Научная статья на тему 'PPAR gamma Agonists – Plant Compounds'

PPAR gamma Agonists – Plant Compounds Текст научной статьи по специальности «Биологические науки»

CC BY
46
12
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
PPAR gamma / type 2 diabetes / natural products

Аннотация научной статьи по биологическим наукам, автор научной работы — Anil Kumar Moola, Shobana G., C. Janani

The nuclear receptor peroxisome proliferator activated receptor-gamma (PPARg) controls adipocyte differentiation, as well as lipid metabolism and insulin sensitivity. As a result, PPARg is a promising candidate gene for a variety of human diseases, such as obesity and type 2 diabetes. Hyperglycemia associated with the metabolic syndrome and type 2 diabetes is treated with agonists of the nuclear receptor PPARg. The PPARg agonists of the thiazolidinedione type, despite being effective in normalizing blood glucose levels, have serious side effects, making the discovery of novel ligands extremely important. Natural products have historically proven to be a promising source of structures for drug development, and recent research has focused on the PPARg-activating potential of a wide range of natural products derived from traditionally used medicinal plants or dietary sources. This review highlights recent studies that have advanced our understanding of this receptor's role in metabolism, with a focus on the effects of compounds in the plant.

i Надоели баннеры? Вы всегда можете отключить рекламу.
iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.
i Надоели баннеры? Вы всегда можете отключить рекламу.

Текст научной работы на тему «PPAR gamma Agonists – Plant Compounds»

Journal of Stress Physiology & Biochemistry, Vol. 19, No. 4, 2023, pp. 163-177 ISSN 1997-0838 Original Text Copyright © 2023 by Moola, Shobana and Janani

REVIEW

PPAR gamma Agonists - Plant Compounds

Anil Kumar Moola 1, Shobana G. 2 and C. Janani 3*

1 Department of Biotechnology, Aditya Degree College, Lakshmi Narayana Nagar, Kakinada, Andhra Pradesh, India - 533 003

2 Department of Biochemistry, Srimad Andavan Arts and Science College, Tiruchirappalli,. Tamil Nadu, India - 622 005

3 Department of Biotechnology, Srimad Andavan Arts and Science College, Tiruchirappalli,. Tamil Nadu, India - 622 005

*E-Mail: janabio. net@gmail.com

Received August 17, 2023

The nuclear receptor peroxisome proliferator activated receptor-gamma (PPARg) controls adipocyte differentiation, as well as lipid metabolism and insulin sensitivity. As a result, PPARg is a promising candidate gene for a variety of human diseases, such as obesity and type 2 diabetes. Hyperglycemia associated with the metabolic syndrome and type 2 diabetes is treated with agonists of the nuclear receptor PPARg. The PPARg agonists of the thiazolidinedione type, despite being effective in normalizing blood glucose levels, have serious side effects, making the discovery of novel ligands extremely important . Natural products have historically proven to be a promising source of structures for drug development, and recent research has focused on the PPARg-activating potential of a wide range of natural products derived from traditionally used medicinal plants or dietary sources. This review highlights recent studies that have advanced our understanding of this receptor's role in metabolism, with a focus on the effects of compounds in the plant

Key words: PPAR gamma, type 2 diabetes, natural products

OPEN

9

ACCESS

Diabetes is a chronic disease that affects a large number of people around the world. It happens when the pancreas doesn't produce enough insulin or when the body can't use the insulin it does produce effectively. Many metabolic changes (obesity, lipid profile, and hypertension) are involved in the etiology of the disease, all of which have a variable impact on the disease's progression. Having the ability to genetically influence diabetes will greatly aid in the proper production of therapeutics and, eventually, the prevention of disease complications, progression, and development. The most common type of diabetes, Type 2 Diabetes Mellitus (T2D), is a complex metabolic disorder that affects 90 percent of diabetic patients worldwide.

Diabetes is a metabolic disease that has grown in importance in modern society as a result of the serious long-term health complications it causes. Type 2 diabetes mellitus (T2DM) has become a global epidemic in the twenty-first century, despite significant advancements in the understanding and management of this complex, multifactorial disease. Type 2 diabetes mellitus (T2DM) is the most common type of diabetes, accounting for over 80% of all cases (Berg et al., 2002; Mlinar et al., 2007; Coman et al., 2012). Disturbances in glucose metabolism are major contributors to diabetes. The hormone responsible for glucose homeostasis is insulin, which is released by pancreatic -cells (Garrett and Grisham, 1999; Sesti, 2006). Insulin stimulates the uptake of glucose from the circulatory system by hepatocytes, myocytes, and adipocytes. Depending on the situation, glucose can be used as an energy source through glycolysis or stored as glycogen in muscle or liver cells. Insulin resistance is defined as the inability of cells to respond to normal levels of circulating insulin (Berg et al., 2002), resulting in the onset of the disease.

Moderate wine intake has been correlated with decrease incidences of cardiovascular sicknesses,

with the aid of using fueloline chromatography and mass spectroscopy with inside the maximum lively wine division. They had an similarity to PPARy much resembling that of the standard pharmaceutical agent rosiglitazone (Zoechling et al., 2011).

Natural compounds may be viable alternatives or complements to currently available treatments for diabetes. They may even lower the disease's risk. A positive aspect is that large amounts can be consumed in everyday diets. A large number of plants and natural biomolecules have been studied for their anti-diabetic properties in the literature. Plants, for example, have been used to prevent diabetes-related conditions since antiquity (Soumyanath, 2006). The mechanism is frequently not fully understood, but more research is being done to better understand the mechanisms of action of various plants and natural compounds. The role of medicinal plants and their active constituents for anti-diabetic agents is discussed in this study. The importance of diabetes management, reducing risks, and using anti-diabetic drugs was also stressed in this research. These extracts/compounds' detailed action mechanisms for their actions are also identified.

Antioxidant Effects in plants

A variety of phytochemicals (phenols, terpenoids, nitrogen-containing alkaloids, and sulphur-containing compounds) present in plants have been linked to anti-diabetic properties (Gothai et al., 2016). Phenolic compounds have been linked to changes in inflammatory behavior (CRP, IL-6, IL-1, and TNF-), transpiration factor enzymes (NF-B, PPAR), and genes associated with T2DM (Anuradha 2013). Ginger (Zingiber officinale Roscoe) remains used as an essential cooking spice and natural remedy across the world. Gingerols, the main smelly additives of ginger, are recognised to enhance diabetes, together with the impact of enhancement towards insulin sensitivity. (Priya et al., 2011)

Researchers have looked at the antioxidant and anti-diabetic effects of various plant sections (Kim et al., 2011; Priya et al., 2011; Unuofin, et al., 2018; Unuofin and Lebelo, 2020). Such antioxidants found in the human body, such as glutathione and thioredoxin, mop up ROS

inflammation, and metabolic sicknesses which includes kind 2 diabetes, obesity, and excessive pressure in blood. We studied binding of ligands from exceptional wines to the peroxisome proliferator-activated receptor y (PPARy), a key component in glucose and lipid metabolism. Ellagic acid and epicatechin gallate (ECG) had been diagnosed

by donating reducing counterparts to free radicals in the shape of a hydrogen atom or an electron, rendering them less dangerous in the body environment. In relation to T2DM therapy, some plant-derived compounds have been ascribed the following properties: stimulate the ERK1/2 and AMPK pathways (Yang etal., 2009; Ohno etal., 2013; Ahad et al., 2014). downregulate COX-2 gene expression, resulting in enhanced proinflammatory mediator liberation (Prabhakar, et al., 2013; Guo, et al., 2014) Increase glucose resistance and insulin sensitivity (Choi, et al., 1991; Tsai, et al., 2012); reduce inflammatory cell influx (Li et al., 2009); lower serum levels of proinflammatory cytokines IL-1, IL-6, and TNF-; inhibit NF-B pathway activation (Ahad et al., 2014) and repress macrophage chemostatic protein (MCP-1) and ICAM expression (Kumar et al., 2013) Figure 1 illustrates the possible antioxidant activity of plant secondary metabolites in the T2DM pathway caused by oxidative stress Via ROS/RNS, oxidative stress findings in diabetes were affected with Insulin resistance.

Antioxidants present in natural phytocompounds have received a lot of attention recently, and they're already being used therapeutically to clean up reactive organisms and avoid oxidative stress-related diabetes. Insulin tolerance, beta cell dysfunction, and insulin secretion are all caused by oxidative stress in diabetics, which can be modulated by phytocompounds with high antioxidant capacity by controlling blood sugar levels or attenuating at least one of the mechanisms linked to insulin resistance: beta cell activity, glucose (re)absorption, and incretin-related pathways (Unuofin and Lebelo 2020).

In a cell culture model of rat adipocytes the treatment of anthocyanins PPAR gamma and target adipocyte specific genes (LPL, aP2, and UCP2) were significantly up-regulated. Leptin and adiponectin as well as their mRNA levels were also increased by anthocyanins resulting from the increased phosphorylated MAPK. The mechanisms of action of anthocyanins in the amelioration of obesity can be mediated by upregulation of the thermogenic mithocondrial uncoupling protein 2 (UCP-2) and the lipolytic enzyme hormone sensitive lipase (HSL) as well as by down-regulation of the nuclear factor plasminogen activator inhibitor-1 (PAI-1) (Jayaprakasam et al., 2005). Some of the previous findings have been

also discovered in human adipocytes treated with anthocyanins (Tsuda et al., 2006). Further studies using a diabetic mouse model KK-Ay-mice have shown similar differences after anthocyanin and cyanidin- 3-glucoside administration. Gene expression of TNF-a and MCP-1 in mesenteric WAT was decreased and GLUT-4 increased, while a novel potential target gene retinol binding protein-4 was significantly decreased by 2 g/kg anthocyanin. in diet. The anthocyanin treatment enhanced the energy expenditure related genes UCP-2 and adiponectin and downregulation of PAI-1 that is induced by IL-6 in obese subjects, which suggests that also anti-inflammatory mechanisms of anthocyanins are involved (Moskaug et al., 2004; Abahusian et al., 1993)

Following food intake, lipid-derived ligands such as unsaturated fatty acids bind to this nuclear receptor, which causes the expression of a large number of genes involved in metabolism. PPARs have been shown to interact with structurally unrelated natural products such as flavonoids, polyphenols (e.g., resveratrol), and organic acids such as punicic acid or abscisic acid in micromolar concentrations (Huang et al., 2005; Hontecillas et al., 2009; Guri et al., 2007). However, due to interactions with a number of other proteins, these molecules do not appear to have clear beneficial molecular or physiological in vivo effects, making further development of these compounds difficult. The antidiabetic thiazolidinediones (TZDs), which include the commonly used medication rosiglitazone (Avandia), stimulate PPAR powerfully. These PPAR activators have recently come under fire due to negative clinical side effects (Anonymous 2010) such as weight gain and other disorders (Rosen 2010).

Nuclear Receptor (PPAR) within the broader nuclear receptor superfamily, PPAR is the third member of a subdivision that also includes PPAR and PPAR (Willson et al., 2000). The name PPAR comes from the discovery that a structurally diverse array of compounds that activate PPAR, the family's first member to be cloned, also increases the size and number of peroxisomes in rodent liver (Issemann & Green, 1990). The fibrate class of lipid-lowering drugs has been shown to target PPAR, which is highly expressed in the liver, kidney, heart, and skeletal muscle. In humans, however, these agents do not promote the proliferation of peroxisomes (Bentley et al.,

1993; Ashby et al., 1994). Although recent studies have suggested that it may be an important regulator of cholesterol trafficking in macrophages, little is known about the physiological role(s) of PPAR, which is expressed in many tissues (Braissant et al., 1996).

In the decade since it was first cloned (Zhu et al., 1993), however, an extensive body of literature relating to the biology of PPAR has emerged. Three mRNA isoforms are produced by different promoter usage and alternate splicing of the gene: PPAR1 and PPAR3 mRNA encode the same protein product; the PPAR2 isoform has an additional 28 amino acids at its N-terminus. PPAR1 is widely expressed, albeit at low levels, in adipose tissue, whereas PPAR2 and PPAR3 are highly expressed. Indeed, the receptor is important for fat cell differentiation, as it promotes the formation of mature lipid-laden adipocytes by inducing the expression of adipocyte-specific genes and inducing the expression of adipocyte-specific genes (Tontonoz et al.1995; Ntambi and Young-Cheul 2000; Janani et al., 2019).

Recent studies with mice genetically engineered to lack PPAR have confirmed its critical role in the development of both white and brown adipocytes in vivo (Barak et al., 1999; Kubota et al., 1999; Rosen et al., 1999; Janani and Ranjitha Kumari 2017), and clinical phenotypes of patients with PPAR gene mutations suggest a similar role in the regulation of human adipose tissue mass (Chatterjee, 2000; Agarwal & Garg, 2002; Hegele et al., 2002).

Role in Type II Diabetes Mellitus

The thiazolidinediones (TZDs) are synthetic compounds that can normalize elevated plasma glucose levels in obese, diabetic rodents and may be efficacious therapeutic agents for the treatment of noninsulin-dependent diabetes mellitus (Christensen et al., 2009)

Thiazolidinediones are insulin sensitizing drugs that target the peroxisome proliferator-activated receptor (PPAR) y. An n-hexane extract of the flowers of Echinacea purpurea was found to activate PPARy without stimulating adipocyte differentiation. Bioassay-guided fractionations yielded five alkamides, of which one was new, and three fatty acids that all activated PPARy. The new alkamide hexadeca-2E,9Z,12Z,14E-tetraenoic acid isobutylamide

(5) was identified by analysis of spectroscopic data and found to activate PPARy with no concurrent stimulation of adipocyte differentiation. Compound five was further shown to increase insulin-stimulated glucose uptake. The data suggest that flowers of E. purpurea contain compounds with potential to manage insulin resistance and type 2 diabetes (Christensen et al., 2009).

The use of TZDs to treat type-2 diabetes mellitus is complicated by systemic fluid retention. Guan et al. (2005) found that treatment of mice with amiloride, a collecting duct-specific diuretic, reversed the enhanced renal Na+ absorption, edema, and water weight gain caused by TZDs. Deletion of Pparg in mouse collecting duct blocked TZD-induced weight gain, decreased renal Na+ avidity, and increased plasma aldosterone. Treatment of cultured mouse collecting ducts with TZDs increased amiloride-sensitive Na+ absorption and Scnn1g (600761) mRNA expression through a Pparg-dependent pathway. Guan et al. (2005) concluded that SCNN1G is a PPARG target gene in the collecting duct and that activation of this pathway mediates fluid retention associated with TZDs.

Choi et al. (2010) showed that obesity induced in mice by high fat feeding activates the protein kinase CDK5 (123831) in adipose tissues. This results in phosphorylation of the nuclear receptor PPARG, a dominant regulator of adipogenesis and fat cell gene expression, at ser273. This modification of PPARG does not alter its adipogenic capacity, but leads to dysregulation of a large number of genes whose expression is altered in obesity, including a reduction in the expression of the insulin-sensitizing adipokine adiponectin (605441). The phosphorylation of PPARG by CDK5 is blocked by antidiabetic PPARG ligands such as rosiglitazone and MRL24. This inhibition works both in vitro and vivo, and is completely independent of classic receptor transcriptional agonism. Similarly, inhibition of PPARG phosphorylation in obese patients by rosiglitazone was very tightly associated with the antidiabetic effects of this drug. Choi et al. (2010) concluded that these results suggested that CDK5-mediated phosphorylation of PPARG may be involved in the pathogenesis of insulin resistance and presented an opportunity for development of an improved generation of antidiabetic drugs through PPARG.

Choi et al. (2011) described novel synthetic compounds that have a unique mode of binding to PPAR-gamma, completely lack classic transcriptional agonism, and block the Cdk5-mediated phosphorylation in cultured adipocytes and in insulin-resistant mice. Moreover, one such compound, SR1664, has potent antidiabetic activity without causing the fluid retention and weight gain that are serious side effects of many of the PPAR-gamma drugs. Also, unlike TZDs, SR1664 does not interfere with bone formation in culture. Choi et al. (2011) concluded that new classes of antidiabetes drugs can be developed by specifically targeting the Cdk5-mediated phosphorylation of PPAR-gamma. Dutchak et al. (2012) reported that FGF21 (609436) is an inducible, fed-state autocrine factor in adipose tissue that functions in a feed-forward loop to regulate the activity of PPAR-gamma. FGF21 knockout (KO) mice displayed defects in PPAR-gamma signaling including decreased body fat and attenuation of PPAR-gamma-dependent gene expression. Moreover, FGF21-KO mice were refractory to both the beneficial insulin-sensitizing effects and the detrimental weight d edema side effects of the PPAR-gamma agonist rosiglitazone. A9-tetrahydrocannabinol (THC), reasons acute vasorelaxation in diverse arteries. Here we display for the primary time that THC additionally reasons slowly growing vasorelaxation thru activation of peroxisome proliferator-activated receptors gamma (PPARy). In vitro, THC (10 MM) triggered time-established vasorelaxation of rat remoted arteries. Time-established vasorelaxation to THC become just like that produced with the aid of using the PPARy agonist rosiglitazone and become inhibited with the aid of using the PPARy antagonist GW9662 (1 mM), however now no longer the cannabinoid CB1 receptor antagonist AM251 (1 mM). Time-established vasorelaxation to THC calls for an intact endothelium, nitric oxide, manufacturing of hydrogen peroxide, and de novo protein synthesis. This lack of characteristic in FGF21-KO mice become coincident with a marked boom withinside the sumoylation of PPAR-gamma, which reduces its transcriptional activity. Adding lower back FGF21 averted sumoylation and restored PPAR-gamma activity. Dutchak et al. (2012) concluded that FGF21 is a key mediator of the physiologic and pharmacologic movements of PPAR-gamma.

Jonker et al. (2012) diagnosed FGF1 (131220) as a important transducer withinside the manner of metabolic homeostasis via dinner party or famine in mice. Jonker et al. (2012) connected the law of FGF1 to the nuclear receptor PPAR-gamma. FGF1 is the prototype of the 22-member FGF own circle of relatives of proteins and has been implicated in a number of physiologic processes, consisting of development, wound healing, and cardiovascular changes. Surprisingly, FGF1 knockout mice displayed no extensive phenotype beneathneath wellknown laboratory conditions. Jonker et al. (2012) confirmed that FGF1 changed into incredibly prompted in adipose tissue in reaction to a high-fats weight-reduction plan and that mice missing FGF1 advanced an competitive diabetic phenotype coupled to aberrant adipose enlargement whilst challenged with a high-fats weight-reduction plan. Further evaluation of adipose depots in FGF1-poor mice discovered a couple of histopathologies withinside the vasculature network, an accentuated inflammatory reaction, aberrant adipocyte length distribution, and ectopic expression of pancreatic lipases. On withdrawal of the high-fats weight-reduction plan, this infected adipose tissue did not well resolve, ensuing in significant fats necrosis. In phrases of mechanisms, Jonker et al. (2012) confirmed that adipose induction of FGF1 withinside the fed kingdom is regulated via way of means of PPAR-gamma appearing via an evolutionarily conserved promoter-proximal PPAR reaction element (PPRE) withinside the FGF1 gene. The discovery of a phenotype for the FGF1 knockout mouse mounted the PPAR-gamma-FGF1 axis as important for retaining metabolic homeostasis and insulin sensitization.

These fundamental insights have culminated with TZDs being recently approved for use in the management of type 2 diabetes. Troglitazone, the first TZD in widespread clinical use, was withdrawn following concerns regarding its hepatotoxicity (Watkins & Whitcomb, 1998); subsequent studies have suggested, however, that this potentially fatal idiosyncratic adverse event is unlikely to be a class effect, but rather reflects the generation of a specific metabolite unique to troglitazone (Willson et al., 2000).

Although originally synthesized as derivatives of clofibrate, unexpectedly the TZDs were found to exhibit

insulin-sensitizing actions in rodent models of T2DM as far back as the early 1980s, a finding that was later confirmed in man (1999). It is only relatively recently, however, that the molecular basis for this action has been elucidated. In the mid-1990s Kliewer and colleagues reported that TZDs function as selective high-affinity ligands for PPARy (Lehmann et al., 1995). Furthermore, the rank order of their potencies for receptor activation in vitro correlates closely with their glucose lowering activity in vivo (Berger et al., 1996; Willson et al., 1996). Taken together, these data strongly suggested PPARy to be the molecular target for the antidiabetic actions of the TZDs. The therapeutic efficacy of the currently available TZDs is, however, limited, and several side-effects have been reported (Schoonjans & Auwerx, 2000). Accordingly, selective non-TZD PPARy ligands (e.g. tyrosine agonists) have been developed and shown to exert potent antidiabetic effects in preclinical studies and early clinical trials (Fiedorek et al., 2000; Willson et al., 2000), whilst compounds that activate RXR (the heterodimeric partner for PPARy) also improve insulin sensitivity in vivo (Mukherjee et al., 1997), findings which support the contention that this receptor is indeed a key regulator of insulin action. Moreover, a unique chemically distinct PPARy ligand, FMOC-L-leucine (F-L-Leu) has been reported to promote differential cofactor recruitment leading to a modified pattern of target gene activation, which promotes insulin sensitization, yet with less adipogenic activity than other PPARy ligands (Rocchi et al., 2001), a finding which has obvious attractions from the therapeutic standpoint.

Therefore studies have focused on their heterozygous littermates, who appear to exhibit greater insulin sensitivity in hyperinsulinaemic euglycaemic clamp studies (Miles et al., 2000) and are protected against the development of insulin resistance when subjected to high fat feeding (Kubota et al., 1999), as compared with their wild-type counterparts. At first glance these findings may appear counterintuitive, i.e. how can a reduction in PPARy activity in heterozygous null mice protect against insulin resistance when PPARy activators such as TZDs are potent insulin sensitizers. A reduction in PPARy activity, due to heterozygous PPARy deficiency, decreases the triglyceride content of white adipose tissue, skeletal

muscle and liver (possibly as a consequence of enhanced leptin expression and fatty acid combustion), thereby ameliorating high fat diet-induced obesity and insulin resistance. Moreover, although heterozygous PPARy deficiency and TZD treatment exert opposing effects on total white adipose tissue mass, the former also decreases lipogenesis, whereas the latter stimulates adipocyte differentiation and apoptosis, thereby both protecting against the development of adipocyte hypertrophy and, accordingly, alleviating insulin resistance (Yamauchi et al., 2001).

Additional proof of a key function for PPARy in figuring out human adiposity has been furnished with the aid of using the identity of 4 unrelated German topics in whom a missense mutation at codon 115 (Pro115Gln) in PPARy2 became observed to be related to extreme obesity (BMIs starting from 37 to forty seven kg/m2; Ristow et al., 1998). With the previous expertise that phosphorylation of PPARy2 at an adjoining serine residue (Ser114) had formerly been proven to lessen the capacity of the receptor to mediate adipocyte differentiation and lipid accumulation (Hu et al., 1996; Adams et al., 1997), the authors speculated that the Pro115Gln mutation would possibly impair such phosphorylation, main to a receptor with stronger transcriptional activity, adipogenic motion and consequently obesity. Indeed, overexpression of the mutant receptor in murine fibroblasts brought about multiplied differentiation of the cells into adipocytes and extra mobile accumulation of triglyceride whilst in comparison with the wild-kind receptor. Interestingly, the authors speculated that such an activating PPARy mutation is probably anticipated to be related to much less insulin resistance than could commonly be predicted for the diploma of obesity. In help of this contention, they located that overweight topics bearing the Pro115Gln mutation had decrease fasting serum insulin concentrations than different overweight topics of their cohort. However, it ought to be mentioned that 3 in their affected people had already evolved T2DM, and a greater specific estimate of insulin resistance and beta cellular characteristic could be required to verify this hypothesis. Identification of isosilybin a from milk thistle seeds as an agonist of peroxisome proliferator-activated receptor gamma. Likewise some of the plant secondary metabolites

acts as agonist of PPARg expression were listed in Table 1.

Table. 1. PPAR gamma agonists

S.N o Name of the plant Name of the Plant Family Name of the compound Mechanism of action of compound References

1. Cannabis sativa Cannabaceae Ajulemic acid Agonist Lee et al., 2009

Apigenin Chrysin Kaempferol Liang et al., 2001

2. Poria cocos Wolf Polyporaceae Dehydrotrametenoli c acid Agonist Sato et al., 2002

3. Glycyrrhiza uralensis Fisher Fabaceae Prenylflavonoid (Glycycoumarin, glycyrin, dehydroglyasperin C and dehydroglyasperin D Agonist Kuroda et al., 2003

4. Saururus chinensis Saururaceae Saurufuran Agonist Atanas et al., 2013

5. Notopterygium incisum Apiaceae Falcarindiol Agonist Hwang et al., 2002

6. Hydrangea macrophylla var. thunbergii Hydrangeaceae Hydrangeic acid Agonist Zhang et al., 2009

7. Amorpha fruticosa L. Fabaceae Amorfrutins Agonist Weidner et al., 2012

8. Astragalus membranaceusMoench Fabaceae Formononetin Agonist Shen et al., 2006

9. Bixa orellana L. Bixaceae Bixin and norbixin Agonist Takahashi et al., 2009

10. Camellia sinensis (L.) Kuntze Theaceae (-)-Catechin Agonist Shin et al., 2009

11. Cannabis sativaL. Cannabaceae A9- Agonist O'Sullivan et al.,

Tetrahydrocannabin ol 2005

12. Chromolaena odorata (L.) Asteraceae (9S,13R)-12-Oxo-phytodienoic acid Agonist Dat et al., 2009

13. Coix lacryma-jobi var. ma-yuen Poaceae Hydroxy unsaturated fatty acids Agonist Yokoi et al.,2009

14. Cornus alternifolia L.f. Cornaceae Kaempferol-3-O-jB-glucopyranoside Agonist He et al. ,2012

15. Commiphora mukul (Hook. ex Stocks) Burseraceae Commipheric acid Agonist Cornick et al., 2009

16. Cymbopogon citratus (DC.) Stapf Poaceae Citral Agonist Katsukawa et al., 2010

17. Echinacea purpurea (L.) Moench Asteraceae Alkamides Agonist Christensen et al., 2009

18. Elaeis guineensis Jacq. Arecaceae Tocotrienols Agonist Fang et al., 2010

19. Elephantopus scaber L. Asteraceae Deoxyelephantopin Agonist Zou et al.,2008

20. Epimedium elatum C. Morren & Decne. Berberidaceae) Acylated flavonol glycosides Agonist Tantry et al., 2012

21. Euonymus alatus(Thunb.) Siebold Celastraceae Kaempferol and quercetin Agonist Fang et al., 2010

22. Glycine max (L.) Merr. Fabaceae Genistein Agonist Dang et al., 2003

23. Glycyrrhiza glabra L. Fabaceae 5'-Formylglabridin, (2R,3R)-3,4',7- Agonist Kuroda et al., 2010

trihydroxy-3'-prenylflavane, echinatin, (3R)-

2',3',7-trihydroxy-4'-

methoxyisoflavan, kanzonol X, kanzonol W, shinpterocarpin, licoflavanone A, glabrol, shinflavanone, gancaonin L, glabrone

24. Glycyrrhiza inflata Batalin Fabaceae Licochalcone E Agonist Park et al., 2012

25. Glycyrrhiza uralensis Fisch. ex Fabaceae Flavonoids and 3- Agonist Kuroda et al.,

DC. arylcoumarins 2003

26. Limnocitrus littoralis (Miq.) Rutaceae Meranzin Agonist Do et al., 2007

Swingle

27. Magnolia officinalis Rehder & Magnoliaceae Magnolol Agonist Choi et

E.H. Wilson al., 2009

28. Momordica charantia L. Cucurbitaceae Cucurbitane-type Agonist Nhiem et al.,

triterpene glycoside 2012

29. Notopterygium incisum C.T. Ting ex H.T. Chang Apiaceae Polyacetylenes Agonist Atanasov et al., 2013

3G. Origanum vulgare L. Lamiaceae Biochanin A Agonist Mueller et al., 2008

31. Panax ginsengC.A. Mey. Araliaceae Ginsenoside 20(S)- Agonist Han et al., 2006

protopanaxatriol

32. Pseudolarix amabilis (J. Pinaceae Pseudolaric acid B Agonist Jaradat et al.,

Nelson) Rehder 2002

33. Robinia pseudoacaciavar. umbraculifer DC. Fabaceae Amorphastilbol Agonist Kim et al. ,2012

34. Pueraria thomsonii Benth. Fabaceae Daidzein Agonist Shen et al.,

2006

35. Sambucus nigraL. Adoxaceae a-Linolenic acid, Agonist Christensen etal

linoleic acid, and naringenin .,2010

36. Silybum marianum (L.) Gaertn. Asteraceae Isosilybin A Agonist Pferschy-

Wenzig et al., 2014

37. Terminalia bellerica Roxb. Combretaceae Gallotannins Agonist Yang et al.,

2013

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

38. Thymus vulgarisL. Lamiaceae Carvacrol Agonist Hotta et al., 2010

39. Vitis vinifera L. Vitaceae Ellagic acid, Agonist Zoechling et al.,

epicatechin gallate 2011

40. Wolfiporia extensa (Peck) Polyporaceae Dehydrotrametenoli Agonist Sato et al., 2002

Ginns c acid

41. Zingiber officinaleRoscoe Zingiberaceae 6-Shogaol Agonist Isa et al., 2008

42. Pistacia lentiscus L. Anacardiaceae Oleanonic acid Agonist Petersen et al., 2011

43. Piper chaba Piperaceae Retrofractamide A Agonist Mourad et al., 2013

44. Kaempferia parviflora Zingiberaceae Methoxyflavonols Agonist Matsuda et al., 2011

45. Stevia rebaudiana Asteraceae Austroinulin Agonist Metibemu et

al., 2017

Reactive Oxygen Species Reactive Nitrogen Species

Endogenous Antioxidants like glutathione, lipoic acid, bilirubin, ferritin, superoxide dismutase. catalase. and glutathione peroxidase.

Oxidative Stress "Nitrosative Stress

i

Insulin resistance

Hyperglycemia

Exogenous Antioxidants like Tocopherol. Ascorbate. (5 Carotene, Flavonoids.

Type II Diabetes

Figure. 1. Potential of antioxidants in Type II Diabetes

CONCLUSION

The study of the reviewed information of the numerous PPAR gamma will become obvious that, despite the fact that those dealers are classed collectively in keeping with their important PPAR target, they have got very unique and on occasion conflicting scientific benefit and unfavorable occasion profiles. It is speculated that that is due to their unique residences and specificities for the unique PPAR receptors (every of which goals specific genes). The maximum placing is the conflicting kind 2 diabetes. In scientific practice, PPAR-a agonists, consisting of the fibrates, enhance dyslipidaemia, even as the PPAR-y agonists, consisting of the plant secondary metabolites will enhance insulin resistance and diabetes.

CONFLICT OF INTERESTS

The authors declare that they have no potential conflicts of interest.

REFERENCES

Abahusian MA, Wright J, Dickerson JWT and Vol EB (1993) Retinol, a-tocopherol and carotenoids in diabetes. Eur J Clin Nutr 53, 630-635.

Agarwal, A.K. and Garg, A. (2002) A novel heterozygous mutation in peroxisome proliferator-activated receptor-y gene in a patient with familial partial lipodystrophy. Journal of Clinical Endocrinological Metabolism, 87, 408-411.

Ahad, A. Ganai, A. A. Mujeeb, M. and Siddiqui, W. A. (2014) Chrysin, an anti-inflammatory molecule, abrogates renal dysfunction in type 2 diabetic rats. Toxicology and Applied Pharmacology, 279, 1, 1-7,.

Ahad, A. Mujeeb, M. Ahsan, H. and Siddiqui W. A. (2014). Prophylactic effect of baicalein against renal dysfunction in type 2 diabetic rats. Biochimie, 106, 101-110.

Anonymous (2010) Regulators restrict Avandia in the US and suspend it in the EU. Nat Rev Drug Discov 9, 828-828.

Anuradha, C. V. (2013). Phytochemicals targeting genes relevant for type 2 diabetes. Canadian Journal of Physiology and Pharmacology, 91, 6, 397-411.

Ashby J, Brady A, Elcombe CR, Elliott BM, Ishmael J, Odum J, Tugwood JD, Kettle S, Purchase IF. (1994). Mechanistically-based human hazard assessment of peroxisome proliferator-induced hepatocarcinogenesis. Hum Exp Toxicol 13 Suppl 2: S1-117.

Atanasov A.G., Blunder M., Fakhrudin N., Liu X., Noha S.M., Malainer C. 2013. Polyacetylenes from Notopterygium incisum--new selective partial agonists of peroxisome proliferator-activated receptor-gamma. PLoS One. 8, e61755.

Barak Y, Nelson MC, Ong ES, Jones YZ, Ruiz-Lozano P, Chien KR, Koder A, Evans RM (1999) PPARy is required for placental, cardiac, and adipose tissue development. Mol Cell 4, 585-595.

Bentley P, Calder I, Elcombe C, Grasso P, Stringer D, Wiegand HJ. (1993). Hepatic peroxisome proliferation in rodents and its significance for humans. Food Chem Toxicol 31, 857-907.

Berg JM, Tymoczko JL, Stryer L (2002). Biochemistry, 5th Ed. W. H. Freeman and Company, New York.

Braissant OL, Foufelle F, Scotto CH, Dau?a MI, Wahli WA. (1996) Differential expression of peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPAR-alpha,-beta, and-gamma in the adult rat. Endocrinology. 137(1), 354-66.

Chang, C. L. T. Lin, Y. Bartolome, A. P. Chen, Y. C. Chiu, S. C. and Yang, W. C. (2013) Herbal therapies for type 2 diabetes mellitus: chemistry, biology, and potential application of selected plants and compounds. Evidence-based Complementary and Alternative Medicine. 2013, 1-33.

Chatterjee, V.K.K. (2000) Resistance to thyroid hormone, and peroxisome proliferator activated receptor y resistance. Biochemistry Society of

Transactions, 29, 227-231.

Choi JH, Banks AS, Estall JL, Kajimura S, Boström P, Laznik D, Ruas JL, Chalmers MJ, Kamenecka TM, Blüher M, Griffin PR. (2010). Anti-diabetic drugs inhibit obesity-linked phosphorylation of PPARy by Cdk5. Nature. 466(7305), 451-6.

Choi JH, Banks AS, Kamenecka TM, Busby SA, Chalmers MJ, Kumar N, Kuruvilla DS, Shin Y, He Y, Bruning JB, Marciano DP. (2011) Antidiabetic actions of a non-agonist PPARy ligand blocking Cdk5-mediated phosphorylation. Nature. 477(7365), 477-81.

Choi, J. Yokozawa, T. and Oura, H. (1991). Improvement of hyperglycemia and hyperlipemia in streptozotocin-diabetic rats by a methanolic extract of Prunus davidiana stems and its main component, prunin. Planta Medica, 57, 3, 208- 211.

Choi S.S., Cha B.Y., Lee Y.S., Yonezawa T., Teruya T., Nagai K. (2009) Magnolol enhances adipocyte differentiation and glucose uptake in 3T3-L1 cells. Life Sci. 84, 908-914.

Christensen K.B., Petersen R.K., Kristiansen K., Christensen L.P. (2010) Identification of bioactive compounds from flowers of black elder (Sambucus nigra L.) that activate the human peroxisome proliferator-activated receptor (PPAR)

gamma. Phytother Res. 24(Suppl 2), S129-S132.

Christensen K.B., Petersen R.K., Petersen S., Kristiansen K., Christensen L.P. (2009). Activation of PPARgamma by metabolites from the flowers of purple coneflower (Echinacea purpurea) J Nat Prod. 72, 933-937.

Christensen KB, Petersen RK, Petersen S, Kristiansen K, Christensen LP. (2009) Activation of PPARy by metabolites from the flowers of purple coneflower (Echinacea purpurea). Journal of natural products. 72(5), 933-937.

Coman C, Rugina OD, Socaciu C. (2012). Plants and natural compounds with antidiabetic action. Notulae Botanicae Horti Agrobotanici Cluj-Napoca. 40(1), 314-325.

Cornick C.L., Strongitharm B.H., Sassano G., Rawlins C., Mayes A.E., Joseph A.N. (2009). Identification of a novel agonist of peroxisome proliferator-activated receptors alpha and gamma that may contribute to

the anti-diabetic activity of guggulipid in Lep(ob)/Lep(ob) mice. J Nutr Biochem. 20, 806-815.

Lowik C.W. (2003). Peroxisome proliferator-activated receptor gamma (PPARgamma) as a molecular target for the soy phytoestrogen genistein. J Biol Chem. 278, 962-967.

(2009). A peroxisome proliferator-activated receptor-gamma agonist and other constituents from Chromolaena odorata. Planta Medica. 75, 803-807.

Do Q.T., Lamy C., Renimel I., Sauvan N., Andre P., Himbert F. (2007). Reverse pharmacognosy: identifying biological properties for plants by means of their molecule constituents: application to meranzin. Planta Medica. 73, 1235-1240.

Dutchak PA, Katafuchi T, Bookout AL, Choi JH, Ruth TY, Mangelsdorf DJ, Kliewer SA. (2012). Fibroblast growth factor-21 regulates PPARy activity and the antidiabetic actions of thiazolidinediones. Cell.

Baumgartner L., Markt P. (2010). Computer-aided discovery, validation, and mechanistic characterization of novel neolignan activators of peroxisome proliferator-activated receptor gamma. Mol Pharmacol. 77, 559-566.

improve insulin sensitivity through activating peroxisome proliferator-activated receptors. Mol Nutr Food Res. 54,, 345-352.

Fiedorek, F.T., Wilson, G.G., Frith, L., Patel, J. & Abou-Donia, M. (2000) Monotherapy with G1262570, a tyrosine-based non-thiazolidinedione PPARy agonist, improves metabolic control in type 2 diabetes mellitus patients. Diabetes, 49 (Suppl. 1), Abstract 157-OR.

Garrett RH, Grisham CM (1997). Biochemistry, 2nd Ed. Harcourt Brace College Publishers, Orlando, Florida.

Gothai, S. Ganesan, P. Park, S Y.. Fakurazi, S. Choi, D. K. and Arulselvan, P. (2016). Natural phyto-bioactive compounds for the treatment of type 2 diabetes: inflammation as a target. Nutrients,. 8. 8, 461.

Guan, Y. Hao, C. Cha D. R. (2005). Thiazolidinediones

expand body fluid volume through PPAR/ stimulation of ENaC-mediated renal salt absorption. Nature Medicine, 11, 8, 861-866.

Guo, R. Liu, B. Wang, K. S. Zhou, W. Li, and Y. Xu, (2014). Resveratrol ameliorates diabetic vascular inflammation and macrophage infiltration in db/db mice by inhibiting the NF-kB pathway. Diabetes and Vascular Disease Research, 11, 2,. 92-102.

Guri AJ, Hontecillas R, Si H, Liu D, Bassaganya-Riera J (2007) Dietary abscisic acid ameliorates glucose tolerance and obesity-related inflammation in db/db mice fed high-fat diets. Clin Nutr 26, 107-116.

Han K.L., Jung M.H., Sohn J.H., Hwang J.K. (2006) Ginsenoside 20S-protopanaxatriol (PPT) activates peroxisome proliferator-activated receptor gamma (PPARgamma) in 3T3-L1 adipocytes. Biol Pharmaceut Bull. 29, 110-113.

Hegele, R.A., Henian, C., Frankowski, C., Mathews, S.T. & Leff, T. (2002) PPARG F388L, a transactivation-deficient mutant, in familial partial lipodystrophy. Diabetes, 51, 3586-3590.

Hontecillas R, O'Shea M, Einerhand A, Diguardo M, Bassaganya-Riera J (2009) Activation of PPAR gamma and alpha by punicic acid ameliorates glucose tolerance and suppresses obesity-related inflammation. J Am Coll Nutr 28, 184-195.

Hotta M., Nakata R., Katsukawa M., Hori K., Takahashi S., Inoue H. (2010) Carvacrol, a component of thyme oil, activates PPARalpha and gamma and suppresses COX-2 expression. J Lipid Res. 51, 132139.

Huang TH, Kota BP, Razmovski V, Roufogalis BD (2005) Herbal or natural medicines as modulators of peroxisome proliferator-activated receptors and related nuclear receptors for therapy of metabolic syndrome. Basic Clin Pharmacol 96, 3-14.

Hwang B.Y., Lee J.H., Nam J.B., Kim H.S., Hong Y.S., Lee J.J. (2002) Two new furanoditerpenes from Saururus chinenesis and their effects on the activation of peroxisome proliferator-activated receptor gamma. J Nat Prod. 65, 616-617.

Hwang, B. Y., J. H. Lee, J. B. Nam, H. S. Kim, Y. S. Hong & J. J. Lee: (2002). Two new furanoditerpenes

Dang Z.C., Audinot V., Papapoulos S.E., Boutin J.A.,

Dat N.T., Lee K., Hong Y.S., Kim Y.H., Minh C.V., Lee J.J.

148(3), 556-567. Fakhrudin N., Ladurner A., Atanasov A.G., Heiss E.H.,

Fang F., Kang Z., Wong C. (2010). Vitamin E tocotrienols

from Saururus chinenesis and their effects on the activation of peroxisome proliferator-activated receptor gamma. J. Nat. Prod. , 65, 616- 617.

He Y.Q., Ma G.Y., Peng J.N., Ma Z.Y., Hamann M.T. (2012) Liver X receptor and peroxisome proliferator-activated receptor agonist from Cornus alternifolia. Biochim Biophys Acta. 1820, 1021-1026.

Kawada T. (2008) 6-Shogaol and 6-gingerol, the pungent of ginger, inhibit TNF-alpha mediated downregulation of adiponectin expression via different mechanisms in 3T3-L1 adipocytes. Biochem Biophys Res Commun. 373, 429-434.

Issemann I, Green S. (1990). Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature. 347(6294), 645-50.

Janani C and Kumari BR. (2015). PPAR gamma gene-a review. Diabetes & Metabolic Syndrome: Clinical Research & Reviews. 1;9(1), 46-50.

Janani C, and Kumari BR. (2017). Antidiabetic activity of methanolic leaves extracts of transformed soybean in 3T3-L1 preadipocytes cell line. Int J Pharm Sci Res. 1;8(11), 4732-4744.

Janani C, Sundararajan B, Kumari BR. (2019) Construction and transformation of peroxisome proliferator activated receptor gamma (RnPPARy) gene using Agrobacterium tumefaciens into Glycine max L. Merr. Gene Reports. 16, 100427.

Jaradat M.S., Noonan D.J., Wu B., Avery M.A., Feller D.R. (2002) Pseudolaric acid analogs as a new class of peroxisome proliferator-activated receptor agonists. Planta Medica. 68, 667-671.

Jayaprakasam B, Vareed SK, Olson LK and Nair MG (2005) Insulin secretion by bioactive anthocyanins and anthocyanidins present in fruits. J Agric Food Chem 53, 28-31.

Jonker JW, Suh JM, Atkins AR, Ahmadian M, Li P, Whyte J, He M, Juguilon H, Yin YQ, Phillips CT, Ruth TY. PS21-100. (2012). A PPAR-FGF1 axis is required for adaptive adipose remodelling and metabolic homeostasis. Nederlands Tijdschrift voor Diabetologie. 10(3), 170-170.

Katsukawa M., Nakata R., Takizawa Y., Hori K., Takahashi S., Inoue H. (2010). Citral, a component of lemongrass oil, activates PPARalpha and gamma and suppresses COX-2 expression. Biochim Biophys Acta. 1801, 1214-1220.

Kim T., Lee W., Jeong K.H., Song J.H., Park S.H., Choi P. (2012) Total synthesis and dual PPARalpha/gamma agonist effects of amorphastilbol and its synthetic derivatives. Bioorg Med Chem Lett. 22, 4122-4126.

Kim, S. Shin, B. C. Lee, M. S. Lee, H. and Ernst, E. (2011) "Red ginseng for type 2 diabetes mellitus: a systematic review of randomized controlled trials," Chinese Journal of Integrative Medicine, 17. 12, 937944.

Kubota N et al. (1999) PPARy mediates high-fat diet-induced adipocyte hypertrophy and insulin resistance. Mol Cell 4, 597-609.

Kumar, B. Gupta, S. K. Srinivasan B. P. (2013). Hesperetin rescues retinal oxidative stress, neuroinflammation and apoptosis in diabetic rats. Microvascular Research, 87, 65-74.

Kuroda M., Mimaki Y., Honda S., Tanaka H., Yokota S., Mae T. (2010). Phenolics from Glycyrrhiza glabra roots and their PPAR-gamma ligand-binding activity. Bioorg Med Chem. 18, 962-970.

Kuroda, M., Y. Mimaki, Y. Sashida, T. Mae, H. Kishida, T. Nishiyama, M. Tsukagawa, E. Konishi, K. Takahashi, T. Kawada, K. Nakagawa M. (2003). Kitahara: Phenolics with PPAR-gamma ligand-binding activity obtained from licorice (Glycyrrhiza uralensis roots) and ameliorative effects of glycyrin on genetically diabetic KK-A(y) mice. Bioorg. Med. Chem. Lett. 13, 4267- 4272.

Lee, C. H., Olson P., Evans R. M. (2003). Minireview: lipid metabolism, metabolic diseases, and peroxisome proliferator activated receptors. Endocrinology, 144, 2201- 2207.

Li, J. J. Lee, S. H. Kim D. K. (2009). Colchicine attenuates inflammatory cell infiltration and extracellular matrix accumulation in diabetic nephropathy. American Journal of Physiology-Renal Physiology, 297, 1, F200-F209,.

Liang, Y. C., Tsai S. H., Tsai D. C., Lin Shiau S. Y., Lin J.

Isa Y., Miyakawa Y., Yanagisawa M., Goto T., Kang M.S.,

K. (2001). Suppression of inducible cyclooxygenase and nitric oxide synthase through activation of peroxisome proliferator activated receptor gamma by flavonoids in mouse macrophages. FEBS Lett., 496, 12- 18.

Matsuda H, Kogami Y, Nakamura S, Sugiyama T, Ueno T, Yoshikawa M. (2011) Structural requirements of flavonoids for the adipogenesis of 3T3-L1 cells. Bioorg. Med. Chem., 19, 2835-2841.

Metibemu DS, Akinloye OA, Akinbamiwa BA, et al. (2017) Austroinulin from Stevia rebaudiana a Potential Agonist of the Peroxisome Proliferator-activated Receptor (PPARy): an in-silico Study. J Syst Biol Proteome Res., 1(1), 10-15

Miles, P.D., Barak, Y., He, W., Evans, R.M. & Olefsky, J.M. (2000) Improved insulin-sensitivity in mice heterozygous for PPARy deficiency. Journal of Clinical Investigations, 105, 287-292.

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

Mlinar B, Marc J, Janez A, Pfeifer M (2007). Molecular mechanisms of insulin resistance and associated diseases. Clin Chim Acta 375:20-35

Moskaug JO, Carlsen H, Myhrstad M & Blomhoff R (2004) Molecular imaging of the biological effects of quercetin and quercetin-rich foods. Mech Ageing Dev 125, 315-324.

Mourad AA, Nakamura S, Ueno T, Minami T, Yagi T, Yasue H, Komatsu R, Yoshikawa M, Taye AM, El-Moselhy MA, Khalifa MM, Matsuda H. (2013). Adipogenetic effects of retrofractamide A derivatives in 3T3-L1 cells. Bioorg. Med. Chem. Lett., 23, 4813-

A.R., Jungbauer A. (2008). Oregano: a source for peroxisome proliferator-activated receptor gamma antagonists. J Agr Food Chem. 56, 11621-11630.

Mukherjee, R., Davies, P.J.A., Crombie, D.L., Dischoff, E.D., Cesario, R.M., Jow, L., Hamann, L.G., Mondon, C.E., Nadzan, A.M., Paterniti, J.J.R. & Heyman, R.A. (1997) Sensitization of diabetic and obese mice to insulin by retinoid X receptor agonists. Nature, 386,

Minh C.V. (2012). Inhibition of nuclear transcription

factor-kappaB and activation of peroxisome proliferator-activated receptors in HepG2 cells by cucurbitane-type triterpene glycosides from Momordica charantia. J Med Food. 15, 369-377.

Ntambi JM, Young-Cheul K. (2000) Adipocyte differentiation and gene expression. The Journal of nutrition. 130(12), 3122S-6S.

Ohno, M. Shibata, C. Kishikawa T. (2013). The flavonoid apigenin improves glucose tolerance through inhibition of microRNA maturation in miRNA103 transgenic mice. Scientific Reports, 3, 1, 2013.

O'Sullivan S.E., Tarling E.J., Bennett A.J., Kendall D.A., Randall M.D. (2005). Novel time-dependent vascular actions of Delta9-tetrahydrocannabinol mediated by peroxisome proliferator-activated receptor gamma. Biochem Biophys Res Commun. 337, 824831.

Park H.G., Bak E.J., Woo G.H., Kim J.M. , Quan Z., Yoon

H.K. (2012). Licochalcone E has an antidiabetic

effect. J Nutr Biochem. 23, 759-767.

Petersen R.K., Christensen K.B., Assimopoulou A.N.,

Frette X., Papageorgiou V.P., Kristiansen K. (2011) Pharmacophore-driven identification of PPARgamma agonists from natural sources. J Comput-Aid Mol Des. 25, 107-116.

Pferschy-Wenzig E.M., Atanasov A.G., Malainer C., Noha S.M., Kunert O., Schuster D. (2014) Identification of isosilybin a from milk thistle seeds as an agonist of peroxisome proliferator-activated receptor gamma. J Nat Prod. 77, 842-847.

Prabhakar, O. (2013) Cerebroprotective effect of resveratrol through antioxidant and anti-inflammatory effects in diabetic rats. Naunyn-Schmiedeberg's Archives of Pharmacology, 386, 8, 705-710.

Priya RM, Padmakumari KP, Sankarikutty B, Lijo Cherian O, Nisha VM, Raghu KG. (2011). Inhibitory potential of ginger extracts against enzymes linked to type 2 diabetes, inflammation and induced oxidative stress. International Journal of Food Sciences and Nutrition. 62(2), 106-110.

Rau O., Wurglics M., Paulke A., Zitzkowski J., Meindl N., Bock A. (2006) Carnosic acid and carnosol, phenolic diterpene compounds of the labiate herbs rosemary

4816.

Mueller M., Lukas B., Novak J., Simoncini T., Genazzani

407-410.

Nhiem N.X., Yen P.H., Ngan N.T., Quang T.H., Kiem P.V.,

and sage, are activators of the human peroxisome proliferator-activated receptor gamma. Planta Medica. 72, 881-887.

Rocchi, S., Picard, F., Vamecq, J., Gelman, L., Potier, N., Zeyer, D., Dubuquoy, L., Bac, P., Champy, M.-F., Plunket, K.D., Leesnitzer, L.M., Blanchard, S.G., Desreumaux, P., Moras, D., Renaud, J.-P. & Auwerx, J. (2001) A unique PPARy ligand with potent insulin-sensitising yet weak adipogenic activity. Molecular Cell, 8, 737-747.

Rosen CJ (2010) Revisiting the rosiglitazone story-lessons learned. N Engl J Med 363, 803-806

Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, Spiegelman BM, Mortensen RM (1999) PPARy is required for the differentiation of adipose tissue in vivo and in vitro. Mol Cell 4, 611-

Tanaka K. (2002) Dehydrotrametenolic acid induces preadipocyte differentiation and sensitizes animal models of noninsulin-dependent diabetes mellitus to insulin. Biol Pharmaceut Bull. 25, 81-86.

Sato, M., Tai T., Nunoura Y., Yajima Y., Kawashima S. & Tanaka K. (2002). Dehydrotrametenolic acid induces preadipocyte differentiation and sensitizes animal models of noninsulin- dependent diabetes mellitus to insulin. Biol. Pharm. Bull., 25, 81- 86.

Schoonjans K, Auwerx J. (2000). Thiazolidinediones: an update. The Lancet. 355(9208), 1008-1010.

Sesti G (2006). Pathophysiology of insulin resistance. Beat Pract Res Clin Endocrinol Metab 20, 665-679.

Shen P., Liu M.H., Ng T.Y., Chan Y.H., Yong E.L. (2006). Differential effects of isoflavones, from Astragalus membranaceus and Pueraria thomsonii, on the activation of PPARalpha, PPARgamma, and adipocyte differentiation in vitro. J Nutr. 136, 899905.

Shin D.W., Kim S.N., Lee S.M., Lee W., Song M.J., Park S.M. 2009 (-)-Catechin promotes adipocyte differentiation in human bone marrow mesenchymal stem cells through PPAR gamma

Soumyanath A (2006). Traditional Medicines for Modern Times - Antidiabetic Plants, Ed. CRC Press, Boca Raton, London, New York.

Takahashi N., Goto T., Taimatsu A., Egawa K., Katoh S., Kusudo T. (2009). Bixin regulates mRNA expression involved in adipogenesis and enhances insulin sensitivity in 3T3-L1 adipocytes through PPARgamma activation. Biochem Biophys Res Commun. 390, 1372-1376.

Tantry M.A., Dar J.A., Idris A., Akbar S., Shawl A.S. (2012). Acylated flavonol glycosides from Epimedium elatum, a plant endemic to the Western Himalayas. Fitoterapia. 83, 665-670.

Tontonoz P, Hu E, Spiegelman BM. (1995) Regulation of adipocyte gene expression and differentiation by peroxisome proliferator activated receptor y. Current opinion in genetics & development. 5(5), 571-576.

Tsai, S.-J. Huang, C.-S. Mong, M.-C. Kam, W.-Y. Huang, H.-Y. and Yin, M.-C. (2012) Anti-inflammatory and antifibrotic effects of naringenin in diabetic mice. Journal of Agricultural and Food Chemistry, 60, 1, 514-521,.

Tsuda T, Ueno Y, Yoshikawa T, Kojo H & Osawa T (2006). Microarray profiling of gene expression in human adipocytes in response to anthocyanins. Biochem Pharmacol 71, 1184-1197.

Unuofin J. O., Otunola, GA. and Afolayan, A. J. (2018) In vitro a-amylase, a-glucosidase, lipase inhibitory and cytotoxic activities of tuber extracts of Kedrostis africana (L.) Cogn. Heliyon. 4, 9, e00810,.

Unuofin JO, Lebelo SL. (2020). Antioxidant effects and mechanisms of medicinal plants and their bioactive compounds for the prevention and treatment of type 2 diabetes: an updated review. Oxidative medicine and cellular longevity. 13; 1-36.

Watkins PB, Whitcomb RW. (1998). Hepatic dysfunction associated with troglitazone. New England Journal of Medicine. 338(13), 916-917.

Weidner C., de Groot J.C., Prasad A., Freiwald A., Quedenau C., Kliem M. (2012). Amorfrutins are potent antidiabetic dietary natural products. Proc Natl

617.

Sato M., Tai T., Nunoura Y., Yajima Y., Kawashima S.,

transactivation. Biochem Pharmacol. 77, 125-133. Acad Sci USA. 109, 7257-7262.

Weidner C., de Groot J.C., Prasad A., Freiwald A., Quedenau C., Kliem M. (2012). Amorfrutins are potent antidiabetic dietary natural products. Proc Natl Acad Sci USA. 109, 7257-7262.

Willson TM, Brown PJ, Sternbach DD, Henke BR. (2000). The PPARs: from orphan receptors to drug discovery. Journal of medicinal chemistry. 43(4), 52750.

Yamauchi, T., Kamon, J., Waki, H., Murakami, K., Motojima, K., Komeda, K., Ide, T., Kubota, N., Terauchi, Y., Tobe, K., Miki, H., Tsuchida, A., Akanuma, Y., Nagai, R., Kimura, S. & Kadowaki, T. (2001) The mechanisms by which both heterozygous peroxisome proliferator-activated receptor gamma (PPARy) deficiency and PPARy agonist improve insulin resistance. Journal of Biological Chemistry, 276, 41245-41254

Yang M.H., Vasquez Y., Ali Z., Khan I.A., Khan S.I. (2013) Constituents from Terminalia species increase PPARalpha and PPARgamma levels and stimulate glucose uptake without enhancing adipocyte differentiation. J Ethnopharmacol. 149, 490-498.

Yang, L.-P. Sun, H.-l. Wu L.-M. (2009). Baicalein reduces inflammatory process in a rodent model of diabetic retinopathy. Investigative Opthalmology & Visual Science, 50(5), 2319.

Yokoi H., Mizukami H., Nagatsu A., Ohno T., Tanabe H.,

Inoue M. (2009) Peroxisome proliferator-activated receptor gamma ligands isolated from adlay seed (Coix lacryma-jobi L. var. ma-yuen STAPF.) Biol Pharmaceut Bull. 32, 735-740.

Zhang H, Matsuda H, Yamashita C, Nakamura S, Yoshikawa M. (2009) Hydrangeic acid from the processed leaves of Hydrangea macrophylla var. thunbergii as a new type of anti-diabetic compound.Eur J Pharmacol. 606(1-3), 255-61.

Zhu Y, Alvares K, Huang Q, Rao MS, Reddy JK. (1993). Cloning of a new member of the peroxisome proliferator-activated receptor gene family from mouse liver. Journal of Biological Chemistry. 268(36), 26817-26820.

Zoechling A, Liebner F, Jungbauer A. (2011). Red wine: a source of potent ligands for peroxisome proliferator-activated receptor y. Food & function., 2(1), 28-38.

Zoechling A., Liebner F., Jungbauer A. (2011) Red wine: a source of potent ligands for peroxisome proliferator-activated receptor gamma. Food Funct. 2, 28-38.

Zou G., Gao Z., Wang J., Zhang Y., Ding H., Huang J. (2008) Deoxyelephantopin inhibits cancer cell proliferation and functions as a selective partial agonist against PPARgamma. Biochem Pharmacol. 75, 1381-1392.

i Надоели баннеры? Вы всегда можете отключить рекламу.