Ursolic acid (UA): A metabolite with promising therapeutic potential

Ursolic acid (UA): A metabolite with promising therapeutic potential

    Ursolic acid (UA): A metabolite with promising therapeutic potential Dharambir Kashyap, Hardeep Singh Tuli, Anil K. Sharma PII: DOI: ...

1MB Sizes 152 Downloads 276 Views

    Ursolic acid (UA): A metabolite with promising therapeutic potential Dharambir Kashyap, Hardeep Singh Tuli, Anil K. Sharma PII: DOI: Reference:

S0024-3205(16)30017-0 doi: 10.1016/j.lfs.2016.01.017 LFS 14654

To appear in:

Life Sciences

Received date: Revised date: Accepted date:

18 September 2015 11 January 2016 12 January 2016

Please cite this article as: Kashyap Dharambir, Tuli Hardeep Singh, Sharma Anil K., Ursolic acid (UA): A metabolite with promising therapeutic potential, Life Sciences (2016), doi: 10.1016/j.lfs.2016.01.017

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Ursolic acid (UA): a metabolite with promising therapeutic potential

1

PT

Dharambir Kashyap1, Hardeep Singh Tuli2*, Anil K. Sharma2

Department of Medical Microbiology, Postgraduate Institute of Medical Education and

Department of Biotechnology, Maharishi Markandeshwar University, Mullana-Ambala,

SC

2

RI

Research (PGIMER), Chandigarh (Punjab), India-160012

MA

NU

Haryana, India -133207.

AC CE P

TE

D

Running Title: Therapeutic potential of Ursolic acid

Corresponding Author: Dr. Hardeep Singh Tuli, Assistant Professor, Department of Biotechnology, Maharishi Markandeshwar University, Mullana (Ambala) Haryana, India133207.Email: [email protected]

1

ACCEPTED MANUSCRIPT

PT

ABSTRACT

RI

Plants are known to produce a variety of bioactive metabolites which are being used to cure various life threatening and chronic diseases. The molecular mechanism of action of such

SC

bioactive molecules, may open up new avenues for the scientific community to develop or

NU

improve novel therapeutic approaches to tackle dreadful diseases such as cancer, cardiovascular and neurodegenerative disorders etc. Ursolic acid (UA) is one among the categories of such

MA

plant-based therapeutic metabolites having multiple intracellular and extracellular targets that play role in apoptosis, metastasis, angiogenesis and inflammatory processes. Moreover, the

TE

D

synthetic derivatives of UA have also been seen to be involved in a range of pharmacological applications, which are associated with prevention of diseases. Evidences suggest that UA could

AC CE P

be used as a potential candidate to develop a comprehensive competent strategy towards the treatment and prevention of health disorders. The review article herein describes the possible therapeutic effects of UA along with putative mechanism of action.

Key Words: Ursolic acid; apoptosis; anti-metastasis; anti-inflammatory; anti-oxidant; derivatives

2

ACCEPTED MANUSCRIPT

PT

Introduction

RI

Plants are the only producers in the ecosystems which have been deliberated to influence mankind throughout life. The bioactive products derived from plants are being considered to be

SC

an unparalleled source to design novel and effective therapeutic agents for the treatment of

NU

dreadful diseases including cancer, cardiovascular and neural disorders [1]. Among the categories of natural products, triterpenoids represent a large family of compounds and comprise

MA

more than 20,000 identified terpenoids including UA with promising remedial value [2,3,4]. UA is a pentacyclic triterpinoid compound which exists either as free acid or as aglycone of

D

saponins. The presence of UA has been confirmed in numerous classes of medicinal plants, such

TE

as Eriobotryajaponica, Rosmarinus offıcinalis, Hedyotis diffusa, Ligustrum lucidum and

AC CE P

Glechoma hederaceae, Arctostaphylos uva-ursi, Vaccinium macrocarpon, Rhododendron hymenanthes Makino, Calluna vulgaris, Ocimum sanctum, Eugenia jambolana and in wax coating of some fruits including apples, pears, prunes [5]. For the last few decades, there has been an extensive research to elucidate the UA mediated pharmacological potential. It has been reported that UA has the ability to modulate a variety of signaling pathways associated with cancer survival and progression, cardiovascular and neural injuries [2]. Moreover, researchers have been able to synthesize derivatives of UA by making chemical modifications in the basic structure which further leads to improve its therapeutic potential and the required active dose. However, in-spite of the availability of such bioactive natural compounds, the cure of inflammation/ or oxidative stress-associated diseases (cancer, cardiovascular and neurodegeneration) remains to be defined. Therefore, in-depth knowledge of 3

ACCEPTED MANUSCRIPT mechanism of action of therapeutic agents such as UA is required so that the scientific community could better understand the modulated signaling pathways during disease

PT

development and would help them out to design some novel therapeutic strategies. The present

RI

review summarized the various therapeutic applications of UA with the possible molecular mechanism of its action.

SC

Chemistry of UA

NU

UA, or 3β-hydroxy-urs-12-en-28-oic-acid, is a pentacyclic triterpenoid (Fig. 1), molecular formula C30H48O3, molecular weight 456.70032 g/mol, acidic, needle-like or crystalline solid,

MA

melting point 283°C-285°C, with a maximum UV absorption wavelength of ~450 nm. The structure of UA comprises of C-30 isoprenoid in the form of pentacyclic triterpenoid. UA has

TE

D

poor solubility in water, but soluble in hot glacial acetic acid and alcoholic NaOH. The biosynthesis of UA is mainly achieved through the folding and cyclization of squalene into

AC CE P

dammarenyl which further undergoes ring expansion and extra cyclization so as to form the fifth ring of UA. It can behave as a bi or tri dentate ligand because of the presence of 3 oxygen atoms and can easily donate lone pairs of electrons to transition metal atoms [6].

Figure-1 Chemical structure of UA 4

ACCEPTED MANUSCRIPT

PT

Induction of apoptosis and cell cycle arrest

RI

Apoptosis a programmed cell death process, characterized by a series of distinct morphological and biochemical changes resulting in natural cell death. It plays an important role in the control

SC

of various biological processes, such as immune responses, hematopoiesis, and embryonic

NU

development [1,7]. Nowadays anticancer therapies are mainly designed to target principle apoptotic pathways to inhibit the growth and proliferation of tumor. A large number of

MA

anticancer agents, including UA is being used to evolve an effective anticancer therapy (Fig. 2). The studies on HepG2 (Human hepatocellular carcinoma) cell line revealed that UA induces

TE

D

apoptosis by releasing the cytochrome C from mitochondria and activating the caspase-3 [8]. DNA fragmentation and other morphological changes including cell shrinkage, cytoplasmic and

AC CE P

nuclear membrane blebbing and an increase in intracellular Ca2+ levels are the hallmark of the apoptosis, which were determined in HL-60 (Human promyelocytic leukemia) cells and Daudi (Human B-lymphoblastoid cell) cells after UA treatment [2,9]. In HepG2 & K562 (Human erythroleukemic cell line) cells, UA was found to inactivate pro-survival proteins (PI3K (phosphoinositide 3 kinase) /Akt) along with down regulation of Bcl2 (B-cell lymphoma 2) expression which are associated with proliferation of cancer cells [10,11]. Using M4Beu (Metastatic pigmented malignant melanoma) cell line Harmand et al., (2005) and Duval et al., (2008), investigated that UA inhibits cell proliferation via altering mitochondrial transmembrane potential as well as Bax (Bcl2 associated protein X)/Bcl-2 balance and proposed UA as a promising candidate in the treatment/ or prevention of skin cancer [12,13]. The expression of molecular proteins linked with extrinsic pathways (Fas/Fas ligand, caspase-8 and PARP (poly 5

ACCEPTED MANUSCRIPT ADP Ribose polymerase)) of apoptotic cell death has also been found to be down regulated by UA in HPV (Human papilloma virus) associated cervical carcinoma cell line [14]. The receptor

PT

for glucocorticoid hormones have also been found to be modulated in the presence of UA which

RI

resulted in the reduction of anti-apoptotic proteins (Bcl2) in human MCF-7 (Michigan cancer foundation-7 cell line) breast cancer cells [15]. Similarly, in hormonal refractory PC-3 (prostate cell

line)

and

androgen-dependent/or

independent

SC

cancer

LNCap

(Human

prostate

NU

adenocarcinoma cancer cell line) cells, UA not only significantly reduced the cell viability but also overcome Bcl-2-mediated apoptotic resistance by down-regulating the expression of Bcl2

MA

protein [16,17]. In a study on HT-29 (Human colorectal cancer-29 cell line) cells, Jian-zhen shan et al., investigated the apoptotic action of UA through the suppression of EGFR/MAPK

TE

D

(Epidermal growth factor receptor /mitogen activated protein kinase) pathway [18]. Moreover, anti-proliferative and apoptotic effects of UA on MDR (Multi drug resistance) SW480

AC CE P

(Colorectal adenocarcinoma cancer cell line) cells were observed via dose dependent downregulation of apoptotic antagonistic molecules [19]. The in vivo/ in vitro studies on gastrointestinal cancer showed that UA modulates the expression of executioner caspase (C-3, C8, C-9) proteins involved in intrinsic as well extrinsic pathways of apoptosis [20]. In addition, UA was found to up-regulate the expression of death receptor in HCT116 (Human colon cancer cell line) and LNCap cells by activating ROS (Reactive oxygen species), JNK (c-Jun N-terminal kinase) and CHOP (C/EBP homologous protein)-dependent pathways [21, 22].The studies carried

out by Zheng et al., (2013) and Gai et al., (2013) on T-24 (Human bladder cancer cell line) cells revealed the apoptotic effect of UA through activation of ER (Endoplasmic reticulum) stress and suppression of Akt (Protein kinase B)/NF-kβ(Nuclear factor-kappa-light chain enhancer of activated β) signaling pathways respectively [23,24]. Another group of researchers reported that 6

ACCEPTED MANUSCRIPT UA, down-regulates survival protein via AMPK (AMP-activated protein kinase) activation and mTOR (Mammalian target of rapamycin) complex 1 inactivation in T24 cells [25]. The UA

PT

treated cells of human leukemia had PKB (Protein kinase B) inactivation, which resulted in JNK

RI

activation followed by suppression of Mcl-1 (Myeloid cell leukemia 1) molecule production [26]. The studies on gemcitabine-resistant PaCa-2, PANC-1 and Capan-1 (Human pancreatic

SC

cancer cell lines) cells reported UA mediated significant growth inhibition via inactivation of the

NU

PI3K/Akt/NF-kβ signaling pathways [27].

In addition to induction of apoptosis, UA has also been known to regulate the cell cycle

MA

regulatory molecules such as cyclin /or CDK (Cyclin dependent kinase). Cell cycle studies of UA treated HepG2 cell line revealed the G0/G1 cell cycle arrest via up-regulation of p21

TE

D

(WAF1) expression [8]. Similarly, HCT15 (Human colon cancer cell line) cells were found to accumulate in G0/G1 phase after 36 h treatment of UA [28]. In a study using MCF-7 cancer

AC CE P

cells, Wang et al., (2010) observed the pro-apoptotic effect of UA in correlation to cyclin D1/CDK4 inactivation, which is further known to play a critical role in cell cycle progression [29]. Another study using SNG-2 and HEC108 (Endometrial adenocarcinoma cancer cell lines) cells has also shown the inhibitory effect of UA on G1 phase regulatory proteins (Cyclin D1) of the cell cycle via modulating MAPK signaling pathways [30]. More recently, Wang and his colleagues investigated the S-phase cell cycle arrest of GBC-SD and SGC-996 (Gallbladder cancer cell lines) cells concomitant with mitochondrial apoptotic cell death [31]. The evidence generally indicates that UA promotes cancer cell apoptosis by inducing cell cycle arrest and modulating associated molecular targets, thereby exerting its oncostatic properties.

7

AC CE P

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

Figure-2: Schematic representation of UA targeted extrinsic and intrinsic apoptotic pathways by activation of caspase-8 and cytochrome-c respectively. Anti-metastatic effect

Metastasis is the vigorous behavior of the growing cancer, related to the major cause of cancer related mortality in the population. Such metastatic activities of cancerous cells are known to ruin the surgery and radiotherapy strategies leaving chemotherapy the only way to cure cancer. Concerning to this fact, the number of chemicals as well as natural compounds are being identified to inhibit the metastatic effect of cancer cells [1,32]. For the past two decades, UA has been progressively used in the cancer research to determine its role as anti-metastatic agent (Fig. 3). The activities of proteases including urokinase and cathepsin B, which are known to be associated with tumor invasion and metastasis have been significantly inhibited by UA [33]. The 8

ACCEPTED MANUSCRIPT IL-1β (interleukin-1 β) /or TNF-α (Tumor necrotic factor-a), induced expression of MMPs (Matrix metallo-proteases) was found to be reduced by UA in C6 glioma cells via inactivating

PT

NF-kβ transcriptional factor through up-regulation of Iκβα (I kappa βalpha) inhibitors [34].

RI

Similarly, dose and time dependent UA treatment in human breast cancer cells led to inactivation of NF-kβ which further resulted in down-regulation of MMP-2, u-PA and up-regulation of

SC

plasminogen activator inhibitor-1 via dephosphorylation of JNK, Akt and m-TOR [35]. In a

NU

couple of studies on TRAMP mice (Transgenic adenocarcinoma of mouse prostate) model, Shanmugam and his colleagues, revealed that UA significantly inhibits the tumor growth by

MA

suppressing pro-inflammatory as well as CXCR4/CXCL12 (Chemokine (C-X-C motif) receptor 4 / chemokine (C-X-C motif) ligand 12) mediated signaling pathways [36]. Also, the

TE

D

concentration dependent anti-invasive and anti-metastatic effect of UA was reported in A549, H3255 and Calu-6 (Human non-small cell lung carcinoma cell lines) cell lines through inhibition

AC CE P

of Na(+) –K(+)-ATPase, TGF-β1 (Transforming growth factor beta1) and ICAM-1 (Intercellular adhesion molecule-1) expression [37]. Similarly the inhibition of molecular proteins, involved in adhesion, invasion and migration of cancer cells have been reported in UA treated in vitro SW620 (Human colonic adenocarcinoma cell line), B16-F10 (Mouse melanoma cell line) and HepG2 cancer cells. Furthermore, using in vivo metastatic melanoma lung cancer C57BL/6 mice model, UA was proposed as a promising anti-metastatic drug [38]. MAPK/P38, is another signaling pathway which is involved in the up-regulation of MMPs expression, and found to be suppressed in SNU-484 (Seoul national university-484) human gastric cancer) cells by UA [3]. Clearly, UA has the capability of suppressing cancer metastasis through several mechanisms, thereby this molecule can be used to control the growth of various cancers.

9

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

AC CE P

Figure-3. Schematic representation of anti-metastatic behavior of UA. UA inhibits the invasiveness of the cancer cell through targeting various pathways, including JNK, NF-kβ, and MAPK/Akt, which subsequently activates the MMPs. Anti-angiogenesis

Angiogenesis, the formation of new capillaries from preexisting vessels, is essential not only for tumor growth and survival but also to enhance its invasive behavior. Several studies indicated that tumor cells create a microenvironment in its surrounding and secrete variety of supportive chemokines for angiogenesis [39,40]. There are many steps during angiogenesis, which could be targeted to inhibit the cancer cell growth (Fig. 4). The in vivo CAM (Chorioallantoic membrane) assay, revealed the promising role of UA in the inhibition of the newly formed vascularization system [41]. The studies on C57BL/6 mice demonstrated that UA suppresses the expression of VEGF (Vascular endothelial growth factor) and iNOS (inducible Nitric oxide synthase) 10

ACCEPTED MANUSCRIPT associated with angiogenesis, at a nontoxic concentration towards endothelial cells [42]. Further, studies on Hep3B, Huh7 and HA22T (Human liver cancer cell lines) cell lines, researchers

PT

demonstrated that UA suppresses the HIF-1α (Hypoxia-inducible factor-1α) which have been

RI

known to increase the expression of angiogenic factors such as VEGF and βFGF (basic Fibroblast growth factor). This down-regulation of HIF-1α expression might be associated with

SC

the antioxidant activity of UA against ROS and NO (Nitric oxide) [43]. In another study using

NU

swiss albino mice model with EAC (Ehrlich ascites carcinoma) tumor Saraswati et al., (2013), investigated that the UA not only down regulates the angiogenesis promoting proteins (VEGF,

MA

iNOS, TNF-α) but also significantly inhibited the growth of cancer cells [44]. Also, Lin et al., (2013), found in vivo (CRC mouse xenograft model) as well as in vitro (HT-29) the anti-

TE

D

angiogenic effect of UA via down-regulating VEGF-A and βFGF through multiple signaling pathways [45]. Authors reported that UA treatment resulted in inhibition of STAT3 (Signal

AC CE P

transducer and activator of transcription-3), Akt/p70S6K (70 kDa ribosomal protein S6 kinase 1) and SHH (Sonic hedgehog) signaling pathways which are associated with tumor survival, proliferation, invasion and angiogenesis [45].Clearly, UA has the potential to suppress tumor angiogenesis via numerous mechanisms (Fig. 4), thereby this molecule could be considered as a potential tumor inhibiting agent in the years to come.

11

AC CE P

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

Figure-4: Illustrates the angiogenesis inhibition by UA, acting at multiple targets. It prevents endothelial cell induction factors such as HIF-1α and VEGF through modulating the different signaling pathways as mentioned. Antioxidant effect

The inability of the cellular immune system to detoxify the ROS results in the expansion of oxidative stress in the micro-environment of cells. Recent evidences have proven the involvement of ROS in the progression of many diseases like cancer, Parkinson’s, Alzheimer’s, atherosclerosis, myocardial infarction and chromosomal disorders [46]. The natural anti-oxidants are now being looked up as convincing therapeutic agents to neutralize free radicals (Fig. 5). The in vivo as well as in vitro studies on CCl4-induced rat and primary culture of rat hepatocytes respectively, suggested that the UA not only reversed the reduced SOD (Superoxide dismutase), 12

ACCEPTED MANUSCRIPT CAT (Catalase), GSH (Glutathione reductase), and GPx (Glutathione peroxidase) activities but also significantly improved the survival rate [47]. Similarly, Saravanan et al., (2006),

PT

investigated the hepato-protective role of UA against ethanol-mediated oxidative stress in rat by

RI

increasing and decreasing the oxidative stress markers such as glutathione, ascorbic acid, αtocopherol and lipid peroxidation respectively [48]. Furthermore, the UA was found to reverse

SC

the β-galactose induced oxidative neurological damage and positively improved the level of

NU

GAP43 (Growth-associated protein 43) in the treated mice [49]. In a study using human blood lymphocytes, Ramachandran et al., (2008), observed that pretreatment of UA normalized the

MA

oxidative effect of UVB (Ultra violet B) irradiation by inhibiting lipid peroxidation and proposed it as a potent candidate for photo-induced skin diseases treatment [50]. In a study conducted on

D

PC12 cell line (Neurological cell model) indicated that UA attenuated the H2O2 (hydrogen

TE

peroxide)/ or MPP+ (1-methyl-4-phenylpyridinium ion) induced cell death as well the formation

AC CE P

of MDA [51] (Malonyldialdehyde). Ramos et al., (2010) confirmed an improvement in the DNA repair mechanism by the UA application via increasing the DNA incision activity [52]. The oxidative stress has also been found to be associated with increased expression of receptor for RAGE (Advanced glycation end-products) which subsequently promotes the progression of various types of cancer and cardiovascular diseases. Using, streptozotocin-induced diabetic rat as an experimental model, Xiang et al, determined that UA may show anti-oxidant activity by modulating the RAGE-NADPH (Nicotinamide adenine dinucleotide phosphate) oxidase-NFkβsignaling pathway [53]. Nrf2 (Nuclear factor erythroid 2-related factor 2), a key molecule responsible for the up-regulation of enzymes (GST and NQO1 (NAD (P) H dehydrogenase, quinone 1)) involved in detoxification of oxidative species, was activated by UA in CSE (Cigarette smoke extracts) induced human bronchial epithelial cell culture model [154,155]. To 13

ACCEPTED MANUSCRIPT conclude, a growing body of evidence suggests its global action on oxidative stress mediated

AC CE P

TE

D

MA

NU

SC

RI

PT

disorders which makes UA to be of great interest for future clinical research on public health.

Figure-5: Illustration of the role of UA to reduce oxidative stress by modulating RAGE NADPH, Nrf2 and different catalases expression. Anti-inflammatory effect Inflammation is a complex phenomenon which is associated with progression of various diseases such as neurodegenerative, cardiovascular and cancer. The transcriptional factor NF-kβ is being considered to be a major cell signaling pathway which up regulates the expression of various inflammatory genes [56]. Variety of soluble and membrane-bound extracellular ligands TNFR (Tumor necrosis factor receptor), TLR (Toll like receptor), and IL-1R (Interleukin receptor-1)) are known to activate the NF-kβ mediated inflammatory pathways (Fig. 6). The anti14

ACCEPTED MANUSCRIPT inflammatory activity of UA at 0.14 mM concentration, was analyzed using croton oil-induced oedema in albino swiss mice [57]. The release of pro-inflammatory cytokines such as IL-2

PT

(Interleukin-2), IFN-γ (Interferon-γ) and TNF-α from Th-2 (T-helper-2) cell of arthritic balb/c

RI

mice was reduced by UA [58]. The pro-inflammatory enzymes like sPLA2 (Secretory phospholipase A 2) which hydrolyzes the phospholipids into a diverse range of inflammatory

SC

mediators have been found to be deactivated in the presences of UA [59]. In a study using

NU

HUVECs (Human umbilical vein endothelial cells), Takada et al., (2010), reported UA mediated down-regulation in the expression of E-selectin via inhibition NF-kβ translocation into the

MA

nucleus [60]. The in vivo studies on prefrontal cortex of mouse suggested that the UA not only reduced the formation of AGEs (advanced glycation end products) and ROS but also

TE

D

significantly suppressed the expression of iNOS and COX-2 (Cyclooxigenase-2), IL-1β, IL-6, and TNF-α [61]. Similarly, Wang and his colleagues reported the neuro-protective role of UA in

AC CE P

LPS (Lipopolysaccharide) induced cognitive deficit mice [62]. In addition to inhibition of NFkβ, UA also suppress the other transcriptional factors such as NF-AT (Nuclear factor activated T cells) and AP-1 (Activator protein-1) in lymphocytes [63]. In another study on TRAMP mice, UA reduced the tumor growth and increased the survival rate of mice by down-regulating the activation of inflammatory mediators including STAT3, AKT and IKKa/b [4,64]. These findings suggest that UA could have tremendous potential in the development of an effective antiinflammatory drug. These anti-inflammatory actions seem not to be exclusively mediated by the free radical scavenging properties of UA which projects this molecule as a new class of potential anti-inflammatory agents.

15

AC CE P

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

Figure-6: The anti-inflammatory property of the UA via SATA3, Akt/mTOR, and NF-kβ mediated pathways.

Antimicrobial and Other therapeutic functions Variety of pathogens exist in the ecosystem which are responsible for spreading numerous lifethreatening infectious diseases. The antimicrobial properties of UA has been extensively studied against several harmful pathogens including bacteria, virus, fungi, and parasites (Table 1). The developing drug resistance in pathogenic strains of microbes is consistently prompting researchers to look for better alternatives as antimicrobial agents. The naturally occurring bioactive compounds may serve as a promising tool to resolve drug resistant related issues and to reduce the side effects of antibiotics. It has been reported that UA modulates microbial genes 16

ACCEPTED MANUSCRIPT expression, induces stress responses and cell autolysis, inhibits biofilm formation and peptidoglycan turnover [65]. Besides, UA has been known to possess a variety of other

PT

beneficial pharmacological applications, which have been summarized in table 2, along with the

RI

proposed molecular mechanism of action.

3

Anti-parasitic

                                                

NU

Antiviral

Escherichia coli Escherichia coli (UPEC) Escherichia coli (ATCC 700336) Escherichia coli (ATCC 25922) Escherichia coli (ATCC 27) Escherichia coli (MDREC-KG4) Bacillus cereus (ATCC 33018) Bacillus subtilis Bacillus sphaericus Pseudomonas aeruginosa (ATCC 15442) Pseudomonas syringae Aeromonas caveae (ATCC 15468) Klebsiella pneumoniae (ATCC 10031) Shigella flexneri (ATCC 12022) Vibrio colorize (ATCC 15748) Listeria monocytogenes (ATCC 19117) Staphylococcus aureus (ATCC 12692) Staphylococcus aureus (ATCC 12624) Staphylococcus aureus (ATCC 6538) Staphylococcus aureus (NCTC 8325) Staphylococcus aureus,MRSA Streptococcus mutans (ATCC 25175T ) Streptococcus mutans Streptococcus sobrinus (ATCC 33478 T) Streptococcus pneumoniae Salmonella typhi Helicobacter pylori (ATCC 43504) Vancomycin-resistantEnterococcus Mycobacterium tuberculosis H37Ra Mycobacterium tuberculosis H37Rv (27294) Mycobacterium tuberculosis RMPr Mycobacterium tuberculosis INHr (35822) Mycobacterium tuberculosis RIF-R (35838) Mycobacterium tuberculosis EMB-R (35837) Mycobacterium tuberculosis STR-R (35820) Mycobacterium tuberculosis MMDO Mycobacterium tuberculosis MTY147 Pig cytomegalovirus (GPCMV) Human papillomavirus type 11 Influenza virus type A (H1N1) Avian influenza type A (H5N1) Hepatitis C virus (HCV) Human immunodeficiency virus (HIV) Setaria cervi Wuchereria bancrofti Trypanosoma cruzi (Y strain) Trypanosoma brucei rhodesiense Plasmodium falciparum K1 Plasmodium falciparum 3D7

MA

2

                                                

D

Antibacterial

Organism

TE

Effect

AC CE P

S. No. 1

SC

Table 1: An overview of antimicrobial activities mediated by UA.

17

MIC

3 to 4°10−4 M 10 µg/mL 256 µg/mL 64 µg/mL 512 µg/mL 1000 µg/mL 1024 µg/mL 25 µg/mL 50 µg/mL 512 µg/mL 25 µg/mL 1024 µg/mL 64 µg/mL 64 µg/mL 1024 µg/mL ≥1024 µg/mL 1024 µg/mL ≥1024 µg/mL ≥32 µg/mL 6 mg/mL 30 mg/mL 4 µg/mL 0.1, 0.2, 0.5 wt% 8 µg/mL 4 µg/mL 50 µg/mL 50 µg/mL 3 µg/mL 10 µg/mL 6.25 & 25 µg/mL 25 µg/mL 6.25 & 25 µg/mL 25 µg/mL 25 µg/mL 12.5 µg/mL 25 µg/mL 25 µg/mL 6.8 µg/mL 12.41 µg/mL >200 µg/mL 6.00-9.25 mM 58.4 l µg/mL 0.3 µM 5 µg/mL 5 µg/mL 25 mg/kg 1.5 µg/mL 49 µg/mL 12.7 µg/mL

References                                                 

[66] [67] [68] [69] [69] [70] [69] [71] [71] [69] [71] [69] [69] [69] [69] [69] [69] [69] [69] [72] [73] [74] [75] [74] [76] [71] [71] [71] [77] [78,79] [78] [78,79] [79] [79] [79] [79] [79] [80] [81] [81] [82] [83] [84,85,86] [87] [87] [88,89] [90] [90] [90]

ACCEPTED MANUSCRIPT 4



Anti-fungal

 0.84 µg/mL

Saccharomyces cerevisiae

 [91]

Table-2: An overview of UA mediated therapeutic effects along with possible mechanism of action.

  

4

Anti-hyperlipidemia



 

5

Anti-lipase lipolytic activities



6

Cardioprotective

 

  

7

Hepatoprotective

      

PT

Male rat

β-adrenergic antagonistic activity Attenuate β-adrenergic induced ANP secretion, ↑ cAMP levels and atrial dynamics, ↑ acetylcholineinduced increase in ANP secretion and ↓ pulse pressure Blunt RAGE-NADPH oxidase-NF-kβ signal transduction pathway Down-regulate miR-21 and p-ERK/ERK Inhibit monocyte dysfunction, improve kidney function Apoptosis, ↓ phosphorylation level of Akt, and inhibit nuclear localization of NF-kβ Inhibit plasma AST and ALT, ↓ plasma creatinine and blood urea nitrogen level, ↑ GHS redox status, improve mitochondrial function ↓ Serum enzyme level, SGPT, SGOT, SAKP and SBIL ↓ Liver weight, serum level of ALT/AST and hepatic steatosis, ↑ lipid β-oxidation and inhibit the hepatic ER stress. ↑ Cyclin D1, cyclin E and C/EBPβ protein expression level ↑ Nrf2, HO-1, NQO1 and GST, ↓ TNF-α, PGE2, iNOS Autophagy via an AMPK/mTOR pathway

40 mg/kg 30 µM

Rat Rabbit

[107] [108]

50 mg/kg

Diabetic rat

[53]

---0.3–10 µM

Mice Female LDLR−/− mice

[109] [110,111]

40 µM

Female SD rat

[112]

0.35-0.7 mg/kg

Long rats

Evan's

[113]

-------

db/db mice/ L02 cells ----------

[114]

------------

Mice

[116]

25, 50 mg/kg

ICR mice

[55]

0, 10, 20, 40µm

HepG2 cells Male ICR

[117]

RI

100 mg/kg, 51.21 µM

AC CE P



References

64 µM 0.01-0.1 mg/kg, and 10 mg/kg, 0.05% wt/wt

SC

 

Anti-diabetic

Experimental model CHO Cell Male Swiss mice

Block Aβ-CD36 interactions, ↓ ROS production. Interaction with the dopaminergic system, through the activation of dopamine D1 and D2 receptors. Serotonergic and noradrenergic system involvement Inhibit SDH and aldose reductase activity, ↑ glucokinase activity, hepatic fatty acid β-oxidation and CPT1, FAS activity, ↓ G6P activity Inhibit PTP1B ↓ Blood glucose, organ coefficient of kidney, BUN and Cr, level of MDA and TNF-α, IL-6, ↓SOD activities ↓ Hepatic G6P, glucokinase activity, ↑ glucokinase/G6P ratio, GLUT2 mRNA level and glycogen content, ↑ reductase activity, ↓SDH activity Normalized serum marker enzymes such as creatine kinase, creatine kinase-MB and LDH, expression of LDH 1 and LDH 2 isoenzymes, ↓ level of plasma TC, low density lipoprotein-cholesterol, very low density lipoprotein-cholesterol, TG, FFA, PL and atherogenic index, ↑ level of high density lipoprotein-cholesterol, ↓ DNA damage, myocardial infarct size. Inactivation of acetyl/malonyl transferase and via NADPH and KR domain Reduce AST and ALT activity, down-regulate SREBP-1c, FAS and ACC and CPT1 and PPAR-α Reduced protein expression of C/EBPb, PPARc, C/EBPa and SREBP-1c, ↑ phosphorylation of ACC and CPT1, ↓ protein expression of FAS and FABP4, ↑ phosphorylation of AMPK and protein expression of Sirt1 Inhibit pancreatic lipase and enhance lipolysis in fat cells

NU

 

Dose

MA

Alzheimer disease Antidepressant/ Anxiolytic-like

Mechanism

D

3

Therapeutic effects

TE

S. No 1 2

18

2.0 µM 35 mg/kg

0.01-0.05%

40 mg/kg

Male diabetic mice CHO/hIR cell and L6 myotube Male Kunming mice Mice Male albino

[92] [93,94], [95] [96]

[97,98]

[99]

[100] [101]

6.0 µg/mL

Wistar rat Male

[102,103]

5 mg/kg

C57BL/6J mice

[104]

Rabbit

[105]

25 mg/kg

0.14% w/w/ 1030 µg/mL

Wistar

[106]

[115]

ACCEPTED MANUSCRIPT 25, 50 mg/kg

mice

[118]



↓ Elevation in the level of Serum urea, serum uric acid, serum creatinine and blood urea nitrogen ↓Urine albumin excretion, renal oxidative stress level, NF-kβ activity, and P-selectin expression GABAa receptor activation Attenuate including brain edema, blood–brain barrier disruption, neural cell apoptosis, and neurological deficient, up-regulate the antioxidative level Inhibit PEP ↑S100 mRNA expression and protein ↓ the expressions of ICAM-1, TLR4,NF-kβ, P65, IL1β, TNF-α, IL-6, iNOS and MMP-9 ↓8-hydroxy-2- deoxyguanosine level, TNF-α, IL-6, IL-17, COX-2, modulate STAT3 and NF-kβ signaling pathways. Influence L - type Ca2+ channels, and reduce the Ca2+ influx.

2, 5, 10 mg/kg

Wistar albino rat HSC

[119]

Male ICR mice Male SD rat

[121] [122]

-----------BALB/c mice SD rats

[123] [124]

9

Neuroprotective

     

0.2% 0.3 mg/kg 25-50 mg/kg

PT



IC50 17.2 10, 5, mg/kg 50 mg/kg

RI

Nephroprotective

↓CYP2E1, TNF-α, IL-1β, COX-2, activation of JNK, p38 MAPK, ERK, NF- kβ

2.5

SC

8



[120]

[125]

25, 50 mg/kg

Male ICR

[64]

1 x 10(7) mol/L - 5 x 10(5) mol/ L

---------

[126]

Gastro-protective



11

Anti-asthmatic



Reduce Th2 cytokines (IL-5 and IL-13), ovalbuminspecific IgE production, and eosinophil infiltration via the Th2-GATA-3, STAT6, and IL-17β, NF-kβ pathway

--------

BALB/c mice

[127]

12

Anti-hormone



↓mRNA level of GREB1, estrogen-responsive protein, mRNA and protein level of ERα, enhanced prostate-specific antigen promoter activity Induced apoptosis, JNK pathway activation

--------

---------

[128]

10, 20, 40 µM

T20 cells

[129]

Reduce infiltration of leukocytes and proteins, myeloperoxidase activity, and malondialdehyde content, ↓ the serum level of TNF-α, IL-6, and IL-1β, inhibited the expression of iNOS and COX-2 ↑ Fas expression, apoptosis, IL-10, ↑ CD4+ Foxp3+ and CD25+ Foxp3+ T cells, ↓ IL17α, IgG2b antiAchR Suppress leukocyte migration and PGE2 production, hyperalgesia and spinal Fos expression

10 mg/kg

Rat

[130]

20-100 mg/kg

MG rat

[131]

50 mg/kg

Rat

[132]

Inhibit NF-kβ and JNK-related signaling pathways, mRNA and protein expression of NFATc1, ↓ TRAP+ osteoclasts, attenuate Ti-particle-induced mouse calvarial bone loss. Target Tph-1 and suppress serotonin biosynthesis ↑ Anti-aging biomarkers such as SIRT1 and PGC-1α, ↓ cellular energy charges such as ATP and ADP, enhance neomyogenesis, ↑ myoglobin expression

5 mM

Male C57BL/6

[133]

10-20 mg/kg 200 mg/kg

OVX rat C57BL mice

[134] [135]

Attenuate the hepatic malondialdehyde level and reduced the plasma AST and ALT level after trauma– hemorrhagic shock, inhibit superoxide anion generation and elastase release in human neutrophils ↑ Ceramide and collagen content, ↓ keratin 1, keratin 10 and involucrin ↑ Protein expression of PPAR-α, involucrin, loricrin and filaggrin, Induce epidermal keratinocyte differentiation via PPAR-α

1, 3, 10 mM

Male SD rat

[136]

1.0 %

NHEK, NHDF HaCaT cells

[137]

0.5%

Rat

[139]



↑ FFA uptake and β-oxidation via an UCP3/AMPKdependent pathway Sustain resistance exercise-induced mTORC1 activity

25 mg/mL

Male SD rat

[140]



Most likely by antioxidant effect

80 mg/kg

Balb/c mice

[141]



14

Autoimmune disease



15

Anti-arthritic

16

Osteo-protective

17

Aging-metabolic phenotype

18

TraumaHemorrhage Shock



19

Dermo-protection



20

Improve epidermal permeability barrier



21

Metabolism



22

MA

D

Sepsis

AC CE P

13

TE



NU

10

Anti-mutagenicity

 

 

19

0.1 mg/mL, 10 µmol/L

[138]

ACCEPTED MANUSCRIPT 23

Target CSC



Inhibit Oct4, Tert, Bmi1, β-catenin, ABCG2, and Ep300 gene, ↓ CK19+ cells and ↑ CK8/18+ cells

50 µM

PLC/PRF/5, Huh7 HCC cells

[142]

24

Colitis



Inhibit pro-inflammatory cytokines, phosphorylation/ degradation and NF-kβ

20 mg/kg

C57BL/6

[143]

25

Epigenetic modification



Inhibit DNMT1

20 µM

HCC cell line

[144]

PT

IkBα

AC CE P

TE

D

MA

NU

SC

RI

Abbreviations- Normal human epidermal keratinocytes (NHEK), Normal human dermal fibroblasts (NHDF), Prostaglandin E2 (PGE2), Hepatic stellate cells (HSCs), Protein tyrosine phosphatase 1B (PTP1B), Lactate dehydrogenease (LDH), Total cholesterol (TC), Triglycerides (TG), Free fatty acids (FFA), Phospholipid (PL), Reactive oxygen species (ROS), Nicotinamide adenine dinucleotide phosphate (NADPH), Nuclear factor kappalight chain enhancer of activated B (NF-kβ), Protein kinase B (Akt), Chinese hamster ovary cell (CHO), Peroxisome proliferator- activated receptor alpha (PPAR-α), Rafampicin resistance (RMPr), Myasthenia gravis (MG), c-Jun Nterminal kinase (JNK), Titanium (Ti), Tryptophan hydroxylase 1 (Tph-1), Ovariectomized (OVX), Sprague- Dawley (SD), Imprinting Control Region (ICR), Prolyl endopeptidase (PEP), Cyclooxygenase-2 (COX-2), inducible Nitric oxide synthase (iNOS), Interleukin-6 (IL-6), Interleukin-1b (IL-1b), Endoplasmic reticulum (ER), CCAAT element binding protein b (C/EBPb), Peroxisome proliferator-activated receptor c (PPARc), CCAAT element binding protein a (C/EBPa), Sterol regulatory element binding protein 1c (SREBP-1c), Acetyl-CoA carboxylase (ACC), Carnitine palmitoyltransferase 1 (CPT1), Fatty acid synthase (FAS), Fatty acid-binding protein 4 (FABP4), AMPactivated protein kinase (AMPK), Silent mating type information regulation 2, homolog 1 (Sirt1), Aspartate aminotransferase (AST), Alanine aminotransferase (ALT), Mammalian target of rapamycin complex 1 (mTORC1), T helper 1 cell (Th2), Immunoglobulin E (IgE), Signal transducer and activator of transcription 6 (STAT6), Growth regulation by estrogen in breast cancer 1 (GREB1), Uncoupling protein 3 (UCP3), Peroxisome proliferator-activated receptor-γ coactivator (PGC-1α), Nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 1 (NFATC1), Tartrate-resistant acid phosphatase (TRAP), Phosphoenolpyruvic acid (PEP), γ-aminobutyric acid a (GABAa), Serum glutamate-pyruvate transaminase (SGPT), Serum glutamic oxaloacetic transaminase (SGOT), Serum alkaline phosphatase (SAKP), Serum biluribin (SBIL), Extracellular signal-regulated kinases (ERK), Receptor for advanced glycation end products (RAGE), Atrial natriuretic peptide (ANP), Lactate dehydrogenase 1&2 (LDH 1&2), Glucose-6-phosphatase (G6P), Glucose transporter 2 (GLUT2), Sorbitol dehydrogenase (SDH), Blood Urea Nitrogen (BUN), Protein-tyrosine phosphatase 1B (PTP1B), Malondialdehyde (MDA).

Derivatives of UA

Though, UA has been declared to exhibit potent therapeutic activities against different kinds of disorders, however, the major restriction to use UA in clinical applications is associated with its less target specificity and bioavailability in the biological systems. Therefore, synthesis of UA derivatives by chemical modifications might be used as an important tool to enhance its biological and pharmacological potential [145]. It has been reported that modifications of UA at the C-3 and/or C-28 positions using functional group not only improve the bioactivity but also enhances the bioavailability rate. In a study using heterocyclic derivatives of UA, Leal and his colleagues investigated the improved anti-cancer action against AsPC-1 (Human pancreatic 20

ACCEPTED MANUSCRIPT cancer) cells [146]. Similarly, Rashid and his colleagues synthesized the triazolyl-derivatives of UA with higher bioactivity as compared to UA [114]. Another group of researchers have

PT

investigated that the addition of an acyl piperazine group at C-28 position of UA can increase its

RI

anticancer potential [147]. Table 3, summarizes the other clinically active UA derivatives along with their reported therapeutic potential and proposed molecular mechanism of action.

SC

Table-3: Summary of various UA based derivatives.

Role with mechanism



N-[3β-acetoxy-urs-12-en-28-oyl]-aminoN-(3,4,5-trimethoxyphenyl) piperazine1-carbothioamide



Apoptosis, cell arrest in G1 phase

cycle

2



N-[3β-acetoxy-urs-12-en-28-oyl]-2aminodiethanol



Apoptosis, cell arrest in S phase

cycle

3





4



N-(3β-hydroxyurs-12-en-28-oyl)-4aminobutyric acid 3-Oxo-urs-12-en-28-oic acid methyl ester

Inhibit aldose reductase (AR) Cytotoxicity

5



6



7

MA

Derivative

NU

S. No. 1

D



Experimental model MGC-803, HCT116, T24, HepG2 A549, HL-7702 cell lines HepG2, BGC-823, SH-SY5Y, HeLa, HELF cell lines E. coliBL21 strain (DE3) HL0-60, BGC, Bel7402, Hela cell lines HeLa, BGC-823, SKOV3 cell lines

Dose

Reference

27.08-38.78 µM

[147]

>100 mM

[148]

20 µM

[146]

10-100 µg/ml

[149]

10 µM

[150]

HepG2, HeLa, L02, HELF cell lines

0.5, 1.5, 5, 10, 20, 40, 80 mM

[151]

Rat C6 glioma, A431 cell line NTUB1 cells

10-100 µM

[152]

4, 20, 50 µM

[153]

HepG2, H22 cell lines

33.12-68.82 mM

[154]

[68]



Apoptosis and arrest cell cycle in S-phase





7,24-dihydroxy ursolic acid



Deplete ATP, ↓ lactate production, cell cycle arrest in S and G2/M phase, down-regulate Bcl2 and HKII, upregulate Bax and p53, suppression glucose metabolism Cytotoxicity

8



Isopropyl 3β-hydroxyurs-12-en-28-oate and Methyl 3,4-seco-ursan-4



Apoptosis, production

9



3β-acetoxy-urs-12-en-28-oic hexamethylenediamine

acid



Apoptosis glucolysis

10



cis-3-O-p-hydroxycinnamoyl acid

Ursolic



Induced inflammation

ATCC 700336, ATCC 25922 (E coli strains)

256 µg/mL

11



Corosolic acid (2α-hydroxyursolic acid)



3-epi-corosolic acid





N-[3β-Acetoxy-urs-11-oxo-12-en-28acyl]aniline N-[3-Acetoxy-urs-11-oxo-12-en-28acyl]-4-methylpiperazine N-[3-Oxo-urs-12-en-28-oyl]-3-amino-1propanol N-[3-Oxo-urs-12-en-28-oyl]-4

 

Colon cancer cell line old female IRC mice HeLa cells, HeLa, BGC-823 and SKOV3 cells.

40 µM



Inhibit Wnt/β-catenin pathway inhibit expression of IL1a, IL-1b, IL-6 and IL-23 Anti-tumor Anti-proliferative

13

  

AC CE P

TE

N-[3β-Acetoxy urs-12-en-28-oyl]-2aminoethanol acetate and N-[3βAcetoxy-urs-12-en-28-oyl]-2-amino-1propanol acetate N-[3β-acetoxy-urs-12-en-28-oyl]-amino2-methylpiperazine

21

ROS

and

target

[155,156]

200µL 10 μmol/L

[157]

 14

 

15



Cyano-3-oxo-12a-fluoro-urs1-en-13,28bolide



Anti-neoplastic



Anti-proliferation , cell cycle arrest in G1 phase, ↑ p21waf , NOXA and ↓ cFLIP

HeLa BGC-823

SKOV3

2.16±0.26, >10, 9.7±1.01

[158]

PC-1, MIA PaCa-2, PANC-1, MCF7, PC-3, HepG2, A549

0.7, 0.9, 1.8µm and 1µm

[159]

RI



SC



methoxyaniline N-[3β-Acetoxyurs-12-en-28-oyl]-3’,4’difluorobenzylamine N-[3β-Acetoxyurs-12-en-28-oyl]-3morpholin-4-yl-1-propylamine Methyl N-[3β-butyryloxyl-urs-12-en-28oyl]-2-amine acetate N-[3βbutyryloxyl-urs-12-en-28-oyl]benzyl amine N-[3β-acetoxy-urs-12-en-28-oyl]morpholine N-[3β-butyroxy -urs-12-en-28-oyl]morpholine

NU



PT

ACCEPTED MANUSCRIPT

MA

Harmful effects of UA

As discussed in the former sections, UA has both in vitro and in vivo therapeutic functions

TE

D

including anticancer, antioxidant and anti-inflammatory activities. Besides these positively published reports, there have been some undesirable pro-inflammatory effects noticed.

AC CE P

Investigations in macrophages suggested the enhanced expression of iNOS and TNF-alpha mRNA via NF-kβ activations by the UA [160]. Furthermore, Ikeda and his colleagues published a series of articles, demonstrating the in vitro as well in vivo pro-inflammatory effects of UA. Ikeda et al., (2005), first reported that UA increases the release of macrophage migration inhibitory factor (MIF) by activating extracellular signal-regulated kinase (ERK)-2 [161]. Their other studies on mouse skin reported a significant increase in the expression of cyclooxygenase COX-1, COX-2, and TNF-alpha factors UA treatment [162]. Interleukin-1β (IL-1), a well-known pro-inflammatory mediator has also been found to be up regulated in UA treated macrophages and colonic mucosa of ICR mice [163]. In another toxicological study, Jing-Bo (2009), has reported the LD50 dose (9.26 g/ kg) of UA extract and suggested it as a safe candidate without any genetic toxicity [164]. More recently to investigate the toxicity and pharmacokinetics of UA 22

ACCEPTED MANUSCRIPT liposomes (UAL) a study was carried out on 63 subjects including patients with advanced solid tumor and healthy volunteers. Results have revealed that UAL treatment possessed manageable

PT

toxicity with maximum tolerated dose (MTD) of 98 mg/m2 and liver associated dose-limiting

RI

toxicity (DLT) [165]. Conclusions and future perspectives

SC

For the last few decades people seem to be more conscious about the usage of herbal medicines

NU

(for e.g. UA) over the synthetic ones. The present review has brought forward the variety of therapeutic roles of UA. Evidences suggest that UA has the ability to modulate multiple

MA

molecular signaling pathways associated with cancer, inflammation, neurological, and cardiovascular diseases. However, further research work is needed to explore other intra as well

TE

D

as extra-cellular targets of UA using QSAR models [166]. The synthesis of UA derivatives could be used as a promising technology to overcome the evolutionary development of cellular

AC CE P

resistance and to improve the therapeutic efficacy towards the targeted diseases. The beneficial effects of UA can be increased many fold by using synergistic approaches with other chemopreventive or therapeutic molecules [167-170]. Cho et al., (2015), reported significant inhibition of 12-O-tetradecanoylphorbol-13-acetate induced skin tumor on using a combination of UA and Resveratrol [171]. The combination of UA and leucine may inhibit the age related muscular damages by initiating the myogenic differentiation [172]. Furthermore the role of nanobiotechnology could confer the revolutionary impact not only to increase the target specificity and bio-availability of UA but also to minimize the requirement of active doses for the treatment.

Conflict of interest None 23

ACCEPTED MANUSCRIPT Acknowledgments The authors would like to acknowledge Maharishi Markandeshwar University, Mullana-

PT

Ambala, for providing the requisite facilities to complete this study. We would also like to

RI

acknowledge, Dr. Shilpa Thakur, Department of biochemistry, Postgraduate Institute of Medical

SC

Education and Research (PGIMER), Chandigarh (Punjab), for her assistance in the preparation

NU

of revised manuscript. REFERENCES

H.S. Tuli, A.K. Sharma, S.S. Sandhu, D. Kashyap, Cordycepin: A bioactive metabolite with therapeutic potential, Life Sci. 93 (2013) 863–869. doi:10.1016/j.lfs.2013.09.030.

[2]

J.H. Baek, Y.S. Lee, C.M. Kang, J. a Kim, K.S. Kwon, H.C. Son, et al., Intracellular Ca2+ release mediates ursolic acid-induced apoptosis in human leukemic HL-60 cells., Int. J. Cancer. 73 (1997) 725–728. doi:10.1002/(SICI)1097-0215(19971127)73:5<725::AIDIJC19>3.0.CO;2-4.

[3]

E. Kim, A. Moon, Ursolic acid inhibits the invasive phenotype of SNU-484 human gastric cancer cells, Oncol. Lett. (2014) 897–902. doi:10.3892/ol.2014.2735.

[4]

M.K. Shanmugam, T.H. Ong, A.P. Kumar, C.K. Lun, P.C. Ho, P.T.H. Wong, et al., Ursolic acid inhibits the initiation, progression of prostate cancer and prolongs the survival of TRAMP mice by modulating pro-inflammatory pathways, PLoS One. 7 (2012) 1–9. doi:10.1371/journal.pone.0032476.

[5]

G. Li, X. Zhang, J. You, C. Song, Z. Sun, L. Xia, et al., Highly sensitive and selective precolumn derivatization high-performance liquid chromatography approach for rapid determination of triterpenes oleanolic and ursolic acids and application to Swertia species: Optimization of triterpenic acids extraction an, Anal. Chim. Acta. 688 (2011) 208–218. doi:10.1016/j.aca.2011.01.010.

[6]

H.S. Tuli, S.S. Sandhu, D. Kashyap, A.K. Sharma, Optimization of extraction condition and antimicrobial potential of a bioactive metabolite, cordycepin from cordyceps militaris 3936, 3 (2014) 1525–1535.

[7]

H.S. Tuli, G. Kumar, S.S. Sandhu, A.K. Sharma, D. Kashyap, Apoptotic effect of cordycepin on A549 human lung cancer cell line, Turkish J. Biol. 39 (2015) 306–311. doi:10.3906/biy-1408-14.

AC CE P

TE

D

MA

[1]

24

ACCEPTED MANUSCRIPT D.K. Kim, J.H. Baek, C.M. Kang, M.A. Yoo, J.W. Sung, D.K. Kim, et al., Apoptotic activity of ursolic acid may correlate with the inhibition of initiation of DNA replication, Int. J. Cancer. 87 (2000) 629–636. doi:10.1002/1097-0215(20000901)87:5<629::AIDIJC2>3.0.CO;2-P.

[9]

F. Lauthier, L. Taillet, P. Trouillas, C. Delage, a Simon, Ursolic acid triggers calciumdependent apoptosis in human Daudi cells., Anticancer. Drugs. 11 (2000) 737–745. doi:10.1097/00001813-200010000-00011.

RI

PT

[8]

NU

SC

[10] C. Tang, Y.-H. Lu, J.-H. Xie, F. Wang, J.-N. Zou, J.-S. Yang, et al., Downregulation of survivin and activation of caspase-3 through the PI3K/Akt pathway in ursolic acidinduced HepG2 cell apoptosis., Anticancer. Drugs. 20 (2009) 249–258. doi:10.1097/CAD.0b013e328327d476.

MA

[11] B. Wu, X. Wang, Z.F. Chi, R. Hu, R. Zhang, W. Yang, et al., Ursolic acid-induced apoptosis in K562 cells involving upregulation of PTEN gene expression and inactivation of the PI3K/Akt pathway, Arch. Pharm. Res. 35 (2012) 543–548. doi:10.1007/s12272012-0318-1.

TE

D

[12] P.O. Harmand, R. Duval, C. Delage, A. Simon, Ursolic acid induces apoptosis through mitochondrial intrinsic pathway and caspase-3 activation in M4Beu melanoma cells, Int. J. Cancer. 114 (2005) 1–11. doi:10.1002/ijc.20588.

AC CE P

[13] R.E. Duval, P.O. Harmand, C. Jayat-Vignoles, J. Cook-Moreau, A. Pinon, C. Delage, et al., Differential involvement of mitochondria during ursolic acid-induced apoptotic process in HaCaT and M4Beu cells, Oncol. Rep. 19 (2008) 145–149. [14] E.K. Yim, M.J. Lee, K.H. Lee, S.J. Um, J.S. Park, Antiproliferative and antiviral mechanisms of ursolic acid and dexamethasone in cervical carcinoma cell lines, Int. J. Gynecol. Cancer. 16 (2006) 2023–2031. doi:10.1111/j.1525-1438.2006.00726.x. [15] E. Kassi, Z. Papoutsi, H. Pratsinis, N. Aligiannis, M. Manoussakis, P. Moutsatsou, Ursolic acid, a naturally occurring triterpenoid, demonstrates anticancer activity on human prostate cancer cells, J. Cancer Res. Clin. Oncol. 133 (2007) 493–500. doi:10.1007/s00432-007-0193-1. [16] E. Kassi, T.G. Sourlingas, M. Spiliotaki, Z. Papoutsi, H. Pratsinis, N. Aligiannis, et al., Ursolic acid triggers apoptosis and Bcl-2 downregulation in MCF-7 breast cancer cells., Cancer Invest. 27 (2009) 723–733. doi:10.1080/07357900802672712. [17] Y.X. Zhang, C.Z. Kong, L.H. Wang, J.Y. Li, X.K. Liu, B. Xu, et al., Ursolic acid overcomes Bcl-2-mediated resistance to apoptosis in prostate cancer cells involving activation of JNK-Induced Bcl-2 phosphorylation and degradation, J. Cell. Biochem. 109 (2010) 764–773. doi:10.1002/jcb.22455.

25

ACCEPTED MANUSCRIPT [18] J. Shan, Y. Xuan, S. Zheng, Q. Dong, S. Zhang, Ursolic acid inhibits proliferation and induces apoptosis of HT-29 colon cancer cells by inhibiting the EGFR/MAPK pathway., J. Zhejiang Univ. Sci. B. 10 (2009) 668–674. doi:10.1631/jzus.B0920149.

RI

PT

[19] J.-Z. Shan, Y.-Y. Xuan, S.-Q. Ruan, M. Sun, Proliferation-inhibiting and apoptosisinducing effects of ursolic acid and oleanolic acid on multi-drug resistance cancer cells in vitro., Chin. J. Integr. Med. 17 (2011) 607–611. doi:10.1007/s11655-011-0815-y.

SC

[20] X. Wang, F. Zhang, L. Yang, Y. Mei, H. Long, X. Zhang, et al., Ursolic acid inhibits proliferation and induces apoptosis of cancer cells in vitro and in vivo., J. Biomed. Biotechnol. 2011 (2011) 419343. doi:10.1155/2011/419343.

MA

NU

[21] S. Prasad, V.R. Yadav, R. Kannappan, B.B. Aggarwal, Ursolic acid, a pentacyclin triterpene, potentiates trail-induced apoptosis through p53-independent up-regulation of death receptors: Evidence for the role of reactive oxygen species and JNK, J. Biol. Chem. 286 (2011) 5546–5557. doi:10.1074/jbc.M110.183699.

D

[22] S.W. Shin, J.W. Park, Ursolic acid sensitizes prostate cancer cells to TRAIL-mediated apoptosis, Biochim. Biophys. Acta - Mol. Cell Res. 1833 (2013) 723–730. doi:10.1016/j.bbamcr.2012.12.005.

AC CE P

TE

[23] Q.Y. Zheng, P.P. Li, F.S. Jin, C. Yao, G.H. Zhang, T. Zang, et al., Ursolic acid induces ER stress response to activate ASK1-JNK signaling and induce apoptosis in human bladder cancer T24 cells, Cell. Signal. 25 (2013) 206–213. doi:10.1016/j.cellsig.2012.09.012. [24] L. Gai, N. Cai, L. Wang, X. Xu, X. Kong, Ursolic acid induces apoptosis via Akt/NF-κB signaling suppression in T24 human bladder cancer cells., Mol. Med. Rep. 7 (2013) 1673– 7. doi:10.3892/mmr.2013.1364. [25] Q.Y. Zheng, F.S. Jin, C. Yao, T. Zhang, G.H. Zhang, X. Ai, Ursolic acid-induced AMPactivated protein kinase (AMPK) activation contributes to growth inhibition and apoptosis in human bladder cancer T24 cells, Biochem. Biophys. Res. Commun. 419 (2012) 741– 747. doi:10.1016/j.bbrc.2012.02.093. [26] N. Gao, S. Cheng, A. Budhraja, Z. Gao, J. Chen, E.H. Liu, et al., Ursolic acid induces apoptosis in human leukaemia cells and exhibits anti-leukaemic activity in nude mice through the PKB pathway, Br. J. Pharmacol. 165 (2012) 1813–1826. doi:10.1111/j.14765381.2011.01684.x. [27] X. Liang, Ursolic acid inhibits growth and induces apoptosis in gemcitabine-resistant human pancreatic cancer via the JNK and PI3K/Akt/NF-κB pathways, Oncol. Rep. (2012). doi:10.3892/or.2012.1827.

26

ACCEPTED MANUSCRIPT [28] J. Li, W.-J. Guo, Q.-Y. Yang, Effects of ursolic acid and oleanolic acid on human colon carcinoma cell line HCT15., World J. Gastroenterol. 8 (2002) 493–5. http://www.ncbi.nlm.nih.gov/pubmed/12046077 (accessed September 1, 2015).

RI

PT

[29] J. Wang, T. Ren, T. Xi, Ursolic acid induces apoptosis by suppressing the expression of FoxM1 in MCF-7 human breast cancer cells, Med. Oncol. 29 (2012) 10–15. doi:10.1007/s12032-010-9777-8.

SC

[30] Y. Achiwa, K. Hasegawa, Y. Udagawa, Effect of ursolic acid on MAPK in cyclin D1 signaling and RING-type E3 ligase (SCF E3s) in two endometrial cancer cell lines., Nutr. Cancer. 65 (2013) 1026–33. doi:10.1080/01635581.2013.810292.

NU

[31] H. Weng, Z.-J. Tan, Y.-P. Hu, Y.-J. Shu, R.-F. Bao, L. Jiang, et al., Ursolic acid induces cell cycle arrest and apoptosis of gallbladder carcinoma cells, Cancer Cell Int. 14 (2014) 96. doi:10.1186/s12935-014-0096-6.

MA

[32] G. Kumar, H.S. Tuli, S. Mittal, J.K. Shandilya, A. Tiwari, S.S. Sandhu, Isothiocyanates: a class of bioactive metabolites with chemopreventive potential., Tumour Biol. 36 (2015) 4005–16. doi:10.1007/s13277-015-3391-5.

TE

D

[33] A. Jedinák, M. Mučková, D. Košt’álová, T. Maliar, I. Mašterová, Antiprotease and antimetastatic activity of ursolic acid isolated from Salvia officinalis, Zeitschrift Fur Naturforsch. - Sect. C J. Biosci. 61 (2006) 777–782.

AC CE P

[34] H.-C. Huang, C.-Y. Huang, S.-Y. Lin-Shiau, J.-K. Lin, Ursolic acid inhibits IL-1beta or TNF-alpha-induced C6 glioma invasion through suppressing the association ZIP/p62 with PKC-zeta and downregulating the MMP-9 expression., Mol. Carcinog. 48 (2009) 517–31. doi:10.1002/mc.20490. [35] C.-T. Yeh, C.-H. Wu, G.-C. Yen, Ursolic acid, a naturally occurring triterpenoid, suppresses migration and invasion of human breast cancer cells by modulating c-Jun Nterminal kinase, Akt and mammalian target of rapamycin signaling., Mol. Nutr. Food Res. 54 (2010) 1285–1295. doi:10.1002/mnfr.200900414. [36] M.K. Shanmugam, K. a. Manu, T.H. Ong, L. Ramachandran, R. Surana, P. Bist, et al., Inhibition of CXCR4/CXCL12 signaling axis by ursolic acid leads to suppression of metastasis in transgenic adenocarcinoma of mouse prostate model, Int. J. Cancer. 129 (2011) 1552–1563. doi:10.1002/ijc.26120. [37] C.-Y. Huang, C.-Y. Lin, C.-W. Tsai, M.-C. Yin, Inhibition of cell proliferation, invasion and migration by ursolic acid in human lung cancer cell lines., Toxicol. In Vitro. 25 (2011) 1274–1280. doi:10.1016/j.tiv.2011.04.014. [38] L. Xiang, T. Chi, Q. Tang, X. Yang, M. Ou, X. Chen, A pentacyclic triterpene natural product , ursolic acid and its prodrug US597 inhibit targets within cell adhesion pathway and prevent cancer metastasis, (2015). 27

ACCEPTED MANUSCRIPT

PT

[39] H.S. Tuli, S.S. Sandhu, A.K. Sharma, P. Gandhi, Anti-angiogenic activity of the extracted fermentation broth of an entomopathogenic fungus, Cordyceps militaris 3936, Int. J. Pharm. Pharm. Sci. 6 (2014). http://innovareacademics.in/journals/index.php/ijpps/article/view/2021 (accessed September 2, 2015).

RI

[40] H.S. Tuli, D. Kashyap, A.K. Sharma, S.S. Sandhu, Molecular aspects of melatonin (MLT)-mediated therapeutic effects., Life Sci. 135 (2015) 147–157. doi:10.1016/j.lfs.2015.06.004.

NU

SC

[41] C. Cárdenas, A.R. Quesada, M.Á. Medina, Effects of ursolic acid on different steps of the angiogenic process, Biochem. Biophys. Res. Commun. 320 (2004) 402–408. doi:10.1016/j.bbrc.2004.05.183.

MA

[42] M. Kanjoormana, G. Kuttan, Antiangiogenic activity of ursolic acid., Integr. Cancer Ther. 9 (2010) 224–235. doi:10.1177/1534735410367647.

D

[43] C.-C. Lin, C.-Y. Huang, M.-C. Mong, C.-Y. Chan, M.-C. Yin, Antiangiogenic potential of three triterpenic acids in human liver cancer cells., J. Agric. Food Chem. 59 (2011) 755– 62. doi:10.1021/jf103904b.

AC CE P

TE

[44] S. Saraswati, S.S. Agrawal, A. a. Alhaider, Ursolic acid inhibits tumor angiogenesis and induces apoptosis through mitochondrial-dependent pathway in Ehrlich ascites carcinoma tumor, Chem. Biol. Interact. 206 (2013) 153–165. doi:10.1016/j.cbi.2013.09.004. [45] J. Lin, Y. Chen, L. Wei, Z. Hong, T.J. Sferra, J. Peng, Ursolic acid inhibits colorectal cancer angiogenesis through suppression of multiple signaling pathways, Int. J. Oncol. 43 (2013) 1666–1674. doi:10.3892/ijo.2013.2101. [46] H.S. Tuli, S.S. Sandhu, A.K. Sharma, Pharmacological and therapeutic potential of Cordyceps with special reference to Cordycepin, 3 Biotech. 4 (2013) 1–12. doi:10.1007/s13205-013-0121-9. [47] S. Martin-Aragón, B. de las Heras, M.I. Sanchez-Reus, J. Benedi, Pharmacological modification of endogenous antioxidant enzymes by ursolic acid on tetrachloride-induced liver damage in rats and primary cultures of rat hepatocytes., Exp. Toxicol. Pathol. 53 (2001) 199–206. doi:10.1078/0940-2993-00185. [48] R. Saravanan, P. Viswanathan, K.V. Pugalendi, Protective effect of ursolic acid on ethanol-mediated experimental liver damage in rats., Life Sci. 78 (2006) 713–8. doi:10.1016/j.lfs.2005.05.060. [49] J. Lu, Y.-L. Zheng, D.-M. Wu, L. Luo, D.-X. Sun, Q. Shan, Ursolic acid ameliorates cognition deficits and attenuates oxidative damage in the brain of senescent mice induced by D-galactose., Biochem. Pharmacol. 74 (2007) 1078–90. doi:10.1016/j.bcp.2007.07.007. 28

ACCEPTED MANUSCRIPT

PT

[50] S. Ramachandran, Modulation of UVB-induced oxidative stress by ursolic acid in human blood lymphocytes, Asian J. …. (2008). http://www.docsdrive.com/pdfs/academicjournals/ajb/2008/11-18.pdf (accessed September 2, 2015).

RI

[51] S.-J. Tsai, M.-C. Yin, Antioxidative and anti-inflammatory protection of oleanolic acid and ursolic acid in PC12 cells., J. Food Sci. 73 (2008) H174–8. doi:10.1111/j.17503841.2008.00864.x.

SC

[52] A.A. Ramos, C. Pereira-Wilson, A.R. Collins, Protective effects of ursolic acid and luteolin against oxidative DNA damage include enhancement of DNA repair in Caco-2 cells., Mutat. Res. 692 (2010) 6–11. doi:10.1016/j.mrfmmm.2010.07.004.

MA

NU

[53] M. Xiang, J. Wang, Y. Zhang, J. Ling, X. Xu, Attenuation of aortic injury by ursolic acid through RAGE-Nox-NFκB pathway in streptozocin-induced diabetic rats., Arch. Pharm. Res. 35 (2012) 877–86. doi:10.1007/s12272-012-0513-0.

TE

D

[54] S. Martin-Aragón, B. de las Heras, M.I. Sanchez-Reus, J. Benedi, R. Saravanan, P. Viswanathan, et al., Ursolic acid inhibits cigarette smoke extract-induced human bronchial epithelial cell injury and prevents development of lung cancer, Life Sci. 88 (2015) 188– 197. doi:10.1016/j.pbb.2007.07.004.

AC CE P

[55] J.-Q. Ma, J. Ding, L. Zhang, C.-M. Liu, Protective effects of ursolic acid in an experimental model of liver fibrosis through Nrf2/ARE pathway., Clin. Res. Hepatol. Gastroenterol. 39 (2015) 188–97. doi:10.1016/j.clinre.2014.09.007. [56] H.S. Tuli, D. Kashyap, A.K. Sharma, S.S. Sandhu, Molecular aspects of melatonin (MLT)-mediated therapeutic effects, Life Sci. 135 (2015) 147–157. doi:10.1016/j.lfs.2015.06.004. [57] D. Baricevic, S. Sosa, R. Della Loggia, A. Tubaro, B. Simonovska, A. Krasna, et al., Topical anti-inflammatory activity of Salvia officinalis L. leaves: the relevance of ursolic acid, J. Ethnopharmacol. 75 (2001) 125–132. doi:10.1016/S0378-8741(00)00396-2. [58] S.F. Ahmad, B. Khan, S. Bani, K. a. Suri, N.K. Satti, G.N. Qazi, Amelioration of adjuvant-induced arthritis by ursolic acid through altered Th1/Th2 cytokine production, Pharmacol. Res. 53 (2006) 233–240. doi:10.1016/j.phrs.2005.11.005. [59] A. Nataraj, C. Raghavendra Gowda, R. Rajesh, B. Vishwanath, Group IIA Secretory PLA2 Inhibition by Ursolic Acid: A Potent Anti-Inflammatory Molecule, Curr. Top. Med. Chem. 7 (2007) 801–809. doi:10.2174/156802607780487696. [60] K. Takada, T. Nakane, K. Masuda, H. Ishii, Ursolic acid and oleanolic acid, members of pentacyclic triterpenoid acids, suppress TNF-??-induced E-selectin expression by cultured umbilical vein endothelial cells, Phytomedicine. 17 (2010) 1114–1119. doi:10.1016/j.phymed.2010.04.006. 29

ACCEPTED MANUSCRIPT

PT

[61] J. Lu, D.M. Wu, Y.L. Zheng, B. Hu, Z.F. Zhang, Q. Ye, et al., Ursolic acid attenuates Dgalactose-induced inflammatory response in mouse prefrontal cortex through inhibiting AGEs/RAGE/NF-κB pathway activation, Cereb. Cortex. 20 (2010) 2540–2548. doi:10.1093/cercor/bhq002.

RI

[62] Y.-J. Wang, J. Lu, D. Wu, Z. Zheng, Y.-L. Zheng, X. Wang, et al., Ursolic acid attenuates lipopolysaccharide-induced cognitive deficits in mouse brain through suppressing p38/NF-κB mediated inflammatory pathways., Neurobiol. Learn. Mem. 96 (2011) 156– 65. doi:10.1016/j.nlm.2011.03.010.

NU

SC

[63] R. Checker, S.K. Sandur, D. Sharma, R.S. Patwardhan, S. Jayakumar, V. Kohli, et al., Potent anti-inflammatory activity of ursolic acid, a triterpenoid antioxidant, is mediated through suppression of NF-κB, AP-1 and NF-AT, PLoS One. 7 (2012). doi:10.1371/journal.pone.0031318.

MA

[64] J.Q. Ma, J. Ding, Z.H. Xiao, C.M. Liu, Ursolic acid ameliorates carbon tetrachlorideinduced oxidative DNA damage and inflammation in mouse kidney by inhibiting the STAT3 and NF-κB activities, Int. Immunopharmacol. 21 (2014) 389–395. doi:10.1016/j.intimp.2014.05.022.

TE

D

[65] A. Kurek, A.M. Grudniak, M. Szwed, A. Klicka, L. Samluk, K.I. Wolska, et al., Oleanolic acid and ursolic acid affect peptidoglycan metabolism in Listeria monocytogenes., Antonie Van Leeuwenhoek. 97 (2010) 61–8. doi:10.1007/s10482-009-9388-6.

AC CE P

[66] M. Broniatowski, M. Flasiński, K. Hąc-Wydro, Antagonistic effects of α-tocopherol and ursolic acid on model bacterial membranes., Biochim. Biophys. Acta. 1848 (2015) 2154– 62. doi:10.1016/j.bbamem.2015.05.009. [67] W. Dorota, K. Marta, T.G. Dorota, Effect of asiatic and ursolic acids on morphology, hydrophobicity, and adhesion of UPECs to uroepithelial cells, Folia Microbiol. (Praha). 58 (2013) 245–252. doi:10.1007/s12223-012-0205-7. [68] Y. Huang, D. Nikolic, S. Pendland, B.J. Doyle, T.D. Locklear, G.B. Mahady, Effects of cranberry extracts and ursolic acid derivatives on P-fimbriated Escherichia coli, COX-2 activity, pro-inflammatory cytokine release and the NF-κβ transcriptional response in vitro, Pharm. Biol. (2009). http://www.tandfonline.com/doi/abs/10.1080/13880200802397996#.Vebb_H1A6f4 (accessed September 2, 2015). [69] P.G.G. Nascimento, T.L.G. Lemos, A.M.C. Bizerra, Â.M.C. Arriaga, D. a Ferreira, G.M.P. Santiago, et al., Antibacterial and Antioxidant Activities of Ursolic Acid, (2014) 1317–1327. doi:10.3390/molecules19011317. [70] G.R. Dwivedi, A. Maurya, D.K. Yadav, F. Khan, M.P. Darokar, S.K. Srivastava, Drug Resistance Reversal Potential of Ursolic Acid Derivatives against Nalidixic Acid- and 30

ACCEPTED MANUSCRIPT Multidrug-resistant Escherichia coli, Chem. Biol. Drug Des. 226015 (2015) n/a–n/a. doi:10.1111/cbdd.12491.

PT

[71] K.I. Wolska, A.M. Grudniak, B. Fiecek, A. Kraczkiewicz-Dowjat, A. Kurek, Antibacterial activity of oleanolic and ursolic acids and their derivatives, Cent. Eur. J. Biol. 5 (2010) 543–553. doi:10.2478/s11535-010-0045-x.

SC

RI

[72] B. Micota, B. Sadowska, A. Podsędek, M. Redzynia, B. Różalska, Leonurus cardiaca L . herb — a derived extract and an ursolic acid as the factors affecting the adhesion capacity of Staphylococcus aureus in the context of infective endocarditis *, 61 (2014) 385–388.

NU

[73] N. Qin, X. Tan, Y. Jiao, L. Liu, W. Zhao, S. Yang, et al., RNA-Seq-based transcriptome analysis of methicillin-resistant Staphylococcus aureus biofilm inhibition by ursolic acid and resveratrol., Sci. Rep. 4 (2014) 5467. doi:10.1038/srep05467.

MA

[74] M.J. Kim, C.S. Kim, J. Park, Y.K. Lim, S. Park, S. Ahn, Antimicrobial Effects of Ursolic Acid against Mutans Streptococci Isolated from Koreans, Int. J. 36 (2011) 7–11.

TE

D

[75] S. Kim, M. Song, B.-D. Roh, S.-H. Park, J.-W. Park, Inhibition of Streptococcus mutans biofilm formation on composite resins containing ursolic acid., Restor. Dent. Endod. 38 (2013) 65–72. doi:10.5395/rde.2013.38.2.65.

AC CE P

[76] K. Horiuchi, S. Shiota, T. Hatano, T. Yoshida, T. Kuroda, T. Tsuchiya, Antimicrobial activity of oleanolic acid from Salvia officinalis and related compounds on vancomycinresistant enterococci (VRE)., Biol. Pharm. Bull. 30 (2007) 1147–1149. doi:10.1248/bpb.30.1147. [77] M.A. Jyoti, T. Zerin, T.-H. Kim, T.-S. Hwang, W.S. Jang, K.-W. Nam, et al., In vitro effect of ursolic acid on the inhibition of Mycobacterium tuberculosis and its cell wall mycolic acid., Pulm. Pharmacol. Ther. 33 (2015) 17–24. doi:10.1016/j.pupt.2015.05.005. [78] D. Martins, L. Carrion, D. Ramos, K. Salomé, P. da Silva, B. Andersson, et al., Antituberculosis activity of oleanolic and ursolic acid isolated from the dichloromethane extract of leaves from Duroia macrophylla, BMC Proc. 8 (2014) P3. doi:10.1186/17536561-8-S4-P3. [79] A. Jiménez-Arellanes, J. Luna-Herrera, J. Cornejo-Garrido, S. López-García, M.E. CastroMussot, M. Meckes-Fischer, et al., Ursolic and oleanolic acids as antimicrobial and immunomodulatory compounds for tuberculosis treatment., BMC Complement. Altern. Med. 13 (2013) 258. doi:10.1186/1472-6882-13-258. [80] J. Zhao, J. Chen, T. Liu, J. Fang, J. Wan, J. Zhao, W. Li, J. Liu, X. Zhao, S. Chen, Antiviral effects of urosolic acid on guinea pig cytomegalovirus in vitro., J. Huazhong. Univ. Sci. Technolog. Med. Sci. 32 (2012) 883–887. doi:10.1007/s11596-012-1052-0.

31

ACCEPTED MANUSCRIPT [81] O.B. Kazakova, G. V. Giniyatullina, E.Y. Yamansarov, G. a. Tolstikov, Betulin and ursolic acid synthetic derivatives as inhibitors of Papilloma virus, Bioorganic Med. Chem. Lett. 20 (2010) 4088–4090. doi:10.1016/j.bmcl.2010.05.083.

RI

PT

[82] G. Song, X. Shen, S. Li, Y. Li, Y. Liu, Y. Zheng, et al., Structure–activity relationships of 3-O-β-chacotriosyl ursolic acid derivatives as novel H5N1 entry inhibitors, Eur. J. Med. Chem. 93 (2015) 431–442. doi:10.1016/j.ejmech.2015.02.029.

SC

[83] L. Kong, S. Li, Q. Liao, Y. Zhang, R. Sun, X. Zhu, et al., Oleanolic acid and ursolic acid: Novel hepatitis C virus antivirals that inhibit NS5B activity, Antiviral Res. 98 (2013) 44– 53. doi:10.1016/j.antiviral.2013.02.003.

NU

[84] Y. Kashiwada, T. Nagao, A. Hashimoto, Y. Ikeshiro, H. Okabe, L.M. Cosentino, et al., Anti-AIDS Agents 38. Anti-HIV Activity of 3- O -Acyl Ursolic Acid Derivatives 1, J. Nat. Prod. 63 (2000) 1619–1622. doi:10.1021/np990633v.

MA

[85] L. Quéré, T. Wenger, H.J. Schramm, Triterpenes as potential dimerization inhibitors of HIV-1 protease., Biochem. Biophys. Res. Commun. 227 (1996) 484–8. doi:10.1006/bbrc.1996.1533.

TE

D

[86] L.-C. Chiang, L.-T. Ng, P.-W. Cheng, W. Chiang, C.-C. Lin, Antiviral activities of extracts and selected pure constituents of Ocimum basilicum., Clin. Exp. Pharmacol. Physiol. 32 (2005) 811–6. doi:10.1111/j.1440-1681.2005.04270.x.

AC CE P

[87] P. Saini, P. Gayen, D. Kumar, A. Nayak, N. Mukherjee, S. Mukherjee, et al., Antifilarial effect of ursolic acid from Nyctanthes arbortristis: Molecular and biochemical evidences, Parasitol. Int. 63 (2014) 717–728. doi:10.1016/j.parint.2014.06.008. [88] D. Da Silva Ferreira, V.R. Esperandim, M.P.A. Toldo, C.C. Kuehn, J.C. Do Prado Júnior, W.R. Cunha, et al., In vivo activity of ursolic and oleanolic acids during the acute phase of Trypanosoma cruzi infection, Exp. Parasitol. 134 (2013) 455–459. doi:10.1016/j.exppara.2013.04.005. [89] J. Eloy, J. Saraiva, S. Albuquerque, J.M. Marchetti, Solid Dispersion of Ursolic Acid in Gelucire 50/13: a Strategy to Enhance Drug Release and Trypanocidal Activity, AAPS PharmSciTech. 13 (2012) 1436–1445. doi:10.1208/s12249-012-9868-2. [90] C. Van Baren, I. Anao, P.D.L. Lira, S. Debenedetti, P. Houghton, S. Croft, et al., Triterpenic acids and flavonoids from Satureja parvifolia, evaluation of their antiprotozoal activity, Zeitschrift Fur Naturforsch. - Sect. C J. Biosci. 61 (2006) 189–192. [91] T.S. Jeong, E.I. Hwang, H.B. Lee, E.S. Lee, Y.K. Kim, B.S. Min, et al., Chitin synthase II inhibitory activity of ursolic acid, isolated from Crataegus pinnatifida., Planta Med. 65 (1999) 261–3. doi:10.1055/s-2006-960474.

32

ACCEPTED MANUSCRIPT [92] K. Wilkinson, J.D. Boyd, M. Glicksman, K.J. Moore, J. El Khoury, A high content drug screen identifies ursolic acid as an inhibitor of amyloid  ?? protein interactions with its receptor CD36, J. Biol. Chem. 286 (2011) 34914–34922. doi:10.1074/jbc.M111.232116.

RI

PT

[93] D.G. MacHado, V.B. Neis, G.O. Balen, a. Colla, M.P. Cunha, J.B. Dalmarco, et al., Antidepressant-like effect of ursolic acid isolated from Rosmarinus officinalis L. in mice: Evidence for the involvement of the dopaminergic system, Pharmacol. Biochem. Behav. 103 (2012) 204–211. doi:10.1016/j.pbb.2012.08.016.

SC

[94] A.R.S. Colla, J.M. Rosa, M.P. Cunha, A.L.S. Rodrigues, Anxiolytic-like effects of ursolic acid in mice, Eur. J. Pharmacol. 758 (2015) 171–176. doi:10.1016/j.ejphar.2015.03.077.

MA

NU

[95] A.R.S. Colla, Á. Oliveira, F.L. Pazini, J.M. Rosa, L.M. Manosso, M.P. Cunha, et al., Serotonergic and noradrenergic systems are implicated in the antidepressant-like effect of ursolic acid in mice, Pharmacol. Biochem. Behav. 124 (2014) 108–116. doi:10.1016/j.pbb.2014.05.015.

D

[96] S.M. Jang, M.J. Kim, M.S. Choi, E.Y. Kwon, M.K. Lee, Inhibitory effects of ursolic acid on hepatic polyol pathway and glucose production in streptozotocin-induced diabetic mice, Metabolism. 59 (2010) 512–519. doi:10.1016/j.metabol.2009.07.040.

AC CE P

TE

[97] W. Zhang, D. Hong, Y. Zhou, Y. Zhang, Q. Shen, J.-Y. Li, et al., Ursolic acid and its derivative inhibit protein tyrosine phosphatase 1B, enhancing insulin receptor phosphorylation and stimulating glucose uptake., Biochim. Biophys. Acta. 1760 (2006) 1505–1512. doi:10.1016/j.bbagen.2006.05.009. [98] I. Kazmi, M. Rahman, M. Afzal, G. Gupta, S. Saleem, O. Afzal, et al., Anti-diabetic potential of ursolic acid stearoyl glucoside: A new triterpenic gycosidic ester from Lantana camara, Fitoterapia. 83 (2012) 142–146. doi:10.1016/j.fitote.2011.10.004. [99] M.-Y. Qi, J.-J. Yang, B. Zhou, D.-Y. Pan, X. Sun, [Study on the protective effect of ursolic acid on alloxan-induced diabetic renal injury and its underlying mechanisms]., Zhongguo Ying Yong Sheng Li Xue Za Zhi. 30 (2014) 445–8. http://europepmc.org/abstract/med/25571638 (accessed September 2, 2015). [100] NISCAIR ONLINE PERIODICALS REPOSITORY (NOPR)  : Effects of ursolic acid on glucose metabolism, the polyol pathway and dyslipidemia in non-obese type 2 diabetic mice, (n.d.). http://nopr.niscair.res.in/handle/123456789/29041 (accessed September 2, 2015). [101] T. Radhiga, C. Rajamanickam, S. Senthil, K.V. Pugalendi, Effect of ursolic acid on cardiac marker enzymes, lipid profile and macroscopic enzyme mapping assay in isoproterenol-induced myocardial ischemic rats, Food Chem. Toxicol. 50 (2012) 3971– 3977. doi:10.1016/j.fct.2012.07.067.

33

ACCEPTED MANUSCRIPT [102] Y. Liu, W. Tian, X. Ma, W. Ding, Evaluation of inhibition of fatty acid synthase by ursolic acid: Positive cooperation mechanism, Biochem. Biophys. Res. Commun. 392 (2010) 386–390. doi:10.1016/j.bbrc.2010.01.031.

RI

PT

[103] I. Kazmi, M. Afzal, S. Rahman, M. Iqbal, F. Imam, F. Anwar, Antiobesity potential of ursolic acid stearoyl glucoside by inhibiting pancreatic lipase., Eur. J. Pharmacol. 709 (2013) 28–36. doi:10.1016/j.ejphar.2013.02.032.

SC

[104] A. Sundaresan, T. Radhiga, K.V. Pugalendi, Effect of ursolic acid and Rosiglitazone combination on hepatic lipid accumulation in high fat diet-fed C57BL/6J mice, Eur. J. Pharmacol. 741 (2014) 297–303. doi:10.1016/j.ejphar.2014.07.032.

NU

[105] Y.L. Wang, Z.J. Wang, H.L. Shen, M. Yin, K.X. Tang, Effects of artesunate and ursolic acid on hyperlipidemia and its complications in rabbit, Eur. J. Pharm. Sci. 50 (2013) 366– 371. doi:10.1016/j.ejps.2013.08.003.

MA

[106] J. Kim, D.S. Jang, H. Kim, J.S. Kim, Anti-lipase and lipolytic activities of ursolic acid isolated from the roots of Actinidia arguta, Arch. Pharm. Res. 32 (2009) 983–987. doi:10.1007/s12272-009-1702-3.

TE

D

[107] L.I. Somova, F.O. Shode, M. Mipando, Cardiotonic and antidysrhythmic effects of oleanolic and ursolic acids, methyl maslinate and uvaol., Phytomedicine. 11 (2004) 121– 129. doi:10.1078/0944-7113-00329.

AC CE P

[108] H.Y. Kim, H.R. Choi, Y.J. Lee, H.Z. Cui, S.N. Jin, K.W. Cho, et al., Accentuation of ursolic acid on muscarinic receptor-induced ANP secretion in beating rabbit atria, Life Sci. 94 (2014) 145–150. doi:10.1016/j.lfs.2013.12.001. [109] X. Dong, S. Liu, L. Zhang, S. Yu, L. Huo, M. Qile, et al., Downregulation of miR-21 is Involved in Direct Actions of Ursolic Acid on the Heart: Implications for Cardiac Fibrosis and Hypertrophy., Cardiovasc. Ther. 33 (2015) 161–7. doi:10.1111/1755-5922.12125. [110] S.L. Ullevig, Q. Zhao, D. Zamora, R. Asmis, Ursolic acid protects diabetic mice against monocyte dysfunction and accelerated atherosclerosis, Atherosclerosis. 219 (2011) 409– 416. doi:10.1016/j.atherosclerosis.2011.06.013. [111] L.R. Tannock, Ursolic acid effect on atherosclerosis: Apples and apples, or apples and oranges?, Atherosclerosis. 219 (2011) 397–398. doi:10.1016/j.atherosclerosis.2011.09.029. [112] X. Wang, K. Ikejima, K. Kon, K. Arai, T. Aoyama, K. Okumura, et al., Ursolic acid ameliorates hepatic fibrosis in the rat by specific induction of apoptosis in hepatic stellate cells, J. Hepatol. 55 (2011) 379–387. doi:10.1016/j.jhep.2010.10.040. [113] J. Chen, H.S. Wong, H.Y. Leung, P.K. Leong, W.M. Chan, N. Chen, et al., An ursolic acid-enriched extract of Cynomorium songaricum protects against carbon tetrachloride 34

ACCEPTED MANUSCRIPT hepatotoxicity and gentamicin nephrotoxicity in rats possibly through a mitochondrial pathway: A comparison with ursolic acid, J. Funct. Foods. 7 (2014) 330–341. doi:10.1016/j.jff.2014.01.027.

RI

PT

[114] T. Sultana, M.A. Rashid, M.A. Ali, S.F. Mahmood, Hepatoprotective and Antibacterial Activity of Ursolic Acid Extracted from Hedyotis corymbosa L., Bangladesh J. Sci. Ind. Res. 45 (2010) 27–34. doi:10.3329/bjsir.v45i1.5174.

SC

[115] J.-S. Li, W.-J. Wang, Y. Sun, Y.-H. Zhang, L. Zheng, Ursolic acid inhibits the development of nonalcoholic fatty liver disease by attenuating endoplasmic reticulum stress., Food Funct. 6 (2015) 1643–51. doi:10.1039/c5fo00083a.

NU

[116] Y.-R. Jin, J.-L. Jin, C.-H. Li, X.-X. Piao, N.-G. Jin, Ursolic acid enhances mouse liver regeneration after partial hepatectomy., Pharm. Biol. 50 (2012) 523–8. doi:10.3109/13880209.2011.611143.

MA

[117] F. Meng, H. Ning, Z. Sun, F. Huang, Y. Li, X. Chu, et al., Ursolic acid protects hepatocytes against lipotoxicity through activating autophagy via an AMPK pathway, J. Funct. Foods. 17 (2015) 172–182. doi:10.1016/j.jff.2015.05.029.

TE

D

[118] J.-Q. Ma, J. Ding, L. Zhang, C.-M. Liu, Ursolic acid protects mouse liver against CCl4induced oxidative stress and inflammation by the MAPK/NF-κB pathway., Environ. Toxicol. Pharmacol. 37 (2014) 975–83. doi:10.1016/j.etap.2014.03.011.

AC CE P

[119] P.G. Pai, S. Chamari Nawarathna, A. Kulkarni, U. Habeeba, S. Reddy C., S. Teerthanath, et al., Nephroprotective Effect of Ursolic Acid in a Murine Model of Gentamicin-Induced Renal Damage, ISRN Pharmacol. 2012 (2012) 1–6. doi:10.5402/2012/410902. [120] C. Ling, L. Jinping, L. Xia, Y. Renyong, Ursolic Acid Provides Kidney Protection in Diabetic Rats, Curr. Ther. Res. - Clin. Exp. 75 (2013) 59–63. doi:10.1016/j.curtheres.2013.07.001. [121] S.J. Jeon, H.J. Park, Q. Gao, I.J. Dela Pena, S.J. Park, H.E. Lee, et al., Ursolic acid enhances pentobarbital-induced sleeping behaviors via GABAergic neurotransmission in mice, Eur. J. Pharmacol. 762 (2015) 443–448. doi:10.1016/j.ejphar.2015.06.037. [122] T. Zhang, J. Su, K. Wang, T. Zhu, X. Li, Ursolic acid reduces oxidative stress to alleviate early brain injury following experimental subarachnoid hemorrhage, Neurosci. Lett. 579 (2014) 12–17. doi:10.1016/j.neulet.2014.07.005. [123] Y. Park, H. Jang, Y. Paik, Et Al., Prolyl Endopeptidase Inhibitory Activity of Ursolic and Oleanolic Acids from Corni Fructus, Agric. Chem. Biotechnol. 48 (2005) 207. http://www.ksabc.or.kr/admin/contribute/journal_jabc/epaper/2005_48_4_207.pdf.

35

ACCEPTED MANUSCRIPT [124] B. Liu, Y. Liu, G. Yang, Z. Xu, J. Chen, Ursolic acid induces neural regeneration after sciatic nerve injury., Neural Regen. Res. 8 (2013) 2510–9. doi:10.3969/j.issn.16735374.2013.27.002.

RI

PT

[125] T. Zhang, J. Su, B. Guo, T. Zhu, K. Wang, X. Li, Ursolic acid alleviates early brain injury after experimental subarachnoid hemorrhage by suppressing TLR4-mediated inflammatory pathway, Int. Immunopharmacol. 23 (2014) 585–591. doi:10.1016/j.intimp.2014.10.009.

SC

[126] N. Prissadova, P. Bozov, K. Marinkov, H. Badakov, A. Kristev, Effects of ursolic acid on contractile activity of gastric smooth muscles., Nat. Prod. Commun. 10 (2015) 565–6. http://www.ncbi.nlm.nih.gov/pubmed/25973477 (accessed September 2, 2015).

MA

NU

[127] S.-H. Kim, J.-H. Hong, Y.-C. Lee, Ursolic acid, a potential PPARγ agonist, suppresses ovalbumin-induced airway inflammation and Penh by down-regulating IL-5, IL-13, and IL-17 in a mouse model of allergic asthma., Eur. J. Pharmacol. 701 (2013) 131–43. doi:10.1016/j.ejphar.2012.11.033.

TE

D

[128] H.-I. Kim, F.-S. Quan, J.-E. Kim, N.-R. Lee, H.J. Kim, S.J. Jo, et al., Inhibition of estrogen signaling through depletion of estrogen receptor alpha by ursolic acid and betulinic acid from Prunella vulgaris var. lilacina., Biochem. Biophys. Res. Commun. 451 (2014) 282–7. doi:10.1016/j.bbrc.2014.07.115.

AC CE P

[129] Y.-Y. Gong, Y.-Y. Liu, S. Yu, X.-N. Zhu, X.-P. Cao, H.-P. Xiao, Ursolic acid suppresses growth and adrenocorticotrophic hormone secretion in AtT20 cells as a potential agent targeting adrenocorticotrophic hormone-producing pituitary adenoma., Mol. Med. Rep. 9 (2014) 2533–9. doi:10.3892/mmr.2014.2078. [130] Z. Hu, Z. Gu, M. Sun, K. Zhang, P. Gao, Q. Yang, et al., Ursolic acid improves survival and attenuates lung injury in septic rats induced by cecal ligation and puncture, J. Surg. Res. 194 (2015) 528–536. doi:10.1016/j.jss.2014.10.027. [131] H. Xu, M. Zhang, X.-L. Li, H. Li, L.-T. Yue, X.-X. Zhang, et al., Low and high doses of ursolic acid ameliorate experimental autoimmune myasthenia gravis through different pathways, J. Neuroimmunol. 281 (2015) 61–67. doi:10.1016/j.jneuroim.2015.02.010. [132] S.-Y. Kang, S.-Y. Yoon, D.-H. Roh, M.-J. Jeon, H.-S. Seo, D.-K. Uh, et al., The antiarthritic effect of ursolic acid on zymosan-induced acute inflammation and adjuvantinduced chronic arthritis models., J. Pharm. Pharmacol. 60 (2008) 1347–1354. doi:10.1211/jpp/60.10.0011. [133] C. Jiang, F. Xiao, X. Gu, Z. Zhai, X. Liu, W. Wang, et al., Inhibitory effects of ursolic acid on osteoclastogenesis and titanium particle-induced osteolysis are mediated primarily via suppression of NF-κB signaling., Biochimie. 111 (2015) 107–18. doi:10.1016/j.biochi.2015.02.002. 36

ACCEPTED MANUSCRIPT [134] H.-J. Fu, Y. Zhao, Y.-R. Zhou, B.-H. Bao, Y. Du, J.-X. Li, Ursolic acid derivatives as bone anabolic agents targeted to tryptophan hydroxylase 1 (Tph-1), Eur. J. Pharm. Sci. 76 (2015) 33–47. doi:10.1016/j.ejps.2015.04.021.

RI

PT

[135] N. Bakhtiari, S. Hosseinkhani, A. Tashakor, R. Hemmati, Ursolic acid ameliorates agingmetabolic phenotype through promoting of skeletal muscle rejuvenation., Med. Hypotheses. 85 (2015) 1–6. doi:10.1016/j.mehy.2015.02.014.

SC

[136] T.-L. Hwang, H.-I. Shen, F.-C. Liu, H.-I. Tsai, Y.-C. Wu, F.-R. Chang, et al., Ursolic acid inhibits superoxide production in activated neutrophils and attenuates trauma-hemorrhage shock-induced organ injury in rats., PLoS One. 9 (2014) e111365. doi:10.1371/journal.pone.0111365.

MA

NU

[137] D.M. Both, K. Goodtzova, D.B. Yarosh, D. a Brown, Liposome-encapsulated ursolic acid increases ceramides and collagen in human skin cells., Arch. Dermatol. Res. 293 (2002) 569–575. doi:10.1007/s00403-001-0272-0.

TE

D

[138] S.W. Lim, S.P. Hong, S.W. Jeong, B. Kim, H. Bak, H.C. Ryoo, et al., Simultaneous effect of ursolic acid and oleanolic acid on epidermal permeability barrier function and epidermal keratinocyte differentiation via peroxisome proliferator-activated receptor-??, J. Dermatol. 34 (2007) 625–634. doi:10.1111/j.1346-8138.2007.00344.x.

AC CE P

[139] X. Chu, X. He, Z. Shi, C. Li, F. Guo, S. Li, et al., Ursolic acid increases energy expenditure through enhancing free fatty acid uptake and β-oxidation via an UCP3/AMPK-dependent pathway in skeletal muscle., Mol. Nutr. Food Res. 59 (2015) 1491–503. doi:10.1002/mnfr.201400670. [140] R. Ogasawara, K. Sato, K. Higashida, K. Nakazato, S. Fujita, Ursolic acid stimulates mTORC1 signaling after resistance exercise in rat skeletal muscle., Am. J. Physiol. Endocrinol. Metab. 305 (2013) E760–5. doi:10.1152/ajpendo.00302.2013. [141] F. Aparecida Resende, C.A.M. de Andrade Barcala, M.C. da Silva Faria, F.H. Kato, W.R. Cunha, D.C. Tavares, Antimutagenicity of ursolic acid and oleanolic acid against doxorubicin-induced clastogenesis in Balb/c mice, Life Sci. 79 (2006) 1268–1273. doi:10.1016/j.lfs.2006.03.038. [142] R. Lin, L. Gong, L. Fan, Z. Zhao, S. Yang, Role of ursolic acid chalcone , a synthetic analogue of ursolic acid , in inhibiting the properties of in liver stem cells, 8 (2015) 1427– 1434. [143] J. Chun, C. Lee, S.W. Hwang, J.P. Im, J.S. Kim, Ursolic acid inhibits nuclear factor-κB signaling in intestinal epithelial cells and macrophages, and attenuates experimental colitis in mice., Life Sci. 110 (2014) 23–34. doi:10.1016/j.lfs.2014.06.018. [144] Y. Yie, S. Zhao, Q. Tang, F. Zheng, J. Wu, L. Yang, et al., Ursolic acid inhibited growth of hepatocellular carcinoma HepG2 cells through AMPKα-mediated reduction of DNA 37

ACCEPTED MANUSCRIPT methyltransferase 1, Mol. Cell. Biochem. 402 (2015) 63–74. doi:10.1007/s11010-0142314-x.

PT

[145] A. Batool, K. Shahid, M. Muddasir, Research Article Synthesis of Aniline Derivative of Ursolic Acid, its Metal Complexes, Characterization and Bioassay, 30 (2015) 25–32.

RI

[146] E.H. Lee, S. a. Popov, J.Y. Lee, a. V. Shpatov, T.P. Kukina, S.W. Kang, et al., Inhibitory effect of ursolic acid derivatives on recombinant human aldose reductase, Russ. J. Bioorganic Chem. 37 (2011) 569–577. doi:10.1134/S1068162011050050.

NU

SC

[147] S.-X. Hua, R.-Z. Huang, M.-Y. Ye, Y.-M. Pan, G.-Y. Yao, Y. Zhang, et al., Design, synthesis and in vitro evaluation of novel ursolic acid derivatives as potential anticancer agents., Eur. J. Med. Chem. 95 (2015) 435–452. doi:10.1016/j.ejmech.2015.03.051.

MA

[148] J.-W. Shao, Y.-C. Dai, J.-P. Xue, J.-C. Wang, F.-P. Lin, Y.-H. Guo, In vitro and in vivo anticancer activity evaluation of ursolic acid derivatives., Eur. J. Med. Chem. 46 (2011) 2652–2661. doi:10.1016/j.ejmech.2011.03.050.

TE

D

[149] C.-M. Ma, S.-Q. Cai, J.-R. Cui, R.-Q. Wang, P.-F. Tu, M. Hattori, et al., The cytotoxic activity of ursolic acid derivatives., Eur. J. Med. Chem. 40 (2005) 582–589. doi:10.1016/j.ejmech.2005.01.001.

AC CE P

[150] Y.Q. Meng, D. Liu, L.L. Cai, H. Chen, B. Cao, Y.Z. Wang, The synthesis of ursolic acid derivatives with cytotoxic activity and the investigation of their preliminary mechanism of action, Bioorganic Med. Chem. 17 (2009) 848–854. doi:10.1016/j.bmc.2008.11.036. [151] H. Dong, X. Yang, J. Xie, L. Xiang, Y. Li, M. Ou, et al., UP12, a novel ursolic acid derivative with potential for targeting multiple signaling pathways in hepatocellular carcinoma., Biochem. Pharmacol. 93 (2015) 151–162. doi:10.1016/j.bcp.2014.11.014. [152] K. Mazumder, E.R.O. Siwu, S. Nozaki, Y. Watanabe, K. Tanaka, K. Fukase, Ursolic acid derivatives from Bangladeshi medicinal plant, Saurauja roxburghii: Isolation and cytotoxic activity against A431 and C6 glioma cell lines, Phytochem. Lett. 4 (2011) 287– 291. doi:10.1016/j.phytol.2011.04.019. [153] H.-Y. Tu, A.-M. Huang, B.-L. Wei, K.-H. Gan, T.-C. Hour, S.-C. Yang, et al., Ursolic acid derivatives induce cell cycle arrest and apoptosis in NTUB1 cells associated with reactive oxygen species., Bioorg. Med. Chem. 17 (2009) 7265–7274. doi:10.1016/j.bmc.2009.08.046. [154] J. Wang, Z. Jiang, L. Xiang, Y. Li, M. Ou, X. Yang, et al., Synergism of ursolic acid derivative US597 with 2-deoxy-D-glucose to preferentially induce tumor cell death by dual-targeting of apoptosis and glycolysis., Sci. Rep. 4 (2014) 5006. doi:10.1038/srep05006.

38

ACCEPTED MANUSCRIPT

PT

[155] J.H. Kim, Y.H. Kim, G.Y. Song, D.E. Kim, Y.J. Jeong, K.H. Liu, et al., Ursolic acid and its natural derivative corosolic acid suppress the proliferation of APC-mutated colon cancer cells through promotion of β-catenin degradation, Food Chem. Toxicol. 67 (2014) 87–95. doi:10.1016/j.fct.2014.02.019.

RI

[156] A.T. Nelson, A.M. Camelio, K.R. Claussen, J. Cho, L. Tremmel, J. DiGiovanni, et al., Synthesis of oxygenated oleanolic and ursolic acid derivatives with anti-inflammatory properties, Bioorg. Med. Chem. Lett. (2015) 19–21. doi:10.1016/j.bmcl.2015.07.029.

SC

[157] Y. Meng, Y. Song, Z. Yan, Y. Xia, Synthesis and in vitro cytotoxicity of novel ursolic acid derivatives, Molecules. 15 (2010) 4033–4040. doi:10.3390/molecules15064033.

NU

[158] L.C. and Y.Z. Yanqiu Meng, Synthesis of Ursolic Acid Derivatives and Research on Their Cytotoxic Activities, Life Sci. J. 9 (2012).

MA

[159] A.S. Leal, R. Wang, J. a R. Salvador, Y. Jing, Semisynthetic Ursolic Acid Fluorolactone Derivatives Inhibit Growth with Induction of p21waf1 and Induce Apoptosis with Upregulation of NOXA and Downregulation of c-FLIP in Cancer Cells, ChemMedChem. 7 (2012) 1635–1646. doi:10.1002/cmdc.201200282.

TE

D

[160] H.J. You, C.Y. Cho, J. Y. Kim, S.J. Park, K.S. Hahm, H.G. Jeong, Ursolic acid enhances nitric oxide and tumor necrosis factor-alpha production via nuclear factor-kappaB activation in the resting macrophages, FEBS Lett. 509 (2001) 156–160.

AC CE P

[161] Y. Ikenda, A. Murakami, H. Ohigashi, Ursolic acid promotes the release of macrophage migration inhibitory factor via ERK2 activation in resting mouse macrophages, Biochem. Pharmacol. 67 (2005) 1497–1505. [162] Y. Ikeda, A. Murakami, T. Nishizawa, H. Ohigashi, Ursolic acid enhances cyclooxygenases and tumor necrosis factoralpha expression in mouse skin.Biosci, Biotechnol. Biochem. 70 (2006) 1033–1037. [163] Y. Ikeda, A. Murakami, Y. Fujimura, H. Tachibana,et al, Aggregated ursolic acid, a natural triterpenoid, induces IL-1beta release from murine peritoneal macrophages: role of CD36, J. Immunol. 178 (2007) 4854–4864. [164] L.I.U. Jing-Bo, Acute and Genetic Toxicity of Ursolic Acid Extract from Ledum pulastre L.[J], Food Sci. 30 (2009) 250-252. [165] X.H. Wang, S.Y. Zhou, Z.Z. Qian, H.L. Zhang, L.H. Qiu, Z. Song, J. Zhao, P. Wang, X.S. Hao, H.Q. Wang, Evaluation of toxicity and single-dose pharmacokinetics of intravenous ursolic acid liposomes in healthy adult volunteers and patients with advanced solid tumors, Expert Opin Drug Metab Toxicol. 9 (2013) 117-25. [166] X. Yang, Y. Li, W. Jing, M. Ou, Y. Chen,Y. Xu, Q. Wu, Q. Zheng, F. Wu, L. Wang, W. Zou, Y.J. Zhang, J. Shao, Synthesis and Biological Evaluation of Novel Ursolic acid 39

ACCEPTED MANUSCRIPT Derivatives as Potential Anticancer Prodrugs, Chem Biol Drug Des. (2015) doi: 10.1111/cbdd.12608.

PT

[167] J. Cho, O. Rho, J. Junco, S. Carbajal, D. Siegel, T. J. Slaga, J. DiGiovanni, Effect of combined treatment with ursolic acid and resveratrol on skin tumor promotion by 12-otetradecanoylphorbol-13-acetate, Cancer Prev. Res. (Phila). 8 (2015) 817-25.

SC

RI

[168] Y. Zou, Y. Lee, J. Huh, J.-W. Park, Synergistic effect of xylitol and ursolic acid combination on oral biofilms., Restor. Dent. Endod. 39 (2014) 288–95. doi:10.5395/rde.2014.39.4.288.

NU

[169] J.J. Junco, A. Mancha-Ramirez, G. Malik, S.-J. Wei, D.J. Kim, H. Liang, et al., Ursolic acid and resveratrol synergize with chloroquine to reduce melanoma cell viability., Melanoma Res. 25 (2015) 103–12. doi:10.1097/CMR.0000000000000137.

MA

[170] D. Wojnicz, D. Tichaczek-goska, M. Kicia, Pentacyclic triterpenes combined with ciprofloxacin help to eradicate the biofilm formed in vitro by Escherichia coli, (2015) 343–353.

TE

D

[171] J. Cho, O. Rho, J. Junco, S. Carbajal, D. Siegel, T.J. Slaga, et al., Effect of Combined Treatment with Ursolic Acid and Resveratrol on Skin Tumor Promotion by 12-OTetradecanoylphorbol-13-Acetate., Cancer Prev. Res. (Phila). 8 (2015) 817–25. doi:10.1158/1940-6207.CAPR-15-0098.

AC CE P

[172] R.-X. Zhu, L. Zhao, Y.-K. Zhang, P. Luo, S.-H. Liu, J.-G. Wang, Sequential treatment with ursolic acid chlorophenyl triazole followed by 5-fluorouracil shows synergistic activity in small cell lung cancer cells, Bangladesh J. Pharmacol. 10 (2015) 197–204. doi:10.3329/bjp.v10i1.21641.

40

AC CE P

Graphical abstract

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

41