Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 4038 2023-10-23 08:23:38 |
2 format correct Meta information modification 4038 2023-10-23 10:12:24 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Kadasah, S.F.; Radwan, M.O. Ursolic Acid Pharmacodynamics towards Nuclear Receptor. Encyclopedia. Available online: https://encyclopedia.pub/entry/50657 (accessed on 05 May 2024).
Kadasah SF, Radwan MO. Ursolic Acid Pharmacodynamics towards Nuclear Receptor. Encyclopedia. Available at: https://encyclopedia.pub/entry/50657. Accessed May 05, 2024.
Kadasah, Sultan F., Mohamed O. Radwan. "Ursolic Acid Pharmacodynamics towards Nuclear Receptor" Encyclopedia, https://encyclopedia.pub/entry/50657 (accessed May 05, 2024).
Kadasah, S.F., & Radwan, M.O. (2023, October 23). Ursolic Acid Pharmacodynamics towards Nuclear Receptor. In Encyclopedia. https://encyclopedia.pub/entry/50657
Kadasah, Sultan F. and Mohamed O. Radwan. "Ursolic Acid Pharmacodynamics towards Nuclear Receptor." Encyclopedia. Web. 23 October, 2023.
Ursolic Acid Pharmacodynamics towards Nuclear Receptor
Edit

Nuclear receptors (NRs) form a family of druggable transcription factors that are regulated by ligand binding to orchestrate multifaceted physiological functions, including reproduction, immunity, metabolism, and growth. NRs represent attractive and valid targets for the management and treatment of a vast array of ailments. Pentacyclic triterpenes (PTs) are ubiquitously distributed natural products in medicinal and aromatic plants, of which ursolic acid (UA) is an extensively studied member, due to its diverse bio-pertinent activities against different cancers, inflammation, aging, obesity, diabetes, dyslipidemia, and liver injury.

ursolic acid nuclear receptors NASH metabolic disorders

1. Modulation of Peroxisome-Proliferator-Activated Receptors (PPARs)

PPARs involve three subtypes (PPARα; NR1C1, PPARβ; NR1C2, and PPARγ; NR1C3) that control insulin sensitivity, resistance, and lipid homeostasis, making them valid targets for alleviating metabolic syndrome, hyperlipidemia, and diabetes [1][2][3][4]. PPARα reduces the formation of blood lipids and plays a role in cancer [5], and PPARβ also plays a role in managing blood lipid levels and insulin sensitivity [6][7]. PPARγ controls insulin sensitivity, adipogenesis, neuroprotection [8][9], and inflammation [10]. Fibrates are PPARα modulators used for hyperlipidemia therapy and are represented by fenofibrate and pemafibrate, whereas thiazolidinediones, such as pioglitazone and rosiglitazone, are used for the treatment of diabetes through PPARγ agonism [11][12]. Among different NRs, the UA effect on PPARs is the most explored [13].

1.1. UA Effect on PPARα

The first report on PT modulation of PPARα and their potential pharmaceutical and cosmeceutical role in dermatology was released in 2005 [14]. Concomitantly, in 2007, Lim et al. showed that topical application of UA to hairless adult mice models enhanced keratinocyte differentiation and led toa subsequent recovery of the epidermal permeability barrier. This effect was clearly observed by examining a biopsy specimen using a light microscope and an electron microscope. The enhanced recovery was hypothesized to be due to PPARα activation. This was validated by immunoblot analysis of PPARα and the keratinocyte differentiation markers involucrin, loricrin, and filaggrin, in human skin keratinocyte cell line HaCaT cells. The test confirmed that UA treatment upregulated the tested protein levels, leading to accelerated recovery. It is worth noting that OA exhibited a similar therapeutic effect [15].
Interestingly, UA’s agonistic effect on PPARα played a pivotal role in its cytotoxic activity against skin cancer through the AMPK pathway. In the mouse squamous carcinoma model, Ca3/7, UA enhanced AMPK phosphorylation at cytotoxic levels, which was reversed by using an AMPK small molecule inhibitor or by AMPK knockdown. As PPARα upregulation has a partial role in skin cancer therapy, the authors investigated this mechanism for UA [5]. Indeed, using the PPARα antagonist GW6471, or the less potent MK886, for one hour prior to adding UA, elevated IC50 values of the latter against Ca3/7 or mouse skin papilloma cells MT1/2, as shown by MTT assay. This suggests that the UA cytotoxic effect is partially mediated by PPARα activation [16].
Likewise, Jia et al. confirmed UA-induced activation of PPARα in terms of alleviating hypertriglyceridemia. Having said that, they could not confirm that UA directly binds to PPARα LBD using Biacore surface plasmon resonance (SPR) analysis. However, UA treatment remarkably promoted PPARα mRNA concentration in cultured hepatocytes (HepG2), as shown by qPCR. A luciferase reporter gene assay in the same cells revealed that UA is a PPARα activity upregulator. Furthermore, a 20 µM concentration of UA enhanced PPARα binding to its response element in the responsive genes by 46% and promoted PPARα transactivation consequently. In a dose-dependent manner, UA treatment was significantly proved to have significant hypolipidemic effects by reducing intracellular triglycerides (TGL) and cholesterol accumulation in HepG2 cells. This was accompanied by significant upregulation of the fatty acid transport protein 4 (FATP4) gene in both mRNA and protein levels; FATP4 is a major fatty acid transporter in the liver and a known target gene PPARα. The authors emphasized that UA promotes PPARα transactivation by indirect mechanisms, other than simply binding to its LBD [17].
The same research group moved forward with in vivo validation of their previous in vitro results. They found out that UA can regulate lipid and glucose metabolism in high-fat diet (HFD)-fed mice. UA intake reduced lipid accumulation in adipose tissues and the liver, while increasing muscle mass. Biochemical analysis confirmed that plasma levels of TGL and low- density lipoprotein (LDL) levels were reduced in contrast to high-density lipoprotein (HDL) levels. This was accompanied by improved glucose tolerance and insulin sensitivity. In mice tissue, UA treatment resulted in the over-expression of mRNA and protein levels of PPARα, the activation of its responsive genes that regulate fatty acids uptake and β-oxidation, and the suppression of lipogenic genes [18]. Additionally, UA induced the hepatic expression of the autophagy marker LC3-II, which could partially participate in the hypoglycemic and hypolipidemic role of UA in HFD-fed mice [19].
The anti-hyperlipidemic effect of UA (25 mg/kg) or artesunate (25 mg/kg) alone or in combination (12.5 + 12.5 mg/kg) was assessed in a New Zealand rabbit model on a Western-style diet. UA administration for a couple of months significantly reduced TGL and cholesterol levels in a comparable manner to atorvastatin without a significant effect on LDL level, which was efficiently lowered in the case of UA/artesunate combination. UA alone alleviated hepatocyte steatosis; meanwhile, the combination completely prevented it in the same way as atorvastatin, as displayed by histopathological examination using hematoxylin and eosin (H&E) stains [20]. In liver tissue, UA alone or in combination upregulated mRNA expression of PPARα, which is in agreement with previous studies.
The potential role of UA in NASH therapy was further investigated by the Li group using the obese NASH Sprague Dawley rat model. UA administration significantly reversed HFD-induced lipid accumulation, NASH, and liver injury and reduced serum ALT, AST, TGL, FFA, and LDL levels in a dose-dependent manner, as revealed by hepatocyte morphologic, histological, and serum biochemical examination. In the same context, UA promoted mRNA and protein levels of PPARα whose knockdown interrupted UA-induced hepatoprotective effect. UA reduced the expression of hepatic inflammatory cytokines, including different interleukins and the tumor necrosis factor α (TNFα). In this model, UA did not affect the activity of PPARγ, farnesoid X receptor (FXR), or liver X receptor (LXR) [21]. Meanwhile, the authors studied the beneficial effect of UA in the human hepatic cell line (HL-7702) model, where it stimulates PPARα mRNA, showing an anti-steatosis effect that was interrupted by PPARα knocking down [21].
Another research group studied the PPARα upregulation effect on alleviating peripheral inflammation and inflammatory hyperalgesia in obese animals. Following the injection of carrageenan into obese Sprague Dawley rats, systemic UA administration mitigated thermal hyperalgesia and paw edema, compared with the control group. At the molecular level, UA lowered the expression of inflammatory mediators, including IL-1β, TNF-α, and NF-κB P65 in the spinal cord of the rats, as shown by the Western blot test. Carrageenan injection into rats’ paws significantly reduced spinal cord PPARα levels in the control HFD groups prior to UA administration, which reversed the process and restored PPARα levels. This means that UA could restore PPARα levels in obese rats’ spinal cords and reduce the expression of inflammatory mediators due to peripheral inflammatory stimulation [22].
UA-induced activation of PPARα is not only beneficial in skin diseases and metabolic disorders, but also in right ventricle hypertrophy (RVH) and remodeling [23]. In a Sprague Dawley monocrotaline-induced RV dysfunction rat model, UA significantly reduced RVH and RV fibrosis, promoted ventricle function, and lowered the increase in cardiomyocyte size and mRNA level of hypertrophic genes and apoptotic cells. From a metabolic aspect, monocrotaline injection remarkably decreased PPARα and PPARγ gene expression; however, UA pretreatment only reversed the abnormal PPARα expression in RV tissue. This opens the way for harnessing UA in the alleviation of RV disorders through the PPARα pathway [24].

1.2. UA Effect on PPARγ

The UA effect on PPARγ was described in different aspects of biological activities. PPARγ agonism is well-known to alleviate inflammations in asthmatic animal models [25]. In the BALB/c mice model of allergic bronchial asthma facing methacholine challenge, UA nebulization mitigated methacholine-induced airway hypersensitivity and alleviated airway inflammation. In the ovalbumin-challenged asthma model, ursolic acid (20 mg/kg) reduced eosinophilia bronchoalveolar lavage fluid, neutrophils, and eosinophils, within peripheral blood mononuclear cells (PBMC). Furthermore, it suppressed cytokine, IL-5, IL-13, and IL-17 release, and reduced the level of anti-ovalbumin IgE, in comparison to untreated cells. The authors showed that UA significantly upregulated PPARγ mRNA expression in lung tissue. PPARγ activation was further validated in EL4 T cells and RAW 264.7 macrophages, via qPCR and Western blotting [26].
Wang et al. explored UA neuroprotection effects through the PPARγ pathway in a model of male Sprague Dawley rats with middle cerebral artery occlusion and reperfusion. UA treatment improved the neurological deficit score, promoted the number of intact healthy neurons, and minimized the infarct size compared to the control animals in a dose-dependent manner. This is accompanied by the upregulation of PPARγ, the downregulation of the inflammatory mediators, matrix metalloproteinase-2/9 (MMP2 and MMP9), the increment of the anti-inflammatory factor tissue inhibitor matrix metalloproteinase (1TIMP1), and the interruption of MAPK signaling pathways in brain tissue, as revealed by Western blot and qRT-PCR. Hence, UA can serve as a neuroprotective therapeutic agent, acting via PPARγ agonism, and optimizing the metalloprotease/anti-metalloprotease balance [27].
Another confirmatory report on UA anti-inflammatory activity in the central nervous system (CNS) via PPARγ activation has been recently published. UA enhanced the phenotypic switch of BV2 cells (murine microglia) from M1 polarization, the pro-inflammatory, to M2 polarization, the anti-inflammatory, through the promotion of PPARγ protein expression. Meanwhile, PPARγ activation resulted in the suppression of MMP2 and MMP9 secretion and the increment of 1TIMP1 secretion, which supports the previous results [27]. Notably, those effects were not observed in the case of the co-addition of UA and the potent selective PPARγ antagonist GW9662. In a word, UA protects against neuro-inflammation through the PPARγ pathway, opening the way for its application in ischemic stroke therapy [28].
A potential dual role of UA in the treatment of multiple sclerosis (MS) through immunomodulation and neuroregeneration via PPARγ agonism was disclosed by Zhang et al. [29]. In MS mice model using experimental autoimmune encephalomyelitis (EAE), a 25 mg/kg/d of UA reduced CNS inflammation and demyelination; furthermore, in Th1- and Th17-polarizing cultures, UA reduced their differentiation, implying an immunomodulatory effect. At the chronic stage of EAE, UA intake not only hinders further spinal cord myelin damage, but also supports myelin recovery, and protects neurons and axons by promoting oligodendrocyte progenitor cell maturation in CNS lesions. The remyelination-enhancer effect was consistent incuprizone-induced demyelination model in a completely PPARγ-agonistic pathway that disappears incase of PPARγ knockout. In an ex vivo model of lysophosphatidylcholine (LPC)-induced demyelination in organotypic cerebellar slices, UA reduced the expression of inflammatory factors and upregulated anti-inflammatory cytokines and neurotrophins, especially mRNA and the protein level of ciliary neurotrophic factor (CNTF), which, in turn, promoted remyelination. CNTF expression was significantly promoted in astrocytes by UA, and this induction was highly opposed by the PPARγ antagonist GW-9662 [29].
Collectively, UA has a highly promising PPARγ-agonistic character that can be employed for the management of a multitude of diseases, including bronchial asthma, CNS ischemia, and neuro-inflammatory diseases such as MS. Table 1 summarizes the mentioned effects of UA on PPARs and the other NRs in this study and the related bioactivity.
Table 1. Summary of ursolic acid (UA) pharmacodynamic effect on nuclear receptors (NRs) and the resulting corresponding therapeutic effect.

2. Modulation of Liver X Receptors (LXRs)

The hydrophobic oxysterols are the endogenous ligands of LXRs, which have two subtypes (LXRα; NR1H3 and LXRβ; NR1H2). Both have shared approximately 70% homology with PPARs. They play a paramount role in lipid and glucose homeostasis, atherosclerosis, and NASH development by regulating hepatic de novo lipogenesis [36][37][38]. Activation of LXRα transactivates hepatic lipogenic genes and LXRα is found to be upregulated in the case of non-alcoholic fatty liver disease (NAFLD); thus, LXR antagonists might be useful for NASH therapy [39][40]. On the contrary, LXRα agonists alleviate the atherosclerotic effect, which is accompanied by severe adverse effects such as hepatic steatosis; this hinders the development of the potent LXRα agonist T090.
Kuding tea or Ku-Ding-Cha leaves are mainly from Ilex latifolia Thunb and Ilex kudingcha C.J. Tseng, of the family Aquifoliaceae. This bitter-tasting tea contains high amounts of ursolic acid and has been widely used in China for more than 2000 years as a healthy beverage for the management of obesity, cardiovascular disease, hypertension, and hyperlipidemia [41]. Fan et al. explored the mechanism of action of Kuding tea alcoholic extract [30]. In cell culture, it could interrupt the later stages of 3T3-L1 adipocyte differentiation. Indeed, in the high-fat diet C57BL/6 mice model, the extract reduced weight gain, blood glucose level, and lipid accumulation in hepatocytes. The authors found that the resulting benefits were partially attributed to LXR antagonism. However, they did not figure out which components in the extract were responsible for the activity.
Later on, Lin et al. identified UA as an LXRα antagonist, in a similar fashion to its oleanane-type analog OA [31][42]. In a dose-dependent manner, UA opposed T090-induced transactivation of LXRα in human hepatocarcinoma cells, as shown by a luciferase reporter assay using an LX response element and SREBP-1c promoter. Consistently, co-treatment with UA attenuated T090-inducedupregulation of LXRα lipogenic target genes, including SREBP-1c, SCD, and FAS. Microscopic examination of Oil Red O stained hepatocytes showed a reduction in TGL and cholesterol accumulation by UA. To validate the present data, the authors went through further in vivo tests using male C57BL/6 mice. Histopathologic examination of the mice liver section, using H&E staining and Oil Red O staining, showed elevated lipid and TG accumulation accompanied by microsteatosis due to T090. This was significantly reversed by the co-administration of UA. In mice hepatocytes, UA showed a similar downregulation effect on lipogenic genesto that in human hepatocytes [31].
Molecular docking calculations of UA and T090 into the LXRα ligand binding site (Protein Data Bank ID:1UHL) [43] using the CDOCKER module of Discovery Studio (DS) revealed useful theoretical information on the potential binding pattern. The CDOCKER binding energy of T090, the co-crystalized ligand, was −45.7965 kcal/mol, whereas UA also fitted snugly in the same hydrophobic pocket, with a comparable energy parameter of −37.5211 kcal/mol, reflecting an optimal interaction. UA displayed strong hydrophobic interactions with Phe326, Phe257, Leu331, Trp443, Leu439, Phe254, and Ala261, with a different binding mode from T090.
UA activity was assessed in human intestinal cells LS174T, and surprisingly, it upregulated ABCA1 and ABCG1 expression instead of the anticipated downregulation. It is worth noting that ABCG1 gene expression decreased upon co-treatment with UA and T090 in HepaRG cells, confirming the cellular-context paradox. This is further confirmed by the lack of UA effect on cellular contents of TG in LS174T cells. It can be deduced that UA suppressed LXRα activation in hepatocytes but not in intestinal cells, suggesting that UA controls LXRα signaling in a cell- and tissue-specific manner due to differential effects on the recruitment of coregulators [31].
The possible clinical application of UA to mitigate the lipogenic side effects of the ani-epileptic drug valproate was tested. De facto, UA significantly opposed valproate induced LXRα transactivation, lipogenic gene expression, and lipid accumulation in HepaRG cells.

3. Modulation of Pregnane X Receptors (PXR) and Constitutive Androstane Receptors (CAR)

Alongside CAR (NR1I3), PXR (NR1I2) is mainly responsible for xenobiotic detoxification by regulating the expression of the metabolic enzyme cytochrome P450 (CYP 450), including the two main types, CYP3A4 and CYP2B6 [44][45]. PXR can be modulated by numerous exogenous and endogenous ligands such as bile acids, steroids, antibiotics like rifampicin, and antimycotics like clotrimazole [46]. Dysregulation of PXR/CAR leads to drug-induced hepatotoxicity, as in the cases of acetaminophen- and isoniazid-induced hepatic injury.
Using a dual-luciferase reporter gene assay in HepaRG cells, UA, alongside carnosol from Rosmarinus officinalis, activated mouse, rat, and human PXR. In terms of human PXR activation, the EC50 values of UA and carnosol were 10.77 and 2.22 µM, respectively. UA was confirmed to bind within PXR LBD and activate luciferase activity in cells transfected with a plasmid expressing human PXR LBD. In human colon adenocarcinoma cells, LS180, UA promoted the mRNA expression of the major metabolizing enzyme CYP3A4 and a multi-drug resistance protein 1called ATP binding cassette B1 (ABCB1). In the intestine, this effect enhanced the first-pass metabolism and reduced the oral bioavailability of chemicals metabolized by CYP3A4 and transported by ABCB1 [47].
In 2017, two different reports came out, reporting the promising role of UA and OA in attenuating rifampicin/isoniazid-induced cytotoxicity viamodulation of PXR and its sister NR CAR [32][48]. The presented results were in discrepancy with the above-mentioned outcome of PXR activation. Herein, in human PXR-expressing and CYP3A4 reporter plasmid-transfected HepaRG cells, UA antagonized PXR activity and significantly attenuated the transactivation of the CYP3A4 promoter in a concentration-dependent manner. This inhibitory effect was remarkable incase of co-treatment with the activator rifampicin. Indeed, UA inhibited CYP3A4 mRNA and protein expression. Likewise, UA, through a CAR-dependent mechanism, was involved in the downregulation of the target gene CYP2B6 on both mRNA and protein levels [32]. The catalytic activity of CYP3A4 and CYP2B6 under only UA, or under rifampicin co-treatment, was significantly attenuated. Interestingly, the well-known rifampin-mediated and isoniazid-induced cytotoxicity was reduced by UA co-treatment, as shown by the HepaRG cell viability test. Additionally, UA elevated the intracellular glutathione levels and regeneration capacity in a concentration-dependent manner [32].
A supporting claim for the outcome for PXR antagonism by UA was introduced by the same research group in 2018; they evaluated the UA effect on PXR transactivation of lipogenic genes, including S14, SCD, FAS, and FAE. It was revealed that UA could effectively oppose the transient activation of promoters S14 and SCD by rifampicin, as shown by reporter assay. In the presence of rifampicin, UA reduced the mRNA and protein expression of S14, SCD, FAS, and FAE genes. Histopathological examination of stained HepaRG cells by phase-contrast microscope showed rifampicin-induced lipid accumulation and steatosis, which was significantly interrupted by UA [31]. Therefore, UA could serve to lessen the unwanted interactions between transcriptional inducers of CYP450 enzymes and drugs [49].

4. Modulation of Retinoic Acid Receptor-Related Orphan Receptors (ROR)

ROR has three subtypes that possess indispensable roles in immunity, development, and metabolic homeostasis. It is worth noting that the RORγt type is only expressed in immune cells, especially Th17 lymphocytes, where it controls their development and differentiation from CD4+ cells [50]. Th17 cells secrete different inflammatory interleukins (ILs), like IL-17 and IL-21, that fight against pathogenic invaders. Nevertheless, its upregulation is linked to autoimmune diseases such as systemic lupus erythematosus, lupus nephritis, psoriasis, rheumatoid arthritis, and MS; thereby, RORγt is a potential target for managing such obstinate diseases [51][52]. Recently, RORγ overexpression was related to the progress of different types of advanced cancers of breast, prostate, and lung [53][54][55]. Endogenous hydroxycholesterols, which have structural similarity to PTs, can bind and modulate RORγt-dependent biological processes [56][57].
Indeed, methyl corosolate, uvaol, and OA are three triterpenes found in loquat leaves with in vitro inhibitory effects against RORγt, accompanied by an interruption of Th17 differentiation, with a potential application in lupus nephritis [58]. Furthermore, the titled PTs ameliorated skin inflammation, epidermal hyperplasia, and aberrant keratinocyte proliferation in an imiquimod-induced psoriasis animal model [59]. Digoxin, with its similar structure to hydroxycholesterols, is a well-established RORγt inverse agonist that was co-crystalized with it (Protein Data Bank ID: 3B0W) [60].
The first report claiming that UA acts as a strong and selective inhibitor of RORγt function came out in 2011 by Xu et al. A preliminary high throughput screening of 2000 compounds identified UA as a human and mouse Th17 development and differentiation inhibitor in a dose-dependent manner. At 2 µM, UA inhibited RORγt-mediated, but not RORαt-mediated, IL-17 and IL-17 expression to almost background level in Th17 cells. UA, in a dose-dependent manner, interrupted the binding of RORγt-LBD, but not RORαt-LBD, to its co-activator peptide. Further experiments led to the conclusion that UA exclusively antagonizes RORγt function with IC50 of 0.68 µM, while its IC50 on Th17 cells was determined to be 0.56 µM. As a result, UA abrogated IL-17 secretion from differentiated Th17 cells of both mouse and human origin. In experimental autoimmune encephalomyelitis mice as an MS model, UA treatment slowed the onset of disease in mice and significantly alleviated the symptoms in comparison with the control group. The CNS of UA-treated mice contained fewer IL-17+ and IFN-γ+ cells, and their spleen showed less IL-17 production. The study paved the way for UA application in autoimmune disorders and Th17-mediated inflammatory diseases [33].
In addition, UA administration significantly reduced the incidence and severity of collagen-induced autoimmune arthritis in mice models, partially via the inhibition of Th17 differentiation, as shown by flow cytometry. In a dose-dependent manner, mRNA expression of IL-17, IL-21, and RORγt was downregulated in the splenocytes [34]. Owing to its pronounced RORγt antagonism, UA was used as a positive control when testing new inverse agonists [50].
A recent interesting report by Zou et al. emphasized the RORγ-dependent anti- proliferative role of UA against triple negative breast cancer (TNBC) cells, HCC70, and prostate cancer (PCa) cell lines C4-2B and 22Rv1 [35]. In the test cell lines, UA lowered RORγ activation, as shown by a luciferase reporter assay, in a dose-dependent manner. In PCa, UA interrupted RORγ-mediated androgen receptors’ (AR) expression and signaling; this was also observed for the variant AR-V7 in C4-2B and 22RV1 cells. The strong anticancer effect of UA was more remarkable in the AR-positive PCa cell line LNCaP but not in the AR-negative PCa cell lines PC3 and DU145. In TNBC, RNA-seq, qRT-PCR, and Western blot analysis showed that UA treatment suppressed the RORγ-mediated mRNA and protein expression of most of the genes controlling cholesterol biosynthesis. Concomitantly, UA disrupted RORγ-controlled apoptosis/cell cycle genes. In conclusion, UA elicits its antiproliferative effect against PCa and TNBC, in part, via targeting RORγ [35].

5. Modulation of Farnesoid X Receptors FXRs

FXRs are involved in lipid and bile acid homeostasis, with a significant role in hepatic inflammation and fibrosis, and are widely distributed in organs such as the liver, kidney, intestinal tract, and adrenal gland [61][62][63][64]. Bile acids, the natural ligands of FXR, were identified as potential promoters of colon cancer [65][66]. FXRα; NR1H4 represents a valid target for mitigating primary biliary cirrhosis (PBC), NASH, diabetes [67], and atherosclerosis [67][68][69][70][71]. An FXR modulator, obeticholic acid, was approved for PBC therapy, and further clinical trials are underway to assess it against NASH [72].
A recent report revealed that UA can modulate FXR in rat models with alcoholic liver injury. UA intervention reduced the pathological changes in hepatocytes when examined by hematoxylin–eosin staining. The reduced hepatic steatosis was accompanied by improved cell inflammation and infiltration. In biochemical terms, alanine aminotransferase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP), and total bile acid (TBA) levels in serum were significantly lessened in comparison to the untreated group. Concomitantly, UA upregulated FXR protein expression and downregulated CYP7A1 and SREBP-1c expression [73].

References

  1. Berger, J.; Moller, D.E. The mechanisms of action of PPARs. Annu. Rev. Med. 2002, 53, 409–435.
  2. Zhang, F.; Lavan, B.E.; Gregoire, F.M. Selective Modulators of PPAR-gamma Activity: Molecular Aspects Related to Obesity and Side-Effects. PPAR Res. 2007, 2007, 32696.
  3. Allen, T.; Zhang, F.; Moodie, S.A.; Clemens, L.E.; Smith, A.; Gregoire, F.; Bell, A.; Muscat, G.E.; Gustafson, T.A. Halofenate is a selective peroxisome proliferator-activated receptor gamma modulator with antidiabetic activity. Diabetes 2006, 55, 2523–2533.
  4. Zheng, W.; Feng, X.; Qiu, L.; Pan, Z.; Wang, R.; Lin, S.; Hou, D.; Jin, L.; Li, Y. Identification of the antibiotic ionomycin as an unexpected peroxisome proliferator-activated receptor γ (PPARγ) ligand with a unique binding mode and effective glucose-lowering activity in a mouse model of diabetes. Diabetologia 2013, 56, 401–411.
  5. Thuillier, P.; Anchiraico, G.J.; Nickel, K.P.; Maldve, R.E.; Gimenez-Conti, I.; Muga, S.J.; Liu, K.-L.; Fischer, S.M.; Belury, M.A. Activators of peroxisome proliferator-activated receptor-alpha partially inhibit mouse skin tumor promotion. Mol. Carcinog. 2000, 29, 134–142.
  6. Leone, T.C.; Weinheimer, C.J.; Kelly, D.P. A critical role for the peroxisome proliferator-activated receptor alpha (PPARalpha) in the cellular fasting response: The PPARalpha-null mouse as a model of fatty acid oxidation disorders. Proc. Natl. Acad. Sci. USA 1999, 96, 7473–7478.
  7. Sanderson, L.M.; Degenhardt, T.; Koppen, A.; Kalkhoven, E.; Desvergne, B.; Müller, M.; Kersten, S. Peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) but not PPARalpha serves as a plasma free fatty acid sensor in liver. Mol. Cell. Biol. 2009, 29, 6257–6267.
  8. Cai, W.; Yang, T.; Liu, H.; Han, L.; Zhang, K.; Hu, X.; Zhang, X.; Yin, K.-J.; Gao, Y.; Bennett, M.V.; et al. Peroxisome proliferator-activated receptor γ (PPARγ): A master gatekeeper in CNS injury and repair. Prog. Neurobiol. 2018, 163–164, 27–58.
  9. Hou, G.; Yin, Y.; Han, D.; Wang, Q.-Y.; Kang, J. Rosiglitazone attenuates the metalloprotease/anti-metalloprotease imbalance in emphysema induced by cigarette smoke: Involvement of extracellular signal-regulated kinase and NFκB signaling. Int. J. Chron Obs. Pulmon Dis. 2015, 10, 715–724.
  10. Fajas, L.; Auboeuf, D.; Raspé, E.; Schoonjans, K.; Lefebvre, A.-M.; Saladin, R.; Najib, J.; Laville, M.; Fruchart, J.-C.; Deeb, S.; et al. The Organization, Promoter Analysis, and Expression of the Human PPARγ Gene*. J. Biol. Chem. 1997, 272, 18779–18789.
  11. Yang, C.; Li, Q.; Li, Y. Targeting Nuclear Receptors with Marine Natural Products. Mar. Drugs 2014, 12, 601–635.
  12. Orasanu, G.; Ziouzenkova, O.; Devchand, P.R.; Nehra, V.; Hamdy, O.; Horton, E.S.; Plutzky, J. The PPARγ Agonist Pioglitazone Represses Inflammation In A PPARα-Dependent Manner In Vitro and In Vivo In Mice. J. Am. Coll. Cardiol. 2008, 52, 869–881.
  13. Enayati, A.; Ghojoghnejad, M.; Roufogalis, B.D.; Maollem, S.A.; Sahebkar, A. Impact of Phytochemicals on PPAR Receptors: Implications for Disease Treatments. PPAR Res. 2022, 2022, 4714914.
  14. Lee, H.K.; Nam, G.W.; Kim, S.H.; Lee, S.H. Phytocomponents of triterpenoids, oleanolic acid and ursolic acid, regulated differently the processing of epidermal keratinocytes via PPAR-α pathway. Exp. Dermatol. 2006, 15, 66–73.
  15. Lim, S.W.; Hong, S.P.; Jeong, S.W.; Kim, B.; Bak, H.; Ryoo, H.C.; Lee, S.H.; Ahn, S.K. Simultaneous effect of ursolic acid and oleanolic acid on epidermal permeability barrier function and epidermal keratinocyte differentiation via peroxisome proliferator-activated receptor-α. J. Dermatol. 2007, 34, 625–634.
  16. Junco, J.J.; Cho, J.; Mancha, A.; Malik, G.; Wei, S.-J.; Kim, D.J.; Liang, H.; DiGiovanni, J.; Slaga, T.J. Role of AMPK and PPARα in the anti-skin cancer effects of ursolic acid. Mol. Carcinog. 2018, 57, 1698–1706.
  17. Jia, Y.; Bhuiyan, M.J.H.; Jun, H.-J.; Lee, J.H.; Hoang, M.H.; Lee, H.-J.; Kim, N.; Lee, D.; Hwang, K.Y.; Hwang, B.Y.; et al. Ursolic acid is a PPAR-α agonist that regulates hepatic lipid metabolism. Bioorg. Med. Chem. Lett. 2011, 21, 5876–5880.
  18. Kimura, S.; Noda, T.; Yoshimori, T. Dissection of the autophagosome maturation process by a novel reporter protein, tandem fluorescent-tagged LC3. Autophagy 2007, 3, 452–460.
  19. Jia, Y.; Kim, S.; Kim, J.; Kim, B.; Wu, C.; Lee, J.H.; Jun, H.-J.; Kim, N.; Lee, D.; Lee, S.-J. Ursolic acid improves lipid and glucose metabolism in high-fat-fed C57BL/6J mice by activating peroxisome proliferator-activated receptor alpha and hepatic autophagy. Mol. Nutr. Food Res. 2015, 59, 344–354.
  20. Wang, Y.L.; Wang, Z.J.; Shen, H.L.; Yin, M.; Tang, K.X. Effects of artesunate and ursolic acid on hyperlipidemia and its complications in rabbit. Eur. J. Pharm. Sci. 2013, 50, 366–371.
  21. Li, S.; Meng, F.; Liao, X.; Wang, Y.; Sun, Z.; Guo, F.; Li, X.; Meng, M.; Li, Y.; Sun, C. Therapeutic role of ursolic acid on ameliorating hepatic steatosis and improving metabolic disorders in high-fat diet-induced non-alcoholic fatty liver disease rats. PLoS ONE 2014, 9, e86724.
  22. Zhang, Y.; Song, C.; Li, H.; Hou, J.; Li, D. Ursolic acid prevents augmented peripheral inflammation and inflammatory hyperalgesia in high-fat diet-induced obese rats by restoring downregulated spinal PPARα. Mol. Med. Rep. 2016, 13, 5309–5316.
  23. Xu, Y.; Gu, Q.; Liu, N.; Yan, Y.; Yang, X.; Hao, Y.; Qu, C. PPARγ Alleviates Right Ventricular Failure Secondary to Pulmonary Arterial Hypertension in Rats. Int. Heart J. 2017, 58, 948–956.
  24. Gao, X.; Zhang, Z.; Li, X.B.; Wei, Q.; Li, H.; Li, C.; Chen, H.; Liu, C.; He, K. Ursolic Acid Improves Monocrotaline-Induced Right Ventricular Remodeling by Regulating Metabolism. J. Cardiovasc. Pharmacol. 2020, 75, 545–555.
  25. Hirasawa, H.; Chiba, T.; Ueki, S.; Kamada, Y.; Ito, W.; Takeda, M.; Fujita, M.; Kato, H.; Kayaba, H.; Chihara, J. The synthetic PPARgamma agonist troglitazone inhibits eotaxin-enhanced eosinophil adhesion to ICAM-1-coated plates. Int. Arch. Allergy Immunol. 2008, 146 (Suppl. 1), 11–15.
  26. Kim, S.-H.; Hong, J.-H.; Lee, Y.-C. Ursolic acid, a potential PPARγ agonist, suppresses ovalbumin-induced airway inflammation and Penh by down-regulating IL-5, IL-13, and IL-17 in a mouse model of allergic asthma. Eur. J. Pharmacol. 2013, 701, 131–143.
  27. Wang, Y.; He, Z.; Deng, S. Ursolic acid reduces the metalloprotease/anti-metalloprotease imbalance in cerebral ischemia and reperfusion injury. Drug Des. Devel. Ther. 2016, 10, 1663–1674.
  28. Wang, Y.; Qiu, L.; Deng, S.; Liu, F.; He, Z.; Li, M.; Wang, Y. Ursolic acid promotes microglial polarization toward the M2 phenotype via PPARγ regulation of MMP2 transcription. Neurotoxicology 2023, 96, 81–91.
  29. Zhang, Y.; Li, X.; Ciric, B.; Curtis, M.T.; Chen, W.-J.; Rostami, A.; Zhang, G.-X. A dual effect of ursolic acid to the treatment of multiple sclerosis through both immunomodulation and direct remyelination. Proc. Natl. Acad. Sci. USA 2020, 117, 9082–9093.
  30. Fan, S.; Zhang, Y.; Hu, N.; Sun, Q.; Ding, X.; Li, G.; Zheng, B.; Gu, M.; Huang, F.; Sun, Y.-Q.; et al. Extract of Kuding tea prevents high-fat diet-induced metabolic disorders in C57BL/6 mice via liver X receptor (LXR) β antagonism. PLoS ONE 2012, 7, e51007.
  31. Lin, Y.-N.; Wang, C.C.N.; Chang, H.-Y.; Chu, F.-Y.; Hsu, Y.-A.; Cheng, W.-K.; Ma, W.-C.; Chen, C.-J.; Wan, L.; Lim, Y.-P. Ursolic Acid, a Novel Liver X Receptor α (LXRα) Antagonist Inhibiting Ligand-Induced Nonalcoholic Fatty Liver and Drug-Induced Lipogenesis. J. Agric. Food Chem. 2018, 66, 11647–11662.
  32. Chang, H.-Y.; Chen, C.-J.; Ma, W.-C.; Cheng, W.-K.; Lin, Y.-N.; Lee, Y.-R.; Chen, J.-J.; Lim, Y.-P. Modulation of pregnane X receptor (PXR) and constitutive androstane receptor (CAR) activation by ursolic acid (UA) attenuates rifampin-isoniazid cytotoxicity. Phytomedicine 2017, 36, 37–49.
  33. Xu, T.; Wang, X.; Zhong, B.; Nurieva, R.I.; Ding, S.; Dong, C. Ursolic Acid Suppresses Interleukin-17 (IL-17) Production by Selectively Antagonizing the Function of RORγt Protein*. J. Biol. Chem. 2011, 286, 22707–22710.
  34. Baek, S.-Y.; Lee, J.; Lee, D.-G.; Park, M.-K.; Lee, J.; Kwok, S.-K.; Cho, M.-L.; Park, S.-H. Ursolic acid ameliorates autoimmune arthritis via suppression of Th17 and B cell differentiation. Acta Pharmacol. Sin. 2014, 35, 1177–1187.
  35. Zou, H.; Yang, Y.; Chen, H.-W. Natural compounds ursolic acid and digoxin exhibit inhibitory activities to cancer cells in RORγ-dependent and -independent manner. Front. Pharmacol. 2023, 14, 1146741.
  36. Dixon, E.D.; Nardo, A.D.; Claudel, T.; Trauner, M. The Role of Lipid Sensing Nuclear Receptors (PPARs and LXR) and Metabolic Lipases in Obesity, Diabetes and NAFLD. Genes 2021, 12, 645.
  37. Lin, Z.; Zhang, Y.; Zhang, Y.; Shen, H.; Hu, L.; Jiang, H.; Shen, X. Oleanolic acid derivative NPLC441 potently stimulates glucose transport in 3T3-L1 adipocytes via a multi-target mechanism. Biochem. Pharmacol. 2008, 76, 1251–1262.
  38. Edwards, P.A.; Kennedy, M.A.; Mak, P.A. LXRs; oxysterol-activated nuclear receptors that regulate genes controlling lipid homeostasis. Vasc. Pharmacol. 2002, 38, 249–256.
  39. Higuchi, N.; Kato, M.; Shundo, Y.; Tajiri, H.; Tanaka, M.; Yamashita, N.; Kohjima, M.; Kotoh, K.; Nakamuta, M.; Takayanagi, R.; et al. Liver X receptor in cooperation with SREBP-1c is a major lipid synthesis regulator in nonalcoholic fatty liver disease. Hepatol. Res. 2008, 38, 1122–1129.
  40. Griffett, K.; Burris, T.P. Development of LXR inverse agonists to treat MAFLD, NASH, and other metabolic diseases. Front. Med. 2023, 10, 1102469.
  41. Wüpper, S.; Lüersen, K.; Rimbach, G. Chemical Composition, Bioactivity and Safety Aspects of Kuding Tea—From Beverage to Herbal Extract. Nutrients 2020, 12, 2796.
  42. Lin, Y.-N.; Chang, H.-Y.; Wang, C.C.N.; Chu, F.-Y.; Shen, H.-Y.; Chen, C.-J.; Lim, Y.-P. Oleanolic Acid Inhibits Liver X Receptor Alpha and Pregnane X Receptor to Attenuate Ligand-Induced Lipogenesis. J. Agric. Food Chem. 2018, 66, 10964–10976.
  43. Svensson, S.; Östberg, T.; Jacobsson, M.; Norström, C.; Stefansson, K.; Hallén, D.; Johansson, I.C.; Zachrisson, K.; Ogg, D.; Jendeberg, L. Crystal structure of the heterodimeric complex of LXRα and RXRβ ligand-binding domains in a fully agonistic conformation. EMBO J. 2003, 22, 4625–4633.
  44. Wada, T.; Gao, J.; Xie, W. PXR and CAR in energy metabolism. Trends Endocrinol. Metab. 2009, 20, 273–279.
  45. Kliewer, S.A.; Goodwin, B.; Willson, T.M. The nuclear pregnane X receptor: A key regulator of xenobiotic metabolism. Endocr. Rev. 2002, 23, 687–702.
  46. Ekins, S.; Chang, C.; Mani, S.; Krasowski, M.D.; Reschly, E.J.; Iyer, M.; Kholodovych, V.; Ai, N.; Welsh, W.J.; Sinz, M.; et al. Human pregnane X receptor antagonists and agonists define molecular requirements for different binding sites. Mol. Pharmacol. 2007, 72, 592–603.
  47. Seow, C.L.; Lau, A.J. Differential activation of pregnane X receptor by carnosic acid, carnosol, ursolic acid, and rosmarinic acid. Pharmacol. Res. 2017, 120, 23–33.
  48. Lin, Y.-N.; Chen, C.-J.; Chang, H.-Y.; Cheng, W.-K.; Lee, Y.-R.; Chen, J.-J.; Lim, Y.-P. Oleanolic Acid-Mediated Inhibition of Pregnane X Receptor and Constitutive Androstane Receptor Attenuates Rifampin-Isoniazid Cytotoxicity. J. Agric. Food Chem. 2017, 65, 8606–8616.
  49. Zhang, Z.-H.; Tang, J.-H.; Zhan, Z.-L.; Zhang, X.-L.; Wu, H.-H.; Hou, Y.-N. Cellular toxicity of isoniazid together with rifampicin and the metabolites of isoniazid on QSG-7701 hepatocytes. Asian Pac. J. Trop. Med. 2012, 5, 306–309.
  50. Prà, M.D.; Carta, D.; Szabadkai, G.; Suman, M.; Frión-Herrera, Y.; Paccagnella, N.; Castellani, G.; De Martin, S.; Ferlin, M.G. Targeting RORs nuclear receptors by novel synthetic steroidal inverse agonists for autoimmune disorders. Bioorg. Med. Chem. 2018, 26, 1686–1704.
  51. Pastwińska, J.; Karaś, K.; Sałkowska, A.; Karwaciak, I.; Chałaśkiewicz, K.; Wojtczak, B.A.; Bachorz, R.A.; Ratajewski, M. Identification of Corosolic and Oleanolic Acids as Molecules Antagonizing the Human RORγT Nuclear Receptor Using the Calculated Fingerprints of the Molecular Similarity. Int. J. Mol. Sci. 2022, 23, 1906.
  52. Solt, L.A.; Burris, T.P. Action of RORs and Their Ligands in (Patho)physiology. Trends Endocrinol. Metab. 2012, 23, 619–627.
  53. Zou, H.; Yang, N.; Zhang, X.; Chen, H.-W. RORγ is a context-specific master regulator of cholesterol biosynthesis and an emerging therapeutic target in cancer and autoimmune diseases. Biochem. Pharmacol. 2022, 196, 114725.
  54. Zou, H.; Zou, H.; Yang, Y.; Yang, Y.; Shi, Z.; Shi, Z.; Wu, X.; Wu, X.; Liu, R.; Liu, R.; et al. Nuclear receptor RORγ inverse agonists/antagonists display tissue- and gene-context selectivity through distinct activities in altering chromatin accessibility and master regulator SREBP2 occupancy. Pharmacol. Res. 2022, 182, 106324.
  55. Wang, Y.; Huang, Z.; Chen, C.Z.; Liu, C.; Evans, C.P.; Gao, A.C.; Zhou, F.; Chen, H.-W. Therapeutic Targeting of MDR1 Expression by RORγ Antagonists Resensitizes Cross-Resistant CRPC to Taxane via Coordinated Induction of Cell Death Programs. Mol. Cancer Ther. 2020, 19, 364–374.
  56. Jin, L.; Martynowski, D.; Zheng, S.; Wada, T.; Xie, W.; Li, Y. Structural basis for hydroxycholesterols as natural ligands of orphan nuclear receptor RORgamma. Mol. Endocrinol. 2010, 24, 923–929.
  57. Kojetin, D.J.; Burris, T.P. REV-ERB and ROR nuclear receptors as drug targets. Nat. Rev. Drug Discov. 2014, 13, 197–216.
  58. Zhou, X.; Chen, H.; Wei, F.; Zhao, Q.; Su, Q.; Lei, Y.; Yin, M.; Tian, X.; Liu, Z.; Yu, B.; et al. The Inhibitory Effects of Pentacyclic Triterpenes from Loquat Leaf against Th17 Differentiation. Immunol. Investig. 2020, 49, 632–647.
  59. Tian, X.; Tang, L.; Wei, F.; Chen, H.; Sheng, L.; Yang, Y.; Zhou, X.; Li, Y.; Xu, X.; Zhang, B.; et al. Pentacyclic triterpene compounds from loquat leaves reduce skin inflammation and epidermal hyperplasia in psoriasis via inhibiting the Th17 cells. Mol. Immunol. 2021, 132, 30–40.
  60. Fujita-Sato, S.; Ito, S.; Isobe, T.; Ohyama, T.; Wakabayashi, K.; Morishita, K.; Ando, O.; Isono, F. Structural Basis of Digoxin That Antagonizes RORγt Receptor Activity and Suppresses Th17 Cell Differentiation and Interleukin (IL)-17 Production. J. Biol. Chem. 2011, 286, 31409–31417.
  61. Lamers, C.; Schubert-Zsilavecz, M.; Merk, D. Medicinal chemistry and pharmacological effects of Farnesoid X Receptor (FXR) antagonists. Curr. Top. Med. Chem. 2014, 14, 2188–2205.
  62. Sun, L.; Cai, J.; Gonzalez, F.J. The role of farnesoid X receptor in metabolic diseases, and gastrointestinal and liver cancer. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 335–347.
  63. Jiang, L.; Zhang, H.; Xiao, D.; Wei, H.; Chen, Y. Farnesoid X receptor (FXR): Structures and ligands. Comput. Struct. Biotechnol. J. 2021, 19, 2148–2159.
  64. Makishima, M.; Okamoto, A.Y.; Repa, J.J.; Tu, H.; Learned, R.M.; Luk, A.; Hull, M.V.; Lustig, K.D.; Mangelsdorf, D.J.; Shan, B. Identification of a nuclear receptor for bile acids. Science 1999, 284, 1362–1365.
  65. Shishodia, S.; Azu, N.; Rosenzweig, J.A.; Jackson, D.A. Guggulsterone for Chemoprevention of Cancer. Curr. Pharm. Des. 2016, 22, 294–306.
  66. Parks, D.J.; Blanchard, S.G.; Bledsoe, R.K.; Chandra, G.; Consler, T.G.; Kliewer, S.A.; Stimmel, J.B.; Willson, T.M.; Zavacki, A.M.; Moore, D.D.; et al. Bile acids: Natural ligands for an orphan nuclear receptor. Science 1999, 284, 1365–1368.
  67. Zhang, Y.; Lee, F.Y.; Barrera, G.; Lee, H.; Vales, C.; Gonzalez, F.J.; Willson, T.M.; Edwards, P.A. Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice. Proc. Natl. Acad. Sci. USA 2006, 103, 1006–1011.
  68. Fang, Y.; Hegazy, L.; Finck, B.N.; Elgendy, B. Recent Advances in the Medicinal Chemistry of Farnesoid X Receptor. J. Med. Chem. 2021, 64, 17545–17571.
  69. Pellicciari, R.; Costantino, G.; Fiorucci, S. Farnesoid X receptor: From structure to potential clinical applications. J. Med. Chem. 2005, 48, 5383–5403.
  70. Shi, X.; Chen, Y.; Zhou, T.; Wang, J.; Xu, X.; Chen, L.; Hu, L.; Shen, X. HS218 as an FXR antagonist suppresses gluconeogenesis by inhibiting FXR binding to PGC-1α promoter. Metabolism 2018, 85, 126–138.
  71. Festa, C.; Finamore, C.; Marchianò, S.; Di Leva, F.S.; Carino, A.; Monti, M.C.; del Gaudio, F.; Ceccacci, S.; Limongelli, V.; Zampella, A.; et al. Investigation around the Oxadiazole Core in the Discovery of a New Chemotype of Potent and Selective FXR Antagonists. ACS Med. Chem. Lett. 2019, 10, 504–510.
  72. Mudaliar, S.; Henry, R.R.; Sanyal, A.J.; Morrow, L.; Marschall, H.; Kipnes, M.; Adorini, L.; Sciacca, C.I.; Clopton, P.; Castelloe, E.; et al. Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease. Gastroenterology 2013, 145, 574–582.e1.
  73. Sun, Y.; Zhang, W.; Li, N.; Guo, S.; Gao, L.; Ge, N. Effect of Ursolic Acid Extracted from Hippophae rhamnoides L. on FXR Signaling Pathway in Liver of Rats with Alcoholic Liver Injury. Sci. Technol. Food Ind. 2023, 44, 363–370.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 311
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 23 Oct 2023
1000/1000