Next Article in Journal
Physiological Characteristics of Photosynthesis in Yellow-Green, Green and Dark-Green Chinese Kale (Brassica oleracea L. var. alboglabra Musil.) under Varying Light Intensities
Next Article in Special Issue
Secondary Bioactive Metabolites from Plant-Derived Food Byproducts through Ecopharmacognostic Approaches: A Bound Phenolic Case Study
Previous Article in Journal
So Uncommon and so Singular, but Underexplored: An Updated Overview on Ethnobotanical Uses, Biological Properties and Phytoconstituents of Sardinian Endemic Plants
Previous Article in Special Issue
In Vitro Antimicrobial and Antiproliferative Activities of the Root Bark Extract and Isolated Chemical Constituents of Zanthoxylum paracanthum Kokwaro (Rutaceae)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Medicinal Properties of Lilium candidum L. and Its Phytochemicals

1
Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
2
Eastern R&D Center, Kiryat Arba 9010000, Israel
3
Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
4
Research & Development Authority, Barzilai University Medical Center, Ashkelon 7830604, Israel
*
Author to whom correspondence should be addressed.
Plants 2020, 9(8), 959; https://doi.org/10.3390/plants9080959
Submission received: 9 July 2020 / Revised: 26 July 2020 / Accepted: 27 July 2020 / Published: 29 July 2020
(This article belongs to the Special Issue Composition and Biological Activities of Plant Secondary Metabolites)

Abstract

:
Lilium candidum L., known as Madonna, meadow, or white lily, is a bulbous plant from the Liliaceae family, originating in the Middle East. L. candidum has been abundantly used in folk medicine since ancient times to relieve a variety of ailments, including age-related diseases, burns, ulcers, and coughs. The aim of this article is to investigate the anti-inflammatory and anti-diabetic activities of L. candidum extracts and its active phytochemicals. Some active volatile phytochemicals were identified using gas chromatography–mass spectrometry (GC-MS) analysis. Significant (p < 0.001) anti-diabetic properties of the extracts kaempferol, linalool, citronellal, and humulene were demonstrated by an elevation in glucose uptake by adipocytes. The significant (p < 0.01) effect of the plant extracts kaempferol, citronellal, and humulene on the secretion of pro-inflammatory cytokines interleukin 6 (IL-6) and interleukin 8 (IL-8) was demonstrated using enzyme-linked immunosorbent assay. Altogether, L. candidum and its rich collection of phytochemicals hold promising medicinal potential, and further investigations of its therapeutic prospects are encouraged.

1. Introduction

The prevalence of age-related diseases (ARDs) including cardiovascular diseases, cancer, type 2 diabetes, neurodegenerative diseases, and obesity is rapidly increasing worldwide [1]. Recent studies provide strong evidence suggesting an essential role of chronic inflammation in the pathogenesis of the above-mentioned ARDs [2,3,4]. In addition, genotoxic stress, which is a component of a wide variety of pathological conditions, not only causes extensive DNA damage but also activates pathways leading to chronic inflammation (the ERK, JNK, and p38 MAPK pathways) and transcription of pro-inflammatory cytokines (TNF-α, IL-1β, interleukin 6 (IL-6)), chemokines (interleukin 8 (IL-8)), adhesion molecules (VCAM-1, ICAM-1, P-, E-selectin), and other pro-inflammatory enzymes including iNOS and COX-2 [5]. This in turn leads to an elevated pro-inflammatory status that is likely to set the stage for increased vulnerability to many ARDs [6].
Unfortunately, current therapeutic agents have inadequate efficacy and many serious adverse effects in treating all kinds of ARDs [7]. All possible options should be considered in order to develop new drugs that are more effective. In fact, many medicinal plants are able to cope with inducers and/or consequences of stress such as thermal or oxidative insults, ionizing radiation, DNA damage, exposure to carcinogens, and inflammatory burden, which is one of the important determinants of survival and longevity [8,9].
Lilium candidum L. has been well known in folk medicine for a long time, not only in the plant’s native regions (Balkans, Middle East) but also in other parts of the world in which it was naturalized, such as various European countries, North Africa, and Mexico. In folk medicine worldwide, L. candidum is prominently associated with dermal conditions, cosmetics, and anti-inflammatory remedies [10,11,12]. A vast ethnopharmacological research study performed in the Campidano Valley and Urzulei district in Italy revealed many medicinal benefits of L. candidum. Among them were application of lily petals and a decoction of bulbs soaked in milk as pectoral poultices, application of petals soaked in spirit as a wound-healing remedy, and the use of oil prepared from flowers as a treatment for mastitis [13]. In addition, ethnopharmacological research in Lucca province in Italy demonstrated the use of L. candidum bulbs as an anti-viral agent to treat shingles (Herpes zoster), and its bulbs and flowers for treatment of skin and articular diseases [14]. L. candidum was also successfully used in anti-inflammatory and dermatological remedies in the Catalan district of the Eastern Pyrenees [14].
L. candidum L., commonly known as Madonna, meadow, or white lily, is a geophyte from the Liliaceae family growing in the wild in several countries of the Middle East. The origin of L. candidum is believed to be in Lebanon and Israel, as well as several parts of Greece [15,16,17], thus Israel represents the Southern border of L. candidum distribution and only few populations are found in the Carmel and the Galilee regions. It is considered an endangered species and as such the plants are protected [15,18]. A collection of wild L. candidum ecotypes from different locations exhibited genetic variation in morphologic and phenologic traits, as well as in phytotoxicity of their leaf extracts [18,19].
Effective delivery of herbal compounds and plant extracts is a very important issue, since drawbacks such as hydrophobicity, insolubility in water, high volatility, and instability pose a challenge [20,21]. The application of innovative drug delivery systems including phytosomes, nanoparticles, hydrogels, microspheres, transferosomes and ethosomes, self-nanoemulsifying drug delivery systems (SNEDDS), self-microemulsifying drug delivery systems (SMEDDS) and so on may improve the biopharmaceutical features of the delivered compounds [22,23].
Owing to its rare beauty, its fragrance, and its glorious symbolism, L. candidum appears to be a fascinating plant. It is therefore not surprising that Madonna lily has also been sought for therapeutic reasons. However, most L. candidum therapeutic properties known from folk tradition have not yet been investigated by scientific methods except anti-fungal [24], anti-cancer [25,26], and anti-viral [27] properties. Thus, the present study is aimed at identifying the presence of selected volatile compounds of the plant, assessing the medicinal potential of all known compounds using bioinformatics, and investigating anti-inflammatory and anti-diabetic activities of L. candidum extract and its active phytochemicals.

2. Results

We identified the presence of many novel volatile compounds using gas chromatography–mass spectrometry (GC-MS) analysis, which, to the best of our knowledge, were not mentioned in the literature. Among those, we found linalool, citronellal, caryophyllene, humulene (Figure 1), and neridiol (not represented), which were isolated and identified not only from L. candidum but from other plants as well.
Table 1 provides a list of these phytochemicals and their medicinal properties.
While it is clear that L. candidum possesses many valuable compounds with considerable therapeutic potential, to the best of our knowledge, no bioinformatical research has been carried out to assess their medicinal potential. In order to close this gap, we analyzed the potential human therapeutic targets (proteins and other biomolecules) of the above-mentioned compounds, using the STITCH database (http://stitch.embl.de/) [63,64,65].
In the case of kaempferol, a significant number of interacting protein targets were found to be associated with three large groups: UGT (uridine 5’-diphospho-glucuronosyltransferase), AHR (aryl hydrocarbon receptor), and CYP1B1 (cytochrome P450 family 1 subfamily B member 1) (Figure 2). The majority of the targets are connected to the UGT enzymes, which participate in cellular detoxification in different tissues of the digestive system [66]. Their involvement in various cancer-associated processes is also well-known [66,67]. AHR is a ligand-activated transcription factor, which may interact with different pathways regulating cellular homeostasis including cellular regeneration in the context of aging and diseases [68]. In addition, AHR regulates CYP1B1 [69], which belongs to the cytochrome P450 superfamily of enzymes. Cytochrome P450 plays an important role in cellular detoxification and in the formation of reactive intermediates of thousands of chemicals [70]. Thus, the medicinal effects of kaempferol are apparently mediated by its direct involvement in many pathological processes.
It is well acknowledged that diabetes mellitus (DM) pathogenesis is linked to oxidative stress [71]. Although many phytochemicals from L. candidum have anti-oxidant properties (Table 1), to the best of our knowledge, no research on its anti-diabetic properties has been performed so far. Thus, we investigated the anti-diabetic activity of bulbs and leaves of L. candidum. For this purpose, we treated adipocytes with an ethanolic extract from L. candidum bulbs and leaves, while the glucose uptake of the adipocytes was estimated after these treatments, as previously. L. candidum extracts increased glucose uptake in 3T3-L1A cells better than insulin. Importantly, the anti-diabetic activity of leaf extracts was higher than those from bulb extracts and of insulin, which was used as a positive control (Figure 3). The difference between negative control (untreated adipocytes) and cells that were treated with extracts was highly significant (p < 0.001). All known phytochemicals of L. candidum were tested in this experiment. As seen in Figure 3, kaempferol, linalool, citronellal, and humulene significantly increased glucose uptake in 3T3-L1 adipocytes (p < 0.001).
Elevated levels of circulating inflammatory mediators including cytokines and chemokines are hallmarks of chronic inflammation and progression of metabolic diseases [2]. The extracts and their phytochemicals (identified by us and known) were investigated from the perspective of chronic inflammation. As seen in Figure 4, plant extracts, kaempferol, citronellal, and humulene significantly decreased secretion of IL-6 and IL-8 cytokines by senescent human pulmonary fibroblasts (HPFs) and human dermal fibroblasts (HDFs) as measured by enzyme-linked immunosorbent assay (ELISA) (p < 0.01).

3. Discussion

Taking into account the therapeutic properties of L. candidum known from folk tradition, we expected to discover anti-inflammatory and anti-diabetic activities in L. candidum extracts. It is important to emphasize here that senescent cells (HPFs and HDFs) are one of the most widely used models for studying inflammatory processes. Accumulation of senescent cells in an organism leads to disruption of tissues and cellular structure and function [72]. The phenomenon of cellular senescence has been demonstrated to play a causal role in driving ageing [72] and chronic diseases [7,9,72]. Namely, the above-mentioned cells are accepted models [7,9] used to investigate the effect of plant extracts and pure compounds on inflammation.
Our hypothesis was confirmed, as the plant extracts indeed possess anti-inflammatory and anti-diabetic properties (Figure 3 and Figure 4). We also identified selected phytochemicals behind these properties: anti-diabetic properties were associated with kaempferol, linalool, citronellal, and humulene (Figure 3). In turn, kaempferol, citronellal, and humulene had a significant impact on the secretion of pro-inflammatory cytokines IL-6 and IL-8 (p < 0.01), (Figure 4). It is known that the above-mentioned cytokines are directly connected to delay in the wound healing process and are known to be secreted by HPFs [9,73]. Thus, our results might provide an explanation of the effect of L. candidum extract on wound healing, as described in ethnopharmacological publications [13,14]. Further investigation is important in order to establish which phytochemicals are connected with wound healing directly, and to elucidate their modes of action.
Although the therapeutic activity of L. candidum extract and its phytochemicals was demonstrated [74,75,76], their mechanism of action is still unknown.
In addition, pro-inflammatory cytokines IL-6 and IL-8 are associated with various diseases including ARDs, such as cancer, diabetes, cardiovascular diseases, multiple sclerosis, asthma, rheumatoid arthritis, and so on [7,8,9]. It is obvious that the stand-alone and combinational anti-inflammatory effects of kaempferol, citronellal, and humulene warrant further investigation. These results are in agreement with those obtained by Vachálková et al. [76], which show that some compounds of L. candidum (spirostanol saponins, two pyroline derivatives, jatropham and its glucoside, 2-fenylethyl-alpha-L-arabinopyranosyl-(1-->6)-beta-D-glucopyranoside, 2-phenylethylpalmitate, methylsuccinic acid and kaempferol) had significant anti-cancer properties [76].
With its rich collection of phytochemicals, L. candidum has promising therapeutic potential. Our results demonstrate a bright future for this plant and its compounds as prophylactic and therapeutic agents. However, additional clinical studies are warranted in order to establish the effectiveness of compounds from L. candidum in the treatment of chronic diseases.

4. Materials and Methods

4.1. Bioinformatic Assessment

For bioinformatical assessment, we selected the STITCH database found at http://stitch.embl.de/. This database contains most of the available information regarding proteins’ interaction with different chemicals. It can be searched according to the name of the chemical or its PubChem ID [63,64,65].

4.2. Preparation of Plant Material

Aerial parts of L. candidum were collected from the greenhouse at Ben-Gurion University of the Negev, Beer-Sheva, Israel. Leaves, flowers, and bulbs of L. candidum were dried by lyophilization and grounded for GC-MS analysis.
Ethanolic extracts were prepared from leaves and bulbs of L. candidum as described previously [27]. Plant tissues were homogenized, incubated at room temperature for 48 h in ethanol, centrifuged at 2000 rpm for 10 min, and the supernatant was evaporated by lyophilization. The pellet was dissolved in a minimal amount of 95% ethanol (0.5 mL) and diluted with water to a final concentration of 10 mg/mL.

4.3. GC-MS Analysis

Identification of volatile compounds is described in our previous publication [7].

4.4. Cell Cultures

All cell culture reagents including Dulbecco’s modified Eagle’s medium (DMEM), fetal bovine serum (FBS), L-glutamine, and antibiotics were purchased from Biological Industries (Kibbutz Beit Haemek, Israel).
Human pulmonary fibroblasts (HPFs), human dermal fibroblasts (HDFs), and 3T3-L1 adipocytes were propagated in DMEM supplemented with 10% FBS, 1% L-glutamine, and 1% antibiotic mixture, which included a combination of penicillin, streptomycin, and nystatin. The cells were grown in an incubator at 37 °C. The relative humidity was set at 95%, and the CO2 content was 5%.

4.5. Cytotoxicity Examination

Nontoxic concentrations of Lilium candidum extracts and compounds were determined by XTT assay. Succinctly, metabolically active cells reduce yellow salt, XTT (sodium 3’-[1-(phenylaminocarbonyl)-3,4-tetrazolium]-bis (4-methoxy6-nitro) benzene sulfonic acid hydrate), to form an orange formazan compound, as described in [7]. For measuring IL-6 and IL-8 levels, only non-toxic concentrations were used.

4.6. Measurement of Anti-Diabetic Activity

Ethanolic bulb and leaf extracts of L. candidum were tested for their anti-diabetic properties. The glucose uptake of the 3T3 adipocytes was determined as described previously [7,71]. Briefly, before the measurement, adipocytes were transferred to low-glucose serum-free media. After an overnight incubation, the cells were treated with bulb or leaf ethanol extracts of L. candidum for 1 h. For negative control, adipocytes were treated with the vehicle only. Insulin at a concentration of 100 nM was used as a positive control. After the 1-h incubation, the fluorescent glucose analog 2-(N-(7-nitrobenz-2-oxa1,3-diazol-4-yl) amino)-2-deoxyglucose (2-NBDG) was added for 30 min. The cells were then rinsed with PBS, and the fluorescence of intracellular 2-NBDG with excitation at 467 nm and emission at 538 nm was measured using a fluorescence microplate reader (POLARstar Omega, BMG LABTECH GmbH, Ortenberg, Germany).

4.7. Measurement of Interleukin Release

Senescent HPFs or HDFs were treated with specified concentrations of the extracts or compounds. After 3 days of exposure, we collected the medium and measured the concentrations of interleukin 6 and 8. The measurement was performed with R&D Systems ELISA kits according to the manufacturer’s instructions. For each experiment, standard curves were built, and the concentrations of interleukins were calculated.

4.8. Statistical Analysis

Experiments were repeated at least three times. All data were analyzed using Statistica, version 7, for Windows software (StatSoft, Inc., Tulsa, Oklahoma), and p < 0.05 was chosen as the minimal acceptable level of significance. Simple regression models were subsequently used to eliminate non-significant effects. Values are presented as means ± SD.

Author Contributions

Conceptualization, L.Y. and S.B.-S.; methodology, L.Y., B.K., and A.B.; validation, L.Y., B.K., and A.B.; resources, L.Y. and J.G.; data curation, B.K. and A.B.; writing—original draft preparation, M.Z. and A.D.; writing—review and editing, M.Z., A.D., L.Y., B.K., and A.B.; visualization, B.K.; supervision, L.Y. and S.B.-S.; project administration, L.Y. and J.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We are pleased to acknowledge Yulia Solomonov for her assistance in data analysis and preparation of the figures and Rima Kozlov for preparation of plant extracts.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Robbins, G.R.; Wen, H.; Ting, J.P. Inflammasomes and metabolic disorders: Old genes in modern diseases. Mol. Cell 2014, 54, 297–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Accardi, G.; Virruso, C.; Balistreri, C.R.; Emanuele, F.; Licastro, F.; Monastero, R.; Porcellini, E.; Vasto, S.; Verga, S.; Caruso, C.; et al. SHIP2: A “new” insulin pathway target for aging research. Rejuvenation Res. 2014, 17, 221–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Thammisetty, S.S.; Pedragosa, J.; Weng, Y.C.; Calon, F.; Planas, A.; Kriz, J. Age-related deregulation of TDP-43 after stroke enhances NF-κB-mediated inflammation and neuronal damage. J. Neuroinflamm. 2018, 15, 312. [Google Scholar] [CrossRef]
  5. Kim, D.H.; Bang, E.; Arulkumar, R.; Ha, S.; Chung, K.W.; Park, M.H.; Choi, Y.J.; Yu, B.P.; Chung, H.Y. Senoinflammation: A major mediator underlying age-related metabolic dysregulation. Exp. Gerontol. 2020, 134, 110891. [Google Scholar] [CrossRef]
  6. Rea, I.M.; Gibson, D.S.; McGilligan, V.; McNerlan, S.E.; Alexander, H.D.; Ross, O.A. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front. Immunol. 2018, 9, 586. [Google Scholar] [CrossRef]
  7. Yarmolinsky, L.; Budovsky, A.; Ben-Shabat, S.; Khalfin, B.; Gorelick, J.; Bishitz, Y.; Miloslavski, R.; Yarmolinsky, L. Recent updates on the phytochemistry and pharmacological properties of Phlomis viscosa Poiret. Rejuvenation Res. 2019, 22, 282–288. [Google Scholar] [CrossRef]
  8. Budovsky, A.; Fraifeld, V.E. Medicinal plants growing in the Judea region network approach for searching potential therapeutic targets. Netw. Biol. 2012, 2, 84–94. [Google Scholar]
  9. Budovsky, A.; Shteinberg, A.; Maor, H.; Duman, O.; Yanai, H.; Wolfson, M.; Fraifeld, V.E. Uncovering the geroprotective potential of medicinal plants from the Judea region of Israel. Rejuvenation Res. 2014, 17, 134–139. [Google Scholar] [CrossRef] [Green Version]
  10. Eisenreichova, E.; Haladova, M.; Mucaji, P.; Budĕsínský, M.; Ubik, K. A new steroidal saponin from the bulbs of Lilium candidum. Pharmazie 2000, 55, 549–550. [Google Scholar]
  11. Jarić, S.; Mačukanović-Jocić, M.; Djurdjević, L.; Mitrović, M.; Kostić, O.; Karadžić, B.; Pavlović, P. An ethnobotanical survey of traditionally used plants on Suva Planina mountain (south-eastern Serbia). J. Ethnopharmacol. 2015, 175, 93–108. [Google Scholar] [CrossRef] [PubMed]
  12. Rigat, M.; Vallès, J.; Gras, A.; Iglésias, J.; Garnatje, T. Plants with topical uses in the ripollès district (Pyrenees, Catalonia, Iberian Peninsula): Ethnobotanical survey and pharmacological validation in the literature. J. Ethnopharmacol. 2015, 164, 162–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Bruni, A.; Ballero, M.; Poli, F. Quantitative ethnopharmacological study of the campidano valley and urzulei district, Sardinia, Italy. J. Ethnopharmacol. 1997, 57, 97–124. [Google Scholar] [CrossRef]
  14. Pieroni, A. Medicinal plants and food medicines in the folk traditions of the upper Lucca province, Italy. J. Ethnopharmacol. 2000, 70, 235–273. [Google Scholar] [CrossRef]
  15. Feinbrun-Dothan, N.; Danin, A.L. Candidum . In Analytical Flora of Eretz-Israel; Plitman, U., Ed.; CANA Publishing: Jerusalem, Israel, 1991; p. 779. [Google Scholar]
  16. Polunin, O. Flowers of Greece and the Balkans; Oxford University Press: Oxford, UK, 1987; pp. 1–592. [Google Scholar]
  17. Mouterde, P. Lilium candidum. In Nouvelle Flore du Liban et de la Syrie; Imprimerie, C., Ed.; Editions de l’Impr. Catholique: Beyrouth, Lebanon, 1966; Volume 1, p. 236. [Google Scholar]
  18. Zaccai, M.; Ram, A.; Mazor, I. Lilium candidum: Flowering characterization of wild Israeli ecotypes. Israel J. Plant Sci. 2010, 57, 297–302. [Google Scholar] [CrossRef]
  19. Rubin, N.; Huleihel, M.; Zaccai, M. Stress conditions during plant growth increase the anti-herpetic properties of Lilium candidum leaf extracts and fractions. J. Med. Plants Res. 2015, 9, 954–961. [Google Scholar]
  20. Trinetta, V.; Morgan, M.T.; Coupland, J.N.; Yucel, U. Essential oils against pathogen and spoilage microorganisms of fruit juices: Use of versatile antimicrobial delivery systems. J. Food Sci. 2017, 82, 471–476. [Google Scholar] [CrossRef]
  21. Carbone, C.; Martins-Gomes, C.; Caddeo, C.; Silva, A.M.; Musumeci, T.; Pignatello, R.; Puglisi, G.; Souto, E.B. Mediterranean essential oils as precious matrix components and active ingredients of lipid nanoparticles. Int. J. Pharm. 2018, 548, 217–226. [Google Scholar]
  22. Bruni, N.; Stella, B.; Giraudo, L.; Della Pepa, C.; Gastaldi, D.; Dosio, F. Nanostructured delivery systems with improved leishmanicidal activity: A critical review. Int. J. Nanomed. 2017, 12, 5289–5311. [Google Scholar] [CrossRef] [Green Version]
  23. Ben-Shabat, S.; Yarmolinsky, L.; Porat, D.; Dahan, A. Antiviral effect of phytochemicals from medicinal plants: Applications and drug delivery strategies. Drug Deliv. Transl. Res. 2020, 10, 354–367. [Google Scholar] [CrossRef] [Green Version]
  24. Mucaji, P.; Hudecova, D.; Haladová, M.; Eisenreichová, E. Anti-yeast activity of ethanol extracts of Lilium candidum L. Ceska Slov. Farm. Cas. Ceske Farm. Spol. Slov. Farm. Spol. 2002, 51, 297–300. [Google Scholar]
  25. Galova, E.; Kopaskova, M.; Sevcovicova, A.; Hadjo, L.; Yankulova, B.; Gregan, F.; Chankova, S.; Miadokova, E. The role of antioxidants from Lilium candidum L. and Salvia officinalis L. Extracts in phytomedicine. Toxicol. Lett. 2011, 205, S60. [Google Scholar] [CrossRef]
  26. Tokgun, O.; Akca, H.; Mammadov, R.; Aykurt, C.; Deniz, G. Convolvulus galaticus, Crocus antalyensis, and Lilium candidum extracts show their antitumor activity through induction of p53-mediated apoptosis on human breast cancer cell line MCF-7 cells. J. Med. Food 2012, 15, 1000–1005. [Google Scholar] [CrossRef] [PubMed]
  27. Yarmolinsky, L.; Zaccai, M.; Ben-Shabat, S.; Mills, D.; Huleihel, M. Antiviral activity of ethanol extracts of Ficus binjamina and Lilium candidum in vitro. New Biotechnol. 2009, 26, 307–313. [Google Scholar] [CrossRef] [PubMed]
  28. Bolaños, V.; Díaz-Martínez, A.; Soto, J.; Marchat, L.A.; Sanchez-Monroy, V.; Ramírez-Moreno, E. Kaempferol inhibits Entamoeba histolytica growth by altering cytoskeletal functions. Mol. Biochem. Parasitol. 2015, 204, 16–25. [Google Scholar] [CrossRef]
  29. Choi, J.H.; Park, S.E.; Kim, S.J.; Kim, S. Kaempferol inhibits thrombosis and platelet activation. Biochimie 2015, 115, 177–186. [Google Scholar] [CrossRef]
  30. Devi, K.P.; Malar, D.S.; Nabavi, S.F.; Sureda, A.; Xiao, J.; Nabavi, S.M.; Daglia, M. Kaempferol and inflammation: From chemistry to medicine. Pharmacol. Res. 2015, 99, 1–10. [Google Scholar] [CrossRef]
  31. Hoang, M.H.; Jia, Y.; Mok, B.; Jun, H.J.; Hwang, K.Y.; Lee, S.J. Kaempferol ameliorates symptoms of metabolic syndrome by regulating activities of liver x receptor-β. J. Nutr. Biochem. 2015, 26, 868–875. [Google Scholar] [CrossRef]
  32. Li, H.; Yang, L.; Zhang, Y.; Gao, Z. Kaempferol inhibits fibroblast collagen synthesis, proliferation and activation in hypertrophic scar via targeting tgf-β receptor type i. Biomed. Pharmacother. 2016, 83, 967–974. [Google Scholar] [CrossRef]
  33. Shin, D.; Park, S.H.; Choi, Y.J.; Kim, Y.H.; Antika, L.D.; Habibah, N.Y.; Kang, M.K.; Kang, Y.H. Dietary compound kaempferol inhibits airway thickening induced by allergic reaction in a bovine serum albumin-induced model of asthma. Int. J. Mol. Sci. 2015, 16, 29980–29995. [Google Scholar] [CrossRef] [Green Version]
  34. Suchal, K.; Malik, S.; Gamad, N.; Malhotra, R.K.; Goyal, S.N.; Bhatia, J.; Arya, D.S. Kampeferol protects against oxidative stress and apoptotic damage in experimental model of isoproterenol-induced cardiac toxicity in rats. Phytomedicine 2016, 23, 1401–1408. [Google Scholar] [CrossRef] [PubMed]
  35. Suchal, K.; Malik, S.; Gamad, N.; Malhotra, R.K.; Goyal, S.N.; Chaudhary, U.; Bhatia, J.; Ojha, S.; Arya, D.S. Kaempferol attenuates myocardial ischemic injury via inhibition of mapk signaling pathway in experimental model of myocardial ischemia-reperfusion injury. Oxid. Med. Cell. Longev. 2016, 2016, 7580731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Alves, S.; Duarte, A.; Sousa, S.; Domingues, F.C. Study of the major essential oil compounds of coriandrum sativum against acinetobacter baumannii and the effect of linalool on adhesion, biofilms and quorum sensing. Biofouling 2016, 32, 155–165. [Google Scholar] [CrossRef] [PubMed]
  37. Dutra, F.L.; Oliveira, M.M.; Santos, R.S.; Silva, W.S.; Alviano, D.S.; Vieira, D.P.; Lopes, A.H. Effects of linalool and eugenol on the survival of Leishmania (L.) infantum chagasi within macrophages. Acta Trop. 2016, 164, 69–76. [Google Scholar] [CrossRef]
  38. Li, X.J.; Yang, Y.J.; Li, Y.S.; Zhang, W.K.; Tang, H.B. A-pinene, linalool, and 1-octanol contribute to the topical anti-inflammatory and analgesic activities of frankincense by inhibiting cox-2. J. Ethnopharmacol. 2016, 179, 22–26. [Google Scholar] [CrossRef]
  39. Mehri, S.; Meshki, M.A.; Hosseinzadeh, H. Linalool as a neuroprotective agent against acrylamide-induced neurotoxicity in wistar rats. Drug Chem. Toxicol. 2015, 38, 162–166. [Google Scholar] [CrossRef]
  40. Park, H.; Seol, G.H.; Ryu, S.; Choi, I.Y. Neuroprotective effects of (−)-linalool against oxygen-glucose deprivation-induced neuronal injury. Arch. Pharmacal Res. 2016, 39, 555–564. [Google Scholar] [CrossRef]
  41. Sabogal-Guáqueta, A.M.; Osorio, E.; Cardona-Gómez, G.P. Linalool reverses neuropathological and behavioral impairments in old triple transgenic alzheimer’s mice. Neuropharmacology 2016, 102, 111–120. [Google Scholar] [CrossRef] [Green Version]
  42. Seol, G.H.; Kang, P.; Lee, H.S.; Seol, G.H. Antioxidant activity of linalool in patients with carpal tunnel syndrome. BMC Neurol. 2016, 16, 17. [Google Scholar] [CrossRef] [Green Version]
  43. Souto-Maior, F.N.; Fonsêca, D.V.D.; Salgado, P.R.R.; Monte, L.D.O.; de Sousa, D.P.; de Almeida, R.N. Antinociceptive and anticonvulsant effects of the monoterpene linalool oxide. Pharm. Biol. 2017, 55, 63–67. [Google Scholar]
  44. Du, E.J.; Ahn, T.J.; Choi, M.S.; Kwon, I.; Kim, H.W.; Kwon, J.Y.; Kang, K. The mosquito repellent citronellal directly potentiates drosophila trpa1, facilitating feeding suppression. Mol. Cells 2015, 38, 911. [Google Scholar]
  45. Maßberg, D.; Simon, A.; Häussinger, D.; Keitel, V.; Gisselmann, G.; Conrad, H.; Hatt, H. Monoterpene (−)-citronellal affects hepatocarcinoma cell signaling via an olfactory receptor. Arch. Biochem. Biophys. 2015, 566, 100–109. [Google Scholar] [CrossRef] [PubMed]
  46. Singh, S.; Fatima, Z.; Hameed, S. Citronellal-induced disruption of membrane homeostasis in Candida albicans and attenuation of its virulence attributes. Rev. Soc. Bras. Med. Trop. 2016, 49, 465–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Basha, R.H.; Sankaranarayanan, C. Β-caryophyllene, a natural sesquiterpene lactone attenuates hyperglycemia mediated oxidative and inflammatory stress in experimental diabetic rats. Chem. Biol. Interact. 2016, 245, 50–58. [Google Scholar] [CrossRef] [PubMed]
  48. De Oliveira, C.C.; de Oliveira, C.V.; Grigoletto, J.; Ribeiro, L.R.; Funck, V.R.; Grauncke, A.C.B.; de Souza, T.L.; Souto, N.S.; Furian, A.F.; Menezes, I.R.A. Anticonvulsant activity of β-caryophyllene against pentylenetetrazol-induced seizures. Epilepsy Behav. 2016, 56, 26–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Di Giacomo, S.; Mazzanti, G.; Di Sotto, A. Mutagenicity of cigarette butt waste in the bacterial reverse mutation assay: The protective effects of β-caryophyllene and β-caryophyllene oxide. Environ. Toxicol. 2016, 31, 1319–1328. [Google Scholar] [CrossRef] [PubMed]
  50. Fidyt, K.; Fiedorowicz, A.; Strządała, L.; Szumny, A. Β-caryophyllene and β-caryophyllene oxide—Natural compounds of anticancer and analgesic properties. Cancer Med. 2016, 5, 3007–3017. [Google Scholar] [CrossRef]
  51. Govindarajan, M.; Benelli, G. A-humulene and β-elemene from syzygium zeylanicum (myrtaceae) essential oil: Highly effective and eco-friendly larvicides against anopheles subpictus, aedes albopictus, and culex tritaeniorhynchus (diptera: Culicidae). Parasitol. Res. 2016, 115, 2771–2778. [Google Scholar] [CrossRef]
  52. Govindarajan, M.; Rajeswary, M.; Hoti, S.; Bhattacharyya, A.; Benelli, G. Eugenol, α-pinene and β-caryophyllene from plectranthus barbatus essential oil as eco-friendly larvicides against malaria, dengue and japanese encephalitis mosquito vectors. Parasitol. Res. 2016, 115, 807–815. [Google Scholar] [CrossRef]
  53. Kelany, M.E.; Abdallah, M.A. Protective effects of combined β-caryophyllene and silymarin against ketoprofen-induced hepatotoxicity in rats. Can. J. Physiol. Pharmacol. 2016, 94, 739–744. [Google Scholar] [CrossRef]
  54. Ojha, S.; Javed, H.; Azimullah, S.; Haque, M.E. Β-caryophyllene, a phytocannabinoid attenuates oxidative stress, neuroinflammation, glial activation, and salvages dopaminergic neurons in a rat model of parkinson disease. Mol. Cell. Biochem. 2016, 418, 59–70. [Google Scholar] [CrossRef] [PubMed]
  55. Pieri, F.A.; Souza, M.C.; Vermelho, L.L.; Vermelho, M.L.; Perciano, P.G.; Vargas, F.S.; Borges, A.P.; da Veiga-Junior, V.F.; Moreira, M.A. Use of β-caryophyllene to combat bacterial dental plaque formation in dogs. BMC Vet. Res. 2016, 12, 216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Tian, X.; Peng, J.; Zhong, J.; Yang, M.; Pang, J.; Lou, J.; Li, M.; An, R.; Zhang, Q.; Xu, L. B-caryophyllene protects in vitro neurovascular unit against oxygen-glucose deprivation and re-oxygenation-induced injury. J. Neurochem. 2016, 139, 757–768. [Google Scholar] [CrossRef] [Green Version]
  57. Lan, Y.H.; Wu, Y.C.; Wu, K.W.; Chung, J.G.; Lu, C.C.; Chen, Y.L.; Wu, T.S.; Yang, J.S. Death receptor 5-mediated TNFR family signaling pathways modulate γ-humulene-induced apoptosis in human colorectal cancer HT29 cells. Oncol. Rep. 2011, 25, 419–424. [Google Scholar] [PubMed] [Green Version]
  58. Rogerio, A.P.; Andrade, E.L.; Leite, D.F.; Figueiredo, C.P.; Calixto, J.B. Preventive and therapeutic anti-inflammatory properties of the sesquiterpene α-humulene in experimental airways allergic inflammation. Br. J. Pharmacol. 2009, 158, 1074–1087. [Google Scholar] [CrossRef] [Green Version]
  59. Baldissera, M.D.; Souza, C.F.; Grando, T.H.; Moreira, K.L.; Schafer, A.S.; Cossetin, L.F.; da Silva, A.P.; da Veiga, M.L.; da Rocha, M.I.; Stefani, L.M.; et al. Nerolidol-loaded nanospheres prevent behavioral impairment via ameliorating Na+, K+-ATPase and AChE activities as well as reducing oxidative stress in the brain of Trypanosoma evansi-infected mice. Naunyn Schmiedebergs Arch. Pharmacol. 2017, 390, 139–148. [Google Scholar] [PubMed]
  60. Ferreira, M.O.G.; Leite, L.L.R.; de Lima, I.S.; Barreto, H.M.; Nunes, L.C.C.; Ribeiro, A.B.; Osajima, J.A.; da Silva Filho, E.C. Chitosan Hydrogel in combination with Nerolidol for healing wounds. Carbohydr. Polym. 2016, 152, 409–418. [Google Scholar] [CrossRef]
  61. Javed, H.; Azimullah, S.; Abul Khair, S.B.; Ojha, S.; Haque, M.E. Neuroprotective effect of nerolidol against neuroinflammation and oxidative stress induced by rotenone. BMC Neurosci. 2016, 17, 58. [Google Scholar] [CrossRef] [Green Version]
  62. Kaur, D.; Pahwa, P.; Goel, R.K. Protective effect of nerolidol against pentylenetetrazol-induced kindling, oxidative stress and associated behavioral comorbidities in mice. Neurochem. Res. 2016, 41, 2859–2867. [Google Scholar]
  63. Kuhn, M.; von Mering, C.; Campillos, M.; Jensen, L.J.; Bork, P. STITCH: Interaction networks of chemicals and proteins. Nucleic Acids Res. 2008, 36, 684–688. [Google Scholar] [CrossRef]
  64. Kuhn, M.; Szklarczyk, D.; Franceschini, A.; Campillos, M.; von Mering, C.; Jensen, L.J.; Beyer, A.; Bork, P. STITCH 2: An interaction network database for small molecules and proteins. Nucleic Acids Res. 2010, 38, 552–556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kuhn, M.; Szklarczyk, D.; Pletscher-Frankild, S.; Blicher, T.H.; von Mering, C.; Jensen, L.J.; Bork, P. STITCH 4: Integration of protein-chemical interactions with user data. Nucleic Acids Res. 2014, 42, 401–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Strassburg, C.P.; Strassburg, A.; Nguyen, N.; Qing, L.; Manns, M.P.; Tukey, R.H. Regulation and function of family 1 and family 2 udp-glucuronosyltransferase genes (ugt1a, ugt2b) in human oesophagus. Biochem. J. 1999, 338, 489–498. [Google Scholar] [CrossRef] [PubMed]
  67. Yilmaz, L.; Borazan, E.; Aytekin, T.; Baskonus, I.; Aytekin, A.; Oztuzcu, S.; Bozdag, Z.; Balik, A. Increased ugt1a3 and ugt1a7 expression is associated with pancreatic cancer. Asian Pac. J. Cancer Prev. 2014, 16, 1651–1655. [Google Scholar]
  68. Casado, F.L. The aryl hydrocarbon receptor relays metabolic signals to promote cellular regeneration. Stem Cells Int. 2016, 2016, 4389802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Rasmussen, M.K.; Balaguer, P.; Ekstrand, B.; Daujat-Chavanieu, M.; Gerbal-Chaloin, S. Skatole (3-methylindole) is a partial aryl hydrocarbon receptor agonist and induces cyp1a1/2 and cyp1b1 expression in primary human hepatocytes. PLoS ONE 2016, 11, e0154629. [Google Scholar]
  70. Nebert, D.W.; Dalton, T.P. The role of cytochrome p450 enzymes in endogenous signalling pathways and environmental carcinogenesis. Nat. Rev. Cancer 2006, 6, 947–960. [Google Scholar] [CrossRef]
  71. Gorelick, J.; Kitron, A.; Pen, S.; Rosenzweig, T.; Madar, Z. Anti-diabetic activity of Chiliadenus iphionoides. J. Ethnopharmacol. 2011, 137, 1245–1249. [Google Scholar] [CrossRef]
  72. Song, P.; An, J.; Zou, M.H. Immune Clearance of Senescent Cells to Combat Ageing and Chronic Diseases. Cells 2020, 9, 671. [Google Scholar] [CrossRef] [Green Version]
  73. Budovsky, A.; Yarmolinsky, L.; Ben-Shabat, S. Effect of medicinal plants on wound healing. Wound Repair Regen. 2015, 23, 171–183. [Google Scholar]
  74. Patocka, J.; Navratilova, Z.; Yokozawa, T. Bioactivity of Lilium candidum L.—A Mini Review. Biomed. J. Sci. Tech. Res. 2019, 18, 13859–13862. [Google Scholar] [CrossRef] [Green Version]
  75. Haladova, M.; Eisenreichova, E.; Mucaji, P.; Budesinsky, M.; Ubik, K. Steroidal Saponins from Lilium candidum L. Collect. Czechoslov. Chem. Commun. 1998, 63, 205–210. [Google Scholar] [CrossRef]
  76. Vachálková, A.; Eisenreichová, E.; Haladová, M.; Mucaji, P.; Józová, B.; Novotný, L. Potential carcinogenic and inhibitory activity of compounds isolated from Lilium candidum L. Neoplasma 2000, 47, 313–318. [Google Scholar]
Figure 1. Chemical structures of compounds isolated from L. candidum. A computerized GC-MS (GC-6890N) equipped with a mass selective (MS)—5973 network (electron ionization 70 eV) detector from Agilent Technologies (Santa Clara, CA, USA) was used. Component recognition was performed by a comparison of the retention time index (RI) of the components to commercial standards and the samples’ mass spectrum with GC-MS libraries: Adams 2001, NIST 98, and QuadLib 1607.
Figure 1. Chemical structures of compounds isolated from L. candidum. A computerized GC-MS (GC-6890N) equipped with a mass selective (MS)—5973 network (electron ionization 70 eV) detector from Agilent Technologies (Santa Clara, CA, USA) was used. Component recognition was performed by a comparison of the retention time index (RI) of the components to commercial standards and the samples’ mass spectrum with GC-MS libraries: Adams 2001, NIST 98, and QuadLib 1607.
Plants 09 00959 g001
Figure 2. Direct protein–protein and protein–compound interactions between human targets of kaempferol. This figure was constructed using the default settings of the STITCH database (http://stitch.embl.de/). The uridine 5’-diphospho-glucuronosyltransferase (UGT), aryl hydrocarbon receptor (AHR), and cytochrome P450 family 1 subfamily B member 1 (CYP1B1) proteins groups are dominant.
Figure 2. Direct protein–protein and protein–compound interactions between human targets of kaempferol. This figure was constructed using the default settings of the STITCH database (http://stitch.embl.de/). The uridine 5’-diphospho-glucuronosyltransferase (UGT), aryl hydrocarbon receptor (AHR), and cytochrome P450 family 1 subfamily B member 1 (CYP1B1) proteins groups are dominant.
Plants 09 00959 g002
Figure 3. Anti-diabetic activity of L. candidum extracts and compounds. The glucose uptake of the 3T3 adipocytes was determined. Adipocytes were treated with ethanol extracts of L. candidum and with the compounds detected in those extracts. Negative control consisted of untreated adipocytes; insulin was used as a positive control. Data from three independent experiments are shown (mean ± SD). *** p < 0.001.
Figure 3. Anti-diabetic activity of L. candidum extracts and compounds. The glucose uptake of the 3T3 adipocytes was determined. Adipocytes were treated with ethanol extracts of L. candidum and with the compounds detected in those extracts. Negative control consisted of untreated adipocytes; insulin was used as a positive control. Data from three independent experiments are shown (mean ± SD). *** p < 0.001.
Plants 09 00959 g003
Figure 4. Effect of L. candidum ethanolic extracts and compounds on the release of pro-inflammatory cytokines interleukin 6 (IL-6) (A,C) and interleukin 8 (IL-8) (B,D) from human pulmonary fibroblasts (HPFs) (A,B) or human dermal fibroblasts (HDFs) (C,D). The ethanolic extracts were diluted in medium to a final ethanol concentration of 0.1%. Cells treated only with 0.1% ethanol (the vehicle) were used as controls to exclude the effect of ethanol on the cells. Data from three independent experiments are shown (mean ± SE). ** p < 0.01, *** p < 0.001.
Figure 4. Effect of L. candidum ethanolic extracts and compounds on the release of pro-inflammatory cytokines interleukin 6 (IL-6) (A,C) and interleukin 8 (IL-8) (B,D) from human pulmonary fibroblasts (HPFs) (A,B) or human dermal fibroblasts (HDFs) (C,D). The ethanolic extracts were diluted in medium to a final ethanol concentration of 0.1%. Cells treated only with 0.1% ethanol (the vehicle) were used as controls to exclude the effect of ethanol on the cells. Data from three independent experiments are shown (mean ± SE). ** p < 0.01, *** p < 0.001.
Plants 09 00959 g004
Table 1. Medicinal properties of phytochemicals present in Lilium candidum.
Table 1. Medicinal properties of phytochemicals present in Lilium candidum.
CompoundMedicinal UsesReferences
KaempferolAnti-apoptotic, pro-wound healing, anti-cancer, cardioprotective, anti-oxidant, pro-apoptotic, anti-allergic, anti-parasitic, anti-diabetic, anti-adipogenic, anti-thrombotic, anti-inflammatory, anti-metabolic syndrome, anti-bacterial, immunoregulatory, hepatoprotective, anti-atherosclerosis[28,29,30,31,32,33,34,35]
LinaloolAnti-parasitic, anti-convulsant, anti-cancer, anti-bacterial, neuroprotective, anti-oxidant, anti-inflammatory, anti-Alzheimer, anxiolytic, hepatoprotective, anti-hyperalgesic, neuroprotective[36,37,38,39,40,41,42,43]
CitronellalAnti-fungal, insect repellant, hepatoprotective, anti-nociceptive, anti-inflammatory, anti-bacterial[44,45,46]
CaryophylleneAnti-cancer, anti-mutagenic, anti-bacterial, oxygen deprivation protective, neuroprotective, hepatoprotective, anti-convulsant, anti-diabetic, anti-microbial, anti-Alzheimer, pro-longevity, analgesic, nephroprotective[47,48,49,50,51,52,53,54,55,56]
HumuleneInsecticidal, anti-cancer, anti-inflammatory[51,57,58]
NeridiolAnti-parasitic, antioxidant, neuroprotective, pro-wound healing, anti-microbial[59,60,61,62]

Share and Cite

MDPI and ACS Style

Zaccai, M.; Yarmolinsky, L.; Khalfin, B.; Budovsky, A.; Gorelick, J.; Dahan, A.; Ben-Shabat, S. Medicinal Properties of Lilium candidum L. and Its Phytochemicals. Plants 2020, 9, 959. https://doi.org/10.3390/plants9080959

AMA Style

Zaccai M, Yarmolinsky L, Khalfin B, Budovsky A, Gorelick J, Dahan A, Ben-Shabat S. Medicinal Properties of Lilium candidum L. and Its Phytochemicals. Plants. 2020; 9(8):959. https://doi.org/10.3390/plants9080959

Chicago/Turabian Style

Zaccai, Michele, Ludmila Yarmolinsky, Boris Khalfin, Arie Budovsky, Jonathan Gorelick, Arik Dahan, and Shimon Ben-Shabat. 2020. "Medicinal Properties of Lilium candidum L. and Its Phytochemicals" Plants 9, no. 8: 959. https://doi.org/10.3390/plants9080959

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