Next Article in Journal
Does Ecological Agriculture Moderate the Relationship between Wine Tourism and Economic Performance? A Structural Equation Analysis Applied to the Ribera del Duero Wine Context
Next Article in Special Issue
In Vivo Hypolipidemic Effects and Antioxidant Capacity of Pinus morrisonicola Hay Extracts by Supercritical Carbon Dioxide Extraction
Previous Article in Journal
Research on Laying Hens Feeding Behavior Detection and Model Visualization Based on Convolutional Neural Network
Previous Article in Special Issue
Anti-Proliferative and Anti-Metastatic Effects of Ethanol Extract from Cajanus cajan (L.) Millsp. Roots and its Sub-Fractions in Oral Squamous Cell Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Application and Development of Natural Plant Metabolite Oleanolic Acid in the Nano Era

1
College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
2
Joint International Research Laboratory of Agriculture & Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou 225009, China
*
Author to whom correspondence should be addressed.
Agriculture 2022, 12(12), 2142; https://doi.org/10.3390/agriculture12122142
Submission received: 16 November 2022 / Revised: 7 December 2022 / Accepted: 12 December 2022 / Published: 13 December 2022

Abstract

:
Like other pentacyclic triterpenoids, oleanolic acid, a natural plant metabolite prevalent in plant peels, stems, and leaves, is regarded as a possible drug candidate. A growing number of studies have shown that oleanolic acid exhibits a variety of beneficial properties, including antiviral, anti-inflammatory, antioxidant, anticancer, and hepatoprotective effects. Additionally, the rapid advance of nanotechnology has dramatically improved oleanolic acid’s bioavailability and minimized its disadvantages, leading to unexpected changes in its pharmacological activity and use. Therefore, our aim was to review the progress of research on the distribution and biological properties of oleanolic acid in plants and to discuss new pharmaceutical approaches for oleanolic acid.

1. Introduction

It is widely recognized that terpenoids are among the very common and most complex natural chemicals found, richly, in plants in nature, and they have a wide range of pharmacological effects [1]. They are recognized as potential drug compounds, among which sesquiterpenoids, represented by artemisinin, are well-known for their remarkable therapeutic effects on malaria [2]. In recent years, researchers have been focusing more on the extraction, analysis, and structural modification of the isolated active ingredients from the natural products of terpene-containing plants. Additionally, the recent decades have seen a surge in interest in triterpene-rich plant extracts, particularly pure triterpenoids such as bioactive phytochemicals [3,4,5,6].
There are two subclasses of triterpenoids based on their chemical structure: pentacyclic triterpenes and tetracyclic triterpenes [7]. Among them, oleanolic acid (OA, Figure 1) and its isomer ursolic acid (UA, Figure 1) are pentacyclic triterpenoid bioactive substances commonly found in fruits and vegetables, and both possess similar physicochemical and biological properties, such as anti-inflammatory, antioxidant, antiviral, anticancer, antidiabetic, hepatoprotective, and cardioprotective effects [8,9,10,11,12,13,14,15]. OA research has gained increasing attention due to its rich biological properties. In the pharmaceutical field, however, its development is hindered, and it is unable to fully exploit its therapeutic effects as a result of its poor water solubility and low bioavailability when taken orally [16]. To address these challenges, a number of new technologies have been developed by researchers, such as preparing new dosage forms of OA and modifying its molecular formula. In particular, nanotechnology, embodied by nanoparticles, as a novel and efficient method of drug preparation, exhibits remarkable effects on improving the dissolution, penetration, and absorption of OA, and its unique combination makes drugs containing OA achieve precise targeting effects [17,18,19].
In this review, we first examine the most recent findings of OA in vivo and in vitro experimental models, its biological features, and the state of related research and then describe and assess the various methods used to manufacture OA and their bioavailability. The purpose of this study is to serve as a reference for the future research, development, and application of OA.

2. Sources of Oleanolic Acid

Oleanolic acid (3β-hydroxyolean-12-en-28-oic acid) is a natural pentacyclic triterpenoid, with a name that is derived from the plant Olea europaea, the primary source of commercial OA preparations at the moment [20]. Pentacyclic triterpenes, important phytochemicals synthesized by the cyclization of squalene in plants, are known as a large class of plant secondary metabolites composed of isoprene (2-methylbutadiene) units [21]. Oleanane, hopane, friedelane, ursane, gammacerane, and lupine are major pentacyclic triterpenes that mainly exhibit antiviral, antitumor, anti-inflammatory, and antioxidant activities [22].
Oleanolic acid can be extracted from different parts of Araliaceae, Asteraceae, Ericaceae, Lamiaceae, Myrtaceae, Oleaceae, Rosaceae, Rubiaceae, Saxifragaceae, and Verbenaceae plants, but its content differs from one plant to another and in different parts of the same plant (Table 1). For instance, studies have shown significant differences in OA content in the fresh leaves and persimmon pulp of different peony varieties [23,24]. Additionally, certain plants have only one organ that contains OA. In birch, for instance, OA is exclusively found in the bark (up to 11 mg/g DW) [25]. OA can also be found in garden thyme and clove plants. Some of the fruit plants where OA has been found and isolated are apple, loquat, grape, elderberry, and sage [25,26]. Notably, a growing number of researchers have examined the abundance of OA in apples, particularly in apple peels, due to their rich content of triterpenoids’ active substances and their ubiquity in the daily diet. As shown by the test results, OA and UA are the main triterpenoid active substances in apple peel extract [27,28]. The level of OA in apple flesh is found to reach 0.28 g/100 g in published research [25]. Although apples are an important food source for humans and a raw material in the food industry, only a small fraction of their leaves are used in certain foods, and the vast majority are underutilized as agricultural waste. To explore new sources of natural antioxidants, anticancer, and anti-inflammatory drugs, researchers have developed new methods for the extraction and detection of OA from apples, such as techniques to enhance extraction rates, including ultrasound-assisted extraction, microwave-assisted extraction, and supercritical fluid carbon dioxide [29,30,31]. Chromatographic techniques that can be used to assess the quantitative and qualitative composition of triterpenoids include high-performance liquid chromatography, gas chromatography, and thin-layer chromatography combined with tandem mass spectrometry [32,33].

3. Pharmacological Effects of Oleanolic Acid

In spite of its wide range of pharmacological properties, OA’s therapeutic potential has only been partially exploited. Studies on triterpenoids have shown that OA exerts beneficial effects through various signaling pathways, such as antiviral, anti-HIV, antibacterial, anticancer, antidiabetic, anti-inflammatory, hepatoprotective, gastroprotective, hypolipidemic, and anti-atherosclerotic effects (Table 2). Apart from its beneficial role in preventing cardiovascular disease, obesity, and metabolic syndrome, OA improves insulin response, maintains beta cell function and survival, and prevents diabetic complications [63,64].

3.1. Anti-Inflammatory Effect of OA

The capacity of OA to alter a number of anti-inflammatory pathways has been demonstrated in animal studies and cellular research. It can significantly reduce inflammation and inhibit numerous inflammatory diseases, such as vasculitis, enteritis, and bronchitis [90,91,92,93]. The anti-inflammatory effect of OA is exerted through a variety of complex mechanisms.

3.1.1. Inhibition of the Production of Pro-Inflammatory Cytokines

In the context of inflammation and oxidative stress, treatment with OA and its derivatives significantly inhibits the production of pro-inflammatory cytokines (IL-1β, IL-6, IL-8, TNF-α, and MCP-1) [68,94,95,96]. Researchers found that high-fat diet C57BL/6J female mice given oleanolic acid in water feeders at 0.005% (w/v) for 16 weeks had a significant reduction in the expression levels of IL-1β, IL-6, and TNFα in adipose tissue [97]. The researchers pretreated BV2 cells with OA (0.5 to 10 μM) for 1 h and then with LPS (100 ng/mL) for 24 h at 37 °C. The results showed that OA reduced the expression levels of TNF-α, IL-1β, and IL-6 in BV2 cells [98]. Researchers have demonstrated that OA ameliorates experimental autoimmune myocarditis (EAM) in several ways, including by promoting anti-inflammatory cytokines (IL-10 and IL-35), interfering with cardiac-specific autoantibody production, and exerting direct protective effects on cardiac cells [15]. In female C57BL/J6 mice with experimental autoimmune encephalomyelitis, OA has also been revealed to decrease pro-inflammatory mediators (TNF-α, IL-1β, IL-23, IL-17A, chemokine KC, and the growth factor IGF-1) both in serum and colonic tissue, prevent lipid peroxidation and superoxide anion accumulation in intestinal tissue, and induce the expression of the ROS scavenger Sestrin-3 [99].

3.1.2. Increase Antioxidant Production

The anti-inflammatory effect of OA is related to the regulation of antioxidant production. This mechanism is related to the expression of transcription factors Nrf2 and MAP kinase that are oxidative-stress-sensitive [100,101]. OA has also been proven to protect rats from ethanol-induced liver injury by inducing Nrf2-associated antioxidants, thereby maintaining redox homeostasis and modulating ethanol metabolism and inflammatory pathways [102]. As a ubiquitous nuclear transcription factor, NF-κB participates in the transcription of target genes and regulates cell proliferation, differentiation, growth, and apoptosis. As reported in previous studies, OA suppresses inflammation by inhibiting NF-κB signaling and regulating pro-inflammatory cytokines such as TLR-9 and IL-18 [66,103].

3.1.3. Inhibition of Activation of Mitochondria-Associated Inflammatory Vesicles

Mitochondria may be a basic target when OA improves inflammation, as they are organelles that contribute to oxidative stress and neuroinflammation [104]. In mitochondrial organelles, an essential component of innate immune response is DNA synthesis, and mitochondrial DNA binds to NLRP3 containing inflammasome to activate inflammasome [105,106]. There is evidence that OA ameliorates carotid artery injury in diabetic Sprague Dawley rats by inhibiting the NLRP3 inflammasome signaling pathway. Researchers induced a diabetic rat model with streptozotocin (60 mg/kg), followed by 2 weeks of treatment with OA (100 mg/kg/day) injection. OA reversed the hyperglycemia-induced upregulation of the NLRP3 inflammasome components in diabetic rats. In addition, the levels of TNF-α, interleukin (IL)-1β, IL-6, and IL-18 were downregulated in the serum of OA-treated diabetic rats [70]. Additionally, through the inhibition of NLRP3 inflammasome activation, OA attenuates microglia activation and oxidative stress, thereby achieving the neuroprotective effect of OA against ischemic stroke. Researchers found that OA (10 mg/kg, p.o.) attenuates NLRP3 inflammasome activation in a male ICR mouse model of transient middle cerebral artery occlusion. Moreover, OA inhibits the assembly of NLRP3 inflammasome in the injured brain after ischemic stroke [71]. Since various diseases are strongly associated with the activation of NLRP3 and its related molecular regulatory signaling pathways, NLRP3 is an exciting frontier for clinical drug research and development [107]. Effects such as anti-inflammatory or antitumor, which are attributed to inhibiting the signaling pathway of NLRP3 inflammasome, reveal potential targets of OA as an anti-inflammatory agent. In contrast, the highly variable nature of NLRP3 inflammasome agonists allows them to be triggered by many different stimuli, and the specific mechanisms behind this need to be identified in more detail.

3.2. Antitumor Effect of Oleanolic Acid

Compared with synthetic compounds, the natural product OA exhibits rich structural diversity and better preventive and anticancer-drug-like properties. It has become one of the research hotspots for anticancer drug development. OA was found to be effective in treating various tumor cell lines, such as MCF-7 and MCF-7/ADR human breast cancer cells, the 1321N1 astrocytoma cell line, hepatocellular carcinoma, and HCT-116 colorectal cancer cells [108]. The antitumor mechanisms of OA mainly include anticancer cell proliferation, induction of apoptosis, induction of autophagy, regulation of cell cycle regulatory proteins, inhibition of vascular endothelial growth, anti-angiogenesis, and inhibition of tumor cell migration and invasion (Figure 2). More importantly, the use of OA for cancer treatment was found to be less toxic to cancer cells than to normal cells and more biosafe than other treatments [109]. Therefore, OA exhibits considerable potential in anticancer applications.

3.2.1. Inhibition of Tumor Cell Proliferation

Oleanolic acid exhibits inhibition of tumor cell proliferation by participating in multiple anticancer signaling pathways and intracellular substance regulation, such as the Akt/mTOR/S6K pathway and the inhibition of macrophage M2 polarization. Mouse model experiments have shown that OA inhibits the proliferation of KRAS-transformed normal cells by attenuating the Akt/mTOR/S6K signaling cascade. In addition, OA also inhibits cancer cell proliferation by suppressing both the mTOR signaling pathway and PI3K/Akt/mTOR signaling pathway [110]. According to experimental findings, OA significantly inhibits the expression of CD163, one of the phenotypic markers of M2 macrophages, and the secretion of IL-10, an anti-inflammatory cytokine produced by M2 macrophages, suggesting that OA’s inhibition of cell proliferation occurs through the suppression of the M2 polarization of macrophages [111].
Notably, the reduction in aerobic glycolysis and glycolytic enzymes is suggested to be a mechanism by which OA inhibits tumor cell proliferation. As shown in studies, there is a positive correlation between aerobic glycolysis and cancer cell proliferation, and a high-sugar environment promotes both aerobic glycolysis and cancer cell proliferation [112]. In gastric cancer cells, the proliferation of gastric tumor cells is largely dependent on aerobic glycolysis, whereas OA can reduce HIF-1α-mediated aerobic glycolysis by inhibiting YAP. The specific mechanism is that OA can inhibit glucose uptake and consumption and downregulate the expression of the glycolytic-rate-limiting enzymes HK2 and PFK1 [11]. It is noteworthy that a novel signaling pathway called Hippo-Yap plays a critical role in the growth and progression of tumors, making it a potential therapeutic target for OA in cancer treatment.
There are multiple pathways by which OA inhibits tumor cell proliferation, including reducing the expression of Bcl-2, Cyclin D1, and CKD4, promoting the expression of Bax and p21, enhancing the activation of p53, etc. [113]. The proliferation of human glioblastoma cells U373 was found to be inhibited by OA when OA activates STAT3 in human macrophages and glioblastoma cells [110]. Moreover, a reduction in gastric cancer cells proliferation is spotted when OA inhibits the expression of cell cycle protein A and cell cycle protein-dependent kinase 2 [114].

3.2.2. Induction of Apoptosis in Tumor Cells

One of the key pathways in tumor therapy is the induction of apoptosis. There is evidence that pentacyclic triterpenoids interfere with multiple stages of cancer development, inhibit tumorigenesis and evolution, and induce apoptosis in several cancers [115,116]. For instance, a variety of tumor cells are induced to undergo apoptosis by OA, including HepG2 cells, human breast cancer MCF-7 cells, HT-29 colon cancer cells, and prostate cancer cells [85,117].
Multiple signaling pathways are involved in the OA-induced apoptosis in tumor cells. One of them is the inhibition of NF-κB activity [118]. OA was shown to inhibit NF-κB expression and regulate the mRNA expression level of the X-linked inhibitor of the apoptosis protein (XIAP) in HuH7 cells compared to untreated cells [119]. Another proven mechanism that promotes apoptosis is that OA inhibits COX-2 overexpression in tumor cells [120]. Here, COX-2 is overexpressed in a variety of tumor cells as a rate-limiting enzyme for prostaglandin synthesis, suggesting its participation in cancer promotion and progression [121]. In human colon cancer HT-29 cells and HepG2 cells, OA is reported to decrease COX-2 protein activity and inducible nitric oxide synthase (INOS) expression, thereby inducing apoptosis [73,122].

3.2.3. Induction of Autophagy

There is an association between OA’s anticancer properties and cellular autophagy. As evidenced by the elevated ROS levels in cells after OA treatment, OA-triggered cellular autophagy proceeds in an ROS-dependent manner. A significant inhibition of the growth of HepG2 and SMC7721 cells is achieved as a result of autophagy induction and inhibition of the PI3K/Akt1/mTOR signaling pathway in response to OA treatment [123]. According to another study in bladder cancer T24 cells (ULK1), the mechanisms through which OA causes autophagy include the activation of AMP-activated protein kinase (AMPK), the blocking of the mTOR molecular target, and the increase in UNC-51-like autophagy-activated kinase 1 [124]. There is further evidence that the inhibition of the Akt/mTOR/S6K pathway contributes to OA-induced autophagy [110]. Moreover, most natural compounds were found to induce autophagy primarily through the Akt/mTOR/S6K pathway [125]. There are other autophagy regulatory pathways, such as ERK and JNK, which were shown to be activated in response to OA treatment, indicating their possible association with OA-induced autophagy [126]. It is, therefore, imperative to conduct more research to understand the mechanism of OA-induced apoptosis, which is crucial for the creation of OA-related anticancer medications.

3.2.4. Regulation of Cell Cycle Regulatory Proteins

In different cancers, the expression of cell cycle regulatory proteins may be affected by OA in different ways, resulting in cell cycle arrest at different stages and apoptosis induction in cancer cells. In prostate cancer PC-3, DU145, and LNCaP cells, OA is proven to promote G0/G1 phase cell cycle arrest in a dose-dependent manner by regulating the expression levels of cell-cycle-related proteins. The researchers discovered that certain doses of OA independently inhibit cell cycle progression. In cell lines pretreated with different doses of OA (15, 30, 45, and 60 μM), there was a significant increase in the percentage of cells in the G0/G1 phase but a considerate reduction in the percentage of cells in S phase, as compared to controls [42]. According to previous findings, OA-treated DU145 cells are arrested in G2 because of the activation of p-AKT, p-JNK, p21, and p27, accompanied by the downregulation of p-ERK, cyclin B1, and CDK2 expression. OA-treated MCF-7 cells are reported to be arrested in G1 owing to the activation of p-JNK, p-ERK, p21, and p27 and the reduction in p-AKT, cyclin D1, CDK4, cyclin E, and CDK2 expression. Moreover, OA-treated U87 cells also exhibit G1 phase arrest caused by the upregulation of p-ERK, p-JNK, p-AKT, p21, and p27 and the downregulation of cyclin D1, CDK4, cyclin E, and CDK2. It is, thus, concluded that OA arrests the cell cycle at different phases and induces apoptosis in cancer cells [74]. According to these results, different cancers are affected differently by OA’s effects on the expression of cell cycle regulatory proteins.
In addition, when it acts on certain receptors or enzymes, OA induces different damages to cellular DNA as an active immunomodulatory component, thereby inhibiting the growth migration, invasion, and progression of tumor cells [127].

3.3. Other Pharmacological Effects

OA is not only protective against acute chemical liver injury, but also against chronic liver-disease-induced fibrosis and cirrhosis [128]. There are several altered gene expression patterns and transcription factors that may contribute to this pathway, including farnesoid x receptor (FXR) and Nrf2-, and MT-related genes [129,130]. By encouraging Nrf2 nuclear accumulation, OA can cause Nrf2-dependent gene deinduction, thus protecting the liver from acetaminophen-induced hepatotoxicity [131].
As demonstrated in another study, after 30 days of oral treatment with OA (10 mg/kg/day) and ethanol (4 g/kg/day), a significant reduction in histopathological damage and serum lipid abnormalities as well as biochemical indices of ethanol-induced oxidative stress, such as elevated lipid hydrogen peroxide in the liver, GSH depletion, and decreased antioxidant enzyme activity, was observed in mice [102]. Based on these findings, OA significantly protects against the development of liver injury through various pathways.
Notably, OA displays great potential as a natural compound for the synthesis of potential antidepressant drugs [132]. It is also evidenced to have low side effects when used as an antidepressant, which further excites researchers [133,134]. Thus, OA is increasingly used by researchers to develop antidepressant drugs. In mice exposed to chronic stress, researchers found that OA (20 mg/kg) can activate the hippocampal brain-derived neu-rotrophic factor (BDNF)-ERK-CREB signaling pathway by regulating miR-132, which produced an antidepressant-like effect [135,136]. Another study revealed that OA (40 mg/kg) exhibited antidepressant-like effects in corticosterone-induced depression through downregulation of SGK1 and GR expression and upregulation of the hippocampal BDNF-AKT/mTOR signaling pathway [137].
Other properties of OA, such as anti-prostate cell proliferation, anti-muscle atrophy, and anti-influenza, have also been demonstrated [137,138,139,140,141]. The effects of OA on cardiovascular activities include anti-arrhythmic, immunomodulatory, anti-hyperlipidemic, vasodilatory, anti-inflammatory, and antioxidant [142,143,144]. In the liver, OA was shown to control glucose 6 phosphates and forkhead box protein O1, thereby normalizing blood glucose levels in rodents with diet-induced obesity or diabetes [145,146]. It was also found that OA inhibits atherosclerosis by downregulating the expression of inducible nitric oxide synthase in apolipoprotein E knockout mice [147]. In conclusion, OA exerts diverse and complex pharmacological effects, making it a promising drug ingredient, but its mechanism requires further investigation.

4. Development and Utilization of Oleanolic Acid

In the biopharmaceutical classification system, pentacyclic triterpenoids are categorized as Class IV medications. As they are almost insoluble in water and have difficulty penetrating biofilms, their pharmacological actions are limited [148]. Another well-known issue of triterpenoids is their low bioavailability. This is primary because their high lipophilicity and water insolubility can significantly decrease the effectiveness of medication absorption in the gastrointestinal system [149]. Thus, researchers are working on exploring new methods to enhance the biopharmaceutical properties of OA. Two principal ways have been found to improve its water solubility, permeability, and bioavailability. One is to modify the molecular formula of OA to obtain higher biological activity and a wide range of derivatives, which can serve as the basis for the development of new drugs [85,128]. The other is the preparation of new dosage forms of OA, such as nanoparticles, liposomes, solid dispersions, and phospholipid complexes, to improve its dissolution, penetration, and absorption [18]. Among them, nanoparticle drugs are becoming increasingly popular among researchers due to their extended drug circulation time, high bioavailability, targeted delivery, and higher bioavailability.

4.1. Structural Modification of Oleanolic Acid

It is possible to improve bioavailability and expand the scope of application of OA by chemically altering its structure, which is an effective way to develop new drugs. The C-28 position, A ring, and C ring are the main structural modification sites of OA, and derivatives modified at different positions exhibit a variety of pharmacological activities (Figure 3) [128]. Its diverse biological properties, coupled with its high availability and low production cost, make OA an excellent semi-synthetic modified precursor molecule [150].
Many analogs of OA have been synthesized and have exhibited excellent biological activity [128]. Two compounds were prepared from OA isolated from Syzygium aromaticum by methylation and acetylation, which have exhibited better in vivo/in vitro anti-inflammatory and membrane stabilization properties, respectively [151,152,153]. A newly synthesized OA derivative was revealed to have a significant inhibitory effect in vitro and substantially reduced blood glucose levels compared with OA in in vivo experiments [154]. In lung cancer cells, recently synthesized OA family compound 2 (OLO-2) olean-28 and 13b-olide are shown to promote apoptosis by activating caspase-3, producing ROS, causing DNA damage, and blocking the activation of the ERK, STAT3, AKT, and NF-κB pathways [155]. Another OA derivative, named SZC017, was also prepared by modifying the molecular structure. It can inhibit levels of Akt, phosphorylated-Akt (p-Akt), p-IκBα, total p65, and total p-p65 p-p65 in both the cytoplasm and nucleus and the p65 nuclear translocation to suppress Akt/NF-κB signaling and topoisomerase I and IIα proteins, thus initiating the intrinsic apoptosis of gastric cancer cells [156].
Recently developed OA derivatives have been proven to mitigate OA’s weak water solubility, allowing it to demonstrate greater bioavailability. A new OA derivative was revealed to be nearly 20 times more soluble than OA in aqueous solution, release higher amounts of NO in HCC than the normal compound, and exhibit potent anti-HCC activity with little effect on normal hepatocytes [157]. Cyclodextrin encapsulation technology significantly contributes to the synthesis of OA. Tetra-ethylene pentaamine-β-cyclodextrin, a synthetic long-chain amino-β-cyclodextrin derivative, was adopted to prepare OA inclusion complexes, and the finding showed a 2100-fold increase in the water solubility of OA [128]. Researchers also found a drastic promotion of the water solubility of OA by inclusion complexation with amino-appended β-cyclodextrins (ACDs). Moreover, the in vitro anticancer activities of OA against human cancer cell lines HepG2, HT29, and HCT116 are significantly enhanced after the formation of inclusion complexes, and the apoptotic-response results indicate their induction during the apoptosis of cancer cells [158]. This could provide a novel approach to developing novel pharmaceutical formulations of OA. It is noteworthy that the new OA-preparation technique does improve solubility, but its metabolic absorption and bioavailability in vivo have not been sufficiently studied.
While great progress has been made in the structural modification of OA, there are still some issues that need to be explored. Newly synthesized derivatives, for instance, have had few innovative studies, modified dosage forms have not been biologically tested, and OA derivatives have reduced or not significantly improved activity. Taken together, it is necessary to investigate functional groups, pharmacological effects, and unnecessary substituents in chemical-structure modification to make accurate and reasonable modifications.

4.2. Nanoscale Preparation

Nanotechnology has revolutionized the pharmaceutical and biotechnology industries in the 21st century and is seen as a powerful tool for basic research, imaging, and, particularly, enhanced drug delivery. With nano-drug delivery systems, drugs can be delivered at lower doses but perform better therapeutic effects, while improving product bioavailability, safety, and patient compliance [159]. These systems also offer the possibility to deliver highly lipophilic or chemically unstable drugs, thus improving OA’s bioavailability, which is hindered by the highly lipophilic and water-insoluble nature of triterpenoids. Moreover, several disadvantages of many natural compounds with poor therapeutic applicability can be addressed by nanocarriers, such as in vivo instability, reduced bioavailability and solubility, low absorption, lack of targeted delivery, and adverse effects. Moreover, nano-based formulations can significantly improve pharmacokinetic parameters and reduce interactions with intracellular proteins, thereby improving the bioavailability of highly lipophilic drugs and achieving targeted delivery to specific sites [160]. In recent years, nanotechnology has been adopted by many researchers to prepare OA.

4.2.1. Nanoliposomes

Liposomes are one of the various nanoparticles that have attracted particular attention because of their structural similarity to biological membranes, high drug loading, and high transport capacity. They possess unique physical and chemical properties as well as high potency and the ability to encapsulate a large number of different molecules [161]. In addition, this type of nanoformulation is highly flexible in preparation, making it possible to undergo different surface modifications for a wide range of applications in skin applications and gene therapy or as drug delivery systems for different diseases [162]. Nanoliposomes developed from OA show slow release, controlled release, high targeting, reduced toxic side effects, and enhanced antitumor effects [163].
In addition, OA’s bioavailability is enormously enhanced after being combined with liposomes (Figure 4). Researchers encapsulated OA in PEGylated liposomes, resulting in good stability, solubility, and diffusion permeability, along with significant drug-carrying capacity and slow in vitro drug release. These OA liposomes have shown superior anticancer activity compared to pure OA and longer drug action through avoidance of Opsonization and macrophage uptake [164]. Chitosan-mediated gold-nanoshell-encapsulated OA liposomes (GNOLs) were also developed by researchers, obtaining a product with an average diameter of 172.03 nm. With a suitable zeta potential, this product is more likely to accumulate in tumor cells. Based on results of in vivo evaluation experiments of GNOLs in U14 tumor-bearing mice, GNOLs exhibit significant inhibitory and pro-apoptotic effects on tumor tissue [163]. Further, liposomes-encapsulated OA is reported to show stronger antitumor activity by synthetic drugs on HeLa cells compared to free OA [158]. The nanoliposome-covered OA is also proved to enhance the anticancer effect by inhibiting proliferation, migration, and invasion [165].
Oleanolic acid liposomes can attenuate the organ toxicity of related chemotherapeutic drugs. As a result of the combination of OA liposomes and doxorubicin, the effective dose of both compounds is reduced, and the organ toxicity of doxorubicin is also eliminated without reducing its anticancer effect. Based on histopathological evaluation, liver, kidney, or heart tissues do not show toxic activity, which can be attributed to OA’s protective antioxidant properties that protect organs from oxidative stress [166]. A combination of doxorubicin and OA exhibits limited cardiotoxicity without any evidence of histopathological changes in major organs, making it a promising strategy for the future treatment of hepatocellular carcinoma [167].
Notably, some researchers have developed a multivesicular liposome containing OA (OA-MVLs) as a treatment for hepatocellular carcinoma. Compared with pure OA, this compound inhibits the growth of human HepG2 cells and mouse H22 hepatoma more effectively. It exhibits a prolonged drug-circulation time, as a result of the continuous release of active drugs from liposome nanoparticles in in vitro and in vivo experiments. Moreover, OA-MVLs can inhibit the adhesion, migration, and invasion of hepatocellular carcinoma cells at low doses without damaging normal hepatocytes [168].
Taken together, OA-MVLs are considered as a potential drug candidate for cancer treatment in the future as a result of their simple preparation and promising biological effects.

4.2.2. Nanoparticles

Biodegradable polymeric nanoparticles (NPs) offer an excellent option for developing and utilizing OA. In comparison with conventional drug formulations, NPs can slow down the drug release rate, prolong the drug-circulation time, reduce the required dose of drug delivery, and improve the pharmacodynamic effects of a drug in preclinical models [18,128]. In particular, since NPs have an increased permeability and retention effect (EPR), they preferentially collect in tumor and inflammatory tissues, avoiding clearance in the spleen (Figure 5) [169,170]. Incorporating OA into NPs, we can achieve slow release and targetivity, improve antitumor effects, enhance drug safety, and expand drug delivery [17,171,172]. Researchers have developed a novel OA NP-loaded lactoferrin nano-delivery system. OA-NPs disintegrate twice as fast as control OA in in vitro dissolution experiments, and pharmacokinetic studies in rats show a 320.5% relative bioavailability of OA NPs [172]. The ring-opening polymerization (ROP) method was used to synthesize a series of amphiphilic carboxylated cellulose-graft-Poly (L-lactide) (CC-g-PLLA) copolymers that can self-assemble into NPs for delivery of anticancer drug OA. The copolymer (DSPLLA 2.03) NPs display higher drug loading efficiency (24.76 ± 0.58%). In addition, NPs exhibit better water solubility (16.9 mg/mL) and a prolonged drug release (120 h). In vitro and in vivo studies indicate that NPs maintain cytotoxicity to 4T1 cells and MCF-7 cells and display high antitumor efficiency [173].
Notably, there is a growing interest in the combination therapy with NPs in antitumor studies. It is essential for drug-loaded NPs to avoid elimination by the reticuloendothelial system (RES) to achieve long cycle times. Hydrophilic agents such as polyethylene glycol (PEG) are usually used to modify the surface of NPs to avoid clearance by phagocytosis. This mechanism alters the physicochemical properties of NPs, thus changing the properties of NPs, such as the drug-release profile, biodistribution, and pharmacokinetics [164]. A hydrophobic OA core and a hydrophilic PEG shell were used by researchers to successfully synthesize mPEG-OA NPs that are also capable of encapsulating another anticancer drug hydroxycamptothecin (HCPT) to achieve synergistic effects. As a result, mPEG-OA/HCPT NPs exhibit significantly enhanced anticancer efficacy compared to free drug formulations [174]. Additionally, a range of desirable properties are also exhibited by these co-assembled NPs for co-delivery of anticancer drugs, such as good water solubility, appropriate size, low side effects, and greater bioavailability. Further, researchers constructed a cancer cell membrane-decorated zeolitic-imidazolate framework hybrid nanoparticle (HP) to codeliver cisplatin (DDP) and OA; the results revealed that this platform (HP/DDP/OLA) displays positive feedback in the treatment of bladder cancer (SW780). Moreover, it can enhance apoptosis, while reversing multidrug resistance in SW780 cells, compared to free drugs alone or monodelivery systems [175].
A pure natural nanomedicine-cum-carrier (NMC) drug delivery system was also designed and fabricated based on bioactive nanomaterials. This system can co-assemble OA and GA into NPs with new morphological characteristics. Natural small-molecule drug carriers retain biological activity and have stronger antitumor effects than non-bioactive drug carriers. Both in vivo and in vitro experiments have confirmed that OA and GA work synergistically to significantly enhance the therapeutic efficacy on tumor therapeutic efficacy. A major reason for this is that OA and GA kill tumors in different mechanisms. In addition, OA/GA NPs are capable of loading up to 15% paclitaxel (PTX). Compared to OA/GA NPs, OA/GA/PTX NPs exhibit enhanced antitumor activity [17].

4.2.3. Other Nanoscale Preparation Methods

An ethanol film hydration was used to prepare OA-loaded hybrid micelles, which were then characterized and evaluated for in vitro release and in vivo drug efficacy. As shown in in vitro drug release studies, approximately 80% of OA was released from the dialysis bag within 24 h, while only 40% of OA micelles was, indicating that the OA micelles achieved a sustained release. In addition, compared to free OA, higher antitumor efficacy was observed with the OA micelles. They also significantly reduced tumor volume and inhibited tumor invasion and migration [176]. Herein, it is safe to conclude that polymer micelles are a promising anticancer-drug delivery system.
OA-loaded nanoemulsions with an average particle size of less than 60 nm were prepared. They were proven to have excellent and stable physical properties based on stability test studies. As evidenced in in vivo experiments, their formulation was non-toxic and non-irritating to the skin. It also had a high skin-penetration ability and can enhance the anti-inflammatory effect of OA. After absorption through the skin, the nanoemulsion made from OA provided a better anti-inflammatory effect [177]. After formulating OA into a nanosuspension, there was an increase of about 550 times in the saturation solubility of OA [178].
In comparison with conventional therapies, nanotechnology has led to greater drug development potential for OA in the treatment of diseases such as cancer. It is possible to further exploit this potential by selecting suitable fabrication methods and nanocarrier-delivery routes to achieve better bioavailability.

5. Conclusions

Oleanolic acid is a natural pentacyclic triterpenoid that can be extracted from various parts of a wide range of plants. Over the past few decades, the potential anti-inflammatory, anticancer, hepatoprotective, and cardioprotective effects of OA and its derivatives have attracted increasing attention from researchers. According to prior studies, OA exerts anti-inflammatory effects by inhibiting the production of pro-inflammatory cytokines, which increases the production of antioxidants or inhibits the activation of mitochondria-associated inflammatory vesicles. Its inflammatory regulatory process involves signaling pathways such as NF-ΚB and NLRP3. Aside from inhibiting tumor cell proliferation, OA induces apoptosis and autophagy in tumor cells and regulates cell cycle regulatory proteins. It is also possible that the anticancer activity of OA is associated with cell death due to the activation or sensitization of the intracellular pathways of pentacyclic triterpenoids.
Like many other plant compounds, OA possesses many beneficial pharmacological properties. However, its obvious drawbacks cannot be ignored, such as poor water solubility and low bioavailability, which are possibly attributed to the lipophilic nature and low water solubility of OA. Various techniques have been investigated to overcome these drawbacks, such as appropriate structural modification of OA and the application of bio-nanotechnology. In recent years, great progress has been made in the chemical structure modification of OA, and new formulations of OA and its derivatives have sprung up. While sharing some pharmacological properties with OA to some extent, OA derivatives display some distinctive advantages, such as less toxicity, stronger therapeutic effects, and greater bioavailability. As OA NPs are made by nanotechnology, their surface possesses a high modulation that allows them to transport hydrophilic and lipophilic molecules, thus ensuring excellent bioavailability of a drug at specific sites of action. In addition, OA NPs can significantly enhance the solubility of OA and contribute to the distribution of the drug within the body. As compared to monomeric systems, co-assembled NPs and co-loaded hybrid NPs exhibit improved stability and biocompatibility, better responsiveness of drug release, and better performance.
Overall, OA, which is widely found in nature, displays strong biological activity and great research potential. However, there are still some challenges remaining to be addressed in its application into drug development, such as its loading capacity, stability, toxicity, and ability to overcome biological barriers. It is important to utilize appropriate technologies to enhance its biomedical activity. We are confident that with further research and the discovery of new dosage forms, the research and application of OA will be greatly facilitated. More researchers need to explore products that benefit human beings as soon as possible.

Author Contributions

Writing—original draft preparation, K.W.; writing—review and editing, Q.L., X.C., Y.W. (Yuhao Wang), and Y.W. (Yanni Wu); supervision, Z.C. and Z.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This review was supported by the Jiangsu Agricultural Industry Technology System, JATS [2022]486, and the Seed Industry Vitalization Program of Jiangsu Province, JBGS [2021]115.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

None of the authors have any conflict of interest to declare.

References

  1. Laszczyk, M.N. Pentacyclic triterpenes of the lupane, oleanane and ursane group as tools in cancer therapy. Planta Med. 2009, 75, 1549–1560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Tu, Y. The discovery of artemisinin (qinghaosu) and gifts from Chinese medicine. Nat. Med. 2011, 17, 1217–1220. [Google Scholar] [CrossRef] [PubMed]
  3. Fan, Z.; Wang, Y.; Yang, C.; Zhou, Z.; Wang, P.; Yan, X. Identification of a novel multifunctional oxidosqualene cyclase from Zea mays sheds light on the biosynthetic pathway of three pentacyclic triterpenoids. Synth. Syst. Biotechnol. 2022, 7, 1167–1172. [Google Scholar] [CrossRef]
  4. Kashyap, D.; Tuli, H.S.; Sharma, A.K. Ursolic acid (UA): A metabolite with promising therapeutic potential. Life Sci. 2016, 146, 201–213. [Google Scholar] [CrossRef] [PubMed]
  5. Proshkina, E.; Plyusnin, S.; Babak, T.; Lashmanova, E.; Maganova, F.; Koval, L.; Platonova, E.; Shaposhnikov, M.; Moskalev, A. Terpenoids as Potential Geroprotectors. Antioxidants 2020, 9, 529. [Google Scholar] [CrossRef] [PubMed]
  6. Li, J.C.; Li, S.Y.; Tang, J.X.; Liu, D.; Feng, X.Y.; Rao, K.R.; Zhao, X.D.; Li, H.M.; Li, R.T. Triterpenoids, steroids and other constituents from Euphorbia kansui and their anti-inflammatory and anti-tumor properties. Phytochemistry 2022, 204, 113449. [Google Scholar] [CrossRef]
  7. Sharma, H.; Kumar, P.; Deshmukh, R.R.; Bishayee, A.; Kumar, S. Pentacyclic triterpenes: New tools to fight metabolic syndrome. Phytomed. Int. J. Phytother. Phytopharm. 2018, 50, 166–177. [Google Scholar] [CrossRef]
  8. Gutiérrez-Rebolledo, G.A.; Siordia-Reyes, A.G.; Meckes-Fischer, M.; Jiménez-Arellanes, A. Hepatoprotective properties of oleanolic and ursolic acids in antitubercular drug-induced liver damage. Asian Pac. J. Trop. Med. 2016, 9, 644–651. [Google Scholar] [CrossRef] [Green Version]
  9. Sen, A. Prophylactic and therapeutic roles of oleanolic acid and its derivatives in several diseases. World J. Clin. Cases 2020, 8, 1767–1792. [Google Scholar] [CrossRef]
  10. Kim, S.; Lee, H.; Lee, S.; Yoon, Y.; Choi, K.H. Antimicrobial action of oleanolic acid on Listeria monocytogenes, Enterococcus faecium, and Enterococcus faecalis. PLoS ONE 2015, 10, e0118800. [Google Scholar] [CrossRef]
  11. Guo, Y.; Han, B.; Luo, K.; Ren, Z.; Cai, L.; Sun, L. NOX2-ROS-HIF-1α signaling is critical for the inhibitory effect of oleanolic acid on rectal cancer cell proliferation. Biomed. Pharmacother. = Biomed. Pharmacother. 2017, 85, 733–739. [Google Scholar] [CrossRef] [PubMed]
  12. Oprean, C.; Mioc, M.; Csányi, E.; Ambrus, R.; Bojin, F.; Tatu, C.; Cristea, M.; Ivan, A.; Danciu, C.; Dehelean, C.; et al. Improvement of ursolic and oleanolic acids’ antitumor activity by complexation with hydrophilic cyclodextrins. Biomed. Pharmacother. = Biomed. Pharmacother. 2016, 83, 1095–1104. [Google Scholar] [CrossRef]
  13. Pattnaik, B.; Lakshma Nayak, V.; Ramakrishna, S.; Venkata Mallavadhani, U. Synthesis of ring-C modified oleanolic acid derivatives and their cytotoxic evaluation. Bioorg. Chem. 2016, 68, 152–158. [Google Scholar] [CrossRef] [PubMed]
  14. Chouaïb, K.; Hichri, F.; Nguir, A.; Daami-Remadi, M.; Elie, N.; Touboul, D.; Ben Jannet, H.; Hamza, M.A. Semi-synthesis of new antimicrobial esters from the natural oleanolic and maslinic acids. Food Chem. 2015, 183, 8–17. [Google Scholar] [CrossRef]
  15. Martín, R.; Cordova, C.; San Román, J.A.; Gutierrez, B.; Cachofeiro, V.; Nieto, M.L. Oleanolic acid modulates the immune-inflammatory response in mice with experimental autoimmune myocarditis and protects from cardiac injury. Therapeutic implications for the human disease. J. Mol. Cell. Cardiol. 2014, 72, 250–262. [Google Scholar] [CrossRef] [PubMed]
  16. Castellano, J.M.; Ramos-Romero, S.; Perona, J.S. Oleanolic Acid: Extraction, Characterization and Biological Activity. Nutrients 2022, 623. [Google Scholar] [CrossRef] [PubMed]
  17. Wang, J.; Zhao, H.; Qiao, W.; Cheng, J.; Han, Y.; Yang, X. Nanomedicine-Cum-Carrier by Co-Assembly of Natural Small Products for Synergistic Enhanced Antitumor with Tissues Protective Actions. ACS Appl. Mater. Interfaces 2020, 12, 42537–42550. [Google Scholar] [CrossRef] [PubMed]
  18. Milan, A.; Mioc, A.; Prodea, A.; Mioc, M.; Buzatu, R.; Ghiulai, R.; Racoviceanu, R.; Caruntu, F.; Şoica, C. The Optimized Delivery of Triterpenes by Liposomal Nanoformulations: Overcoming the Challenges. Int. J. Mol. Sci. 2022, 23, 1140. [Google Scholar] [CrossRef]
  19. Valdés, K.; Morales, J.; Rodríguez, L.; Günther, G. Potential use of nanocarriers with pentacyclic triterpenes in cancer treatments. Nanomedicine 2016, 11, 3139–3156. [Google Scholar] [CrossRef]
  20. Sporn, M.B.; Liby, K.T.; Yore, M.M.; Fu, L.; Lopchuk, J.M.; Gribble, G.W. New synthetic triterpenoids: Potent agents for prevention and treatment of tissue injury caused by inflammatory and oxidative stress. J. Nat. Prod. 2011, 74, 537–545. [Google Scholar] [CrossRef]
  21. Khwaza, V.; Oyedeji, O.O.; Aderibigbe, B.A. Antiviral Activities of Oleanolic Acid and Its Analogues. Molecules 2018, 2300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Xiao, S.; Tian, Z.; Wang, Y.; Si, L.; Zhang, L.; Zhou, D. Recent progress in the antiviral activity and mechanism study of pentacyclic triterpenoids and their derivatives. Med. Res. Rev. 2018, 38, 951–976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Zhou, C.; Zhang, Y.; Sheng, Y.; Zhao, D.; Lv, S.; Hu, Y.; Tao, J. Herbaceous peony (Paeonia lactiflora Pall.) as an alternative source of oleanolic and ursolic acids. Int. J. Mol. Sci. 2011, 12, 655–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zhou, C.; Sheng, Y.; Zhao, D.; Wang, Z.; Tao, J. Variation of oleanolic and ursolic acid in the flesh of persimmon fruit among different cultivars. Molecules 2010, 15, 6580–6587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Jäger, S.; Trojan, H.; Kopp, T.; Laszczyk, M.N.; Scheffler, A. Pentacyclic triterpene distribution in various plants—Rich sources for a new group of multi-potent plant extracts. Molecules 2009, 14, 2016–2031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Onoja, E.; Ndukwe, I.G. Isolation of oleanolic acid from chloroform extract ofBorreria stachydea[(DC) Hutch. and Dalziel]. J. Nat. Prod. Plant Resour. 2013, 3, 57–60. [Google Scholar]
  27. Odun-Ayo, F.; Chetty, K.; Reddy, L. Determination of the ursolic and oleanolic acids content with the antioxidant capacity in apple peel extract of various cultivars. Braz. J. Biol. = Rev. Brasleira De Biol. 2022, 82, e258442. [Google Scholar] [CrossRef]
  28. Wojdyło, A.; Nowicka, P.; Turkiewicz, I.P.; Tkacz, K.; Hernandez, F. Comparison of bioactive compounds and health promoting properties of fruits and leaves of apple, pear and quince. Sci. Rep. 2021, 11, 20253. [Google Scholar] [CrossRef]
  29. Fan, J.-P.; Liao, D.-D.; Zhang, X.-H. Ultrasonic assisted extraction of ursolic acid from apple pomace: A novel and facile technique. Sep. Sci. Technol. 2016, 51, 1344–1350. [Google Scholar] [CrossRef]
  30. Terigar, B.G.; Balasubramanian, S.; Boldor, D.; Xu, Z.; Lima, M.; Sabliov, C.M. Continuous microwave-assisted isoflavone extraction system: Design and performance evaluation. Bioresour. Technol. 2010, 101, 2466–2471. [Google Scholar] [CrossRef]
  31. Wang, C.; Duan, Z.; Fan, L.; Li, J. Supercritical CO₂ Fluid Extraction of Elaeagnus mollis Diels Seed Oil and Its Antioxidant Ability. Molecules 2019, 24, 911. [Google Scholar] [CrossRef] [PubMed]
  32. Vilkickyte, G.; Raudone, L. Optimization, Validation and Application of HPLC-PDA Methods for Quantification of Triterpenoids in Vaccinium vitis-idaea L. Molecules 2021, 26, 1645. [Google Scholar] [CrossRef]
  33. Naumoska, K.; Vovk, I. Analysis of triterpenoids and phytosterols in vegetables by thin-layer chromatography coupled to tandem mass spectrometry. J. Chromatogr. A 2015, 1381, 229–238. [Google Scholar] [CrossRef] [PubMed]
  34. Rastogi, S.; Pandey, M.M.; Kumar Singh Rawat, A. Medicinal plants of the genus Betula--traditional uses and a phytochemical-pharmacological review. J. Ethnopharmacol. 2015, 159, 62–83. [Google Scholar] [CrossRef]
  35. Zhou, C.; Chen, K.; Sun, C.; Chen, Q.; Zhang, W.; Li, X. Determination of oleanolic acid, ursolic acid and amygdalin in the flower of Eriobotrya japonica Lindl. by HPLC. Biomed. Chromatogr. BMC 2007, 21, 755–761. [Google Scholar] [CrossRef] [PubMed]
  36. Sánchez, M.; Theoduloz, C.; Schmeda-Hirschmann, G.; Razmilic, I.; Yáñez, T.; Rodríguez, J.A. Gastroprotective and ulcer-healing activity of oleanolic acid derivatives: In vitro-in vivo relationships. Life Sci. 2006, 79, 1349–1356. [Google Scholar] [CrossRef]
  37. Pang, Z.; Zhi-yan, Z.; Wang, W.; Ma, Y.; Feng-ju, N.; Zhang, X.; Han, C. The advances in research on the pharmacological effects of Fructus Ligustri Lucidi. BioMed Res. Int. 2015, 2015, 281873. [Google Scholar] [CrossRef] [Green Version]
  38. Xia, E.Q.; Yu, Y.Y.; Xu, X.R.; Deng, G.F.; Guo, Y.J.; Li, H.B. Ultrasound-assisted extraction of oleanolic acid and ursolic acid from Ligustrum lucidum Ait. Ultrason. Sonochem. 2012, 19, 772–776. [Google Scholar] [CrossRef]
  39. Banik, R.M.; Pandey, D.K. Optimizing conditions for oleanolic acid extraction from Lantana camara roots using response surface methodology. Ind. Crops Prod. 2008, 27, 241–248. [Google Scholar] [CrossRef]
  40. Petrisor, G.; Motelica, L.; Craciun, L.N.; Oprea, O.C.; Ficai, D.; Ficai, A. Melissa officinalis: Composition, Pharmacological Effects and Derived Release Systems-A Review. Int. J. Mol. Sci. 2022, 23, 3591. [Google Scholar] [CrossRef] [PubMed]
  41. Silva, A.M.; Alvarado, H.L.; Abrego, G.; Martins-Gomes, C.; Garduño-Ramirez, M.L.; García, M.L.; Calpena, A.C.; Souto, E.B. In Vitro Cytotoxicity of Oleanolic/Ursolic Acids-Loaded in PLGA Nanoparticles in Different Cell Lines. Pharmaceutics 2019, 11, 362. [Google Scholar] [CrossRef] [PubMed]
  42. Li, X.; Song, Y.; Zhang, P.; Zhu, H.; Chen, L.; Xiao, Y.; Xing, Y. Oleanolic acid inhibits cell survival and proliferation of prostate cancer cells in vitro and in vivo through the PI3K/Akt pathway. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2016, 37, 7599–7613. [Google Scholar] [CrossRef] [PubMed]
  43. Fu, Q.; Zhang, L.; Cheng, N.; Jia, M.; Zhang, Y. Extraction optimization of oleanolic and ursolic acids from pomegranate (Punica granatum L.) flowers. Food Bioprod. Process. 2014, 92, 321–327. [Google Scholar] [CrossRef]
  44. Jesus, J.A.; Lago, J.H.; Laurenti, M.D.; Yamamoto, E.S.; Passero, L.F. Antimicrobial activity of oleanolic and ursolic acids: An update. Evid.-Based Complement. Altern. Med. Ecam 2015, 2015, 620472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Cláudio, A.F.M.; Cognigni, A.; de Faria, E.L.P.; Silvestre, A.J.D.; Zirbs, R.; Freire, M.G.; Bica, K. Valorization of olive tree leaves: Extraction of oleanolic acid using aqueous solutions of surface-active ionic liquids. Sep. Purif. Technol. 2018, 204, 30–37. [Google Scholar] [CrossRef]
  46. Kowalski, R. Studies of selected plant raw materials as alternative sources of triterpenes of oleanolic and ursolic acid types. J. Agric. Food Chem. 2007, 55, 656–662. [Google Scholar] [CrossRef]
  47. Szczuka, D.; Nowak, A.; Zakłos-Szyda, M.; Kochan, E.; Szymańska, G.; Motyl, I.; Blasiak, J. American Ginseng (Panax quinquefolium L.) as a Source of Bioactive Phytochemicals with Pro-Health Properties. Nutrients 2019, 11, 1041. [Google Scholar] [CrossRef] [Green Version]
  48. Ali, H.; Houghton, P.J.; Soumyanath, A. alpha-Amylase inhibitory activity of some Malaysian plants used to treat diabetes; with particular reference to Phyllanthus amarus. J. Ethnopharmacol. 2006, 107, 449–455. [Google Scholar] [CrossRef]
  49. Yan, M.; Zhu, Y.; Zhang, H.J.; Jiao, W.H.; Han, B.N.; Liu, Z.X.; Qiu, F.; Chen, W.S.; Lin, H.W. Anti-inflammatory secondary metabolites from the leaves of Rosa laevigata. Bioorg. Med. Chem. 2013, 21, 3290–3297. [Google Scholar] [CrossRef]
  50. Kong, L.; Li, S.; Liao, Q.; Zhang, Y.; Sun, R.; Zhu, X.; Zhang, Q.; Wang, J.; Wu, X.; Fang, X.; et al. Oleanolic acid and ursolic acid: Novel hepatitis C virus antivirals that inhibit NS5B activity. Antivir. Res. 2013, 98, 44–53. [Google Scholar] [CrossRef]
  51. Gleńsk, M.; Czapińska, E.; Woźniak, M.; Ceremuga, I.; Włodarczyk, M.; Terlecki, G.; Ziółkowski, P.; Seweryn, E. Triterpenoid Acids as Important Antiproliferative Constituents of European Elderberry Fruits. Nutr. Cancer 2017, 69, 643–651. [Google Scholar] [CrossRef] [PubMed]
  52. Razboršek, M.I.; Vončina, D.B.; Doleček, V.; Vončina, E. Determination of Oleanolic, Betulinic and Ursolic Acid in Lamiaceae and Mass Spectral Fragmentation of Their Trimethylsilylated Derivatives. Chromatographia 2008, 67, 433–440. [Google Scholar] [CrossRef]
  53. Ratajac, R.; Stojanovic, D.; Petrović, J.; Milanov, D.; Vasić, R.; Stojanov, I.M.; Lako, B. Antibacterial activity of the essential oil of Mountain Savory (Satureja montana) against Arcanobacterium pyogenes. Planta Med. 2011, 77, PN5. [Google Scholar] [CrossRef]
  54. Chen, X.Q.; Li, Y.; He, J.; Cheng, X.; Wang, K.; Li, M.M.; Pan, Z.H.; Peng, L.Y.; Zhao, Q.S. Triterpenoids and diterpenoids from Viburnum chingii. Chem. Pharm. Bull. 2011, 59, 496–498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Niampoka, C.; Suttisri, R.; Bavovada, R.; Takayama, H.; Aimi, N. Potentially cytotoxic triterpenoids from the root bark of Siphonodon celastrineus Griff. Arch. Pharmacal Res. 2005, 28, 546–549. [Google Scholar] [CrossRef] [PubMed]
  56. Kowalski, R. Antimicrobial activity of essential oils and extracts of rosinweed (Silphium trifoliatum and Silphium integrifolium) plants used by the American Indians. Flavour Fragr. J. 2008, 23, 426–433. [Google Scholar] [CrossRef]
  57. Horiuchi, K.; Shiota, S.; Hatano, T.; Yoshida, T.; Kuroda, T.; Tsuchiya, T. Antimicrobial activity of oleanolic acid from Salvia officinalis and related compounds on vancomycin-resistant enterococci (VRE). Biol. Pharm. Bull. 2007, 30, 1147–1149. [Google Scholar] [CrossRef] [Green Version]
  58. Ghorbani, A.; Esmaeilizadeh, M. Pharmacological properties of Salvia officinalis and its components. J. Tradit. Complement. Med. 2017, 7, 433–440. [Google Scholar] [CrossRef]
  59. Shimada, A.; Ueno, H.; Inagaki, M. Glutaminase inhibitory activities of pentacyclic triterpenes isolated from Thymus vulgaris L. Nat. Prod. Res. 2022, 36, 2864–2868. [Google Scholar] [CrossRef]
  60. Rali, S.; Oyedeji, O.O.; Aremu, O.O.; Oyedeji, A.O.; Nkeh-Chungag, B.N. Semisynthesis of Derivatives of Oleanolic Acid from Syzygium aromaticum and Their Antinociceptive and Anti-Inflammatory Properties. Mediat. Inflamm. 2016, 2016, 8401843. [Google Scholar] [CrossRef] [Green Version]
  61. Jäger, S.; Winkler, K.; Pfüller, U.; Scheffler, A. Solubility studies of oleanolic acid and betulinic acid in aqueous solutions and plant extracts of Viscum album L. Planta Med. 2007, 73, 157–162. [Google Scholar] [CrossRef]
  62. Chen, J.; Shao, J.; Zhao, C.; Shen, J.; Dong, Z.; Liu, W.; Zhao, M.; Fan, J. Chemical constituents from Viburnum fordiae Hance and their anti-inflammatory and antioxidant activities. Arch. Pharmacal Res. 2018, 41, 625–632. [Google Scholar] [CrossRef]
  63. Yunoki, K.; Sasaki, G.; Tokuji, Y.; Kinoshita, M.; Naito, A.; Aida, K.; Ohnishi, M. Effect of dietary wine pomace extract and oleanolic acid on plasma lipids in rats fed high-fat diet and its DNA microarray analysis. J. Agric. Food Chem. 2008, 56, 12052–12058. [Google Scholar] [CrossRef]
  64. Gamede, M.; Mabuza, L.; Ngubane, P.; Khathi, A. Plant-Derived Oleanolic Acid (OA) Ameliorates Risk Factors of Cardiovascular Diseases in a Diet-Induced Pre-Diabetic Rat Model: Effects on Selected Cardiovascular Risk Factors. Molecules 2019, 24, 340. [Google Scholar] [CrossRef] [Green Version]
  65. Yuan, L.; Zhang, L.; Yao, N.; Wu, L.; Liu, J.; Liu, F.; Zhang, H.; Hu, X.; Xiong, Y.; Xia, C. Upregulation of UGT1A1 expression by ursolic acid and oleanolic acid via the inhibition of the PKC/NF-κB signaling pathway. Phytomed. Int. J. Phytother. Phytopharm. 2021, 92, 153726. [Google Scholar] [CrossRef]
  66. Zhang, X.; Wang, H.; Xu, Y.; Luan, M.; Zhao, F.; Meng, Q. Advances on the Anti-Inflammatory Activity of Oleanolic Acid and Derivatives. Mini Rev. Med. Chem. 2021, 21, 2020–2038. [Google Scholar] [CrossRef]
  67. Bao, J.; Yan, W.; Xu, K.; Chen, M.; Chen, Z.; Ran, J.; Xiong, Y.; Wu, L. Oleanolic Acid Decreases IL-1β-Induced Activation of Fibroblast-Like Synoviocytes via the SIRT3-NF-κB Axis in Osteoarthritis. Oxidative Med. Cell. Longev. 2020, 2020, 7517219. [Google Scholar] [CrossRef]
  68. Lu, X.; Li, Y.; Yang, W.; Tao, M.; Dai, Y.; Xu, J.; Xu, Q. Inhibition of NF-κB is required for oleanolic acid to downregulate PD-L1 by promoting DNA demethylation in gastric cancer cells. J. Biochem. Mol. Toxicol. 2021, 35, e22621. [Google Scholar] [CrossRef]
  69. Li, W.; Zeng, H.; Xu, M.; Huang, C.; Tao, L.; Li, J.; Zhang, T.; Chen, H.; Xia, J.; Li, C.; et al. Oleanolic Acid Improves Obesity-Related Inflammation and Insulin Resistance by Regulating Macrophages Activation. Front. Pharmacol. 2021, 12, 697483. [Google Scholar] [CrossRef]
  70. An, Q.; Hu, Q.; Wang, B.; Cui, W.; Wu, F.; Ding, Y. Oleanolic acid alleviates diabetic rat carotid artery injury through the inhibition of NLRP3 inflammasome signaling pathways. Mol. Med. Rep. 2017, 16, 8413–8419. [Google Scholar] [CrossRef] [Green Version]
  71. Sapkota, A.; Choi, J.W. Oleanolic Acid Provides Neuroprotection against Ischemic Stroke through the Inhibition of Microglial Activation and NLRP3 Inflammasome Activation. Biomol. Ther. 2022, 30, 55–63. [Google Scholar] [CrossRef] [PubMed]
  72. Feng, J.; Zhang, P.; Chen, X.; He, G. PI3K and ERK/Nrf2 pathways are involved in oleanolic acid-induced heme oxygenase-1 expression in rat vascular smooth muscle cells. J. Cell. Biochem. 2011, 112, 1524–1531. [Google Scholar] [CrossRef] [PubMed]
  73. Wang, X.; Bai, H.; Zhang, X.; Liu, J.; Cao, P.; Liao, N.; Zhang, W.; Wang, Z.; Hai, C. Inhibitory effect of oleanolic acid on hepatocellular carcinoma via ERK-p53-mediated cell cycle arrest and mitochondrial-dependent apoptosis. Carcinogenesis 2013, 34, 1323–1330. [Google Scholar] [CrossRef]
  74. Kim, G.J.; Jo, H.J.; Lee, K.J.; Choi, J.W.; An, J.H. Oleanolic acid induces p53-dependent apoptosis via the ERK/JNK/AKT pathway in cancer cell lines in prostatic cancer xenografts in mice. Oncotarget 2018, 9, 26370–26386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Lu, Y.; Zhu, M.; Chen, W.; Yin, L.; Zhu, J.; Chen, N.; Chen, W. Oleanolic acid induces apoptosis of MKN28 cells via AKT and JNK signaling pathways. Pharm. Biol. 2014, 52, 789–795. [Google Scholar] [CrossRef] [PubMed]
  76. Kim, S.H.; Hong, J.H.; Lee, Y.C. Oleanolic acid suppresses ovalbumin-induced airway inflammation and Th2-mediated allergic asthma by modulating the transcription factors T-bet, GATA-3, RORγt and Foxp3 in asthmatic mice. Int. Immunopharmacol. 2014, 18, 311–324. [Google Scholar] [CrossRef]
  77. Phull, A.R.; Eo, S.H.; Kim, S.J. Oleanolic acid (OA) regulates inflammation and cellular dedifferentiation of chondrocytes via MAPK signaling pathways. Cell. Mol. Biol. (Noisy-Le-Grand Fr.) 2017, 63, 12–17. [Google Scholar] [CrossRef]
  78. Wu, Y.; Gao, L.J.; Fan, Y.S.; Chen, Y.; Li, Q. Network Pharmacology-Based Analysis on the Action Mechanism of Oleanolic Acid to Alleviate Osteoporosis. ACS Omega 2021, 6, 28410–28420. [Google Scholar] [CrossRef]
  79. Zhao, X.; Liu, M.; Li, D. Oleanolic acid suppresses the proliferation of lung carcinoma cells by miR-122/Cyclin G1/MEF2D axis. Mol. Cell. Biochem. 2015, 400, 1–7. [Google Scholar] [CrossRef]
  80. Chen, F.F.; Wang, J.T.; Zhang, L.X.; Xing, S.F.; Wang, Y.X.; Wang, K.; Deng, S.L.; Zhang, J.Q.; Tang, L.; Wu, H.S. Oleanolic acid derivative DKS26 exerts antidiabetic and hepatoprotective effects in diabetic mice and promotes glucagon-like peptide-1 secretion and expression in intestinal cells. Br. J. Pharmacol. 2017, 174, 2912–2928. [Google Scholar] [CrossRef] [Green Version]
  81. Yu, Y.; Ma, T.; Lv, L.; Jia, L.; Ruan, H.; Chen, H.; Zhang, J.; Gao, L. Oleanolic acid targets the regulation of PI3K/AKT/mTOR pathway and activates autophagy in chondrocytes to improve osteoarthritis in rats. J. Funct. Foods 2022, 94, 105144. [Google Scholar] [CrossRef]
  82. Zhang, Z.; Jiang, M.; Xie, X.; Yang, H.; Wang, X.; Xiao, L.; Wang, N. Oleanolic acid ameliorates high glucose-induced endothelial dysfunction via PPARδ activation. Sci. Rep. 2017, 7, 40237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Li, L.; Lin, J.; Sun, G.; Wei, L.; Shen, A.; Zhang, M.; Peng, J. Oleanolic acid inhibits colorectal cancer angiogenesis in vivo and in vitro via suppression of STAT3 and Hedgehog pathways. Mol. Med. Rep. 2016, 13, 5276–5282. [Google Scholar] [CrossRef]
  84. Niu, G.; Sun, L.; Pei, Y.; Wang, D. Oleanolic Acid Inhibits Colorectal Cancer Angiogenesis by Blocking the VEGFR2 Signaling Pathway. Anti-Cancer Agents Med. Chem. 2018, 18, 583–590. [Google Scholar] [CrossRef] [PubMed]
  85. Tang, Z.Y.; Li, Y.; Tang, Y.T.; Ma, X.D.; Tang, Z.Y. Anticancer activity of oleanolic acid and its derivatives: Recent advances in evidence, target profiling and mechanisms of action. Biomed. Pharmacother. = Biomed. Pharmacother. 2022, 145, 112397. [Google Scholar] [CrossRef]
  86. Zhou, Z.; Dong, Y.; Li, N.; Niu, M.; Wang, S.; Zhou, Y.; Sun, Z.; Chu, P.; Tang, Z. An oleanolic acid derivative, K73-03, inhibits pancreatic cancer cells proliferation in vitro and in vivo via blocking EGFR/Akt pathway. Cell Biol. Int. 2022, 46, 1801–1813. [Google Scholar] [CrossRef]
  87. Castellano, J.M.; Guinda, A.; Delgado, T.; Rada, M.; Cayuela, J.A. Biochemical basis of the antidiabetic activity of oleanolic acid and related pentacyclic triterpenes. Diabetes 2013, 62, 1791–1799. [Google Scholar] [CrossRef] [Green Version]
  88. Dinkova-Kostova, A.T.; Liby, K.T.; Stephenson, K.K.; Holtzclaw, W.D.; Gao, X.; Suh, N.; Williams, C.; Risingsong, R.; Honda, T.; Gribble, G.W.; et al. Extremely potent triterpenoid inducers of the phase 2 response: Correlations of protection against oxidant and inflammatory stress. Proc. Natl. Acad. Sci. USA 2005, 102, 4584–4589. [Google Scholar] [CrossRef] [Green Version]
  89. Wang, X.; Liu, R.; Zhang, W.; Zhang, X.; Liao, N.; Wang, Z.; Li, W.; Qin, X.; Hai, C. Oleanolic acid improves hepatic insulin resistance via antioxidant, hypolipidemic and anti-inflammatory effects. Mol. Cell. Endocrinol. 2013, 376, 70–80. [Google Scholar] [CrossRef]
  90. Fernández-Aparicio, Á.; Correa-Rodríguez, M.; Castellano, J.M.; Schmidt-RioValle, J.; Perona, J.S.; González-Jiménez, E. Potential Molecular Targets of Oleanolic Acid in Insulin Resistance and Underlying Oxidative Stress: A Systematic Review. Antioxidants 2022, 11, 1517. [Google Scholar] [CrossRef]
  91. Lee, W.; Yang, E.J.; Ku, S.K.; Song, K.S.; Bae, J.S. Anti-inflammatory effects of oleanolic acid on LPS-induced inflammation in vitro and in vivo. Inflammation 2013, 36, 94–102. [Google Scholar] [CrossRef] [PubMed]
  92. Dong, N.; Xue, C.; Zhang, L.; Zhang, T.; Wang, C.; Bi, C.; Shan, A. Oleanolic acid enhances tight junctions and ameliorates inflammation in Salmonella typhimurium-induced diarrhea in mice via the TLR4/NF-κB and MAPK pathway. Food Funct. 2020, 11, 1122–1132. [Google Scholar] [CrossRef] [PubMed]
  93. Jehangir, A.; Shahzad, M.; Shahid, K.; Waheed, A.; Ayub, F. Zinc and iron complexes of oleanolic acid, (OA) attenuate allergic airway inflammation in rats. Inflammopharmacology 2019, 27, 1179–1192. [Google Scholar] [CrossRef] [PubMed]
  94. Huang, T.H.; Yang, Q.; Harada, M.; Li, G.Q.; Yamahara, J.; Roufogalis, B.D.; Li, Y. Pomegranate flower extract diminishes cardiac fibrosis in Zucker diabetic fatty rats: Modulation of cardiac endothelin-1 and nuclear factor-kappaB pathways. J. Cardiovasc. Pharmacol. 2005, 46, 856–862. [Google Scholar] [CrossRef]
  95. Takada, K.; Nakane, T.; Masuda, K.; Ishii, H. Ursolic acid and oleanolic acid, members of pentacyclic triterpenoid acids, suppress TNF-α-induced E-selectin expression by cultured umbilical vein endothelial cells. Phytomed. Int. J. Phytother. Phytopharm. 2010, 17, 1114–1119. [Google Scholar] [CrossRef]
  96. Hassan Mir, R.; Godavari, G.; Siddiqui, N.A.; Ahmad, B.; Mothana, R.A.; Ullah, R.; Almarfadi, O.M.; Jachak, S.M.; Masoodi, M.H. Design, Synthesis, Molecular Modelling, and Biological Evaluation of Oleanolic Acid-Arylidene Derivatives as Potential Anti-Inflammatory Agents. Drug Des. Dev. Ther. 2021, 15, 385–397. [Google Scholar] [CrossRef]
  97. Djeziri, F.Z.; Belarbi, M.; Murtaza, B.; Hichami, A.; Benammar, C.; Khan, N.A. Oleanolic acid improves diet-induced obesity by modulating fat preference and inflammation in mice. Biochimie 2018, 152, 110–120. [Google Scholar] [CrossRef]
  98. Castellano, J.M.; Garcia-Rodriguez, S.; Espinosa, J.M.; Millan-Linares, M.C.; Rada, M.; Perona, J.S. Oleanolic Acid Exerts a Neuroprotective Effect Against Microglial Cell Activation by Modulating Cytokine Release and Antioxidant Defense Systems. Biomolecules 2019, 9, 683. [Google Scholar] [CrossRef] [Green Version]
  99. Gutierrez, B.; Gallardo, I.; Ruiz, L.; Alvarez, Y.; Cachofeiro, V.; Margolles, A.; Hernandez, M.; Nieto, M.L. Oleanolic acid ameliorates intestinal alterations associated with EAE. J. Neuroinflamm. 2020, 17, 363. [Google Scholar] [CrossRef]
  100. Wang, X.; Ye, X.L.; Liu, R.; Chen, H.L.; Bai, H.; Liang, X.; Zhang, X.D.; Wang, Z.; Li, W.L.; Hai, C.X. Antioxidant activities of oleanolic acid in vitro: Possible role of Nrf2 and MAP kinases. Chem.-Biol. Interact. 2010, 184, 328–337. [Google Scholar] [CrossRef]
  101. Peng, F.; Li, G.; Xie, Y.; Yin, H.Y.; Li, X.J.; Yang, Y.D. Compositional characterization of Pyrus ussuriensis Maxim and their antioxidant activities and induction of apoptosis in Bel-7402 cell. J. Food Biochem. 2020, 44, e13222. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, J.; Wang, X.; Liu, R.; Liu, Y.; Zhang, T.; Fu, H.; Hai, C. Oleanolic acid co-administration alleviates ethanol-induced hepatic injury via Nrf-2 and ethanol-metabolizing modulating in rats. Chem.-Biol. Interact. 2014, 221, 88–98. [Google Scholar] [CrossRef] [PubMed]
  103. Iskender, H.; Dokumacioglu, E.; Terim Kapakin, K.A.; Yenice, G.; Mohtare, B.; Bolat, I.; Hayirli, A. Effects of oleanolic acid on inflammation and metabolism in diabetic rats. Biotech. Histochem. Off. Publ. Biol. Stain Comm. 2022, 97, 269–276. [Google Scholar] [CrossRef] [PubMed]
  104. Fricker, M.; Tolkovsky, A.M.; Borutaite, V.; Coleman, M.; Brown, G.C. Neuronal Cell Death. Physiol. Rev. 2018, 98, 813–880. [Google Scholar] [CrossRef]
  105. Zhang, T.; Hou, D.; He, J.; Zeng, X.; Liu, R.; Liu, L.; Li, T.; Xiao, Y.; Ma, R.; Huang, H.; et al. Oxidative-Damaged Mitochondria Activate GABARAPL1-Induced NLRP3 Inflammasomes in an Autophagic-Exosome Manner after Acute Myocardial Ischemia. Oxidative Med. Cell. Longev. 2022, 2022, 7958542. [Google Scholar] [CrossRef] [PubMed]
  106. Lawrence, G.; Holley, C.L.; Schroder, K. Parkinson’s disease: Connecting mitochondria to inflammasomes. Trends Immunol. 2022, 43, 877–885. [Google Scholar] [CrossRef] [PubMed]
  107. He, Y.; Hara, H.; Núñez, G. Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends Biochem. Sci. 2016, 41, 1012–1021. [Google Scholar] [CrossRef] [Green Version]
  108. Shanmugam, M.K.; Dai, X.; Kumar, A.P.; Tan, B.K.; Sethi, G.; Bishayee, A. Oleanolic acid and its synthetic derivatives for the prevention and therapy of cancer: Preclinical and clinical evidence. Cancer Lett. 2014, 346, 206–216. [Google Scholar] [CrossRef] [Green Version]
  109. Potočnjak, I.; Šimić, L.; Vukelić, I.; Batičić, L.; Domitrović, R. Oleanolic acid induces HCT116 colon cancer cell death through the p38/FOXO3a/Sirt6 pathway. Chem.-Biol. Interact. 2022, 363, 110010. [Google Scholar] [CrossRef]
  110. Liu, J.; Zheng, L.; Ma, L.; Wang, B.; Zhao, Y.; Wu, N.; Liu, G.; Lin, X. Oleanolic acid inhibits proliferation and invasiveness of Kras-transformed cells via autophagy. J. Nutr. Biochem. 2014, 25, 1154–1160. [Google Scholar] [CrossRef]
  111. Fujiwara, Y.; Komohara, Y.; Kudo, R.; Tsurushima, K.; Ohnishi, K.; Ikeda, T.; Takeya, M. Oleanolic acid inhibits macrophage differentiation into the M2 phenotype and glioblastoma cell proliferation by suppressing the activation of STAT3. Oncol. Rep. 2011, 26, 1533–1537. [Google Scholar] [CrossRef]
  112. Birts, C.N.; Banerjee, A.; Darley, M.; Dunlop, C.R.; Nelson, S.; Nijjar, S.K.; Parker, R.; West, J.; Tavassoli, A.; Rose-Zerilli, M.J.J.; et al. p53 is regulated by aerobic glycolysis in cancer cells by the CtBP family of NADH-dependent transcriptional regulators. Sci. Signal. 2020, 13, eaau9529. [Google Scholar] [CrossRef] [PubMed]
  113. Li, L.; Wei, L.; Shen, A.; Chu, J.; Lin, J.; Peng, J. Oleanolic acid modulates multiple intracellular targets to inhibit colorectal cancer growth. Int. J. Oncol. 2015, 47, 2247–2254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Li, Y.; Xu, Q.; Yang, W.; Wu, T.; Lu, X. Oleanolic acid reduces aerobic glycolysis-associated proliferation by inhibiting yes-associated protein in gastric cancer cells. Gene 2019, 712, 143956. [Google Scholar] [CrossRef] [PubMed]
  115. Kuo, R.Y.; Qian, K.; Morris-Natschke, S.L.; Lee, K.H. Plant-derived triterpenoids and analogues as antitumor and anti-HIV agents. Nat. Prod. Rep. 2009, 26, 1321–1344. [Google Scholar] [CrossRef] [Green Version]
  116. Yamai, H.; Sawada, N.; Yoshida, T.; Seike, J.; Takizawa, H.; Kenzaki, K.; Miyoshi, T.; Kondo, K.; Bando, Y.; Ohnishi, Y.; et al. Triterpenes augment the inhibitory effects of anticancer drugs on growth of human esophageal carcinoma cells in vitro and suppress experimental metastasis in vivo. Int. J. Cancer 2009, 125, 952–960. [Google Scholar] [CrossRef] [PubMed]
  117. Juan, M.E.; Planas, J.M.; Ruiz-Gutierrez, V.; Daniel, H.; Wenzel, U. Antiproliferative and apoptosis-inducing effects of maslinic and oleanolic acids, two pentacyclic triterpenes from olives, on HT-29 colon cancer cells. Br. J. Nutr. 2008, 100, 36–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Kuttan, G.; Pratheeshkumar, P.; Manu, K.A.; Kuttan, R. Inhibition of tumor progression by naturally occurring terpenoids. Pharm. Biol. 2011, 49, 995–1007. [Google Scholar] [CrossRef]
  119. Shyu, M.H.; Kao, T.C.; Yen, G.C. Oleanolic acid and ursolic acid induce apoptosis in HuH7 human hepatocellular carcinoma cells through a mitochondrial-dependent pathway and downregulation of XIAP. J. Agric. Food Chem. 2010, 58, 6110–6118. [Google Scholar] [CrossRef]
  120. Wang, Y.Y.; Yang, Y.X.; Zhao, R.; Pan, S.T.; Zhe, H.; He, Z.X.; Duan, W.; Zhang, X.; Yang, T.; Qiu, J.X.; et al. Bardoxolone methyl induces apoptosis and autophagy and inhibits epithelial-to-mesenchymal transition and stemness in esophageal squamous cancer cells. Drug Des. Dev. Ther. 2015, 9, 993–1026. [Google Scholar] [CrossRef] [Green Version]
  121. Ghosh, N.; Chaki, R.; Mandal, V.; Mandal, S.C. COX-2 as a target for cancer chemotherapy. Pharmacol. Rep. PR 2010, 62, 233–244. [Google Scholar] [CrossRef]
  122. Janakiram, N.B.; Indranie, C.; Malisetty, S.V.; Jagan, P.; Steele, V.E.; Rao, C.V. Chemoprevention of colon carcinogenesis by oleanolic acid and its analog in male F344 rats and modulation of COX-2 and apoptosis in human colon HT-29 cancer cells. Pharm. Res. 2008, 25, 2151–2157. [Google Scholar] [CrossRef]
  123. Shi, Y.; Song, Q.; Hu, D.; Zhuang, X.; Yu, S.; Teng, D. Oleanolic acid induced autophagic cell death in hepatocellular carcinoma cells via PI3K/Akt/mTOR and ROS-dependent pathway. Korean J. Physiol. Pharmacol. Off. J. Korean Physiol. Soc. Korean Soc. Pharmacol. 2016, 20, 237–243. [Google Scholar] [CrossRef] [Green Version]
  124. Song, Y.; Zhang, P.; Sun, Y.; Li, X.; Chen, L.; Xiao, Y.; Xing, Y. AMPK activation-dependent autophagy compromises oleanolic acid-induced cytotoxicity in human bladder cancer cells. Oncotarget 2017, 8, 67942–67954. [Google Scholar] [CrossRef] [Green Version]
  125. Sun, H.; Wang, Z.; Yakisich, J.S. Natural products targeting autophagy via the PI3K/Akt/mTOR pathway as anticancer agents. Anti-Cancer Agents Med. Chem. 2013, 13, 1048–1056. [Google Scholar] [CrossRef]
  126. Wang, X.; Chen, H.L.; Liu, J.Z.; Liao, N.; Yu, W.H.; Zhang, X.D.; Zhang, T.; Li, W.L.; Hai, C.X. Protective effect of oleanolic acid against beta cell dysfunction and mitochondrial apoptosis: Crucial role of ERK-NRF2 signaling pathway. J. Biol. Regul. Homeost. Agents 2013, 27, 55–67. [Google Scholar]
  127. Lúcio, K.A.; Rocha Gda, G.; Monção-Ribeiro, L.C.; Fernandes, J.; Takiya, C.M.; Gattass, C.R. Oleanolic acid initiates apoptosis in non-small cell lung cancer cell lines and reduces metastasis of a B16F10 melanoma model in vivo. PLoS ONE 2011, 6, e28596. [Google Scholar] [CrossRef] [Green Version]
  128. Feng, A.; Yang, S.; Sun, Y.; Zhang, L.; Bo, F.; Li, L. Development and Evaluation of Oleanolic Acid Dosage Forms and Its Derivatives. BioMed Res. Int. 2020, 2020, 1308749. [Google Scholar] [CrossRef]
  129. Liu, J.; Wu, Q.; Lu, Y.F.; Pi, J. New insights into generalized hepatoprotective effects of oleanolic acid: Key roles of metallothionein and Nrf2 induction. Biochem. Pharm. 2008, 76, 922–928. [Google Scholar] [CrossRef]
  130. She, J.; Gu, T.; Pang, X.; Liu, Y.; Tang, L.; Zhou, X. Natural Products Targeting Liver X Receptors or Farnesoid X Receptor. Front. Pharmacol. 2021, 12, 772435. [Google Scholar] [CrossRef]
  131. Reisman, S.A.; Aleksunes, L.M.; Klaassen, C.D. Oleanolic acid activates Nrf2 and protects from acetaminophen hepatotoxicity via Nrf2-dependent and Nrf2-independent processes. Biochem. Pharm. 2009, 77, 1273–1282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Fajemiroye, J.O.; Polepally, P.R.; Chaurasiya, N.D.; Tekwani, B.L.; Zjawiony, J.K.; Costa, E.A. Oleanolic acid acrylate elicits antidepressant-like effect mediated by 5-HT1A receptor. Sci. Rep. 2015, 5, 11582. [Google Scholar] [CrossRef] [PubMed]
  133. Liu, J. Pharmacology of oleanolic acid and ursolic acid. J. Ethnopharmacol. 1995, 49, 57–68. [Google Scholar] [CrossRef] [PubMed]
  134. Liu, J. Oleanolic acid and ursolic acid: Research perspectives. J. Ethnopharmacol. 2005, 100, 92–94. [Google Scholar] [CrossRef]
  135. Yi, L.T.; Li, J.; Liu, B.B.; Luo, L.; Liu, Q.; Geng, D. BDNF-ERK-CREB signalling mediates the role of miR-132 in the regulation of the effects of oleanolic acid in male mice. J. Psychiatry Neurosci. JPN 2014, 39, 348–359. [Google Scholar] [CrossRef] [Green Version]
  136. Yi, L.T.; Li, J.; Liu, Q.; Geng, D.; Zhou, Y.F.; Ke, X.Q.; Chen, H.; Weng, L.J. Antidepressant-like effect of oleanolic acid in mice exposed to the repeated forced swimming test. J. Psychopharmacol. 2013, 27, 459–468. [Google Scholar] [CrossRef]
  137. Dong, S.Q.; Wang, S.S.; Zhu, J.X.; Mu, R.H.; Li, C.F.; Geng, D.; Liu, Q.; Yi, L.T. Oleanolic acid decreases SGK1 in the hippocampus in corticosterone-induced mice. Steroids 2019, 149, 108419. [Google Scholar] [CrossRef]
  138. Xie, B.P.; Shi, L.Y.; Li, J.P.; Zeng, Y.; Liu, W.; Tang, S.Y.; Jia, L.J.; Zhang, J.; Gan, G.X. Oleanolic acid inhibits RANKL-induced osteoclastogenesis via ER alpha/miR-503/RANK signaling pathway in RAW264.7 cells. Biomed. Pharmacother. = Biomed. Pharmacother. 2019, 117, 109045. [Google Scholar] [CrossRef]
  139. Cheon, S.Y.; Jin, B.R.; Kim, H.J.; An, H.J. Oleanolic Acid Ameliorates Benign Prostatic Hyperplasia by Regulating PCNA-Dependent Cell Cycle Progression In Vivo and In Vitro. J. Nat. Prod. 2020, 83, 1183–1189. [Google Scholar] [CrossRef]
  140. Cui, W.; Liu, C.X.; Wang, J.; Zhang, Y.C.; Shen, Q.; Feng, Z.H.; Wu, J.; Li, J.X. An oleanolic acid derivative reduces denervation-induced muscle atrophy via activation of CNTF-mediated JAK2/STAT3 signaling pathway. Eur. J. Pharmacol. 2019, 861, 172612. [Google Scholar] [CrossRef]
  141. Su, Y.; Meng, L.; Sun, J.; Li, W.; Shao, L.; Chen, K.; Zhou, D.; Yang, F.; Yu, F. Design, synthesis of oleanolic acid-saccharide conjugates using click chemistry methodology and study of their anti-influenza activity. Eur. J. Med. Chem. 2019, 182, 111622. [Google Scholar] [CrossRef] [PubMed]
  142. Rodriguez-Rodriguez, R. Oleanolic acid and related triterpenoids from olives on vascular function: Molecular mechanisms and therapeutic perspectives. Curr. Med. Chem. 2015, 22, 1414–1425. [Google Scholar] [CrossRef] [PubMed]
  143. Somova, L.O.; Nadar, A.; Rammanan, P.; Shode, F.O. Cardiovascular, antihyperlipidemic and antioxidant effects of oleanolic and ursolic acids in experimental hypertension. Phytomed. Int. J. Phytother. Phytopharm. 2003, 10, 115–121. [Google Scholar] [CrossRef]
  144. Sun, N.; Li, D.; Chen, X.; Wu, P.; Lu, Y.J.; Hou, N.; Chen, W.H.; Wong, W.L. New Applications of Oleanolic Acid and its Derivatives as Cardioprotective Agents: A Review of their Therapeutic Perspectives. Curr. Pharm. Des. 2019, 25, 3740–3750. [Google Scholar] [CrossRef] [PubMed]
  145. Camer, D.; Yu, Y.; Szabo, A.; Huang, X.F. The molecular mechanisms underpinning the therapeutic properties of oleanolic acid, its isomer and derivatives for type 2 diabetes and associated complications. Mol. Nutr. Food Res. 2014, 58, 1750–1759. [Google Scholar] [CrossRef] [Green Version]
  146. Gao, D.; Li, Q.; Li, Y.; Liu, Z.; Fan, Y.; Liu, Z.; Zhao, H.; Li, J.; Han, Z. Antidiabetic and antioxidant effects of oleanolic acid from Ligustrum lucidum Ait in alloxan-induced diabetic rats. Phytother. Res. PTR 2009, 23, 1257–1262. [Google Scholar] [CrossRef]
  147. Buus, N.H.; Hansson, N.C.; Rodriguez-Rodriguez, R.; Stankevicius, E.; Andersen, M.R.; Simonsen, U. Antiatherogenic effects of oleanolic acid in apolipoprotein E knockout mice. Eur. J. Pharmacol. 2011, 670, 519–526. [Google Scholar] [CrossRef]
  148. Oboh, M.; Govender, L.; Siwela, M.; Mkhwanazi, B.N. Anti-Diabetic Potential of Plant-Based Pentacyclic Triterpene Derivatives: Progress Made to Improve Efficacy and Bioavailability. Molecules 2021, 26, 7243. [Google Scholar] [CrossRef]
  149. NA, J.C.F.; Pirson, L.; Edelberg, H.; Miranda, L.M.; Loira-Pastoriza, C.; Preat, V.; Larondelle, Y.; André, C.M. Pentacyclic Triterpene Bioavailability: An Overview of In Vitro and In Vivo Studies. Molecules 2017, 22, 400. [Google Scholar] [CrossRef] [Green Version]
  150. Pollier, J.; Goossens, A. Oleanolic acid. Phytochemistry 2012, 77, 10–15. [Google Scholar] [CrossRef]
  151. Bishayee, A.; Ahmed, S.; Brankov, N.; Perloff, M. Triterpenoids as potential agents for the chemoprevention and therapy of breast cancer. Front. Biosci. 2011, 16, 980–996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Xu, R.; Fazio, G.C.; Matsuda, S.P. On the origins of triterpenoid skeletal diversity. Phytochemistry 2004, 65, 261–291. [Google Scholar] [CrossRef] [PubMed]
  153. Nkeh-Chungag, B.N.; Oyedeji, O.O.; Oyedeji, A.O.; Ndebia, E.J. Anti-inflammatory and membrane-stabilizing properties of two semisynthetic derivatives of oleanolic acid. Inflammation 2015, 38, 61–69. [Google Scholar] [CrossRef] [PubMed]
  154. Zhao, H.; Holmes, S.S.; Baker, G.A.; Challa, S.; Bose, H.S.; Song, Z. Ionic derivatives of betulinic acid as novel HIV-1 protease inhibitors. J. Enzym. Inhib. Med. Chem. 2012, 27, 715–721. [Google Scholar] [CrossRef] [Green Version]
  155. Zhu, B.; Ren, C.; Du, K.; Zhu, H.; Ai, Y.; Kang, F.; Luo, Y.; Liu, W.; Wang, L.; Xu, Y.; et al. Olean-28,13b-olide 2 plays a role in cisplatin-mediated apoptosis and reverses cisplatin resistance in human lung cancer through multiple signaling pathways. Biochem. Pharm. 2019, 170, 113642. [Google Scholar] [CrossRef]
  156. Gao, L.; Xu, Z.; Wang, Y.; Sun, B.; Song, Z.; Yang, B.; Liu, X.; Lin, Y.; Peng, J.; Han, G.; et al. Anticancer effect of SZC017, a novel derivative of oleanolic acid, on human gastric cancer cells. Oncol. Rep. 2016, 35, 1101–1108. [Google Scholar] [CrossRef] [Green Version]
  157. Cheng, K.G.; Su, C.H.; Yang, L.D.; Liu, J.; Chen, Z.F. Synthesis of oleanolic acid dimers linked at C-28 and evaluation of anti-tumor activity. Eur. J. Med. Chem. 2015, 89, 480–489. [Google Scholar] [CrossRef]
  158. Ren, Y.; Liu, Y.; Yang, Z.; Niu, R.; Gao, K.; Yang, B.; Liao, X.; Zhang, J. Solid inclusion complexes of oleanolic acid with amino-appended β-cyclodextrins (ACDs): Preparation, characterization, water solubility and anticancer activity. Mater. Sci. Eng. C Mater. Biol. Appl. 2016, 69, 68–76. [Google Scholar] [CrossRef]
  159. Chaturvedi, V.K.; Singh, A.; Singh, V.K.; Singh, M.P. Cancer Nanotechnology: A New Revolution for Cancer Diagnosis and Therapy. Curr. Drug Metab. 2019, 20, 416–429. [Google Scholar] [CrossRef]
  160. Zhao, Z.; Ukidve, A.; Kim, J.; Mitragotri, S. Targeting Strategies for Tissue-Specific Drug Delivery. Cell 2020, 181, 151–167. [Google Scholar] [CrossRef]
  161. Gregoriadis, G. Engineering liposomes for drug delivery: Progress and problems. Trends Biotechnol. 1995, 13, 527–537. [Google Scholar] [CrossRef] [PubMed]
  162. Lamichhane, N.; Udayakumar, T.S.; D’Souza, W.D.; Simone, C.B.; Raghavan, S.R.; Polf, J.; Mahmood, J. Liposomes: Clinical Applications and Potential for Image-Guided Drug Delivery. Molecules 2018, 23, 288. [Google Scholar] [CrossRef] [PubMed]
  163. Luo, L.; Bian, Y.; Liu, Y.; Zhang, X.; Wang, M.; Xing, S.; Li, L.; Gao, D. Combined Near Infrared Photothermal Therapy and Chemotherapy Using Gold Nanoshells Coated Liposomes to Enhance Antitumor Effect. Small 2016, 12, 4103–4112. [Google Scholar] [CrossRef] [PubMed]
  164. Gao, D.; Tang, S.; Tong, Q. Oleanolic acid liposomes with polyethylene glycol modification: Promising antitumor drug delivery. Int. J. Nanomed. 2012, 7, 3517–3526. [Google Scholar] [CrossRef] [Green Version]
  165. Fan, R.; Wang, H.; Zhang, L.; Ma, T.; Tian, Y.; Li, H. Nanocrystallized Oleanolic Acid Better Inhibits Proliferation, Migration and Invasion in Intracranial Glioma via Caspase-3 Pathway. J. Cancer 2020, 11, 1949–1958. [Google Scholar] [CrossRef]
  166. Sarfraz, M.; Afzal, A.; Yang, T.; Gai, Y.; Raza, S.M.; Khan, M.W.; Cheng, Y.; Ma, X.; Xiang, G. Development of Dual Drug Loaded Nanosized Liposomal Formulation by A Reengineered Ethanolic Injection Method and Its Pre-Clinical Pharmacokinetic Studies. Pharmaceutics 2018, 10, 151. [Google Scholar] [CrossRef] [Green Version]
  167. Sarfraz, M.; Afzal, A.; Raza, S.M.; Bashir, S.; Madni, A.; Khan, M.W.; Ma, X.; Xiang, G. Liposomal co-delivered oleanolic acid attenuates doxorubicin-induced multi-organ toxicity in hepatocellular carcinoma. Oncotarget 2017, 8, 47136–47153. [Google Scholar] [CrossRef] [Green Version]
  168. Wang, Y.; Luo, Y.; Li, C.; Zhang, X.; Pi, C.; Yu, L.; Wang, S.; Zhong, Z. Optimized formulation of multivesicular liposomes loaded with oleanolic acid enhanced anticancer effect in vitro. Drug Des. Dev. Ther. 2017, 11, 955–968. [Google Scholar] [CrossRef] [Green Version]
  169. Jurj, A.; Braicu, C.; Pop, L.A.; Tomuleasa, C.; Gherman, C.D.; Berindan-Neagoe, I. The new era of nanotechnology, an alternative to change cancer treatment. Drug Des. Dev. Ther. 2017, 11, 2871–2890. [Google Scholar] [CrossRef] [Green Version]
  170. Zhi, K.; Wang, J.; Zhao, H.; Yang, X. Self-assembled small molecule natural product gel for drug delivery: A breakthrough in new application of small molecule natural products. Acta Pharm. Sin. B 2020, 10, 913–927. [Google Scholar] [CrossRef]
  171. Gao, M.; Xu, H.; Bao, X.; Zhang, C.; Guan, X.; Liu, H.; Lv, L.; Deng, S.; Gao, D.; Wang, C.; et al. Oleanolic acid-loaded PLGA-TPGS nanoparticles combined with heparin sodium-loaded PLGA-TPGS nanoparticles for enhancing chemotherapy to liver cancer. Life Sci. 2016, 165, 63–74. [Google Scholar] [CrossRef] [PubMed]
  172. Xia, X.; Liu, H.; Lv, H.; Zhang, J.; Zhou, J.; Zhao, Z. Preparation, characterization, and in vitro/vivo studies of oleanolic acid-loaded lactoferrin nanoparticles. Drug Des. Dev. Ther. 2017, 11, 1417–1427. [Google Scholar] [CrossRef] [PubMed]
  173. Wang, Y.; Zhu, P.; Li, G.; Zhu, S.; Liu, K.; Liu, Y.; He, J.; Lei, J. Amphiphilic carboxylated cellulose-g-poly(l-lactide) copolymer nanoparticles for oleanolic acid delivery. Carbohydr. Polym. 2019, 214, 100–109. [Google Scholar] [CrossRef] [PubMed]
  174. Wang, Y.; Tian, Y.; Pengbo, Z.; Yunyun, M.; He, J.; Lei, J. Self-assembled nanoparticles based on poly(ethylene glycol)–oleanolic acid conjugates for co-delivery of anticancer drugs. RSC Adv. 2017, 7, 29591–29598. [Google Scholar] [CrossRef] [Green Version]
  175. Chen, D.; Cai, L.; Guo, Y.; Chen, J.; Gao, Q.; Yang, J.; Li, Y. Cancer Cell Membrane-Decorated Zeolitic-Imidazolate Frameworks Codelivering Cisplatin and Oleanolic Acid Induce Apoptosis and Reversed Multidrug Resistance on Bladder Carcinoma Cells. ACS Omega 2020, 5, 995–1002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Wu, H.; Zhong, Q.; Zhong, R.; Huang, H.; Xia, Z.; Ke, Z.; Zhang, Z.; Song, J.; Jia, X. Preparation and antitumor evaluation of self-assembling oleanolic acid-loaded Pluronic P105/d-α-tocopheryl polyethylene glycol succinate mixed micelles for non-small-cell lung cancer treatment. Int. J. Nanomed. 2016, 11, 6337–6352. [Google Scholar] [CrossRef] [Green Version]
  177. Alvarado, H.L.; Abrego, G.; Souto, E.B.; Garduño-Ramirez, M.L.; Clares, B.; García, M.L.; Calpena, A.C. Nanoemulsions for dermal controlled release of oleanolic and ursolic acids: In vitro, ex vivo and in vivo characterization. Colloids Surf. B Biointerfaces 2015, 130, 40–47. [Google Scholar] [CrossRef]
  178. Li, W.; Das, S.; Ng, K.Y.; Heng, P.W. Formulation, biological and pharmacokinetic studies of sucrose ester-stabilized nanosuspensions of oleanolic Acid. Pharm. Res. 2011, 28, 2020–2033. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of oleanolic acid and ursolic acid (drawn according to [1]).
Figure 1. Chemical structures of oleanolic acid and ursolic acid (drawn according to [1]).
Agriculture 12 02142 g001
Figure 2. Anticancer ways of OA.
Figure 2. Anticancer ways of OA.
Agriculture 12 02142 g002
Figure 3. Synthesis of derivatives of oleanolic acid [128].
Figure 3. Synthesis of derivatives of oleanolic acid [128].
Agriculture 12 02142 g003
Figure 4. Common modifications and intracellular delivery modes of nanoliposomes.
Figure 4. Common modifications and intracellular delivery modes of nanoliposomes.
Agriculture 12 02142 g004
Figure 5. NPs accumulate at tumor sites through EPR effects and targeted delivery systems of NPs.
Figure 5. NPs accumulate at tumor sites through EPR effects and targeted delivery systems of NPs.
Agriculture 12 02142 g005
Table 1. Natural sources of OA.
Table 1. Natural sources of OA.
Plant SpeciesFamilyPlant PartBiological ActivityReference
Betula albaBetulaceaeBarkAnti-inflammatory, anti-bacterial, anti-viral, antitumor[25,34]
Crataegus pinnatifidaRosaceaeLeavesAnti-inflammatory, anti-bacterial, anti-viral, antitumor[25]
Eriobotrya japonicaRosaceaeFlowersNot mentioned[35]
Fabiana imbricataSolanaceaeLeaves and flowersAntiviral, antitumor, antihyperlipidemic[36]
Ligustrum lucidum AitOleaceaeFruits, leavesAnti-hepatitis, anti-inflammatory, antioxidative, antiprotozoal, antimutagenic, anticancer[37,38,39]
Gentiana luteaGentianaceaeRhizomeAntimicrobial[14]
Lavandula angustifoliaLamiaceaeHerbsAnti-inflammatory, anti-bacterial[25]
Lantana camaraVerbenaceaeLeaves and flowersAnti-inflammatory, antioxidative, antiprotozoal[39]
Melissa officinalisLamiaceaeHerbsAntiviral, hepatoprotective[25,40,41]
Nerium oleanderApocynaceaeLeavesNot mentioned[25]
Olea europaea L. OleaceaeFruits, bark, leavesAnticancer, antimicrobial, anti-diabetic[25,42,43,44,45]
Origanum majoranaLamiaceaeHerbsNot mentioned[25]
Panax quinquefoliumAraliaceaeRootsAnticancer, anti-diabetes, neuroprotection, anti-Aging[46,47]
Phyllanthus amarusPhyllanthaceaeLeaves, aerialsAnti-diabetes[48]
Punica granatum L.LythraceaeFruitAntioxidant activity[13]
Rosmarinus officinalis L.LamiaceaeLeaves, flowers, stems, branchesAnti-inflammatory, hepatoprotective, gastroprotective, antiulcer[43]
Rosa laevigataRosaceaeLeavesAnti-inflammatory[49]
Syzygium aromaticumMyrtaceaeLeaves, flower budsAntinociceptive, Anti-inflammatory, antihypertensive, antioxidant[36,50]
Sambucus nigraAdoxaceaeLeaves, barkAnticancer[25,51]
Satureja montanaLamiaceaeHerbsAnticancer, anti-bacterial[25,52,53]
Siphonodon celastrineusCelastraceaeRoot bark, stemsAnti-inflammatory[54,55]
Silphium trifoliatumAsteraceaeLeavesAnti-bacterial[46,56]
Salvia officinalisLamiaceaeHerbsAnti-bacterial, anti-inflammatory, anticancer, antioxidative[25,57,58]
Thymus vulgarisLamiaceaeHerbsGlutaminase inhibitor[25,59]
Viscum albumSantalaceaeLeaves, stemsAntitumor, analgesic, anti-inflammatory[39,60,61]
Viburnum chingiiAdoxaceaeLeavesAntimicrobial[54,62]
Table 2. Pharmacological actions and signaling pathways of OA.
Table 2. Pharmacological actions and signaling pathways of OA.
Signaling PathwayBiological ActivityReference
Nuclear factor-κB (NF-κB) signaling pathwayAnti-inflammatory, antitumor[65,66,67,68]
Nod-like receptor pyrin domain containing 3 (NLRP3) signaling pathwayAnti-inflammatory, neuroprotection[69,70,71]
Extracellular-signal-regulated kinase (ERK) signaling pathwayLiver protection, antitumor[72,73]
Protein kinase B/Akt signaling pathwayAntitumor, liver protection[72,74,75]
Jun N-terminal kinases (JNK) signaling pathwayAntitumor[1,75]
Orphan receptor γ t signaling pathwayAnti-inflammatory, anti-asthma[76]
Mitogen-activated protein kinases (MAPK) signaling pathwayAnti-inflammatory[77,78]
MiR-122/cyclin G1/myocyte enhancer factor 2D (miR-122/CCNG1/MEF2D) signaling pathwayAntitumor[79]
Cyclic adenosine 3′,5′-monophosphate/protein kinase A (cAMP/PKA) signaling pathwayLowers blood sugar and blood lipids, protects pancreatic islets[80]
Phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathwayAnti-osteoarthritis[81]
Endothelial nitric oxide synthase/Akt/nitric oxide (eNOS/Akt/NO) signaling pathwayAmeliorates high glucose-induced endothelial dysfunction[82]
Signal transducer and activator of transcription 3 (STAT3) and sonic hedgehog (SHH) signaling pathwayInhibits colorectal cancer[83]
Mitogen-activated protein kinase kinase (MEK)/ERK/JNK signaling pathwayAnticancer[84]
Hippo-Yes-associated protein (Hippo-Yap) signaling pathwayAnti-stomach cancer[85]
Epidermal growth factor (EGFR)/AKT signaling pathwayAnti-pancreatic cancer[86]
Nuclear factor erythroid 2-related factor 2 (Nrf-2) signaling pathwayLiver protection, antidiabetic, anti-inflammatory, maintenance of redox and protein homeostasis[87,88,89]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wang, K.; Lu, Q.; Cao, X.; Wang, Y.; Wu, Y.; Chen, Z.; Yang, Z. Application and Development of Natural Plant Metabolite Oleanolic Acid in the Nano Era. Agriculture 2022, 12, 2142. https://doi.org/10.3390/agriculture12122142

AMA Style

Wang K, Lu Q, Cao X, Wang Y, Wu Y, Chen Z, Yang Z. Application and Development of Natural Plant Metabolite Oleanolic Acid in the Nano Era. Agriculture. 2022; 12(12):2142. https://doi.org/10.3390/agriculture12122142

Chicago/Turabian Style

Wang, Kun, Qinyue Lu, Xiang Cao, Yuhao Wang, Yanni Wu, Zhi Chen, and Zhangping Yang. 2022. "Application and Development of Natural Plant Metabolite Oleanolic Acid in the Nano Era" Agriculture 12, no. 12: 2142. https://doi.org/10.3390/agriculture12122142

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

Article Metrics

Back to TopTop