, ,

ANTI-ADIPOGENIC – Adipocyte Differentiation Inhibitor

(6 customer reviews)

$275.00

& Free Shipping
SKU: N/A Categories: , ,
Guaranteed Safe Checkout

DISCOUNTS AND INCENTIVES :

  • Free $50 Interstellar Mug On Orders Over $500 (Choose Color At Checkout)
  • Free $95 Value Rush Worldwide Shipping On All Orders $750 Or More
  • Free $333 Value 75mm (3") Sterling Silver Interstellar Statement Pendant With $1500 Or More In Cart
  • 35% Off With $1000 Or More In Cart - Use Code "1000" (Excludes Combos)
  • 40% Off With $2000 Or More In Cart - Use Code "2000" (Excludes Combos)
  • 45% Off With $3000 Or More In Cart - Use Code "3000" (Excludes Combos)
  • 50% Off With $4000 Or More In Cart - Use Code "4000" (Excludes Combos)
  • 55% Off With $5000 Or More In Cart - Use Code "5000" (Excludes Combos)
  • 60% Off With $7500 Or More In Cart - Use Code "7500" (Excludes Combos)

 


Adipogenesis refers to the process of formation of adipose tissue. It is a multistep process starting with clonal expansion of mesenchymal cells and the Differentiation of these mesenchymal cells into Pre-Adipocytes and finally into mature Adipocytes.

Herbal Remedies against Adipogenesis

Many phytochemicals and herbal extracts show Inhibitory effects on Adipogenesis in cell models and mouse models. However, lack of clinical evidence and epidemiological data to support their roles in Inhibiting Adipogenesis hindered their development to become an Anti-Obesity therapeutic agent. Furthermore, many of these phytochemicals have low bioavailability. Chemical structural modification or targeted delivery system may help to increase their Anti-Adipogenesis or Anti-Obesity efficacies. Nevertheless, many of these phytochemicals are abundantly found in our daily food, or they are the herbs for cooking or seasoning. For example, resveratrol is rich in red wine and grapes; genistein is rich in soy; quercetin in apples and onions, and curcumin is a well-known component of the cook seasoning. Therefore, a healthy diet can definitely help us to increase the uptake of these phytochemicals in our daily life. Moreover, combination of phytochemicals or herbal extracts may act synergistically to Inhibit Adipogenesis.

Indeed, Obesity is not simply an excess accumulation of white Adipose Tissue but is usually associated with insulin resistance and an increased production of metabolic hormones coupled with chronic low-grade state of Inflammation . To effectively Reduce Obesity , a holistic strategy with the consumption of phytochemicals or herbal extracts can be designed not only to Reduce Adipose Tissue Mass, but also increase thermogenic energy expenditure, improve insulin sensitivity, Reduce plasma lipids which can help to Reduce the Obesity -associated dyslipidemia and other comorbid conditions.

Featuring:

Açai (Euterpe Oleracea Martius) Anthocyanins • Acer Okamotoanum Nakai Leaf • Achyranthes Bidentata Blum • Acorn (Quercus Acutissima Carr.) Shell • Adenanthin, A Natural Ent-Kaurane Diterpenoid Isolated From The Herb Isodon Adenantha • Aicar • Ajoene Sulfur-Containing Compound • Allium Hookeri Root • Andrographolide • Anthocyanins • Antofine Alkaloid • Antrodia Cinnamomea • Apigetrin (Apigenin-7-O-Glucoside) • Arctigenin • Arctiin • Aristolochia Manshuriensis Kom • Aristotelia Chilensis • Artemisia Capillaris • Aster Yomena (Kitam.) Honda Leaves • Astilbe Chinensis Franch. Et Savet • Astragalin, (3-O-Glucoside Of Kaempferol) • Averrhoa Carambola L. Peel • Avicularin • Bacaba (Oenocarpus Bacaba Mart.) Phenolic (Bpe) • Baicalin • Bamboo (Phyllostachys Bambusoides) Leaf • Benincasa Hispida • Berberine • Bergamottin • Bestatin (Ubenimex) • Betanin (Betanin-5-O-Β-Glucoside) • Bisdemethoxycurcumin • Blumea Balsamifera • Boldine • Boussingaulti Gracilis Miers Var. Pseudobaselloides Bailey • Butein Chalconoid • Capsaicin • Carduus Crispus • Carnosic Acid • Carnosine (L-Carnosine) • Chromolaena Odorata Leaves • Chrysanthemum Zawadskii Extact • Chrysin • Cinnamomum Verum • Cirsium Brevicaule A. Gray • Cirsium Setidens Nakai • Citrus Aurantium Peel • Clitoria Ternatea Flower Petal • Cocoa Tea (Camellia Ptilophylla) • Colocynth (Citrullus Colocynthis) Flesh • Conjugated Linoleic Acid (Cla) • Coptis Chinensis (Coptidis Rhizoma) • Cordycepin • Coumestrol • Cranberries (Oxycoccus Quadripetalus) • Crocin • Cryptotanshinone • Curcumin • Curcumin-3,4-Dichloro Phenyl Pyrazole (Cdpp) • Cyanidine-3-O-Galactoside Enriched Aronia Melanocarpa • Cyclopia Subternata • Delphinidin • Delphinidin-3-O-Β-Glucoside • Dhea • Diallyl Trisulphide • Dioscin (Ds) A Steroidal Saponin • Djulis (Chenopodium Formosanum) • Dolichos Lablab L. Seeds • Ecklonia Cava • Ecliptal, Isolated From Eclipta Alba • Egcg • Ellagic Acid • Epiberberine • Erigeron Annuus (L.) Pers. • Esculetin • Euphorbia Lunulata • Evodiamine • Ferulic Acid • Ficus Deltoidea Var. Deltoidea • Fisetin • Foenumoside B From Lysimachia Foenum-Graecum • Formononetin • Fucoxanthinol • Gallotannin Derivatives From Mango (Mangifera Indica L.) • Ganoderma Applanatum • Ganoderma Lucidum • Garcinia Cambogia • Garcinol • Gelidium Elegans • Genistein • Gentiopicroside • Gentiopicroside Isolated From Gentiana Scabra Bge. • Ginkgetin, A Biflavone From Ginkgo Biloba Leaves • Ginkgo Biloba Seed Coat • Ginkgolide C, Isolated From Ginkgo Biloba Leaves • Ginsenoside Rg1 • Ginsenoside Rg2 • Ginsenoside Rg3 • Glehnia Littoralis Root • Guarana (Paullinia Cupana) • Guggulsterone • Heshouwu (Polygonum Multiflorum Thunb) • Hesperidin • Hibiscus Rosa Sinensis Flower • Hibiscus Sabdariffa L • Hippophae Rhamnoides L. Leaves • Hwangryunhaedok-Tang • Hyperoside • Icaritin Hydrolytic Product Of Icarin From Epimedium Genus • Irvingia Gabonensis Seed • Ivy Gourd (Coccinia Grandis L. Voigt) Root • Kaempferol • Komulkosiraegi [Gracilaria Vermiculophylla (Ohmi) Papenfuss] • Lindera Obtusiloba • Lupenone • Lupeol • Lysimachia Foenum-Graecum • Magnolia Officinalis Magnolol • Mai Tong Fang (Radix Astragali • Mangifera Indica L. Peel • Mangiferin From Iris Rossii Baker • Maté (Ilex Paraguariensis) • Melissa Officinalis • Metadina Trichotoma • Methyl Cinnamate • Mollugin (From Rubia Cordifolia L Roots) • Momordica Charantia • Moringa Oleifera Leaves (Drumstick Tree) • Morus Alba • Murta (Myrceugenia Euosma) Dried Branches And Leaves • Myrica Gale • Nam Doc Mai • Naringenin • Nobiletin • Nonivamide • Oleanolic Acid • Oligonol • Orientin • Oroxylin A Flavonoid • Panicum Miliaceum L. • Pelargonidin • Petalonia Binghamiae • Peucedanum Japonicum Thunberg L. • Phyllanthus Emblica Fruit • Piperine • Platycodin D • Platyphylloside • Pleurotus Eryngii Var. Ferulae ‘Beesan No. 2’ (Cebt) • Pluchea Indica (L.) Less. Tea • Polygonum Cuspidatum • Porphyra Yezoensis • Prieurianin • Procyanidin B2 • Pterostilbene • Punicalagin • Quercetin • Raspberry Ketone  • Rehmannia Glutinosa • Resveratrol • Retinoic Acid • Rhamnetin • Rhein Glycoside • Rhizoma Polygonati Falcatum • Rosmarinic Acid • S-Petasin • Salicortin • Sargassum Micracanthum • Sargassum Serratifolium • Saururus Chinensis • Securigera Securidaca (Seeds) • Sesamol • Shikonin  • Sibiraea Angustata • Silibinin • Siphonaxanthin • Soyasaponin Ab • Stearidonic Acid (Sda) • Stinging Nettles (Utrica Dioica L.) • Styrax Japonica Fruit • Sulforaphene • Sulfuretin Is A Natural Flavonoid Found In The Plant Rhus Verniciflua Stokes. • Tannic Acid • Taraxacum Officinale (Dandelion) • Tocotrienol • Tricin • Trigonella Foenum-Graecum (Seeds) • Triticum Aestivum • Tyrosol (From Rhodiola Crenulata ) • Ulmus Pumila • Ursolic Acid • Vaccinium Floribundum • Vanillic Acid • Veratrum Nigrum • Viburnum Opulus L. • Vitexilactone, A Constituent From Vitex Trifolia L • Vitexin From Mung Bean • Vitisin A From Iris Lactea Pall. Var. Chinensis (Fisch.) Koidz • Wasabi (Wasabia Japonica Matsum.) Leaves • Wedelolactone Furanocoumarin • Widdrol • Xanthohumol • Zeaxanthin • Zizyphus Jujube • Α-Mangostin • Β-Asarone • Β-Cryptoxanthin • 13-Methylberberine • 17Β-Glycyrrhetinic Acid • 2,6-Dimethoxy-1,4-Benzoquinone • 25 Hydroxycholesterol • 3-Chloro-4,5-Dihydroxybenzaldehyde • 3,5-Dicaffeoyl-Epi-Quinic Acid Isolated From Edible Halophyte Atriplex Gmelinii • 3,5,6,7,7,3′,4′-Heptamethoxyflavone (Hmf), A Naturally Occurring Polymethoxyflavone Found In Citrus Peel • 6-Gingerol • 6,7,4′-Trihydroxyisoflavone 

Potential of natural products in the Inhibition of Adipogenesis through regulation of PPARγ expression and/or its transcriptional activity

Obesity is a global health problem characterized as an increase in the Mass of Adipose Tissue . Adipogenesis is one of the key pathways that increases the Mass of Adipose Tissue , by which preAdipocyte s mature into Adipocytes through cell Differentiation . Peroxisome proliferator-activated receptor γ (PPARγ ), the chief regulator of Adipogenesis , has been acutely investigated as a molecular target for natural products in the development of anti-Obesity treatments. In this review, the regulation of PPARγ expression by natural products through Inhibition of CCAAT/enhancer-binding protein β (C/EBPβ) and the farnesoid X receptor (FXR), increased expression of GATA-2 and GATA-3 and activation of the Wnt/β-catenin pathway were analyzed.

Furthermore, the regulation of PPARγ transcriptional activity associated with natural products through the antagonism of PPARγ and activation of Sirtuin 1 (Sirt1) and AMP-activated protein kinase (AMPK) were discussed. Lastly, regulation of mitogen-activated protein kinase (MAPK) by natural products, which might regulate both PPARγ expression and PPARγ transcriptional activity, was summarized. Understanding the role natural products play, as well as the mechanisms behind their regulation of PPARγ activity is critical for future research into their therapeutic potential for fighting Obesity .

Effects of Flavonoids and Phenolic Acids on the Inhibition of Adipogenesis in 3T3-L1 Adipocytes

Obesity has become a global epidemic in both developed and developing countries, and it is a significant risk factor for various diseases such as Diabetes , cancer, heart disease, and hypertension. In the present study, the effect of naturally occurring Antioxidants (Flavonoids and Phenolic Acids ) on the Inhibition of Adipogenesis in 3T3-L1 Adipocytes was investigated. The results showed that o-coumaric acid and rutin had the highest Inhibition on intracellular triglyceride (61.3 and 83.0%, respectively) among 15 Phenolic Acids and 6 Flavonoids tested. However, the oil red o stained material (OROSM) showed that cell number in 3T3-L1 Adipocytes was not influenced by those compounds.

For glycerol-3-phosphate dehydrogenase (GPDH) activity, the data indicated that o-coumaric acid and rutin had the highest Inhibition on GPDH activity (54.2 and 66.8%, respectively) among the compounds tested. o-Coumaric acid and rutin also Inhibited the expression of PPARγ , C/EBPα and leptin and then up-regulated expression of adiponectin at the protein level. Some naturally occurring Antioxidants efficiently suppressed Adipogenesis in 3T3-L1 Adipocytes. These results suggest that o-coumaric acid and rutin targeted for Adipocyte functions could be effective in improving the symptoms of Metabolic Syndrome.

INGREDIENTS: 


açai (Euterpe oleracea Martius)


Anti-lipidaemic and anti-inflammatory effect of açai (Euterpe oleracea Martius) Polyphenols on 3T3-L1 Adipocytes


The anti-lipidaemic and anti-inflammatory effects of açai Polyphenols in 3T3-L1 mouse Adipocytes were investigated. Pre-Adipocytes were differentiated with and without açai-Polyphenols at concentrations of 2.5, 5 and 10 µg gallic acid equivalents (GAE)/mL. Results showed that açai Polyphenols Reduce the accumulation of intracellular lipids in differentiated Adipocytes in a dose-dependent manner and downregulated PPARγ 2. The gene-expression of Adipogenic transcription factors C/EBPα, C/ebpβ, Klf5 and Srebp1c was decreased.

This was accompanied by a reduction of Adipogenic genes, including aP2, LPL, FATP1 and FAS, leptin and total PAI and an increase of adiponectin. Additionally, açai Polyphenols protected cells against the production of ROS and decreased the expression of mRNA and protein levels of pro-inflammatory cytokines when 3T3-L1 cells were challenged with TNF-α. Thus, these results indicate that açai Polyphenols may be useful in the prevention of Adipogenesis, oxidative stress and Inflammation.


AntiAdipogenic effects of açai seed extract on high fat diet-fed mice and 3T3-L1 Adipocytes : A potential mechanism of action

Açai seed extract (ASE) exerts antiobesogenic effects.•ASE regulates lipogenic signaling pathways.•ASE is promising functional food ingredient for the management of Obesity.

Body Adiposity is an important risk factor for the development of chronic non-transmissible diseases. Studies on the process of Adipogenesis from ScienceDirect’s AI-generated Topic PagesAdipogenesis have been extensively performed in vivo and in vitro models to describe the molecular and cellular bases of Adipose Tissue development and the effect of natural products in this process. The açai seed extract (ASE) has been evidenced as a potential regulator of body Mass. In our work high-fat diet–fed mice treated with ASE (300 mg/Kg/d) (HFD-ASE) showed a lower adipose index (−32.63%, p < 0.001) than the high-fat diet–fed mice group (HFD) and the Adipocytes from the HFD group were considerably enlarged (p < 0.001) compared to those in the control group (CG) and HFD-ASE group (+175% and +123%, respectively).
We also evaluated the effects of ASE on the modulation of Adipogenesis in 3T3-L1 cells. ASE exposure (25 and 100 μg/mL) led to a decrease of 26.6 (p < 0.05) in proliferation and also Inhibited pre-Adipocyte Differentiation through the decreasing expression (p < 0.05) of transcription factors and Adipogenic proteins such as PPARɣ, SREBP-1, and FAS. These results show that the ASE Reduce Adipogenesis and suppress lipid accumulation in the in vivo model and in 3T3-L1 Adipocytes and reinforce ASE as a potential strategy to modulate Adipogenesis.


Acer okamotoanum Nakai Leaf

Acer okamotoanum Nakai Leaf Extract Inhibits Adipogenesis Via Suppressing Expression of PPAR γ and C/EBP α in 3T3-L1 cells


The genus Acer contains several species with various bioactivities including antioxidant, antitumor and anti-inflammatory properties. However, Acer okamotoanum Nakai, one species within this genus has not been fully studied yet. Therefore, in this study, we investigated the Anti-Adipogenic activities of leaf extract from A. okamotoanum Nakai (LEAO) on 3T3-L1 preadipocytes. Adipogenesis is one of the cell Differentiation processes, which converts preadipocytes into mature Adipocytes . Nowadays, Inhibition of Adipogenesis is considered as an effective strategy in the field of Anti-Obesity research. In this study, we observed that LEAO decreased the accumulation of lipid droplets during Adipogenesis and down-regulated the expression of key Adipogenic transcription factors such as peroxisome proliferator-activated receptor γ (PPAR γ) and CCAAT/enhancer binding protein α (C/EBP α). In addition, LEAO inactivated PI3K/Akt signaling and its downstream factors that promote Adipogenesis by inducing the expression of PPAR γ. LEAO also activated β-catenin signaling, which prevents the Adipogenic program by suppressing the expression of PPAR γ. Therefore, we found that treatment with LEAO is effective for attenuating Adipogenesis in 3T3-L1 cells . Consequently, these findings suggest that LEAO has the potential to be used as a therapeutic agent for preventing Obesity.

Keywords: Acer okamotoanum, Adipogenesis, Anti-Obesity, 3T3-L1 preadipocytes



Achyranthes bidentata Blum


The present study investigated the AntiObesity effect of Achyranthes bidentata Blume root water extract in a 3T3-L1 Adipocyte Differentiation model and rats fed with a high-fat diet. To investigate the effect of Achyranthes bidentata Blume on Adipogenesis in vitro, differentiating 3T3-L1 cells in Adipocyte -induction media were treated every two days with Achyranthes bidentata Blume at various concentrations (1 to 25 μg/mL) for eight days. We found that Achyranthes bidentata Blume root Inhibited 3T3-L1 Adipocyte Differentiation without affecting cell viability and Western blot analysis revealed that phospho-Akt expression was markedly decreased, whereas there was no significant change in perilipin expression.

Furthermore, administration of Achyranthes bidentataBlume root (0.5 g/kg body weight for six weeks) to rats fed with a high-fat diet significantly Reduced body Weight Gain without affecting food intake, and the level of triglyceride was significantly decreased when compared to those in rats fed with only a high-fat diet. These results suggest that Achyranthes bidentata Blume root water extract could have a beneficial effect on Inhibition of Adipogenesis and controlling body weight in rats fed with a high-fat diet.



Acorn (Quercus acutissima CARR.) shell

Anti-oxidant and Anti-Adipogenic effects of acorn (Quercus acutissima CARR.) shell extracts via regulation of wnt signaling in 3T3-L1 cells


Acorn (Quercus acutissima CARR.) is a nut from the Fagaceae family that has been used in traditional medicine for many years. However, shells from acorns are regarded as a by-product and are mostly discarded. Anti-Adipogenic activities of acorn shells were investigated using 3T3-L1 cells and methanol shell extracts (AE-M). AE-M demonstrated Cu2+-chelation activities and anti-oxidant activities via reduction of oxidative stress levels induced using AAPH. Six days after Adipocyte Differentiation, 50 and 100 μg/mL AE-M completely suppressed 3T3-L1 Adipogenesis and the Anti-Adipogenic effect was stronger than for the positive control 50 μM quercetin.

Treatment with AE-M in 3T3-L1 cells Reduced mRNA expression levels of Adipogenic genes. AE-M-Inhibition was found in Pre-Adipogenic, early, and intermediate stages of Adipogenesis in 3T3-L1 cells. The Wnt/β-catenin signaling pathway is required for AE-M-Inhibition of 3T3-L1 Adipogenesis.



Adenanthin

(Isodon adenantha)



Adenanthin, a natural ent-kaurane diterpenoid extracted from the herb Isodon adenantha, has been reported to increase intracellular reactive oxygen species in leukemic and hepatocellular carcinoma cells. However, the function and mechanism of the compound in Adipogenesis and the development of Obesity is still unknown. In this study, we demonstrated that adenanthin Inhibited Adipogenesis of 3T3-L1 and mouse embryonic fibroblasts, and the underlying mechanism included two processes: a delayed mitotic clonal expansion via G0/G1 cell cycle arrest by Inhibiting the RB-E2F1 signaling pathway and a Reduced C/EBPβ signaling by Inhibiting the expression and activity of C/EBPβ during mitotic clonal expansion. Furthermore, adenanthin significantly Reduced the growing body weight and Adipose Tissue Mass during high-fat diet-inducing Obesity of mice, indicating the beneficial effects of adenanthin as a potential agent for prevention of Obesity .
In this work, we showed that adenanthin functioned its Anti-Adipogenic effect by increasing the intracellular ROS level during MCE and then affected two processes: (1) Inhibited the RB-E2F1 signaling pathway and results in a G0/G1 cell cycle arrest during MCE at the early stage of Adipocyte Differentiation ; (2) Reduced C/EBPβ transcriptional activity by Inhibiting the expression and activity of C/EBPβ during MCE. These results were in line with the Inhibitory effect of adenanthin on HFD-induced development of Obesity in mice.
ROS, oxygen-derived small molecules, react readily with various chemical structures including proteins, lipids, sugars, and nucleic acids [36]. In recent years, there has been an accumulating understanding of ROS as signaling molecules [37]. Most research groups now believe that a regulated basal level of ROS is necessary and advantageous for maintenance of cell functions, such as proliferation, Differentiation, and survival [38,39]. Increasing evidence has indicated that ROS plays a key role for Adipogenesis, but whether the increased ROS is positive or negative for Adipogenesis is still controversial. Some studies demonstrate that ROS promotes Adipocytes to differentiate [22,23], however, other studies show that ROS is indicated to Inhibit Adipocyte Differentiation by reducing the DNA-binding activity of C/EBPβ [24] and suppressing MCE [25]. Consistent with these findings [24,25], in our case, increased ROS Inhibited Adipogenesis, which was rescued by treatment of cells with adenanthin supplemented with H2O2 cleaner NAC (Figure 4). Furthermore, we exogenously treated cells with various doses of H2O2 (50 to 1200 μM) and found that lower doses (100, 200, and 400 μM) of H2O2 promoted 3T3-L1 Differentiation , whereas higher doses (800, 1000, and 1200 μM) of H2O2 Inhibited 3T3-L1 Differentiation (Supplementary Figure S1a–c), implying that “positive” or “negative” roles of H2O2 relies on its level as previous report [26].
It is well known that Adipogenesis occurs in several stages and involves a cascade of transcription factors [40]. Our results showed that adenanthin functions in MCE (Figure 3 and Figure 4). When it was induced to Differentiation, growth-arrested 3T3-L1 preadipocytes synchronously re-enter the cell cycle and undergo MCE followed by expression of genes that produce the Adipocyte phenotype. MCE is a prerequisite for Differentiation of 3T3-L1 preadipocytes into Adipocytes [16]. Therefore, cell cycle regulation at the early stage of Adipogenesis has been considered as a strategy for modulating Adipogenesis [41]. Recently, many natural products have been reported to be Anti-Obesity agents that can Inhibit MCE of 3T3-L1 Adipocytes [41,42,43]. In our work, we found that adenanthin could attenuate cell cycle progression through a G0/G1 arrest during MCE by decreasing protein levels of the phosphorylation of RB, E2F1, Cyclin B1, and Cdk1 and modestly increased protein levels of P21 and P18 (Figure 3 and Figure 4d–f). As the cells cross the G1/S checkpoint, C/EBPβ acquires DNA-binding activity. Coincident with the acquisition of DNA-binding activity, C/EBPβ binds to centromeres through consensus C/EBP-binding sites in centromeric satellite DNA [15,20]. Consistent with a previous study [43], our results showed that adenanthin Inhibits the expression and activity of C/EBPβ during MCE (Figure 4b,c).
Finally, our results demonstrated that adenanthin Reduced the growing body weight and Adipose Tissue Mass during the early stage of induction of Obesity (Figure 5a–d). Moreover, to evaluate the safety of adenanthin, the serum of HFD fed mice after a 30-day exposure to adenanthin was collected and serum aspartate amino transferase (AST), alamine amino transferase (ALT), and creatinine levels between vehicle and adenanthin-treated mice were evaluated, and the results were not significantly changed (Supplementary Table S1). Whether adenanthin is active during the late stage of Obesity or has an Anti-Obesity effect is still unknown. In the future, we can monitor the activity of adenanthin during different stages of HFD-induced Obesity.
In summary, adenanthin, a natural ent-kaurane diterpenoid isolated from the herb Isodon adenantha in 1987 [27], was able to Inhibit Adipogenesis of 3T3-L1 and mouse embryonic fibroblasts (MEFs) through regulation of ROS and significantly Reduced the growing body weight and Adipose Tissue Mass during HFD-induced development of Obesity in mice, revealing adenanthin may be a potential agent for prevention of Obesity.


AICAR (5-aminoimidazole-4-carboxamide ribonucleoside)

The Effects of AICAR on Adipocyte Differentiation of 3T3-L1 cells


The AMP-activated protein kinase (AMPK) activator, 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR), has been found to Inhibit the Differentiation of 3T3-L1 Adipocytes, if added at an early phase of Differentiation. AICAR blocks the expression of the late Adipogenic markers, fatty acid synthase and acetyl-CoA carboxylase, and of the transcription factors, C/EBPα and PPARγ. It also Inhibits early clonal expansion of Pre-Adipocytes, prevents the fall in C/EBPβ expression during the intermediate stage of Differentiation and Inhibits the late phase expression of CHOP-10, an antagonist of C/EBPβ. These data suggest a possible Inhibitory role for AMPK in the process of adipose Differentiation and suggest that AMPK might be a target to block Adipogenesis.

AMP-activated protein kinase
AICAR (5-aminoimidazole-4-carboxamide ribonucleoside)
3T3-L1 Adipocytes
Acetyl-CoA carboxylase
C/EBP (CAAT/enhancer binding protein)
PPAR (peroxisome proliferator-activated receptor)

AICAR Inhibits Adipocyte Differentiation in 3T3L1 and restores metabolic alterations in diet-induced Obesity mice model

Background: Obesity is one of the principal causative factors involved in the development of Metabolic Syndrome. AMP-activated protein kinase (AMPK) is an energy sensor that regulates cellular metabolism. The role of AMP-activated protein kinase in Adipocyte Differentiation is not completely understood, therefore, we examined the effect of 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR), a pharmacological activator of AMP-activated protein kinase (AMPK) on Adipocyte Differentiation in 3T3L1 cells and in a mouse D iet induced o besity (DIO) model.

Methods: To examine the effect of AICAR on Adipocyte Differentiation in 3T3L1 cells and in a mouse Diet i nduced o besity (DIO) model, 3T3L1 cells were differentiatied in the presence or absence of different concentration of AICAR and neutral lipid content and expression of various Adipocyte-specific transcription factors were examined. In vivo study, treated and untreated mice with AICAR (0.1–0.5 mg/g body weight) were fed high-fat diet (60% kcal% fat) to induce DIO and several parameters were studied.

Results: AICAR blocked Adipogenic conversion in 3T3L1 cells along with significant decrease in the neutral lipid content by downregulating several Adipocyte -specific transcription factors including peroxisome proliferators-activated receptor γ (PPARγ), C/EBPα and ADD1/SREBP1, which are critical for Adipogenesis in vitro. Moreover, intraperitoneal administration of AICAR (0.5 mg g/body weight) to mice fed with high-fat diet (60% kcal% fat) to induce DIO, significantly blocked the body Weight Gain and total content of epididymal fat in these mice over a period of 6 weeks. AICAR treatment also restored normal adipokine levels and resulted in significant improvement in glucose tolerance and insulin sensitivity. The reduction in Adipose Tissue content in AICAR treated DIO mice was due to reduction in lipid accumulation in the pre-existing Adipocytes. However, no change was observed in the expression of PPARγ, C/EBPα and ADD1/SREBP1 transcription factors in vivo though PGC1α expression was significantly induced.

Conclusion: Our study demonstrates that AICAR treatment significantly attenuates adipoctye Differentiation in vitro. However, its administration restricted the body weight, epididymal fat content and normalized metabolic alteration mediated by diet induced Obesity in mice. Increase in phosphorylation of ACC and AMPKα during Adipocyte Differentiation and AMPK activity in epididymal Adipose Tissue in DIO mice raises doubts about the involvement of AMPK in this process.



Ajoene Sulfur



This paper describes effects of several sulfur-containing compounds from garlic on the cell viability, apoptosis and Adipogenesis in 3T3-L1 Adipocytes. In both preadipocytes and mature Adipocytes, 100 and 200 microM ajoene significantly decreased cell viability and increased apoptosis. The effect on apoptosis was further confirmed with Hoechst staining. In contrast, diallyl sulfide, diallyl disulfide, diallyl trisulfide, deoxyalliin, and allyl methyl sulfide had no significant effect on cell viability or apoptosis in either preadipocytes or mature Adipocytes. In maturing preadipocytes ajoene significantly decreased lipid accumulation in a dose-dependent manner and these results were further confirmed by a decrease in lipid droplet number and lipid content through Oil Red O staining. There was no significant change in lipid accumulation in maturing preadipocytes treated with other garlic derivatives.
Thus, despite the same source of origin, garlic, ajoene was the only one with potent effects on cell viability, apoptosis and Adipogenesis in 3T3-L1 Adipocytes.


Allium hookeri root


Effects of Allium hookeri root water extracts on Inhibition of Adipogenesis and GLUT-4 expression in 3T3-L1 Adipocytes


Anti-Adipogenic and AntiDiabetic activities of Allium hookeri root water extracts (ARW) were assessed. Oil Red O staining showed that treatment with ARW caused a dose-dependent reduction in lipid accumulation. ARW was also involved in Adipocyte Lipolysis via LPL activity, and in the concentration of glycerol in a culture medium. On the basis of the concentration of adipokines following ARW treatment, ARW appeared to Inhibit expression of PPAR-γ, to Reduce concentrations of leptin and resistin, to increase the concentration of adiponectin, and to Inhibit lipid accumulation. ARW modulated adipokine expression associated with insulin resistance and sensitivity. 3T3-L1 Adipocytes treated with ARW showed increased GLUT-4 expression with increased glucose uptake into Adipocytes. ARW showed effectiveness for improvement of Diabetic conditions.


Allium hookeri (AH) is widely consumed as a herbal medicine. It possesses biological activity against metabolic diseases. The objective of this study was to investigate effects of AH root water extract (AHR) on Adipogenesis in 3T3-L1 cells and in high-fat diet (HFD)-induced obese mice. AHR Inhibited lipid accumulation during Adipocyte Differentiation by downregulation of gene expression, such as hormone sensitive lipase (HSL), lipoprotein lipase (LPL) and an Adipogenic gene, CCAAT/enhancer binding protein-α in 3T3-L1 preadipocytes.
Oral administration of AHR significantly suppressed body Weight Gain, Adipose Tissue weight, serum leptin levels, and Adipocyte cell size in HFD-induced obese mice. Moreover, AHR significantly decreased hepatic mRNA expression levels of cholesterol synthesis genes, such as 3-hydroxy-3-methylglutaryl CoA reductase, sterol regulatory element-binding transcription factor (SREBP)-2, and low-density lipoprotein receptor, as well as fatty acid synthesis genes, such as SREBP-1c and fatty acid synthase. Serum triglyceride levels were also lowered by AHR, likely as a result of the upregulating gene involved in fatty acid β-oxidation, carnitine palmitoyltransferase 1a, in the liver. AHR treatment activated gene expression of peroxisome proliferator-activated receptor-γ, which might have promoted HSL and LPL-medicated Lipolysis, thereby reducing white Adipose Tissue weight. In conclusion, AHR treatment can improve metabolic alterations induced by HFD in mice by modifying expression levels of genes involved in Adipogenesis, lipogenesis, and Lipolysis in the white Adipose Tissue and liver. View Full-Text

Anti-Obesity Effect of Allium hookeri Leaf Extract in High-Fat Diet-Fed Mice



Andrographolide (Andrographis paniculata)


Andrographolide Inhibits Adipogenesis of 3T3-L1 cells by suppressing C/EBPβ expression and activation


•Andrographolide is a diterpenoid phytochemical.

•Andrographolide Inhibits Adipogenesis of 3 T3-L1 Adipocytes.

•Andrographolide Suppresses Differentiation cocktail-induced C/EBPβ expression.

•Andrographolide attenuates ERK and GSK3β-dependent C/EBPβ activation.

•Andrographolide arrests 3 T3-L1 Adipocytes at G0/G1 phase.

Andrographolide, a diterpenoid, is the most abundant terpenoid in Andrographis paniculata, a popular Chinese herbal medicine. Andrographolide displays diverse biological activities including hypoglycemia, hypolipidemia, anti-Inflammation, and anti-tumorigenesis. Recent evidence indicates that andrographolide displays Anti-Obesity property by Inhibiting lipogenic gene expression, however, the underlying mechanisms remain to be elucidated. In this study, the effects of andrographolide on transcription factor cascade and mitotic clonal expansion in 3T3-L1 Preadipocyte Differentiation into Adipocyte were determined. Andrographolide dose-dependently (0–15 μM) Inhibited CCAAT/enhancer-binding protein α (C/EBPα ) and C/EBPβ mRNA and protein expression as well as peroxisome proliferator-activated receptor γ (PPARγ) protein level during the Adipogenesis of 3T3-L1 cells.

Concomitantly, fatty acid synthase and stearoyl-CoA desaturase expression and lipid accumulation were attenuated by andrographolide. Oil-red O staining further showed that the first 48 h after the initiation of Differentiation was critical for andrographolide Inhibition of Adipocyte formation. Andrographolide Inhibited the phosphorylation of PKA and the activation of cAMP response element-binding protein (CREB) in response to a Differentiation cocktail, which led to attenuated C/EBPβ expression. In addition, ERK and GSK3β-dependent C/EBPβ phosphorylation was attenuated by andrographolide.

Moreover, andrographolide suppressed cyclin A, cyclin E, and CDK2 expression and impaired the progression of mitotic clonal expansion (MCE) by arresting the cell cycle at the Go/G1 phase. Taken together, these results indicate that andrographolide has a potent Anti-Obesity action by Inhibiting PKA-CREB-mediated C/EBPβ expression as well as C/EBPβ transcriptional activity, which halts MCE progression and attenuates C/EBPα and PPARγ expression.



Anthocyanins (Vitis coignetiae Pulliat)


Regulation of Adipocyte Function by Anthocyanins ; Possibility of Preventing the Metabolic Syndrome


Obesity is defined as the accumulation of excess Adipose Tissue resulting from various metabolic disorders. Adipocyte dysfunction is strongly associated with the development of Obesity and insulin resistance. Metabolic Syndrome is characterized by a group of metabolic risk factors in one person. Abdominal Obesity and Adipocyte dysfunction play an important role in the development of this syndrome.

Anthocyanins are used as a food coloring, and they are widely distributed in human diets including berries, suggesting that large amounts of Anthocyanins are ingested from plant-based foods. This study shows that Anthocyanins have a significant potency of AntiObesity and ameliorate Adipocyte function in in vitro and in vivo systems and also that they have important implications for preventing Metabolic Syndrome.

KEYWORDS:

•Anthocyanins

•cyanidin 3-glucoside

•cyanidin

•Obesity

•Diabetes

Metabolic Syndrome

•Adipocyte


Anthocyanins Inhibit Lipogenesis During Adipocyte Differentiation of 3T3-L1 preadipocytes


Anthocyanins have been shown to suppress body weight and fat Mass in animal studies. However, the effect of Anthocyanins on the process of lipid accumulation during Adipocyte Differentiation is not fully understood and the lipogenic transcription factors regulated by Anthocyanins have not been identified. We investigated the effects of Anthocyanins on lipogenesis pathways during Adipocyte Differentiation in 3T3-L1 cells. Anthocyanins Reduced triglyceride (TG) accumulation in a dose-dependent manner during Adipocyte Differentiation. Accumulation of TG was rapidly reversed by anthocyanin withdrawal.

Anthocyanins markedly Reduced gene and protein expression levels of lipogenic transcription factors such as liver X receptor α, sterol regulatory element-binding protein-1c, peroxisome proliferators-activated receptor-γ, and CCAAT enhancer-binding protein-α. In addition, the target gene and protein expression of these lipogenic transcription factors such as fatty acid synthase, stearoyl-CoA desaturase-1, and acetyl-CoA carboxylase α were markedly suppressed by Anthocyanins. Thus, Anthocyanins suppress lipid accumulation in Adipocytes due to broad Inhibition of the transcription factors regulating lipogenesis. This may partially explain the mechanism by which Anthocyanins exert their Anti-Obesity effect.



Antofine Alkaloid


Antofine (ANTF) is a phenanthroindolizidine alkaloid isolated from the root of Cynanchum paniculatum Kitagawa (Asclepiadaceae), which is used as an herbal remedy for pain and Inflammation. ANTF also possesses antiviral and antitumorigenic activities. In this study, we investigated the role of ANTF in Adipogenesis. Chronic ABTF administration suppressed Adipocyte Differentiation and marker expression in a dose-dependent manner. Furthermore, acute administration of ANTF at early stages of Differentiation process Inhibited lipid droplet formation and Adipogenic gene expression.
ANTF Treatment decreased expression of PPARγ protein, a master transcription factor in the regulation of Adipocyte Differentiation, leading to a suppression of aP2 promoter activity. These results suggest that ANTF exerts potent Anti-Adipogenic effects via direct suppression of PPARγ protein expression, with consequent downregulation of Adipogenic gene expression.


Antrodia cinnamomea


Aqueous extract of Antrodia cinnamomea Reduced high-fat diet-induced Obesity in mice and suppressed Adipogenesis in 3T3-L1 cells


Antrodia cinnamomea (AC) has been used as a natural dietary supplement for health promotion.

•AC prevents high fat diet-induced Obesity in vivo.

•AC Inhibits Adipogenesis and mitotic clonal expansion in vitro.

•AC has potential applications to treat Obesity , insulin resistance and hepatic steatosis.

This study examined the Anti-Obesity and Anti-Adipogenic effects of Antrodia cinnamomea. Oral administration of aqueous extract of A. cinnamomea (ACW) significantly Reduced high-fat diet-induced body Weight Gain and relative perirenal and mesenteric fat weight in C57BL/6J mice. Administration of ACW significantly Reduced serum insulin, leptin, aspartate aminotransferase levels, HOMA-IR index, and hepatic cholesterol and triacylglycerol levels. Next, investigation of whether ACW and its polysaccharides (PS) and non-polysaccharides (NPS) subfractions possess Anti-Adipogenic action was conducted. Evidence showed that ACW, PS and NPS significantly Inhibited lipid deposits in 3T3-L1 Adipocytes.
Both ACW and NPS significantly Inhibited mitotic clonal expansion process of Adipocyte Differentiation. ACW and NPS also significantly decreased the expressions of PPARγ, C/EBPα, aP2 and FAS genes, while PS markedly Inhibited PPARγ and aP2 gene expression. Our data indicated that ACW Inhibits Preadipocyte Differentiation and Adipogenesis. These results suggested that ACW may have therapeutic potential for Obesity and related metabolic disorders.


Apigetrin (apigenin-7-O-glucoside) (Cosmos bipinnata Cav.)


Apigetrin Inhibits Adipogenesis in 3T3-L1 cells by downregulating PPARγ and CEBP-αAbstract


Background: Apigetrin, a flavonoid found in many plant leaves and seeds, has been known to possess antimutagenic, anti-cancer, antioxidant and anti-inflammatory properties. Here, we are investigating the effect of the apigetrin on Adipocytes Differentiation in 3T3-L1 Adipocytes, and elucidating the mechanism of its action.

Methods: Lipids accumulation was measured by Oil Red O staining and cell cycle was analyzed by flow cytometry. The antioxidant effect of apigetrin was evaluated against hydrogen peroxide. The expression of various genes, involved in Adipogenesis and Inflammation, was studied by real-time PCR.

Results: Our results showed that apigterin treatment Inhibited significantly lipid accumulation without effect on cell viability at 100 μM, and it exerted the Anti-Adipogenic effect during the early stages of Differentiation. Flow cytometry analysis showed that apigenin-7-O-glucoside (Ap7G) Inhibited cell proliferation during mitotic clonal expansion and caused cell cycle delay. Quantitative PCR analysis revealed that the mRNA levels of C/EBP-α, PPAR-γ, SREBP-1c and FAS were suppressed after apigetrin treatment at 100 μM. Moreover, the mRNA level of pro-inflammatory genes (TNF-α and IL-6) were suppressed after apigterin treatment, at high concentration Preadipocyte cells.

Conclusion: Taken together, these results indicated that apigenin-7-O-glucoside Inhibits Adipogenesis of 3T3-L1 preadipocytes at early stage of Adipogenesis.



Arctigenin


Although arctigenin (ARC) has been reported to have some pharmacological effects such as anti‐Inflammation, anti‐cancer, and antioxidant, there have been no reports on the anti‐Obesity effect of ARC. The aim of this study is to investigate whether ARC has an anti‐Obesity effect and mediates the AMP‐activated protein kinase (AMPK) pathway. We investigated the anti‐Adipogenic effect of ARC using 3T3‐L1 pre‐Adipocytes and human Adipose Tissue‐derived mesenchymal stem cells (hAMSCs). In high‐fat diet (HFD)‐induced obese mice, whether ARC can Inhibit Weight Gain was investigated. We found that ARC Reduced Weight Gain, fat pad weight, and triglycerides in HFD‐induced obese mice.
ARC also Inhibited the expression of peroxisome proliferator‐activated receptor gamma (PPARγ ) and CCAAT/enhancer‐binding protein alpha (C/EBPα) in in vitro and in vivo. Furthermore, ARC induced the AMPK activation resulting in down‐modulation of Adipogenesis‐related factors including PPARγ, C/EBPα, fatty acid synthase, Adipocyte fatty acid‐binding protein, and lipoprotein lipase. This study demonstrates that ARC can Reduce key Adipogenic factors by activating the AMPK in vitro and in vivo and suggests a therapeutic implication of ARC for Obesity treatment.


Arctiin (Arctium lappa L.seed)

Arctiin Inhibits Adipogenesis in 3T3-L1 cells and decreases Adiposity and body weight in mice fed a high-fat diet



Aristolochia Manshuriensis Kom


Aristolochia Manshuriensis Kom Inhibits Adipocyte Differentiation by Regulation of ERK1/2 and Akt Pathway


Aristolochia manshuriensis Kom (AMK) is a traditional medicinal herb used for the treatment of arthritis, rheumatism, hepatitis, and Anti-Obesity. Because of nephrotoxicity and carcinogenicity of AMK, there are no pharmacological reports on Anti-Obesity potential of AMK. Here, we showed AMK has an Inhibitory effect on Adipocyte Differentiation of 3T3-L1 cells along with significantly decrease in the lipid accumulation by downregulating several Adipocyte-specific transcription factors including peroxisome proliferation-activity receptor γ (PPAR-γ), CCAAT/enhancer binding protein α (C/EBP-α) and C/EBP-β, which are critical for Adipogenesis in vitro.

AMK also markedly activated the extracellular signal-regulated protein kinase 1/2 (ERK1/2) pathway including Ras, Raf1, and mitogen-activated protein kinase kinase 1 (MEK1), and significantly suppressed Akt pathway by Inhibition of phosphoinositide-dependent kinase 1 (PDK1). Aristolochic acid (AA) and ethyl acetate (EtOAc) fraction of AMK with AA were significantly Inhibited TG accumulation, and regulated two pathway (ERK1/2 and Akt) during Adipocyte Differentiation, and was not due to its cytotoxicity. These two pathways were upstream of PPAR-γ and C/EBPα in the Adipogenesis. In addition, gene expressions of secreting factors such as fatty acid synthase (FAS), adiponectin, lipopreotein lipase (LPL), and aP2 were significantly Inhibited by treatment of AMK during Adipogenesis. We used the high-fat diet (HFD)-induced Obesity mouse model to determine the Inhibitory effects of AMK on Obesity.

Oral administration of AMK (62.5 mg/kg/day) significantly decreased the fat tissue weight, total cholesterol (TC), and low density lipoprotein-cholesterol (LDL-C) concentration in the blood. The results of this study suggested that AMK Inhibited lipid accumulation by the down-regulation of the major transcription factors of the adipogensis pathway including PPAR-γ and C/EBP-α through regulation of Akt pathway and ERK 1/2 pathway in 3T3-L1 Adipocytes and HFD-induced Obesity mice, and AA may be main act in Inhibitory effects of AMK during Adipocyte Differentiation.


Herbal Remedies against Adipogenesis

Aristolochia manshuriensis Kom is a traditional medicinal herb used for treatment of arthritis, rheumatism, hepatitis. Its extract Inhibited Adipocyte Differentiation by regulating ERK1/2 and Akt pathway. Besides, expressions of FAS, LPL and aP2 were significantly Reduced by the extract treatment during Adipogenesis.



Aristotelia chilensis


Antioxidant Capacity and in Vitro Inhibition of Adipogenesis and Inflammation by Phenolic Extracts of Vaccinium floribundum and Aristotelia chilensis


Interest in berries from South America has increased due to their potential health benefits. The objective of this study was to characterize the Anthocyanins and proanthocyanidins of Vaccinium floribundum and Aristotelia chilensis, total phenolics, and antioxidant capacity and to evaluate, in vitro, the ability of their phenolic extracts to Reduce Adipogenesis and lipid accumulation in 3T3-L1 Adipocytes. The anti-inflammatory property of these extracts on RAW 264.7 macrophages was also investigated. Antioxidant capacity, measured as oxygen radical scavenging capacity and expressed as Trolox equivalents, was higher in the berries of A. chilensis. Phenolic extracts Inhibited lipid accumulation by 4.0−10.8% when Adipocytes were treated at maturity and by 5.9−37.9% when treated throughout Differentiation.

Furthermore, a proanthocyanidin-enriched fraction from V. floribundum significantly increased Pref-1 expression in preadipocytes. Phenolic extracts decreased the production of nitric oxide (3.7−25.5%) and prostaglandin E2 (9.1−89.1%) and the expression of inducible nitric oxide synthase (9.8−61.8%) and cycloxygenase-2 (16.6−62.0%) in lipopolysaccharide-stimulated RAW 264.7 macrophages. V. floribundum and A. chilensisphytochemicals limit Adipogenesis and inflammatory pathways in vitro, warranting further in vivo studies.


KEYWORDS (keywords):

•Aristotelia chilensis

•Vaccinium floribundum

•Adipogenesis

•Anthocyanins

•Inflammation

•proanthocyanidins

•Obesity



Artemisia capillaris

Obesity has increased continuously in western countries during the last several decades and recently become a problem in developing countries. Currently, Anti-Obesity drugs originating from natural products are being investigated for their potential to overcome adverse effects associated with chemical drugs. Artemisinic acid, which was isolated from the well-known anti-malaria herb Artemisia annua (AA) L., was recently shown to possess Anti-Adipogenic effects in vitro. However, the Anti-Adipogenic effects of AA in animal models have not yet been investigated.

Therefore, we conducted daily oral administration with AA water extract in a diet-induced Obesity animal model and treated 3T3-L1 cells with AA to confirm the Anti-Adipogenic effects in the related protein expressions. We then evaluated the physiology, Adipose Tissue histology and mRNA expressions of many related genes. Inhibition of Adipogenesis by the AA water extract was observed in vitro. In the animal model, Weight Gain was significantly lower in the AA treated group, but there were no changes in food intake volume or calories. Reductions in lipid droplet size and mRNA expression associated with Adipogenesis were also observed in animal epididymal fat. This study is the first to report that AA has an anti-obese effects in vivo.



Aster yomena (Kitam.) Honda leaves


Ethanol extracts of Aster yomena (Kitam.) Honda Inhibit Adipogenesis through the activation of the AMPK signaling pathway in 3T3-L1 preadipocytes


The leaves of Aster yomena (Kitam.) Honda have long been used as a traditional herb for treating disorders including coughs, asthma, and insect bites. According to recent studies, A. yomena leaf extracts have several pharmacological properties, including anti-inflammatory, antioxidant, and anti-asthmatic activities. However, little information is available regarding their Anti-Obesity effect. In this study, we investigated the Inhibitory effect of the ethanol extracts of A. yomena leaves (EEAY) on Adipocyte Differentiation and Adipogenesis using 3T3-L1 preadipocytes. When 3T3-L1 preadipocytes were treated with various concentrations of EEAY (ranging from non-toxic), the number of lipid droplets, lipid content, and triglyceride production, the typical characteristics of Adipocytes, were suppressed in a concentration-dependent manner.

During this process, EEAY significantly Reduced the expression of Adipogenic transcription factors, including peroxisome proliferator-activated receptor-γ, CCAAT/enhancer-binding protein α and β, and sterol regulatory element-binding protein-1c. In addition, EEAY was also found to potently Inhibit the expression of Adipocyte-specific genes, including Adipocyte fatty acid-binding protein and leptin. In particular, EEAY treatment effectively enhanced the activation of the AMP-activated protein kinase (AMPK) signaling pathway; however, the co-treatment with compound C, an Inhibit or of AMPK, significantly restored the EEAY-induced Inhibition of Pro-Adipogenic transcription factors and Adipocyte-specific genes. These results indicate that EEAY may exert an Anti-Obesity effect by controlling the AMPK signaling pathway, suggesting that the leaf extract of A. yomena may be a potential Anti-Obesity agent.



Astilbe chinensis Franch. et Savet


AntiObesity Effect of Astilbe chinensis Franch. et Savet. Extract through Regulation of Adipogenesis and AMP-Activated Protein Kinase Pathways in 3T3-L1 Adipocyte and High-Fat Diet-Induced C57BL/6N Obese Mice


Astilbe chinensis Franch. et Savat. (AC) has been used in traditional medicine for the treatment of chronic bronchitis, arthralgia, and gastralgia. In this study, we investigated the AntiObesity effect of AC extract on 3T3-L1 preadipocytes and high-fat-diet-fed C57BL/6N obese mice. We found that AC extracts dramatically decreased the lipid content of 3T3-L1 cells in a concentration-dependent manner without cytotoxicity. The action mechanism of AC extract was demonstrated to be the Inhibition of lipid accumulation and dose-dependent decrease in the expression of CCAAT/enhancer-binding protein α (C/EBPα), peroxisome proliferator-activated receptor-γ (PPAR-γ), and sterol regulatory element-binding protein 1 (SREBP1).

Furthermore, AC extract increased the mitochondrial phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC), mitochondrial biogenesis, and Lipolysis -related factors. In amice model of high-fat-diet-induced Obesity, the mice administered AC extract experienced significant decrease of 64% in Weight Gain, 55% in insulin resistance index, 22% in plasma triglycerides (TG), 56% in total cholesterol (TC), and 21% in nonesterified fatty acid (NEFA) levels compared with those in the high-fat diet-fed control mice. Collectively, these results indicated that AC extract exerted antiobesogenic activity through the modulation of the AMPK signaling pathway, Inhibition of Adipogenesis, decreased lipid content, and Reduced Adipocyte size.



Astragalin, (3-O-glucoside of kaempferol) (persimmon leaves)


AntiObesity Effect of Astilbe chinensisFranch. et Savet. Extract through Regulation of Adipogenesis and AMP-Activated Protein Kinase Pathways in 3T3-L1 Adipocyte and High-Fat Diet-Induced C57BL/6N Obese Mice


Astilbe chinensis Franch. et Savat. (AC) has been used in traditional medicine for the treatment of chronic bronchitis, arthralgia, and gastralgia. In this study, we investigated the AntiObesity effect of AC extract on 3T3-L1 preadipocytes and high-fat-diet-fed C57BL/6N obese mice. We found that AC extracts dramatically decreased the lipid content of 3T3-L1 cells in a concentration-dependent manner without cytotoxicity. The action mechanism of AC extract was demonstrated to be the Inhibition of lipid accumulation and dose-dependent decrease in the expression of CCAAT/enhancer-binding protein α (C/EBPα), peroxisome proliferator-activated receptor-γ (PPAR-γ), and sterol regulatory element-binding protein 1 (SREBP1).

Furthermore, AC extract increased the mitochondrial phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC), mitochondrial biogenesis, and Lipolysis-related factors. In amice model of high-fat-diet-induced Obesity, the mice administered AC extract experienced significant decrease of 64% in Weight Gain, 55% in insulin resistance index, 22% in plasma triglycerides (TG), 56% in total cholesterol (TC), and 21% in nonesterified fatty acid (NEFA) levels compared with those in the high-fat diet-fed control mice.

Collectively, these results indicated that AC extract exerted antiobesogenic activity through the modulation of the AMPK signaling pathway, Inhibition of Adipogenesis, decreased lipid content, and Reduced Adipocyte size.



Averrhoa carambola L. peel


Averrhoa carambola L. peel extract Suppresses Adipocyte Differentiation in 3T3-L1 cells


Obesity is associated with an increased risk of many chronic diseases. Recently, a growing body of evidence has shown that phytochemicals may Inhibit Adipogenesis and Obesity. In this study, we report for the first time, the ability of Averrhoa carambola L. peel extract commonly known as star fruit (SFP) to effectively suppress Adipocyte Differentiation in 3T3-L1 preadipocytes and therefore, address it as a potential candidate to treat Obesity and its related diseases. (−)-Epicatechin was identified as a bioactive compound likely responsible for this suppression.

As the genetic expression studies revealed that the Adipogenic activity of SFP extract was due to the simultaneous downregulation of the C/EBPα and PPARγ as well as the upregulation of PPARα receptor genes, a detailed computational docking study was also elucidated to reveal the likely binding mode of (−)-epicatechin to the receptor of interest, accounting for the likely mechanism that results in the overall suppression of Adipocyte Differentiation.



Avicularin (Guava leaves)


Avicularin, a Plant Flavonoid, Suppresses Lipid Accumulation through Repression of C/EBPα -Activated GLUT4-Mediated Glucose Uptake in 3T3-L1 cells


Avicularin (quercetin-3-O-α-l-arabinofuranoside) is a plant flavonoid and a quercetin glycoside. In this study, we found that avicularin suppressed the accumulation of intracellular lipids through repression of glucose transporter 4 (GLUT4)-mediated glucose uptake in mouse adipocytic 3T3-L1 cells. Avicularin was highly purified (purity of more than at least 99%) from Taxillus kaempferi (DC.) Danser (Loranthaceae) by high-performance liquid chromatography, and its structure was determined by nuclear magnetic resonance and Mass spectrometry. Avicularin decreased the intracellular triglyceride level along with a reduction in the expression of Adipogenic genes such as peroxisome proliferator-activated receptor γ, CCAAT/enhancer-binding protein (C/EBP) α, and aP2 (fatty acid-binding protein 4).

In contrast, avicularin did not affect the expression of lipogenic and lipolytic genes. Interestingly, the expression of the GLUT4 gene was significantly suppressed in an avicularin-concentration-dependent manner. Moreover, the binding of C/EBPα to the promoter region of the GLUT4 gene was repressed by adding avicularin to the medium in 3T3-L1 cells, as demonstrated by the results of a chromatin immunoprecipitation assay. These results indicate that avicularin Inhibited the accumulation of the intracellular lipids by decreasing C/EBPα-activated GLUT4-mediated glucose uptake in Adipocytes.


Bacaba (Oenocarpus bacaba Mart.) phenolic (BPE)


Bacaba phenolic extract attenuates Adipogenesis by down-regulating PPARγ and C/EBPα in 3T3-L1 cells


•Bacaba (Oenocarpus bacaba Mart.) phenolic extract (BPE) Inhibits Differentiation in 3T3-L1 preadipocytes.

•BPE down-regulates protein expression of PPARγ and C/EBPα in a dose-dependent manner.

•During mitotic clonal expansion (early stage of Differentiation) BPE Inhibits lipid accumulation.

•Longer BPE incubation period is needed to effect protein expression.

Bacaba (Oenocarpus bacaba Mart.) is a native Brazilian palm fruit with a high amount of Polyphenol ics, reported to have an apoptotic effect on cancer cells [1]. Here, we examined the effect of Bacaba phenolic extract (BPE) on Adipogenesis using 3T3-L1 preadipocytes . Proliferating and differentiating Adipocytes were incubated with BPE at 6, 12, and 24 μg of gallic acid equivalents (GAE)/ml. BPE Reduced accumulation of intracellular lipids and protein expression of Adipogenic markers including PPARγ, C/EBPα, FABP4, IR-β, and adiponectin in a dose-dependent manner during Differentiation of 3T3-L1 cells into Adipocytes.

Furthermore, lipid accumulation decreased with BPE (24 μg of GAE/ml) during the early stage of mitotic clonal expansion (Days 0–2). In contrast, the Inhibition of protein expression of Adipogenic markers needed a longer duration (Days 0–4, 0–7, 2–7) of BPE incubation. These results suggest that BPE Inhibits adipogenisis in vitro via targeting transcriptional factors during the early and middle stages of Differentiation.


Bacaba (Oenocarpus bacaba Mart.) phenolic extract
3T3-L1 cells
Adipogenesis
PPARγ
C/EBPα


Baicalin (Scutellaria baicalensis)


AntiObesity effect of baicalin involves the modulations of proAdipogenic and AntiAdipogenic regulators of the Adipogenesis pathway


In this study, the AntiObesity effects of baicalin, 5,6‐dihydroxyflavone‐7‐glucuronic acid, were characterized using an in vitro system of Adipogenesis, i.e. fat cell formation. Baicalin‐treatment of 3T3‐L1 preadipocytes was shown to Inhibit triglyceride accumulation and lipid droplet formation during induced Adipogenesis. Microarray analyses showed that baicalin modulated the expression of genes located in pathways such as Adipogenesis, cholesterol biosynthesis, focal adhesion and others. In the Adipogenesis pathway, treatment with baicalin significantly down‐regulated terminal Differentiation markers of Adipocytes including fatty acid binding protein 4. The effects of baicalin on the core part of the Adipogenesis pathway, however, were paradoxical; the expression levels of CCAAT/enhancer binding protein (C/EBP)β and C/EBPδ were up‐regulated, while the expression levels of the peroxisome proliferator‐activated receptor (PPAR)γ and C/EBPα were down-regulated.

The antiAdipogenic mechanisms of baicalin can be explained by its effects on the upstream part of Adipogenesis pathway; baicalin not only up‐regulates the anti Adipogenic regulators, C/EBPγ, C/EBP homologous protein and Kruppel‐like factor (KLF)2, but also down‐regulates the pro Adipogenic regulator, KLF15. The overall effects of baicalin on these upstream regulators of Adipogenesis were antiAdipogenic, resulting in the down‐regulation of downstream genes and the Inhibition of cellular Fat Accumulation.



Bamboo (Phyllostachys bambusoides) leaf


Bamboo (Phyllostachys bambusoides) leaf extracts Inhibit Adipogenesis by regulating Adipogenic transcription factors and enzymes in 3T3-L1 Adipocytes


In this study, the Inhibitory effects of bamboo leaf extracts on Adipogenesis were investigated by evaluating their activity against Adipogenic transcription factors and enzymes in 3T3-L1 Adipocytes. Bamboo leaf extracts significantly decreased triglyceride levels, and increased glycerol release in Adipocytes. Cells treated with the water extract showed significantly higher glycerol release as well as lower triglyceride contents than those treated with the ethanol extract. Both bamboo leaf extracts significantly Inhibited the expression of Adipogenic transcription factors and enzymes, such as CCAAT/enhancer-binding protein α, sterol regulatory element binding protein 1c, peroxisome proliferator-activated receptor γ, acetyl-coenzyme A carboxylase, and fatty acid synthase, and increased the expression of phospho-adenosine monophosphate-activated protein kinase.

These results show that bamboo leaf extracts Inhibited Adipogenesis in 3T3-L1 Adipocytes and that the water extract was more efficacious than the ethanol extract.



Banaba (Lagerstroemia speciosa L.)


An Extract of Lagerstroemia speciosa L. Has Insulin-Like Glucose Uptake–Stimulatory and Adipocyte Differentiation–Inhibitory Activities in 3T3-L1 Cells


The effects of extracts isolated from Lagerstroemia speciosa L. (banaba) on glucose transport and adipocyte differentiation in 3T3-L1 cells were studied. Glucose uptake–inducing activity of banaba extract (BE) was investigated in differentiated adipocytes using a radioactive assay, and the ability of BE to induce differentiation in preadipocytes was examined by Northern and Western blot analyses. The hot water BE and the banaba methanol eluent (BME) stimulated glucose uptake in 3T3-L1 adipocytes with an induction time and a dose-dependent response similar to those of insulin. Furthermore, there were no additive or synergistic effects found between BE and insulin on glucose uptake, and the glucose uptake activity of insulin could be reduced to basal levels by adding increasing amounts of BE.

Unlike insulin, BE did not induce adipocyte differentiation in the presence of 3-isobutyl-1-methylxanthine (IBMX) and dexamethasone (DEX). BE inhibited the adipocyte differentiation induced by insulin plus IBMX and DEX (IS-IBMX-DEX) of 3T3-L1 preadipocytes in a dose-dependent manner. The differences in the glucose uptake and differentiation inhibitory activities between untreated cells and those treated with BE were significant (P < 0.01). The inhibitory activity was further demonstrated by drastic reductions of peroxisome proliferator-activated receptor γ2 (PPARγ2) mRNA and glucose transporter-4 (GLUT4) protein in cells induced from preadipocytes with IS-IBMX-DEX in the presence of BE. The unique combination of a glucose uptake stimulatory activity, the absence of adipocyte differentiation activity and effective inhibition of adipocyte differentiation induced by IS-IBMX-DEX in 3T3-L1 cells suggest that BE may be useful for prevention and treatment of hyperglycemia and obesity in type II diabetics.



Benincasa hispida


Effects of Fractions from Benincasa hispida on Inhibition of Adipogenesis in 3T3-L1 preadipocytes


The effects of three fractions, hexane (BHHH), chloroform (BHHC), and ethyl acetate (BHHE), from water extract of Benincasa hispida on the underlying mechanisms of Adipogenesis were investigated in 3T3-L1 cells. Intracellular lipid droplets were stained with Oil Red O dye and quantified. Compared to control, lipid accumulation significantly decreased by 11% and 13% upon treatment with BHHC and BHHE, respectively at a concentration of 50 . Intracellular triglyceride (TG) levels were also Reduced by 21% and 16%, respectively, at the same concentration. To determine the mechanism behind the reductions in TG content and lipid accumulation, glycerol release and expression levels of Adipogenic marker genes were measured. The levels of free glycerol released into culture medium increased by 13% and 17% upon treatment with BHHC and BHHE, respectively.

In subsequent measurements using real-time polymerization chain reaction, the mRNA levels of , C/, and leptin significantly decreased upon treatment with BHHE (45%, 67%, and 35%) in comparison with non-treated control. These results suggest that BHHE Inhibits Adipocyte Differentiation by blocking , C/, and leptin gene expression in 3T3-L1 cells , resulting in Reduced lipid accumulation, increased glycerol release, and intracellular triglycerides.



Berberine


Berberine Inhibits Adipocyte Differentiation , proliferation and Adiposity through down-regulating galectin-3


This study is designed to investigate the effects of berberine (BBR) on galectin-3 (Gal-3) and the relationships to its suppressive activities on Adipocyte Differentiation , proliferation and Adiposity . Our results showed that BBR greatly suppressed the Differentiation and proliferation of mouse primary preadipocytes isolated from epididymal white Adipose Tissue (eWAT), during which the expression level of Gal-3 was down-regulated significantly. Overexpression of Gal-3 totally abolished the suppressive activities of BBR on Gal-3 expression, Preadipocyte Differentiation and proliferation. BBR Reduced Gal-3 promoter activity, destabilized its mRNA and Inhibited firefly luciferase activity of a recombinant plasmid containing the Gal-3 3′ untranslated region (UTR). Furthermore, BBR up-regulated microRNA (miRNA) let-7d expression and the suppressive activity on Gal-3 3′UTR was abolished by point mutation on the let-7d binding site. In mice fed a high-fat diet (HFD), BBR up-regulated let-7d and down-regulated Gal-3 expression in eWAT; it also suppressed Adipocyte Differentiation and proliferation and Reduced Adiposity greatly.

In summary, our study proves that BBR Inhibits the Differentiation and proliferation of Adipocytes through down-regulating Gal-3, which is closely associated with its Anti-Obesity effect. Our results may support the future clinical application of BBR for the treatment of Obesity or related diseases.



Bergamottin (Bergamot)


Bergamottin Inhibits Adipogenesis in 3T3-L1 cells and Weight Regulation in Diet-Induced Obese Mice



Blumea balsamifera


Anti-Obesity Effect of Blumea balsamifera Extract in 3T3-L1 preadipocytes and Adipocytes


Obesity, the leading metabolic disease in the world, is a serious health problem in industrialized countries. We investigated the Anti-Obesity effect of Blumea balsamifera extract on Adipocyte Differentiation of 3T3-L1 preadipocytes and Anti-Obesity effect of 3T3-L1 Adipocytes. We found that treatment with an extract of Blumea balsamifera suppressed lipid accumulation and glycerol-3-phosphate dehydrogenase (GPDH) activity without affecting cell viability in 3T3-L1 preadipocytes and Adipocytes.

Furthermore, Blumea balsamifera extract brought significant attenuation of expressions of key Adipogenic transcription factors, including peroxisome proliferator-activated receptor (PPAR)γ, CCAAT element binding protein (C/EBPs) and leptin, however, induced up-regulation of adiponectin at the protein level in 3T3-L1 preadipocytes and Adipocytes. These results suggest that Blumea balsamifera extract may block Adipogenesis, at least in part, by decreasing key Adipogenic transcription factors in 3T3-L1 preadipocytes and may have antiatherogenic, anti-inflammatory, and AntiDiabetic effects through up-regulation of adiponectin in 3T3-L1 Adipocytes.


Boldine (Peumus boldus Molina leaf)


The Aporphine Alkaloid Boldine Induces Adiponectin Expression and Regulation in 3T3-L1 cells


Adiponectin is an adipokine secreted by differentiated Adipocytes . Clinical studies suggest a negative correlation between oxidative stress and adiponectin levels in patients with Metabolic Syndrome or cardiovascular disease. Natural compounds that can Prevent oxidative stress mediated Inhibition of adiponectin may be potentially therapeutic. Boldine, an aporphine alkaloid abundant in the medicinal plant Peumus boldus, is a powerful antioxidant. The current study demonstrates the effects of boldine on the expression of adiponectin and its regulators, CCAAT/enhancer binding protein-α (C/EBPα) and peroxisome proliferator-activated receptor (PPAR)-γ, in 3T3-L1 cells.

Differentiated 3T3-L1 Adipocytes were exposed to either hydrogen peroxide (H2O2) (100 μM) or tumor necrosis factor-α (TNFα) (1 ng/mL) for 24 hours in the presence or absence of increasing concentrations of boldine (5–100 μM). Quantitative polymerase chain reaction showed that both the oxidants decreased the mRNA levels of adiponectin, PPARγ, and C/EBPα to half of the control levels. Boldine, at all concentrations, counteracted the Inhibitory effect of H2O2 or TNFα and increased the expression of adiponectin and its regulators. The effect of boldine on adiponectin expression was biphasic, with the lower concentrations (5–25 μM) having a larger inductive effect compared to higher concentrations (50–100 μM). Boldine treatment alone in the absence of H2O2 or TNFα was also able to induce adiponectin at the inductive phase of Adipogenesis.

Peroxisome proliferator response element-luciferase promoter transactivity analysis showed that boldine interacts with the PPAR response element and could potentially modulate PPAR responsive genes. Our results indicate that boldine is able to modulate the expression of adiponectin and its regulators in 3T3-L1 cells and has the potential to be beneficial in Obesity-related cardiovascular disease.


Boussingaulti gracilis Miers var. pseudobaselloides Bailey


Anti-Obesity Effects of Boussingaulti gracilis Miers var. pseudobaselloides Bailey via Activation of AMP-Activated Protein Kinase in 3T3-L1 cells


In a previous study, we demonstrated the Anti-Obesity and hypolipidemic effects of Boussingaulti gracilis Miers var. pseudobaselloides Bailey in high-fat diet-induced obese rats. The present study investigated the molecular mechanisms by which B. gracilis Miers var. pseudobaselloides Bailey ethanol extract (BGE) conferred antiDifferentiation and Anti-Adipogenic effects in the 3T3-L1 Preadipocyte Differentiation model. BGE treatment significantly and dose-dependently suppressed lipid accumulation and down-regulated the expression of major transcription factors involved in Adipogenesis, such as peroxisome proliferator-activated receptor-γ, CCAAT/enhancer binding protein α, sterol regulatory element-binding proteins, and their target genes.

It is important that treatment with BGE increased phosphorylation of AMP-activated protein kinase (AMPK), which is one of the rate-limiting enzymes in the fatty acid synthesis pathway, and its direct downstream protein, acetyl-coenzyme A carboxylase. These results suggest that BGE may exert Anti-Adipogenic effects through regulation of AMPK activity and expression of genes involved in lipogenesis.



Bromophenol (5‐bromo‐3,4‐dihydroxybenzaldehyde) isolated from red alga Polysiphonia morrowii



The aim of the present study was to investigate the effect of 5‐bromo‐3,4‐dihydroxybenzaldehyde (BD) isolated from Polysiphonia morrowii on Adipogenesis and Differentiation of 3T3‐L1 preadipocytes into mature Adipocytes and its possible mechanism of action. Levels of lipid accumulation and triglyceride were significantly lower in BD treated cells than those in untreated cells. In addition, BD treatment Reduced protein expression levels of peroxisome proliferator‐activated receptor‐γ, CCAAT/enhancer‐binding proteins α, and sterol regulatory element‐binding protein 1 compared with control (no treatment).

It also Reduced expression levels of adiponectin, leptin, fatty acid synthase, and fatty acid binding protein 4. AMP‐activated protein kinase activation was found to be one specific mechanism involved in the effect of BD. These results demonstrate that BD possesses Inhibitory effect on Adipogenesis through activating AMP‐activated protein kinase signal pathway.



Butein Chalconoid


Butein is a novel Anti-Adipogenic compound


Rhus verniciflua Stokes (RVS) has been used as a traditional herbal medicine for its various biological activities including Anti-Adipogenic effects. Activity-guided separation led to the identification of the Anti-Adipogenic functions of butein. Butein, a novel Anti-Adipogenic compound, robustly suppressed lipid accumulation and Inhibited expression of Adipogenic markers. Molecular studies showed that activated transforming growth factor-β (TGF-β) and suppressed signal transducer and activator of transcription 3 (STAT3) signaling pathways were mediated by butein. Analysis of the temporal expression profiles suggests that TGF-β signaling precedes the STAT3 in the butein-mediated Anti-Adipogenic cascade.

Small interfering RNA-mediated silencing of STAT3 or SMAD2/3 blunted the Inhibitory effects of butein on Adipogenesis indicating that an interaction between two signaling pathways is required for the action of butein. Upon butein treatments, stimulation of TGF-β signaling was still preserved in STAT3 silenced cells, whereas regulation of STAT3 signaling by butein was significantly impaired in SMAD2/3 silenced cells, further showing that TGF-β acts upstream of STAT3 in the butein-mediated anti-Adipogenesis.

Taken together, the present study shows that butein, a novel Anti-Adipogenic compound from RVS, Inhibits Adipocyte Differentiation through the TGF-β pathway followed by STAT3 and peroxisome proliferator-activated receptor γ signaling, further implicating potential roles of butein in TGF-β- and STAT3-dysregulated diseases.



Capsaicin


Effects of Capsaicin on Induction of Apoptosis and Inhibition of Adipogenesis in 3T3-L1 cells


Currently, at the beginning of the 21st century, Obesity has become the leading metabolic disease in the world. It is a serious health problem in industrialized countries. Previous research has suggested that decreased Preadipocyte Differentiation and proliferation and decreased lipogenesis are mechanisms to Reduce Obesity. In the present study, the effects of capsaicin on the induction of apoptosis and Inhibition of lipid accumulation in 3T3-L1 preadipocytes and Adipocytes were investigated. The results demonstrated that capsaicin decreased cell population growth of 3T3-L1 preadipocytes, assessed with the MTT assay. Flow cytometric analysis of 3T3-L1 preadipocytes exposed to capsaicin showed that apoptotic cells increased in a time- and dose-dependent manner. Treatment with capsaicin decreased the number of normal cells and increased the number of early apoptotic and late apoptotic cells in a dose-dependent manner.

The treatment of cells with capsaicin caused the loss of mitochondria membrane potential (ΔΨm). The induction of apoptosis in 3T3-L1 preadipocytes by capsaicin was mediated through the activation of caspase-3, Bax, and Bak, and then through the cleavage of PARP and the down-regulation of Bcl-2. Moreover, capsaicin significantly decreased the amount of intracellular triglycerides and glycerol-3-phosphate dehydrogenase (GPDH) activity in 3T3-L1 Adipocytes. Capsaicin also Inhibited the expression of PPARγ, C/EBPα, and leptin, but induced up-regulation of adiponectin at the protein level. These results demonstrate that capsaicin efficiently induces apoptosis and Inhibits Adipogenesis in 3T3-L1 preadipocytes and Adipocytes.

Keywords: Capsaicin; Adipogenesis; 3T3-L1 cells; apoptosis; protein expression.



Carduus crispus


Inhibition of Adipocyte Differentiation by MeOH Extract from Carduus crispus through ERK and p38 MAPK Pathways


In this study, the effects of a methanol (MeOH) extract of Carduus crispus L. (Asteraceae) on Adipogenesis was investigated in 3T3-L1 cells. To differentiate preadipocytes to Adipocytes, confluent 3T3-L1 preadipocytes were treated with a hormone mixture, which included isobutylmethylxanthine, dexamethasone, and insulin (MDI). The methanol extract of C. crispus significantly decreased Fat Accumulation by Inhibiting Adipogenic signal transcriptional factors in MDI-induced 3T3-L1 cells in a dose-dependent manner.

In MTT assays and on PI-staining, methanol extract of C. crispus Inhibited the proliferation of 3T3-L1 cells during mitotic clonal expansion (MCE). The Anti-Adipogenic effect of the Carduus extract seemed to be associated with the up-regulation of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) pathways within the first 2 days after MDI treatment. These results suggest that methanol extract of C. crispus might be beneficial for the treatment of Obesity.



Carnosic acid (Salvia japonica Thunb.)


Carnosic acid and carnosol Inhibit Adipocyte Differentiation in mouse 3T3-L1 cells through induction of phase2 enzymes and activation of glutathione metabolism


In the previous studies, we reported that carnosic acid (CA) and carnosol (CS) originating from rosemary protected cortical neurons by activating the Keap1/Nrf2 pathway, which activation was initiated by S-alkylation of the critical cysteine thiol of the Keap1 protein by the “electrophilic”quinone-type of CA or CS. Here, we found that CA and CS Inhibited the in vitro Differentiation of mouse preadipocytes, 3T3-L1 cells, into Adipocytes. In contrast, other physiologically-active and rosemary-originated compounds were completely negative. These actions seemed to be mediated by activation of the antioxidant-response element(ARE) and induction of phase2 enzymes. This estimation is justified by our present findings that only CA and CS among rosemary-originated compounds significantly activated the ARE and induced the phase2 enzymes. Next, we performed cDNA microarray analysis in order to identify the gene(s) responsible for these biological actions and found that phase2 enzymes (Gsta2, Gclc, Abcc4, and Abcc1), all of which are involved in the metabolism of glutathione (GSH), constituted 4 of the top 5 CA-induced genes.

Furthermore, CA and CS, but not the other compounds tested, significantly increased the intracellular level of total GSH. Thus, we propose that the stimulation of GSH metabolism may be a critical step for the Inhibition of Adipocyte Differentiation in 3T3-L1 cells and suggest that pro-electrophilic compounds such as CA and CS may be potential drugs against Obesity-related diseases.


AntiAdipogenic effect of carnosic acid is exerted through the C/EBPs and PPARγ pathways at the onset of the Differentiation program


•The antiAdipogenic mechanism of carnosic acid is multifactorial.

•Carnosic acid Inhibits mitotic clonal expansion of 3T3-L1 cells.

•The expression of C/EBPα , PPARγ and FABP2 are Inhibited by carnosic acid.
Carnosic acid alters the ratio of C/EBPβ forms and its subnuclear distribution.
•Carnosic acid is a potential agent for the treatment/prevention of Obesity.

Background: Obesity is a serious health problem all over the world, and Inhibition of Adipogenesis constitutes one of the therapeutic strategies for its treatment. Carnosic acid (CA), the main bioactive compound of Rosmarinus officinalis extract, Inhibits 3T3-L1 preadipocytes Differentiation. However, very little is known about the molecular mechanism responsible for its antiAdipogenic effect.

Methods: We evaluated the effect of CA on the Differentiation of 3T3-L1 preadipocytes analyzing the process of mitotic clonal expansion, the level of Adipogenic markers, and the subcellular distribution of C/EBPβ.

Results: CA treatment only during the first day of 3T3-L1 Differentiation process was enough to Inhibit Adipogenesis. This Inhibition was accompanied by a blockade of mitotic clonal expansion. CA did not interfere with C/EBPβ and C/EBPδ mRNA levels but blocked PPARγ , and FABP4 expression. C/EBPβ has different forms known as LIP and LAP. CA induced an increase in the level of LIP within 24 h of Differentiation, leading to an increment in LIP/LAP ratio. Importantly, overexpression of LAP restored the capacity of 3T3-L1 preadipocytes to differentiate in the presence of CA. Finally, CA promoted subnuclear de-localization of C/EBPβ.

Conclusions: CA exerts its Anti-Adipogenic effect in a multifactorial manner by interfering mitotic clonal expansion, altering the ratio of the different C/EBPβ forms, inducing the loss of C/EBPβ proper subnuclear distribution, and blocking the expression of C/EBPα and PPARγ.

General significance: Understanding the molecular mechanism by which CA blocks Adipogenesis is relevant because CA could be new a food additive beneficial for the prevention and/or treatment of Obesity.



Carnosine (L-Carnosine) (β – alanine and L-histidine)


Synergistic effect of carnosine on browning of Adipose Tissue in exercised obese rats; a focus on circulating irisin levels


The recent appreciation of the energy burning capacity of brown Adipose Tissue turns it to an attractive target for anti‐Obesity therapy. We sought to evaluate the effect of L‐carnosine on browning of white Adipose Tissue in exercised obese rats. Sixty adult male Wistar albino rats, 7–8 week‐old weighing 130–150 g, were allocated into six groups; with 10 rats in each, for an experimentation period of 12 weeks: (i) normal control rats fed a standard fat diet (SFD/control), (ii) normal control rats fed a standard diet and injected with L‐carnosine (250 mg/kg, i.p,) for 6 weeks (SFD/CAR), (iii) high‐fat diet (HFD)‐induced obese rats for 12 weeks, (iv) HFD rats subjected to exercise training (HFD/EXE) for 6 weeks, (v) HFD rats injected with L‐carnosine (250 mg/kg,i.p.) for 6 weeks (HFD/CAR) and, (vi) HFD rats subjected to exercise training and L‐carnosine (HFD/EXE/CAR). At the end of the 12‐week‐experiment, the body weights and the serum levels of lipid profile, oxidative stress, and inflammatory markers as well as circulating myokines were investigated. Gastrocnemius muscles and inguinal Adipose Tissues were excised for the measurement of gene expression of muscle irisin, Adipose Tissue uncoupling protein1 (UCP1), CD137 and the protein level of p38MAPK.

In addition, histopathological examination for the studied groups was performed. Both exercise training for 6 weeks and carnosine treatment significantly decreased body Weight Gain, ameliorated Obesity‐induced dyslipidemia, Reduced the thiobarbituric acid reactive species (TBARS) and TNF‐α, while increased total antioxidant capacity and IL‐10. Furthermore, increases in serum irisin levels and the expression of adipose uncoupling protein‐1 (UCP‐1), adipose CD137, p38 MAPK, and muscular fibronectin type III domain‐containing protein 5(FNDC5), the precursor of irisin gene expression, were correlated with these carnosine‐ and exercise‐induced physiological improvements. The highest improvement was evident in the combined exercise and carnosine group which indicates that their beneficial effects in obese animals were synergistic. These findings suggest that L‐carnosine may induce browning of Adipose Tissue through irisin stimulation, a phenomenon that could be related to its antioxidant, Anti‐Inflammatory, and anti‐Obesity effects.



Celastrol (Tripterygium wilfordii)


Cascade regulation of PPARγ2 and C/EBPα signaling pathways by celastrol impairs adipocyte differentiation and stimulates lipolysis in 3T3-L1 adipocytes


Objective: Celastrol, a triterpene from the root bark of the Chinese medicinal plant Tripterygium wilfordii, has been shown to exhibit anti-oxidant, anti-inflammatory, anti-cancer and insecticidal activities. Also, it has been demonstrated that celastrol has obesity-controlling effects in diet-induced obesity mice. However, direct evidence that celastrol contributes to the development of adipocyte differentiation and lipolysis has not been fully elucidated. Moreover, no previous studies have evaluated whether celastrol may regulate adipogenic transcriptional markers in adipocytes.

Materials/Methods: In order to address the questions above, we extended previous observations and investigated in vitro celastrol signaling study whether celastrol may regulate differentiation, lipolysis and key adipogenic transcriptional pathways in 3T3-L1 adipocytes.

Results: Treatment of celastrol not only inhibited adipocyte differentiation (lipid accumulation, glyceraldehyde-3-phosphate dehydrogenase activity and triglyceride content) but also increased lipolysis (glycerol release and free fatty acid release) in 3T3-L1 adipocytes. In addition, all celastrol-regulated functional activities were controlled by PPARγ2 and C/EBPα signaling pathways in duration of celastrol’s treatment in 3T3-L1 adipocytes.

Conclusion: Our initial data from in vitro celastrol signaling studies suggest novel insights into the role of PPARγ2 and C/EBPα as probable mediators of the action of celastrol in regulating adipocyte differentiation and lipolysis in 3T3-L1 adipocytes.



Chromolaena odorata leaves


Anti-Adipogenic effect of Flavonoids from Chromolaena odorata leaves in 3T3-L1 Adipocytes


Objective: The leaves of Chromolaena odorata, a highly invasive shrub found growing wild worldwide, are traditionally used for wound healing. Due to its high flavonoidcontents, we aimed to find a new application for this plant. Preliminary tests using its ethanolic leaf extract showed that it could suppress the accumulation of lipids in Adipocytes. We therefore studied the Anti-Adipogenic effect of several C. odorata leaf extracts and the relationship between molecular structure and bio-activity of its isolated flavonoid constituents using 3T3-L1 preadipocytes /Adipocytes as a model.

Methods: Three leaf extracts and thirteen Flavonoids isolated from C. odorata were tested for their effect on lipid accumulation in 3T3-L1 Adipocytes using AdipoRed reagent, with quercetin as the positive control. The effects of active Flavonoids on the Adipocytes were confirmed by oil red O staining and visualized under a light microscope.

Results: n-Hexane and ethyl acetate extracts of C. odorata leaves displayed Anti-Adipogenic activity. The latter extract was the more potent one, especially at 40 µg/mL. Four Flavonoids, pectolinarigenin, kaempferide, 4,2′-dihydroxy-4′,5′,6′-trimethoxychalcone and dillenetin, exhibited significant, concentration-dependent Inhibitory effects on lipid accumulation in 3T3-L1 Adipocytes . The most potent flavonoid obtained in this study was 4,2′-dihydroxy-4′,5′,6′-trimethoxychalcone, which caused 75% and 90% Inhibition of cellular lipid accumulation at 30 and 50 µmol/L, respectively. Both kaempferide and 4,2′-dihydroxy-4′,5′,6′-trimethoxychalcone were major constituents in the ethyl acetate extract of this plant.

Conclusion: C. odorata leaves contained several Flavonoids with Anti-Adipogenic effects against lipid accumulation in 3T3-L1 Adipocytes. The plant, normally considered a useless weed, may actually provide an abundant source of biologically active Flavonoids.



Chrysanthemum zawadskii


Anti-Adipogenic Effects of Ethanol Extracts Prepared from Selected Medicinal Herbs in 3T3-L1 cells



Chrysin (Oroxylum indicum(L.)Vent.seed)


Chrysin induces brown fat–like phenotype and enhances lipid metabolism in 3T3-L1 Adipocytes


Objectives: Many studies have to do with promising therapeutic phytochemicals such as Flavonoids to treat Obesity and related complications, and a number of dietary compounds have been proposed as tools for increasing energy expenditure and decreasing Fat Accumulation in mammals. Here, we show that the flavonoid chrysin induces browning of 3T3-L1 Adipocytes via enhanced expression of brown fat–specific genes and proteins as well as enhances lipid metabolism.

Methods: Chrysin-induced fat browning was investigated by determining expression levels of brown fat–specific genes and proteins by real-time polymerase chain reaction and immunoblot analysis, respectively.

Results: Chrysin enhanced expression of brown fat–specific markers and increased protein levels of peroxisome proliferator-activated receptor (PPAR)α, PPARγ, PPARδ, phosphorylated AMP-activated protein kinase (p-AMPK), phosphorylated acetyl-CoA carboxylase, hormone sensitive lipase, perilipin, carnitine palmitoyltransferase 1, acyl-coenzyme A oxidase 1, peroxisome proliferator-activated receptor-1 alpha (PGC-1α), and uncoupling protein 1 (UCP-1), suggesting its possible role in augmentation of Lipolysis, fat oxidation, and thermogenesis as well as reduction of lipogenesis. Increased expression of UCP-1 and other brown fat–specific markers was possibly mediated by chrysin-induced activation of AMPK based on the fact that Inhibition of AMPK by dorsomorphin abolished expression of PR domain-containing 16, UCP-1, and PGC-1α while the activator 5-aminoimidazole-4-carboxamide ribonucleotide elevated expression of these brown marker proteins.

Conclusion: Our findings suggest that chrysin plays a dual modulatory role in the form of inducing the brown-like phenotype as well as enhancing lipid metabolism and thus may be explored as a potentially promising food additive for prevention of Obesity.



Cinnamomum verum


Inhibition of Adipocyte Differentiation and Adipogenesis by supercritical fluid extracts and marc from Cinnamomum verum


This study was performed to evaluate the AntiObesity effect of supercritical fluid extracts (SFC) and marc methanol extracts (SFM) from Cinnamomum verum in 3T3-L1 preadipocytes. In inducing the Differentiation of 3T3-L1 preadipocytes in the presence of an Adipogenic cocktail, iso-butylmethylanthine (IBMX), dexamathasone, and insulin, treatment with fraction residue SFC and SFM. SFC significantly Reduced the mRNA expression of the transcription factor peroxisome proliferator-activatedreceptor-γ (PPARγ), the sterol regulatory-element-binding protein-1c (SREBP1c), and the CCAAT enhancer-binding-protein α (C/EBPα) in a concentration-dependent manner.

Moreover, SFC markedly down-regulated acyl-CoA synthetase-1 (ASC1), fatty acid synthesis (FAS), fatty acid transport-1 (FATP1), fatty acid binding protein 4 (FABP4), and perilipin. These findings suggest that SFC may be a potential therapeutic adjunct for Obesity by targeting the Differentiation of preadipocytes, as well as their functions.



Cirsium brevicaule A. GRAY


Cirsium brevicaule A. GRAY leaf Inhibits Adipogenesis in 3T3-L1 cells and C57BL/6 mice


Background: Various Flavonoids obtained from the genus Cirsium have been reported to exhibit beneficial effects on health. The present study evaluated the AntiObesity effects of Cirsium brevicaule A. GRAY leaf (CL) by using 3T3-L1 cells and C57BL/6 mice that were fed a high-fat diet (HFD).

Methods: Dried CL powder was serially extracted with solvents of various polarities, and these extracts were tested for antiAdipogenic activity using 3T3-L1 Adipocytes . Mice were fed experimental HFD supplemented with dried CL powder for 4 wk. Lipid levels and mRNA levels of genes related to lipid metabolism were determined in 3T3-L1 Adipocytes and the white Adipose Tissue (WAT) and liver of mice fed on a HFD.

Results: Treatment of 3T3-L1 Adipocytes with a hexane extract of CL significantly Reduced cellular lipid accumulation and expression of the fatty acid synthase (FASN) gene. Dietary CL Reduced the serum levels of non-esterified fatty acids in HFD-fed mice. Significant decreases in subcutaneous WAT weight and associated FASN gene expression were observed in the mice fed the experimental CL diet. Dietary CL also Reduced the hepatic lipid and serum levels of a hepatopathic indicator in the HFD-fed mice. A significant reduction in mRNA levels of FASN and HMG-CoA reductase were observed in the livers of the CL-diet group. Dietary CL, on the other hand, increased in the hepatic mRNA levels of genes related to β-oxidation, namely peroxisome proliferator-activated receptor α, calnitine palmitoyltrasferase 1A, and uncoupling protein 2. Expression of the insulin receptor gene was also significantly increased in the livers of mice-fed the CL diet.

Conclusions: The present study therefore demonstrated that CL Suppresses lipid accumulation in the WAT and liver partly through Inhibiting mRNA levels of FASN gene and enhancing the Lipolysis -related gene expression.



Cirsium setidens Nakai


Cirsium setidens Nakai, a wild perennial herb, grows mainly in Gangwon province, Korea, and has been reported to contain bioactive ingredients with various medicinal activities, including the treatment of edema, bleeding, and hemoptysis. However, the potential AntiObesity effects of C. setidens Nakai have not been fully investigated. This study evaluated the AntiObesity effect of standardized C. setidens Nakai ethanolic extract (CNE) in 3T3-L1 Adipocytes and in obese C57BL/6J mice fed a high-fat diet. CNE suppressed the expression of lipogenic genes and increased the expression of lipolytic genes. The AntiAdipogenic and antilipogenic effects of CNE appear to be mediated by the Inhibition of peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein (C/EBP) expressions.

Moreover, CNE stimulated fatty acid oxidation in an AMPK-dependent manner. CNE-treated groups of C57BL/6J mice showed Reduced body weights and Adipose Tissue weight and improved serum lipid profiles through the downregulation of PPARγ, C/EBPα, fatty acid binding protein 4 (FABP4), sterol regulatory element binding protein-1c (SREBP-1c), and fatty acid synthase (FAS) and the upregulation of adiponectin and carnitine palmitoyltransferase-1 (CPT-1) in obese C57BL/6J mice fed a high-fat diet. These results suggest that CNE may have an AntiObesity effect on Adipogenesis and lipid metabolism in vitro and in vivo and present the possibility of developing a treatment for Obesity with nontoxic natural resources.


Assessment of the pectolinarin content and the radical scavenging-linked AntiObesity activity of Cirsium setidens Nakai extracts


Cirsium setidens Nakai contains bioactive compounds that exert biological activities. Method validation for analysis of the pectolinarin content in Cirsium setidens extracts (CSE) and radical scavenging-linked AntiObesity activities using 3T3-L1 cells and C57BL/6 mice were performed. The pectolinarin content of CSE was 2.81±0.01 mg/g with a high degree of linearity in calibration curves (R2=0.9999).

CSE exhibited free radical-scavenging activities and a reducing power. CSE and pectolinarin Inhibited lipid accumulation during Adipogenesis of 3T3-L1 cells via down-regulation of Adipogenic transcription factors. CSE supplementation suppressed body weight in C57BL/6 mice fed a high fat diet and Reduced plasma total cholesterol, triglyceride, insulin, and glucose levels. Pectolinarin-enriched CSE can be considered as a good source of natural Antioxidants and AntiObesity ingredients.



Citrus aurantium peel


Citrus aurantium Flavonoids Inhibit Adipogenesis through the Akt signaling pathway in 3T3-L1 cells


Background: Obesity is a health hazard that is associated with a number of diseases and metabolic abnormalities, such as type-2 Diabetes , hypertension, dyslipidemia, and coronary heart disease. In the current study, we investigated the effects of Citrus aurantium Flavonoids (CAF) on the Inhibition of Adipogenesis and Adipocyte Differentiation in 3T3-L1 cells.

Methods: During Adipocyte Differentiation, 3T3-L1 cells were treated with 0, 10, and 50 μg/ml CAF, and then the mRNA and protein expression of Adipogenesis-related genes was assayed. We examined the effect of CAF on level of phosphorylated Akt in 3T3-L1 cells treated with CAF at various concentrations during Adipocyte Differentiation.

Results: The insulin-induced expression of C/EBPβ and PPARγ mRNA and protein were significantly down-regulated in a dose-dependent manner following CAF treatment. CAF also dramatically decreased the expression of C/EBPα, which is essential for the acquisition of insulin sensitivity by Adipocytes. Moreover, the expression of the aP2 and FAS genes, which are involved in lipid metabolism, decreased dramatically upon treatment with CAF. Interestingly, CAF diminished the insulin-stimulated serine phosphorylation of Akt (Ser473) and GSK3β (Ser9), which may Reduce glucose uptake in response to insulin and lipid accumulation. Furthermore, CAF not only Inhibited triglyceride accumulation during Adipogenesis but also contributed to the Lipolysis of Adipocytes.

Conclusions: In the present study, we demonstrate that CAF suppressed Adipogenesis in 3T3-L1 Adipocytes . Our results indicated that CAF down-regulates the expression of C/EBPβ and subsequently Inhibits the activation of PPARγ and C/EBPα. The Anti-Adipogenic activity of CAF was mediated by the Inhibition of Akt activation and GSK3β phosphorylation, which induced the down-regulation of lipid accumulation and lipid metabolizing genes, ultimately Inhibiting Adipocyte Differentiation.



Clitoria ternatea Flower Petal


Clitoria ternatea Flower Petal Extract Inhibits Adipogenesis and Lipid Accumulation in 3T3-L1 preadipocytes by Downregulating Adipogenic Gene Expression


Clitoria ternatea (commonly known as blue pea) flower petal extract (CTE) is used as a natural colorant in a variety of foods and beverages. The objective of study was to determine the Inhibitory effect of CTE on Adipogenesis in 3T3-L1 preadipocytes. The phytochemical profiles of CTE were analyzed by liquid chromatography and tandem Mass spectrometry (LC-MS/MS). Anti-Adipogenesis effect of CTE was measured by using Oil Red O staining, intracellular triglyceride assay, quantitative real-time PCR and western blot analysis in 3T3-L1 Adipocytes. Cell cycle studies were performed by flow cytometry. Lipolysis experiments were performed using a colorimetric assay kit. In early stages, CTE demonstrated Anti-Adipogenic effects through Inhibition of proliferation and cell cycle retardation by suppressing expression of phospho-Akt and phospho-ERK1/2 signaling pathway.

The results also showed that CTE Inhibited the late stage of Differentiation through diminishing expression of Adipogenic transcription factors including PPARγ and C/EBPα. The Inhibitory action was subsequently attenuated in downregulation of fatty acid synthase and acetyl-CoA carboxylase, causing the reduction of TG accumulation. In addition, CTE also enhanced catecholamine-induced Lipolysis in Adipocytes. These results suggest that CTE effectively attenuates Adipogenesis by controlling cell cycle progression and downregulating Adipogenic gene expression.



Cocoa tea (Camellia ptilophylla)


Cocoa tea (Camellia ptilophylla) water extract Inhibits Adipocyte Differentiation in mouse 3T3-L1 preadipocytes


Cocoa tea (Camellia ptilophylla) is a naturally decaffeinated tea plant. Previously we found that cocoa tea demonstrated a beneficial effect against high-fat diet induced Obesity , hepatic steatosis and hyperlipidemia in mice. The present study aimed to investigate the Anti-Adipogenic effect of cocoa tea in vitro using preadipocytes 3T3-L1. Adipogenic Differentiation was confirmed by Oil Red O stain, qPCR and Western blot. Our results demonstrated that cocoa tea significantly Inhibited triglyceride accumulation in mature Adipocytes in a dose-dependent manner. Cocoa tea was shown to suppress the expressions of key Adipogenic transcription factors, including peroxisome proliferator-activated receptor gamma (PPAR γ) and CCAAT/enhancer binding protein (C/EBP α).

The tea extract was subsequently found to Reduce the expressions of Adipocyte-specific genes such as sterol regulatory element binding transcription factor 1c (SREBP-1c), fatty acid synthase (FAS), Acetyl-CoA carboxylase (ACC), fatty acid translocase (FAT) and stearoylcoenzyme A desaturase-1 (SCD-1). In addition, JNK, ERK and p38 phosphorylation were Inhibited during cocoa tea Inhibition of 3T3-L1 Adipogenic Differentiation. Taken together, this is the first study that demonstrates cocoa tea has the capacity to suppress Adipogenesis in pre-Adipocyte 3T3-L1 similar to traditional green tea



Colocynth (Citrullus colocynthis) Flesh


Colocynth (Citrullus colocynthis) seed extracts attenuate Adipogenesis by down-regulating PPARγ / SREBP-1c and C/EBPα in 3T3-L1 cells


Adipogenesis is the overall process of Adipocyte Differentiation. With a positive energy balance, intracellular accumulation of triglyceride increases through the generation of functional Adipocytes. This occurs with Adipogenesis of undifferentiated preadipocytes. Citrullus colocynthis (CC), a member of the Cucurbitaceae family, is a desert viny plant native to North Africa. The effect of ethanolic seed extracts of colocynth (SCEE) on Adipogenesis was investigated using 3T3-L1 preadipocytes. Cell viability was measured using the MTT assay; triglycerides were stained with Oil Red O and Adipogenesis-related gene expressions were quantified using qRT-PCR. Results showed that SCEE helps to Inhibit intracellular triglyceride accumulation during Adipogenesis without affecting cell viability. Likewise, SCEE not only showed Anti-Adipogenic activities, essentially during the early stage, but their effects during the middle and late stages were very low.

These effects have been confirmed by the down-regulation of C/EBPβ and C/EBPδ at 24, 48 and 72 h. In addition, SCEE treatment in 3T3-L1 cells induced down-regulation of the transcription factors CCAAT/enhancer binding protein α (C/EBPα), peroxisome proliferator activated receptor γ (PPARγ), and sterol regulatory element-binding protein 1c (SREBP-1c) and their target genes. UPLC-ESI-MS/MS profiling showed 10 bioactive compounds including quinic acid, isovitexin, scoparin, vitexin-2’’-O-rhamnoside, reserpine, digitoxin, triprolidine, naringenin, linoleic acid and oleic acid. Thus, these results suggested that the seeds of colocynth may have beneficial health effects.



Conjugated linoleic acid (CLA) (Ruminant fat)


Impact of conjugated linoleic acid on bone physiology: proposed mechanism involving Inhibition of Adipogenesis


Conjugated linoleic acid (CLA) supplementation decreases adipose Mass and increases bone Mass in mice. Recent clinical studies demonstrate a beneficial effect of CLA on reducing weight and adipose Mass in humans. This article reviews possible biological mechanisms of action of CLA on bone metabolism, focusing on modulation of nuclear receptor peroxisome proliferator‐activated receptor gamma activity to steer mesenchymal stem cell Differentiation toward an adipose and away from an osteoblast lineage. Clinical studies of the effects of CLA on bone Mass and clinical implications of the effects of CLA on bone health in humans are summarized and discussed.



Coptis chinensis (Coptidis Rhizoma)


Effect of Atractylodes Rhizoma Alba, Houttuyniae Herba, Lonicerae Flos, Scutellariae Radix and Coptidis Rhizoma Extracts Combined with Metformin on the Antioxidant and Adipocyte Differentiate Inhibition


Objectives: This study was to investigate the antioxiative capacity, AntiObesity effects of Atractylodes Rhizoma Alba, Houttuyniae Herba, Lonicerae Flos, Scutellariae Radix, and Coptidis Rhizoma on Raw 264.7 and 3T3-L1 cell lines.

Methods: Three different types of herb extracts (A. Rhizoma Alba, H. Herba, L. Flos, S. Radix, and C. Rhizoma; water 100%, ethanol 30%, ethanol 100%) were used in this study. Total Polyphenol compound, 1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging, reactive oxygen species (ROS) activity, NO production and cell proliferation were measured.

Results: Total Polyphenol compound measurement of L. Flos, A. Rhizogenes, and C. Rhizoma extracts were higher than A. Rhizoma Alba, H. Herba. DPPH radical scavenging activity, ROS activity and NO production of S. Radix, C. Rhizoma extracts were lower than L. Flos, A. Rhizoma, and H. Herba.

Conclusions: Metformin and S. Radix, C. Rhizoma, A. Rhizoma Alba, and L. Flos extracts combination groups showed synergistic effect on Adipocyte Differentiation Inhibition and antioxidative activity.



Cordycepin (Cordyceps sinensis)


The Effects of Cordyceps sinensis (Berk.) Sacc. and Gymnema inodorum (Lour.) Decne. Extracts on Adipogenesis and Lipase Activity In Vitro


This study aimed to investigate the effects of Cordyceps sinensis extract (CSE) and Gymnema inodorum extract (GIE), used alone and combined, on AntiAdipogenesis in 3T3-L1 cells . Oil Red O staining was used to examine the effects of these extracts on Inhibition of intracellular lipid accumulation in 3T3-L1 Adipocytes and on lipid droplet morphology. Fourier transform-infrared (FTIR) microspectroscopy was used to examine biomolecular changes in 3T3-L1 Adipocytes . The pancreatic lipase assay was used to evaluate the Inhibitory effects of CSE and GIE on pancreatic lipase activity.

Taken together, the results indicated that CSE, GIE, and their combination suppressed lipid accumulation. The FTIR microspectroscopy results indicated that CSE, GIE, and their combination had Inhibitory effects on lipid accumulation in the Adipocytes. Compared with the untreated Adipocytes, the signal intensity and integrated areas of glycogen and other carbohydrates, the acyl chain of phospholipids, and the lipid/protein ratios of the CSE, GIE, alone, and combined treated Adipocytes were significantly lower (p < 0.05). Combination treatment resulted in a synergistic effect on lipid accumulation reduction in the Adipocytes. Principal component analysis of the biomolecular changes revealed six distinct clusters in the FTIR spectra of the sample cells. The pancreatic lipase assay results indicated that CSE and GIE Inhibited the pancreatic lipase activity in a dose-dependent manner (mean ± standard error of the mean IC50 values, 2312.44 ± 176.55 μg mL−1 and 982.24 ± 44.40 μg mL−1, resp.). Our findings indicated that FTIR microspectroscopy has potential application for evaluation of the effectiveness of medicinal plants and for the development of infrared biochemical Obesity markers useful for treating patients with Obesity.

These results suggested that use of CSE and GIE alone and in combination may be efficacious as a complementary therapy for hyperlipidemia and Obesity management. However, clinical trials in animals and humans must first be completed.



Coumestrol (Tonka bean)


Coumestrol modulates Akt and Wnt/β-catenin signaling during the attenuation of Adipogenesis


Coumestrol is a natural phytochemical present in plants such as red clover and soy, and has been reported to stimulate the estrogen receptor as a major phytoestrogen. While the molecular mechanisms responsible for the Anti-Adipogenic effects of phytoestrogens such as genistein and daidzein have been previously investigated, the effects of coumestrol on Adipogenesis remain to be elucidated. We observed that coumestrol dose-dependently attenuates MDI (mixture of 3-isobutyl-1-methylxanthine, dexamethasone, and insulin)-induced lipid accumulation, consistent with an earlier study, while significantly Inhibiting MDI-induced Adipogenesis in the first 48 hours of Differentiation, a critical time window for Anti-Adipogenic effects. Coumestrol treatment suppressed MDI-induced protein expression of PPARγ and C/EBPα in Adipocytes, leading to the subsequent downregulation of FAS and aP2 expression. Akt and GSK3β were phosphorylated shortly after MDI stimulation, and these responses were Inhibited by coumestrol treatment.

Coumestrol also increased LRP6 protein expression, resulting in the recovery of β-catenin downregulation by MDI, while attenuating MDI-induced downregulation of Wnt10b. In addition, mRNA and protein expression of c-Myc and cyclin D1, target genes of β-catenin, were both recovered by coumestrol treatment. These results suggest that coumestrol Inhibits Adipocyte Differentiation via regulation of Akt and Wnt/β-catenin signaling and may have potential for development as an agent to Prevent Adipogenesis.



Cranberries (Oxycoccus quadripetalus)


Cranberries (Oxycoccus quadripetalus) Inhibit Adipogenesis and lipogenesis in 3T3-L1 cells


Cranberries (Oxycoccus quadripetalus) are a valuable source of bioactive substances with high antioxidant potential and well documented beneficial health properties. In the present study, the activity of cranberries, in terms of the Inhibiting effects of Adipogenesis, was investigated using the 3T3-L1 cell line. The obtained results showed that cranberries Reduced proliferation and viability of 3T3-L1 preadipocytes in a dose-dependent manner.
Treatment with cranberries decreased the number of Adipocytes and Reduced lipid accumulation in maturing 3T3-L1 preadipocytes, demonstrating an Inhibitory effect on lipogenesis. Moreover, it was found that cranberries directly induced Lipolysis in Adipocytes and down-regulated the expression of major transcription factors of the Adipogenesis pathway, such as PPARγ, C/EBPα and SREBP1. These findings indicate that cranberries are capable of suppressing Adipogenesis and therefore they seem to be natural bioactive factors effective in Adipose Tissue Mass modulation.


Crocin (saffron extract)


Obesity has become a severe public health problem worldwide. Crocin, a natural product, has been reported to have a number of pharmacological activities, including anti-inflammatory, anti-cancer, neuroprotective, antihypertensive, and cardioprotective action. The aims of the current study were to identify the beneficial effects of crocin on Obesity, Adipocyte Differentiation, and Lipolysis and to evaluate the possible role of AMPK. Results indicated that crocin significantly increased AMPK phosphorylation in differentiated Adipocytes in vitro and in Adipose Tissue in db/db mice. Crocin Reduced lipid accumulation in differentiated Adipocytes. In addition, crocin Inhibited the expression of mRNA of important Adipogenesis -related regulators, including CEBPα, CEBPβ, PPARγ, aP2, FAS, and CD36, in both differentiated Adipocytes and Adipose Tissue in db/db mice. Crocin increased the expression of mRNA of key Lipolysis -associated factors, including PPARα, LPL, and HSL, in both differentiated Adipocytes and Adipose Tissue in db/db mice.
In Adipocytes, knockdown of AMPK significantly suppressed the crocin-induced Inhibition of Adipocyte Differentiation and increase in Lipolysis. BML-275 is an Inhibit or of AMPK. In Adipose Tissue in db/db mice, BML-275 suppressed crocin-induced Inhibition of fat formation and alleviation of a metabolic disorder. The current results suggest that crocin alleviates Obesity in db/db mice and that it Inhibits Adipocyte Differentiation in preadipocytes. Crocin Inhibits Adipogenesis and promotes Lipolysis via activation of AMPK. Therefore, crocin may have promise as an option for the clinical treatment for Obesity and associated metabolic diseases.


Cryptotanshinone, a compound of Salvia miltiorrhiza


Cryptotanshinone, a compound of Salvia miltiorrhiza Inhibits Pre-Adipocytes Differentiation by regulation of Adipogenesis -related genes expression via STAT3 signaling


Background: Cryptotanshinone (CT), a major tanshinonefound in Salvia miltiorrhiza Bunge (Lamiaceae), has various pharmacological effects such as antitumor, anti-inflammatory, and antioxidant properties. Despite its well-documented benefits in a wide range of diseases, the effect of CT on Adipocyte Differentiation has not been well characterized.

Purpose: The present study was designed to determine the in vitro Anti-Adipogenic effect and underlying molecular mechanisms of CT using 3T3-L1 murine Pre-Adipocytes .

Methods: We measured the levels of intracellular triglyceride accumulation and mRNA and protein expression of key Adipogenic transcription factors and their target genes.

Results: Treatment with CT drastically Reduced lipid accumulation in a dose- and time-dependent manner. Molecular assays showed that CT effectively suppressed the expression of C/EBPβ, C/EBPα , and PPARγ and of their target Adipocyte -specific genes aP2, adiponectin, and GLUT4 but activated the expression of Anti-Adipogenic genes such as GATA2, CHOP10, and TNF-α. CT treatment also Inhibited the phosphorylation of STAT3 in the early phase of Adipogenesis . A small-interfering-RNA-mediated knock-down of STAT3 potentiated the Anti-Adipogenic effect of CT.

Conclusion: Taken together, the results suggest that CT may be a good Anti-Adipogenic candidate because it regulates STAT3 during early Adipogenesis .



Curcumin


Modification of Curcumin with Polyethylene Glycol Enhances the Delivery of Curcumin in preadipocytes and Its AntiAdipogenic Property


Conjugation of curcumin (CCM) by polyethylene glycol (PEG) has been previously developed to improve water solubility of the natural form of CCM and its antiproliferative role in some human cancer cell lines. This study examined the cellular uptake kinetics of the natural form of CCM and CCM−PEG. Their cytotoxic effect in proliferating preadipocytes and AntiAdipogenic property in differentiating preadipocytes had also been investigated. CCM and CCM−PEG were found to be differently absorbed in 3T3-L1 preadipocytes and Adipocytes with a limited amount of CCM−PEG absorption in the cell. The improved water solubility of CCM−PEG was correlated with increased cellular retention of CCM in 3T3-L1 cells, particularly in preadipocytes.

Consequently, CCM−PEG treatment sensitized proliferating preadipocytes to CCM-induced cell toxicity. Furthermore, incubation of differentiating 3T3-L1 cells with CCM−PEG resulted in improvement of the Inhibitory role of CCM in Adipocyte Differentiation with no toxic effect. These results suggest that pegylation-improved water solubility and cellular retention of CCM may be uniquely useful for improving the delivery of CCM in preadipocytes and its AntiAdipogenic ability.

Curcumin Inhibits Adipocyte Differentiation through modulation of mitotic clonal expansion


Adipocyte Differentiation is a key process in determining the number of mature Adipocytes in the development of Obesity. Here, we examined the function of curcumin, a dietary Polyphenol found in turmeric, and its underlying mechanisms in Adipocyte Differentiation. Our study reveals that curcumin exerts an Anti-Adipogenic function both in 3T3-L1 murine cells and in human primary preadipocytes as determined by intracellular lipid accumulation assay, quantitative analysis of Adipocyte marker gene expression and a noninvasive multimodal Coherent Anti-Stokes Raman Scattering (CARS) microscopic analysis of intracellular curcumin. The Inhibitory action of curcumin was largely limited to the early stage of Adipocyte Differentiation, where curcumin was found to Inhibit mitotic clonal expansion (MCE) process as evidenced by impaired proliferation, cell-cycle entry into S phase and the S to G2/M phase transition of confluent cells, and levels of cell cycle-regulating proteins with no significant effect on cell viability and cytotoxicity.

This, in turn, resulted in Inhibition of mRNA levels of early Adipogenic transcription factors, particularly Krüppel-like factor 5 (KLF5), CCAAT/enhancer binding proteinα (C/EBPα ) and peroxisome proliferator-activated receptorγ (PPARγ), in the early stage of Adipocyte Differentiation. Supplementation with rosiglitazone, a PPARγ ligand, during the early stage of Adipocyte Differentiation partially rescued curcumin-Inhibited Adipocyte Differentiation. Collectively, our results show that curcumin is an Anti-Adipogenic dietary bioactive component largely involved in the modulation of the MCE process during the early stage of Adipocyte Differentiation.



In summary, curcumin, the major Polyphenol found in turmeric, effectively Inhibits Preadipocyte Differentiation. Dephosphorylation of MAPKs may participate in the Anti-Adipogenic effects of curcumin. Curcumin also Inhibited the induction of Adipogenic transcription factors such as PPARγ and C/EBPα. This study is the first to demonstrate that the Wnt/β-catenin signaling pathway plays a crucial role in curcumin-induced suppression of Adipogenesis.


Curcumin Inhibits Adipogenesis in 3T3-L1 Adipocytes and angiogenesis and Obesity in C57/BL mice


Angiogenesis is necessary for the growth of Adipose Tissue. Dietary Polyphenols may suppress growth of Adipose Tissue through their antiangiogenic activity and by modulating Adipocyte metabolism. We investigated the effect of curcumin, the major Polyphenol in turmeric spice, on angiogenesis, Adipogenesis, Differentiation, apoptosis, and gene expression involved in lipid and energy metabolism in 3T3-L1 Adipocyte in cell culture systems and on body Weight Gain and Adiposity in mice fed a high-fat diet (22%) supplemented with 500 mg curcumin/kg diet for 12 wk. Curcumin (5–20 μmol/L) suppressed 3T3-L1 Differentiation, caused apoptosis, and Inhibited adipokine-induced angiogenesis of human umbilical vein endothelial cells.

Supplementing the high-fat diet of mice with curcumin did not affect food intake but Reduced body Weight Gain, Adiposity, and microvessel density in Adipose Tissue, which coincided with Reduced expression of vascular endothelial growth factor (VEGF) and its receptor VEGFR-2. Curcumin increased 5′AMP-activated protein kinase phosphorylation, Reduced glycerol-3-phosphate acyl transferase-1, and increased carnitine palmitoyltransferase-1 expression, which led to increased oxidation and decreased fatty acid esterification. The in vivo effect of curcumin on the expression of these enzymes was also confirmed by real-time RT-PCR in subcutaneous Adipose Tissue. In addition, curcumin significantly lowered serum cholesterol and expression of PPARγ and CCAAT/enhancer binding protein α, 2 key transcription factors in Adipogenesis and lipogenesis. The curcumin suppression of angiogenesis in Adipose Tissue together with its effect on lipid metabolism in Adipocytes may contribute to lower body fat and body Weight Gain. Our findings suggest that dietary curcumin may have a potential benefit in preventing Obesity.


Curcumin and Obesity


Turmeric has been long recognized for its Anti‐Inflammatory and health‐promoting properties. Curcumin is one of the principal Anti‐Inflammatory and healthful components of turmeric comprising 2–8% of most turmeric preparations. Experimental evidence supports the activity of curcumin in promoting weight loss and reducing the incidence of Obesity‐related diseases. With the discovery that Obesity is characterized by chronic low‐grade metabolic Inflammation, phytochemicals like curcumin which have Anti‐Inflammatory activity are being intensely investigated. Recent scientific research reveals that curcumin directly interacts with white Adipose Tissue to suppress chronic Inflammation. In Adipose Tissue, curcumin Inhibits macrophage infiltration and nuclear factor κB (NF‐κB) activation induced by inflammatory agents. Curcumin Reduces the expression of the potent pro inflammatory adipokines tumor necrosis factor‐α (TNFα), monocyte chemoattractant protein‐1 (MCP‐1), and plasminogen activator Inhibit or type‐1 (PAI‐1), and it induces the expression of adiponectin, the principal Anti‐Inflammatory agent secreted by Adipocytes . Curcumin also has effects to Inhibit Adipocyte Differentiation and to promote antioxidant activities. Through these diverse mechanisms curcumin Reduces Obesity and curtails the adverse health effects of Obesity.

The direct role of Wnt/β‐catenin signaling in the antiAdipogenic activity of curcumin was examined using mouse 3T3‐L1 Preadipocyte cells [72] … All of these activities driven by curcumin were associated with suppressed Adipogenesis in 3T3‐L1 cells.


Curcumin Inhibits 3T3-L1 Preadipocyte proliferation by mechanisms involving post-transcriptional p27 regulation


Inhibition of Differentiation resulted from suppression of PPARγ and C/EBPα, the dominant transcriptional regulators of Adipogenesis.  Further, data presented here elucidate an early antiAdipogenic role for curcumin via post-transcriptional regulation of p27.

Curcumin Inhibited early stages of Adipogenesis during cell cycle progression.

Curcumin increased p27 protein through post-translational mechanisms.

Curcumin suppressed Skp2 protein which directs p27 ubiquitylation.
p27 accumulation may account for late G1 arrest and suppression of Adipogenesis.

Potential mechanism regarding Anti-Obesity effects of curcumin.

Previous reports from our lab have shown that Skp2 is necessary for p27 degradation and cell cycle progression during Adipocyte Differentiation. Data presented here demonstrate that the anti-inflammatory, Anti-Obesity phytochemical curcumin blocked Skp2 protein accumulation during early Adipocyte hyperplasia. In addition, curcumin dose-dependently induced p27 protein accumulation andG1 arrest of synchronously replicating 3T3-L1 preadipocytes. Of note, p27 protein accumulation occurred in the presence of decreased p27 mRNA suggesting a role for post-transcriptional regulation. In support of this hypothesis, curcumin markedly increased p27 protein half-life as well as attenuated ubiquitin proteasome activity suggesting that Inhibition of targeted p27 proteolysis occurred through curcumin-mediated attenuation of Skp2 and 26S proteasome activity. While we observed no cytotoxic effects for curcumin at doses less than 20 µM, it is important to note an increase in apoptotic signaling at concentrations greater than 30 µM.

Finally, data presented here demonstrate that the anti-proliferative effect of curcumin was critical for the suppression of Adipocyte Differentiation and the development of the mature Adipocyte. Collectively, our data demonstrate that curcumin-mediated post-transcriptional accumulation of p27 accounts in part for the anti-proliferative effect observed in 3T3-L1 preadipocytes.



Curcumin-3,4-Dichloro Phenyl Pyrazole (CDPP)


Curcumin-3, 4-Dichloro Phenyl Pyrazole (CDPP) overcomes curcumin’s low bioavailability and Inhibits Adipogenesis


Adipocyte dysfunction, Obesity and associated metabolic disorders are of prime healthcare concern worldwide. Among available medications, natural products and inspired molecules hold 40% space in clinically prescribed medicines. In queue, this study overcomes the drawback of curcumin’s low bioavailability with potent Anti-Adipogenic and anti-dyslipidemic activity.

Methods: To evaluate the role of CDPP on Adipocyte Differentiation, 3T3-L1 Adipocytes were used as an in-vitro model. Flow cytometry was performed for cell cycle analysis. Syrian golden hamsters were used to study pharmacokinetic profile and dyslipidemic activity exhibited by CDPP.

Result: CDPP was found to be a potent Inhibit or of Adipogenesis in-vitro. It blocked mitotic clonal expansion by causing cell cycle arrest. CDPP showed marked improvement in gastrointestinal stability and bioavailability in-vivo as compared to curcumin. Administration of CDPP (100 mg/kg) significantly improved HFD induced dyslipidemic profile in hamsters and activated reverse cholesterol transportmachinery.

Conclusion: CDPP could be used as a potential drug candidate against Adipogenesis and dyslipidemia with enhanced gastrointestinal stability and bioavailability.



Cyanidine-3-O-Galactoside Enriched Aronia melanocarpa


Cyanidin-3-O-galactoside-enriched Aronia melanocarpa extract attenuates Weight Gain and Adipogenic pathways in high-fat diet-induced obese C57BL/6 mice


Aronia melanocarpa are a rich source of Anthocyanins that have received considerable interest for their relations to human health. In this study, the Anti-Adipogenic effect of cyanidin-3-O-galactoside-enriched Aronia melanocarpa extract (AM-Ex) and its underlying mechanisms were investigated in an in vivo system. Five-week-old male C57BL/6N mice were randomly divided into five groups for 8-week feeding with a control diet (CD), a high-fat diet (HFD), or a HFD with 50 (AM-Ex 50), 100 (AM-Ex 100), or 200 AM-Ex (AM-Ex 200) mg/kg body weight/day. HFD-fed mice showed a significant increase in body weight compared to the CD group, and AM-Ex dose-dependently Inhibited this Weight Gain. AM-Ex significantly Reduced the food intake and the weight of white fat tissue, including epididymal fat, retroperitoneal fat, mesenteric fat, and inguinal fat. Treatment with AM-Ex (50 to 200 mg/kg) Reduced serum levels of leptin, insulin, triglyceride, total cholesterol, and low density lipoprotein (LDL)-cholesterol.

Real-time reverse transcription-polymerase chain reaction (RT-PCR) analysis revealed that AM-Ex suppressed Adipogenesis by decreasing CCAAT/enhancer binding protein α, peroxisome proliferator-activated receptor γ, sterol regulatory element-binding protein-1c, peroxisome proliferator-activated receptor gamma coactivator-1α, acetyl-CoA carboxylase 1, ATP-citrate lyase, fatty acid synthase, and Adipocyte protein 2 messenger RNA (mRNA) expressions. These results suggest that AM-Ex is potentially beneficial for the suppression of HFD-induced Obesity by modulating multiple pathways associated with Adipogenesis and food intake.


Cyanidine-3-O-Galactoside Enriched Aronia melanocarpa Extract Inhibits Adipogenesis and Lipogenesis via Down-Regulation of Adipogenic Transcription Factors and Their Target Genes in 3T3-L1 cells


Our results showed that the Anti-Obesity effect of AM-Ex occured through down-regulation of the transcription factors PPARγ, C/EBPα, SREBP-1c, and Adipogenesis and lipogenesis-related genes, aP2, LPL, ACC1, ACL, and FAS. These results demonstrate that AM-Ex can be used as a preventive or therapeutic agent for Obesity. Future, animal and human studies are needed to further investigate the mechanism and proper concentration of AM to be used as Anti-Obesity agents.



Cyclopia subternata


Cyclopia maculata and Cyclopia subternata (honeybush tea) Inhibits Adipogenesis in 3T3-L1 Pre-Adipocytes


The stems, leaves and flowers of Cyclopia have been consumed as aherbal tea ‘honeybush tea’ to treat various medical ailments since the 19th century. Plant Polyphenols are reported to Inhibit Adipogenesis in cell and animal models of Obesity. The aim of this study was to assess the effect of hot water extracts of two Cyclopia species, C. maculata and C. subternata on Obesity in an in vitro model. The total Polyphenol content of unfermented C. subternata, unfermented C. maculata and fermented C. maculata extracts was 25.6, 22.4 and 10.8 g GAE/100 g, respectively.

The major compounds present in the extracts were: the flavonoid, phloretin-3′,5′-di-C-glucoside in C. subternata, the xanthone, mangiferin in unfermented C. maculata and the flavanone, hesperidin in fermented C. maculata. All of the plant extracts Inhibited intracellular triglyceride and Fat Accumulation, and decreased PPARγ 2 expression. The higher concentrations of unfermented C. maculata (800 and 1600 μg/ml) and C. subternata(1600 μg/ml) were cytotoxic in terms of decreased mitochondrialdehydrogenase activity. Both fermented and unfermented C. maculata, at concentrations greater than 100 μg/ml, decreased cellular ATPcontent. Cyclopia maculata and C. subternata Inhibit Adipogenesis in vitro, suggesting their potential as Anti-Obesity agents.


Cyclopia maculata (honeybush tea) stimulates Lipolysis in 3T3-L1 Adipocytes


We have previously, for the first time, demonstrated that hot water extracts of Cyclopia maculata and Cyclopia subternata, endemic South African plants that are consumed as herbal teas, Inhibit Adipogenesis in 3T3-L1 Adipocytes. The aim of this study was to extend the Anti-Obesity investigations of these plants by quantifying Lipolysis in mature 3T3-L1 Adipocytes. Glycerol concentration in culturesupernatants was used as a marker of Adipocyte LipolysisIsoproterenol, a β-adrenergic agonist and a known lipolytic agent, was used as a positive control in our assays. Lipolysis was stimulated by all extracts, although statistical significance was noted for fermented (oxidised) C. maculata only. A concentration of 80 μg/ml ofC. maculata extract induced maximal Lipolysis (1.8-fold, p < 0.001).

The increased Lipolysis was accompanied by an increase in the expression of hormone sensitive lipase (1.6-fold, p < 0.05) and perilipin (1.6-fold,p < 0.05). The plant extracts, at the concentration range assayed (0–100 μg/ml), were not cytotoxic in terms of mitochondrialdehydrogenase and adenosine-5′-triphosphate activity. These results showed that C. maculata stimulates Lipolysis in mature 3T3-L1 Adipocytes, providing further support for the Anti-Obesity effects ofCyclopia spp.


A Polyphenol -enriched fraction of Cyclopia intermedia decreases lipid content in 3T3-L1 Adipocytes and Reduce s body Weight Gain of obese db/db mice


Extracts of Cyclopia species, indigenous South African fynbos plants used for the production of honeybush tea, have potential as Anti-Obesity nutraceuticalingredients. Previously, we demonstrated that aqueous extracts of C. maculata and C. subternata exhibited Anti-Obesity effects in 3T3-L1 Adipocytes. In this study, we further explored these Anti-Obesity effects of C. maculata and C. subternata as well as C. intermedia for the first time. Extracts were prepared using a 40% methanol–water mixture (40% MeOH) in order to enhance the Polyphenol ic content of the extracts.

Moreover, these extracts were separated into aqueous and organic fractions using liquid–liquid partitioning with n-butanol and water to further enrich the Polyphenol content of the organic fractions. Extracts of all three Cyclopia species decreased the lipid content in 3T3-L1 Adipocytes, although differences in bioactivity of their aqueous and organic fractions were observed. The organic fraction of C. intermedia was further investigated. This fraction dose-dependently decreased the lipid content in 3T3-L1 Adipocytes without affecting cell viability, while increasing mRNA expression of HSL (1.57-fold, P < 0.05), SIRT1 (1.5-fold, P = 0.07), UCP3 (1.5-fold, P < 0.05) and PPARγ (1.29-fold, P < 0.05). Daily treatment of obese db/db mice with 351.5 mg/kg bodyweight of the organic C. intermedia fraction for 28 days decreased body Weight Gain by 21% (P < 0.05) without any effect on food or water consumption.

The organic fraction was enriched in phenolic content relative to the extract with neoponcirin, a flavanone not previously identified in Cyclopia species, mangiferin, isomangiferin and hesperidin comprising 17.37% of the organic fraction of C. intermedia compared to 4.96% of its “large scale” prepared 40% MeOH extract. Their specific roles as Anti-Obesity agents in these models needs to be studied to guide product development.


Polyphenols have a range of health promoting effects against chronic diseases such as Obesity and type 2 Diabetes mellitus. Previous research in our group showed that Cyclopia species (honeybush), endemic South African plants, display Anti-Obesity effects. The aim of this study was to investigate the Anti-Obesity potential of Polyphenol-enriched extracts of three Cyclopia spp. (C. subternata, C. intermedia and C. maculata) using bioactivity guided fractionation to facilitate the identification of Anti-Obesity Polyphenols. Aqueous methanol extracts of C. subternata, C. intermedia and C. maculata were prepared and separated into their aqueous and organic fractions, which were tested for their Anti-Obesity effects in 3T3-L1 Adipocytes using the Oil Red O, glycerol release (marker of Lipolysis), and triglyceride assays. Cytotoxicity was evaluated using the 3-[4,5-Dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) and adenosine triphosphate (ATP) assays. The Anti-Obesity properties of the organic fraction of the aqueous methanol extract of C. intermedia were evaluated in obese Leprdb/db mice. Thereafter, the organic fraction was separated into four fractions (CCC F1 to CCC F4) using high performance counter-current chromatography (HPCCC), which were tested for their Anti-Obesity and cytotoxic effects in 3T3-L1 Pre-Adipocytes and in mature Adipocytes. Liquid chromatography tandem Mass spectrometry (LC-MS/MS) and quantitative high performance liquid chromatography diode array detection (HPLC-DAD) were used for determination of phenolic composition.
The molecular mechanisms of action of the organic fraction and its CCC fractions were assessed with quantitative real-time PCR and western blot analysis. The Polyphenol-enriched extracts of C. subternata, C. intermedia and C. maculata, and their aqueous and organic fractions exhibited differences in phenolic composition and bioactivity. The aqueous fractions of C. maculata and C. subternata, containing lower phenolic content than the organic fractions, decreased lipid content in 3T3-L1 Adipocytes compared to their organic counterparts. However, the organic fraction of C. intermedia, with higher phenolic content than its aqueous fraction, decreased lipid content compared to its aqueous fraction in 3T3-L1 Adipocytes. In Leprdb/db mice, the organic fraction of C. intermedia decreased body Weight Gain, without affecting food or water consumption.
Further fractionation of the organic fraction of C. intermedia by HPCCC provided four fractions with differences in phenolic composition and Anti-Obesity effects. None of the extracts or fractions, except for the aqueous C. intermedia and organic C. maculata fractions which decreased ATP content, affected cell viability as measured with the MTT and ATP assays during acute treatment. The highest concentrations of CCC F1 and CCC F3 decreased ATP content during chronic treatment in Pre-Adipocytes . In 3T3-L1 Adipocytes, the organic fraction of C. intermedia and CCC fractions modulated the expression of genes implicated in lipid and energy metabolism, whereas the organic fraction of C. intermedia had no effect on the expression of these genes in Leprdb/db mice. In conclusion, the organic fraction of C. intermedia exhibited Anti-Obesity properties in vitro and in vivo. A flavanone with Anti-Obesity potential, neoponcirin, was identified in C. intermedia, the first discovery of this compound in Cyclopia spp. Bioactivity guided fractionation of the organic fraction of C. intermedia resulted in CCC fractions retaining different Polyphenols with varying Anti-Obesity effects.
These results demonstrate that the Anti-Obesity potential of C. intermedia is due to the presence of more than one compound, with different mechanisms of action, or the synergistic effects of more than one compound, may contribute to the bioactivity of C. intermedia.


Delphinidin (Consolida ajacis (L.) Schur)

Delphinidin-3-Oβ-glucoside (D3G) is a health-promoting anthocyanin whose Anti-Obesity activity has not yet been thoroughly investigated. We examined the effects of D3G on Adipogenesis and lipogenesis in 3T3-L1 Adipocytes and primary white Adipocytes using real-time RT-PCR and immunoblot analysis. D3G significantly Inhibited the accumulation of lipids in a dose-dependent manner without displaying cytotoxicity. In the 3T3-L1 Adipocytes, D3G downregulated the expression of key Adipogenic and lipogenic markers, which are known as peroxisome proliferator-activated receptor gamma (PPARγ), sterol regulatory element-binding transcription factor 1 (SREBP1), CCAAT/enhancer-binding protein alpha (C/EBPα ), and fatty acid synthase (FAS).

Moreover, the relative protein expression of silent mating type information regulation 2 homolog 1 (SIRT1) and carnitine palmitoyltransferase-1 (CPT-1) were increased, alongside Reduced lipid levels and the presence of several small lipid droplets. Furthermore, D3G increased the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC), which suggests that D3G may play a role in AMPK and ACC activation in Adipocytes. Our data indicate that D3G attenuates Adipogenesis and promotes lipid metabolism by activating AMPK-mediated signaling, and, hence, could have a therapeutic role in the management and treatment of Obesity.



Delphinidin-3-O-β-Glucoside


Anti-Adipogenic effects of delphinidin3Oβglucoside in 3T3-L1 preadipocytes and primary white Adipocytes


Abstract Delphinidin3Oβglucoside (D3G) is a health-promoting anthocyanin whose antiObesity activity has not yet been thoroughly investigated. We examined the effects of D3G on Adipogenesis and lipogenesis in 3T3-L1 Adipocytes and primary white Adipocytes.



DHEA


Dehydroepiandrosterone (DHEA) treatment in vitro Inhibits Adipogenesis in human omental but not subcutaneous Adipose Tissue


Dehydroepiandrosterone (DHEA), a precursor sex steroid, circulates in sulphated form (DHEAS). Serum DHEAS concentrations are inversely correlated with Metabolic Syndrome components and in vivo/in vitro studies suggest a role in modulating adipose Mass. To investigate further, we assessed the in vitro biological effect of DHEA in white (3T3-L1 ) and brown (PAZ6) Preadipocyte cell lines and human primary preadipocytes.

DHEA (from 10−8 M) caused concentration-dependent proliferation Inhibition of 3T3-L1 and PAZ6 preadipocytes. Cell cycle analysis demonstrated unaltered apoptosis but indicated blockade at G1/S or G2/M in 3T3-L1 and PAZ6, respectively. Preadipocyte cell-line Adipogenesis was not affected.

In human primary subcutaneous and omental preadipocytes, DHEA significantly Inhibited proliferation from 10−8 M. DHEA 10−7 M had opposing effects on Adipogenesis in the two fat depots. Subcutaneous Preadipocyte Differentiation was unaffected or increased whereas omental preadipocytes showed significantly Reduced Adipogenesis.

We conclude that DHEA exerts fat depot-specific differences which modulate body composition by limiting omental fat production.



Diallyl trisulphide (onion)


AntiObesity effects of quercetin-rich onion peel extract on the Differentiation of 3T3-L1 preadipocytes and the Adipogenesis in high fat-fed rats


The aim of the present study was to examine the effect of quercetin-rich onion peel extract (OPE) on anti-Differentiation in 3T3-L1 preadipocytes and the AntiObesity in high-fat fed rats. We found that lipid accumulations and TG contents in 3T3-L1 cells were markedly suppressed by OPE. The mRNA levels of activating protein (AP2) were down-regulated and those of carnitine palmitoyl transferase-1 α (CPT-1α) and fatty acid binding protein 4 (FABP4) were up-regulated by 75 and 100 μg/ml OPE. Body weight, retroperitoneal and mesenteric fat weights of SD rats were significantly lower in the 8 week high fat (HF) diet + 0.72% OPE group than in the HF group.

Peroxisome proliferator-activated receptor (PPAR)γ mRNA levels were down-regulated in the epididymal fat of OPE than those of control and HF, and significant down-regulation of CCAAT/enhancer binding protein (C/EBP)α mRNA levels in OPE was also observed than the control. The mRNA levels of CPT-1α and uncoupling protein-1 (UCP-1) were up-regulated by the OPE, while those of fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC) were down-regulated in HF and OPE groups compared to control group. These results suggest that quercentin-enriched OPE may have AntiObesity effects by suppressing Preadipocyte Differentiation and Inhibiting Adipogenesis.



Dioscin (DS) a steroidal saponin (Dioscorea japonica Thunb.)


Dioscin Inhibits Adipogenesis through the AMPK/MAPK pathway in 3T3-L1 cells and modulates Fat Accumulation in obese mice


Dioscin (DS) is a steroidal saponin present in a number of medicinal plants and has been shown to exert anticancer, antifungal and antiviral effects. The present study aimed to deternube the effects DS on the regulation of Adipogenesis and to elucidate the underlying mechanisms. In vitro experiments were performed using differentiating 3T3-L1 cells treated with various concentrations (0-4 µM) of DS for 6 days. A cell viability assay was performed on differentiating cells following exposure to DS. Oil Red O staining and triglyceride content assay were performed to evaluate the lipid accumulation in the cells. We also carried out the following experiments: i) flow cytometry for cell cycle analysis, ii) quantitative reverse transcription polymerase chain reaction for measuring Adipogenesis-related gene expression, and iii) western blot analysis to measure the expression of Adipogenesis transcription factors and AMP-activated protein kinase (AMPK), acetyl-CoA carboxylase (ACC) and mitogen-activated protein kinase (MAPK) phosphorylation.

In vivo experiements were performed using mice with Obesity induced by a high-fat diet (HFD) that were treated with or without DS for 7 weeks. DS suppressed lipid accumulation in the 3T3-L1 cells without affecting viability at a dose of up to 4 µM. It also delayed cell cycle progression 48 h after the initiation of Adipogenesis. DS Inhibited Adipocyte Differentiation by the downregulation of Adipogenic transcription factors and attenuated the expression of Adipogenesis-associated genes. In addition, it enhanced the phosphorylation of AMPK and its target molecule, ACC, during the Differentiation of the cells. Moreover, the Inhibition of Adipogenesis by DS was mediated through the suppression of the phosphorylation of MAPKs, such as extracellular-regulated kinase 1/2 (ERK1/2) and p38, but not c-Jun-N-terminal kinase (JNK).

DS significantly Reduced Weight Gain in the mice with HFD-induced Obesity; this was evident by the suppression of Fat Accumulation in the abdomen. the present study reveals an Anti-Adipogenic effect of DS in vitro and in vivo and highlights AMPK/MAPK signaling as targets for DS during Adipogenesis.



Djulis (Chenopodium formosanum)


The Inhibitory Effects of Djulis (Chenopodium formosanum) and Its Bioactive Compounds on Adipogenesis in 3T3-L1 Adipocytes


The aim of this study was to provide new insights into the role of the ethanolic extracts of Djulis (Chenopodium formosanum, EECF) and its bioactive compounds in preventing Adipogenesis in 3T3-L1 Adipocytes. The results demonstrated EECF significantly Inhibited oil red O-stained material (OROSM), triglyceride levels and glycerol-3-phosphate dehydrogenase (GPDH) activity in 3T3-L1 Adipocytes. The expression of the critical molecules involved in lipid synthesis such as PPARγ, C/EBPα and SREBP-1c was attenuated in EECF-treated cells. According to HPLC-DAD and HPLC-MS/MS analysis, rutin, kaempferol, betanin and another nine compounds were present in EECF. The suppression of lipid accumulation by rutin, kaempferol and betanin occurred by decreasing the gene expression of PPARγ, C/EBPα and SREBP-1c. Taken together, these findings suggest the presence of bioactive compounds in EECF may partly account for the anti-Adipogenesis of EECF and EECF is therefore a potentially lipid lowering functional food.



Dolichos lablab L. Seeds


AntiObesity activities of chikusetsusaponin IVa and Dolichos lablab L. seeds


Obesity, a condition where excess body fat accumulates to the extent, causes a negative effect on health. Previously, we reported the extract of Dolichos lablab L. (DLL-Ex) Inhibited high-fat diet (HFD)-induced increases in body weight and body fat Mass and ameliorated increases in body weight. In the present work, we studyed the molecular mechanism for the Inhibitory effect of DLL-Ex or Chikusetsusaponin IVa (CS-IVa), as isolated from Dolichos lablab L. (DLL) seeds extract, on Adipocyte Differentiation . We evaluated the effect of DLL-Ex, an Anti-Obesity agent, and CS-IVa, an active component of DLL-Ex, on 3T3-L1 cell Differentiation via Oil red O assay and Q-PCR, along with their effects on CCAAT element binding protein alpha (C/EBPα ), peroxisome proliferator-activated receptor gamma (PPARγ), fatty acid synthase (FAS), and fatty acid-binding protein 4 (FABP4) mRNA transcriptions. FAS and FABP4 protein expression levels after exposure to CS-IVa were also tested. The results showed that DLL-Ex and CS-IVa have potent Inhibitory activity on Adipocyte Differentiation. Therefore, DLL and CS-IVa may be developed as a functional food material to treat Obesity.



Ecklonia Cava


Dieckol, a phlorotannin isolated from a brown seaweed, Ecklonia cava, Inhibits Adipogenesis through AMP-activated protein kinase (AMPK) activation in 3T3-L1


•The Ecklonia cava extract tested herein evidenced profound Adipogenesis Inhibition.

•The three Polyphenol compounds of phlorotannins were isolated from E. cava.

•Dieckol exhibited greatest potential Adipogenesis Inhibition.

•Dieckol significantly Reduced the expression of Adipogenic-specific genes and proteins.

•Dieckol Inhibits Adipogenesis by activating the AMPK pathway.

In this study, we assessed the potential Inhibitory effect of 5 species of brown seaweeds on Adipogenesis the Differentiation of 3T3-L1 preadipocytes into mature Adipocytes by measuring Oil-Red O staining. The Ecklonia cava extract tested herein evidenced profound Adipogenesis Inhibitory effect, compared to that exhibited by the other four brown seaweed extracts. Thus, E. cava was selected for isolation of active compounds and finally the three Polyphenol compounds of phlorotannins were obtained and their Inhibitory effect on Adipogenesis was observed. Among the phlorotannins, dieckol exhibited greatest potential Adipogenesis Inhibition and down-regulated the expression of peroxisome proliferator-activated receptor-γ (PPARγ), CCAAT/enhancer-binding proteins (C/EBPα), sterol regulatory element-binding protein 1 (SREBP1) and fatty acid binding protein 4 (FABP4) in a dose-dependent manner.

The specific mechanism mediating the effects of dieckol was confirmed by AMP-activated protein kinase (AMPK) activation. These results demonstrate Inhibitory effect of dieckol compound on Adipogenesis through the activation of the AMPK signal pathway.


Anti-Adipogenic effect of dioxinodehydroeckol via AMPK activation in 3T3-L1 Adipocytes


Dioxinodehydroeckol (DHE) isolated from Ecklonia cava, has previously been investigated for its Inhibition of the Differentiation of 3T3-L1 preadipocytes into Adipocytes. Levels of lipid accumulation were measured, along with changes in the expression of genes and proteins associated with Adipogenesis and Lipolysis. Confluent 3T3-L1 preadipocytes in medium with or without different concentrations of DHE for 7 days were differentiated into Adipocytes. Lipid accumulation was quantified by measuring direct triglyceride contents and Oil-Red O staining. The expression of genes and proteins associated with Adipogenesis and Lipolysis was measured using RT-PCR, quantitative real-time RT-PCR and Western blotting analysis. It was found that the presence of DHE significantly Reduced lipid accumulation and down-regulated the expression of peroxisome proliferator-activated receptor-γ (PPARγ), sterol regulatory element-binding protein 1 (SREBP1) and CCAAT/enhancer-binding proteins (C/EBPα) in a dose-dependent manner.
Moreover, DHE suppressed regulation of the Adipocyte-specific gene promoters such as fatty acid binding protein (FABP4), fatty acid transport protein (FATP1), fatty acid synthase (FAS), lipoprotein lipase (LPL), acyl-CoA synthetase 1 (ACS1), leptin, perilipin and HSL compared to control Adipocytes. The specific mechanism mediating the effects of DHE was confirmed by activation of phosphorylated AMP-activated protein kinase (pAMPK). Therefore, these results suggest that DHE exerts Anti-Adipogenic effect on Adipocyte Differentiation through the activation and modulation of the AMPK signaling pathway.

Anti-hyperlipidemic effect of Polyphenol extract (Seapolynol™) and dieckol isolated from Ecklonia cava in in vivo and in vitro models


The Inhibitory effect of Polyphenol extracts (Seapolynol, SPN) of the marine brown algae Ecklonia cavaand dieckol, a major component of SPN, on hyperlipidemia was investigated in ICR mice fed a high-fat diet (HFD) for five weeks. For analysis of the anti-hyperlipidemic effects of SPN and dieckol, these two agents were given orally on a daily basis to HFD-fed mice for four weeks, starting one week after the beginning of HFD feeding. Groups administered with SPN as well as dieckol showed lower body Weight Gains than the HFD only group. Administration of SPN and dieckol also resulted in a significant reduction of the level of total cholesterol (TCHO), triglyceride (TG), and low-density lipoprotein (LDL) cholesterol in the serum of HFD-fed mice. In Oil Red O staining using 3T3-L1 preadipocytes, it was shown that both SPN and dieckol markedly Inhibited lipid accumulation of 3T3-L1 cells. Furthermore, SPN and dieckol (50 μg/mL) significantly Inhibited 3-hydroxyl-methyl glutaryl coenzyme A (HMGCoA) reductase activity in vitro.
Taken together, these results suggest that Polyphenols of Ecklonia cava (SPN) and dieckol Reduce body Weight Gain and Fat Accumulation in HFD-induced obese mice, and that their hypolipidemic effect is related to the Inhibition of Adipogenesis of Adipocytes and HMGCoA reductase activity.

Enzyme-treated Ecklonia cava extract Inhibits Adipogenesis through the downregulation of C/EBPα in 3T3-L1 Adipocytes


In this study, we examined the Inhibitory effects of enzyme- treated Ecklonia cava (EEc) extract on the Adipogenesis of 3T3-L1 Adipocytes. The components of Ecklonia cava (E. cava) were first separated and purified using the digestive enzymes pectinase (Rapidase® X‑Press L) and cellulase (Rohament® CL). We found that the EEc extract contained three distinct phlorotannins: eckol, dieckol and phlorofucofuroeckol-A. Among the phlorotannins, dieckol was the most abundant in the EEc extract at 16 mg/g. Then we examined the Inhibitory effects of EEc extract treatment on Differentiation‑related transcription factors and on Adipogenesis‑related gene expression in vitro using 3T3-L1 Adipocytes. 3T3‑L1 pre‑Adipocytes were used to determine the concentrations of the EEc extract and Garcinia cambogia (Gar) extract that did not result in cytotoxicity. Glucose utilization and triglyceride (TG) accumulation in the EEc‑treated Adipocytes were similarly Inhibited by 50 µg/ml EEc and 200 µg/ml Gar, and these results were confirmed by Oil Red O staining.

Protein expression of Adipogenesis Differentiation‑related transcription factors following treatment with the EEc extract was also examined. Only the expression of CCAAT/enhancer‑binding protein (C/EBP)α was decreased, while there was no effect on the expression of C/EBPβ, C/EBPδ, and peroxisome proliferator‑activated receptor γ (PPARγ). Treatment with the EEc extract decreased the expression levels of Adipogenesis‑related genes, in particular sterol regulatory element binding protein‑1c (SREBP‑1c), Adipocyte fatty acid binding protein (A‑FABP), fatty acid synthase (FAS) and adiponectin. These results suggest that EEc extract treatment has an Inhibitory effect on Adipogenesis, specifically by affecting the activation of the C/EBPα signaling pathway and the resulting Adipogenesis-related gene expression.


Edible Brown Alga Ecklonia cava Derived Phlorotannin‐Induced AntiAdipogenic Activity in Vitro


The present study performed the bioactivity‐monitored isolation of anti‐Adipogenic phlorotannins from E cklonia cava as a part of the ongoing research to develop AntiObesity nutraceuticals from natural origin. Extracts were partitioned into n‐hexane, 85% aqueous MeOH , n‐BuOH and water fractions. Their Adipogenic activities were examined by measuring glycerol release level and Adipogenic‐related gene expression in differentiating 3T 3‐L 1 preadipocytes. Among them, n‐BuOH fractions significantly increased glycerol secretion and Reduced the regulation of Adipogenic transcription factors, peroxisome proliferator‐activated receptor‐γ (PPAR γ) and CCAAT /enhancer‐binding protein α (C/EBP α), as well as tumor necrosis factor α (TNF α). Further separation from n‐BuOH fraction led to the isolation of phlorotannins, triphlorethol‐A, eckol and dieckol.

Phlorotannins increased the glycerol secretion and Reduced the glucose consumption levels of 3T 3‐L 1 Adipocytes. Moreover, the phlorotannins Reduced the expression levels of PPAR γ, C /EBP α and Differentiation‐dependent factor 1/sterol regulatory element‐binding protein 1c, as well as downstream genes such as fatty acid binding protein‐4, fatty acid transport protein‐1, fatty acid synthase, L eptin and acyl‐CoA synthetase 1. In addition, phlorotannins increased the mRNA expression of hormone‐sensitive lipase while suppressing perilipin and TNF α expressions.


Evaluation of Inhibitory effect of phlorotannins from Ecklonia cava on triglyceride accumulation in Adipocyte


In the present study, a methanolic extract of Ecklonia cava and its solvent-partitioned fractions were evaluated for their AntiAdipogenic effect in 3T3-L1 Adipocytes. One of them, the n-BuOH fraction, effectively Reduced lipid accumulation and glucose consumption. In addition, the presence of the n-BuOH fraction in Adipocytes suppressed the regulations of Adipogenic transcription factors, PPARγ and SREBP1c, and Adipogenic specific genes, FABP4, FABP1, FAS, LPL, HSL, and ACS1. Further purification of n-BuOH fractions led to the isolation of six phlorotannins. The six phlorotannins effectively suppressed triglyceride accumulation. Comparative analysis showed that lipid accumulation in Adipocytes was dramatically attenuated in the presence of eckstolonol.

Phlorotannins isolated from the edible brown alga Ecklonia stolonifera exert Anti-Adipogenic activity on 3T3-L1 Adipocytes by downregulating C/EBPα and PPARγ


The dramatic increase in Obesity-related diseases emphasizes the need to elucidate the cellular and molecular mechanisms underlying fat metabolism. Inhibition of Adipocyte Differentiation has been suggested to be an important strategy for preventing or treating Obesity. In our previous study, we characterized an Ecklonia stolonifera extract and non-polar fractions thereof, including dichloromethane and ethyl acetate fractions. We showed that these fractions Inhibited Adipocyte Differentiation and lipid formation/accumulation in 3T3-L1 preadipocytes, as assessed by Oil Red O staining. As part of our ongoing search for Anti-Obesity agents derived from E. stolonifera, in this work, we characterized five known phlorotannins, including phloroglucinol, eckol, dieckol, dioxinodehydroeckol, and phlorofucofuroeckol A, all of which were isolated from the active ethyl acetate fraction of E. stolonifera.
We determined the chemical structures of these phlorotannins through comparisons of published nuclear magnetic resonance (NMR) spectral data. Furthermore, we screened these phlorotannins for their abilities to Inhibit Adipogenesis over a range of concentrations (12.5–100 μM). Of these five phlorotannins, phloroglucinol, eckol, and phlorofucofuroeckol A significantly concentration-dependently Inhibited lipid accumulation in 3T3-L1 cells without affecting cell viability. In addition, the five isolated phlorotannins also significantly Reduced the expression levels of several Adipocyte marker genes, including proliferator activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα), although they did so to different extents.
These results suggest that the molecular weight of a phlorotannin is an important factor affecting its ability to Inhibit Adipocyte Differentiation and modulate the expression levels of Adipocyte marker genes.

Seapolynol extracted from Ecklonia cava Inhibits Adipocyte Differentiation in vitro and decreases Fat Accumulation in vivo


Our study showed that SN Inhibited lipid accumulation in 3T3-L1 cells and high-fat-diet-fed zebrafish and mice. Furthermore, we examined the underlying mechanisms of the SN-induced AntiAdipogenic effect including Inhibition of MCE via cell cycle arrest, and activation of AMPKα signaling. Ecklonia cava has been shown to Inhibit Adipogenesis in cell lines. Several Polyphenols (dieckol, phloroeckol, dioxinodehydroeckol) derived from Ecklonia cava were shown to have AntiAdipogenic effects in Adipocytes. A clinical study on Ecklonia cava Polyphenols was also performed in overweight subjects. In particular, the most abundant Ecklonia cava Polyphenol, dieckol, has been identified to have AntiAdipogenic effects in cells and animal models. SN, a standardized extract of Ecklonia cava, includes active compounds including the Polyphenols mentioned above, and is commercially available; SN contains dieckol, eckol, and bieckol derivatives. Recent studies have shown the biological effects of this extract. Ecklonia cava extract or Ecklonia cava-derived Polyphenols have been reported to have antioxidant effects. Kang et al., showed the potential of Ecklonia cava Polyphenols as chemopreventive agents [25], and the antioxidant and anti-inflammatory effects of Ecklonia cava in osteoarthritis were evaluated. Ecklonia cava extract was shown to have antibacterial effects on antibiotic-resistant food-borne pathogens. Additionally, Heo et al., showed protective effects of Ecklonia cavaphlorotannins against photo-oxidative stress from UV-B radiation.

Shibata et al., reported the Inhibitory effect of Ecklonia cava phlorotannins on pro-inflammatory responses. In particular, an AntiAdipogenic effect of SN has been reported recently. However, the mechanisms underlying the AntiAdipogenic effects of SN should be investigated. The current study is the first to show the AntiAdipogenic effect of SN in two animal models with early Adipogenic and signaling mechanisms, in which zebrafish was used to show the effects of SN in early Adipogenesis. Lipid accumulation in zebrafish is known to be detectable from 15 dpf. Zebrafish were grown for 17–20 dpf to show early Fat Accumulation by Nile-red staining; SN Inhibited Fat Accumulation in zebrafish from an early stage.

In addition, the diet induced obese mice has been used here were 5 weeks old, fed on HFD for 10 weeks. The HFD mice caused a marked increase the lipid accumulation in the liver or TG and total cholesterol in the blood compared to control diet mice, while SN dramatically repressed the lipid accumulation in the liver or TG and total cholesterol in the blood. In addition, SN significantly decreased HFD-induced abnormal expression of Adipogenic and fatty liver associated genes. On the other hand, the high levels of glucose and insulin concentration are generally occurring in HFD-induced obese mice [30]. However, we did not observed the sign of hyperglycemia and hyperinsulinemia in HFD-induced obese mice compared to the control diet group in this study.
The afore-mentioned evidence prompt us to speculate that some of the factors such as mouse strain, ages, diet composition or duration of HFD may affect to develop the intermediate stage Obesity between Obesity and type 2 Diabetes. This model may occur the some of genetic changes in the tissues due to the HFD consumption in mice, but it has not yet appeared the hyperglycemia and hyperinsulinemia. However, additional animal study should be analyzed in further studies.
Our data showed that SN induced Inhibition in early Adipogenesis via regulating early Adipogenic factors, such as KLF2, KLF4, and C/EBPβ. SN-mediated Inhibition of early Adipogenesis was associated with the Inhibition of MCE via cell cycle arrest, and the Inhibition of ERK and Akt signaling was underlined as a reason for the SN-induced Inhibition of early Adipogenesis. Recent studies have shown that several phytochemicals induce Inhibition of MCE via cell cycle arrest as one of the major mechanisms of an AntiAdipogenic effect in 3T3-L1 cells. In particular, our data showed that SN activates AMPKα, a sensor in energy metabolism, and ACC, a major enzyme in fatty acid synthesis, in both Adipocytes and a mouse model. Such AMPKα activation was also shown previously in a study of dieckol. Accordingly, the SN-induced activation of AMPKα is likely due to dieckol, a major component of SN. However, the other compounds in SN should also be investigated. The SN-mediated repression of ACC could contribute to the suppression of TG accumulation in Adipocytes and mice models. Our Inhibit or study showed SN-induced repression of ACC was mediated via activation of AMPKα signaling.
Our study showed that SN supplementation effectively improved the plasma lipid status of mice fed HFD, as was seen with dieckol. However, SN was superior to dieckol in terms of the suppression of lipid accumulation and cholesterol levels. SN decreased triglyceride levels by 40%—while dieckol Reduced them by only ~10%—in mice fed, an HFD. LDL cholesterol levels were Reduced by 66% in SN-supplemented HFD-fed mice, while dieckol supplementation showed a 54% reduction in LDL cholesterol [32]. This result was supported by the study of Yeo et al., which showed SN is more effective than dieckol on Inhibition of lipid accumulation and weight increase. Given that dieckol comprises 8% of SN, other SN-derived compounds likely contribute to the difference in effects on lipid accumulation and cholesterol levels between SN and dieckol. SN showed significant effects in decreasing triglyceride and cholesterol levels in HFD-fed mice.
The reductions in triglycerides and lipid accumulation by SN are supported by its suppression of Adipogenic factors. However, the SN-induced reduction of (LDL) cholesterol level was not fully addressed in this study. In particular, downstream targets of AMPKα include various factors—including HMGCoA and SREBP1, which are responsible for cholesterol metabolism. Although SREBF-1 was shown to be regulated by SN in zebrafish, other cholesterol-related factors should be examined. AMPKα also regulates carbohydrate metabolism via factors such as ChREBP, but glucose and insulin levels were unaffected by SN, suggesting that SN does not greatly affect carbohydrate metabolism.
AMPKα is known to be associated with factors related to the energy expenditure, such as the NAD-dependent protein deacetylase sirtuin 1 (SIRT1) and peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α). These signaling processes are known to activate thermogenic responses, such as heat production with upregulation of uncoupling proteins. A recent study showed that a phytochemical, indole-3-cabinol, directly activated SIRT1 via binding in 3T3-L1 cells [34]. Thus, the roles of SN and SN-derived compounds should be investigated in thermogenic signaling in future studies.
We also used the zebrafish model, and showed SN-mediated Inhibition in early Adipogenesis and lipid accumulation. Zebrafish provides various advantages for in vivoresearch but also has several limitations. Full sets of antibodies for lipid metabolism studies have not yet been established. Further studies should include data for the AntiAdipogenic effects of SN in protein levels. Finally, our data demonstrated that SN effectively suppressed HFD-induced lipid accumulation in cell line, zebrafish, and mouse models.
Ecklonia cava contains various Polyphenols, known as phlorotannins. Phloroeckol, dioxinodehydroeckol, and dieckol have been shown to have AntiAdipogenic activities in Adipocytes and animals, and dieckol, a major compound of SN, could give an important contribution to AntiAdipogenic effects of SN. However, the effect may be derived from the synergistic or combinational effects of constituents in SN. Since other Ecklonia cava-derived Polyphenols, which may also have AntiAdipogenic effects, have yet to be fully investigated, further study should be performed on SN-derived compounds, additionally, comparative and combinational studies on SN-derived compounds would be executed in the future.


Ecliptal, isolated from Eclipta alba


Ecliptal, a promising natural lead isolated from Eclipta alba modulates Adipocyte function and Ameliorates Metabolic Syndrome


A swift increase has been observed in the number of individuals with metabolic syndrome worldwide. A number of natural compounds have been identified towards combating metabolic syndrome. Adding to this premise, here we report the pleiotropic activities of Ecliptal (EC); a natural compound isolated from the herb Eclipta alba. Administration of EC was shown to have prominent anti-adipogenic effects in 3T3-L1 and hMSC derived adipocytes. It was shown to activate Wnt-pathway and alter AKT signaling. Additionally, it caused cell cycle arrest and inhibited mitotic clonal expansion. EC treatment augmented mitochondrial biogenesis as well as function as estimated by expression of PGC1α, UCP-1, mitochondrial complexes and estimation of oxygen consumption rate. EC also reduced LPS-induced inflammation and tunicamycin induced ER stress.

Further, EC enhanced insulin sensitivity by increasing AKT phosphorylation, inhibiting PKCα/βII phosphorylation and reducing leptin/adiponectin ratio. Finally, EC administration in Syrian golden hamsters was shown to have potent anti-dyslipidemic effects. Cumulatively, encompassing pleiotropic activities of EC, it could prove to be a potential drug candidate against obesity, insulin resistance and related metabolic syndrome.


Ethyl acetate fraction of Eclipta alba: a potential phytopharmaceutical targeting Adipocyte Differentiation


Natural products have always fascinated mankind for their miraculous properties. Eclipta alba (E. alba), a medicinal herb has long been used in traditional medicine for curing several pathologies. It has been shown to have anti-diabetic effect as well as hepato-protective activity. Here, in order to address metabolic derangements, the study was designed to evaluate the efficacy of E. alba and its fractions in adipogenesis inhibition and dys-lipidemia. Of the crude extract and fractions screened, ethyl acetate fraction of E. alba inhibited adipocyte differentiation in 3T3-L1 pre-adipocytes and hMSC derived adipocytes.
It inhibited mitotic clonal expansion and caused cell cycle arrest in G1 and S phase as suggested by western blot analysis and flow cytometry. It was also shown to have lipolytic effects. Oral administration of ethyl acetate fraction of E. alba to hamsters unveiled its anti-adipogenic as well as anti-dyslipidemic activity in-vivo. Mass spectrometry analysis of ethyl acetate fraction confirmed the presence of several bioactive components, projecting it as an effective phytopharmaceutical agent. In conclusion, ethyl acetate fraction of E. alba possesses potent anti-adipogenic as well as anti-dyslipidemic activity and could be projected as an herbal formulation towards obesity.

Ethnopharmacological significance of Eclipta alba (L.) hassk.(Asteraceae)


Eclipta alba can be found growing wild in fallow lands of Bangladesh where it is considered as a weed by farmers. Traditional medicinal systems of the Indian subcontinent countries as well as tribal practitioners consider the plant to have diverse medicinal values and use it commonly for treatment of gastrointestinal disorders, respiratory tract disorders (including asthma), fever, hair loss and graying of hair, liver disorders (including jaundice), skin disorders, spleen enlargement, and cuts and wounds. The plant has several phytoconstituents like wedelolactone, eclalbasaponins, ursolic acid, oleanolic acid, luteolin, and apigenin.
Pharmacological activities of plant extracts and individual phytoconstituents have revealed anticancer, hepatoprotective, snake venom neutralizing, anti-inflammatory, and antimicrobial properties. Phytoconstituents like wedelolactone and ursolic and oleanolic acids as well as luteolin and apigenin can form the basis of new drugs against cancer, arthritis, gastrointestinal disorders, skin diseases, and liver disorders.


Egcg


The Anti-Adipogenic effects of (-) epigallocatechin gallate are dependent on the WNT/β-catenin pathway


(-)Epigallocatechin gallate (EGCG) is the most abundant catechin in green tea and reportedly has anti-obesity and anti-adipogenic effects. In this study, we determined that the up-regulation of the WNT/β-catenin pathway is the anti-adipogenic mechanisms of EGCG in 3T3-L1 cells. EGCG treatment down-regulates the expression of major genes involved in the adipogenesis pathway including peroxisome proliferator-activated receptor (PPAR)γ, CCAAT/enhancer binding protein (C/EBP)α, fatty acid binding protein (FABP)4 and fatty acid synthase (FASN), while up-regulating the nuclear level of β-catenin. Knockdown of β-catenin using small interfering (si) RNA attenuated the inhibitory effects of EGCG on intracellular lipid accumulation. β-catenin siRNA transfection also recovered terminal adipocyte markers such as FABP4, FASN, lipoprotein lipase and adiponectin, which were down-regulated by EGCG. The DNA binding activities as well as the expression levels of PPARγ and C/EBPα, which were down-regulated by EGCG, were significantly restored by β-catenin siRNA transfection. In addition, we found that EGCG efficiently up-regulates the WNT/β-catenin pathway. Among the members of the WNT/β-catenin pathway, the expressions of low density lipoprotein receptor-related protein (LRP)5, LRP6, disheveled (DVL)2 and DVL3 were significantly up-regulated, while AXIN expression was down-regulated by EGCG, and the phosphorylation of glycogen synthase kinase 3β was increased. These results suggest that EGCG activates the WNT/β-catenin pathway, resulting in the up-regulation of β-catenin, which down-regulates the major genes of the adipogenesis pathway. Taken together, our findings clearly show that the anti-adipogenic effects of EGCG are, at least partially, dependent on the WNT/β-catenin pathway.


Genistein, EGCG, and capsaicin Inhibit Adipocyte Differentiation process via activating AMP-activated protein kinase


Phytochemicals such as soy isoflavone genistein have been reported to possess therapeutic effects for obesity, diabetes, and cardiovascular diseases. In the present study, the molecular basis of selective phytochemicals with emphasis on their ability to control intracellular signaling cascades of AMP-activated kinase (AMPK) responsible for the inhibition of adipogenesis was investigated. Recently, the evolutionarily conserved serine/threonine kinase, AMPK, emerges as a possible target molecule of anti-obesity. Hypothalamic AMPK was found to integrate nutritional and hormonal signals modulating feeding behavior and energy expenditure.

We have investigated the effects of genistein, EGCG, and capsaicin on adipocyte differentiation in relation to AMPK activation in 3T3-L1 cells. Genistein (20-200muM) significantly inhibited the process of adipocyte differentiation and led to apoptosis of mature adipocytes. Genistein, EGCG, and capsaicin stimulated the intracellular ROS release, which activated AMPK rapidly. We suggest that AMPK is a novel and critical component of both inhibition of adipocyte differentiation and apoptosis of mature adipocytes by genistein or EGCG or capsaicin further implying AMPK as a prime target of obesity control.


Comparison of cytotoxicity and the Anti-Adipogenic effect of green tea Polyphenols with epigallocatechin-3-gallate in 3T3-L1 preadipocytes


Recent studies have demonstrated the effects of green tea polyphenols (GTP) and epigallocatechin-3-gallate (EGCG) on obesity. However, high doses of EGCG have also exhibited cytotoxicity. The aim of this study was to compare total GTP with purified EGCG on cytotoxicity, and to investigate the effects and the molecular mechanism of total GTP and EGCG on adipogenesis. Cytotoxicity was determined by cell viability assay. For the adipogenesis study, 3T3-L1 preadipocytes were incubated with three doses of GTP (1, 10, and 100 μg/ml) and the effect of EGCG (6.8 μg/ml) was compared with 10 μg/ml GTP containing 68% EGCG. Oil Red O staining and triglyceride content assay were carried out 10 days after differentiation and treatment. Adipogenic regulators CCAAT element binding protein α (C/EBPα), peroxisome proliferator-activated receptor gamma (PPARγ) and sterol regulatory element-binding protein-1c (SREBP-1c) were determined by qRT-PCR and immunoblotting. GTP at 1000 μg/ml and EGCG (68 and 680 μg/ml) significantly affected cell viability.
Purified EGCG had greater cytotoxicity than corresponding doses of GTP. About 10 μg/ml of GTP showed stronger reduction in triglyceride accumulation than EGCG treatment. Transcriptional factors of C/EBPα, SREBP-1c and PPARγ were markedly decreased in both GTP and EGCG-treated cells and GTP exhibited stronger inhibitory effects on C/EBPα and PPARγ protein expression than EGCG (p < 0.05). In conclusion, total GTP exerted greater inhibitory effects than purified EGCG on adipogenesis through down-regulating the adipogenic factor C/EBPα, SREBP-1c and PPARγ expression. These findings support that a polyphenol mixture is safer and more effective than EGCG alone for preventing obesity and obesity-related chronic diseases.

Epigallocatechin-3-gallate Inhibits Adipogenesis through down-regulation of PPARγ and FAS expression mediated by PI3K-AKT signaling in 3T3-L1 cells


Epigallocatechin-3-gallate (EGCG), a major component in green tea, functions as extensive bioactivities including anti-inflammation, anti-oxidation, and anti-cancer. However, little is known about its anti-adipogenesis and underlying mechanisms. The purport of this study sought to investigate effects of EGCG on 3T3-L1 preadipocyte differentiation and to explore its possible mechanisms. The 3T3-L1 cells were induced to differentiate under the condition of pro-adipogenic cocktail with or without indicated EGCG concentrations (10, 50, 100, 200µM) for 2, 4, 6 and 8 days, respectively. Also, another batch of 3T3-L1 cells was induced under the optimal EGCG concentration (100µM) with or without SC3036 (PI3K activator, 10µM) or SC79 (AKT activator, 0.5µM) for 8 days.

Subsequently, the cell viability was examined by MTT assay and the cell morphology was visualized by Oil red O staining. Finally, the mRNA levels including peroxisome proliferator activated receptor γ (PPARγ) and fatty acid synthase (FAS) were detected by quantitative real time PCR, while the protein levels of PPARγ, FAS, phosphatidylinositol 3 kinase (PI3K), insulin receptor substrate1(IRS1), AKT, and p-AKT were measured by immunoblotting analysis. Our results showed that EGCG inhibited adipogenesis of 3T3-L1 preadipocyte in a concentration-dependent manner. Moreover, the inhibitory effects were reversed by SC3036 or SC79, suggesting that the inhibitory effects of EGCG are mediated by PI3K-AKT signaling to down-regulate PPARγ and FAS expression levels. The findings shed light on EGCG anti-adipogenic effects and its underlying mechanism and provide a novel preventive-therapeutic potential for obesity subjects as a compound from Chinese green tea.

Keywords: Adipocyte; Adipogenesis; Epigallocatechin-3-gallate; Peroxisome proliferator-activated receptor gamma; Phosphatidylinositol 3-kinase.


AntiObesity effect of EGCG and glucosamine-6-phosphate through decreased expression of genes related to Adipogenesis and cell cycle arrest in 3T3-L1


Purpose: Several studies have proven that EGCG, the primary green tea catechin, and glucosamine-6-phosphate (PGlc) reduce triglyceride contents in 3T3-L1 adipocytes. The objective of this study is to evaluate the combination effect of EGCG and PGlc on decline of accumulated fat in differentiated 3T3-L1 adipocytes.
Methods: EGCG and PGlc were administered for 6 day for differentiation of 3T3-L1 adipocytes. Cell viability was measured using the CCK assay kit. In addition, TG accumulation in culture 3T3-L1 adipocytes was investigated by Oil Red O staining. We examined the expression level of several genes and proteins associated with adipogenesis and lipolysis using real-time RT-PCR and Western blot analysis. A flow cytometer Calibar was used to assess the effect of EGCG and PGluco on cell-cycle progression of differentiating 3T3-L1 cells.
Results: Intracelluar lipid accumulation was significantly decreased by combination treatment with EGCG 60 μM and PGlc 200 μg/m compared with control and EGCG treatment alone. In addition, use of combination treatment resulted in directly decreased expression of PPARγ, C/EBPα, and SREBP1. In addition, it inhibited adipocyte differentiation and adipogenesis through downstream regulation of adipogenic target genes such as FAS, ACSL1, and LPL, and the inhibitory action of EGCG and PGlc was found to inhibit the mitotic clonal expansion (MCE) process as evidenced by impaired cell cycle entry into S phase and the S to G2/M phase transition of confluent cells and levels of cell cycle regulating proteins such as cyclin A and CDK2.
Conclusion: Combination treatment of EGCG and PGlc inhibited adipocyte differentiation through decreased expression of genes related to adipogenesis and adipogenic and cell cycle arrest in early stage of adipocyte differentiation.


Ellagic Acid (Rhus chinensis Mill.)

Fatty acid synthase (FAS) has been recognized as a potential therapeutic target for Obesity. In this study, for the first time, the Inhibitory effect of pomegranate husk extract, punicalagin and ellagic acid on FAS was investigated. We found them potently Inhibiting the activity of FAS with half-Inhibitory concentration values (IC50) of 4.1 μg/ml (pomegranate husk extract), 4.2 μg/ml (4.50 μM, punicalagin) and 1.31 μg/ml (4.34 μM, ellagic acid), respectively. Moreover, they all exhibited time-dependent inactivation of FAS.

Punicalagin and ellagic acid Inhibited FAS with different mechanisms compared to previously reported Inhibitors, through inactivating acetyl/malonyl transferase and β-ketoacyl synthase domains, respectively. Additionally, 100 μg/ml pomegranate husk extract, 5.24 μg/ml (5 μM) punicalagin and 4.5 μg/ml (15 μM) ellagic acid effectively Reduced lipid accumulation inside FAS over-expressed 3T3-L1 Adipocytes. Since FAS plays a key role in the biosynthesis pathway of fatty acid, these findings suggest that pomegranate husk extract, punicalagin and ellagic acid have potential in the prevention and treatment of Obesity.



Epiberberine (Berberis thunbergii DC)

It has been reported that alkaloids derived from Coptis chinensis exert Anti-Adipogenic activity on 3T3-L1 Adipocytes by downregulating peroxisome proliferation-activity receptor-γ (PPAR-γ) and CCAAT/enhancer binding protein-α (C/EBP-α). However, the signaling-based mechanism of the Inhibitory role of epiberberine in the early stages of 3T3-L1 Adipocyte Differentiation is uncharacterized. Here, we show that epiberberine had Inhibitory effects on Adipocyte Differentiation and significantly decreased lipid accumulation by downregulating an Adipocyte -specific transcription factor, sterol regulatory element-binding protein-1 (SREBP-1).

Furthermore, we observed that epiberberine markedly suppressed the Differentiation -mediated phosphorylation of components of both the Raf/mitogen-activated protein kinase 1 (MEK1)/extracellular signal-regulated protein kinase 1/2 (ERK1/2) and AMP-activated protein kinase-α1 (AMPKα)/Akt pathways. In addition, gene expression of fatty acid synthase (FAS) was significantly Inhibited by treatment with epiberberine during Adipogenesis . These results indicate that the Anti-Adipogenic mechanism of epiberberine is associated with Inhibition of phosphorylation of Raf/MEK1/ERK1/2 and AMPKα/Akt, followed by downregulation of the major transcription factors of Adipogenesis , such as PPAR-γ, C/EBP-α, and SREBP-1, and FAS. Taken together, this study suggests that the Anti-Adipogenic effect of epiberberine is mediated by downregulation of the Raf/MEK1/ERK1/2 and AMPKα/Akt pathways during 3T3-L1 Adipocyte Differentiation . Moreover, the Anti-Adipogenic effects of epiberberine were not accompanied by modulation of β-catenin.



Erigeron annuus (L.) Pers.

Obesity is one of the major public health problems in the world because it is implicated in Metabolic Syndrome s, such as type 2 Diabetes , hypertension, and cardiovascular diseases. The objective of this study was to investigate whether Erigeron annuus (L.) Pers. (EAP) extract Suppresses reactive oxygen species (ROS) production and Fat Accumulation in 3T3-L1 cells by activating an AMP-dependent kinase (AMPK) signaling pathway. Our results showed that EAP water extract significantly Inhibits ROS production, Adipogenesis , and lipogenesis during Differentiation of 3T3-L1 preadipocytes . In addition, EAP decreased mRNA and protein levels of proliferator-activated receptor γ (PPARγ ) and CCAAT/enhancer-binding protein alpha (C/EBPα ). Moreover, EAP suppressed mRNA expressions of fatty acid synthase (FAS), lipoprotein lipase (LPL), Adipocyte protein 2 (aP2) in a dose-dependent manner. Whereas, EAP upregulated adiponectin expression, phosphorylation levels of AMPK and carnitine palmitoyltransferase 1 (CPT-1) protein level during Differentiation of 3T3-L1 preadipocytes.

These results suggest that EAP water extract can exert ROS-linked Anti-Obesity effect through the mechanism that might involve Inhibition of ROS production, Adipogenesis and lipogenesis via an activating AMPK signaling pathway.



Esculetin


Esculetin Induces Apoptosis and Inhibits Adipogenesis in 3T3‐L1 Cells


Objective: To determine the effects of esculetin, a plant phenolic compound with apoptotic activity in cancer cells, on 3T3‐L1 Adipocyte apoptosis and Adipogenesis .

Research Methods and Procedures: 3T3‐L1 pre‐confluent preadipocytes and lipid‐filled Adipocytes were incubated with esculetin (0 to 800 μM) for up to 48 hours. Viability was determined using the Cell Titer 96 Aqueous One Solution cell proliferation assay; apoptosis was quantified by measurement of single‐stranded DNA. Post‐confluent preadipocytes were incubated with esculetin for up to 6 days during maturation. Adipogenesis was quantified by measuring lipid content using Nile Red dye; cells were also stained with Oil Red O for visual confirmation of effects on lipid accumulation.

Results: In mature Adipocytes , esculetin caused a time‐ and dose‐related increase in Adipocyte apoptosis and a decrease in viability. Apoptosis was increased after only 6 hours by 400 and 800 μM esculetin (p < 0.05), and after 48 hours, as little as 50 μM esculetin increased apoptosis (p < 0.05). In preadipocytes , apoptosis was detectable only after 48 hours (p < 0.05) with 200 μM esculetin and higher concentrations. However, results of the cell viability assay indicated a reduction in Preadipocyte number in a time‐ and dose‐related manner, beginning as early as 6 hours with 400 and 800 μM esculetin (p < 0.05). Esculetin also Inhibited Adipogenesis of 3T3‐L1 preadipocytes . Esculetin‐mediated Inhibition of Adipocyte Differentiation occurred during the early, intermediate, and late stages of the Differentiation process. In addition, esculetin induced apoptosis during the late stage of Differentiation .

Discussion: These findings suggest that esculetin can alter fat cell number by direct effects on cell viability, Adipogenesis, and apoptosis in 3T3‐L1 cells.



Euphorbia lunulata


Syzygium aqueum leaf extract and its bioactive compounds enhances pre-Adipocyte Differentiation and 2-NBDG uptake in 3T3-L1 cells


The insulin-like and/or insulin-sensitising effects of Syzygium aqueum leaf extract and its six bioactive compounds; 4-hydroxybenzaldehyde, myricetin-3-O-rhamnoside, europetin-3-O-rhamnoside, phloretin, myrigalone-G and myrigalone-B were investigated in 3T3-L1 Adipocytes. We observed that, S. aqueum leaf extract (0.04–5 μg/ml) and its six bioactive compounds (0.08–10 μM) at non-cytotoxic concentrations were effectively enhance Adipogenesis, stimulate glucose uptake and increase adiponectin secretion in 3T3-L1 Adipocytes. Clearly, the compounds myricetin-3-O-rhamnoside and europetin-3-O-rhamnoside showed insulin-like and insulin-sensitising effects on Adipocytes from a concentration of 0.08 μM.

These compounds were far better than rosiglitazone and the other isolated compounds in enhancing Adipogenesis, stimulating 2-NBDG uptake and increasing adiponectin secretion at all the concentrations tested. These suggest the AntiDiabetic potential of S. aqueum leaf extract and its six bioactive compounds. However, further molecular interaction studies to explain the mechanisms of action are highly warranted.



Evodiamine (Evodia rutaecarpa)


Evodiamine Inhibits Adipogenesis via the EGFR–PKCα–ERK signaling pathway


The molecular mechanism of the Anti-Adipogenic effect of evodiamine (which has several capsaicin-like pharmacological actions) was investigated. The evodiamine effect was not blocked by the specific TRPV1 antagonist capsazepine in 3T3-L1 preadipocytes , whereas its effect was greatly curtailed by Inhibitors of protein kinase C (PKC) and epidermal growth factor receptor (EGFR). Signal analyses showed that evodiamine stimulated the phosphorylation of EGFR, PKCα, and ERK, all of which were Reduced by an Inhibitors EGFR Inhibitor.

Silencing experiments of EGFR mRNA supported the involvement of these signaling molecules in the Inhibitory effect of evodiamine. An unidentified mechanism whereby evodiamine Inhibits Adipogenesis via the EGFR–PKCα–ERK signaling pathway was revealed.



Ferulic Acid (Ferula assafoetida L)


Aqueous extracts of hulled barley containing coumaric acid and ferulic acid Inhibit Adipogenesis in vitro and Obesity in vivo


World epidemic Obesity is a major contributing factor to metabolic diseases including insulin resistance and cardiovascular diseases. In this study, aqueous and ethanolic extracts of hulled barley with roasting temperatures of up to 250 °C were investigated for their Anti-Adipogenic effects in vitro and in vivo. An aqueous extract of hulled barley roasted at 210 °C (AHB210) effectively Inhibited Adipocyte Differentiation. Intraperitoneal injections of 15 or 50 mg/kg AHB210 dose dependently prevented body Weight Gain, fat Mass increase, and dysregulated lipid profiles in high fat diet-induced obese male mice. In addition, oral administration of AHB210 to ovariectomized rats also prevented body Weight Gain.

A high performance liquid chromatographic analysis identified coumaric acid and ferulic acid as primary Anti-Obesity mediators. The presence of beta glucan in AHB210 was less likely to be responsible for the lipid accumulating actions. Taken together, AHB210 may be useful to Prevent Obesity and its related metabolic diseases.



Ficus deltoidea var. deltoidea


Anti-Adipogenic effects of extracts of Ficus deltoidea var. deltoidea and var. angustifolia on 3T3-L1 Adipocytes


Objective: This study examined the Anti-Adipogenic effects of extracts of Ficus deltoidea var. deltoidia and var. angustifolia, a natural slimming aid, on 3T3-L1 Adipocytes.

Methods: Methanol and water extracts of leaves of the F. deltoidea varieties were analyzed to determine their total flavonoid content (TFC) and total phenolic content (TPC), respectively. The study was initiated by determining the maximum non-toxic dose (MNTD) of the methanol and water extracts for 3T3-L1 preadipocytes. Possible Anti-Adipogenic effects were then examined by treating 2-d post confluent 3T3-L1 preadipocytes with either methanol extract or water extract at MNTD and half MNTD (½MNTD), after which the preadipocytces were induced to form mature Adipocytes. Visualisation and quantification of lipid content in mature Adipocytes were carried out through oil red O staining and measurement of optical density (OD) at 520 nm, respectively.

Results: The TFCs of the methanol extracts were 1.36 and 1.97 g quercetin equivalents (QE)/100 g dry weight (DW), while the TPCs of the water extracts were 5.61 and 2.73 g gallic acid equivalents (GAE)/100 g DW for var. deltoidea and var. angustilofia, respectively. The MNTDs determined for methanol and water extracts were (300.0±28.3) and (225.0±21.2) µg/ml, respectively, for var. deltoidea, while much lower MNTDs [(60.0±2.0) µg/ml for methanol extracts and (8.0±1.0) µg/ml for water extracts] were recorded for var. angustifolia. Studies revealed that the methanol extracts of both varieties and the water extracts of var. angustifolia at either MNTD or ½MNTD significantly Inhibited the maturation of preadipocytes.

Conclusions: The Inhibition of the formation of mature Adipocytes indicated that leaf extracts of F. deltoidea could have potential Anti-Obesity effects.



Fisetin (Rhus succedanea L)


Inhibition of mitotic clonal expansion mediates fisetin-exerted prevention of Adipocyte Differentiation in 3T3-L1 cells


Adipocytes are the key player in Adipose Tissue Inflammation and subsequent systemic insulin resistance and its development involves complex process of proliferation and Differentiation of preadipocytes. Fistein, a Polyphenol flavonoid, is known to exert anti-inflammatory, anti-carcinogenic and anti-Diabetic effects. In this study, we aimed to investigate the effect of fisetin on Adipocyte proliferation and Differentiation in 3T3-L1 Preadipocyte cell line and its mechanism of action. We found that fisetin Inhibits Adipocyte Differentiation in a concentration dependent manner, which were evidenced by Oil Red O staining and the protein expression of mature Adipocyte marker genes fatty acid synthase and peroxisome proliferator-activated receptor γ.

Moreover, the proliferation of preadipocytes was also markedly suppressed by treatment of fisetin for 24 and 48 h in the Differentiation medium. We also found that fisetin Inhibition of Adipocyte Differentiation was largely due to the effect on mitotic clonal expansion. Fisetin suppression of Preadipocyte proliferation at early stage of Differentiation was accompanied by the changes of expression of a series of cell cycle regulatory proteins. Altogether, our results suggest that the Inhibition of Adipocyte Differentiation by fisetin may be at least in part mediated by cell cycle arrest during Adipogenesis.



Foenumoside B from Lysimachia foenum-graecum


Foenumoside B from Lysimachia foenumgraecum Inhibits Adipocyte Differentiation and Obesity induced by high-fat diet


We have previously reported anti-obesity effects of Lysimachia foenum-graecum in high-fat diet (HFD)-induced obesity model. Here we isolated a triterpene saponin foenumoside B as an active component of L. foenum-graecum. Foenumoside B blocked the differentiation of 3T3-L1 preadipocytes in a dose-dependent manner with an IC50 of 0.2 μg/ml in adipogenesis assay and suppressed the induction of PPARγ, the master regulator of adipogenesis. Foenumoside B induced the activation of AMP-activated protein kinase (AMPK), and modulated the expression of genes involved in lipid metabolism towards lipid breakdown in differentiated adipocytes. In mouse model, oral administration of foenumoside B (10mg/kg/day for 6 weeks) reduced HFD-induced body weight gain significantly without affecting food intake. Treatment of foenumoside B suppressed lipid accumulation in white adipose tissues and the liver, and lowered blood levels of glucose, triglycerides, ALT, and AST in HFD-induced obese mice.

Consistent with the in vitro results, foenumoside B activated AMPK signaling, suppressed the expression of lipogenic genes, and enhanced the expression of lipolytic genes in vivo. Foenumoside B also blocked HFD-induced proinflammatory cytokine production in adipose tissue, suggesting its protective role against insulin resistance. Taken together, these findings demonstrate that foenumoside B represents the anti-obesity effects of L. foenum-graecum, and suggest therapeutic potential of foenumoside B in obesity and obesity-related metabolic diseases.


Suppression of Adipocyte Differentiation by Foenumoside B from Lysimachia foenumgraecum Is Mediated by PPARγ Antagonism


Lysimachia foenum-graecum extract (LFE) and its active component foenumoside B (FSB) have been shown to inhibit adipocyte differentiation, but their mechanisms were poorly defined. Here, we investigated the molecular mechanisms responsible for their anti-adipogenic effects. Both LFE and FSB inhibited the differentiation of 3T3-L1 preadipocytes induced by peroxisome proliferator-activated receptor-γ (PPARγ) agonists, accompanied by reductions in the expressions of the lipogenic genes aP2, CD36, and FAS. Moreover, LFE and FSB inhibited PPARγ transactivation activity with IC50s of 22.5 μg/ml and 7.63 μg/ml, respectively, and showed selectivity against PPARα and PPARδ. Rosiglitazone-induced interaction between PPARγ ligand binding domain (LBD) and coactivator SRC-1 was blocked by LFE or FSB, whereas reduced NCoR-1 binding to PPARγ by rosiglitazone was reversed in the presence of LFE or FSB.

In vivo administration of LFE into either ob/ob mice or KKAy mice reduced body weights, and levels of PPARγ and C/EBPα in fat tissues. Furthermore, insulin resistance was ameliorated by LFE treatment, with reduced adipose tissue inflammation and hepatic steatosis. Thus, LFE and FSB were found to act as PPARγ antagonists that improve insulin sensitivity and metabolic profiles. We propose that LFE and its active component FSB offer a new therapeutic strategy for metabolic disorders including obesity and insulin resistance.


Foenumoside B isolated from Lysimachia foenumgraecum extract Suppresses LPS-induced inflammatory response via NF-κB/AP-1 inactivation in murine


Foenumoside B (FSB), a bioactive component isolated from the Lysimachia foenum-graecum extract (LFE), has been shown to possess anti-inflammatory effects, but the underlying molecular mechanisms involved have not been elucidated. Accordingly, the authors investigated the mechanisms responsible for the anti-inflammatory effects of FSB in murine macrophages activated by LPS. FSB suppressed the LPS-induced expressions of iNOS and COX-2 at protein and mRNA levels and consequently decreased NO and PGE2 production in RAW264.7 and primary macrophages. FSB also reduced the LPS-induced inductions of TNF-α, IL-6 and IL-1β at protein and mRNA levels.

Studies of the molecular mechanisms involved in the anti-inflammatory effects of FSB showed that it inhibited the transcriptional activities of NF-κB and AP-1, and the nuclear translocation of NF-κB via inhibition of the phosphorylations of AKT, p38 and STAT3. In a sepsis model, pretreatment with FSB inhibited the LPS-stimulated mRNA and protein levels of proinflammatory mediators, such as, iNOS, COX-2, TNF-α, IL-6 and IL-1β in plasma and liver. Importantly, FSB increased the survival rate of mice in the LPS-induced sepsis model. Taken together, these results show that the anti-inflammatory effects of FSB against LPS-induced inflammatory conditions are associated with inhibitions of the phosphorylations of AKT, p38 and STAT3 followed by the transcriptional suppressions of NF-κB and AP-1, and thus, reduced expressions of pro-inflammatory genes.



Formononetin (Astragalus membranaceus)

Formononetin, an isoflavone, activates AMP-activated protein kinase/β-catenin signalling to Inhibit Adipogenesis and rescues C57BL/6 mice from high-fat diet-induced Obesity and bone loss


Balance between Adipocyte and osteoblast Differentiation is the key link of disease progression in Obesity and osteoporosis. We have previously reported that formononetin (FNT), an isoflavone extracted from Butea monosperma, stimulates osteoblast formation and protects against postmenopausal bone loss. The inverse relationship between osteoblasts and Adipocytes prompted us to analyse the effect of FNT on Adipogenesis and in vivo bone loss, triggered by high-fat diet (HFD)-induced Obesity. The Anti-Obesity effect and mechanism of action of FNT was determined in 3T3-L1 cells and HFD-induced obese male mice. Our findings show that FNT Suppresses the Adipogenic Differentiation of 3T3-L1 fibroblasts, through down-regulation of key Adipogenic markers such as PPARγ, CCAAT/enhancer-binding protein alpha (C/EBPα) and sterol regulatory element-binding protein (SREBP) and Inhibits intracellular TAG accumulation. Increased intracellular reactive oxygen species levels and AMP-activated protein kinase (AMPK) activation accompanied by stabilisation of β-catenin were attributed to the Anti-Adipogenic action of FNT. In vivo, 12 weeks of FNT treatment Inhibited the development of Obesity in mice by attenuating HFD-induced body Weight Gain and visceral Fat Accumulation. The Anti-Obesity effect of FNT results from increased energy expenditure.

FNT also protects against HFD-induced dyslipidaemia and rescues deterioration of trabecular bone volume by increasing bone formation and decreasing bone resorbtion caused by HFD. FNT’s rescuing action against Obesity-induced osteoporosis commenced at the level of progenitors, as bone marrow progenitor cells, obtained from the HFD mice group supplemented with FNT, showed increased osteogenic and decreased Adipogenic potentials. Our findings suggest that FNT Inhibits Adipogenesis through AMPK/β-catenin signal transduction pathways and protects against HFD-induced Obesity and bone loss.



fucoxanthinol


Fucoxanthin and its metabolite, fucoxanthinol, suppress Adipocyte Differentiation in 3T3-L1 cells


Fucoxanthin is a major carotenoid found in edible seaweed such as Undaria pinnatifida and Hijikia fusiformis. We investigated the suppressive effects of fucoxanthin and its metabolite, fucoxanthinol, on the differentiation of 3T3-L1 preadipocytes to adipocytes. Fucoxanthin inhibited intercellular lipid accumulation during adipocyte differentiation of 3T3-L1 cells. Furthermore, fucoxanthin was converted to fucoxanthinol in 3T3-L1 cells. Fucoxanthinol also exhibited suppressive effects on lipid accumulation and decreased glycerol-3-phosphate dehydrogenase activity, an indicator of adipocyte differentiation.

The suppressive effect of fucoxanthinol was stronger than that of fucoxanthin. In addition, in 3T3-L1 cells treated with fucoxanthin and fucoxanthinol, peroxisome proliferator-activated receptor gamma (PPARgamma), which regulates adipogenic gene expression, was down-regulated in a dose-dependent manner. These results suggest that fucoxanthin and fucoxanthinol inhibit the adipocyte differentiation of 3T3-L1 cells through down-regulation of PPARgamma. Fucoxanthinol had stronger suppressive effects than fucoxanthin on adipocyte differentiation in 3T3-L1 cells.


Simultaneous determination of fucoxanthin and its deacetylated metabolite fucoxanthinol in rat plasma by liquid chromatography-tandem Mass spectrometry


Fucoxanthin and its deacetylated metabolite fucoxanthinol are two major carotenoids that have been confirmed to possess various pharmacological properties. In the present study, fucoxanthinol was identified as the deacetylated metabolite of fucoxanthin, after intravenous (i.v.) and intragastric gavage (i.g.) administration to rats at doses of 2 and 65 mg/kg, respectively, by liquid chromatography-tandem mass spectrometric (LC-MS/MS) analysis. Next, an accurate and precise LC-MS/MS method was developed to quantitatively determine fucoxanthin and fucoxanthinol in rat plasma.

Plasma samples were resolved by LC-MS/MS on a reverse-phase SB-C18 column that was equilibrated and eluted with acetonitrile (A)/aqueous 0.1% formic acid (B; 92/8, v/v) at a flow rate of 0.5 mL/min. Analytes were monitored by multiple-reaction monitoring (MRM) under positive electrospray ionization mode. The precursor/product transitions (m/z) were 659.3→109.0 for fucoxanthin, 617.2→109.0 for fucoxanthinol, and 429.4→313.2 for the internal standard (IS). Calibration curves for fucoxanthin and fucoxanthinol were linear over concentrations ranging from 1.53 to 720 and 1.17 to 600 ng/mL, respectively. The inter- and intraday accuracy and precision were within ±15%. The method was applied successfully in a pharmacokinetic study and the resulting oral fucoxanthin bioavailability calculated.


Overview on the antihypertensive and anti-Obesity effects of secondary metabolites from seaweeds


Hypertension and obesity are two significant factors that contribute to the onset and exacerbation of a cascade of mechanisms including activation of the sympathetic and renin-angiotensin systems, oxidative stress, release of inflammatory mediators, increase of adipogenesis and thus promotion of systemic dysfunction that leads to clinical manifestations of cardiovascular diseases. Seaweeds, in addition to their use as food, are now unanimously acknowledged as an invaluable source of new natural products that may hold noteworthy leads for future drug discovery and development, including in the prevention and/or treatment of the cardiovascular risk factors. Several compounds including peptides, phlorotannins, polysaccharides, carotenoids, and sterols, isolated from brown, red and green macroalgae exhibit significant anti-hypertensive and anti-obesity properties.

This review will provide a comprehensive overview of the recent advances on bioactive pure compounds isolated from different seaweed sources focusing on their potential use as drugs to treat or prevent hypertension and obesity. On the other hand, although it is obvious that macroalgae represent promising sources of antihypertensive and anti-obesity compounds, it is also clear that further efforts are required to fully understand their cellular mechanisms of action, to establish structure-inhibition relationships and mainly to evaluate them in pre-clinical and clinical trials.

Keywords: anti-hypertension; anti-obesity; fucoxanthin; peptides; phlorotannins; seaweeds.


Potential of natural products in the Inhibition of Adipogenesis through regulation of PPARγ expression and/or its transcriptional activity


Obesity is a global health problem characterized as an increase in the mass of adipose tissue. Adipogenesis is one of the key pathways that increases the mass of adipose tissue, by which preadipocytes mature into adipocytes through cell differentiation. Peroxisome proliferator-activated receptor γ (PPARγ), the chief regulator of adipogenesis, has been acutely investigated as a molecular target for natural products in the development of anti-obesity treatments. In this review, the regulation of PPARγ expression by natural products through inhibition of CCAAT/enhancer-binding protein β (C/EBPβ) and the farnesoid X receptor (FXR), increased expression of GATA-2 and GATA-3 and activation of the Wnt/β-catenin pathway were analyzed.

Furthermore, the regulation of PPARγ transcriptional activity associated with natural products through the antagonism of PPARγ and activation of Sirtuin 1 (Sirt1) and AMP-activated protein kinase (AMPK) were discussed. Lastly, regulation of mitogen-activated protein kinase (MAPK) by natural products, which might regulate both PPARγ expression and PPARγ transcriptional activity, was summarized. Understanding the role natural products play, as well as the mechanisms behind their regulation of PPARγ activity is critical for future research into their therapeutic potential for fighting obesity.

Keywords: obesity, adipogenesis, PPARγ, natural products, PPARγ expression, PPARγ transcriptional activity.


Gallotannin derivatives from mango (Mangifera indica L.)


Gallotannin derivatives from mango (Mangifera indica L.) suppress Adipogenesis and increase thermogenesis in 3T3-L1 Adipocytes in part through the AMPK


Expansion of adipose tissue in obesity is associated with dysregulation of adipokines, which can lead to long-term metabolic disorders. Gallotannin derivatives from mango possess anti-inflammatory activities, but their potential role in lipid metabolism is not well investigated. In this study, 3T3-L1 preadipocytes were differentiated into adipocytes and treated with mango polyphenols (MG), or pyrogallol (PG) for 6 days. The anti-adipogenic activity of PG was demonstrated by reduced number of lipid droplets and expressions of adipogenic markers, such as peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer binding protein α (C/EBPα).

In mature adipocytes, PG promoted adipocyte browning and increased the expressions of uncoupling protein 1 (UCP1) and sirtuin 1 (Sirt1). Knockdown of AMP-activated protein kinase (AMPK) α1 with siRNA partially abolished the effect of PG on adipogenesis. Results indicate that gallotannin derivatives modulate lipid metabolism, at least in part, though the AMPK pathway, and possess potential to prevent obesity-related conditions.



Obesity is an escalating global epidemic associated with increased risk of developing type 2 diabetes, cardiovascular diseases and cancer. Dietary modifications, particularly increasing consumption of polyphenol-rich foods, are considered some of the most effective strategies in the prevention of obesity-related chronic diseases. Mango (Mangifera indica L.) contains high content of phenolic compounds (e.g., gallic acid (GA), gallotannin (GT), and galloyl glycosides), showing anti-inflammatory and antiobesogenic potential in chronic diseases. Lactobacillus plantarum (L.plantarum) possesses enzymatic activities to degrade GT into GA and PG, allowing for absorption and excretion. Thus, the interaction between gut microbiota and GT derivatives may affect the subsequent biological activities exerted by the microbial metabolites.

Anti-obesogenic potential of GT derivatives from mango in modulating lipid metabolism was investigated in 3T3-L1 adipocytes. GT derivatives suppressed adipogenesis and increased thermogenesis in adipocytes in part through the interactions with the AMPK-C/EBPα/PPARγ and AMPK-UCP1/Sirt1 axes. In gnotobiotic mice fed a high-fat diet (HFD), GT alone decreased lipid accumulation in white adipose tissue and increased thermogenesis in brown adipose tissue. Intestinal colonization with L.plantarum enhanced these effects and additionally lowered levels of inflammation and insulin resistance. GT and L.plantarum reduced HFD-induced inflammation and insulin resistance and promoted thermogenesis in adipose tissue potentially through the activity of GT-metabolizing bacterial enzymes yielding absorbable bioactive of GT-metabolizing bacterial enzymes yielding absorbable bioactive GT metabolites, which implies the potential role of prebiotic-probiotic interactions in the prevention of diet-induced metabolic disorders.

These findings were expected to translate into a human clinical trial, which examined the influence of 6 weeks of daily mango supplementation on inflammation and metabolic functions. Mango supplementation improved the plasma levels of proinflammatory cytokines and metabolic hormones in obese participants partly due to increased systemic exposure to polyphenolic metabolites. In summary, health benefits of mango-derived polyphenols in obesity and insulin resistance are mainly attributed to the production of microbial metabolites of GT, which is in part through the interactions with the AMPK-C/EBPα/PPARγ and AMPKUCP1/Sirt1 axes in adipose tissue. Improving the abundance of probiotics in gut microbiota may improve the bioavailability of mango-derived polyphenols, resulting in enhanced efficacy of the microbial metabolites in the prevention of lipid accumulation and metabolic dysfunction in obesity.



Ganoderma applanatum Polysaccharide

Effect of Ganoderma applanatum Mycelium Extract on the Inhibition of Adipogenesis in 3T3-L1 Adipocytes


Ganoderma applanatum (GA) and related fungal species have been used for over 2000 years in China to Prevent and treat various human diseases. However, there is no critical research evaluating the functionality of GA grown using submerged culture technology. This study aimed to evaluate the effects of submerged culture GA mycelium (GAM) and its active components (protocatechualdehyde [PCA]) on Preadipocyte Differentiation of 3T3-L1 cells. Mouse-derived Preadipocyte 3T3-L1 cells were treated with Differentiation inducers in the presence or absence of GAM extracts. We determined triglyceride accumulations, glycerol-3-phosphate dehydrogenase (GPDH) activities, and Differentiation makers. PCA, the active component of GAM extract, was also used to treat 3T3-L1 cells. The MTT assay showed that the GAM extract (0.01–1 mg/mL) was not toxic to 3T3-L1 preAdipocyte. Treatment of cells with GAM extracts and its active components significantly decreased the GPDH activity and lipid accumulation, a marker of Adipogenesis, in a dose-dependent manner.

Western blot analysis results showed that the protein expression levels of peroxisome proliferator-activated receptor γ (PPARγ ), CCAAT/enhancer-binding protein α (C/EBPα), and sterol regulatory element-binding protein 1 (SREBP1) were Inhibited by the GAM extract. In addition, Adipogenic-specific genes such as perilipin, fatty acid synthase (FAS), fatty acid transport protein 1 (FATP1), and fatty acid-binding protein 4 (FABP4) decreased in a dose-dependent manner. Quantitative high-performance liquid chromatography analysis showed that the GAM extract contained 1.14 mg/g PCA. GAM extracts suppressed Differentiation of 3T3-L1 preadipocytes, in part, through altered regulation of PPARγ, C/EBPα, and SREBP1. These results suggest that GAM extracts and PCA may suppress Adipogenesis by Inhibiting Differentiation of preadipocytes.



Ganoderma lucidum Polysaccharide

Four new lanostane triterpenes, butyl ganoderate A, butyl ganoderate B, butyl lucidenate N, and butyl lucidenate A, were isolated from the fruiting bodies of Ganoderma lucidum together with 14 known compounds. The structures of the new triterpenes were established by extensive spectroscopic studies and chemical evidence. In addition, the Inhibitory effect of isolated compounds on Adipocyte Differentiation in 3T3-L1 cells was examined.



Garcinia cambogia (HCA(Hydroxy Citric acid)


Results of recent studies on gut microbiota have suggested that obesogenic bacteria exacerbate Obesity and metabolic dysfunction in the host when fed a high fat diet (HFD). In order to explore Obesity-associated bacterial candidates and their response to diet, the composition of faecal bacterial communities was investigated by analyzing 16S rRNA gene sequences in mice. Dietary intervention with probiotics and Garcinia cambogia extract attenuated Weight Gain and Adipocyte size in HFD-fed mice.
To identify Obesity-causative microbiota, two statistical analyses were performed. Forty-eight bacterial species were found to overlap between the two analyses, indicating the commonly identified species as diet-driven and Obesity-associated, which would make them strong candidates for host-microbiome interaction on Obesity.
Finally, correlation based network analysis between diet, microbe, and host revealed that Clostridium aminophilum, a hyper-ammonia-producing bacterium, was highly correlated with Obesity phenotypes and other associated bacteria, and shown to be suppressed by the combination of probiotics and Garcinia cambogia extract. Results of the present study suggest that probiotics and Garcinia cambogia extract alleviate Weight Gain and Adiposity, in part via differentially modulating the composition of gut microbiota in HFD fed mice.


Garcinol (Mangosteen shell)


The aim of this work was to study the effects of garcinol and pterostilbene on cell proliferation and Adipogenesis in 3T3-L1 cells. The results showed that garcinol and pterostilbene decreased the cell population growth and caused cell cycle arrest at the G2/M phase in 3T3-L1 preadipocytes. During Adipocyte Differentiation, both garcinol and pterostilbene had Inhibitory effects on fat droplet formation and triacylglycerol accumulation. The data indicated that garcinol and pterostilbene could Inhibit the glycerol-3-phosphate dehydrogenase (GPDH) activity by 97.8 and 61.5%, respectively, as compared to the control. Both garcinol and pterostilbene significantly attenuated the protein expressions of PPARγ and C/EBPα during 3T3-L1 Adipocyte Differentiation.
Moreover, garcinol and pterostilbene caused an Inhibition of lipid accumulation in the 3T3-L1 Adipocyte Differentiation phase. Garcinol and pterostilbene also significantly up-regulated the gene expression of adiponectin as well as down-regulated the gene expressions of leptin, resistin, and fatty acid synthase (FAS) in 3T3-L1 Adipocyte Differentiation . In 3T3-L1 Adipocytes, garcinol significantly down-regulated the protein expressions of PPARγ and FAS as well as up-regulated the protein expressions of adipose triglyceride lipase (ATGL) and adiponectin. Garcinol also significantly up-regulated the gene expression of adiponectin as well as down-regulated the gene expressions of leptin and FAS. These results suggest that garcinol and pterostilbene have Anti-Adipogenic effects on preadipocytes and Adipocytes.


Gelidium elegans


Altered Gelidium elegans Extract-stimulated Beige-like Phenotype Attenuates Adipogenesis in 3T3-L1 cells


Previously, we showed that Gelidium elegans extract (GE) Suppresses oxidative stress and lipid accumulation. However, the molecular mechanism underlying the Anti-Adipogenic ability of GE is still unclear. The levels of Adipogenesis markers and triglyceride synthesis enzymes were measured by western blot. To evaluate the lipid accumulation in 3T3-L1 cells, oil red o staining was performed. We investigated whether GE induces Lipolysis by measuring Adipocyte triglyceride lipase (ATGL) during Adipocyte Differentiation. We also examined the expression of beige cell-associated genes and the production of carbon dioxide in 3T3-L1 cells. We showed that GE increased the protein expression of CAAT/enhancer binding protein (C/EBP) homologous protein 10 and Inhibited the expression of C/EBPβ. GE discouraged triglyceride synthesis via deregulation of lysophosphatidic acid acyltransferase-θ (LPAATθ) and diacylglycerolacyltransferase 1 (DGAT1) during late-stage Adipogenesis in 3T3-L1 cells. GE also dramatically increased ATGL in 3T3-L1 cells.

Finally, in 3T3-L1 cells treated with GE, markers of beige Adipocytes such as PRDM16 and UCP1 were upregulated, and large amounts of carbon dioxide were produced. These data indicate that GE Suppresses Adipogenesis by stimulating a beige-like phenotype in 3T3-L1 cells.



Genistein


Genistein Mediates the Anti-Adipogenic Actions of Sophora japonica L. Extracts


Previous studies showed that feeding diets containing the mature fruits of Sophora japonica L. prevented body Weight Gain and Reduced fat Mass in high-fat diet-induced obese mice. 


Enhanced pro-apoptotic and Anti-Adipogenic effects of genistein plus guggulsterone in 3T3-L1 Adipocytes


Genistein (G), an isoflavone, and guggulsterone (GS), the active substance in guggulipid, have been reported to possess therapeutic effects for Obesity . In the present study, we investigated the effects of combinations of G plus GS on apoptosis.



genistein’s Anti-Adipogenic effect coincides with the expression of CEBPβ, CEBPα and PPARγ , we hypothesized that genistein Inhibits 3T3-L1 cell …



Microarrays showed that antiAdipogenic effects of genistein were principally attributable to activation of Wnt signalling via ERs‐dependent pathway, such as Erk/JNK signalling and LEF/TCF4 co … Unlike genistein, daidzein Inhibited Adipogenesis through stimulation of …



The AntiAdipogenic effect of genistein was not due to Inhibition of insulin receptor subtrate-1 …Genistein (4′,5,7-trihydroxyisoflavone) and all tissue culture materials were from GIBCO … Rabbit polyclonal anti-signal transducers and activators of transcription (STAT) 3 antibody was …



Genistein has been shown to have potential antiObesity effects, decreasing food intake, bodyweight … to ERα [28], but in a cell-based gene transcription assay, genistein was only … Therefore,

the AntiAdipogenic effect of genistein in our study might be partially mediated through …



Genistein Affects Adipose Tissue Deposition in a Dose-Dependent and Gender-Specific Manner.



compounds genistein (G), quercetin (Q), and resveratrol (R) have been reported to each exhibit Anti-Adipogenic activities in Adipocytes and antiproliferative and pro-apoptotic activities in several cell types. We studied the combined effects of G, Q, and R on Adipogenesis and …


The phytoestrogen genistein enhances osteogenesis and represses Adipogenic Differentiation of human primary bone marrow stromal cells


At d 3 of Adipogenesis , TGFβ1 was strongly up-regulated by genistein in an ER-dependent manner …Adipogenic Differentiation and maturation, on the other hand, were Reduced by genistein (and 17β-estradiol) via an ER-dependent mechanism involving autocrine or …


Genistein, EGCG, and capsaicin Inhibit Adipocyte Differentiation process via activating AMP-activated protein kinase


and apoptotic potential [3]. Also, it has been reported that genistein exhibits Anti-Adipogenic effects in several Adipocytes, although its precise mechanism of action is not known [4]. We have investigated the molecular events leading to the Inhibition of Adipogenesis by genistein


Gentiopicroside isolated from Gentiana scabra Bge.


Gentiopicroside isolated from Gentiana scabra Bge. Inhibits Adipogenesis in 3T3-L1 cells and Reduce s body weight in diet-induced obese mice


Gentiopicroside is a major active component of the Gentiana scabra Bge., which is commonly used as herbal medicine for the treatment of Inflammation in Asia. Gentiopicroside significantly down-regulated expression of key Adipogenic transcription factors (PPARγ, C/EBPα, SREBP-1c) and dose-dependently Inhibited the lipid uptake-related gene (LPL), fatty acid transport-related gene (FABP4) and triglyceride (TG) synthesis-related gene (DGAT2), as well as fatty acid synthesis-related genes (FAS, SCD1), which resulted in Reduced intracellular lipid droplet accumulation and TG content in 3T3-L1 cells.
Gentiopicroside also down-regulated expression of inflammatory cytokine genes (NFκB1, TNFα, IL6) compared with vehicle. Oral administration of gentiopicroside (50 mg/kg) in mice fed with high-fat diet for 12 weeks resulted in Reduced body weight and visceral fat Mass compared with the control group. Overall, the results of this study showed that gentiopicroside had positive Anti-Obesity effects by regulating the expression of Adipogenesis /lipogenesis-related genes and inflammatory genes in 3T3-L1, and that it effectively Reduced body weight and visceral fat Mass in vivo.

AntiObesity Effects of Gentiana lutea Extract on 3T3-L1 preadipocytes and a High-Fat Diet-Induced Mouse Model


Obesity is one of the most common metabolic diseases resulting in Metabolic Syndrome. In this study, we investigated the AntiObesity effect of Gentiana lutea L. (GL) extract on 3T3-L1 preadipocytes and a high-fat-diet (HFD)-induced mouse model. For the induction of preadipocytes into Adipocytes , 3T3-L1 cells were induced by treatment with 0.5 mM 3-isobutyl-1-methylxanthine, 1 mM dexamethasone, and 1 μg/mL insulin. Adipogenesis was assessed based on the messenger ribonucleic acid expression of Adipogenic -inducing genes (adiponectin (Adipoq), CCAAT/enhancer-binding protein alpha (Cebpa), and glucose transporter type 4 (Slc2a4)) and lipid accumulation in the differentiated Adipocytes was visualized by Oil Red O staining. In vivo, obese mice were induced with HFD and coadministered with 100 or 200 mg/kg/day of GL extract for 12 weeks. GL extract treatment Inhibited Adipocyte Differentiation by downregulating the expression of Adipogenic -related genes in 3T3-L1 cells.
In the obese mouse model, GL extract prevented HFD-induced Weight Gain, fatty hepatocyte deposition, and Adipocyte size by decreasing the secretion of leptin and insulin. In conclusion, GL extract shows AntiObesity effects in vitro and in vivo, suggesting that this extract can be beneficial in the prevention of Obesity.


Ginkgetin, a biflavone from Ginkgo biloba leaves


Ginkgetin, a biflavone from Ginkgo biloba leaves, prevents Adipogenesis through STAT5-mediated PPARγ and C/EBPα regulation


Adipogenesis involved in hypertrophy and hyperplasia of Adipocytes is responsible for expanding the Mass of Adipose Tissue s in obese individuals. Peroxisome proliferator-activated receptor γ (PPARγ ) and CCAAT/enhancer-binding protein α(C/EBPα ) are two principal transcription factors induced by delicate signaling pathways, including signal transducer and activator of transcription 5 (STAT5), in Adipogenesis. Here, we demonstrated a novel role of ginkgetin, a biflavone from Ginkgo biloba leaves, as a STAT5 Inhibit or that blocks the Differentiation of preadipocytes into Adipocytes. During the Differentiation of 3T3-L1 cells, ginkgetin treatment during the first 2 days markedly Inhibited the formation of lipid-bearing Adipocytes. PPARγ and C/EBPα expression was decreased in 3T3-L1 cells during Adipogenesis following ginkgetin treatment, whereas no change was observed in C/EBPβ or C/EBPδ expression. Inhibition of PPARγ and C/EBPα expression by ginkgetin occurred through the prevention of STAT5 activation during the initiation phase of Adipogenesis.

In addition, ginkgetin-mediated the Inhibition of Adipogenesis was recapitulated in the Differentiation of primary preadipocytes. Lastly, we confirmed the Inhibitory effects of ginkgetin on the hypertrophy of white Adipose Tissues from high-fat diet-fed mice. These results indicate that ginkgetin is a potential anti-Adipogenesis and Anti-Obesity drug.


Ginkgetin: A natural biflavone with versatile pharmacological activities


Natural products, being richly endowed with curative powers, have become spotlight for biomedical and pharmaceutical research to develop novel therapeutics during recent years. Ginkgetin, a natural non-toxic biflavone, has been shown to exhibit anti-cancer, anti-inflammatory, anti-microbial, Anti-Adipogenic , and neuroprotective activities. Ginkgetin combats cancer progression by arresting cell cycle, inducing apoptosis, stimulating autophagy, and targeting many deregulated signaling pathways such as JAK/STAT and MAPKs. Ginkgetin halts Inflammation mediators like interleukins, iNOS, COX-2, PGE2, NF-κB, and acts as an Inhibit or of PLA2. GK shows strong neuroprotection against oxidative stress-promoted cell death, Inhibits cerebral micro-hemorrhage, decreases neurologic deficits, and halts apoptosis of neurons.

Ginkgetin also acts as anti-fungal, anti-viral, anti-bacterial, leishmanicidal and anti-plasmodial agent. Ginkgetin shows substantial preventive or therapeutic effects in in vivo models of many diseases including atherosclerosis, cancer, neurodegenerative, hepatic, influenza, and inflammatory diseases. Based on various computational, in vitro and in vivo evidences, this article demonstrates the potential of ginkgetin for development of therapeutics against various diseases. Although GK has been systematically studied from pharmacological point of view, a vast field of pharmacokinetics, pre-clinical and clinical studies is still open for the researchers to fully validate its potential for the treatment of various diseases.


Isoginkgetin enhances adiponectin secretion from differentiated adiposarcoma cells via a novel pathway involving AMP-activated protein kinase


The recent discovery of adiponectin and its role in Metabolic Syndrome has pointed to a new direction of therapeutics. The fact that adiponectin levels are inversely correlated with the degree of Obesity and insulin resistance suggests the benefit of lifting plasma adiponectin levels. The feasibility of this approach has been demonstrated in various animal models (Shklyaev et al. 2003, Yamauchi et al. 2003a). We are interested in exploring approaches based on up-regulation of adiponectin using small molecules. At present, the only available small molecules with this property are the synthetic PPARγ agonists (Maeda et al. 2001, Combs et al. 2002). The PPARγ agonists, such as the drug Avandia (rosiglitazone), not only ameliorate insulin resistance but also cause Weight Gain which further increases the risk of type II Diabetes and limits its application for Diabetes. PPARα agonists are also being investigated for this application due to their weight loss effect in rodents (Liu et al. 2005), but the potency of these agonists are generally lower than PPARγ agonists.

In this study, we report a natural compound, isoginkgetin, up-regulating adiponectin production with potency comparable to that of rosiglitazone, but devoid of the potent Adipogenesis-promoting effect of rosiglitazone. Isoginkgetin is one of the major active constituents in G. biloba extract, which has been used as an ancient Chinese remedy for its multiple biological activities, including anti-oxidant, anti-inflammatory, and anti-tumor activities (Yoshikawa et al. 1999, DeFeudis et al. 2003). The cellular signaling induced by this compound is largely unknown. This is the first study to reveal the potential insulin-sensitizing effects of isoginkgetin based on its regulation of adiponectin. Our observation of the effect of isoginkgetin on Lipolysis is different from the results reported by Dell’Agli & Bosisio (2002). This could be due to the different experimental settings, such as incubation time. In addition, Dell’Agli observed a bi-phase effect of isoginkgetin on Lipolysis, an enhancement of Lipolysis was only observed at 0.03–0.3 μM isoginkgetin, and the abolishment of this effect is not explainable by cytotoxicity. Therefore, more detailed studies under different experimental conditions need to be carried out to address this distinction.

The mechanism of isoginkgetin action is apparently distinct from that of rosiglitazone for a number of reasons. First, Inhibition of PPARγ activity did not affect the effect of isoginkgetin, whereas the activity of rosiglitazone was significantly diminished. Second, isoginkgetin and rosiglitazone exhibited differential stimulation kinetics. Third, results on the adiponectin mRNA and protein levels suggest the differential regulation of adiponectin synthesis and secretion by these two compounds. Fourth, although both compounds have comparable effects on adiponectin production, they have differential effects on Adipogenesis and PPARγ activation.

Regulation of adiponectin production seems complex and has not been fully understood. Hormones and cytokines, including insulin, TNF-α, and interleukin-6, exhibited different effects on adiponectin production in vitro, while the signaling pathways mediating these regulations are largely unknown. The results that isoginkgetin potentiates adiponectin secretion in a likely PPARγ -independent manner suggest new types of targets and agents that can be explored for new therapy.

AMPK is a key regulator for energy metabolism in vivo. Activation of AMPK is involved in fat oxidation via the Inhibition of downstream acetyl CoA carboxylase (ACC; Tomas et al. 2002). PPARγ agonists activate AMPK and Inhibit ACC both in vitro and in vivo in skeletal muscle (Saha et al. 2004). Our data suggest that AMPK or genes upstream of the AMPK pathway maybe effectors of isoginkgetin action. Further experiments, for example introducing the kinase-dead AMPK into Adipocytes to investigate its effect on isoginkgetin-regulated adiponectin production, would be helpful to confirm the involvement of AMPK. Whether isoginkgetin activates AMPK by increasing the ratio of AMP:ATP, as rosiglitazone does (Fryer et al. 2002a), needs to be further investigated. An interesting finding that Inhibition of AMPK-Reduced basal adiponectin production in differentiated Adipocytes suggests the general role of AMPK in the regulation of adiponectin. Whatever the mechanism, the activation of AMPK and Inhibition of phosphor-ACC by isoginkgetin will be beneficial for lipid regulation. Moreover, AMPK pathway can be further explored for adiponectin up-regulation.

In conclusion, isoginkgetin is a promising candidate compound for the treatment of insulin resistance based on our in vitro studies. Further in vivo studies are warranted. Analogs of isoginkgetin possessing higher potency in adiponectin production and lower PPARγ agonist activity can also be explored to identify more active compounds in vivo. Currently, insulin-sensitizing drugs mainly include PPARγ agonists and compounds that target the insulin signaling pathway. This study presents for the first time a potential new strategy for the discovery of insulin sensitizers by screening adiponectin production. Moreover, the critical genes involved in isoginkgetin action may provide novel targets for anti-Diabetic therapy.


Potential anti-obesogenic effects of ginkgo biloba observed in epididymal white Adipose Tissue of obese rats


The definition of white Adipose Tissue (WAT) as an inert Mass for energy storage is long gone; over the last two decades the Adipose Tissue has been recognized as a dynamic tissue and key player in the modulation of energy metabolism (1, 2). Adipokines such as leptin, adiponectin, and tumour necrosis factor-α (TNF-α) have a direct effect on energy homeostasis and modulation of low-grade Inflammation (3). The intake of high fat diets has the potential to not only disturb normal adipokine secretion but also to remodel Adipose Tissue by increasing Adipocyte size and/or number, contributing to the development of a pro-inflammatory microenvironment (4, 5). These perturbations have been positively associated with metabolic disorders such as Obesity, type 2 Diabetes , non-alcoholic fatty liver disease (NAFLD), insulin resistance, and cardiovascular diseases (6, 7).

In Obesity , particularly visceral Obesity , enlarged WAT visceral Adipocytes show dysregulated Lipolysis , inducing high levels of circulating non-esterified fatty acids (NEFAs) (8, 9). NEFAs in normal circumstances are utilized as energy by tissues such as liver and muscle; however, when in excess they contribute to the development of insulin resistance (4, 911). Furthermore, in response to overnutrition, hypertrophic Adipocytes contribute to increased circulating triacylglycerol (TAG) levels mainly from de novo lipogenesis, in which fatty acids (FA) are synthetized from non-lipid substrates, particularly carbohydrates, or from FA obtained from ex-situ lipid sources such as chylomicrons and very-low-density lipoproteins (VLDL) (12, 13). Visceral Obesity seems to play a central role in the development of metabolic disorders, being associated with low-grade chronic Inflammation and the production of pro-inflammatory cytokines which have the potential to trigger insulin resistance and endothelial dysfunction (1416).

In this context, several pharmacological approaches have been tried for the treatment of Obesity. However, more often than not such approaches were followed by undesired side effects, including psychiatric manifestations, increased risk of cardiovascular events, and others (17). Considering the dramatic increase in the prevalence of Obesity over the last decades globally, a range of anti-obesogenic alternative supplementation therapies based on plant extracts (18) have been investigated.

More recently, Ginkgo biloba Extract (GbE) has been investigated as an alternative therapy for metabolic disorders associated with Obesity. GbE, a herbal extract containing Flavonoids, terpenoids, and terpene lactones (19), is a well-known phytotherapic compound often employed as coadjuvant supplement in neurodegenerative diseases (20, 21), NAFLD (22, 23), type 1 and 2 Diabetes (24, 25). Previous findings from our research group showed that diet-induced obese (DIO) rats supplemented with GbE showed Reduced food and energy intake, Reduced body Adiposity, improved insulin signalling and sensitivity, enhanced insulin receptor and AKT phosphorylation, and Reduced NFκB-p65 phosphorylation in retroperitoneal Adipose Tissue (26, 27).

GbE may have a potentially therapeutic use for menopause-associated Obesity; supplementation with 500 mg/kg of GbE stimulated hypothalamic serotonergic activity in ovariectomized rats (28). GbE isolated bioactive compounds have been demonstrated to stimulate Lipolysis in 3T3-L1 Adipocytes (29), and to Inhibit Adipogenesis through activation of the AMPK pathway (30). However, the effects of GbE supplementation on metabolic processes of visceral Adipose Tissue in DIO rats remain largely unknown. In view of the considerations highlighted above, the aim of the present study was to investigate the effects of GbE supplementation as a potentially anti-obesogenic effector for improvement in lipid metabolism of epididymal Adipose Tissue of DIO rats.



Ginkgo Biloba


Ginkgetin, a biflavone from Ginkgo biloba leaves, prevents Adipogenesis through STAT5-mediated PPARγ and C/EBPα regulation


Adipogenesis involved in hypertrophy and hyperplasia of Adipocytes is responsible for expanding the Mass of Adipose Tissue s in obese individuals. Peroxisome proliferator-activated receptor γ (PPARγ ) and CCAAT/enhancer-binding protein α (C/EBPα ) are two principal transcription factors induced by delicate signaling pathways, including signal transducer and activator of transcription 5 (STAT5), in Adipogenesis. Here, we demonstrated a novel role of ginkgetin, a biflavone from Ginkgo biloba leaves, as a STAT5 Inhibit or that blocks the Differentiation of preadipocytes into Adipocytes.

During the Differentiation of 3T3-L1 cells, ginkgetin treatment during the first 2 days markedly Inhibited the formation of lipid-bearing Adipocytes . PPARγ and C/EBPα expression was decreased in 3T3-L1 cells during Adipogenesis following ginkgetin treatment, whereas no change was observed in C/EBPβ or C/EBPδ expression. Inhibition of PPARγ and C/EBPα expression by ginkgetin occurred through the prevention of STAT5 activation during the initiation phase of Adipogenesis. In addition, ginkgetin-mediated the Inhibition of Adipogenesis was recapitulated in the Differentiation of primary preadipocytes. Lastly, we confirmed the Inhibitory effects of ginkgetin on the hypertrophy of white Adipose Tissues from high-fat diet-fed mice. These results indicate that ginkgetin is a potential anti-Adipogenesis and Anti-Obesity drug.



Ginkgolide C, isolated from Ginkgo biloba leaves


Ginkgolide C Suppresses Adipogenesis in 3T3-L1 Adipocytes via the AMPK signaling pathway


Ginkgolide C, isolated from Ginkgo biloba leaves, is a flavone reported to have multiple biological functions, from decreased platelet aggregation to ameliorating Alzheimer disease. The study aim was to evaluate the AntiAdipogenic effect of ginkgolide C in 3T3-L1 Adipocytes. Ginkgolide C was used to treat differentiated 3T3-L1 cells. Cell supernatant was collected to assay glycerol release, and cells were lysed to measure protein and gene expression related to Adipogenesis and Lipolysis by western blot and real-time PCR, respectively. Ginkgolide C significantly suppressed lipid accumulation in differentiated Adipocytes.

It also decreased Adipogenesis-related transcription factor expression, including peroxisome proliferator-activated receptor and CCAAT/enhancer-binding protein. Furthermore, ginkgolide C enhanced adipose triglyceride lipase and hormone-sensitive lipase production for Lipolysis and increased phosphorylation of AMP-activated protein kinase (AMPK), resulting in decreased activity of acetyl-CoA carboxylase for fatty acid synthesis. In coculture with an AMPK Inhibit or (compound C), ginkgolide C also improved activation of sirtuin 1 and phosphorylation of AMPK in differentiated 3T3-L1 cells. The results suggest that ginkgolide C is an effective flavone for increasing Lipolysis and Inhibiting Adipogenesis in Adipocytes through the activated AMPK pathway.


Ginkgolide C Reduced oleic acid-induced lipid accumulation in HepG2 cells


Ginkgolide C, isolated from Ginkgo biloba, is a diterpene lactone that has multiple biological functions and can improve Alzheimer disease and platelet aggregation. Ginkgolide C also Inhibits Adipogenesis in 3T3-L1 Adipocytes . The present study evaluated whether ginkgolide C Reduced lipid accumulation and regulated the molecular mechanism of lipogenesis in oleic acid-induced HepG2 hepatocytes. HepG2 cells were treated with 0.5 mM oleic acid for 48 h to induce a fatty liver cell model. Then, the cells were exposed to various concentrations of ginkgolide C for 24 h. Staining with Oil Red O and the fluorescent dye BODIPY 493/503 revealed that ginkgolide C significantly Reduced excessive lipid accumulation in HepG2 cells. Ginkgolide C decreased peroxisome proliferator-activated receptor γ and sterol regulatory element-binding protein 1c to block the expression of fatty acid synthase.
Ginkgolide C treatment also promoted the expression of adipose triglyceride lipaseand the phosphorylation level of hormone-sensitive lipase to enhance the decomposition of triglycerides. In addition, ginkgolide C stimulated CPT-1 to activate fatty acid β-oxidation, significantly increased sirt1 and phosphorylation of AMP-activated protein kinase (AMPK), and decreased expression of acetyl-CoAcarboxylase for suppressed fatty acid synthesis in hepatocytes. Taken together, our results suggest that ginkgolide C Reduced lipid accumulation and increased Lipolysis through the sirt1/AMPK pathway in oleic acid-induced fatty liver cells.


Ginsenoside Rg1


AntiObesity effects of ginsenoside Rg1 on 3T3-L1 preadipocytes and high fat diet-induced obese mice mediated by AMPK


Ginsenosides Rg1 is one of the major pharmacologically active saponins in ginseng, which as an antioxidant Reduces oxidative damage in the liver and can also be used to Prevent cardiovascular diseases and Diabetes. However, there is no research targeting the effect of lipid metabolism in high-fat diet (HFD)-induced mice. In this study, we evaluated the Anti-Obesity effects of Rg1 in 3T3-L1 Adipocyte cells and HFD-induced obese C57BL/6J mice. Administration of Rg1 to HFD-induced obese mice significantly decreased body weight, total cholesterol, and total triglyceride levels. In addition to effects in 3T3-L1 cells, Rg1 Reduced the accumulation of lipid droplets in a dose-dependent manner.

Furthermore, Rg1 exhibits an Anti-Adipogenic effect via regulation of the expression of the transcriptional factors and lipid metabolism-related genes in vivo and in vitro. We observed that Rg1 administration significantly increased the phosphorylation level of AMP-activated protein kinase (AMPK) in both epididymal white Adipose Tissue and 3T3-L1 cells. These results indicated that Rg1 works both in an Anti-Adipogenic and Anti-Obesity manner through inducing AMPK activation, Inhibiting lipogenesis, and decreasing intracellular lipid content, Adipocyte size, and adipose weight.



Ginsenoside Rg2


Ginsenoside Rg2 Inhibits Adipogenesis in 3T3-L1 preadipocytes and Suppresses Obesity in high-fat-diet-induced obese mice through the AMPK pathway


Ginsenoside Rg2 is one of the specific ginsenosides in red ginseng, and has been reported to exhibit protective effects against neurotoxicity and memory impairment, and also Inhibition of hepatic glucose production. However, the effect of Rg2 on the prevention of Obesity has not been investigated. In this study, we evaluated the Anti-Obesity and Anti-Adipogenic effects of Rg2 in high-fat diet-induced obese mice (HFD mice) and 3T3-L1 preadipocytes . Oral administration of Rg2 (10 mg kg−1) to HFD mice significantly decreased body Weight Gain, total triglycerides, and free fatty acid levels.
In 3T3-L1 preadipocytes, Rg2 (80 μM) Inhibited Adipocyte Differentiation and Reduced the accumulation of intracellular lipids. Quantitative PCR and western blot analysis revealed that Rg2 decreased the expression levels of Adipogenic transcription factors (PPARγ, C/EBPα, and SREBP1-c), and then regulated target genes such as acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS). Rg2 significantly promoted AMP-activated protein kinase (AMPK) both in vivo and in vitro, which is known to suppress Adipogenesis. It was also found that pretreating with compound C, a typical Inhibit or of AMPK, attenuated the Inhibitory effect of Rg2 on AMPK phosphorylation. These findings suggested that Rg2-induced activation of AMPK leads to a decrease in the expression of Adipogenic transcription factors, and suppression of Adipogenesis in vivo and in vitro. Hence, Rg2 has the potential for the development of healthy foods and the prevention of Obesity.


Ginsenoside Rg3


AntiAdipogenic effects and mechanisms of ginsenoside Rg3 in Pre-Adipocytes and obese mice


Red or black ginseng has been reported more powerful than white/fresh ginseng in dealing with various diseases/conditions including Obesity. The major reason is that heating/steaming, the process of making red or black ginseng, produces large amount of bioactive compounds including ginsenoside Rg3 (Rg3), which are trace in fresh or white ginseng. In the present study, Rg3 was applied both in Pre-Adipocytes and obese mice to investigate the Anti-Adipogenic effects and relevant mechanisms. Our results show that Rg3 dose-dependently Inhibited cell Differentiation both in 3T3-L1 cells (30, 50, and 100 μM) and human primary Pre-Adipocytes (10, 20, and 30 μM).

This Inhibitory effect is accompanied by the attenuation of the expressions of Adipogenic markers including peroxisome proliferator-activated receptor gamma (PPAR-γ), CCAAT/enhancer binding protein alpha (C/EBP-α), fatty acid synthase (FAS), fatty acid binding protein 4 (FABP4) and perilipin. Although dietary intake of Rg3 (0.1 mg Rg3/kg diet, 8 weeks) did not significantly affect body Weight Gain , fat pads and food intake as well as of PPAR-γ expression in fat tissues, we found that hepatic PPAR-γ and C/EBP-α protein expressions and hepatic glutathione reductase and glutathione S-transferase, two major Antioxidants molecules were significantly Reduced by Rg3. These results suggest that ginsenoside Rg3 may be a potential agent in reducing/preventing Obesity.


AntiObesity effect of ginsenoside Rg3 involves the AMPK and PPAR‐γ signal pathways


Ginsenosides, the active component of ginseng, exerts AntiDiabetic and anticancer effects. This study investigated the molecular basis of ginsenoside Rg3, a red ginseng rich constituent, focusing on its ability to Inhibit Adipocyte Differentiation in 3T3‐L1 cells. The data show that ginsenoside Rg3 was effective in the Inhibition of Adipocyte Differentiation.

This Inhibitory effect of ginsenoside Rg3 on Adipocyte Differentiation was accompanied by PPAR‐γ Inhibition in rosiglitazone‐treated cells. The study also tested whether AMP‐activated protein kinase (AMPK) activation was involved in the Inhibitory effects of ginsenoside Rg3. AMPK plays a role in maintaining health in the context of diseases such as type 2 Diabetes, Obesity and cancer. AMPK was reported to control nutritional and hormonal signal modulating. Rg3 significantly and time‐dependently activated AMPK. Taken together, these results suggest that the AntiObesity effect of red ginseng rich constituent, ginsenoside Rg3, involves the AMPK signaling pathway and PPAR‐γ Inhibition.


Ginsenoside Rg3 ameliorated HFD-induced hepatic steatosis through downregulation of STAT5-PPARγ


Ginsenoside Rg3 Reduced lipogenesis in 3T3-L1 cells

The chemical structure of the natural compound ginsenoside Rg3 is shown in Fig. 1A. It has been reported to regulate Adipocyte Differentiation (Hwang et al. 2009), but it is not clear how Rg3 regulates lipogenesis. Healthy Adipose Tissue expansion, which involves the accumulation of lipid inside adipose cells, functions in stark contrast with Obesity-induced pathologies such as Inflammation and insulin resistance (Sun et al. 2011, Kusminski et al. 2016). We first assessed whether Rg3 influences lipogenesis in 3T3-L1 cells. Rg3 has been reported to induce apoptosis and cause cytotoxicity (Kim et al. 2014, Shan et al. 2015, Tian et al. 2016); therefore, we analyzed cell viability to rule out such side effects. The high concentration of 100-μM Rg3 induced cytotoxicity and Reduced the viability of 3T3-L1 cells.

The LC50 value of Rg3 in 3T3-L1 cell lipogenesis is 200 μM, and our working concentrations (5, 25 and 50 μM) of Rg3 Reduced lipogenesis without cell toxicity (Fig. 1B and C). To investigate the Inhibiting effect on lipid accumulation by Rg3, we treated 3T3-L1 cells for 3 or 9 days. Oil Red O staining confirmed lipid accumulation in the cell. At 3 days, the lipogenic cocktail MDI (insulin, dexamethasone and isobutylmethylxanthine) induced in Pre-Adipocytes a very low increase in their accumulated fat deposits. When Rg3 was combined with MDI-induced cells, their lipid accumulation was suppressed (Fig. 1D), in an Rg3 dose-dependent manner, as assessed at 9 days after treatment using Oil Red O staining microscopy data and Oil Red O absorbance at 520 nm (Fig. 1E). In Adipocytes, lipid accumulation is related to the amount of TGs in the cell. Similar to the Oil Red O staining results, TG storage was Reduced by Rg3 treatment in 3T3-L1 cells in a dose-dependent manner (Fig. 1F). These data suggest that Rg3 can Prevent lipogenesis and lipid accumulation in the 3T3-L1 cell line.


Ginsenoside Rg3 induces browning of 3T3-L1 Adipocytes by activating AMPK signaling


Ginsenoside Rg3, one of the major components in Panax ginseng, has been reported to possess several therapeutic effects including Anti-Obesity properties. However, its effect on the browning of mature white Adipocytes as well as the underlying mechanism remains poorly understood. In this study, we suggested a novel role of Rg3 in the browning of mature 3T3-L1 Adipocytes by upregulating browning-related gene expression. The browning effects of Rg3 on differentiated 3T3-L1 Adipocytes were evaluated by analyzing browning-related markers using quantitative PCR, immunoblotting, and immunostaining. In addition, the size and sum area of lipid droplets in differentiated 3T3-L1 Adipocytes were measured using Oil-Red-O staining. In mature 3T3-L1 Adipocytes, Rg3 dose-dependently induced the expression of browning-related genes such as Ucp1, Prdm16, Pgc1α, Cidea, and Dio2.

Moreover, Rg3 induced the expression of beige fat-specific genes (CD137 and TMEM26) and lipid metabolism-associated genes (FASN, SREBP1, and MCAD), which indicated the activation of lipid metabolism by Rg3. We also demonstrated that activation of 5’ adenosine monophosphate-activated protein kinase (AMPK) is required for Rg3-mediated up-regulation of browning gene expression. Moreover, Rg3 Inhibited the accumulation of lipid droplets and Reduced the droplet size in mature 3T3-L1 Adipocytes.

Taken together, this study identifies a novel role of Rg3 in browning of white Adipocytes, as well as suggesting a potential mechanism of an Anti-Obesity effect of Panax ginseng.



Glehnia littoralis Root


Glehnia littoralis root extract Inhibits Fat Accumulation in 3T3-L1 cells and high-fat diet-induced obese mice by downregulating Adipogenic gene expression


Glehnia littoralis has been reported to have several pharmacological properties but no reports describing the AntiAdipogenic effect of this plant have been published. This study was conducted to investigate the effects of Glehnia littoralis root hot water extract (GLE) and its underlying mechanism on 3T3-L1 cell Adipogenesis and in high-fat diet- (HFD-) induced obese mice. We measured intracellular lipid accumulation using oil red O staining in vitro. For in vivo study, twenty-eight C57BL/6J male mice were randomly divided into four groups, Control, HFD, HFD + 1% GLE, and HFD + 5% GLE, which was performed for eight weeks.
We determined the expression levels of the Adipogenesis -related proteins by RT-PCR and western blotting in HFD-induced obese mice. The GLE dose-dependently Inhibited 3T3-L1 Adipocyte Differentiation and intracellular lipid accumulation in differentiated Adipocytes. Further, body Weight Gain and Fat Accumulation were significantly lower in the GLE-treated HFD mice than in the untreated HFD mice. GLE treatment suppressed the expression of Adipogenic genes such as peroxisome proliferator-activated receptor (PPAR) γ, CCAAT/enhancer-binding protein (C/EBP) α, fatty acid synthase (aP2), and fatty acid synthase (FAS).
These results suggest that the GLE Inhibits Adipocyte Differentiation and intracellular lipid accumulation by downregulating the Adipogenic gene expression both in vitro and in vivo.


Guarana (Paullinia cupana)

Guarana (Paullinia cupana) is a plant originated in Brazil that presents a beneficial effect on body weight control and metabolic alterations. The aim of this study was to evaluate the effects of guarana on genes and miRNAs related to Adipogenesis in 3T3L1 cells. The Anti-Adipogenic effect of guarana was evaluated by Oil Red-O staining. Gene and miRNA expression levels were determined by real time PCR. The Cebpα and β-catenin nuclear translocation were evaluated using immunocytochemistry. Our data indicated that the triglyceride-reducing effect of guarana was dose-dependent from 100 to 300 µg/mL (−12%, −20%, −24% and −40%, respectively, p < 0.0001). An up-regulation of the Anti-Adipogenic genes Wnt10b, Wnt3a, Wnt1, Gata3 and Dlk1 and a down-regulation of Pro-Adipogenic genes Cebpα, PPARγ and Creb1 were also observed. Furthermore, guarana repressed mmu-miR-27b-3p, mmu-miR-34b-5p and mmu-miR-760-5p, that contributed for up-regulation of their molecular targets Wnt3a, Wnt1 and Wnt10b.

Additionally, cells treated with guarana presented an increase on β-catenin nuclear translocation (p < 0.0018). In summary, our data indicate that guarana has an Anti-Adipogenic potential due to its ability to modulate miRNAs and genes related to this process. Together our data demonstrate the important role of guarana as a putative therapeutic agent.



Guggulsterone (Commiphora mukul tree)

Guggulsterone Inhibits Adipocyte Differentiation and Induces Apoptosis in 3T3‐L1 Cells


Objective: To determine the effects of guggulsterone (GS), the active substance in guggulipid, on apoptosis, Adipogenesis , and Lipolysis using 3T3‐L1 cells.

Methods and Procedures: For apoptosis and Lipolysis experiments, mature Adipocytes were treated with GS isomers. Viability, apoptosis, and caspase 3/7 activation were quantified using MTS, enzyme‐linked immunosorbent assay (ELISA), caspase‐Glo 3/7 activity assay, respectively. The expression of cytochrome c was demonstrated by western blot. Lipolysis was quantified by measuring the release of glycerol. For Adipogenesis experiments, postconfluent preadipocytes were incubated with GS isomers for up to 6 days during maturation. Adipogenesis was quantified by measuring lipid content using Nile Red dye. Western blot was also used to demonstrate the Adipocyte ‐specific transcription factors peroxisome proliferator–activated receptor γ2 (PPARγ 2), CCAAT/enhancer binding protein α (C/EBPα ), and C/EBPβ.

Results: In mature Adipocytes cis‐GS decreased viability, whereas the trans‐GS isomer had little effect. Both isomers caused dose‐dependent increases in apoptosis and cis‐GS was more effective than trans‐GS in inducing apoptosis. cis‐ and trans‐GS also increased caspase‐3 activity and release of cytochrome c from mitochondria. In maturing preadipocytes , both isomers were equally effective in reducing lipid content. The Adipocyte ‐specific transcription factors PPARγ 2, C/EBPα , and C/EBPβ were downregulated after treatment with cis‐GS during the maturation period. Furthermore, cis‐GS increased basal Lipolysis of mature Adipocytes , but trans‐GS had no effect.

Discussion: These results indicate that GS isomers may exert AntiObesity effects by Inhibiting Differentiation of preadipocytes , and by inducing apoptosis and promoting Lipolysis of mature Adipocytes . The cis‐GS isomer was more potent than the trans‐GS isomer in inducing apoptosis and Lipolysis in mature Adipocytes .



Heshouwu (Polygonum multiflorum Thunb)

Heshouwu (Polygonum multiflorum Thunb.) ethanol extract Suppresses Pre-Adipocytes Differentiation in 3T3-L1 cells and Adiposity in obese mice


This study investigated whether Heshouwu (Polygonum multiflorum Thunb.) root ethanol extract (PME) has Anti-Obesity activity using 3T3-L1 cells and high-fat diet (HFD)-induced obese mice. Treatment with PME (5 and 10 μg/mL) dose-dependently suppressed 3T3-L1 pre-Adipocyte Differentiation to Adipocytes and cellular triglyceride contents. In addition, PME Inhibited mRNA and protein expression of Adipogenic transcription factors such as CCAAT/enhancer-binding protein α (C/EBPα ) and peroxisome proliferator-activated receptor γ (PPARγ ), which led to down-regulation of fatty acid synthase gene expression. After feeding mice PME (0.05%) with HFD for 12 weeks, their visceral fat Mass , size and body weight were significantly Reduced compared with the HFD group.

Furthermore, PME supplementation significantly up-regulated the PPARα, CPT1, CPT2, UCP1 and HSL mRNA levels compared with the HFD group, whereas it down-regulated expression of the PPARγ and DGAT2 genes. Finally, HFD increased serum leptin, insulin, glucose and insulin and glucose levels; however, PME reversed these changes. These results demonstrated that PME might relieve Obesity that occurs via Inhibition of Adipogenesis and lipogenesis as well as through Lipolysis and fatty acid oxidation in 3T3-L1 cells and HFD-induced obese mice.



Hesperidin (Citrus aurantium L)

We explored the potential of hesperidin and capsaicin, separately and in combination, to induce white Adipose Tissue (WAT) browning and to help body weight management in Western diet-fed rats. Adult male Wistar rats were fed for 8 weeks with Western diet and treated daily with hesperidin (100 mg/kg/day), capsaicin (4 mg/kg/day), hesperidin (100 mg/kg/day) + capsaicin (4 mg/kg/day), or the vehicle. Hesperidin and capsaicin separately, but not (or to a lesser extent) the combination, resulted in a decreased size of Adipocytes and induced emergence of multilocular brown-like Adipocytes positive for UCP1 and CIDEA in retroperitoneal WAT.

Expression levels of browning markers, such as Prdm16, in inguinal WAT also increased with capsaicin treatment compared with the vehicle (145% ± 17% vs 92% ± 21%, P < 0.05), but no significant effects were found with the combination (106% ± 12%). Thus, the combination of both bioactives Reduces the effectiveness of each compound to decrease the Adipocyte size and induce WAT browning.



Hibiscus rosa sinensis flower


AMPK activating and anti Adipogenic potential of Hibiscus rosa sinensis flower in 3T3-L1 cells


Ethnopharmacological relevance: The flowers of Hibiscus rosa sinensis has array of pharmacological actions. They are used in preparation of herbal decoction and teas, which have been used traditionally to reduce body weight and for its effect on metabolic syndrome.

Aim of the study: To investigate the anti adipogenic efficacy of major fraction from ethyl acetate extract of the Hibiscus rosa sinensis flower at 25 and 50 µg/mL (HRF 25 and 50 µg/mL) in 3T3-L1 cells and delineate its possible mechanism of action.

Materials and methods: Pre adipocyte 3T3-L1 cells were differentiated in the presence and absence of HRF 25 and 50 µg/mL, their lipid accumulation was measured qualitatively by Oil red O staining and quantitatively by triglyceride estimation. Effect on adipolysis was determined, adipogenic and its regulatory gene and protein expression were studied and effect of HRF 25 and 50 µg/mL on AMPK was confirmed in the presence of dorsomorphin.

Results: Treatment with HRF 25 and 50 µg/mL activated AMP-activated protein kinase (AMPK) and was found to alleviate triglyceride accumulation significantly (p < 0.001) by 1.6 and 2.3 times respectively in pre adipocytes during differentiation. HRF 25 and 50 µg/mL also nonsignificantly reduced lipolysis which releases free fatty acids, a major contributing factor for insulin resistance. Activation of AMPK by phosphorylation has led to reduced gene and protein expression of adipogenic factors Peroxisome proliferator- activated receptor gamma (PPAR-γ), CCAT/enhancer binding protein alpha (C/EBPα), Sterol regulatory element- binding protein-1c (SREBP-1c) and their targets Fatty acid binding protein 4 (FABP4), Fatty acid synthase (FAS), Perilipin and enhanced Adiponectin expression. Treatment with HRF 25 and 50 µg/mL also resulted in inactivation of Acetyl-CoA carboxylase (ACC) by enhancing ACC phosphorylation, which reduced the levels of malonyl-CoA an allosteric inhibitor of carnitine palmitoyl transferase 1 (CPT1). Enhanced CPT1 levels causes induction of fatty acid β- oxidation. Effects of HRF were nullified in the presence of AMPK antagonist dorsomorphin.

Conclusion: In summary, HRF treatments reduced adipogenesis, enhanced factors regulating fatty acid oxidation and this is mediated by AMPK activation. The results conclusively showed anti-obesity potential of HRF and it might be helpful in treatment of associated complications.

Keywords: 3T3-L; AMPK; Adipogenesis; Hibiscus rosa sinensis; Lipolysis.