Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2538 word(s) 2538 2022-03-17 03:17:53 |
2 format correct -7 word(s) 2531 2022-03-21 02:35:16 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Rashid, H. Lemon Verbena (Aloysia citrodora). Encyclopedia. Available online: https://encyclopedia.pub/entry/20759 (accessed on 26 April 2024).
Rashid H. Lemon Verbena (Aloysia citrodora). Encyclopedia. Available at: https://encyclopedia.pub/entry/20759. Accessed April 26, 2024.
Rashid, Hasan. "Lemon Verbena (Aloysia citrodora)" Encyclopedia, https://encyclopedia.pub/entry/20759 (accessed April 26, 2024).
Rashid, H. (2022, March 19). Lemon Verbena (Aloysia citrodora). In Encyclopedia. https://encyclopedia.pub/entry/20759
Rashid, Hasan. "Lemon Verbena (Aloysia citrodora)." Encyclopedia. Web. 19 March, 2022.
Lemon Verbena (Aloysia citrodora)
Edit

Aloysia citrodora (Verbenaceae), an acknowledged medicinal plant, is traditionally used to treat various diseases, including bronchitis, insomnia, anxiety, digestive, and heart problems.

lemon verbena plant extracts traditional medicine antioxidant antiproliferation

1. Introduction

Worldwide estimation of cancer incidence and mortality, created by the International Agency for Research on Cancer, revealed that 19.3 million new cancer cases and almost 10.0 million cancer deaths occurred in 2020. Female breast cancer is the most diagnosed cancer, surpassing lung cancer, with (11.7%) new cases of the total reported cases, followed by lung cancer (11.4%). The global cancer incidences are expected to be 28.4 million cases in 2040, a 47% rise from 2020 [1].
Cancer is a disorder that triggers normal cells’ uncontrolled growth and alters the genome (which causes malignant characteristics in normal cells) [2]. This overgrowth progression impairs the normal biological process of healthy cells by the invasion of nearby tissues and metastasizes to distant tissues [3]. The first options of treatment are chemotherapy, radiation therapy, and surgery. Many adjuvant therapies and strategies such as quality of life changes, antioxidant supplements, herbal medicines, and remedies based on natural products are continuously growing as promising strategies to augment conventional therapies [4][5][6]. Today, the most reliable option for cancer treatment is chemotherapy, and drug resistance appears as a significant limitation [7][8]. These limitations push researchers to find other sources for treatment and direct researchers vision into the richest source of treatments from ancient times, to nature.
Another problem is the free radicals that can cause oxidative damage to any sort of cell component. This type of damage in humans can lead to various degenerative disorders, including cancer and cell aging, directing the interest to the antioxidants [9]. Antioxidant molecules, pharmacologically effective and with few or no adverse effects, are currently being explored for preventive care worldwide and in the food industry. Plants are vulnerable to the harmful effects of active oxygen and produce many antioxidant chemicals in addition to tocopherols. Flavonoids, several other phenolic compounds, and polyphenolics are examples of these substances [9].
All societies around the world have consumed medicinal plants. In the United States and Europe, natural products or their derivatives account for nearly (50%) of all prescription drugs [10]. Many drugs used to treat cancer, infections, and other disorders are entirely derived from plants or are synthetic/semi-synthetic derivatives of plants [11]. According to the World Health Organization (WHO), (11%) of drugs are derived entirely from plants, with a significant percentage of synthetic medications derived from natural precursors [12]. Showing the increasing importance of natural products in drug development.
Approximately 150 plant herbs are still used as a source of traditional herbal therapy in Jordan [13]. One of these herbs is Lemon verbena, scientifically known as Aloysia citrodora Paláu, an acknowledged medicinal plant [14]A. citrodora has a broad range of medicinal, cosmetic, aromatic, and culinary applications [15]A. citrodora extracts and preparations are mostly reported to have antioxidant and anti-microbial biological activities [16][17][18][19]. Phytochemically, the extracts of A. citrodora were found to have an excessive amount of phenolics such as phenylpropanoids, flavonoids, lignans, tannins, and a variety of other nonphenolic compounds [20]. Generally, the major compounds of the oil of A. citrodora were the citral isomers geranial and neral [21][22]. However, limonene, 1,8-cineol, β-caryophyllene, citronellal, and citronellol were frequently listed as major ingredients. Furthermore, studies showed that the oil composition is affected by numerous factors like plants’ genotype, environmental factors, and growth conditions [23][24].

2. Antioxidant Activity

Natural antioxidants found in medicinal herbs are responsible for reducing or inhibiting the harmful effects of oxidative stress and reactive oxygen species (ROS), which have been detected in almost all cancer types and promote their progression, development, survival, and boost its metastatic ability. Novel therapeutic approaches are needed to control the intracellular ROS signaling production and ROS-induced tumor. Then again, the antioxidants could avert initiation events of cancer, where ROS considered a crucial part [25]. The hydroxyl groups of the active compounds permit a scavenging effect on various reactive oxygen and nitrogen species and provide a powerful antioxidant effect [26].
Free radical scavenging activity is determined through several available procedures. However, the radical-scavenging DPPH test has attracted the most attention [27]. It has been utilized for evaluating the antiradical properties of extracts very often because it can accommodate many samples in a short amount of time and is sensitive enough to detect active components at low concentrations [28].
Ethanol extracts were found to be the most potent scavengers against the DPPH radical, followed by water extract. It could be due to the amount of phenols and flavonoids [29]. Depending on the TPC experiment, phenols are abundant in these extracts. Although TPC measures all phenolics, some heterogeneous phenolic compounds could respond differently. F–C values are found to be higher than those obtained by other methods such as HPLC-MS/MS and HPLC-UV [30][31]. A study implied that the antioxidant capability of an extract is determined not only by the amount of polyphenols present but also by the nature of the antioxidants molecules [32]. The potent antioxidant extracts are rich in polyphenols. Numerous investigations have found a link between phenolic composites and radical-scavenging capacity [33][34]. A significant linear correlation was found between the content of phenolic compounds and the extracts’ antioxidant activity in a few plants [35].
Ethyl acetate extract showed lower activity against DPPH. The previous findings on the extract components revealed potent activity when assessed solely [36][37][38]. This activity could be affected by many factors. The constituent of the reacting mixture differs in molecular size, polarity, solubility, chemical structure, concentration, and molecular ratio. The interactions between the components (not only with the active compounds) result in additive, synergistic or antagonistic effects [39]. As in the case of ethyl acetate extract, most probably an antagonistic effect between its components causes the drop in the activity.
The free-radical scavenging activity of A. citrodora EO was also assessed in this entry. The oil did not exhibit any radical scavenging activity at all concentrations examined. Abuhamdah, R., & Mohammed (2014) conducted a study on Jordanian origin A. citrodora fresh and dried leaves identified 83 compounds of essential oil chemical composition. The highest components were limonene (12.14%) caryophyllene oxide (10.44 %), curcumene (9.17 %), spathulenol (7.16 %), 1,8-cineole (7.94%), followed by geranial (4.03 %) and neral (2.55) [40]. Another study of the EOs obtained from dried or fresh Jordanian A. citrodora leaves identified limonene, neral, and geranial as major components. They have also identified in low amounts α-pinene, α-terpinene, sabinene, linalool, and caryophyllene [41]. A study of Jordanian A. citrodora aerial parts by Hudaib et al. (2013) revealed limonene (17.7%) as the highest constituent, together with the citral isomers (neral: 9.8% and geranial: 10.1%), representing more than one-third of the oil. Other major components identified in A. citrodora oil included mainly 1,8-cineole (11.7%), α-curcumene (6.3%), and the oxygenated sesquiterpene components spathulenol (4.6%) and caryophyllene oxide (3.1%) [42].
Studies from several countries in the literature revealed the presence of citral isomers (neral and geranial) in A. citrodora essential oil [43][44][45][46][47]. However, the present entry results did not indicate the occurrence of any common major detected compounds (e.g., neral, geranial, thujone, citronellal, carvone). In a recent article of the A. citrodora EOs from two separate localities, the antioxidant activity of the plant containing more citral resulted in more potent antioxidant activities [48]. The literature emphasizes that various factors could affect EOs composition, such as the age-related stage of the plant, its physiology, and growth conditions. Also, the constituents of the EO could be altered by the isolation and analysis conditions [41][49][50][51].
Another factor affecting EOs antioxidant activity could be the presence of Alpha-pinene and limonene compounds which have poor activity in the DPPH test system. The antioxidative effect of EOs is frequently not attributable to the primary constituents; lesser molecules and synergistic effects may play a substantial role in the activity [52]. Antioxidant activity can be induced by the presence of heteroatom-containing compounds in EOs. Oxygen-containing moieties, such as phenols or hydroxyl, are more effective antioxidants than nitrogen-containing structures like aniline [53].
Variations in the amounts of each compound identified in these extracts had a strong correlation with the extract activity. Jordanian A. citrodora in this analysis revealed high variation in its composition compared to studies from different countries [52][53], even its EO components identified in Jordan 9 years ago [42]. The current findings could indicate the significant effects of these factors (mentioned above) on the quality and quantity of the extracts.

3. Antiproliferative Activity

3.1. In Vitro Study

Ethyl acetate extract established the highest anti-proliferation activity, followed by the EO and ethanol extract. In contrast, water extract exhibited the lowest activity. The difference in activity could result from the various flavonoids and phenolic substances’ chemical structure types found in the extracts. Like in many other plant species, flavonoids might be responsible for the major bioactivities of A. citrodora, such as antimicrobial, neuropsychological, antioxidant, cytoprotective, and anti-cancer effects [54]. Flavonoids can occur naturally either as a compound associated with sugar in conjugated form (glycosides) or without attached sugar as aglycones. The presence or absence of sugar moiety can affect the flavonoids’ solubility, reactivity, and stability [55]. Overall, pharmacokinetic properties can have a major impact on the health-promoting effects of phytochemicals [26].
To the best of researchers knowledge, the current entry is the first attempt to assess the antiproliferative property of the aerial parts of A. citrodora extracted by ethyl acetate (EA) as solvents that had the ability to extract various non-polar active compounds. The LC-MS analysis revealed naringenin (flavanones) as the most abundant compound in EA extract (25.22%). Naringenin is insoluble in water and soluble in organic solvents, like alcohol [56][57]. It has been found in several plants possessing various biological activities like antioxidant, antitumor, antiviral, antibacterial, anti-inflammatory, and cardioprotective effects [36][58][59][60]. In many reports, Naringenin exhibited antiproliferation effect, ability to inhibit cell growth, increase AMP-activated protein kinase phosphorylation, CyclinD1 expression down-regulation, and cell death induction. Other reports include promising results for prostate cancer, melanoma, and gliomas-brain cancer [61][62][63].
Another major detected component was 5,6,4′-Trihydroxy-7,3′-Dimethoxyflavone (5-TDMF) (23.67%). It is proven to have potent antioxidant activity in vitro and ex vivo. One study demonstrates that 5-TDMF has potent antioxidant and anti-inflammatory effects without cytotoxicity observed. The same study suggested that (5-TDMF) could block LPS-induced NF-κB translocation and iNOS and COX-2 expressions by inhibiting the mitogen-activated protein (MAP) kinase and MAPK/ERK signaling pathways. Suggesting potential novel chemo-preventive anti-inflammatory agents [38]. Still, a lack of detailed studies is noticed regarding its molecular mechanism that controls its activities, such as binding to a particular protein structure [64].
Hispidulin (HIS) (22.61%) is another detected compound considered a monomethoxy and trihydroxy flavone compound. In vitro investigations suggested the capability to affect the activation of JNK, p38, and NF-κB [65]. Also, it could control helenalin-induced cytotoxicity [66]. HIS has been reported in numerous studies to have potential antimutagenic, antioxidative, and anti-inflammatory effects [67][68][69]. Besides, many reported anticancer activity against multiple cancer cell lines such as gastric, pancreatic, ovarian, gallbladder, and colorectal. In addition to glioblastoma renal carcinoma, acute myeloid leukemia, Glioblastoma Multiforme, hepatocellular carcinoma cancers [69][70][71][72][73][74][75][76][77][78][79][80][81][82]. In colon cancer, HIS was noticed to inhibit the hypoxia-generated epithelial-mesenchymal transition, which had significantly enhanced the cytotoxicity of anticancer drugs against cancer cells [70].
Other detected components include Eupatilin (5,7-dihydroxy-3′,4′,6-trimethoxyflavone) (13.24%). That is known to possess promising antiproliferative, anti-inflammatory, modest antioxidant, neuroprotective, anti-allergic, and cardioprotective activities [37][83][84][85][86][87][88][89][90][91]. Several studies on eupatilin have explained its anti-cancer property due to its promising capacity to prompt apoptosis in different cancer cell lines [83][84][92][93][94][95][96][97][98]. Same as baicalein bioflavonoid (S7.84%), which can arrest cancerous cells at the G2/M and G1/S cell cycle phases [56]. It also decreases cell proliferation or induces apoptosis in multiple myeloma and cancer types. It has been demonstrated to inhibit cancer cell migration and invasion in many studies [99][100][101][102][103][104].
No available studies have been found related to 5,7-Dihydroxy-2′-Methoxyflavone (2′-Methoxychrysin) (4.91%) activity, although this compound has been separated previously from many species [105][106]. This compound has the physicochemical and pharmacological effects of flavonoids—flavones, and flavon-3-ols [107].
Previous reports revealed different types of flavonoids identified in A. citrodora as Skaltsa used column chromatography to isolate several flavonoids from the leaf extract of A. citrodora in 1988. Flavone structures were found in all of the purified substances [108]. Subsequently, glycosides of previously isolated flavones were also detected, such as apigenin-7-diglucuronide and chrysoeriol7-diglucuronide in the aqueous extract of A. citrodora aerial parts [109]. New flavonoids in the aerial parts (jaceosidin, nepetin, and nepitrin) have also been reported in recent studies, all of which have flavone structures [110].
The occurrence of these compounds simultaneously could indicate a synergistic, additive, or antagonistic effect on the antiproliferative activity of the extract. Ethyl acetate extract had the most potent antiproliferative effect on all used cancer cell lines (IC50 ranging from 136 to 203 μg/mL), even on the normal cell line, which in comparison with cancerous cells and depending on selectivity index, revealed selective toxicity (targeting malignant cells without harmful effect on normal cells) using suitable concentration.
The antiproliferative experiments of the EO of the aerial parts of A. citrodora revealed weak activity compared with other studies (IC50 ranging from 402 to 633 μg/mL). Oukerrou et al. (2017) demonstrated that A. citrodora EO exerted a dose-dependent cytotoxic effect on P815, MCF7, and Vero tumor cell lines, with IC50s ranging from 6.60 to 79.63 μg/mL [111]. Another study observed the potent cytotoxic effect of A. citrodora EO from two different regions of Palestine on HeLa cell lines. The IC50 values were 84.50 and 33.31 μg/mL and compared with Doxorubicin (IC50 value of 22.01 μg/mL) [48]. No notable activity was observed regarding ethanol, and aqueous extracts in this entry suggested the presence of weakly active polar compounds or antagonistic interaction between the phenolics and flavonoids components.

3.2. In Vivo Study

The ethyl acetate extract of A. citrodora was used in the current entry to treat mice implanted with breast cancer cells. A significant reduction in tumor size and a high cure percentage were observed. A. citrodora ethyl acetate extract dosage of 0.162 g/kg showed a high reduction in tumor size by 57.97% and many undetected tumors or curing effects of 44.44%. Reduction in the tumor size may be explained by the cytotoxic phytochemicals that exhibited notable effects on tumor cells.
Many studies showed the effect of phytochemical compounds detected in A. citrodora with cytoprotective, antioxidant, and anti-proliferation activities [54]. Naringenin, 5,6,4′-Trihydroxy-7,3′-Dimethoxyflavone (5- TDMF), and hispidulin were the most concentrated compounds in ethyl acetate extract. These compounds possess promising antiproliferative activities. In vivo studies with naringenin revealed anti-proliferation activity through a delay of tumor growth on ovariectomized C57BL/6 mice injected with E0771 mammary tumor cells [59]. In another in vivo study using breast cancer cells, observations suggested decreased secretion of TGF-β1 and accumulation of intracellular TGF-β1, and inhibition of TGF-β1 transport from the trans-Golgi network and PKC activity [112]. Also, 5,6,4′-Trihydroxy-7,3′-Dimethoxyflavone (5- TDMF), hispidulin in-vivo anti-proliferation activity, and antioxidant properties were reported [38][113]. These compounds may work synergistically to inhibit cancer either by a direct effect on cancer cells or by an indirect effect through antioxidant activity and other mechanisms [114].
Liver and kidney function enzymes such as ALT, AST, and creatinine are the most reflective parameters of toxicity and safety profile at the therapeutic doses because they are significant in eliminating the drugs through metabolism and excretion [115].
The ALT and AST for the treated group were within the normal range and lower than the normal group, which indicates an acceptable safety profile for all the treatments. These results could be justified because the doses used in this entry chose a dose according to LD50 estimation and used Karber method calculation with no toxic outcomes. On the other hand, creatinine levels observed in this entry were higher for the treatment group than the control and normal mice creatinine levels. High kidney enzymes are acceptable as long as they are within the range of normal mice enzymes level. Mainly after no deaths were observed after ten days of treatment, the results might indicate the safety of using the ethyl acetate extract.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249.
  2. MacConaill, L.E.; Garraway, L.A. Clinical implications of the cancer genome. J. Clin. Oncol. 2010, 28, 5219.
  3. Goldenberg, M.M. Trastuzumab, a recombinant DNA-derived humanized monoclonal antibody, a novel agent for the treatment of metastatic breast cancer. Clin. Ther. 1999, 21, 309–318.
  4. Yasueda, A.; Urushima, H.; Ito, T. Efficacy and interaction of antioxidant supplements as adjuvant therapy in cancer treatment: A systematic review. Integr. Cancer Ther. 2016, 15, 17–39.
  5. Molassiotis, A.; Fernadez-Ortega, P.; Pud, D.; Ozden, G.; Scott, J.A.; Panteli, V.; Margulies, A.; Browall, M.; Magri, M.; Selvekerova, S. Use of complementary and alternative medicine in cancer patients: A European survey. Ann. Oncol. 2005, 16, 655–663.
  6. Talib, W.H.; Mahmod, A.I.; Kamal, A.; Rashid, H.M.; Alashqar, A.; Khater, S.; Jamal, D.; Waly, M. Ketogenic Diet in Cancer Prevention and Therapy: Molecular Targets and Therapeutic Opportunities. Curr. Issues Mol. Biol. 2021, 43, 558–589.
  7. Goodman, L.S.; Wintrobe, M.M.; Dameshek, W.; Goodman, M.J.; Gilman, A.; McLennan, M.T. Nitrogen mustard therapy: Use of methyl-bis (beta-chloroethyl) amine hydrochloride and tris (beta-chloroethyl) amine hydrochloride for hodgkin’s disease, lymphosarcoma, leukemia and certain allied and miscellaneous disorders. J. Am. Med. Assoc. 1946, 132, 126–132.
  8. Mansoori, B.; Mohammadi, A.; Davudian, S.; Shirjang, S.; Baradaran, B. The different mechanisms of cancer drug resistance: A brief review. Adv. Pharm. Bull. 2017, 7, 339.
  9. Dubey, N.K. Plants as A Source of Natural Antioxidants; CABI: Varanasi, India, 2014.
  10. Verpoorte, R. Pharmacognosy in the new millennium: Leadfinding and biotechnology. J. Pharm. Pharmacol. 2000, 52, 253–262.
  11. Amin, M.; Anwar, F.; Naz, F.; Mehmood, T.; Saari, N. Anti-Helicobacter pylori and urease inhibition activities of some traditional medicinal plants. Molecules 2013, 18, 2135–2149.
  12. Rates, S.M.K. Plants as source of drugs. Toxicon 2001, 39, 603–613.
  13. Abu-Irmaileh, B.E.; Afifi, F.U. Herbal medicine in Jordan with special emphasis on commonly used herbs. J. Ethnopharmacol. 2003, 89, 193–197.
  14. Healthcare, T. PDR for Herbal Medicines, 4th ed.; PDR.; Thomson Reuters: Toronto, ON, Canada, 2007.
  15. Simon, J.E.; Chadwick, A.F. Herbs: An Indexed Bibliography 1971–1980 the Scientific Literature on Selected Herbs, and Aromatic and Medicinal Plants of the Temperate Zone, 1st ed.; Archon Books: Hamden, CT, USA, 1984.
  16. Ali, H.F.; El Beltagi, H.S.; Nasr, N. Assessment of volatile components, free radical-scavenging capacity and anti-microbial activity of lemon verbena leaves. Res. J. Phytochem. 2008, 2, 84–92.
  17. Funes, L.; Carrera-Quintanar, L.; Cerdán-Calero, M.; Ferrer, M.D.; Drobnic, F.; Pons, A.; Roche, E.; Micol, V. Effect of lemon verbena supplementation on muscular damage markers, proinflammatory cytokines release and neutrophils’ oxidative stress in chronic exercise. Eur. J. Appl. Physiol. 2011, 111, 695–705.
  18. Funes, L.; Fernández-Arroyo, S.; Laporta, O.; Pons, A.; Roche, E.; Segura-Carretero, A.; Fernández-Gutiérrez, A.; Micol, V. Correlation between plasma antioxidant capacity and verbascoside levels in rats after oral administration of lemon verbena extract. Food Chem. 2009, 117, 589–598.
  19. Schmid, R.; Bruneton, J.; Hatton, C.K. Pharmacognosy, phytochemistry, medicinal plants. Taxon 1995, 44, 469.
  20. Ono, M.; Oda, E.; Tanaka, T.; Iida, Y.; Yamasaki, T.; Masuoka, C.; Ikeda, T.; Nohara, T. DPPH radical-scavenging effect on some constituents from the aerial parts of Lippia triphylla. J. Nat. Med. 2008, 62, 101–106.
  21. Escobar, P.; Milena Leal, S.; Herrera, L.V.; Martinez, J.R.; Stashenko, E. Chemical composition and antiprotozoal activities of Colombian Lippia spp essential oils and their major components. Mem. Inst. Oswaldo Cruz 2010, 105, 184–190.
  22. Rojas, L.B.; Velasco, J.; Diaz, T.; Gil Otaiza, R.; Carmona, J.; Usubillaga, A. Chemical composition and antibacterial effect of Aloysia triphylla (L’ Hér.) Britton essential oil against genito-urinary pathogens. Lat. Am. Caribb. Bull. Med. Aromat. Plants 2010, 9, 56–62.
  23. Gil, A.; Van Baren, C.M.; Di Leo Lira, P.M.; Bandoni, A.L. Identification of the genotype from the content and composition of the essential oil of lemon verbena (Aloysia citriodora Palau). J. Agric. Food Chem. 2007, 55, 8664–8669.
  24. Dambolena, J.S.; Zunino, M.P.; Lucini, E.I.; Zygadlo, J.A.; Banchio, E.; Biurrun, F.; Rotman, A.; Ahumada, O. Aromatic plants of northwest Argentina. Constituents of the essential oils of aerial parts of seven Verbenaceae: Lantana and Aloysia. J. Essent. Oil Res. 2010, 22, 289–293.
  25. Liou, G.-Y.; Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 2010, 44, 479–496.
  26. Tošović, J.; Bren, U. Antioxidative action of ellagic acid—A kinetic DFT study. Antioxidants 2020, 9, 587.
  27. Rout, O.P.; Acharya, R.; Mishra, S.K. In-Vitro Antioxidant potentials in leaves of Coleus aromaticus Benth and rhizomes of Zingiber zerumbet (L.) SM. J. Appl. Pharm. Sci. 2011, 1, 194.
  28. Dziri, S.; Hassen, I.; Fatnassi, S.; Mrabet, Y.; Casabianca, H.; Hanchi, B.; Hosni, K. Phenolic constituents, antioxidant and antimicrobial activities of rosy garlic (Allium roseum var. odoratissimum). J. Funct. Foods 2012, 4, 423–432.
  29. Ismail, H.I.; Chan, K.W.; Mariod, A.A.; Ismail, M. Phenolic content and antioxidant activity of cantaloupe (Cucumis melo) methanolic extracts. Food Chem. 2010, 119, 643–647.
  30. Vallverdu-Queralt, A.; Medina-Remon, A.; Martínez-Huélamo, M.; Jauregui, O.; Andres-Lacueva, C.; Lamuela-Raventos, R.M. Phenolic profile and hydrophilic antioxidant capacity as chemotaxonomic markers of tomato varieties. J. Agric. Food Chem. 2011, 59, 3994–4001.
  31. Vallverdú-Queralt, A.; Regueiro, J.; Alvarenga, J.F.R.; Martinez-Huelamo, M.; Leal, L.N.; Lamuela-Raventos, R.M. Characterization of the phenolic and antioxidant profiles of selected culinary herbs and spices: Caraway, turmeric, dill, marjoram and nutmeg. Food Sci. Technol. 2015, 35, 189–195.
  32. Falah, S.; Suzuki, T.; Katayama, T. Chemical constituents from Swietenia macrophylla bark and their antioxidant activity. Pak. J. Biol. Sci. 2008, 11, 2007–2012.
  33. Katsarou, A.; Rhizopoulou, S.; Kefalas, P. Antioxidant potential of the aerial tissues of the mistletoe Loranthus europaeus Jacq. Rec. Nat. Prod. 2012, 6, 394.
  34. Mrvčić, J.; Posavec, S.; Kazazić, S.; Stanzer, D.; Peša, A.; Stehlik-Tomas, V. Spirit drinks: A source of dietary polyphenols. Croat. J. Food Sci. Technol. 2012, 4, 102–111.
  35. Stankovic, M.S. Total phenolic content, flavonoid concentration and antioxidant activity of Marrubium peregrinum L. extracts. Kragujev. J. Sci. 2011, 33, 63–72.
  36. Karim, N.; Jia, Z.; Zheng, X.; Cui, S.; Chen, W. A recent review of citrus flavanone naringenin on metabolic diseases and its potential sources for high yield-production. Trends Food Sci. Technol. 2018, 79, 35–54.
  37. Choi, E.-J.; Oh, H.-M.; Na, B.-R.; Ramesh, T.P.; Lee, H.-J.; Choi, C.-S.; Choi, S.-C.; Oh, T.-Y.; Choi, S.-J.; Chae, J.-R. Eupatilin protects gastric epithelial cells from oxidative damage and down-regulates genes responsible for the cellular oxidative stress. Pharm. Res. 2008, 25, 1355–1364.
  38. Wang, S.-H.; Liang, C.-H.; Liang, F.-P.; Ding, H.-Y.; Lin, S.-P.; Huang, G.-J.; Lin, W.-C.; Juang, S.-H. The inhibitory mechanisms study of 5, 6, 4′-trihydroxy-7, 3′-dimethoxyflavone against the LPS-induced macrophage inflammatory responses through the antioxidant ability. Molecules 2016, 21, 136.
  39. Olszowy-Tomczyk, M. Synergistic, antagonistic and additive antioxidant effects in the binary mixtures. Phytochem. Rev. 2020, 19, 63–103.
  40. Abuhamdah, R.; Mohammed, A.L.I. Chemical, Molecular Pharmacology and Neuroprotective Properties of the Essential Oil Derived from Aloysia citrodora Palau. Ph.D. Thesis, Durham University, Durham, UK, 2014. Available online: http://etheses.dur.ac.uk/10663/ (accessed on 1 January 2022).
  41. Abuhamdah, S.; Abuhamdah, R.; Howes, M.-J.R.; Al-Olimat, S.; Ennaceur, A.; Chazot, P.L. Pharmacological and neuroprotective profile of an essential oil derived from leaves of Aloysia citrodora Palau. J. Pharm. Pharmacol. 2015, 67, 1306–1315.
  42. Hudaib, M.; Tawaha, K.; Bustanji, Y. Chemical profile of the volatile oil of Lemon verbena (Aloysia citriodora Paláu) growing in Jordan. J. Essent. Oil Bear. Plants 2013, 16, 568–574.
  43. Argyropoulou, C.; Daferera, D.; Tarantilis, P.A.; Fasseas, C.; Polissiou, M. Chemical composition of the essential oil from leaves of Lippia citriodora HBK (Verbenaceae) at two developmental stages. Biochem. Syst. Ecol. 2007, 35, 831–837.
  44. Lira, P.D.L.; van Baren, C.M.; Retta, D.; Bandoni, A.L.; Gil, A.; Gattuso, M.; Gattuso, S. Characterization of Lemon verbena (Aloysia citriodora Palau) from Argentina by the essential oil. J. Essent. Oil Res. 2008, 20, 350–353.
  45. Bellakhdar, J.; Idrissi, A., II; Canigueral, S.; Iglesias, J.; Vila, R. Composition of lemon verbena (Aloysia triphylla (L’Herit.) Britton) oil of Moroccan origin. J. Essent. Oil Res. 1994, 6, 523–526.
  46. Ebadi, M.; Sefidkon, F.; Azizi, M.; Ahmadi, N. Packaging methods and storage duration affect essential oil content and composition of lemon verbena (Lippia citriodora Kunth.). Food Sci. Nutr. 2017, 5, 588–595.
  47. Santos-Gomes, P.C.; Fernandes-Ferreira, M.; Vicente, A.M.S. Composition of the essential oils from flowers and leaves of vervain grown in Portugal. J. Essent. Oil Res. 2005, 17, 73–78.
  48. Jaradat, N.; Hawash, M.; Abualhasan, M.N.; Qadi, M.; Ghanim, M.; Massarwy, E.; Ammar, S.A.; Zmero, N.; Arar, M.; Hussein, F. Spectral characterization, antioxidant, antimicrobial, cytotoxic, and cyclooxygenase inhibitory activities of Aloysia citriodora essential oils collected from two Palestinian regions. BMC Complement. Med. Ther. 2021, 21, 143.
  49. Knez Hrnčič, M.; Španinger, E.; Košir, I.J.; Knez, Ž.; Bren, U. Hop compounds: Extraction techniques, chemical analyses, antioxidative, antimicrobial, and anticarcinogenic effects. Nutrients 2019, 11, 257.
  50. Terblanché, F.C.; Kornelius, G. Essential oil constituents of the genus Lippia (Verbenaceae)—A literature review. J. Essent. oil Res. 1996, 8, 471–485.
  51. Montes, M.; Valenzuela, L.; Wilkomirsky, T.; Arrive, M. SUR LA COMPOSITION DE L’ESSENCE D’ALOYSIA TRIPHYLLA (“CEDRON”). Planta Med. 1973, 23, 119–124.
  52. Chizzola, R. Regular monoterpenes and sesquiterpenes (essential oils). Nat. Prod. 2013, 10, 973–978.
  53. Bendary, E.; Francis, R.R.; Ali, H.M.G.; Sarwat, M.I.; El Hady, S. Antioxidant and structure–activity relationships (SARs) of some phenolic and anilines compounds. Ann. Agric. Sci. 2013, 58, 173–181.
  54. Bahramsoltani, R.; Rostamiasrabadi, P.; Shahpiri, Z.; Marques, A.M.; Rahimi, R.; Farzaei, M.H. Aloysia citrodora Paláu (Lemon verbena): A review of phytochemistry and pharmacology. J. Ethnopharmacol. 2018, 222, 34–51.
  55. Santos, E.L.; BHLNS, M.; Ferriani, A.P.; Teixeira, S.D. Flavonoids-From Biosynthesis to Human Health; Books on Demand: Norderstedt, Germany, 2017.
  56. Brglez Mojzer, E.; Knez Hrnčič, M.; Škerget, M.; Knez, Ž.; Bren, U. Polyphenols: Extraction methods, antioxidative action, bioavailability and anticarcinogenic effects. Molecules 2016, 21, 901.
  57. Salehi, B.; Fokou, P.V.T.; Sharifi-Rad, M.; Zucca, P.; Pezzani, R.; Martins, N.; Sharifi-Rad, J. The therapeutic potential of naringenin: A review of clinical trials. Pharmaceuticals 2019, 12, 11.
  58. Yin, J.; Liang, Y.; Wang, D.; Yan, Z.; Yin, H.; Wu, D.; Su, Q. Naringenin induces laxative effects by upregulating the expression levels of c-Kit and SCF, as well as those of aquaporin 3 in mice with loperamide-induced constipation. Int. J. Mol. Med. 2018, 41, 649–658.
  59. Ke, J.; Banh, T.; Hsiao, Y.; Cole, R.M.; Straka, S.R.; Yee, L.D.; Belury, M.A. Citrus flavonoid naringenin reduces mammary tumor cell viability, adipose mass, and adipose inflammation in obese ovariectomized mice. Mol. Nutr. Food Res. 2017, 61, 1600934.
  60. Pinho-Ribeiro, F.A.; Zarpelon, A.C.; Fattori, V.; Manchope, M.F.; Mizokami, S.S.; Casagrande, R.; Verri, W.A., Jr. Naringenin reduces inflammatory pain in mice. Neuropharmacology 2016, 105, 508–519.
  61. Stompor, M.; Uram, Ł.; Podgórski, R. In vitro effect of 8-prenylnaringenin and naringenin on fibroblasts and glioblastoma cells-cellular accumulation and cytotoxicity. Molecules 2017, 22, 1092.
  62. Lim, W.; Park, S.; Bazer, F.W.; Song, G. Naringenin-induced apoptotic cell death in prostate cancer cells is mediated via the PI3K/AKT and MAPK signaling pathways. J. Cell. Biochem. 2017, 118, 1118–1131.
  63. Bouzaiene, N.N.; Chaabane, F.; Sassi, A.; Chekir-Ghedira, L.; Ghedira, K. Effect of apigenin-7-glucoside, genkwanin and naringenin on tyrosinase activity and melanin synthesis in B16F10 melanoma cells. Life Sci. 2016, 144, 80–85.
  64. Lešnik, S.; Bren, U. Mechanistic Insights into Biological Activities of Polyphenolic Compounds from Rosemary Obtained by Inverse Molecular Docking. Foods 2022, 11, 67.
  65. Nepal, M.; Choi, H.J.; Choi, B.-Y.; Yang, M.-S.; Chae, J.-I.; Li, L.; Soh, Y. Hispidulin attenuates bone resorption and osteoclastogenesis via the RANKL-induced NF-κB and NFATc1 pathways. Eur. J. Pharmacol. 2013, 715, 96–104.
  66. Woerdenbag, H.J.; Merfort, I.; Schmidt, T.J.; Passreiter, C.M.; Willuhn, G.; Van Uden, W.; Pras, N.; Konings, A.W.T. Decreased helenalin-induced cytotoxicity by flavonoids from Arnica as studied in a human lung carcinoma cell line. Phytomedicine 1995, 2, 127–132.
  67. Kavvadias, D.; Sand, P.; Youdim, K.A.; Qaiser, M.Z.; Rice-Evans, C.; Baur, R.; Sigel, E.; Rausch, W.; Riederer, P.; Schreier, P. The flavone hispidulin, a benzodiazepine receptor ligand with positive allosteric properties, traverses the blood–brain barrier and exhibits anticonvulsive effects. Br. J. Pharmacol. 2004, 142, 811–820.
  68. Khamar, D.; Devkar, R.; Reshma, K.K.; Shreedhara, C.S.; Setty, M.M.; Hegde, S. Enhanced hispidulin production in vitro from callus culture of millingtonia hortensis L.F. IJPBS 2013, 2, 633–639.
  69. Lin, Y.-C.; Hung, C.-M.; Tsai, J.-C.; Lee, J.-C.; Chen, Y.-L.S.; Wei, C.-W.; Kao, J.-Y.; Way, T.-D. Hispidulin potently inhibits human glioblastoma multiforme cells through activation of AMP-activated protein kinase (AMPK). J. Agric. Food Chem. 2010, 58, 9511–9517.
  70. Wang, Y.; Liu, W.; He, X.; Fei, Z. Hispidulin enhances the anti-tumor effects of temozolomide in glioblastoma by activating AMPK. Cell Biochem. Biophys. 2015, 71, 701–706.
  71. Gao, M.-Q.; Gao, H.; Han, M.; Liu, K.-L.; Peng, J.-J.; Han, Y.-T. Hispidulin suppresses tumor growth and metastasis in renal cell carcinoma by modulating ceramide-sphingosine 1-phosphate rheostat. Am. J. Cancer Res. 2017, 7, 1501.
  72. Gao, H.; Jiang, Q.; Han, Y.; Peng, J.; Wang, C. Hispidulin potentiates the antitumor effect of sunitinib against human renal cell carcinoma in laboratory models. Cell Biochem. Biophys. 2015, 71, 757–764.
  73. Gao, H.; Gao, M.; Peng, J.; Han, M.; Liu, K.; Han, Y. Hispidulin mediates apoptosis in human renal cell carcinoma by inducing ceramide accumulation. Acta Pharmacol. Sin. 2017, 38, 1618.
  74. Yu, C.Y.; Su, K.-Y.; Lee, P.-L.; Jhan, J.-Y.; Tsao, P.-H.; Chan, D.-C.; Chen, Y.-L.S. Potential therapeutic role of hispidulin in gastric cancer through induction of apoptosis via NAG-1 signaling. Evid. Based Complement. Altern. Med. 2013, 2013, 518301.
  75. He, L.; Wu, Y.; Lin, L.; Wang, J.; Wu, Y.; Chen, Y.; Yi, Z.; Liu, M.; Pang, X. Hispidulin, a small flavonoid molecule, suppresses the angiogenesis and growth of human pancreatic cancer by targeting vascular endothelial growth factor receptor 2-mediated PI3K/Akt/mTOR signaling pathway. Cancer Sci. 2011, 102, 219–225.
  76. Yang, J.-M.; Hung, C.-M.; Fu, C.-N.; Lee, J.-C.; Huang, C.-H.; Yang, M.-H.; Lin, C.-L.; Kao, J.-Y.; Way, T.-D. Hispidulin sensitizes human ovarian cancer cells to TRAIL-induced apoptosis by AMPK activation leading to Mcl-1 block in translation. J. Agric. Food Chem. 2010, 58, 10020–10026.
  77. Gao, H.; Wang, H.; Peng, J. Hispidulin induces apoptosis through mitochondrial dysfunction and inhibition of P13k/Akt signalling pathway in HepG2 cancer cells. Cell Biochem. Biophys. 2014, 69, 27–34.
  78. Han, M.; Gao, H.; Ju, P.; Gao, M.; Yuan, Y.; Chen, X.; Liu, K.; Han, Y.; Han, Z. Hispidulin inhibits hepatocellular carcinoma growth and metastasis through AMPK and ERK signaling mediated activation of PPARγ. Biomed. Pharmacother. 2018, 103, 272–283.
  79. Gao, H.; Xie, J.; Peng, J.; Han, Y.; Jiang, Q.; Han, M.; Wang, C. Hispidulin inhibits proliferation and enhances chemosensitivity of gallbladder cancer cells by targeting HIF-1α. Exp. Cell Res. 2015, 332, 236–246.
  80. Gao, H.; Liu, Y.; Li, K.; Wu, T.; Peng, J.; Jing, F. Hispidulin induces mitochondrial apoptosis in acute myeloid leukemia cells by targeting extracellular matrix metalloproteinase inducer. Am. J. Transl. Res. 2016, 8, 1115.
  81. Xie, J.; Gao, H.; Peng, J.; Han, Y.; Chen, X.; Jiang, Q.; Wang, C. Hispidulin prevents hypoxia-induced epithelial-mesenchymal transition in human colon carcinoma cells. Am. J. Cancer Res. 2015, 5, 1047.
  82. Liu, K.; Gao, H.; Wang, Q.; Wang, L.; Zhang, B.; Han, Z.; Chen, X.; Han, M.; Gao, M. Retracted: Hispidulin suppresses cell growth and metastasis by targeting PIM 1 through JAK 2/STAT 3 signaling in colorectal cancer. Cancer Sci. 2018, 109, 1369–1381.
  83. Kim, M.-J.; Kim, D.-H.; Na, H.-K.; Oh, T.Y.; Shin, C.-Y.; Surh, Y.-J. Eupatilin, a pharmacologically active flavone derived from Artemisia plants, induces apoptosis in human gastric cancer (AGS) cells. J. Environ. Pathol. Toxicol. Oncol. 2005, 24, 261–269.
  84. Seo, H.-J.; Surh, Y.-J. Eupatilin, a pharmacologically active flavone derived from Artemisia plants, induces apoptosis in human promyelocytic leukemia cells. Mutat. Res. Toxicol. Environ. Mutagen. 2001, 496, 191–198.
  85. Kim, J.; Kim, Y.; Yi, H.; Jung, H.; Rim, Y.A.; Park, N.; Jung, S.M.; Park, S.-H.; Ju, J.H. Eupatilin ameliorates collagen induced arthritis. J. Korean Med. Sci. 2015, 30, 233–239.
  86. Choi, E.-J.; Lee, S.; Chae, J.-R.; Lee, H.-S.; Jun, C.-D.; Kim, S.-H. Eupatilin inhibits lipopolysaccharide-induced expression of inflammatory mediators in macrophages. Life Sci. 2011, 88, 1121–1126.
  87. Cai, M.; Phan, P.-T.T.; Hong, J.G.; Kim, D.H.; Kim, J.M.; Park, S.J.; Liu, X.; Han, J.E.; Park, H.; Choi, J.W. The neuroprotective effect of eupatilin against ischemia/reperfusion-induced delayed neuronal damage in mice. Eur. J. Pharmacol. 2012, 689, 104–110.
  88. Kim, J.Y.; Kwon, E.Y.; Lee, Y.S.; Kim, W.B.; Ro, J.Y. Eupatilin blocks mediator release via tyrosine kinase inhibition in activated guinea pig lung mast cells. J. Toxicol. Environ. Health Part A 2005, 68, 2063–2080.
  89. Qiao, Z.; Xu, Y.; Yang, J. Eupatilin inhibits the apoptosis in H9c2 cardiomyocytes via the Akt/GSK-3β pathway following hypoxia/reoxygenation injury. Biomed. Pharmacother. 2016, 82, 373–378.
  90. Wei, Q.; Ji, X.Y.; Long, X.S.; Li, Q.R.; Yin, H. Chemical constituents from leaves of “chuju” Chrysanthemum morifolium and their antioxidant activities in vitro. Zhong Yao Cai Zhongyaocai J. Chin. Med. Mater. 2015, 38, 305–310.
  91. Ono, M.; Tsuru, T.; Abe, H.; Eto, M.; Okawa, M.; Abe, F.; Kinjo, J.; Ikeda, T.; Nohara, T. Bisabolane-type sesquiterpenes from the aerial parts of Lippia dulcis. J. Nat. Prod. 2006, 69, 1417–1420.
  92. Lee, S.; Lee, M.; Kim, S.-H. Eupatilin inhibits H2O2-induced apoptotic cell death through inhibition of mitogen-activated protein kinases and nuclear factor-κB. Food Chem. Toxicol. 2008, 46, 2865–2870.
  93. Park, B.B.; Yoon, J.S.; Kim, E.S.; Choi, J.; Won, Y.-W.; Choi, J.H.; Lee, Y.Y. Inhibitory effects of eupatilin on tumor invasion of human gastric cancer MKN-1 cells. Tumor Biol. 2013, 34, 875–885.
  94. Zhong, W.; Wang, X.; Pan, B.; Li, F.; Kuang, L.; Su, Z. Eupatilin induces human renal cancer cell apoptosis via ROS-mediated MAPK and PI3K/AKT signaling pathways. Oncol. Lett. 2016, 12, 2894–2899.
  95. Park, S.C.; Yoon, J.-H.; Kim, W.; Gwak, G.-Y.; Kim, K.M.; Lee, S.-H.; Lee, S.-M.; Lee, H.-S. Eupatilin attenuates bile acid-induced hepatocyte apoptosis. J. Gastroenterol. 2006, 41, 772–778.
  96. Li, Y.; Wu, H.; Dong, Y.; Lin, B.O.; Xu, G.; Ma, Y. Application of eupatilin in the treatment of osteosarcoma. Oncol. Lett. 2015, 10, 2505–2510.
  97. Wang, Y.; Hou, H.; Li, M.; Yang, Y.; Sun, L. Anticancer effect of eupatilin on glioma cells through inhibition of the Notch-1 signaling pathway. Mol. Med. Rep. 2016, 13, 1141–1146.
  98. Al Shawi, A.; Rasul, A.; Khan, M.; Iqbal, F.; Tonghui, M. Eupatilin: A flavonoid compound isolated from the artemisia plant, induces apoptosis and G2/M phase cell cycle arrest in human melanoma A375 cells. Afr. J. Pharm. Pharmacol. 2011, 5, 582–588.
  99. Bonham, M.; Posakony, J.; Coleman, I.; Montgomery, B.; Simon, J.; Nelson, P.S. Characterization of chemical constituents in Scutellaria baicalensis with antiandrogenic and growth-inhibitory activities toward prostate carcinoma. Clin. Cancer Res. 2005, 11, 3905–3914.
  100. Chiu, Y.-W.; Lin, T.-H.; Huang, W.-S.; Teng, C.-Y.; Liou, Y.-S.; Kuo, W.-H.; Lin, W.-L.; Huang, H.-I.; Tung, J.-N.; Huang, C.-Y. Baicalein inhibits the migration and invasive properties of human hepatoma cells. Toxicol. Appl. Pharmacol. 2011, 255, 316–326.
  101. Ma, Z.; Otsuyama, K.; Liu, S.; Abroun, S.; Ishikawa, H.; Tsuyama, N.; Obata, M.; Li, F.-J.; Zheng, X.; Maki, Y. Baicalein, a component of Scutellaria radix from Huang-Lian-Jie-Du-Tang (HLJDT), leads to suppression of proliferation and induction of apoptosis in human myeloma cells. Blood 2005, 105, 3312–3318.
  102. Tong, W.-G.; Ding, X.-Z.; Adrian, T.E. The mechanisms of lipoxygenase inhibitor-induced apoptosis in human breast cancer cells. Biochem. Biophys. Res. Commun. 2002, 296, 942–948.
  103. Wu, B.; Li, J.; Huang, D.; Wang, W.; Chen, Y.; Liao, Y.; Tang, X.; Xie, H.; Tang, F. Baicalein mediates inhibition of migration and invasiveness of skin carcinoma through Ezrin in A431 cells. BMC Cancer 2011, 11, 527.
  104. Zhang, D.Y.; Wu, J.; Ye, F.; Xue, L.; Jiang, S.; Yi, J.; Zhang, W.; Wei, H.; Sung, M.; Wang, W. Inhibition of cancer cell proliferation and prostaglandin E2 synthesis by Scutellaria baicalensis. Cancer Res. 2003, 63, 4037–4043.
  105. Chemesova, I.I.; Iinuma, M.; Budantsev, A.L. Investigation of the flavonoid composition of Scutellaria adenostegia. Chem. Nat. Compd. 1993, 29, 133–134.
  106. Popova, T.P.; Lytvynenko, V.I.; Pakaln, D.A. Phenol compound study of populations of Scutellaria sevanensis and related species. Farm. Zh. 1979, 6, 49–53.
  107. Azimova, S.S.; Vinogradova, V.I. Natural Compounds: Flavonoids; Springer: Berlin/Heidelberg, Germany, 2013.
  108. Skaltsa, H.; Shammas, G. Flavonoids from Lippia citriodora. Planta Med. 1988, 54, 465.
  109. Quirantes-Piné, R.; Funes, L.; Micol, V.; Segura-Carretero, A.; Fernández-Gutiérrez, A. High-performance liquid chromatography with diode array detection coupled to electrospray time-of-flight and ion-trap tandem mass spectrometry to identify phenolic compounds from a lemon verbena extract. J. Chromatogr. A 2009, 1216, 5391–5397.
  110. Zhang, Y.; Chen, Y.; Wang, S.; Dong, Y.; Wang, T.; Qu, L.; Li, N.; Wang, T. Bioactive constituents from the aerial parts of Lippia triphylla. Molecules 2015, 20, 21946–21959.
  111. Oukerrou, M.A.; Tilaoui, M.; Mouse, H.A.; Leouifoudi, I.; Jaafari, A.; Zyad, A. Chemical composition and cytotoxic and antibacterial activities of the essential oil of Aloysia citriodora palau grown in Morocco. Adv. Pharmacol. Sci. 2017, 2017, 7801924.
  112. Zhang, F.; Dong, W.; Zeng, W.; Zhang, L.; Zhang, C.; Qiu, Y.; Wang, L.; Yin, X.; Zhang, C.; Liang, W. Naringenin prevents TGF-β1 secretion from breast cancer and suppresses pulmonary metastasis by inhibiting PKC activation. Breast Cancer Res. 2016, 18, 38.
  113. Ashaq, A.; Maqbool, M.F.; Maryam, A.; Khan, M.; Shakir, H.A.; Irfan, M.; Qazi, J.I.; Li, Y.; Ma, T. Hispidulin: A novel natural compound with therapeutic potential against human cancers. Phyther. Res. 2021, 35, 771–789.
  114. Seca, A.M.L.; Pinto, D.C.G.A. Plant secondary metabolites as anticancer agents: Successes in clinical trials and therapeutic application. Int. J. Mol. Sci. 2018, 19, 263.
  115. Oduola, T.; Bello, I.; Adeosun, G.; Ademosun, A.-W.; Raheem, G.; Avwioro, G. Hepatotoxicity and nephrotoxicity evaluation in Wistar albino rats exposed to Morinda lucida leaf extract. N. Am. J. Med. Sci. 2010, 2, 230.
More
Information
Subjects: Biology; Plant Sciences
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 806
Revisions: 2 times (View History)
Update Date: 21 Mar 2022
1000/1000