Next Article in Journal
Amino Acids as the Potential Co-Former for Co-Crystal Development: A Review
Previous Article in Journal
In Vitro and In Vivo Activity of 14-O-[(4,6-Diamino-pyrimidine-2-yl) thioacetyl] Mutilin against Methicillin-Resistant Staphylococcus aureus
Previous Article in Special Issue
Protective Effects of Polyphenol Enriched Complex Plants Extract on Metabolic Dysfunctions Associated with Obesity and Related Nonalcoholic Fatty Liver Diseases in High Fat Diet-Induced C57BL/6 Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review on Obesity Management through Natural Compounds and a Green Nanomedicine-Based Approach

1
Department of Zoology, Maharshi Dayanand University, Rohtak 124001, India
2
Department of Zoology, Gaur Brahman Degree College, Rohtak 124001, India
3
Department of Zoology, Maharaja Neempal Singh Government College, Bhiwani 127021, India
4
Department of Zoology, Hindu Girls College, Sonipat 131001, India
5
Department of Biotechnology, Kurukshetra University, Kurukshetra 136119, India
*
Author to whom correspondence should be addressed.
Submission received: 27 February 2021 / Revised: 29 April 2021 / Accepted: 21 May 2021 / Published: 28 May 2021
(This article belongs to the Special Issue Anti-obesity Effects of Natural Compounds)

Abstract

:
Obesity is a serious health complication in almost every corner of the world. Excessive weight gain results in the onset of several other health issues such as type II diabetes, cancer, respiratory diseases, musculoskeletal disorders (especially osteoarthritis), and cardiovascular diseases. As allopathic medications and derived pharmaceuticals are partially successful in overcoming this health complication, there is an incessant need to develop new alternative anti-obesity strategies with long term efficacy and less side effects. Plants harbor secondary metabolites such as phenolics, flavonoids, terpenoids and other specific compounds that have been shown to have effective anti-obesity properties. Nanoencapsulation of these secondary metabolites enhances the anti-obesity efficacy of these natural compounds due to their speculated property of target specificity and enhanced efficiency. These nanoencapsulated and naive secondary metabolites show anti-obesity properties mainly by inhibiting the lipid and carbohydrate metabolizing enzymes, suppression of adipogenesis and appetite, and enhancing energy metabolism. This review focuses on the plants and their secondary metabolites, along with their nanoencapsulation, that have anti-obesity effects, with their possible acting mechanisms, for better human health.

1. Introduction

In past few decades, overweight and obese cases among people of all age groups have become a serious health issue. According to the WHO, obesity is mainly considered as excessive and abnormal fat accumulation that can lead to improper health functioning. Approximately 1.9 billion adults were overweight with a basal metabolic index (BMI ≥ 25) and among these, 650 million adults were obese (BMI ≥ 30), in 2016. The frequency of obesity almost tripled from 1975 to 2016. In 2019, an estimated 38.2 million children under 5 years old were obese all over the world and almost 50% of total children in Asia were obese or overweight [1]. Obesity is mainly due to genetic, environment and behavioral factors [2]. It mainly occurs due to an increase in the ratio of calories or energy intake to the calories or energy expenditure, which results from genetic susceptibility and lethargic lifestyle modifications [3]. Obesity and excessive weight gain can be controlled by modifications in diet and increased physical exercise, but these approaches are not rapid, so many people prefer chemical medications over these approaches for effective results [4]. Various chemical medications are available in the market that have anti-obesity property, such as orlistat, fenfluramine, coreaserin, rimonabant, cetlistat, sibutramine, and phentermine with topiramate, with different efficacy to control obesity [5,6]. These medications have several adverse effects on health like cardiometabolic abnormalities, anxiety, high blood pressure and pulse rate, and depressive disorders [7]. Therefore, more sophisticated efforts should be done to discover natural anti-obesity agents with less side effects and more efficiency. Accordingly, several natural secondary metabolites present in different plants like polyphenols, flavonoids, terpenoids, alkaloids, saponins, carboxylic acids, glycosides and tannins are reported to have anti-obesity efficacy through different mechanisms of action. Several bioactive compounds in edible plants, such as green tea with epigallocatechins, nobiletin in citrus peel, resveratrol, pterostilbene in berries, curcumin in turmeric, and anthocyanins in Hibiscus sabdariffa, are reported to suppress the obesity-causing factors [8,9]. Nanotechnology and nanoencapsulation of these secondary metabolites opens up a new horizon for overcoming this aforementioned health issue. These biochemicals can be encapsulated with biocompatible nanoparticles, which enhances their target specificity, bioavailability, stability and aqueous solubility [10]. The basic pathogenesis of obesity mainly includes an increase in appetite and a decrease in calorie expenditure by modulating physical activity and cellular functioning. These abnormalities increase the process of adipogenesis, which in turn increases the release of cytokines and vascular complications leading to cardiovascular system disorders like atherosclerosis and hyperlipidemia. The various factors responsible for the induction of obesity and other related health disorders are shown in Figure 1.

2. Role of Natural Products in Obesity: Regulation of Metabolism

The natural products present in different plant resources alter the regulation of various enzymes and genetic factors through different acting mechanisms (Figure 2). These secondary metabolites inhibit carbohydrate and lipid metabolizing enzymes like α-amylase, α-glucosidases and different lipases present in the gastrointestinal tract. Amylases and glucosidases are the main key enzymes responsible for digesting the carbohydrates and result in release of glucose via glucose transporters [11]. An increase in the level of glucose above normal ultimately results in the release of insulin from pancreatic cells and initiation of three pathways (glycogenesis, glycolysis and de novo lipogenesis) to decrease glucose in the blood [12]. The synthesis of lipids and fatty acids from glucose and esterification of these lipids into triglycerides for storage in adipose tissue causes obesity [13]. On the other hand, lipases, mainly secreted from the different regions of the gastrointestinal tract, are mainly involved in the digestion of fatty acids, phospholipids and triglycerides and hydrolyze them into monoglycerides. These monoglycerides form chylomicrons and the micellar structure with sugars, lysophosphatidic acid and bile salts. This structure then passes into the enterocytes and subsequently results in the synthesis and storage of triglycerides in adipose tissue [14]. The inhibition of these enzymes, after treatment with plant products, ultimately results in the reduction of obesity. These secondary metabolites reduce obesity through modulation of different hormones such as leptin, ghrelin and insulin. Leptin is primarily secreted by white adipose tissue (WAT) [15] and regulates the “brain–gut axis” by activating its receptors in the central nervous system (CNS), subsequently reducing food intake and enhancing the calorie expenditure pathways [16]. Insulin is secreted from the pancreatic beta cells and transforms signals to the brain that result in a reduction in food intake over the long term and more rapid energy expenditure. Signals from both leptin and insulin communicate in a way to reduce the food and energy intake [17]. Plants with anti-obesity properties increase the level of both hormones. Adiponectin (an adipokine secreted from adipose tissue) increases hepatic insulin activity, increases fatty acid oxidation, and enhances glucose uptake in both skeletal muscle and the liver [16]. It mainly acts through the activation of adenosine monophosphate-activated protein kinase (AMPK) activity and AMPK impedes acetyl Co~A carboxylase activity and decreases the content of malonyl Co~A [18].
Ghrelin is also called the hunger hormone and inhibition of the secretion of ghrelin has an anti-obesity effect [19]. The process of adipogenesis and adipocyte differentiation can be interfered by regulating various transcriptional factors involved in different steps of these processes to manage obesity [20]. These transcriptional factors are proliferator-activated receptors (PPAR), sterol regulatory elementary binding proteins (SREBP) and CCAAT/enhancer binding proteins (C/EBP) [21]. Repression at the level of SREBP [22,23] and C/EBP [24] and enhancement of the PPAR level [25] are also strategies to manage obesity by different plant metabolites. Regulation of lipid metabolism at the level of synthesis and lipid degradation by different enzymes and hormones can reduce the obesity effect [26]. SREBP1a (sterol regulatory element binding protein 1a), SREBP2, low density lipoproteins (LDL), and receptors-3-hydroxy-3-methylglutaryl Co~A reductase mainly regulate the process of synthesis of cholesterol from acetyl Co~A [27]. SREBP-1c upregulates the transcription of the lipogenic enzymes stearoyl Co~A desaturase and fatty acid synthase (FAS) [28]. Activation of AMPK interferes with SREBP-1c and FAS and reduces the synthesis of cholesterol and fatty acids [29]. In a similar way, carnitine palmitoyl transferase 1A (CPT1A) decreases the concentration of hepatic triglycerides and increases the process of fatty acid oxidation [30]. Therefore, regulation of all of these factors imparts a beneficial effect in preventing obesity with the help of natural products that are secondary metabolites obtained from plants. Their role in treating obesity is described below.

2.1. Polyphenols

Polyphenols are phenolic compounds with at least one or more aromatic ring/s with a hydroxyl group and other functional groups like glycosides, methyl ethers and esters associated with its chemical structure [31]. On the basis of the number of aromatic rings, polyphenols can be categorized into, among others, tannins, stilbenes, phenolic acid, flavonoids, lignans, lignins and coumarins [32,33]. Among all the categories of phenolic compounds, flavonoids can be distinguished by the presence of two aromatic rings, connected by a 3-C bridge. Resveratrol, catechins, quercetin, procyanidins, epigallocatechins gallate, anthocyanins and procyanidins are gaining much interest due to their significant anti-obesity properties. Several studies have reported anti-obesity efficacy of different phenolic compounds in both animals and cell models. The polyphenols (extracted from Vitis rotundifloia) [34], ellagitannins, and proanthocyanidins extracted from raspberries and strawberries [35] are reported to inhibit pancreatic lipase with an IC50 value of 16.90 and 5 µg/mL, respectively. Gallic acid, epigallocatechin’s gallate (EGCG) and epigallocatechin are reported to inhibit the lipase activity with an IC50 value of 387.2, 273.3 and 39.2 µM, respectively [36]. Liu et al. [37] reported the inhibition of pancreatic lipase, α-amylase and α-glucosidases with an IC50 value of 1.86, 0.38 and 2.20 mg/mL, respectively, by phenolic compounds extracted from Nelumbo nucifera. Polyphenols extracted from Citrus aurantium [38] and Coralluma fimbriate [39] are reported to show appetite suppressive effects. MacLean and Luo [40] reported that Bushman’s Hat (Hoodia Gordoni) extracts increased ATP (adenosine triphosphate) in hypothalamic neurons and positively regulated hunger and food intake in rats. Epigallocatechins-3-gallate (EGCG) extracted from green tea reduced food intake by inhibition of ghrelin hormone and stimulation of adiponectin [41]. Stimulation of thermogenesis and energy consumption is also a significant way to decrease obesity. BAT is unique as it involves release of excess energy by the process of thermogenesis. An uncoupling protein (UCP1) regulates the process of thermogenesis in BAT by reducing the proton gradient and uncoupling ATP synthesis from oxidation [42]. UCP3 is another homologous protein to UCP1 and exerts its anti-obesity action by regulating the level of leptin, thyroid hormones and β-adrenergic agonists [43]. EGCG extracted from green tea is reported to induce thermogenesis and energy consumption [44]. Other compounds like quercetin, isoflavones, gallic acid, resveratrol, and curcumin induce thermogenic activity by modulating the signaling pathway of adenosine monophosphate protein kinase (AMPK), SIRT1 (sirtuin 1), proliferator activated receptor gamma coactivator 1-α (PGC-1α), and catechol O-methyl transferase, which are mainly involved in the regulation of transcription and physiology of adipose tissue (Figure 3). Curcumin, resveratrol, epigallocatechin-3-gallat, and genisten are able to inhibit the process of differentiation of adipocytes [45]. Carvacrol [46] and phenolic compounds from chokeberries [47] inhibited the process of adipocyte differentiation by modulating the level of PPAR-γ, C/EBP-α and SREBP-1c. Lipid and triglyceride accumulation is mainly responsible for excessive weight gain and obesity. The process of synthesis of cholesterol from acetyl Co~A is mainly regulated by SREBP1α, SREBP2, LDL receptors and 3-hydroxy methylglutaryl Co~A reductase [27]. AMPK stimulates fatty acid oxidation and reduces the synthesis of cholesterol by interfering with fatty acid synthase and SREBP-1c [29]. Polyphenols extracted from Hibiscus sabdariffa [48] and combined with the extract of Lippia citridora [49] exhibited anti-obesity efficacy.

2.2. Flavonoids

Flavonoids, commonly present in a variety of plants, are mainly responsible for the flavor and colour of the vegetables and fruits [50]. The chemical structure of flavonoids mainly consists of a heterocyclic pyran ring and two aromatic rings associated with it, which forms a 15-C phenylpropanoid core. Flavonoids are grouped into six categories on the basis of the double bond present in the heterocyclic ring and its oxidation status. These groups are anthoxanthins (flavonols or catechins), anthocyanins (cyanin pigment), flavonones (narigenin and herpertin), flavones (luteolin, apigenin), isoflavones (genistein, flavin) and chalcones (butein, xanthoangelol) [51]. These different groups of flavonoids show demarcated anti-obesity properties with different modes of action. Flavonoids inhibit weight gain by reducing food intake and increasing the feeling of satiety. Flavonoids extracted from the spinach leaf and the combination of flavonoids and procyanidin [52] had a significant effect in treating overweight by reducing the cravings for food and increasing satiety. WAT is specialized in storage of excess energy in the form of triglycerides [53] and BAT is mostly specialized for high metabolism and energy expenditure. It is predominantly present in the suprarenal, spinal and supraclavicular regions [54]. Both adipocytes express UCP-1 protein, which is mainly responsible for thermogenesis and energy consumption. Thermogenesis is mainly regulated by various mechanisms and improves the activity of the sympathetic nervous system, which results in the secretion of norepinephrine, which ultimately results in energy consumption and reduction of fat accumulation [55]. PGC1-α is the main transcription factor that regulates the process of thermogenesis [56]. AMPK and SIRT1 are mainly increased by flavonoids and are the factors mainly responsible for the expression of PGC1α. AMPK/PGC1α signaling is primarily responsible for browning of adipose tissue and thermogenesis [57]. The gastrointestinal tract comprises a diverse bacterial population, including Actinobacteria, Firmicutes, Proteobacteria and Bacteroidetes [58], and out of which 1.9% of total flora are heritable and more than 20% of the diversity mainly depends on environmental factors such as dietary habits [59]. Imbalance in the diversity of the gut microflora may cause endotoxic accumulation in the circulatory system, which in turn may induce chronic inflammation and obesity [60]. Short chain fatty acids (SCFAs) are produced after bacterial fermentation of some indigestible biochemicals such as polyphenols, polysaccharides and protein, which are mainly involved in energy expenditure, oxidation of fatty acids, regulation of sympathetic activity and intestinal gluconeogenesis [61]. Bile acid produced from cholesterol in the liver can be metabolized by intestinal bacteria and bile acid also regulates the composition of microbes and facilitates their growth [62]. The aforementioned factors reveal the role of flavonoids in obesity regulation (Figure 4).

2.3. Diterpenoids

Diterpenoids are chemical compounds that contain two terpene units, with each having four isoprene units with the molecular formula C20H32 [63]. The anti-obesity mechanism of diterpenoids is depicted in Figure 5. Diterpenoids have several therapeutic effects, such as anti-obesity effects from taxanes from Taxus [64] carnosic acid, and steviol and its derivatives [65]. Diterpenoids and their derivatives showed anti-obesity properties through different strategies. Teucrin A isolated from Teucrin chaemodrys and Carnosic acid isolated from Rosmarinus officinalis are reported to reduce body weight in obese Sprague Dawley rats [66] and mice [67], respectively. Secondly, PTP-1B (protein tyrosine phosphatase 1B) was shown to mainly have adverse effects on leptin transduction and insulin signaling, and the inhibition of this enzyme is reported to speed up the insulin signaling pathways and in turn have positive effects in treating obesity [68]. Acanthoic acid, isolated from Acanthopanax koreanum [69] and Ent-16βH, 17-isobutryloxy-kauran-19-oic acid and Ent-16βH, 18-isobutryloxy-kauran-19-oic acid isolated from Siegesbeckia glabrescens [70], and Hueafuranoid A isolated from Huea sp. are reported to inhibit PTP-1B activity in a dose dependent manner (IC50 = 30 µg/mL and 13.9 µM, respectively). Diterpenoids also showed lipase inhibitory effects as they inhibited the activity of pancreatic lipase and triglyceride accumulation. Carnosic acid (CA) (isolated from R. officinalis) and carnasol (isolated from Salvia officinalis [71] showed lipase inhibitory activity and modulated body weight gain [3] and lipoprotein-lipase mRNA expression in mouse adipose tissue [72]. Diterpenoids showed anti-obesity effects by inhibiting adipocyte differentiation. Carnosic acid [72,73] and 14-deoxy-11,12- didehydroandrographolide isolated from Andrographis paniculata [74] are reported to interfere with mitotic clonal expansion, block the expression C/EBPα and PPAR-α, reduce lipoprotein mRNA expression via tumor necrosis factor (TNF-α) and interleukin-6, alter the ratio of different C/EBP-β proteins, and activate the mTOR pathways. In a study on the geranylgeraniol (alcoholic derivatives of diterpenoids), which are mostly found in some herbs and fruits, it was reported that they activate human PPARα and PPARγ in CV1 cells and regulate the expression of target genes mainly responsible for lipid metabolism in 3T3-L1 cells and Hep G2 [75].

3. Anti-Obesity Effects of Different Plant Resources

Different plant extracts with different anti-obesity effects are described in Table 1. The plant extracts of Curcuma longa [76], Camellia sinensis [77], Cosmos cadatus [78], Morniga olifera [79] and Elateriospermum tapos [80] are reported to have anti-obesity efficacy and among these plants Curcuma longa is reported to have the highest anti-obesity potential. It decreased body weight and adipose tissue weight, and more importantly, regulated the expression of transcription factors like PPAR-γ (71 %) and C/EBP-α (38.83%). Fatty acyl synthase (FAS) (66.03%) and acetyl Co~A carboxylase (ACC) (35%) decreased and that of AMPK (12.93%), CPT1 (88%) and adiponectin (49.01%) increased after the treatment. Other plants also regulated the serum lipid profiles, including TG, TC, ALT, AST and ALP concentration and expression of the transcription factors responsible for lipid metabolism. C57BL/6J mice were also implemented as an experimental model for Nelumbo nucifera [81], chlorogenic acid [82], Capsicum annuum [83], Cudrania tricuspidate [84], Morus alba, Ilex paraguariensis, Rosmarinus officinalis, Moringa officinalis [85,86], Ecklonia cava [47], Cirsium cetidens [87], Gymnema sylvestre [88] and Ishige okamurae [89], and among all these plants, the mixed extracts of Morus alba, Ilex paraguariensis and Rosmarinus officinalis showed the maximum anti-obesity effect as it decreased the body weight and relative weight of other body organs up to 98% and regulated the serum lipid profile by decreasing the TC (18.6%), LDL-c (59%), ALT (60.1%), AST (35.2%), insulin (75.9%) and leptin (46.8%). These results are maximum among the plants described above in Table 1 on C57BL/6J mice. The secondary metabolites from all the plants also reduced the expression of those transcription factors (PPAR-γ, C/EBPα, SREBP-1c) that are mainly responsible for the accumulation of lipids in adipose tissue and promoted the expression of those factors (ACC, PPARα, AMPK and adiponectins).
Among the anti-obesity effects of Juniperus communis [104], Ramulus mori [111], Vibrunum opulus [106], Paullinia cupana [96], Peucedanum japonicum [23], Aster yomena [97], Prunus salicina [98], Eclipta alba [99], and Polygonum cuspidatum [92] evaluated on 3T3-L1 cells, Eclipta alba showed the highest activity as it decreased the expression of PPARα (1.9 fold), C/EBPα (1.8 fold), FAS (1.4 fold) and FABP4 (1.8 fold) and also decreased the concentration of lipids in terms of cholesterol and triglycerides in these adipocytes. The above-described plants regulated the transcription factors and lipid accumulation in 3T3-L1 adipocytes. Other biochemical parameters like inhibition of digesting enzymes were also reported in the studies on Solenostema argel, Coralluma quadrangular, Hibiscus sabdariffa [103], Helichrysum sanguineum [110], Vitis vinifera, and Rhus coriaria [100]. Solenostema argel showed the maximum inhibition of pancreatic lipase (97.02 ± 1.4%), α-amylase (69.32 ± 1.14%) and α-glucosidase (89.08 ± 1.1%).

4. Nanotechnology Associated with Anti-Obesity Effects

Nanotechnology and nanoencapsulation of secondary metabolites is an emerging and more beneficial strategy for the treatment of obesity with amplified efficiency and minimized side effects [117]. Various types of phytochemicals such as flavonoids, terpenoids, polyphenols, glycosides and tannins have been reported as promising agents in treating obesity but their low target specificity, low aqueous solubility, stability and toxicity at high dose put some restrictions on their clinical use. These biomolecules are mainly responsible for the reduction of metal ions from their metallic precursor and green synthesis of metallic nanoparticles. These limitations can be overcome by using metallic nanoparticles and nanoencapsulated phytochemicals, as they increase their target specificity, bioavailability, and solubility, and more importantly, prevent them from pre-term degradation [10]. They also have a high surface to volume ratio and tunable surface chemistry. Table 2 shows the list of some nanoparticles synthesized from the biological material and some nanoencapsulated biological molecules with their anti-obesity effects on obese animal models and/or cell lines.
The anti-obesity effect of the nanocellulose compound isolated from Vitis vinifera was reported by Abdelbaky et al. [118] on a rat model. They reported the regulation of body weight, organ weight, blood serum lipid profile and food intake after induction of cellulose NPs in their diet. The nanoparticles isolated from red grape seed, especially cellulose nanocrystals (CNC), had a significant positive effect on obesity and hyperlipidemia compared to grape seeds powder, while the chemical constituents of the crude leaf extract of Vitis vinifera [100] inhibited pancreatic lipase, affecting lipid metabolism and consequently obesity. In a similar way, the gold nanoparticles from Salacia chinensis [119], Smilax glabra [120], Poria cocos [122], Dendropanax morbifera [130] and chitosan NPs [121] were reported to show anti-obesity efficacy by regulating the serum lipid profile and the level of hormones related to lipid metabolism and regulation of the transcriptional factors mainly responsible for the metabolism of lipid digestion and lipid accumulation. Nanoencapsulation increased the delivery of the specific molecule/secondary metabolite to the specific target with much greater efficacy and stability. Nanoencapsulation of quercetin with a succinyl chitosan alginate shell [124] and PLGA [128] in rat models revealed that the serum lipid profile and glucose level were regulated after the induction of these nanoencapsulated structures. On the other hand, quercetin, being one of the chemical constituents in extracts of different plant parts—flowers of Capsicum annuum [94], plums of Prunus salicina [98], leaves of Vitis vinifera [100], fruits of Rhus coriaria [100], leaves of Cosmos cadatus Kunth [78], fruits of Malus prunifolia [102], fruits of Malus huphenesis [102], Hibiscus sabdariffa [103], Solenostemma argel [103], and Vibrunum opulus [105], twigs of Ranulus mori [111], leaves of Nelumbo nucifera [81], soybean embryo and enzymatically modified Isoquercetin [115]—has contributed to obesity management as mentioned above.
EGCG is a secondary metabolite mainly present in green tea and many other plant products and its encapsulation with Soy PC, α-tocopherol and Kolliphor HS15 [126] and Kodia-PC and α-tocopherol [125] was reported in C57BL/6J mice and Human THP1 cells, respectively. In mice, it regulated the serum lipid profile and decreased the surface lesion of aortic arches and inflammatory factors, and in monocytes, it mainly decreased the expression of the transcription factors responsible for lipid accumulation and in turn decreased the cholesterol level in these monocytes.
Similarly, the anti-obesity studies on Curcuma longa [76] and the nanoencapsulated curcumin [72,127] molecule showed that the curcumin molecule capsulated with a PLA-PEG molecule enhanced the anti-obesity effect in terms of modification of the serum lipid profile (p ≤ 0.01) and modulation of transcription factors (p ≤ 0.01) compared to native curcumin molecules [76].
Resveratrol and oxy resveratrol are chemical ingredients of twigs of Ranulus mori [111], root extract of Polygonum cuspidatum [92], and a nanocellulose compound isolated from Vitis vinifera [118]. Trans resveratrol-encapsulated NPs [131] regulated the expression of signal pathways, lipid parameters and conversion of WAT to BAT related to obesity management. These chemicals acted individually (NPs) and/or in combination (crude/extract) in obesity management. Resveratrol was encapsulated with PLGA by Wan et al. [129], who used HepG2 cells to evaluate the anti-obesity efficacy of these nanoencapsulated structures. They found a significant decrease in the lipid accumulation, hepatocellular differentiation and triglyceride accumulation in these hepatocytes.
More studies are required, which will certainly open new avenues and add further to existing knowledge to establish the contribution of the constituent chemical/s (nanoencapsulated or constitutent of extract), individually or synergistically, that may act as efficacious nanomedicines assisting in obesity management, specifically acting through its regulatory parameter/s.

5. Conclusions

The human population has witnessed the health benefits of natural compounds due to the presence of secondary metabolites in medicinal plants. The secondary metabolites have modulating effects in various disorders. The natural compounds have less side effects. Obesity is an ordinary underrated health disorder but is now considered a serious public health issue globally, which leads to the emergence of other chronic health disorders. This review summarizes the natural products in plant extract(s)/isolated or purified compound(s) and the nanomedicine-enhanced effect of these natural compounds to reduce the comorbidities related to obesity. The nanomedicine-based approach showed curative effects for obesity management by their gene target specific activity and this drug delivery system opens up a new horizon to improve the ameliorative efficiency of these natural compounds. To cite an example, the nanoencapsulated curcumin (PLA-PEG) molecule showed significantly enhanced anti-obesity effects compared to the native curcumin molecule. The nanoparticles and nanoencapsulation treatment enhanced the anti-obesity efficacy. This article also aimed to explain the mechanism of these secondary metabolites in regulating the obesity parameters by modulating the gene/transcriptional factors amenable for adipogenesis, adipocyte differentiation, energy and lipid metabolism, and gut microbiota. Future perspective studies of such natural molecules in terms of drug specificity, efficacy and ethical issues/trials need to be conducted for their specific validation in the interest of better human health.

Funding

This work received no external funding.

Acknowledgments

The authors thankfully acknowledge the facilities from Maharshi Dayanand University, Rohtak (India), CSIR & UGC, New Delhi.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AbbreviationAbbreviation Expanded Form
ACCAcetyl Co~A Carboxylase
ALPAlkaline Phosphatase
ALTAlanine Aminotransferase
AMPK Adenosine Monophosphate-Activated Protein Kinase
Ap2Activating Protein 2
ASTAspartate Aminotransferase
BATBrown Adipose Tissue
BMIBasal Metabolic Index
C/EBPCCAAT/Enhancer Binding Proteins
CACarnosic Acid
CDIVKCis-3′,4′-Diisovalerylkhellactone
CNS Central Nervous System
CPT1A Carnitine Palmitoyl Transferase 1A
CREBcAMP Response Element Binding Proteins
CRPCapsicoside G- Rich Protein
CYP7A1Cytochrome P450 Family 7 Subfamily A Member 1
EGCGEpigallocatechin’s Gallate
FABPFAS Binding Proteins
FAS Fatty Acid Synthase
FBGFasting Blood Glucose
FFAFree Fatty Acids
HDLHigh Density Lipoproteins
HFDHigh Fat Diet
ILInterleukin
Jak2Janus kinase 2
JNKcJun N-terminal Kinase
LDLLow Density Lipoproteins
mTORMechanistic Target of Rapamycin
NPYNeuropeptide Y
ORXORX Peptide
PGC1-αProliferator Activated Receptor Gamma Coactivator 1-A
PLA-PEGPolylactic Acid-Polyethylene Glycol
PLGAPolylactic-Co-Glycolic Acid
PPARPeroxisome Proliferator-Activated Receptors
PTP-1BProtein Tyrosine Phosphtase 1B
SCD-1Stearoyl-CoA Desaturase-1
SCFAsShort Chain Fatty Acids
SIRT1Sirtuin 1
SREBPSterol Regulatory Elementary Binding Proteins
STAT 3Signal Transducer and Activator of Transcription 3
TCTotal Cholesterol
TGTriglycerides
TNF-αTumour Necrosis Factor-Alpha
UCP1Uncoupling Protein
WAT White Adipose Tissue

References

  1. World Health Organization. Obesity and Overweight. 2020. Available online: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight (accessed on 7 February 2021).
  2. Rodriguez-Perez, C.; Segura-Carretero, A.; Contreras, D.M. Phenolic compounds as natural and multifunctional anti-obesity agents: A review. Crit. Rev. Food Sci. Nutr. 2019, 59, 1212–1229. [Google Scholar] [CrossRef]
  3. Romo Vaquero, M.; García Villalba, R.; Larrosa, M.; Yáñez-Gascón, M.J.; Fromentin, E.; Flanagan, J.; García-Conesa, M.T. Bioavailability of the major bioactive diterpenoids in a rosemary extract: Metabolic profile in the intestine, liver, plasma, and brain of Zucker rats. Mol. Nutr. Food Res. 2013, 57, 1834–1846. [Google Scholar] [CrossRef]
  4. Urbatzka, R.; Freitas, S.; Palmeira, A.; Almeida, T.; Moreira, J.; Azevedo, C.; Afonso, C.; Correia-da-Silva, M.; Sousa, E.; Pinto, M.; et al. Lipid reducing activity and toxicity profiles of a library of polyphenol derivatives. Eur. J. Med. Chem. 2018, 151, 272–284. [Google Scholar] [CrossRef]
  5. Srivastava, G.; Apovian, C.M. Current pharmacotherapy for obesity. Nat. Rev. Endocrinol. 2018, 14, 12–24. [Google Scholar] [CrossRef]
  6. Kim, K.S.; Jang, M.J.; Fang, S.; Yoon, S.G.; Kim, I.Y.; Seong, J.K.; Hahm, D.H. Anti-obesity effect of taurine through inhibition of adipogenesis in white fat tissue but not in brown fat tissue in a high-fat diet-induced obese mouse model. J. Amino Acids 2019, 51, 245–254. [Google Scholar] [CrossRef]
  7. Despres, J.P. Pleiotropic effects of rimonabant: Clinical implications. Curr. Pharm. Des. 2009, 15, 553–570. [Google Scholar] [CrossRef]
  8. Kim, J.K.; So, H.; Youn, M.J.; Kim, H.J.; Kim, Y.; Park, C.; Kim, S.J.; Ha, Y.A.; Chai, K.Y.; Kim, S.M.; et al. Hibiscus sabdariffa L. water extract inhibits the adipocyte differentiation through the PI3-K and MAPK pathway. J. Ethnopharmacol. 2007, 114, 260–267. [Google Scholar] [CrossRef] [PubMed]
  9. Huang, L.; Chen, J.; Cao, P. Anti-obese effect of glucosamine and chitosan oligosaccharide in high-fat diet-induced obese rats. Mar. Drugs 2015, 13, 2732–2756. [Google Scholar] [CrossRef] [PubMed]
  10. Goktas, Z.; Zu, Y.; Abbasi, M.; Galyean, S.; Wu, D.; Fan, Z.; Wang, S. Recent advances in nanoencapsulation of phytochemicals to combat obesity and its comorbidities. J. Agric. Food Chem. 2020, 68, 8119–8131. [Google Scholar] [CrossRef] [PubMed]
  11. Nyambe-Silavwe, H.; Villa-Rodriguez, J.A.; Ifie, I.; Holmes, M.; Aydin, E.; Jensen, J.M.; Williamson, G. Inhibition of human α-amylase by dietary polyphenols. J. Function. Foods 2015, 19, 723–732. [Google Scholar] [CrossRef]
  12. Wong, S.K.; Chin, K.Y.; Suhaimi, F.H.; Fairus, A.; Ima-Nirwana, S. Animal models of metabolic syndrome: A review. Nutr. Metab. 2016, 13, 65. [Google Scholar] [CrossRef] [Green Version]
  13. Duwaerts, C.C.; Maher, J.J. Macronutrients and the adipose-liver axis in obesity and fatty liver. Cell Mol. Gastroenterol. Hepatol. 2019, 7, 749–761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Birari, R.B.; Bhutani, K.K. Pancreatic lipase inhibitors from natural sources: Unexplored potential. Drug. Discov. 2007, 12, 879–889. [Google Scholar] [CrossRef]
  15. Friedman, J.M.; Halaas, J.L. Leptin and the regulation of body weight in mammals. Nature 1988, 395, 763–770. [Google Scholar] [CrossRef] [PubMed]
  16. Singla, P.; Bardoloi, A.; Parkash, A.A. Metabolic effects of obesity: A review. World J. Diabetes 2010, 1, 76–88. [Google Scholar] [CrossRef]
  17. Ashrafi, K. Obesity and the regulation of fat metabolism. Worm Book 2007, 1–2. [Google Scholar] [CrossRef] [PubMed]
  18. Yamauchi, T.; Kamon, J.; Minokoshi, Y.; Ito, Y.; Waki, H.; Uchida, S.; Yamashita, S.; Noda, M.; Kita, S.; Ueki, K.; et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat. Med. 2002, 8, 1288–1295. [Google Scholar] [CrossRef]
  19. Santiago-García, P.A.; López, M.G. Agavins from Agave angustifolia and Agave potatorum affect food intake, body weight gain and satiety-related hormones (GLP-1 and ghrelin) in mice. Food Funct. 2014, 5, 3311–3319. [Google Scholar] [CrossRef]
  20. Lefterova, M.I.; Lazar, M.A. New developments in adipogenesis. Trends Endocrinol. Metab. 2009, 20, 107–114. [Google Scholar] [CrossRef]
  21. Brey, C.W.; Nelder, M.P.; Hailemariam, T.; Gaugler, R.; Hashmi, S. Krüppel-like family of transcription factors: An emerging new frontier in fat biology. Int. J. Biol. Sci. 2009, 5, 622. [Google Scholar] [CrossRef]
  22. Yamasaki, M.; Ogawa, T.; Wang, L.; Katsube, T.; Yamasaki, Y.; Sun, X.; Shiwaku, K. Anti-obesity effects of hot water extract from Wasabi (Wasabia japonica Matsum.) leaves in mice fed high-fat diets. Nutr. Res. Pract. 2013, 7, 267–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Choi, R.Y.; Nam, S.J.; Ham, J.R.; Lee, H.I.; Yee, S.T.; Kang, K.Y.; Lee, M.K. Anti-adipogenic and anti-diabetic effects of cis-3′, 4′-diisovalerylkhellactone isolated from Peucedanum japonicum Thunb leaves in vitro. Bioorg. Med. Chem. Lett. 2016, 26, 4655–4660. [Google Scholar] [CrossRef]
  24. Imran, A.; Butt, M.S.; Arshad, M.S.; Arshad, M.U.; Saeed, F.; Sohaib, M.; Munir, R. Exploring the potential of black tea-based flavonoids against hyperlipidemia related disorders. J. Nutr. Biochem. 2016, 30, 14–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Tan, S.; Gao, B.; Tao, Y.; Guo, J.; Su, Z.Q. Antiobese effects of capsaicin–chitosan microsphere (CCMS) in obese rats induced by high fat diet. J. Agric. Food Chem. 2014, 8, 1866–1874. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, L.; Zeng, B.; Liu, Z.; Liao, Z.; Zhong, Q.; Gu, L.; Wei, H.; Fang, X. Green tea polyphenols modulate colonic microbiota diversity and lipid metabolism in high-fat diet treated HFA mice. J. Food Sci. 2018, 83, 864–873. [Google Scholar] [CrossRef] [PubMed]
  27. Karagianni, P.; Talianidis, I. Transcription factor networks regulating hepatic fatty acid metabolism. Biochim. Biophys. Acta 2015, 1851, 2–8. [Google Scholar] [CrossRef] [PubMed]
  28. Salvado, L.; Palomer, X.; Barroso, E.; Vazquez-Carrera, M. Targeting endoplasmic reticulum stress in insulin resistance. Trends Endocrinol. Metab. 2015, 26, 438–448. [Google Scholar] [CrossRef] [PubMed]
  29. Srivastava, R.A.; Pinkosky, S.L.; Filippov, S.; Hanselman, J.C.; Cramer, C.T.; Newton, R.S. AMP-activated protein kinase: An emerging drug target to regulate imbalances in lipid and carbohydrate metabolism to treat cardio-metabolic diseases. J. Lipid Res. 2012, 53, 2490–2514. [Google Scholar] [CrossRef] [Green Version]
  30. Takei, K.; Nakagawa, Y.; Wang, Y.; Han, S.I.; Satoh, A.; Sekiya, M.; Matsuzaka, T.; Shimano, H. Effects of K-877, a novel selective PPARalpha modulator, on small intestine contribute to the amelioration of hyperlipidemia in low- density lipoprotein receptor knockout mice. J. Pharmacol. Sci. 2017, 133, 214–222. [Google Scholar] [CrossRef] [Green Version]
  31. Bhuyan, D.J.; Basu, A. Phenolic compounds: Potential health benefits and toxicity. In Utilisation of Bioactive Compounds from Agricultural and Food Production Waste; Vuong, Q.V., Ed.; CRC Press, Taylor & Francis Group: Boca Raton, FL, USA, 2017; pp. 27–59. [Google Scholar]
  32. Naczk, M.; Shahidi, F. Phenolics in cereals, fruits and vegetables: Occurrence, extraction and analysis. J. Pharm. Biomed. Anal. 2006, 41, 1523–1542. [Google Scholar] [CrossRef]
  33. Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [Green Version]
  34. You, Q.; Chen, F.; Wang, X.; Luo, P.G.; Jiang, Y. Inhibitory effects of muscadine an- thocyanins on alpha-glucosidase and pancreatic lipase activities. J. Agric. Food Chem. 2011, 59, 9506–9511. [Google Scholar] [CrossRef]
  35. Mc-Dougall, G.J.; Kulkarni, N.N.; Stewart, D. Berry polyphenols in- hibit pancreatic lipase activity in vitro. Food Chem. 2009, 115, 193–199. [Google Scholar] [CrossRef]
  36. Rahim, A.; Takahashi, Y.; Yamaki, K. Mode of pancreatic lipase inhibition activity in vitro by some flavonoids and non-flavonoid polyphenols. Food Res. Int. 2015, 75, 289–294. [Google Scholar] [CrossRef] [PubMed]
  37. Liu, S.; Li, D.; Huang, B.; Chen, Y.; Lu, X.; Wang, Y. Inhibition of pancre- atic lipase, alpha-glucosidase, alpha-amylase, and hypolipidemic effects of the total flavonoids from Nelumbo nucifera leaves. J. Ethnopharmacol. 2013, 149, 263–269. [Google Scholar] [CrossRef]
  38. Klontz, K.C.; Timbo, B.B.; Street, D. Consumption of dietary supplements containing citrus aurantium (bitter orange)—2004 California behavioral risk factor surveillance survey (BRFSS). Ann. Pharmacother. 2006, 40, 1747–1751. [Google Scholar] [CrossRef]
  39. Kuriyan, R.; Raj, T.; Srinivas, S.K.; Vaz, M.; Rajendran, R.; Kurpad, A.V. Effect of Caralluma fimbriata extract on appetite, food intake and anthropometry in adult Indian men and women. Appetite 2007, 48, 338–344. [Google Scholar] [CrossRef] [PubMed]
  40. MacLean, D.B.; Luo, L.G. Increased ATP content/production in the hypothalamus may be a signal for energy-sensing of satiety: Studies of the anorectic mechanism of a plant steroidal glycoside. Brain Res. 2004, 20, 1–11. [Google Scholar] [CrossRef] [PubMed]
  41. Chen, I.C.; Liu, J.; Chiu, H.H. Therapeutic effect of high dose green tea extract on weight reduction: A randomized, doubleblind, placebo-controlled clinical trial. Clin. Nutr. 2016, 35, 592–599. [Google Scholar] [CrossRef]
  42. Cannon, B.; Nedergaard, J. Brown adipose tissue: Function and physiological significance. Physiol. Rev. 2004, 84, 277–359. [Google Scholar] [CrossRef]
  43. Gong, Y.; Slee, R.B.; Fukai, N.; Rawadi, G.; Roman-Roman, S.; Reginato, A.M. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell 2001, 107, 513–523. [Google Scholar] [CrossRef] [Green Version]
  44. Zhou, J.; Mao, L.; Xu, P.; Wang, Y. Effects of (-)-epigallocatechin gallate (EGCG) on energy expenditure and microglia-mediated hypothalamic inflammation in mice fed a high-fat diet. Nutrients 2018, 10, 1681. [Google Scholar] [CrossRef] [Green Version]
  45. Mele, L.; Bidault, G.; Mena, P.; Crozier, A.; Brighenti, F.; Vidal-Puig, A.; Del Rio, D. Dietary (Poly)phenols, brown adipose tissue activation, and energy expenditure: A narrative review. Adv. Nutr. 2017, 8, 694–704. [Google Scholar] [CrossRef] [Green Version]
  46. Spalletta, S.; Flati, V.; Toniato, E.; Di Gregorio, J.; Marino, A.; Pierdomenico, L.; Marchi-Sio, M.; D’Orazi, G.; Cacciatore, I.; Robuffo, I. Carvacrol reduces adipogenic differentiation by modulating autophagy and ChREBP expression. PLoS ONE 2018, 13, e0206894. [Google Scholar] [CrossRef]
  47. Kim, J.; Choi, M.; Lee, C.; Kwon, T.; Nam, T.J. Anti-obesity effects of pectinase and cellulase enzyme-treated Ecklonia cava extract in high-fat diet-fed C57BL/6N mice. Int. J. Mol. Med. 2018, 41, 924–934. [Google Scholar] [PubMed] [Green Version]
  48. Herranz-Lopez, M.; Olivares-Vicente, M.; Boix-Castejon, M.; Caturla, N.; Roche, E.; Micol, V. Differential effects of a combination of Hibiscus sabdariffa and Lippia citriodora polyphenols in overweight/obese subjects: A randomized controlled trial. Sci. Rep. 2019, 9, 2999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Herranz-Lopez, M.; Olivares-Vicente, M.; Encinar, J.A.; Barrajon-Catalan, E.; Segura-Carretero, A.; Joven, J.; Micol, V. Multi-targeted molecular effects of hibiscus sabdariffa polyphenols: An opportunity for a global approach to obesity. Nutrients 2017, 9, 907. [Google Scholar] [CrossRef]
  50. Leyva-López, N.; Gutierrez-Grijalva, E.P.; Ambriz-Perez, D.L.; Heredia, J.B. Flavonoids as cytokine modulators: A possible therapy for inflammation-related diseases. Int. J. Mol. Sci. 2016, 17, 921. [Google Scholar] [CrossRef] [PubMed]
  51. Galleano, M.; Verstraeten, S.V.; Oteiza, P.I.; Fraga, C.G. Antioxidant actions of flavonoids: Thermodynamic and kinetic analysis. Arch. Biochem. Biophys. 2010, 501, 23–30. [Google Scholar] [CrossRef] [PubMed]
  52. Nieuwenhuizen, A.G.; Laere, K.M.J.V.; Raggers, R.J. Combination of Flavonoids and Procyanidin for the Reduction of the Mammalian Appetite. U.S. Patent Application No. 10/535,258, 22 June 2006. [Google Scholar]
  53. Harms, M.; Seale, P. Brown and beige fat: Development, function and therapeutic potential. Nat. Med. 2013, 19, 1252–1263. [Google Scholar] [CrossRef] [Green Version]
  54. Virtanen, K.A.; Lidell, M.E.; Orava, J.; Heglind, M.; Westergren, R.; Niemi, T.; Nuutila, P. Functional brown adipose tissue in healthy adults. N. Engl. J. Med. Overseas Ed. 2009, 360, 1518–1525. [Google Scholar] [CrossRef] [PubMed]
  55. Dulloo, A.G.; Seydoux, J.; Girardier, L.; Chantre, P.; Vandermander, J. Green tea and thermogenesis: Interactions between catechinpolyphenols, caffeine and sympathetic activity. Int. J. Obes. 2000, 24, 252–258. [Google Scholar] [CrossRef] [Green Version]
  56. Fernandezmarcos, P.J.; Auwerx, J. Regulation of PGC-1α, a nodal regulator of mitochondrial biogenesis. Am. J. Clin. Nutr. 2011, 93, S884–S890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Zhang, X.; Zhang, Q.X.; Wang, X.; Zhang, L.; Qu, W.; Bao, B.; Liu, C.A.; Liu, J. Dietary luteolin activates browning and thermogenesis in mice through an AMPK/PGC1α pathway-mediated mechanism. Int. J. Obes. 2016, 40, 1841–1849. [Google Scholar] [CrossRef]
  58. Davis, H.C. Can the gastrointestinal microbiota be modulated by dietary fibre to treat obesity? Irish J. Med. Sci. 2018, 187, 393–402. [Google Scholar] [CrossRef] [PubMed]
  59. Rothschild, D.; Weissbrod, O.; Barkan, E.; Kurilshikov, A.; Korem, T.; Zeevi, D.; Costea, P.I.; Godnea, A.; Kalka, N.; Bar, I.N. Environment dominates over host genetics in shaping human gut microbiota. Nature 2018, 555, 210–215. [Google Scholar] [CrossRef]
  60. Musso, G.; Gambino, R.; Cassader, M. Interactions between gut microbiota and host metabolism predisposing to obesity and diabetes. Annu. Rev. Med. 2011, 62, 361–380. [Google Scholar] [CrossRef]
  61. Canfora, E.E.; Jocken, J.W.; Blaak, E.E. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat. Rev. Endocrinol. 2015, 11, 577. [Google Scholar] [CrossRef] [PubMed]
  62. Wahlstrom, A.; Sayin, S.I.; Marschall, H.U.; Backhed, F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 2016, 24, 41–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Haslam, D.W.; James, W.P. Obesity—Review. Lancet 2005, 366, 1197–1209. [Google Scholar] [CrossRef]
  64. Rowinsky, M.D.; Eric, K. The development and clinical utility of the taxane class of antimicrotubule chemotherapy agents. Annu. Rev. Med. 1997, 48, 353–374. [Google Scholar] [CrossRef] [PubMed]
  65. Lipina, C.; Hundal, H.S. Carnosic acid stimulates glucose uptake in skeletal muscle cells via a PME-1/PP2A/PKB signalling axis. Cell Signal. 2014, 26, 2343–2349. [Google Scholar] [CrossRef]
  66. De Vincenzi, M.; Maialetti, F.; Silano, M. Constituents of aromatic plants: Teucrin. Fitoterapia 2003, 74, 746–749. [Google Scholar] [CrossRef]
  67. Park, M.Y.; Sung, M.K. Carnosic acid attenuates obesity induced glucose intolerance and hepatic fat accumulation by modulating genes of lipid metabolism in C57BL/6J-ob/ob mice. J. Sci. Food Agric. 2015, 95, 828–835. [Google Scholar] [CrossRef] [PubMed]
  68. Johnson, R.K. Dietary intake—How do we measure what people are really eating? Obesity 2002, 10, 63S. [Google Scholar] [CrossRef]
  69. Na, M.; Oh, W.K.; Kim, Y.H.; Cai, X.F.; Kim, S.; Kim, B.Y.; Ahn, J.S. Inhibition of protein tyrosine phosphatase 1B by diterpenoids isolated from Acanthopanax koreanum. Bioorg. Med. Chem. Lett. 2006, 16, 3061–3064. [Google Scholar] [CrossRef]
  70. Kim, G.N.; Shin, M.R.; Shin, S.H.; Lee, A.R.; Lee, J.Y.; Seo, B.I.; Roh, S.S. Study of antiobesity effect through inhibition of pancreatic lipase activity of Diospyros kaki fruit and Citrus unshiu peel. BioMed Res. Int. 2016, 2016, 1723042. [Google Scholar] [CrossRef] [Green Version]
  71. Ninomiya, K.; Matsuda, H.; Shimoda, H.; Nishida, N.; Kasajima, N.; Yoshino, T.; Yoshikawa, M. Carnosic acid, a new class of lipidabsorption inhibitor from sage. Bioorg. Med. Chem. Lett. 2004, 14, 1943–1946. [Google Scholar] [CrossRef]
  72. Lee, E.J.; Hwang, J.S.; Kang, E.S.; Lee, S.B.; Hur, J.; Lee, W.J.; Choi, M.J.; Kim, J.T.; Seo, H.G. Nanoemulsions improve the efficacy of turmeric in palmitate- and high fat diet-induced cellular and animal models. Biomed. Pharmacother. 2019, 110, 181–189. [Google Scholar] [CrossRef]
  73. Gaya, M.; Repetto, V.; Toneatto, J.; Anesini, C.; Piwien-Pilipuk, G.; Moreno, S. Antiadipogenic effect of carnosic acid, a natural compound present in Rosmarinus officinalis, is exerted through the C/EBPs and PPARγ pathways at the onset of the differentiation program. Biochim. Biophy. Acta 2013, 1830, 3796–3806. [Google Scholar] [CrossRef]
  74. Li, S.; Tan, H.Y.; Wang, N.; Cheung, F.; Hong, M.; Feng, Y. The potential and action mechanism of polyphenols in the treatment of liver diseases. Oxid. Med. Cell. Longev. 2018, 2018, 8394818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Takahashi, Y.; Kushiro, M.; Shinohara, K.; Ide, T. Dietary conjugated linoleic acid reduces body fat mass and affects gene expression of proteins regulating energy metabolism in mice. Comp. Biochem. Phys. B 2002, 133, 395–404. [Google Scholar] [CrossRef]
  76. Kim, J.H.; Kim, O.K.; Yoon, H.G.; Park, J.; You, Y.; Kim, K.; Jun, W. Anti-obesity effect of extract from fermented Curcuma longa L. through regulation of adipogenesis and lipolysis pathway in high-fat diet-induced obese rats. Food Nutr. Res. 2016, 60, 30428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Seo, D.B.; Jeong, H.W.; Kim, Y.J.; Kim, S.; Kim, J.; Lee, J.H.; Lee, S.J. Fermented green tea extract exhibits hypolipidaemic effects through the inhibition of pancreatic lipase and promotion of energy expenditure. Br. J. Nutr. 2017, 117, 177–186. [Google Scholar] [CrossRef] [Green Version]
  78. Rahman, H.A.; Sahib, N.G.; Saari, N.; Abas, F.; Ismail, A.; Mumtaz, M.W.; Hamid, A.A. Anti-obesity effect of ethanolic extract from Cosmos caudatus Kunth leaf in lean rats fed a high fat diet. BMC Complement Altern. Med. 2017, 17, 1–17. [Google Scholar] [CrossRef] [Green Version]
  79. Kilany, O.E.; Abdelrazek, H.M.; Aldayel, T.S.; Abdo, S.; Mahmoud, M.M. Anti-obesity potential of Moringa olifera seed extract and lycopene on high fat diet induced obesity in male Sprauge Dawely rats. Saudi J. Biol. Sci. 2020, 27, 2733–2746. [Google Scholar] [CrossRef]
  80. Perumal, K.V.; Ja’afar, N.L.; Mat Taib, C.N.; Shafie, N.H.; Bahari, H. Antiobesity activity of Elateriospermum tapos shell extract in obesity-induced Sprague dawley rats. Molecules 2021, 26, 321. [Google Scholar] [CrossRef]
  81. Wu, Y.; Tan, F.; Zhang, T.; Xie, B.; Ran, L.; Zhao, X. The anti-obesity effect of lotus leaves on high-fat-diet-induced obesity by modulating lipid metabolism in C57BL/6J mice. Appl. Biol. Chem. 2020, 63, 1–11. [Google Scholar] [CrossRef]
  82. He, X.; Zheng, S.; Sheng, Y.; Miao, T.; Xu, J.; Xu, W.; Zhao, C. Chlorogenic acid ameliorates obesity by preventing energy balance shift in high-fat diet induced obese mice. J. Sci. Food Agric. 2021, 101, 631–637. [Google Scholar] [CrossRef]
  83. Sung, J.; Jeong, H.S.; Lee, J. Effect of the capsicoside G-rich fraction from pepper (Capsicum annuum L.) seeds on high-fat diet-induced obesity in mice. Phyther. Res. 2016, 11, 1848–1855. [Google Scholar] [CrossRef]
  84. Suh, D.H.; Jung, E.S.; Park, H.M.; Kim, S.H.; Lee, S.; Jo, Y.H.; Lee, C.H. Comparison of metabolites variation and antiobesity effects of fermented versus nonfermented mixtures of Cudrania tricuspidata, Lonicera caerulea, and soybean according to fermentation in vitro and in vivo. PLoS ONE 2016, 11, e0149022. [Google Scholar] [CrossRef] [PubMed]
  85. Yimam, M.; Jiao, P.; Hong, M.; Brownell, L.; Lee, Y.C.; Hyun, E.J.; Jia, Q. Appetite suppression and antiobesity effect of a botanical composition composed of Morus alba, Yerba mate, and Magnolia officinalis. J. Obes. 2016, 2016, 4670818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Yimam, M.; Jiao, P.; Hong, M.; Brownell, L.; Lee, Y.C.; Kim, H.J.; Jia, Q. A botanical composition from Morus alba, Ilex paraguariensis, and Rosmarinus officinalis for body weight management. J. Med. Food 2017, 11, 1100–1112. [Google Scholar] [CrossRef] [PubMed]
  87. Cho, B.Y.; Park, M.R.; Lee, J.H.; Ra, M.J.; Han, K.C.; Kang, I.J.; Lee, O.H. Standardized Cirsium setidens Nakai Ethanolic extract suppresses adipogenesis and regulates lipid metabolisms in 3T3-L1 adipocytes and C57BL/6J mice fed high-fat diets. J. Med. Food 2017, 20, 763–776. [Google Scholar] [CrossRef]
  88. Kim, H.J.; Hong, S.H.; Chang, S.H.; Kim, S.; Lee, A.Y.; Jang, Y.; Cho, M.H. Effects of feeding a diet containing Gymnema sylvestre extract: Attenuating progression of obesity in C57BL/6J mice. Asian Pac. J. Trop. Med. 2016, 9, 437–444. [Google Scholar] [CrossRef]
  89. Ding, Y.; Wang, L.; Im, S.; Hwang, O.; Kim, H.S.; Kang, M.C.; Lee, S.H. Anti-obesity effect of diphlorethohydroxycarmalol isolated from brown alga Ishige okamurae in high-fat diet-induced obese mice. Mar. Drugs 2019, 17, 637. [Google Scholar] [CrossRef] [Green Version]
  90. Su, M.; Huang, W.; Zhu, B. Acetylshikonin from Zicao prevents obesity in rats on a high-fat diet by inhibiting lipid accumulation and inducing lipolysis. PLoS ONE 2016, 11, e0146884. [Google Scholar] [CrossRef] [Green Version]
  91. Jeong, E.J.; Jegal, J.; Ahn, J.; Kim, J.; Yang, M.H. Anti-obesity effect of Dioscorea oppositifolia extract in high-fat diet-induced obese mice and its chemical characterization. Biol. Pharm. Bull. 2016. [Google Scholar] [CrossRef] [Green Version]
  92. Choi, D.H.; Han, J.H.; Yu, K.H.; Hong, M.; Lee, S.Y.; Park, K.H.; Kwon, T.H. Antioxidant and anti-obesity activities of Polygonum cuspidatum extract through alleviation of lipid accumulation on 3T3-L1 adipocytes. J. Microbiol. Biotechnol. 2020, 30, 21–30. [Google Scholar] [CrossRef] [PubMed]
  93. Ku, S.K.; Sung, S.H.; Choung, J.J.; Choi, J.S.; Shin, Y.K.; Kim, J.W. Anti-obesity and anti-diabetic effects of a standardized potato extract in ob/ob mice. Exp. Ther. Med. 2016, 12, 354–364. [Google Scholar] [CrossRef] [Green Version]
  94. Marrelli, M.; Menichini, F.; Conforti, F. Hypolipidemic and antioxidant properties of hot pepper flower (Capsicum annuum L.). Plant Foods Hum. Nutr. 2016, 71, 301–306. [Google Scholar] [CrossRef]
  95. Vitalone, A.; Di Sotto, A.; Mammola, C.L.; Heyn, R.; Miglietta, S.; Mariani, P.; Mazzanti, G. Phytochemical analysis and effects on ingestive behaviour of a Caralluma fimbriata extract. Food Chem. Toxicol. 2017, 108, 63–73. [Google Scholar] [CrossRef]
  96. Lima, N.D.S.; Numata, E.D.P.; Mesquita, L.M.D.S.; Dias, P.H.; Vilegas, W.; Gambero, A.; Ribeiro, M.L. Modulatory effects of guarana (Paullinia cupana) on adipogenesis. Nutrients 2017, 9, 635. [Google Scholar] [CrossRef] [Green Version]
  97. Han, M.H.; Jeong, J.S.; Jeong, J.W.; Choi, S.H.; Kim, S.O.; Hong, S.H.; Choi, Y.H. Ethanol extracts of Aster yomena (Kitam.) Honda inhibit adipogenesis through the activation of the AMPK signaling pathway in 3T3-L1 preadipocytes. Drug Discov. Ther. 2017, 5, 281–287. [Google Scholar]
  98. Kyung Choe, W.; Tae Kang, B.; Ok Kim, S. Water-extracted plum (Prunus salicina L. cv. Soldam) attenuates adipogenesis in murine 3T3-L1 adipocyte cells through the PI3K/Akt signaling pathway. Exp. Ther. Med. 2018, 15, 1608–1615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Gupta, A.; Kumar, A.; Kumar, D.; Nandan, S.; Shankar, K.; Varshney, S.; Gaikwad, A.N. Ethyl acetate fraction of Eclipta alba: A potential phytopharmaceutical targeting adipocyte differentiation. Biomed. Pharmacother. 2017, 96, 572–583. [Google Scholar] [CrossRef] [PubMed]
  100. Jaradat, N.; Zaid, A.N.; Hussein, F.; Zaqzouq, M.; Aljammal, H.; Ayesh, O. Anti-lipase potential of the organic and aqueous extracts of ten traditional edible and medicinal plants in Palestine—A comparison study with orlistat. Medicines 2017, 4, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Mangal, P.; Khare, P.; Jagtap, S.; Bishnoi, M.; Kondepudi, K.K.; Bhutani, K.K. Screening of six Ayurvedic medicinal plants for anti-obesity potential: An investigation on bioactive constituents from Oroxylum indicum (L.) Kurz bark. J. Ethnopharmacol. 2017, 197, 138–146. [Google Scholar] [CrossRef]
  102. Wen, C.; Wang, D.; Li, X.; Huang, T.; Huang, C.; Hu, K. Targeted isolation and identification of bioactive compounds lowering cholesterol in the crude extracts of crabapples using UPLC-DAD-MS-SPE/NMR based on pharmacology-guided PLS-DA. J. Pharm. Biomed. Anal. 2018, 150, 144–151. [Google Scholar] [CrossRef]
  103. El-Shiekh, R.A.; Al-Mahdy, D.A.; Hifnawy, M.S.; Abdel-Sattar, E.A. In-vitro screening of selected traditional medicinal plants for their anti-obesity and anti-oxidant activities. S. Afr. J. Bot. 2019, 123, 43–50. [Google Scholar] [CrossRef]
  104. Bais, S.; Patel, N.J. In vitro anti diabetic and anti obesity effect of J. communis extract on 3T3L1 mouse adipocytes: A possible role of MAPK/ERK activation. Obes. Med. 2020, 18, 100219. [Google Scholar] [CrossRef]
  105. Podsędek, A.; Zakłos-Szyda, M.; Polka, D.; Sosnowska, D. Effects of Viburnum opulus fruit extracts on adipogenesis of 3T3-L1 cells and lipase activity. J. Funct. Foods 2020, 73, 104111. [Google Scholar] [CrossRef]
  106. Hussein, M.M.; Samy, M.; Arisha, A.H.; Saadeldin, I.M.; Alshammari, G.M. Anti-obesity effects of individual or combination treatment with Spirulina platensis and green coffee bean aqueous extracts in high-fat diet-induced obese rats. All Life 2020, 13, 328–338. [Google Scholar] [CrossRef]
  107. Wang, L.; Guan, X.Q.; He, R.J.; Huo, P.C.; Qin, W.W.; Cui, L.T.; Ge, G.B. Discovery and characterization of pentacyclic triterpenoid acids in Styrax as potent and reversible pancreatic lipase inhibitors. J. Funct. Foods 2020, 74, 104159. [Google Scholar] [CrossRef]
  108. Lee, J.; Kong, B.; Lee, S.H. Patchouli alcohol, a compound from Pogostemon cablin, inhibits obesity. J. Med. Food 2020, 23, 326–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Coronado-Cáceres, L.J.; Rabadán-Chávez, G.; Mojica, L.; Hernández-Ledesma, B.; Quevedo-Corona, L.; Lugo Cervantes, E. Cocoa seed proteins’(Theobroma cacao L.) anti-obesity potential through lipase inhibition using in silico, in vitro and in vivo models. Foods 2020, 9, 1359. [Google Scholar] [CrossRef]
  110. Jaradat, N.; Qneibi, M.; Hawash, M.; Sawalha, A.; Qtaishat, S.; Hussein, F.; Issa, L. Chemical composition, antioxidant, antiobesity, and antidiabetic effects of Helichrysum sanguineum (L.) Kostel. from Palestine. Arab J. Sci. Eng. 2021, 46, 41–51. [Google Scholar] [CrossRef]
  111. Park, Y.H.; An, M.; Kim, J.K.; Lim, Y.H. Antiobesity effect of ethanolic extract of Ramulus mori in differentiated 3T3-L1 adipocytes and high-fat diet-induced obese mice. J. Ethnopharmacol. 2020, 251, 112542. [Google Scholar] [CrossRef] [PubMed]
  112. Kanwal, S.; Aliya, S.; Xin, Y. Anti-obesity effect of Dictyophora indusiata mushroom polysaccharide (DIP) in high fat diet-induced obesity via regulating inflammatory cascades and intestinal microbiome. Front. Endocrinol. 2020, 11, 558874. [Google Scholar] [CrossRef]
  113. Kang, M.C.; Lee, H.G.; Kim, H.S.; Song, K.M.; Chun, Y.G.; Lee, M.H.; Jeon, Y.J. Anti-obesity effects of Sargassum thunbergii via downregulation of adipogenesis gene and upregulation of thermogenic genes in high-fat diet-induced obese mice. Nutrients 2020, 12, 3325. [Google Scholar] [CrossRef]
  114. Lee, H.G.; Lu, Y.A.; Li, X.; Hyun, J.M.; Kim, H.S.; Lee, J.J.; Jeon, Y.J. Anti-Obesity effects of Grateloupia elliptica, a red seaweed, in mice with high-fat diet-induced obesity via suppression of adipogenic factors in white adipose tissue and increased thermogenic factors in brown adipose tissue. Nutrients 2020, 12, 308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Kim, M.; Im, S.; Choi, C.; Son, Y.; Kwon, D.; Jung, Y.S.; Lee, Y.H. Anti-obesity effects of soybean embryo extract and enzymatically-modified isoquercitrin. Biomolecules 2020, 10, 1394. [Google Scholar] [CrossRef]
  116. Song, M.; Yang, G.; Hoa, T.Q.; Hieu, H.D.; Amin, A.S.M.; Choe, W.; Ha, J. Anti-obesity effect of fermented persimmon extracts via activation of AMP-activated protein kinase. Biol. Pharmaceut. Bull. 2020, 43, 440–449. [Google Scholar] [CrossRef]
  117. Zheng, X.; Zhu, J.; Zhang, X.; Cheng, M.; Zhang, Z.; Cao, J. The modulatory effect of nanocomplexes loaded with EGCG3″Me on intestinal microbiota of high fat diet-induced obesity mice model. J. Food Biochem. 2018, 42, e12501. [Google Scholar] [CrossRef]
  118. Abdelbaky, M.S.M.E.; Ibrahim, H.S.; Hassan, M.L.; Sayed, Z.E. Nanoparticles effects of red grape (Vitis vinifera) seeds and grape seeds powder on obese hyperlipidemic rats. ARC J. Nutr. Growth 2016, 2, 1–15. [Google Scholar]
  119. Gao, L.; Hu, Y.; Hu, D.; Li, Y.; Yang, S.; Dong, X.; Liu, H. Anti-obesity activity of gold nanoparticles synthesized from Salacia chinensis modulates the biochemical alterations in high-fat diet-induced obese rat model via AMPK signaling pathway. Arab. J. Chem. 2020, 13, 6589–6597. [Google Scholar] [CrossRef]
  120. Ansari, S.; Bari, A.; Ullah, R.; Mathanmohun, M.; Veeraraghavan, V.P.; Sun, Z. Gold nanoparticles synthesized with Smilax glabra rhizome modulates the anti-obesity parameters in high-fat diet and streptozotocin induced obese diabetes rat model. J. Photochem. Photobiol. B Biol. 2019, 201, 111643. [Google Scholar] [CrossRef] [PubMed]
  121. Zhang, H.L.; Zhong, X.B.; Tao, Y.; Wu, S.H.; Su, Z.Q. Effects of chitosan and water-soluble chitosan micro-and nanoparticles in obese rats fed a high-fat diet. Int. J. Nanomed. 2012, 7, 4069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Simu, S.Y.; Ahn, S.; Castro-Aceituno, V.; Singh, P.; Mathiyalangan, R.; Jiménez-Pérez, Z.E.; Yang, D.C. Gold nanoparticles synthesized with fresh panax ginseng leaf extract suppress adipogenesis by downregulating PPAR/CEBP signaling in 3T3-L1 mature adipocytes. J. Nanosci. Nanotechnol. 2018, 18, 1–8. [Google Scholar]
  123. Li, W.; Wan, H.; Yan, S.; Yan, Z.; Chen, Y.; Guo, P.; Ning, L. Gold nanoparticles synthesized with Poria cocos modulates the anti-obesity parameters in high-fat diet and streptozotocin induced obese diabetes rat model. Arab. J. Chem. 2020, 13, 5966–5977. [Google Scholar] [CrossRef]
  124. Mukhopadhyay, P.; Maity, S.; Mandal, S.; Chakraborti, A.S.; Prajapati, A.K.; Kundu, P.P. Preparation, characterization and in vivo evaluation of pH sensitive, safe quercetin-succinylated chitosanalginate core-shell-corona nanoparticle for diabetes treatment. Carbohydr. Polym. 2018, 182, 42–51. [Google Scholar] [CrossRef] [PubMed]
  125. Zhang, J.; Nie, S.; Zu, Y.; Abbasi, M.; Cao, J.; Li, C.; Wu, D.; Labib, S.; Brackee, G.; Shen, C.L.; et al. Anti-atherogenic effects of CD36-targeted epigallocatechin gallate-loaded nanoparticles. J. Controll. Release 2019, 303, 263–273. [Google Scholar] [CrossRef] [PubMed]
  126. Zhang, J.; Nie, S.; Martinez-Zaguilan, R.; Sennoune, S.R.; Wang, S. Formulation, characteristics and antiatherogenic bioactivities of CD36-targeted epigallocatechin gallate (EGCG)-loaded nanoparticles. J. Nutr. Biochem. 2016, 30, 14–23. [Google Scholar] [CrossRef] [Green Version]
  127. El-Naggar, M.E.; Al-Joufi, F.; Anwar, M.; Attia, M.F.; El-Bana, M.A. Curcumin-loaded PLA-PEG copolymer nanoparticles for treatment of liver inflammation in streptozotocin-induced diabetic rats. Colloids Surf. B 2019, 177, 389–398. [Google Scholar] [CrossRef] [PubMed]
  128. Chitkara, D.; Nikalaje, S.K.; Mittal, A.; Chand, M.; Kumar, N. Development of quercetin nanoformulation and in vivo evaluation using streptozotocin induced diabetic rat model. Drug Deliv. Transl. Res. 2012, 2, 112–123. [Google Scholar] [CrossRef]
  129. Wan, S.; Zhang, L.; Quan, Y.; Wei, K. Resveratrol-loaded PLGA nanoparticles: Enhanced stability, solubility and bioactivity of resveratrol for non-alcoholic fatty liver disease therapy. R. Soc. Open Sci. 2018, 5, 181457. [Google Scholar] [CrossRef] [Green Version]
  130. Yi, H.M.; Simu, S.Y.; Ahn, S.; Aceituno, V.V.; Wang, C.; Mathhiyalagan, R.; Hurh, J.; Batjikh, I.; Ali, H.; Kim, S.; et al. Anti-obesity effect of gold nanoparticles from Dendropanax morbifera Léveille by suppression of triglyceride synthesis and downregulation of PPARγ and CEBPα signaling pathways in 3T3-L1 mature adipocytes and HepG2 cells. Curr. Nanosci. 2020, 16, 2. [Google Scholar] [CrossRef]
  131. Zu, Y.; Zhao, L.; Hao, L.; Mechref, Y.; Zabet-Moghaddam, M.; Keyel, P.A.; Wang, S. Browning white adipose tissue using adipose stromal cell-targeted resveratrol-loaded nanoparticles for combating obesity. J. Controll. Release 2021, 333, 339–351. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Obesity regulatory factors.
Figure 1. Obesity regulatory factors.
Molecules 26 03278 g001
Figure 2. List of various parameters for assessing the anti-obesity efficacy of various natural products.
Figure 2. List of various parameters for assessing the anti-obesity efficacy of various natural products.
Molecules 26 03278 g002
Figure 3. Diagram showing the different steps mainly regulated by polyphenols in managing obesity.
Figure 3. Diagram showing the different steps mainly regulated by polyphenols in managing obesity.
Molecules 26 03278 g003
Figure 4. Diagram showing the primary mechanism that flavonoids have anti-obesity effects via different pathways.
Figure 4. Diagram showing the primary mechanism that flavonoids have anti-obesity effects via different pathways.
Molecules 26 03278 g004
Figure 5. The anti-obesity mechanisms of diterpenoids.
Figure 5. The anti-obesity mechanisms of diterpenoids.
Molecules 26 03278 g005
Table 1. List of different plants with anti-obesity effects with the list of secondary metabolites responsible for this biological activity.
Table 1. List of different plants with anti-obesity effects with the list of secondary metabolites responsible for this biological activity.
Sr. No.Plant Secondary MetaboliteExperimental ModelImportant Findings References
1.Rhizome of Curcuma longa (Zingiberaceae)CurcuminSprague-Dawley rats Body weight gain, perirenal and epididymal adipose tissue weight decreased (p ≤ 0.05)
PPAR-γ (71%), C/EBPα (38.83%), FAS (66.03%), ACC (35%) level decreased
AMPK (12.93%), Adiponectin (49.01%) an CPT1 (88%) increased
TG and LDL level decreased (p ≤ 0.05)
[76]
2.Leaves of Gymenma sylvestra (Apocynaceae)Deacyl gymnemic acidC57BL/6J miceBody weight gain, epididymal fat decreased, lowers food and energy efficiency ratio
TC (30.44%), TG (18.53%),
LDL (33.69%), Leptin (35.52%) decreased and HDL (20.26%) increased
[88]
3.Root, root cortices and root corks of Zicao (Boraginaceae)AcetylshikoninSprague-Dawley rats and 3T3-L1 adipocytesFood efficiency ratio decreased to 10.0 ± 0.14%,
Body weight gain, Epididymal adipose tissue weight (18.7 ± 0.23%), adipocytes size (p ≤ 0.001) at 100 mg/kg
FFA (35.5%) and serum TG level (39.9%) reduced at 900 mg/kg
Lipolysis (0.85 fold), glycerol secretion (0.50 folds) increased,
PPAR-γ and C/EBP reduced at 1.5 µM (p ≤ 0.05), Adipocytes differentiation decreased
[90]
4.Rhizomes of Dioscorea oppositifolia (Dioscoreaceae)(3R, 5R)-3,5 dihydroxy-1,7-bis (4-hydroxyphenyl)-3,5-heptanediol and 3,5-dimethoxy-2,7-phenanthrenediolMiceBody weight gain (45.2%) and adipose tissue weight (22.5%) decreased,
TG (18.8%), TC (15.4%), hepatic TG (48.1%) and Hepatic TC (46.9%) decreased, HDL-c (34.2%) increased at 100 mg/kg dose
[91]
5.Leaves of Peucedanum japonicum thumb (Umbelliferae)Cis-3′,4′-diisovalerylkhellactone (cDIVK)3T3-L1 adipocytes TG (65%), adipocyte differentiation (80%) decreased
AMPK phosphorylation increased (103%)
C/EBPα, PPARγ and SREB-1c (p ≤ 0.05)
Inhibition of α-glucosidase activity at 50 µM concentration
[92]
6.Seeds of Capsicum annuum (Solanaceae)Capsicoside G-rich protein (CRP-13.35%)C57BL/6J mice Body weight, food consumption and food efficiency decreased (p ≤ 0.05)
Adipocyte and epididymal adipose tissue weight decreased
TG, TC, AST, ALT level decreased (p ≤ 0.05)
C/EBPα, PPARγ, SREBP1c, FAS decreased (p ≤ 0.05)
[83]
7.Tubers of Solanum tuberosum (Solanaceae)Proteinase inhibitors MiceBody weight gain (18.5%), food consumption (39.93%), epididymal fat (52.40%), TC (19.65%), FFA (33.33%), TG (28.60%), leptin (7.35%) and adipocyte differentiation decreased at 300 mg/kg
Adiponectin increased (33.62%)
[93]
8.Fruit of Diospyros kaki (Ebenaceae) and peel of Citrus unshui (Rutaceae)Flavonoids and phenolic compounds MiceBody weight gain decreased (16.62%) visceral fat weight (12.52%) decreased and pancreatic lipase inhibited (about 50%) at 500 µg/mL
TC, TG, LDL decreased and HDL increased (p ≤ 0.05) at 50 mg/kg
[70]
9.Leaves of Cudrania tricuspidata (Moraceae
and Lonicera caerulea (Caprifoliaceae) and whole soyabean plant of Glycine hispida (Fabaceae)
Anthocyanins, flavonoids, phenolic compounds, anthoxanthines, prenylated flavonoids, isoflavone and soyasaponins C57BL/6J miceSubcutaneous fat (37.10%), Adipose area (39.85%), Adipose tissue (25.22%), TC (10.40%), TG (53.80%), LDL (14.56%), ALT (38.46%), Leptin (66.36% decreased)[84]
10.Flower of Capsicum annuum (Solanaceae)Phenolic compound, flavonoids, chlorogenic acid and quercetin Macrophages RAW-264.7Pancreatic lipase inhibited (IC50 = 3.54 ± 0.18) to 82.54% at 10 mg/mL
Inhibition of lipids peroxidation (IC50 = 27.61 ± 2.25 µg/mL) after 30 min and (IC50 = 41.69 ± 1.13 µg/mL) after 60 min of incubation
[94]
11.Leaves of Morus alba (Moraceae), root bark of Yerba mate (Aquifoliaceae) and stem bark of Magnolia officinalis (Magnoliaceae)Xanthines, purines, alkaloids, flavonoids, polyphenols C57BL/6J mice Body weight gain (98.6%) and relative weight of body organs (p ≤ 0.001) decreased,
TC (18.6%), LDL (59%), ALT (60.1%), AST (35.2%), insulin (75.9%), leptin (46.8%) decreased and ghrelin hormones increased (4.2 folds) at 350 mg/kg
[85]
12.Aerial part of Caralluma fimbriata (Asclepiadeceae) Organic acid, amino acid, carbohydrates, pregnane and trigonelline RatDecreased the hypothalamic level of NPY, ORX peptide
Insulin level (29.52%) increased
AST (22.61%), ALT (12.12%) ALP 22.07%) and Lipase (7.5%) decreased
[95]
13.Paullinia cupana (Sapindaceae)Phenolics, flavonoids, caffeine, theobranium, theophylline, tannins, saponins, catechins, epicatechins, proanthocyanins 3T3-L1 adipocytes TG accumulation decreased at 300 µg/mL
Upregulated the expression of anti-adipogenic genes FOXO1, Gata3, Dlk1, Wnt1, Wnt3a, Wnt10b
Decreased the expression of adipogenic markers PPAR-γ and CEBP-α and SREBP-1
[96]
14.Leaves of Cirsium setidens (Asteraceae) Pectolinarin 3T3-L1 adipocytes and C57BL/6J mice Lipid accumulation (56%) and body weight gain (18.46%) decreased at 200 µg/mL,
AST (3.1%), ALT (4.15%), TC (8.73%), TG (22.45%) decreased,
Expression of PPARα, C/EBPα, C/EBP-δ and FABP4 downregulated at 200 µg/mL
[87]
15.Root bark of Morus alba (Moraceae), leaves of Ilex paraguariensis (Aquifoliaceae) and leaves of Rosmarinus officinalis (Lamiaceae)Bioflavonoids, morusin, Kuwanon G, carnosol, carnosic acid, caffeine, polyphenols, xanthines, purines and alkaloids C57BL/6J miceBody weight (30.2%), TC (21.1%), TG (44.6%), LDL (38.2%) decreased at 800 mg/kg,
Level of leptin (16.4%) and ghrelin (2.1 folds) decreased and of insulin (65.5%) increased
[86]
16.Leaves of Aster yomena (Asteraceae)Phenolics compounds and terpenoids 3T3-L1 adipocytes Expression of PPARγ, C/EBPα, C/EBPβ and SREBP-1c decreases significantly in a conc. dependent manner (p ≤ 0.05),
Phosphorylation of AMPK increased,
Lipids accumulation reduced
[97]
17.Leaves, stem and roots of Ecklonia cava (Lessoniaceae)Phlorotannins, fucodiphloroethal C57BL/6N Mice Body weight (0.82 fold), liver weight (2.14 ±0.31 to 0.97 ± 0.14 g), epididymal adipose tissue (0.29 ± 0.02 to 0.20 ± 0.02), perirenal adipose tissue (0.63 ± 0.08 to 0.33 ± 0.05 g), TC (83.6 ± 6.10 to 59.9 ± 16.6 mg/dL), TG (57.9 ± 12.16 to 38.0 ± 5.05 mg/dL) decreased at 150 mg/kg
HDL-c increased (38.4 ± 3.87 to 61.2 ± 7.39 mg/dL), level of SREBP-1c, FABP, FAS decreased and AMPK phosphorylation increased (p ≤ 0.05)
[47]
18.Plums of Prunus salicina (Rosaceae)Phenolic compounds, flavonoids, chlorogenic acid, hydroxycinnamic acid, epicatechin, quercetin3T3-L1 adipocytes Adipocytes differentiation decreased, PPAR-γ, C/EBP-α, β-actin, decreased and AMPK phosphorylation increased (p ≤ 0.05)[98]
19.Whole plant of Eclipta alba (Asteraceae)Demethylwedelolactone derivative, isodemethylwedelolactane, apigenin, isoechinmocystic acid-3-O-glycoside, Echinocystic acid 3T3-L1 adipocytes Expression of PPARα (1.9 folds), C/EBPα (1.8 folds), FAS (1.4 folds), FABP4 (1.8 folds) decreased
TC, TG and LDL decreased (p ≤ 0.05)
[99]
20Leaves of Vitis vinifera (Vitaceae)Phenolic compounds, phenolic acid, flavonoids-trans caffeoyl tartaric acid, myricetin-3-O-glycoside, quercetin-3-O glucoside, kaempferol-3-O-glucoside and isorhamnetin Enzyme/s Pancreatic lipase enzyme inhibited with IC50 14.1 ± 1.9 µg/mL[100]
21.Fruits of Rhus coriaria (Anacardiaceae)Caffeoylquinic acid, quercetin, rhamnetin, myricetin, kaempferol, gallic acid, methyl gallate, m-digallic acid and amenthoflavone Enzyme/s Pancreatic lipase enzyme inhibited with IC50 19.95 ± 1 µg/mL[100]
22.Organic extract of Origanum dayiVolatile oils, linalyl acetate terpinen-4-ol, α-terpineol, (E)-sabinene hydrate acetate,
1,8-cineole, (E)-sabinene hydrate
Enzyme/sPancreatic lipase enzyme inhibited with IC50 26.9 ± 2.80 µg/mL[100]
23. Leaves of Bacillus fermented Camellia sinensis (Theaceae) green tea Catechins, caffeine, gallocatechins, EGCG, Sprague-Dawley ratsInhibition of pancreatic lipase with IC50 of 0.48 mg/mL
Post prandial lipaemia reduced
Expression of PPAR-α (0.2 folds), SREBP-1c (0.7 folds), ACC (1.7 folds), FAS (1.7 folds) decreased
Acetyl Co~A and CPT1 increased
[77]
24.Leaves of Cosmos cadatus Kunth (Asteraceae)Catechins, quercetin, rutin, kaempferol, chlorogenic acid Sprague-Dawley ratsBody weight gain (60%) and visceral fat decreased at 350 mg/ kg
Ghrelin level and leptin level (45.38%) decreased
AST (158.90 ± 48.09 to 93.50 ± 11.25 mg/dL), ALT (45.95 ± 7.51 to 33.25 ± 6.80 mg/dL) and ALP (94.00 ± 11.17 to 79.75 ± 7.59 mg/dL) decreased at 175 mg/kg
[78]
25.Bark of Oroxylum indicum (Bignoniaceae)Oroxylin A, Chrysin and Baicalein 3T3-L1 adipocytes and Enzyme/sInhibition of pancreatic lipase (IC50 = 78.03 ± 1.67 µg/mL) and lipid accumulation decreased (IC50 = 70.13 ± 2.27 µg/mL)
Downregulation of PPAR-γ, C/EBP-α
[101]
26.Fruits of Malus prunifolia (Rosaceae)Citric acid, p-coumaric acid, hyperoside, myricetin, naringenin, quercetin. Kaempferol, gentiopicroside, 8-epiloganic acid MiceRegulated the serum lipid parameters,
TC (p ≤ 0.01), LDL-C (p ≤ 0.01) level decreased and HDL-C (p ≤ 0.05) increased
[102]
27.Fruits of Malus huphenesis (Rosaceae) p-coumaric acid, 8-epiloganic acid, hyperoside, myricetin, naringenin, Kaempferol, quercetin, gentiopicroside, MiceRegulated the serum lipid parameters,
TC (p ≤ 0.05), LDL-C (p ≤ 0.05) level decreased and HDL-C increased
[102]
28.Caralluma quadrangula (Apocyanaceae)Phenolics, flavonoids, tannins and steroidal contents Enzyme/sPancreatic lipase (62.56 ± 0.43%), α-amylase (54.31 ± 0.58%) and α-glucosidase (47.11 ± 1.3%) got inhibited [103]
29. Hibiscus sabdariffa (Malvaceae)Delphinidin-3-sambubioside, cyanidin-3-diglucoside, Delphinidin, Luteolin, quercetin, gossypitrin, chlorogenic acid and protocatechuic acid, ellagic acid, p-coumaric acid, ferulic acid and caffeic acid Enzyme/sPancreatic lipase (67.56 ± 3.63%), α-amylase (64.75 ± 1.2%) and α-glucosidase (64.25 ± 1.7%) got inhibited [103]
30Solenostemma argel (Apocyanaceae) β-sitosterol, β-sitosterol glucoside, stemmoside C, Kaempferol-3-O-neohesperidoside, kaempferol-3-O-glucoside, quercetin-3-O-neohesperidoside,Enzyme/sPancreatic lipase (97.02 ± 1.4%), α-amylase (69.32 ± 1.14%) and α-glucosidase (89.08 ± 1.1%) got inhibited[103]
31.Leaves of Juniperus communis (Cupressaceae) Volatile oils, flavonoids and phenolic compounds3T3-L1 adipocytes α-amylase (97.5%) inhibited with IC50 20.93 ± 1.067 µg/mL and α-glucosidase (98.4%) with IC50 152.93 ± 1.067 µg/mL
Lipase (97.5%) with IC50 69.93 ± 1.067 at 1000 µg/mL
Adipogenesis (67%) at lipids accumulation (69%) inhibited at 160 µg/mL
[104]
32.Fruits of Vibrunum opulus (Adoxaceae)Anthocyanidins, phenolics and flavonoids, quercetin, caffeic acid, dicaffeolquinic acid, neochlorogenic acid, catechin and rutin 3T3-L1 adipocytes Pancreatic lipase (60%) and lipids accumulation (22%) inhibited at 75 µg/mL
Expression of PPAR-γ and leptin decreased (p ≤ 0.001)
[105]
33.Powder of plant Spirrulina platensis (Phormidiaceae) Phospholipids, polyunsaturated fatty acids, provitamins, minerals, proteins and polysaccharidesRatBody weight, liver weight, BMI, hepatosomatic index decreased (p ≤ 0.05)
ALT (134.23 ± 3.56 to 88.79 ± 4.79 U/L), AST (210.50 ± 9.97 to 184.00 ± 9.97 U/L), ALP (157.01 ± 4.43 to 98.27 ± 5.45 U/L) and leptin (0.38 ± 0.02 to 0.26 ± 0.01 ng/mL) decreased
PGC-1α increased, FAS and PPAR-γ and decreased (p ≤ 0.05)
[106]
34.Beans of Coffea arabica (Rubiaceae)p-coumaric acid, quinic acid, chlorogenic acid, caffeine, and caffeic acidRatBody weight, liver weight, BMI, hepatosomatic index decreased (p ≤ 0.05)
ALT (134.23 ± 3.56 to 93.48 ± 5.07 U/L), AST (210.50 ± 9.97 to 193.69 ± 9.55 U/L), ALP (157.01 ± 4.43 to 84.12 ± 2.65 U/L) and leptin (0.38 ± 0.02 to 0.31 ± 0.004 ng/mL) decreased
PGC-1α increased, FAS and PPAR-γ and decreased (p ≤ 0.05)
[106]
35.Mixture of the Spirrulina platensis and Coffea arabicaChlorogenic acid and unsaturated fatty acids like oleic acid, linolenic acid and linoleic acidRatBody weight, liver weight, BMI, hepatosomatic index decreased (p ≤ 0.05)
ALT (134.23 ± 3.56 to 73.37 ± 5.77 U/L), AST (210.50 ± 9.97 to 142.84 ± 9.5 U/L), ALP (157.01 ± 4.43 to 66.77 ± 2.79 U/L) and leptin (0.38 ± 0.02 to 0.17 ± 0.003 ng/mL) decreased
PGC-1α increased, FAS and PPAR-γ and decreased (p ≤ 0.05)
[106]
36.Pentacyclic triterpenoids isolated from the Styrax Betulinic acid, epibetulinic acid, oleanic acid, oleanonic acid, betulonic acid, corosolic acid, maslinic acid, cinnamyl cinnamate, 3-phenylpropyl cinnamate Enzyme/sPancreatic lipase inhibited by Oleanonic acid with IC50-0.49 µM, betulonic acid with IC50-1.48 µM, oleanolic acid with IC50-3.53 µM, maslinic acid with IC50-6.06 µM and corosolic acid with IC50-6.35 µM[107]
37.Pogostemum cablin (Lamiaceae)Sesquiterpene alcohol (Patchouli alcohol)3T3-L1 adipocytes and C57BL/6J miceTG accumulation (71.4%), PPAR-γ (58.8%) and C/EBPα (77.3%) decreased at 100 µM, β-catenin increased (36.6%)
Body weight, epididymal, retroperitoneal, and brown fat weight decreased (p ≤ 0.05)
[108]
38.Seeds oil of Moringa olifera (Moringaceae)Tannins, flavonoids, terpenoids, glycosides and saponinsSprague-Dawley rats Body weight (226.33 ± 8.53 to 201.17 ± 15.82g), Kidney weight (0.30 ± 0.01 to 0.25 ± 0.03 g) and epididymal tissue (0.97 ± 0.05 to 0.90 ± 0.09 g) decreased
TC, ALT, AST, Creatinine and uric acid declined significantly (p ≤ 0.05)
[79]
39.Seeds of Theobroma cocao (Malvaceae)Proteins isolated; Vicillin and albuminMale wistar rats Pancreatic lipase inhibited with IC50 value 1.4 mg /mL
Body weight (301.6 ± 4.60 to 278.5 ± 6.50 g) decreased
Total lipids, TG, TC decreased significantly (p ≤ 0.05) at 150 mg/kg
[109]
40.Chlorogenic acid Phenolic acid C57BL/6J mice ALT, AST, ALP decreased significantly (p ≤ 0.001)
Improves the gut microbiota and contributes to 66 total bacterial species out of 117
Body weight gain decreased (15%) (p ≤ 0.05)
BAT activity increased
[82]
41.Shell extract of Elateriospermum tapos (Euphorbiaceae)3′,4′,5′-trimethoxyflavone, acetyllysine, 7-methoxy chromone, undulatone, aldosterone 18-glucouronide Sprague-Dawley rats Body weight (350.0 ± 15.0 to 288.0 ± 42.8 g), calories intake (9341.9 ± 781.52 to 7971.6 ± 945.46 KJ),
Liver, viscera fat and gonadal fat decreased (p ≤ 0.05),
TC (1.63 ± 0.33 to 1.48 ± 0.16 mMol/L), LDL-C (1.04 ± 0.16 to 0.86 ± 0.20 mMol/L), TG (1.77 ± 0.84 to 1.32 ± 0.23 mMol/L) lipoprotein lipase activity (1.77 ± 0.84 to 1.32 ± 0.23) decreased and HDL-C increased (0.35 ± 0.05 to 0.41 ± 0.08)
[80]
42.Aerial part of Helichrysum sanguineum (Asteraceae)Cardiac glycosides, phenols, volatile oils, tannins,
Steroids, reducing sugar and flavonoids
Enzyme/sα-amylase and pancreatic lipase got inhibited with IC50 value of 28.18 ± 1.04 to 63.09 ± 0.3 µg/mL[110]
43.Twig of Ramulus mori (Lamiaceae)Quercetin 3-β-glycosides, quercetin Mulberroside, oxyresveratrol and resveratrol3T3-L1 adipocytes and MiceBody weight (27.3%), liver weight (17.5%) and epididymal tissue weight (19.5%) decreased at 100 mg/kg
TC (211.5 ± 16.4 to 165.8 ± 7.0 mg/dL), TG (97.7 ± 8.5 to 65.0 ± 14.2 mg/dL), LDL-C (46.3 ± 19.1 to 24.5 ± 13.9 mg/dL) decreased and HDL-C (46.3 ± 19.1 to 24.5 ±13.9 mg/dL) increased
PPAR-γ (0.96 fold), C/EBPα (0.92 fold), SREBP (0.92 fold) decreased and ACC (0.78 fold), FAS (0.96 fold) and SCD (0.84 fold) increased at 40 µg/mL
[111]
44.Leaves of Nelumbo nucifera (Nelumbonaceae)Kaempferitrin, hyperoside, astragalin, phloretin and quercetin C57BL/6J mice ALT, AST, ALP, TG, TC and HDL-C decreased (p ≤ 0.05), HDL-C increased (p ≤ 0.05)
Liver index (4.17 ± 0.22 to 3.93 ± 0.21) and epididymal fat index (1.55 ± 0.28 to 1.14 ± 0.21) decreased,
Expression of PPAR-γ and C/EBPα decreased and PPAR-α, CPT1 and CYP7A1 increased
Level of IL-1β, IL-6, TNFα and INF-γ decreased
IL-4 and IL-10 increased
[81]
45.Dictyophora indusiata (Phallaceae)Polyphenols and polysaccharides Balb-C mice Weight gain (43.5 ± 3 to 38.00 to 3.00 g) decreased significantly (p ≤ 0.0001),
Epididymal, subcutaneous fat accumulation and liver fat reduced (p ≤ 0.01) in dose dependent pattern
PPAR-γ, C/EBPα and SREBP-1c and FAS (p ≤ 0.001) decreased and ACC increased
ALT, AST, TG and FFA decreased (p ≤ 0.001)
Firmicutes decreased and Bacteroidetes increased
[112]
46.Whole plant of Ishige okamurae (Ishigeaceae)Diplorethohydroxycarmalol isolated C57BL/6J male mice TG (137.88 ± 16.24 to 86.73 ± 11.03 mg/dL), LDL-C (22.24 ± 1.40 to 16.82 ± 2.02 mg/dL), leptin (2.04 ± 0.59 to 1.23 ± 0.37 mg/dL) AST (47.11 ± 6.07 to 41.02 ± 1.52 mU/mL) decreased
Expression of AMPK and ACC increased (p ≤ 0.01)
C/EBPα, SREBP-1c, PPAR-γ and FABP4 (p ≤ 0.01) FAS (p ≤ 0.05)
[89]
47.Whole plant of brown algae Sargassum thunbergiiPolyphenols of algae C57BL/6 mice Body weight gain, epididymal fat, subcutaneous fat, mesenteric fat and perirenal fat decreased (p ≤ 0.05)
TG (30.65 ± 2.19 to 23.28 ± 1.87 nmol/µL), TC (84.77 ± 4.05 to 73.76 ± 1.61 µg/µL), Leptin (3521.6 ± 0.09 to 2299.6 ± 0.3 pg/mL) insulin (2.21 ± 0.02 to 1.13± 0.42 ng/mL) decreased
Expression of UCP1 and UCP3 unregulated. PPAR-γ downregulated
[113]
48.Red seaweed Grateloupia elliptica (Cryptonemiaceae)Polyphenols, alkaloids, terpenoids, organosulfur compounds, phytosterols, alginates, fucoidans and phlorotannins Mice and 3T3-L1 adipocytes Lipids accumulation reduced by 61% at 200 µg/mL
Body weight gain suppressed; WAT weight decreased (p ≤ 0.05)
Expression of SREBP-1 (p ≤ 0.001), PPARγ (p ≤ 0.0001), FABP-4 (p ≤ 0.0001),
Expression of UCP-1 and UCP-3 rised
TG (110.78 ± 1.62 to 58.22 ± 0.14 mmol/µL), TC (50.17 ± 0.05 to 29.33 ± 0.07 µg/µL), leptin (4308.13 ± 59.37 to 1896.88 ± 1.87 pg/mL), insulin (4.50 ± 0.18 to 3.97 ± 0.00 ng/mL) decreased
[114]
49.Soybean embryo and enzymatically modified Isoquercetin Isoflavones, daidzein, glycitein, genisten C3H10T12 adipocytes and MiceBody weight gain and fat accumulation decreased significantly
Reduced the lipids contents, expression of HSL and PL1N1 increased that increased PKA-dependent cytosolic lipolysis
Expression of UCP1, CREB, and SIRT1 increased (p ≤ 0.01) at 160 µg/mL
[115]
50.Fermented fruit extracts of Diospyros kaki (Ebenaceae)Vitamins, polyphenols, dietry fibres, gallic acid, epicatechins, gallocatechins, apicatechins gallateC57BL/6N mice and 3T3-L1 adipocytes Body weight gain (15%) and Abdominal fat (27%) and liver mass decreased
TC (p ≤ 0.01), TG (p ≤ 0.05), FBG (p ≤ 0.05), LDL-C (p ≤ 0.01) decreased and HDL-C (p ≤ 0.05) increased
Expression of PPAR-γ, C/EBPα, SREBP-1c and FAS decreased
AMPK phosphorylation increased
[116]
51Root extract of Polygonum cuspidatum (Polygonaceae)Resveratrol, Emodin, Picerid-phenolic acids 3T3-L1 adipocytes Expression of PPAR-γ, C/EBPα, SREBP-1c, aP2, FAS, NF-kβ, p38, p38 mitogen activated protein kinases, JNK significantly decreased at 150 µg/mL and JNK increased (p ≤ 0.05)[92]
Table 2. Biologically synthesized metallic nanoparticles and nanoencapsulated secondary metabolites showing anti-obesity efficacy.
Table 2. Biologically synthesized metallic nanoparticles and nanoencapsulated secondary metabolites showing anti-obesity efficacy.
Sr. No.Type of NanostructureChemical Constituents InvolvedCharacteristics of Nanostructure Experimental Model Important Findings Reference
1.Nanocellulose from Grape seeds (Vitis vinifera)Pyrogallol, Protocatechuic acid, Chlorogenic, e-vanillic, Benzoic, Naringenin, Hisperidin, Rosmarinic acid, Kaempferol4–7 nm in width and 37–45 nm in length Rat Body weight, weight of liver, kidney, heart and spleen reduced (p ≤ 0.05), body weight gain decreased up to 13.73 ± 1.91 and 25.69 ± 2.20% at 2% and 4% nanoparticles diet respectively, Food intake decreased up to 14.00 and 12.00 g/day at 2% and 4% NPs diet respectively, TGs and TC value decreased to 85.97 ± 6.08 and 74.80 ± 3.23 mg/dL at 2% NPs diet [118]
2.Gold nanoparticles of Salacia chinensis Saponins, flavonoids and proanthocyanidinsCrystalline, 20 to 50 nm in size and spherical in shape Rat Adipose index, leptin and resistin level decreased and adiponectin level increased (p ≤ 0.05), BMI and body weight decreased (p ≤ 0.05), AMPKα1 and pAMPKα1 decreased[119]
3.Gold nanoparticles of Smilax glabra Phenols, alkaloids, flavonoids, glycosides, steroids, tannins, resin and volatile oils20 nm in size and Spherical in shape RatBody weight and BMI decreased (p ≤ 0.001), Leptin and resistin level (p ≤ 0.05), adiponectin level increased (p ≤ 0.05), plasma insulin level increased (p ≤ 0.01), TNFα and IL-1β decreased[120]
4.Chitosan NPs and water-soluble chitosan NPsChitosan 400–700 nm size of CTS-NPs, 700–1000 nm of WSC-NPsRat Weight gain reduced (p ≤ 0.05), epididymal and perirenal WAT decreased, TC, TG, LDL, Lipids accumulation decreased significantly, CBPα and PPAR-γ expression decreased [121]
5.Gold nanoparticles of the leaf extract of Panax ginseng Ginsenoside Rh2, Ginsenoside Rg3, Protopanaxatriol saponins and Protopanaxadiol saponins 10–20 nm in size and spherical in shape 3T3-L1 mature adipocytes Triglycerides accumulation decreased (p ≤ 0.001), expression of PPARα, CREBPα and β, SREBP, FAS decreased [122]
6.Gold NPs of Poria cocos Triterpenes, glycosides and phenolic acids Spherical in shape, polydispersed and 20 nm in size RatTC, TG and HDL decreased at the significance level of p ≤ 0.05
Leptin, resistin level decreased and adiponectin level increased
[123]
7.Nanoencapsulated Quercetin Succinyl chitosan- alginate core shell NPs91.58 nm in size, spherical shape, 95% EEMale wistar rat TG level (51.41%), TC (49.01%), AST (32.69%), ALT (39.36%), ALP (39.15%) decreased respectively [124]
8.Nanoencapsulated EGCG α-tocopherol acetate (8.8%), Kolliphor HS15 (45%), Soy PC (36.2%), EGCG (10%)104 nm in size and spherical in shape, polydispersed with 95.0% EEC57BL1/6J mice TC, HDL-C, TG, Plasma TNF-α, MCP-1, IL-6 decreased significantly at p ≤ 0.05, decreased in lesion surface area of aortic arches, secretion of inflammatory factors decreased at 20 µg/mL to 25 mg/kg[125]
9.Nanoencapsulated EGCG1-(Palmitoyl)-2-(5-keto-6-octenedioyl)phosphotidylcholine (Kodia-PC), α-tocopherolSpherical in shape and 108 nm in size, 96% EE, polydispersity index less than 0.3Human monocytes THP1 cellsmRNA and protein level of MCP-1 decreased, macrophage EGCG content increased, Kappa β and p38 mitogen-activated protein kinase decreased, PPAR γ decreased, total macrophage cholesterol level decreased[126]
10Nanoencapsulated curcumin Turmeric loaded nanoemulsion (TE-NE), 20 folds lower curcumin content than turmeric extract136–138 nm in size and spherical in shape, 87.95 ± 0.39 EEBalb/c mice and HepG2 cells SREBP-1, PPARγ decreased at 5% of TE-NE, Cleaved caspase-3 and PARP level increased at 300 mg/kg (p ≤ 0.01)
Body weight gain decreased
Total serum cholesterol level decreased (p ≤ 0.05), TG level decreased (p ≤ 0.01)
[72]
11Nanencapsulated curcumin PLA-PEG polymers Monodispersed, spherical in shape and 117 nm in size with surface charge 35 mVWhite albino ratsALT (63.44%) and AST (54%) decreased
NF-kβ, COX-2, TGF-β level decreased and PPAR-α increased
[127]
12Nanoencapsulated quercetin Polylactic-co-glycolic acid (PLGA)179.9 ± 11.2 nm in size, spherical in shape, 0.128 polydispersity index, 86% EE, Sprague-Dawley diabetic rats Decreased blood glucose level at 150 mg/kg (p ≤ 0.05)[128]
13Nanoemcapsulated Resveratrol Polylactic-co-glycolic acid (PLGA)Spherical morphology, 176.1 nm in size, 97.25% EE, 14.9% drug loading capacity HepG2 hepatocytes Lipids accumulation decreased to 80.77% at 100 µM
Decreased hepatocellular differentiation,
Decreased TG level in a dose dependent manner (p ≤ 0.0001)
[129]
14Gold nanoparticle (Dendropanax morbifera Léveille) Gold nanoparticle (D-AuNPs) Spherical (size 10–20 nm)3T3-L1 adipocytes and HepG2 cellsThe adipogenesis process was negatively controlled by D-AuNPs, with downregulated PPARγ, CEBPα, Jak2, STAT3, and ap2 expression in 3T3-L1 cells and FAS and ACC levels in HepG2 cells. So D-AuNPs exert antiadipogenic properties.Yi, [130]
15Trans Resveratrol-encapsulated NPs
(L-R nano)
DSPE-PEG 5000 peptide incorporated L-R nano with ASC targetsASC-targeted nanoparticle C57BL/6 mice and 3T3L1 adipocytesTargeted delivery of browning agents to adipose stromal cells (ASCs) in subcutaneous WAT[131]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bhardwaj, M.; Yadav, P.; Vashishth, D.; Sharma, K.; Kumar, A.; Chahal, J.; Dalal, S.; Kataria, S.K. A Review on Obesity Management through Natural Compounds and a Green Nanomedicine-Based Approach. Molecules 2021, 26, 3278. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26113278

AMA Style

Bhardwaj M, Yadav P, Vashishth D, Sharma K, Kumar A, Chahal J, Dalal S, Kataria SK. A Review on Obesity Management through Natural Compounds and a Green Nanomedicine-Based Approach. Molecules. 2021; 26(11):3278. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26113278

Chicago/Turabian Style

Bhardwaj, Monika, Poonam Yadav, Divya Vashishth, Kavita Sharma, Ajay Kumar, Jyoti Chahal, Sunita Dalal, and Sudhir Kumar Kataria. 2021. "A Review on Obesity Management through Natural Compounds and a Green Nanomedicine-Based Approach" Molecules 26, no. 11: 3278. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26113278

Article Metrics

Back to TopTop