialdehyde, liver echogenicity and enzymes among patients with nonalcoholic fatty liver disease: A randomized double blind clinical trial. Diabetol. Metab. Syndr. 2021, 13, 112. [Google Scholar] [CrossRef] [PubMed]
  17. Halegoua-DeMarzio, D.; Navarro, V.; Ahmad, J.; Avula, B.; Barnhart, H.; Barritt, A.S.; Bonkovsky, H.L.; Fontana, R.J.; Ghabril, M.S.; Hoofnagle, J.H.; et al. Liver Injury Associated with Turmeric-A Growing Problem: Ten Cases from the Drug-Induced Liver Injury Network [DILIN]. Am. J. Med. 2023, 136, 200–206. [Google Scholar] [CrossRef] [PubMed]
  18. Ajitkumar, A.; Mohan, G.; Ghose, M.; Yarrarapu, S.; Afiniwala, S. Drug-Induced Liver Injury Secondary to Turmeric Use. Eur. J. Case Rep. Intern. Med. 2023, 10, 003845. [Google Scholar] [CrossRef] [PubMed]
  19. Palaniappan, V.; Karthikeyan, K. Turmeric: The Yellow Allergen. Indian Dermatol. Online J. 2022, 14, 459–464. [Google Scholar] [CrossRef] [PubMed]
  20. Lopresti, A.L. Potential Role of Curcumin for the Treatment of Major Depressive Disorder. CNS Drugs 2022, 36, 123–141. [Google Scholar] [CrossRef] [PubMed]
  21. Yang, Z.-J.; Huang, S.-Y.; Zhou, D.-D.; Xiong, R.-G.; Zhao, C.-N.; Fang, A.-P.; Zhang, Y.-J.; Li, H.-B.; Zhu, H.-L. Effects and Mechanisms of Curcumin for the Prevention and Management of Cancers: An Updated Review. Antioxidants 2022, 11, 1481. [Google Scholar] [CrossRef] [PubMed]
  22. Yang, C.; Zhu, Q.; Chen, Y.; Ji, K.; Li, S.; Wu, Q.; Pan, Q.; Li, J. Review of the Protective Mechanism of Curcumin on Cardiovascular Disease. Drug Des. Devel. Ther. 2024, 18, 165–192. [Google Scholar] [CrossRef] [PubMed]
  23. Stati, G.; Rossi, F.; Sancilio, S.; Basile, M.; Di Pietro, R. Curcuma longa Hepatotoxicity: A Baseless Accusation. Cases Assessed for Causality Using RUCAM Method. Front. Pharmacol. 2021, 12, 780330. [Google Scholar] [CrossRef] [PubMed]
  24. Zeng, L.; Yu, G.; Hao, W.; Yang, K.; Chen, H. The efficacy and safety of Curcuma longa extract and curcumin supplements on osteoarthritis: A systematic review and meta-analysis. Biosci. Rep. 2021, 41, BSR20210817. [Google Scholar] [CrossRef] [PubMed]
  25. Filardi, T.; Varì, R.; Ferretti, E.; Zicari, A.; Morano, S.; Santangelo, C. Curcumin: Could This Compound Be Useful in Pregnancy and Pregnancy-Related Complications? Nutrients 2020, 12, 3179. [Google Scholar] [CrossRef] [PubMed]
  26. Hassanzadeh, K.; Buccarello, L.; Dragotto, J.; Mohammadi, A.; Corbo, M.; Feligioni, M. Obstacles against the marketing of curcumin as a drug. Int. J. Mol. Sci. 2020, 21, 6619. [Google Scholar] [CrossRef] [PubMed]
  27. Pezzino, S.; Sofia, M.; Mazzone, C.; Litrico, G.; Greco, L.P.; Gallo, L.; La Greca, G.; Latteri, S. Innovative treatments for obesity and NAFLD: A bibliometric study on antioxidants, herbs, phytochemicals, and natural compounds. Heliyon 2024, 10, e35498. [Google Scholar] [CrossRef] [PubMed]
  28. Ellulu, M.S.; Patimah, I.; Khaza’ai, H.; Rahmat, A.; Abed, Y. Obesity and inflammation: The linking mechanism and the complications. Arch. Med. Sci. AMS 2017, 13, 851–863. [Google Scholar] [CrossRef] [PubMed]
  29. Pérez-Torres, I.; Castrejón-Téllez, V.; Soto, M.E.; Rubio-Ruiz, M.E.; Manzano-Pech, L.; Guarner-Lans, V. Oxidative Stress, Plant Natural Antioxidants, and Obesity. Int. J. Mol. Sci. 2021, 22, 1786. [Google Scholar] [CrossRef] [PubMed]
  30. Westbury, S.; Oyebode, O.; van Rens, T.; Barber, T.M. Obesity stigma: Causes, consequences, and potential solutions. Curr. Obes. Rep. 2023, 12, 10–23. [Google Scholar] [CrossRef] [PubMed]
  31. Novelli, G.; Cassadonte, C.; Sbraccia, P.; Biancolella, M. Genetics: A starting point for the prevention and the treatment of obesity. Nutrients 2023, 15, 2782. [Google Scholar] [CrossRef] [PubMed]
  32. Okunogbe, A.; Nugent, R.; Spencer, G.; Powis, J.; Ralston, J.; Wilding, J. Economic impacts of overweight and obesity: Current and future estimates for 161 countries. BMJ Glob. Health 2022, 7, e009773. [Google Scholar] [CrossRef] [PubMed]
  33. Anekwe, C.V.; Jarrell, A.R.; Townsend, M.J.; Gaudier, G.I.; Hiserodt, J.M.; Stanford, F.C. Socioeconomics of obesity. Curr. Obes. Rep. 2020, 9, 272–279. [Google Scholar] [CrossRef] [PubMed]
  34. Flangea, C.; Vlad, D.; Popescu, R.; Dumitrascu, V.; Rata, A.L.; Tryfon, M.E.; Balasoiu, B.; Vlad, C.S. Cannabis: Zone aspects of raw plant components in sport—A narrative review. Nutrients 2025, 17, 861. [Google Scholar] [CrossRef] [PubMed]
  35. Rodrigues, J.L.; Prather, K.L.J.; Kluskens, L.D.; Rodrigues, L.R. Heterologous Production of Curcuminoids. Microbiol. Mol. Biol. Rev. 2015, 79, 39–60. [Google Scholar] [CrossRef] [PubMed]
  36. Lan, T.T.P.; Huy, N.D.; Luong, N.N.; Nghi, N.V.; Tan, T.H.; Quan, L.V.; Loc, N.H. Identification and characterization of genes in the curcuminoid pathway of Curcuma zedoaria Roscoe. Curr. Pharm. Biotechnol. 2018, 19, 839–846. [Google Scholar] [CrossRef] [PubMed]
  37. Ayer, D.K.; Modha, K.; Parekh, V.; Patel, R.; Vadodariya, G.; Ramtekey, V.; Bhuriya, A. Associating gene expressions with curcuminoid biosynthesis in turmeric. J. Genet. Eng. Biotechnol. 2020, 18, 83. [Google Scholar] [CrossRef] [PubMed]
  38. Rodrigues, J.L.; Gomes, D.; Rodrigues, L.R. A combinatorial approach to optimize the production of curcuminoids from tyrosine in Escherichia coli. Front. Bioeng. Biotechnol. 2020, 8, 59. [Google Scholar] [CrossRef] [PubMed]
  39. Hasan, A.A.; Tatarskiy, V.; Kalinina, E. Synthetic pathways and the therapeutic potential of quercetin and curcumin. Int. J. Mol. Sci. 2022, 23, 14413. [Google Scholar] [CrossRef] [PubMed]
  40. Majeed, M.; Nagabhushanam, K.; Devarajan, T.V.; Saklecha, S.; Reddy, S.V.K.; Mundkur, L. A minor metabolite from Curcuma longa effective against metabolic syndrome: Results from a randomized, double-blind, placebo-controlled clinical study. Food Funct. 2023, 14, 4722–4733. [Google Scholar] [CrossRef] [PubMed]
  41. Majeed, A.; Majeed, M.; Thajuddin, N.; Arumugam, S.; Ali, F.; Beede, K.; Adams, S.J.; Gnanamani, M. Bioconversion of curcumin into calebin-A by the endophytic fungus Ovatospora brasiliensis EPE-10 MTCC 25236 associated with Curcuma caesia. AMB Expr. 2019, 9, 79. [Google Scholar] [CrossRef] [PubMed]
  42. Mueller, A.-L.; Brockmueller, A.; Kunnumakkara, A.B.; Shakibaei, M. Calebin A, a compound of turmeric, down-regulates inflammation in tenocytes by NF-κB/Scleraxis Signaling. Int. J. Mol. Sci. 2022, 23, 1695. [Google Scholar] [CrossRef] [PubMed]
  43. Brockmueller, A.; Buhrmann, C.; Shayan, P.; Shakibaei, M. Calebin A modulates inflammatory and autophagy signals for the prevention and treatment of osteoarthritis. Front. Immunol. 2024, 15, 1363947. [Google Scholar] [CrossRef] [PubMed]
  44. Nan, A.; Dumitrascu, V.; Flangea, C.; Dumitrescu, G.; Puscasiu, D.; Vlad, T.; Popescu, R.; Vlad, C. From chemical composition to antiproliferative effects through in vitro studies: Honey, an ancient and modern hot topic remedy. Nutrients 2025, 17, 1595. [Google Scholar] [CrossRef] [PubMed]
  45. Olas, B. Honey and its phenolic compounds as an effective natural medicine for cardiovascular diseases in humans? Nutrients 2020, 12, 283. [Google Scholar] [CrossRef] [PubMed]
  46. Ayoub, W.S.; Zahoor, R.I.; Dar, A.H.; Farooq, S.; Mir, T.A.; Ganaie, T.A.; Srivastava, S.; Pandey, V.K.; Altaf, A. Exploiting the polyphenolic potential of honey in the prevention of chronic diseases. Food Chem. Adv. 2023, 3, 100373. [Google Scholar] [CrossRef]
  47. Chumroenphat, T.; Somboonwatthanakul, I.; Saensouk, S.; Siriamornpun, S. Changes in curcuminoids and chemical components of turmeric (Curcuma longa L.) under freeze-drying and low-temperature drying methods. Food Chem. 2021, 339, 128121. [Google Scholar] [CrossRef] [PubMed]
  48. Salem, M.A.; El-Shiekh, R.A.; Fernie, A.R.; Alseekh, S.; Zayed, A. Metabolomics-based profiling for quality assessment and revealing the impact of drying of Turmeric (Curcuma longa L.). Sci. Rep. 2022, 12, 10288. [Google Scholar] [CrossRef] [PubMed]
  49. Komonsing, N.; Reyer, S.; Khuwijitjaru, P.; Mahayothee, B.; Müller, J. Drying behavior and curcuminoids changes in turmeric slices during drying under simulated solar radiation as influenced by different transparent cover materials. Foods 2022, 11, 696. [Google Scholar] [CrossRef] [PubMed]
  50. Dei Cas, M.; Ghidoni, R. Dietary Curcumin: Correlation between bioavailability and health potential. Nutrients 2019, 11, 2147. [Google Scholar] [CrossRef] [PubMed]
  51. Hegde, M.; Girisa, S.; BharathwajChetty, B.; Vishwa, R.; Kunnumakkara, A.B. Curcumin formulations for better bioavailability: What we learned from clinical trials thus far? ACS Omega 2023, 8, 10713–10746. [Google Scholar] [CrossRef] [PubMed]
  52. Bertoncini-Silva, C.; Vlad, A.; Ricciarelli, R.; Giacomo Fassini, P.; Suen, V.M.M.; Zingg, J.-M. Enhancing the bioavailability and bioactivity of curcumin for disease prevention and treatment. Antioxidants 2024, 13, 331. [Google Scholar] [CrossRef] [PubMed]
  53. Bučević Popović, V.; Karahmet Farhat, E.; Banjari, I.; Jeličić Kadić, A.; Puljak, L. Bioavailability of oral curcumin in systematic reviews: A methodological study. Pharmaceuticals 2024, 17, 164. [Google Scholar] [CrossRef] [PubMed]
  54. Tabanelli, R.; Brogi, S.; Calderone, V. Improving curcumin bioavailability: Current strategies and future perspectives. Pharmaceutics 2021, 13, 1715. [Google Scholar] [CrossRef] [PubMed]
  55. Sohn, S.I.; Priya, A.; Balasubramaniam, B.; Muthuramalingam, P.; Sivasankar, C.; Selvaraj, A.; Valliammai, A.; Jothi, R.; Pandian, S. Biomedical Applications and Bioavailability of Curcumin-An Updated Overview. Pharmaceutics 2021, 13, 2102. [Google Scholar] [CrossRef] [PubMed]
  56. Laurindo, L.F.; de Carvalho, G.M.; de Oliveira Zanuso, B.; Figueira, M.E.; Direito, R.; de Alvares Goulart, R.; Buglio, D.S.; Barbalho, S.M. Curcumin-based nanomedicines in the treatment of inflammatory and immunomodulated diseases: An evidence-based comprehensive review. Pharmaceutics 2023, 15, 229. [Google Scholar] [CrossRef] [PubMed]
  57. Li, J.; Xu, F.; Dai, Y.; Zhang, J.; Shi, Y.; Lai, D.; Sriboonvorakul, N.; Hu, J. A review of cyclodextrin encapsulation and intelligent response for the release of curcumin. Polymers 2022, 14, 5421. [Google Scholar] [CrossRef] [PubMed]
  58. Kasprzak-Drozd, K.; Oniszczuk, T.; Gancarz, M.; Kondracka, A.; Rusinek, R.; Oniszczuk, A. Curcumin and weight loss: Does it work? Int. J. Mol. Sci. 2022, 23, 639. [Google Scholar] [CrossRef] [PubMed]
  59. Varì, R.; Scazzocchio, B.; Silenzi, A.; Giovannini, C.; Masella, R. Obesity-associated inflammation: Does curcumin exert a beneficial role? Nutrients 2021, 13, 1021. [Google Scholar] [CrossRef] [PubMed]
  60. Masenga, S.K.; Kabwe, L.S.; Chakulya, M.; Kirabo, A. Mechanisms of oxidative stress in metabolic syndrome. Int. J. Mol. Sci. 2023, 24, 7898. [Google Scholar] [CrossRef] [PubMed]
  61. Corkey, B.E. Reactive oxygen species: Role in obesity and mitochondrial energy efficiency. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2023, 378, 20220210. [Google Scholar] [CrossRef] [PubMed]
  62. Ahmed, B.; Sultana, R.; Greene, M.W. Adipose tissue and insulin resistance in obese. Biomed. Pharmacother. 2021, 137, 111315. [Google Scholar] [CrossRef] [PubMed]
  63. Heindel, J.J.; Lustig, R.H.; Howard, S.; Corkey, B.E. Obesogens: A unifying theory for the global rise in obesity. Int. J. Obes. 2024, 48, 449–460. [Google Scholar] [CrossRef] [PubMed]
  64. Cheng, M.; Ding, F.; Li, L.; Dai, C.; Sun, X.; Xu, J.; Chen, F.; Li, M.; Li, X. Exploring the role of curcumin in mitigating oxidative stress to alleviate lipid metabolism disorders. Front. Pharmacol. 2025, 16, 1517174. [Google Scholar] [CrossRef] [PubMed]
  65. Hassan, M.H.; Awadalla, E.A.; Abd El-Kader, A.E.M.; Seifeldin, E.A.; Mahmoud, M.A.; Muddathir, A.R.M.; Abdelsadik, A. Antitoxic effects of curcumin against obesity-induced multi-organs’ biochemical and histopathological abnormalities in an animal model. Evid. Based Complement. Alternat. Med. 2022, 2022, 9707278. [Google Scholar] [CrossRef] [PubMed]
  66. Zhao, D.; Pan, Y.; Yu, N.; Bai, Y.; Ma, R.; Mo, F.; Zuo, J.; Chen, B.; Jia, Q.; Zhang, D.; et al. Curcumin improves adipocytes browning and mitochondrial function in 3T3-L1 cells and obese rodent model. R. Soc. Open Sci. 2021, 8, 200974. [Google Scholar] [CrossRef] [PubMed]
  67. Jin, T. Mechanisms underlying the metabolic beneficial effect of curcumin intervention: Beyond anti-inflammation and anti-oxidative stress. Obes. Med. 2019, 13, 1–5. [Google Scholar] [CrossRef]
  68. Kim, S.W.; Choi, J.H.; Mukherjee, R.; Hwang, K.C.; Yun, J.W. Proteomic identification of fat-browning markers in cultured white adipocytes treated with curcumin. Mol. Cell. Biochem. 2016, 415, 51–66. [Google Scholar] [CrossRef] [PubMed]
  69. Kim, M.B.; Lee, J.; Lee, J.Y. Targeting mitochondrial dysfunction for the prevention and treatment of metabolic disease by bioactive food components. J. Lipid Atheroscler. 2024, 13, 306–327. [Google Scholar] [CrossRef] [PubMed]
  70. Jacob, A.; Wu, R.; Zhou, M.; Wang, P. Mechanism of the Anti-inflammatory Effect of Curcumin: PPAR-gamma Activation. PPAR Res. 2007, 2007, 89369. [Google Scholar] [CrossRef] [PubMed]
  71. Sun, C.; Mao, S.; Chen, S.; Zhang, W.; Liu, C. PPARs-Orchestrated Metabolic Homeostasis in the Adipose Tissue. Int. J. Mol. Sci. 2021, 22, 8974. [Google Scholar] [CrossRef] [PubMed]
  72. He, Y.; Zhang, R.; Yu, L.; Zahr, T.; Li, X.; Kim, T.-W.; Qiang, L. PPARγ Acetylation in Adipocytes Exacerbates BAT Whitening and Worsens Age-Associated Metabolic Dysfunction. Cells 2023, 12, 1424. [Google Scholar] [CrossRef] [PubMed]
  73. Ma, X.; Wang, D.; Zhao, W.; Xu, L. Deciphering the Roles of PPARγ in Adipocytes via Dynamic Change of Transcription Complex. Front. Endocrinol. 2018, 9, 473. [Google Scholar] [CrossRef] [PubMed]
  74. Prasetyawan, S.; Safitri, A.; Atho’illah, M.F.; Rahayu, S. Computational evaluation of bioactive compounds in Curcuma zanthorrhiza targeting SIRT1 and NFκB. BioTechnologia 2023, 104, 171–182. [Google Scholar] [CrossRef] [PubMed]
  75. Hou, D.; Liao, H.; Hao, S.; Liu, R.; Huang, H.; Duan, C. Curcumin simultaneously improves mitochondrial dynamics and myocardial cell bioenergy after sepsis via the SIRT1-DRP1/PGC-1α pathway. Heliyon 2024, 10, e28501. [Google Scholar] [CrossRef] [PubMed]
  76. Cox, F.F.; Misiou, A.; Vierkant, A.; Ale-Agha, N.; Grandoch, M.; Haendeler, J.; Altschmied, J. Protective effects of curcumin in cardiovascular diseases—Impact on oxidative stress and mitochondria. Cells 2022, 11, 342. [Google Scholar] [CrossRef] [PubMed]
  77. Zia, A.; Farkhondeh, T.; Pourbagher-Shahri, A.M.; Samarghandian, S. The role of curcumin in aging and senescence: Molecular mechanisms. Biomed. Pharmacother. 2021, 134, 111119. [Google Scholar] [CrossRef] [PubMed]
  78. Zhong, Y.; Liu, C.; Feng, J.; Li, J.F.; Fan, Z.C. Curcumin affects ox-LDL-induced IL-6, TNF-α, MCP-1 secretion and cholesterol efflux in THP-1 cells by suppressing the TLR4/NF-κB/miR33a signaling pathway. Exp. Therap. Med. 2020, 20, 1856–1870. [Google Scholar] [CrossRef] [PubMed]
  79. Islam, T.; Scoggin, S.; Gong, X.; Zabet-Moghaddam, M.; Kalupahana, N.S.; Moustaid-Moussa, N. Anti-Inflammatory mechanisms of curcumin and its metabolites in white adipose tissue and cultured adipocytes. Nutrients 2024, 16, 70. [Google Scholar] [CrossRef] [PubMed]
  80. Sadeghi, M.; Dehnavi, S.; Asadirad, A.; Xu, S.; Majeed, M.; Jamialahmadi, T.; Johnston, T.P.; Sahebkar, A. Curcumin and chemokines: Mechanism of action and therapeutic potential in inflammatory diseases. Inflammopharmacology 2023, 31, 1069–1093. [Google Scholar] [CrossRef] [PubMed]
  81. Ayub, H.; Islam, M.; Saeed, M.; Ahmad, H.; Al-Asmari, F.; Ramadan, M.F.; Alissa, M.; Arif, M.A.; Rana, M.U.J.; Subtain, M.; et al. On the health effects of curcumin and its derivatives. Food Sci. Nutr. 2024, 12, 8623–8650. [Google Scholar] [CrossRef] [PubMed]
  82. Scarano, F.; Gliozzi, M.; Zito, M.C.; Guarnieri, L.; Carresi, C.; Macrì, R.; Nucera, S.; Scicchitano, M.; Bosco, F.; Ruga, S.; et al. Potential of nutraceutical supplementation in the modulation of white and brown fat tissues in obesity-associated disorders: Role of inflammatory signalling. Int. J. Mol. Sci. 2021, 22, 3351. [Google Scholar] [CrossRef] [PubMed]
  83. Hussain, Y.; Alam, W.; Ullah, H.; Dacrema, M.; Daglia, M.; Khan, H.; Arciola, C.R. Antimicrobial Potential of Curcumin: Therapeutic Potential and Challenges to Clinical Applications. Antibiotics 2022, 11, 322. [Google Scholar] [CrossRef] [PubMed]
  84. Imaizumi, U.; Inaba, K.; Kurahashi, A.; Kuroda, H.; Sanuki, T.; Yoshida, A.; Yoshino, F.; Hamada, N. Effectiveness of curcumin-based antimicrobial photodynamic therapy against Staphylococcus aureus. J. Oral Sci. 2023, 65, 270–274. [Google Scholar] [CrossRef] [PubMed]
  85. Adamczak, A.; Ożarowski, M.; Karpiński, T.M. Curcumin, a Natural Antimicrobial Agent with Strain-Specific Activity. Pharmaceuticals 2020, 13, 153. [Google Scholar] [CrossRef] [PubMed]
  86. Kubizna, M.; Dawiec, G.; Wiench, R. Efficacy of Curcumin-Mediated Antimicrobial Photodynamic Therapy on Candida spp.—A Systematic Review. Int. J. Mol. Sci. 2024, 25, 8136. [Google Scholar] [CrossRef] [PubMed]
  87. Shabbir, U.; Rubab, M.; Daliri, E.B.-M.; Chelliah, R.; Javed, A.; Oh, D.-H. Curcumin, Quercetin, Catechins and Metabolic Diseases: The Role of Gut Microbiota. Nutrients 2021, 13, 206. [Google Scholar] [CrossRef] [PubMed]
  88. Zhu, J.; He, L. The Modulatory Effects of Curcumin on the Gut Microbiota: A Potential Strategy for Disease Treatment and Health Promotion. Microorganisms 2024, 12, 642. [Google Scholar] [CrossRef] [PubMed]
  89. Lamichhane, G.; Olawale, F.; Liu, J.; Lee, D.-Y.; Lee, S.-J.; Chaffin, N.; Alake, S.; Lucas, E.A.; Zhang, G.; Egan, J.M.; et al. Curcumin Mitigates Gut Dysbiosis and Enhances Gut Barrier Function to Alleviate Metabolic Dysfunction in Obese, Aged Mice. Biology 2024, 13, 955. [Google Scholar] [CrossRef] [PubMed]
  90. Neyrinck, A.M.; Sánchez, C.R.; Rodriguez, J.; Cani, P.D.; Bindels, L.B.; Delzenne, N.M. Prebiotic Effect of Berberine and Curcumin Is Associated with the Improvement of Obesity in Mice. Nutrients 2021, 13, 1436. [Google Scholar] [CrossRef] [PubMed]
  91. Peng, S.; Meng, M.; Luo, P.; Liu, J.; Wang, J.; Chen, Y. Tetrahydrocurcumin Alleviates Metabolic Dysfunction-Associated Steatohepatitis in Mice by Regulating Serum Lipids, Bile Acids, and Gut Microbiota. Int. J. Mol. Sci. 2025, 26, 895. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, M.; Wang, K.; Liao, X.; Hu, H.; Chen, L.; Meng, L.; Gao, W.; Li, Q. Carnitine palmitoyltransferase system: A new target for anti-inflammatory and anticancer therapy? Front. Pharmacol. 2021, 12, 760581. [Google Scholar] [CrossRef] [PubMed]
  93. Cao, S.; Liu, M.; Han, Y.; Li, S.; Zhu, X.; Li, D.; Shi, Y.; Liu, B. Effects of saponins on lipid metabolism: The gut–liver axis plays a key role. Nutrients 2024, 16, 1514. [Google Scholar] [CrossRef] [PubMed]
  94. El Bounkari, O.; Zan, C.; Yang, B.; Ebert, S.; Wagner, J.; Bugar, E.; Kramer, N.; Bourilhon, P.; Kontos, C.; Zarwel, M.; et al. An atypical atherogenic chemokine that promotes advanced atherosclerosis and hepatic lipogenesis. Nat. Commun. 2025, 16, 2297. [Google Scholar] [CrossRef] [PubMed]
  95. Tang, H.; Zhang, Y.; Zhao, D.; Guo, M.; Yuan, X.; Wang, X. Unlocking the lipid code: SREBPs as key drivers in gastrointestinal tumour metabolism. Lipids Health Dis. 2025, 24, 190. [Google Scholar] [CrossRef] [PubMed]
  96. Sangouni, A.A.; Taghdir, M.; Mirahmadi, J.; Sepandi, M.; Parastouei, K. Effects of curcumin and/or coenzyme Q10 supplementation on metabolic control in subjects with metabolic syndrome: A randomized clinical trial. Nutr. J. 2022, 21, 62. [Google Scholar] [CrossRef] [PubMed]
  97. Rehman, A.; Mehta, K.J. Betaine in ameliorating alcohol-induced hepatic steatosis. Eur. J. Nutr. 2022, 61, 1167–1176. [Google Scholar] [CrossRef] [PubMed]
  98. Ceja-Galicia, Z.A.; García-Arroyo, F.E.; Aparicio-Trejo, O.E.; El-Hafidi, M.; Gonzaga-Sánchez, G.; León-Contreras, J.C.; Hernández-Pando, R.; Guevara-Cruz, M.; Tovar, A.R.; Rojas-Morales, P.; et al. Therapeutic effect of curcumin on 5/6Nx hypertriglyceridemia: Association with the improvement of renal mitochondrial β-oxidation and lipid metabolism in kidney and liver. Antioxidants 2022, 11, 2195. [Google Scholar] [CrossRef] [PubMed]
  99. Zeng, Y.; Luo, Y.; Wang, L.; Zhang, K.; Peng, J.; Fan, G. Therapeutic Effect of Curcumin on Metabolic Diseases: Evidence from Clinical Studies. Int. J. Mol. Sci. 2023, 24, 3323. [Google Scholar] [CrossRef] [PubMed]
  100. Boretti, A. Curcumin-based fixed dose combination products for cholesterol management: A narrative review. ACS Pharmacol. Transl. Sci. 2024, 7, 300–308. [Google Scholar] [CrossRef] [PubMed]
  101. Yaikwawong, M.; Jansarikit, L.; Jirawatnotai, S.; Chuengsamarn, S. The effect of curcumin on reducing atherogenic risks in obese patients with type 2 diabetes: A randomized controlled trial. Nutrients 2024, 16, 2441. [Google Scholar] [CrossRef] [PubMed]
  102. Tan, C.; Zhou, L.; Wen, W.; Xiao, N. Curcumin promotes cholesterol efflux by regulating ABCA1 expression through miR-125a-5p/SIRT6 axis in THP-1 macrophage to prevent atherosclerosis. J. Toxicol. Sci. 2021, 46, 209–222. [Google Scholar] [CrossRef] [PubMed]
  103. Yang, K.; Chen, J.; Zhang, T.; Yuan, X.; Ge, A.; Wang, S.; Xu, H.; Zeng, L.; Ge, J. Efficacy and safety of dietary polyphenol supplementation in the treatment of non-alcoholic fatty liver disease: A systematic review and meta-analysis. Front. Immunol. 2022, 13, 949746. [Google Scholar] [CrossRef] [PubMed]
  104. Singh, L.; Sharma, S.; Xu, S.; Tewari, D.; Fang, J. Curcumin as a natural remedy for atherosclerosis: A pharmacological review. Molecules 2021, 26, 4036. [Google Scholar] [CrossRef] [PubMed]
  105. Baratzadeh, F.; Butler, A.E.; Kesharwani, P.; Moallem, S.A.; Sahebkar, A. Effects of curcumin on low-density lipoprotein oxidation: From experimental studies to clinical practice. EXCLI J. 2022, 21, 840–851. [Google Scholar] [PubMed]
  106. Reyes-Farias, M.; Fos-Domenech, J.; Serra, D.; Herrero, L.; Sánchez-Infantes, D. White adipose tissue dysfunction in obesity and aging. Biochem. Pharmacol. 2021, 192, 114723. [Google Scholar] [CrossRef] [PubMed]
  107. Zhu, Q.; An, Y.A.; Scherer, P.E. Mitochondrial regulation and white adipose tissue homeostasis. Trends Cell Biol. 2022, 32, 351–364. [Google Scholar] [CrossRef] [PubMed]
  108. Mu, W.-J.; Zhu, J.-Y.; Chen, M.; Guo, L. Exercise-mediated browning of white adipose tissue: Its significance, mechanism and effectiveness. Int. J. Mol. Sci. 2021, 22, 11512. [Google Scholar] [CrossRef] [PubMed]
  109. Cai, J.; Quan, Y.; Zhu, S.; Lin, J.; Zhang, Q.; Liu, J.; Liang, Z.; Liao, Y.; Jiang, W.; He, Y.; et al. The browning and mobilization of subcutaneous white adipose tissue supports efficient skin repair. Cell Metab. 2024, 36, 1287–1301.e7. [Google Scholar] [CrossRef] [PubMed]
  110. Cheng, L.; Wang, J.; Dai, H.; Duan, Y.; An, Y.; Shi, L.; Lv, Y.; Li, H.; Wang, C.; Ma, Q.; et al. Brown and beige adipose tissue: A novel therapeutic strategy for obesity and type 2 diabetes mellitus. Adipocyte 2021, 10, 48–65. [Google Scholar] [CrossRef] [PubMed]
  111. Alalaiwe, A.; Fang, J.-Y.; Lee, H.-J.; Chiu, C.-H.; Hsu, C.-Y. The demethoxy derivatives of curcumin exhibit greater differentiation suppression in 3t3-l1 adipocytes than curcumin: A mechanistic study of adipogenesis and molecular docking. Biomolecules 2021, 11, 1025. [Google Scholar] [CrossRef] [PubMed]
  112. Moetlediwa, M.T.; Ramashia, R.; Pheiffer, C.; Titinchi, S.J.J.; Mazibuko-Mbeje, S.E.; Jack, B.U. Therapeutic effects of curcumin derivatives against obesity and associated metabolic complications: A review of in vitro and in vivo studies. Int. J. Mol. Sci. 2023, 24, 14366. [Google Scholar] [CrossRef] [PubMed]
  113. Torres, J.L.; Usategui-Martín, R.; Hernández-Cosido, L.; Bernardo, E.; Manzanedo-Bueno, L.; Hernández-García, I.; Mateos-Díaz, A.-M.; Rozo, O.; Matesanz, N.; Salete-Granado, D.; et al. PPAR-γ Gene Expression in Human Adipose Tissue Is Associated with Weight Loss After Sleeve Gastrectomy. J. Gastrointest. Surg. 2022, 26, 286–297. [Google Scholar] [CrossRef] [PubMed]
  114. Soares, R.M.V.; da Silva, M.A.; Campos, J.T.A.M.; Lima, J.G. Familial partial lipodystrophy resulting from loss-of-function PPARγ pathogenic variants: Phenotypic, clinical, and genetic features. Front. Endocrinol. 2024, 15, 1394102. [Google Scholar]
  115. Enayati, A.; Ghojoghnejad, M.; Roufogalis, B.D.; Maollem, S.A.; Sahebkar, A. Impact of phytochemicals on PPAR Receptors: Implications for Disease Treatments. PPAR Res. 2022, 2022, 4714914. [Google Scholar] [CrossRef] [PubMed]
  116. Corrales, P.; Vidal-Puig, A.; Medina-Gómez, G. PPARs and Metabolic Disorders Associated with Challenged Adipose Tissue Plasticity. Int. J. Mol. Sci. 2018, 19, 2124. [Google Scholar] [CrossRef] [PubMed]
  117. Wu, L.-Y.; Chen, C.-W.; Chen, L.-K.; Chou, H.-Y.; Chang, C.-L.; Juan, C.-C. Curcumin attenuates adipogenesis by inducing preadipocyte apoptosis and inhibiting adipocyte differentiation. Nutrients 2019, 11, 2307. [Google Scholar] [CrossRef] [PubMed]
  118. Lee, H.J.; Jin, B.Y.; Park, M.R.; Kim, N.H.; Seo, K.S.; Jeong, Y.T.; Wada, T.; Lee, J.S.; Choi, S.H.; Kim, D.H. Inhibition of adipose tissue angiogenesis prevents rebound weight gain after caloric restriction in mice fed a high-fat diet. Life Sci. 2023, 332, 122101. [Google Scholar] [CrossRef] [PubMed]
  119. Pélissier, L.; Bagot, S.; Miles-Chan, J.L.; Pereira, B.; Boirie, Y.; Duclos, M.; Dulloo, A.; Isacco, L.; Thivel, D. Is dieting a risk for higher weight gain in normal-weight individual? A systematic review and meta-analysis. Br. J. Nutr. 2023, 130, 1190–1212. [Google Scholar] [CrossRef] [PubMed]
  120. Wang, L.; Suyama, S.; Lee, S.A.; Ueta, Y.; Seino, Y.; Sharp, G.W.G.; Yada, T. Fasting inhibits excitatory synaptic input on paraventricular oxytocin neurons via neuropeptide Y and Y1 receptor, inducing rebound hyperphagia, and weight gain. Front. Nutr. 2022, 9, 994827. [Google Scholar] [CrossRef] [PubMed]
  121. Tanasie, G.; Bojin, F.; Tatu, R.F.; Sisu, A.M.; Cristea, M.; Puscasiu, D.A.; Nemes, E.A.; Tatu, C.S. In vitro effects of biomaterials on mesenchymal stem cells viability and proliferation. Mater. Plast. 2017, 54, 523–526. [Google Scholar] [CrossRef]
  122. Hurmuz, M.; Bojin, F.; Ionac, M.; Tatu, F.; Puscasiu, D.; Tatu, C. Plastic adherence method for isolation of stem cells derived from infrapatellar fat pad. Mater. Plast. 2016, 53, 553–556. [Google Scholar]
  123. Ikeda, K.; Yamada, T. UCP1 dependent and independent thermogenesis in brown and beige adipocytes. Front. Endocrinol. 2020, 11, 498. [Google Scholar] [CrossRef] [PubMed]
  124. Van Nguyen, T.T.; Van Vu, V.; Van Pham, P. Brown adipocyte and browning thermogenesis: Metabolic crosstalk beyond mitochondrial limits and physiological impacts. Adipocyte 2023, 12, 2237164. [Google Scholar] [CrossRef] [PubMed]
  125. Zhu, Y.; Liu, W.; Qi, Z. Adipose tissue browning and thermogenesis under physiologically energetic challenges: A remodelled thermogenic system. J. Physiol. 2024, 602, 23–48. [Google Scholar] [CrossRef] [PubMed]
  126. Sakers, A.; De Siqueira, M.K.; Seale, P.; Villanueva, C.J. Adipose-tissue plasticity in health and disease. Cell 2022, 185, 419–446. [Google Scholar] [CrossRef] [PubMed]
  127. Chiu, Y.H.; Chou, W.L.; Ko, M.C.; Liao, J.C.; Huang, T.H. Curcumin mitigates obesity-driven dysbiosis and liver steatosis while promoting browning and thermogenesis in white adipose tissue of high-fat diet-fed mice. J. Nutr. Biochem. 2025, 143, 109920. [Google Scholar] [CrossRef] [PubMed]
  128. He, L.; Su, Z.; Wang, S. The anti-obesity effects of polyphenols: A comprehensive review of molecular mechanisms and signal pathways in regulating adipocytes. Front. Nutr. 2024, 11, 1393575. [Google Scholar] [CrossRef] [PubMed]
  129. Zhu, X.; Du, S.; Yan, Q.; Min, C.; Zhou, N.; Zhou, W.; Li, X. Dietary curcumin supplementation promotes browning and energy expenditure in postnatal overfed rats. Nutr. Metab. 2021, 18, 97. [Google Scholar] [CrossRef] [PubMed]
  130. Brockmueller, A.; Mueller, A.L.; Kunnumakkara, A.B.; Aggarwal, B.B.; Shakibaei, M. Multifunctionality of Calebin A in inflammation, chronic diseases and cancer. Front. Oncol. 2022, 12, 962066. [Google Scholar] [CrossRef] [PubMed]
  131. Chalichem, N.S.S.; Jupudi, S.; Yasam, V.R.; Basavan, D. Dipeptidyl peptidase-IV inhibitory action of Calebin A: An in silico and in vitro analysis. J. Ayurveda Integr. Med. 2021, 12, 663–672. [Google Scholar] [CrossRef] [PubMed]
  132. Lee, P.S.; Lu, Y.Y.; Nagabhushanam, K.; Ho, C.T.; Mei, H.C.; Pan, M.H. Calebin-A prevents HFD-induced obesity in mice by promoting thermogenesis and modulating gut microbiota. J. Tradit. Complement. Med. 2022, 13, 119–127. [Google Scholar] [CrossRef] [PubMed]
  133. Buhrmann, C.; Shayan, P.; Banik, K.; Kunnumakkara, A.B.; Kubatka, P.; Koklesova, L.; Shakibaei, M. Targeting NF-κB signaling by Calebin A, a compound of turmeric, in multicellular tumor microenvironment: Potential role of apoptosis induction in CRC cells. Biomedicines 2020, 8, 236. [Google Scholar] [CrossRef] [PubMed]
  134. de, P. Oliveira, A.L.; Martinez, S.E.; Nagabushnam, K.; Majeed, M.; Alrushaid, S.; Sayre, C.L.; Davies, N.M. Calebin A: Analytical Development for Pharmacokinetics Study, Elucidation of Pharmacological Activities and Content Analysis of Natural Health Products. J. Pharm. Pharm. Sci. 2015, 18, 494–514. [Google Scholar]
  135. Sabir, S.M.; Zeb, A.; Mahmood, M.; Abbas, S.R.; Ahmad, Z.; Iqbal, N. Phytochemical analysis and biological activities of ethanolic extract of Curcuma longa rhizome. Braz. J. Biol. 2021, 81, 737–740. [Google Scholar] [CrossRef] [PubMed]
  136. Tran, G.H.; Lee, H.D.; Kim, S.H.; Lee, S.; Lee, S. Profiling of the leaves and stems of Curcuma longa using LC-ESI-MS and HPLC analysis. J. Appl. Biol. Chem. 2023, 66, 338–344. [Google Scholar] [CrossRef]
  137. Yoon, D.S.; Cho, S.Y.; Yoon, H.J.; Kim, S.R.; Jung, U.J. Protective effects of p-coumaric acid against high-fat diet-induced metabolic dysregulation in mice. Biomed. Pharmacother. 2021, 142, 111969. [Google Scholar] [CrossRef] [PubMed]
  138. Alam, M.A.; Subhan, N.; Hossain, H.; Hossain, M.; Reza, H.M.; Rahman, M.M.; Ullah, M.O. Hydroxycinnamic acid derivatives: A potential class of natural compounds for the management of lipid metabolism and obesity. Nutr. Metab. 2016, 13, 27. [Google Scholar] [CrossRef] [PubMed]
  139. Lee, M.; Nam, S.H.; Yoon, H.G.; Kim, S.; You, Y.; Choi, K.C.; Lee, Y.H.; Lee, J.; Park, J.; Jun, W. Fermented Curcuma longa L. prevents alcoholic fatty liver disease in mice by regulating CYP2E1, SREBP-1c, and PPAR-α. J. Med. Food 2022, 25, 456–463. [Google Scholar] [CrossRef] [PubMed]
  140. Muhammad Abdul Kadar, N.N.; Ahmad, F.; Teoh, S.L.; Yahaya, M.F. Caffeic Acid on metabolic syndrome: A review. Molecules 2021, 26, 5490. [Google Scholar] [CrossRef] [PubMed]
  141. Ye, L.; Hu, P.; Feng, L.-P.; Huang, L.-L.; Wang, Y.; Yan, X.; Xiong, J.; Xia, H.-L. Protective effects of ferulic acid on metabolic syndrome: A comprehensive review. Molecules 2023, 28, 281. [Google Scholar] [CrossRef] [PubMed]
  142. Luo, Z.; Li, M.; Yang, J.; Li, J.; Zhang, Y.; Liu, F.; El-Omar, E.; Han, L.; Bian, J.; Gong, L.; et al. Ferulic acid attenuates high-fat diet-induced hypercholesterolemia by activating classic bile acid synthesis pathway. Front. Nutr. 2022, 9, 976638. [Google Scholar] [CrossRef] [PubMed]
  143. Cho, J.; Park, E. Ferulic acid maintains the self-renewal capacity of embryo stem cells and adipose-derived mesenchymal stem cells in high fat diet-induced obese mice. J. Nutr. Biochem. 2020, 77, 108327. [Google Scholar] [CrossRef] [PubMed]
  144. Salazar-López, N.J.; Astiazarán-García, H.; González-Aguilar, G.A.; Loarca-Piña, G.; Ezquerra-Brauer, J.-M.; Domínguez Avila, J.A.; Robles-Sánchez, M. Ferulic Acid on glucose dysregulation, dyslipidemia, and inflammation in diet-induced obese rats: An integrated study. Nutrients 2017, 9, 675. [Google Scholar] [CrossRef] [PubMed]
  145. Dosoky, N.S.; Setzer, W.N. Chemical Composition and Biological Activities of Essential Oils of Curcuma Species. Nutrients 2018, 10, 1196. [Google Scholar] [CrossRef] [PubMed]
  146. Kotha, R.R.; Luthria, D.L. Curcumin: Biological, Pharmaceutical, Nutraceutical, and Analytical Aspects. Molecules 2019, 24, 2930. [Google Scholar] [CrossRef] [PubMed]
  147. Dosoky, N.S.; Satyal, P.; Setzer, W.N. Variations in the Volatile Compositions of Curcuma Species. Foods 2019, 8, 53. [Google Scholar] [CrossRef] [PubMed]
  148. Poudel, D.K.; Ojha, P.K.; Rokaya, A.; Satyal, R.; Satyal, P.; Setzer, W.N. Analysis of Volatile Constituents in Curcuma Species, viz. C. aeruginosa, C. zedoaria, and C. longa, from Nepal. Plants 2022, 11, 1932. [Google Scholar]
  149. Cao, W.; Chen, X.; Xiao, C.; Lin, D.; Li, Y.; Luo, S.; Zeng, Z.; Sun, B.; Lei, S. Ar-turmerone inhibits the proliferation and mobility of glioma by downregulating cathepsin B. Aging 2023, 15, 9377–9390. [Google Scholar] [CrossRef] [PubMed]
  150. Hori, Y.; Tsutsumi, R.; Nasu, K.; Boateng, A.; Ashikari, Y.; Sugiura, M.; Nakajima, M.; Kurauchi, Y.; Hisatsune, A.; Katsuki, H.; et al. Aromatic-Turmerone Analogs Protect Dopaminergic Neurons in Midbrain Slice Cultures through Their Neuroprotective Activities. Cells 2021, 10, 1090. [Google Scholar] [CrossRef] [PubMed]
  151. Duangyod, T.; Rujanapan, N.; Champakam, S.; Charoensup, R. Anti-inflammatory activity and chemical constituents of red limestone. J. Adv. Pharm. Technol. Res. 2021, 12, 185–189. [Google Scholar] [CrossRef] [PubMed]
  152. Gadnayak, A.; Dehury, B.; Nayak, A.; Jena, S.; Sahoo, A.; Panda, P.C.; Ray, A.; Nayak, S. Mechanistic insights into 5-lipoxygenase inhibition by active principles derived from essential oils of Curcuma species: Molecular docking, ADMET analysis and molecular dynamic simulation study. PLoS ONE 2022, 17, e0271956. [Google Scholar] [CrossRef] [PubMed]
  153. Wang, X.; Shen, Y.; Thakur, K.; Han, J.; Zhang, J.-G.; Hu, F.; Wei, Z.-J. Antibacterial Activity and Mechanism of Ginger Essential Oil against Escherichia coli and Staphylococcus aureus. Molecules 2020, 25, 3955. [Google Scholar] [CrossRef] [PubMed]
  154. Zohmachhuana, A.; Malsawmdawngliana; Lalnunmawia, F.; Mathipi, V.; Lalrinzuali, K.; Kumar, N.S. Curcuma aeruginosa Roxb. exhibits cytotoxicity in A-549 and HeLa cells by inducing apoptosis through caspase-dependent pathways. Biomed. Pharmacother. 2022, 150, 113039. [Google Scholar] [CrossRef] [PubMed]
  155. Peng, X.; Luo, R.; Li, J.; He, A.; Wang, X.; Wan, H.; Cai, Y.; Dong, W.; Lin, J. Zingiberene targets the miR-16/cyclin-B1 axis to regulate the growth, migration and invasion of human liver cancer cells. J. BUON 2020, 25, 1904–1910. [Google Scholar] [PubMed]
  156. Chen, J.-J.; Tsai, T.-H.; Liao, H.-R.; Chen, L.-C.; Kuo, Y.-H.; Sung, P.-J.; Chen, C.-L.; Wei, C.-S. New Sesquiterpenoids and Anti-Platelet Aggregation Constituents from the Rhizomes of Curcuma zedoaria. Molecules 2016, 21, 1385. [Google Scholar] [CrossRef] [PubMed]
  157. Yu, Z.; Xu, J.; Shao, M.; Zou, J. Germacrone Induces Apoptosis as Well as Protective Autophagy in Human Prostate Cancer Cells. Cancer Manag. Res. 2020, 12, 4009–4016. [Google Scholar] [CrossRef] [PubMed]
  158. Zhang, R.; Hao, J.; Guo, K.; Liu, W.; Yao, F.; Wu, Q.; Liu, C.; Wang, Q.; Yang, X. Germacrone Inhibits Cell Proliferation and Induces Apoptosis in Human Esophageal Squamous Cell Carcinoma Cells. BioMed Res. Int. 2020, 2020, 7643248. [Google Scholar] [CrossRef] [PubMed]
  159. Dhyani, P.; Sati, P.; Sharma, E.; Attri, D.C.; Bahukhandi, A.; Tynybekov, B.; Szopa, A.; Sharifi-Rad, J.; Calina, D.; Suleria, H.A.R.; et al. Sesquiterpenoid lactones as potential anti-cancer agents: An update on molecular mechanisms and recent studies. Cancer Cell Int. 2022, 22, 305. [Google Scholar] [CrossRef] [PubMed]
  160. Septembre-Malaterre, A.; Bedoui, Y.; Giry, C.; Gasque, P.; Guiraud, P.; Sélambarom, J. Quercetin can reduce viral RNA level of O’nyong-nyong virus and resulting innate immune cytokine responses in cultured human synovial fibroblasts. Sci. Rep. 2021, 11, 6369. [Google Scholar] [CrossRef] [PubMed]
  161. Han, Q.; Zhu, J.; Zhang, P. Mechanisms of main components in Curcuma longa L. on hepatic fibrosis based on network pharmacology and molecular docking: A review. Medicine 2023, 102, e34353. [Google Scholar] [CrossRef] [PubMed]
  162. Jamil, Q.U.A.; Iqbal, S.M.; Jaeger, W.; Studenik, C. Vasodilating, spasmolytic, inotropic and chronotropic activities of curcuminoids from Curcuma longa in isolated organ preparations of guinea pigs. J. Physiol. Pharmacol. 2018, 69, 441–449. [Google Scholar]
  163. Li, J.; Sun, Y.; Li, G.; Cheng, C.; Sui, X.; Wu, Q. The Extraction, Determination, and Bioactivity of Curcumenol: A Comprehensive Review. Molecules 2024, 29, 656. [Google Scholar] [CrossRef] [PubMed]
  164. Li, F.; Qi, Q.; Qiao, Y.; Huang, Y.; Lu, Y.; Gu, K.; Liu, H.; Gao, C.; Liu, S.; Wu, H. Curcumenol inhibits malignant progression and promotes ferroptosis via the SLC7A11/NF-κB/TGF-β pathway in triple-negative breast cancer. Int. J. Mol. Med. 2025, 56, 111. [Google Scholar] [CrossRef] [PubMed]
  165. Unhapipatpong, C.; Polruang, N.; Shantavasinkul, P.C.; Julanon, N.; Numthavaj, P.; Thakkinstian, A. The effect of curcumin supplementation on weight loss and anthropometric indices: An umbrella review and updated meta-analyses of randomized controlled trials. Am. J. Clin. Nutr. 2023, 117, 1005–1016. [Google Scholar] [CrossRef] [PubMed]
  166. Akbari, M.; Lankarani, K.B.; Tabrizi, R.; Ghayour-Mobarhan, M.; Peymani, P.; Ferns, G.; Ghaderi, A.; Asemi, Z. The Effects of Curcumin on Weight Loss Among Patients With Metabolic Syndrome and Related Disorders: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front. Pharmacol. 2019, 10, 649. [Google Scholar] [CrossRef] [PubMed]
  167. Alsharif, F.J.; Almuhtadi, Y.A. The Effect of curcumin supplementation on anthropometric measures among overweight or obese adults. Nutrients 2021, 13, 680. [Google Scholar] [CrossRef] [PubMed]
  168. Khan, S.; Arif, M.; Laraib, H.; Naqvi, S.N.; Shah, O.A.; Farooq, U.; Sami-Ullah, M.; Khan, G.A. The effect of turmeric and black pepper powder incorporated in breakfast on postprandial glycemia, appetite, palatability, and gastrointestinal well-being in normal-weight adults. Food Sci. Nutr. 2024, 12, 2846–2854. [Google Scholar] [CrossRef] [PubMed]
  169. Karandish, M.; Mozaffari-khosravi, H.; Mohammadi, S.M.; Cheraghian, B.; Azhdari, M. Curcumin and zinc co-supplementation along with a loss-weight diet can improve lipid profiles in subjects with prediabetes: A multi-arm, parallel-group, randomized, double-blind placebo-controlled phase 2 clinical trial. Diabetol. Metab. Syndr. 2022, 14, 22. [Google Scholar] [CrossRef] [PubMed]
  170. Batsis, J.A.; Apolzan, J.W.; Bagley, P.J.; Blunt, H.B.; Divan, V.; Gill, S.; Golden, A.; Gundumraj, S.; Heymsfield, S.B.; Kahan, S.; et al. A Systematic Review of Dietary Supplements and Alternative Therapies for Weight Loss. Obesity 2021, 29, 1102–1113. [Google Scholar] [CrossRef] [PubMed]
  171. Alexandru, I.; Nistor, D.; Motofelea, A.C.; Cadar, B.-A.; Crintea, A.; Tatu, C.; Pop, G.N.; Csep, A.N. Vitamins, Coenzyme Q10, and Antioxidant Strategies to Improve Oocyte Quality in Women with Gynecological Cancers: A Comprehensive Review. Antioxidants 2024, 13, 1567. [Google Scholar] [CrossRef] [PubMed]
  172. Munteanu, C.; Schwartz, B. Interactions between Dietary Antioxidants, Dietary Fiber and the Gut Microbiome: Their Putative Role in Inflammation and Cancer. Int. J. Mol. Sci. 2024, 25, 8250. [Google Scholar] [CrossRef] [PubMed]
  173. Park, S.-H. Role of Phytochemicals in Treatment of Aging and Cancer: Focus on Mechanism of FOXO3 Activation. Antioxidants 2024, 13, 1099. [Google Scholar] [CrossRef] [PubMed]
  174. Scafuri, L.; Buonerba, C.; Strianese, O.; de Azambuja, E.; Palleschi, M.; Riccio, V.; Marotta, V.; Scocca, C.; Riccio, G.; Errico, C.; et al. Impact of Dietary Supplements on Clinical Outcomes and Quality of Life in Patients with Breast Cancer: A Systematic Review. Nutrients 2025, 17, 981. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Biosynthesis of curcumin and its derivatives starting from phenylalanine. Synthesis of the precursors p-coumaryl-CoA and feruloyl-CoA, marked in red. Phenolic OH groups are also marked in red (a); interconversions of precursors to enol forms of Bisdesmethoxycurcumin, Desmethoxycurcumin and Curcumin, and enol-ketotautomerization are schematically presented. Phenolic OH groups and precursors p-coumaryl-CoA, and feruloyl-CoA are marked in red (b).
Figure 1. Biosynthesis of curcumin and its derivatives starting from phenylalanine. Synthesis of the precursors p-coumaryl-CoA and feruloyl-CoA, marked in red. Phenolic OH groups are also marked in red (a); interconversions of precursors to enol forms of Bisdesmethoxycurcumin, Desmethoxycurcumin and Curcumin, and enol-ketotautomerization are schematically presented. Phenolic OH groups and precursors p-coumaryl-CoA, and feruloyl-CoA are marked in red (b).
Ijms 26 06880 g001
Figure 2. The transformation of curcumin into calebin A is carried out by catalysis of Baeyer–Villiger-type monooxygenase.
Figure 2. The transformation of curcumin into calebin A is carried out by catalysis of Baeyer–Villiger-type monooxygenase.
Ijms 26 06880 g002
Figure 3. Schematic representation of the effect of curcumin on upregulation of PPAR-γ and promotion of thermogenesis through activation of UCP-1. PPAR-γ—peroxisome-activating receptor γ; UCP-1—uncoupling protein-1; DRP-1—dynamin-related protein 1; SIRT-1—sirtuin 1; RXR—retinoid X receptor; PPRE—proliferation response element; PGC-1α—peroxisome proliferator-activated receptor G coactivator 1-α.
Figure 3. Schematic representation of the effect of curcumin on upregulation of PPAR-γ and promotion of thermogenesis through activation of UCP-1. PPAR-γ—peroxisome-activating receptor γ; UCP-1—uncoupling protein-1; DRP-1—dynamin-related protein 1; SIRT-1—sirtuin 1; RXR—retinoid X receptor; PPRE—proliferation response element; PGC-1α—peroxisome proliferator-activated receptor G coactivator 1-α.
Ijms 26 06880 g003
Figure 4. The changes produced by curcumin. Green illustrates the activating effect on enzymes involved in the removal of free radicals; yellow shows the depressing effect that curcumin exerts on ROS, ROS-producing factors, as well as pro-inflammatory mediators. ROS—reactive oxygen species; MPO—myeloperoxidase; SOD—superoxide dismutase; GPX—glutathione peroxidase; NNT—nicotinamide nucleotide transhydrogenase; GSH—reduced glutathione; NF-kB—nuclear factor-kB.
Figure 4. The changes produced by curcumin. Green illustrates the activating effect on enzymes involved in the removal of free radicals; yellow shows the depressing effect that curcumin exerts on ROS, ROS-producing factors, as well as pro-inflammatory mediators. ROS—reactive oxygen species; MPO—myeloperoxidase; SOD—superoxide dismutase; GPX—glutathione peroxidase; NNT—nicotinamide nucleotide transhydrogenase; GSH—reduced glutathione; NF-kB—nuclear factor-kB.
Ijms 26 06880 g004
Figure 5. Schematic representation of the inhibitory action of curcumin on PPAR-γ and the main biochemical processes stimulated by PPAR-γ in adipogenesis. PPAR-γ—peroxisome-activating receptor γ; C/EBPα—CCAAT/enhancer-binding protein α.
Figure 5. Schematic representation of the inhibitory action of curcumin on PPAR-γ and the main biochemical processes stimulated by PPAR-γ in adipogenesis. PPAR-γ—peroxisome-activating receptor γ; C/EBPα—CCAAT/enhancer-binding protein α.
Ijms 26 06880 g005
Table 1. Human trials involving curcumin and their main outcomes.
Table 1. Human trials involving curcumin and their main outcomes.
PopulationStudy TypeInterventionOutcomeDurationReferences
Middle-aged and elderly overweight participantsRandomized, double-blind, placebo-controlledTurmeric extract mixture (CLE)
-
Body weight, BMI and inflammatory markers decreased
-
Mental health score and anger–hostility improved
12 Weeks[9]
Subjects with mild/moderate knee joint painPilot, randomized, double-blind, placebo-controlled clinical trialB-Turmactive® (a new formulation of dry extracts of turmeric roots) and brewer’s yeast as a placebo
-
Reduced global pain; however, only B-Turmactive® had superior effects in reducing night-time pain
1 week[10]
Obese subjects with type-2 diabetesRandomized, double-blind, placebo-controlled trialCurcumin extract 1500 mg/day
-
Important decrease in fasting blood glucose and HbA1c
-
Improved function of β-cells
-
BMI was lowered
12 months[11]
Healthy males 20–40 years oldRandomized, double-blind, crossover pilot trialCurcuma longa powder; low dose (1.5 g), moderate dose (3 g), high dose (6 g), separated by 7-day washout
-
High doses of C. longa did not substantially influence the antioxidant effect. Moreover, lower doses might optimize antioxidant capacity
3 weeks[12]
Mild to moderate hospitalized
COVID-19 patients
Triple-blind randomized, placebo-controlledOral nanocurcumin formulation (Sinacurmin® soft-gel 40 mg)With the exception of a sore throat, symptoms were reduced considerably faster compared to the placebo treatment4 months[13]
Female subjects with polycystic ovary syndromeRandomized, double-blind, placebo-controlled trialCurcumin 500 mg three times dailySignificantly decreased fasting plasma glucose and insulin, reducing sex hormone levels and hirsutism score12 weeks[14]
Patients with knee osteoarthritis (OA)Randomized, non-inferiority, controlled clinical trialBioavailable turmeric extract (BCM-95®) 500 mg capsules versus paracetamol 650 mg tabletsThe results obtained considering bioavailable turmeric extract were comparable to those observed with paracetamol, improving physical function and pain relief in patients with knee OA6 weeks[15]
Patients with nonalcoholic fatty liver diseaseRandomized, double-blind, placebo-controlled clinical trialTurmeric 2 g/day
-
Substantial reduction in liver enzymes (AST, ALT, GGT)
-
Significant decrease in triglyceride and LDL, HDL, MDA levels
-
No significant change in liver echogenicity, total cholesterol and VLDL levels
8 weeks [16]
Table 2. Other components found in small amounts in turmeric and their main biological activities.
Table 2. Other components found in small amounts in turmeric and their main biological activities.
ComponentStructureMain Activity
Turmerones:
Aromatic (Ar)-turmerone
Ijms 26 06880 i001
-
arrest proliferation and decrease mobility of glioma cells [149]
-
prevention dopaminergic neurodegeneration induced by microglial activation [150]
α-turmeroneIjms 26 06880 i002
-
anti-inflammatory [151,152]
β-turmeroneIjms 26 06880 i003
-
anti-inflammatory [152]
Sesquiterpenes:
α-curcumene
Ijms 26 06880 i004
-
antibacterial (Staphylococcus aureus and E. coli) [153]
-
anti-proliferative (lung adenocarcinoma cells) [154]
CurcumanolideIjms 26 06880 i005
-
anti-inflammatory [151]
ZingibereneIjms 26 06880 i006
-
antibacterial (Staphylococcus aureus and E. coli) [153]
-
anti-proliferative (liver cancer cells) [155]
Germacrane:
Germacrone
Ijms 26 06880 i007
-
inhibition of collagen-induced platelet aggregation [156]
-
anti-proliferative effect on prostate cancer cells [157] and esophageal squamous cell carcinoma cells [158]
GermacreneIjms 26 06880 i008
-
anti-inflammatory [159]
-
cytotoxic activity against liver and breast carcinoma cells [159]
Phenols/polyphenols:
Quercetin
Ijms 26 06880 i009
-
reduces viral replication in synovial fibroblast cells infected with O’nyong-nyong virus [160]
-
adjuvant in hepatic fibrosis [161]
VanillinIjms 26 06880 i010
-
spasmolytic effect [162]
Guaiane:
Curcumenol
Ijms 26 06880 i011
-
anti-tumoral (lung cancer, liver cancer, gastric cancer [163], breast cancer [164]), hepatoprotective [163]
ProcurcumenolIjms 26 06880 i012
-
inhibition of collagen-induced platelet aggregation [156]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Marina, C.D.; Puscasiu, D.; Flangea, C.; Vlad, T.; Cimporescu, A.; Popescu, R.; Moatar, A.E.; Vlad, D.C. Adipo-Modulation by Turmeric Bioactive Phenolic Components: From Curcuma Plant to Effects. Int. J. Mol. Sci. 2025, 26, 6880. https://doi.org/10.3390/ijms26146880

AMA Style

Marina CD, Puscasiu D, Flangea C, Vlad T, Cimporescu A, Popescu R, Moatar AE, Vlad DC. Adipo-Modulation by Turmeric Bioactive Phenolic Components: From Curcuma Plant to Effects. International Journal of Molecular Sciences. 2025; 26(14):6880. https://doi.org/10.3390/ijms26146880

Chicago/Turabian Style

Marina, Cristina Doriana, Daniela Puscasiu, Corina Flangea, Tania Vlad, Adinela Cimporescu, Roxana Popescu, Aurica Elisabeta Moatar, and Daliborca Cristina Vlad. 2025. "Adipo-Modulation by Turmeric Bioactive Phenolic Components: From Curcuma Plant to Effects" International Journal of Molecular Sciences 26, no. 14: 6880. https://doi.org/10.3390/ijms26146880

APA Style

Marina, C. D., Puscasiu, D., Flangea, C., Vlad, T., Cimporescu, A., Popescu, R., Moatar, A. E., & Vlad, D. C. (2025). Adipo-Modulation by Turmeric Bioactive Phenolic Components: From Curcuma Plant to Effects. International Journal of Molecular Sciences, 26(14), 6880. https://doi.org/10.3390/ijms26146880

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop