Volume 8, Issue 3 (2022)                   Pharm Biomed Res 2022, 8(3): 205-224 | Back to browse issues page


XML Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

Rahamouz Haghighi S, Yazdinezhad A, Bagheri K, Sharafi A. Volatile Constituents and Toxicity of Essential Oils Extracted From Aerial Parts of Plantago Lanceolata and Plantago Major Growing in Iran. Pharm Biomed Res 2022; 8 (3) :205-224
URL: http://pbr.mazums.ac.ir/article-1-458-en.html
1- Department of Plant Production and Genetics, Faculty of Agriculture, University of Zanjan, Zanjan, Iran.
2- Department of Pharmacognosy, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran.
3- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
Full-Text [PDF 2375 kb]   (734 Downloads)     |   Abstract (HTML)  (1270 Views)
Full-Text:   (748 Views)
Introduction
Essential oils are used as additives in many types of foods and beverages and various food supplements [1]. The Plantago genus of the Plantaginaceae family includes approximately 300 annual and perennial species, growing worldwide, and specially cultivated in the subtropical regions [2]. According to Iran’s traditional medicine, Plantago species have many medical applications without serious side effects; however, some of the medicinal effects of Plantago lanceolata L. (P. lanceolata) and Plantago major L. (P.major) in Iran’s traditional medicine have not been discovered in modern medicine [3]. 
P. lanceolata and P. major are used to treat wounds, infectious diseases, digestive and respiratory problems, fever, pain, dermatitis, and tumors [4, 5]. Furthermore, Plantago species were used to cure burns, ulcers, and eye diseases, as anti-inflammatory, antipyretic agents, anti-tussive, and purgative for snakebites [6]. Researchers have also reported that P.major mucilage can optimize the drug release in propranolol buccoadhesive tablets [7]. Additionally, they can be used in cosmetics to produce face masks, creams, or lotions for acne-prone and oily skins because of their astringent, anti-septic, and anti-bacterial properties [6]. 
GC/MS is one of the most important instruments used to analyze a sample with volatile constituents as it combines both the chromatographic technique for the efficient separation of sample constituents and mass spectroscopy that identifies the compounds according to their mass-to-charge ratio (m/z) [8]. The above-mentioned properties of these plants provide us with significant reasons to analyze their volatile composition. To date, only a few Plantago species have been investigated for their chemical constituents and biological activities of extracts. Previous studies on the chemical investigation of Plantago L. leaves and seeds extracts demonstrated the presence of polysaccharides, phenolic acids, flavonoids, iridoid glycosides, and vitamins [2]. 
There are few valid studies on the essential oil compositions of P. lanceolata and P. major, considering that these plants contain very small amounts of essential oil. Therefore, in the current study, following our previous studies on these plants, their essential oil compositions were examined. In addition, we evaluated the toxicity effects of the essential oils on colon cancer cells and Artemia salina (A.salina). To the best of our knowledge, there are no reports on the cytotoxicity assay of P. lanceolata and P. major essential oils on colon cancer cell lines.

Materials and Methods
Herbal material 

The aerial parts (leaf and stem) of P. lanceolata and P. major were collected from Zanjan Province, Iran (the geographical coordinates of the collection sites are as follows: 36°41’15.5”N 48°24’02.2”E). The taxonomic identity of species was authenticated at the Department of Botany, University of Zanjan, Iran. All sections were cut into small pieces and were dried in shade and at room temperature separately for one week. 

Isolation of essential oils 
The aerial parts of P. lanceolata and P. major (100 g) were ground to a coarse powder and extracted with 1500 mL of distilled water for hydrodistillation in a Clevenger-type apparatus for 5 to 6 h to arise the volatile composition in the form of essential oils. The essential oils were collected into 1 mL of n-pentane and then poured into a glass and stored at 4°C until further analysis [1]. 

Gas chromatography-mass spectrometry analysis 
The essential oils of the aerial parts of P. lanceolata and P. major were used for GC/MS analysis. GC/MS analysis was performed using the Agilent technologies 5975c. GC/MS analysis was carried out by 1 µL of the materials subjected to analysis. The GC/MS system has been equipped with a capillary column (30 m×250 µm×0.25 µm, Agilent). Helium as the carrier gas was used at the flow rate of (1 mL/min). The injector and the interface temperature were maintained at 250°C. The column temperature was programmed as follows: the initial temperature was 40°C (1 min) and then it increased at a rate of 2°C/min up to 200°C (10 min). The identification of the constituents of P. lanceolata and P. major was performed by comparison with MS literature data (NIST08.L) and retention index (RI) [1]. The mixtures of n-alkanes (C8-C20) were injected using the above temperature program to calculate the RI for each peak. The RI of the compounds was calculated using the following equation

Where: (Ix) is the Kovats retention index; (n) is the number of carbon atoms in the alkane; (tn) and (tn+1) are the retention times of the reference n-alkane hydrocarbons with n and n + 1 carbon atoms; and (tx) is the retention time of the peak of the unknown compound. 
Several peaks did not have RIs for the calculated mixtures of n-alkanes (C8-C20). Thus, compounds with a formula structure less than C8 and more than C20 could not be calculated (these compounds were considered unknown). 

Cell line culture
Human embryonic kidney cell (HEK-293) as a normal cell line and colorectal cancer cell line (HCT-116) provided by the Pasteur Institute of Iran, Tehran were cultured in the Dulbecco’s Modified Eagle Medium with supplementation of penicillin-streptomycin (1%) along with 10% fetal bovine serum incubated in 5% CO2 incubator at 37°C.

Cytotoxicity assay
The MTT assay was performed to evaluate the cytotoxicity of P. lanceolata and P. major essential oils on the cell lines [9]. A 96-well plate with a density of 7 × 103 cells/well were used for cell seeding. The cells were allowed to attach and grow for 24 h. The cells underwent treatment with 25-400 µg/mL concentrations. The HCT-116 were treated with 5-fluorouracil (5-FU) (Austria, Ebewe Pharma) in different doses (2.5-10 μg/mL) for 72 h. The 5-FU and untreated cells were utilized as the positive and negative control, respectively. The addition and incubation of 20 μL of MTT (5 mg/mL) for 4 h took place after 24 to 72 h, followed by removing the medium and adding 200 μL of dimethyl sulfoxide to dissolve the obtained formazan. An ELISA plate reader (Tecan Infinite M200, Austria) at 570 and 690 nm read the absorbance. The cell growth inhibition rates were examined by the following formula:


Toxicity assay on artemia salina
The larvae of brine shrimp (A.salina Leach) were employed to examine the P. lanceolata and P. major essential oils’ overall toxicity [10]. A. salina eggs were provided by Urmia University, the West Azerbaijan Province, Iran. A flask with 35 g of NaCl dissolved in 1 L of distilled water was used for cyst culture, followed by 48 h incubation at 28°C and the larvae hatching after 48 h. Every well in the 96-well microtiter plates having the Roswell Park Memorial Institute (RPMI-1640) received the essential oils (25-400 µg/mL). The next step included the addition of 10 nauplii per well to the 96-well plates and incubation at a temperature of 25°C for 24 h. A binocular microscope was employed to calculate the number of live nauplii in every well after 24 h. All experiments were repeated 3 times. Additionally, the negative control contained only 10 nauplii and artificial seawater. Potassium dichromate (K2Cr2O7) was used as a positive control at the same concentrations as the essential oils. The number of survived samples in the experimental and control wells was used to calculate the percentages of the nauplii morality. The Abbott formula determined the lethality:

Statistical analysis
The data were analyzed using the SPSS software, version 21. The significant differences between means were calculated. Values were expressed as the mean of the 3 replications ± Standard Deviation (SD). The Duncan test at P value<0.05 was used to determine significant differences among treatments. IC50 and LC50 values were analyzed with the ED50 plus v1.0 Software.

Results 
Many peaks were detected in the chromatogram of the essential oils extracted from P. lanceolata and P. major aerial parts by GC/MS and their compositions were identified according to the NIST08.L library. Figure 1 shows the main chromatograms of the essential oils of P.

lanceolata and P. major. The essential oils were rich in amine derivations, alcohols, alkenes, and fatty acids. The essential oils also showed the presence of acids, alkaloids, amino acids, carboxylic acid derivatives, esters, ketones, monoterpenoids, nitriles, oximes, phenols, phenethylamine derivatives, and others (Table 1).

Volatile constituents of p. lanceolata essential oil
Most component of P. lanceolata essential oil is generated by metaraminol (14.04%), bifemelane (8.73%), metossamina (8.16%), and pterin-6-carboxylic acid (5.11%).
In the present study, 106 components belonging to main chemical groups were identified in P. lanceolata essential oil: alcohols (17.56%) with benzyl alcohol; .α.-(1-aminoethyl)-m-hydroxy-, (-)-(14.04) as the main component; amines (14.70%) with phenylephrine (3.71%); alkenes and alkenes (12.28%) with bifemelane (8.73%); ketones (8.70%) with bicyclo [2.2.1] heptan-2-one, 4,7,7-trimethyl-, semicarbazone (2.97%); acids (8.05%) with pterin-6-carboxylic acid (5.11%); alkaloids (5.76%) with 2H-1,2,3-triazole-4-carboxylic acid; 2-(2-fluorophenyl)- (2.12%); esters (4.02) with 2-thiopheneacetic acid; 3,5-difluorophenyl ester (1.53%); amides (3.55%) with propanamide (0.58%); amino acids (2.71%) with histidine; 1, N-dimethyl-4-nitro- (1.76%); monoterpenoids (2.45%) with Linalool (0.97%); phenol (Benzeneethanamine, 2-fluoro-.beta.,5-dihydroxy-N-methyl-) (0.45%); nitriles (0.21%) with propanenitrile, 3-(methylamino)- (0.17%); oximes with ethanone, 1-(4-pyridinyl)-, oxime (0.13%) as the main components and others (21.03%) (Table 2 and 3).

The biological activities of the volatile constituents of P. lanceolata oil are reported in Table 4.

Volatile constituents of the essential oils of p. major
The present study showed that 2-dodecen-1-yl (-) succinic anhydride (15.29%), benzenemethanol,. α.-(1-aminoethyl)-2,5-dimethoxy- (11.83%), dl-phenylephrine (7.51%), nortriptyline (5.15%) were the major constituents (Tables 2 and 3).
In the present study, 79 components belonging to main chemical groups were identified in P. major essential oil: amines (35.74%) with phenylephrine (11.66%) as the main component; alkenes and alkanes (24.88%) with 2-dodecen-1-yl(-)succinic anhydride (15.29%); phenols (10.49%) with dl-phenylephrine (7.51%); esters (6.96%) with sarcosine, N-valeryl-, butyl ester (2.02%); alcohols (5.14%) with cyclobutanol, 2-ethyl- (1.72%); alkaloids (3.97%) with ethylamine, 2-(adamantan-1-yl)-1-methyl- (0.28%); ketones (3.61%) with 3-(E)-hexen-2-one, (5S)-5-[(t-butoxycarbonyl-(R)-alanyl)amino]- (2.65%); amides (2.2%) with [(2,5-dimethoxyphenyl)sulfonyl]ethylamine (0.69%); monoterpenes with isoborneol (1.17%); amino acids (glycine, N-(N-L-alanylglycyl)-) (0.35%) and acid (0.16%) with imidazole-5-carboxylic acid, 2-amino- as the main component. P.major essential oil has many properties and applications that are provided in Table 4.
The essential oils of P. lanceolata and P. major species showed that the predominant compounds were present in both species; however, the amounts of these compounds (%) were different. For example, (-)-Benzyl alcohol, .α.-(1-aminoethyl)-m-hydroxy (14.04% and 1.37%), metossamina (8.16% and 0.17%), benzenemethanol, .α.- (1-aminoethyl) -2,5-dimethoxy- (3.71% and 11.66%), dl-phenylephrine (0.15% and 7.51%), nortriptyline (0.95% and 5.15%) were present in P. lanceolata and P. major, respectively (Figure 2).

Bifemelane (% 8.73), pterin-6-carboxylic acid (5.11%) existed only in P. lanceolata while 2-dodecen-1-yl (-) succinic anhydride (15.29%) were only found in P. major.

Cytotoxic activities
Colorectal cancer cells were incubated after treatment with essential oils to study the cytotoxic activities of P. lanceolata and P. major. The essential oils of P. major exhibited more antiproliferative properties on HCT-116 at 72 h compared to P. lanceolata (IC50: 102.66 µg/mL). IC50 values showed that P. major essential oil had a greater cytotoxic effect on HCT-116 than HEK-293; however, P. lanceolata showed almost the same effect on cancer and normal cells (Table 5). The results indicated that a very low IC50 of 5-FU (4.136 µg/mL) was required to inhibit HCT-116 cell viability compared to the essential oil of P. lanceolata and P. major. 

Toxicity assay on artemia salina
The general toxicity of the essential oils was assessed against A. salina. At 25-100 µg/mL of the essential oils, all of the nauplii were alive, indicating no toxicity (LC50:2242.57 µg/mL and 1783.7 µg/mL) (Table 5). At 400 µg/mL of P. lanceolata and P. major, the percentage of lethality was 8% and 12%, respectively. Although, the K2Cr2O7 has shown to have a toxic effect (LC50 of 58.22 μg/mL).

volatile constituents in the essential oil, respectively. In their study, the main aroma constituents of P. lanceolata leaves were groups of fatty acids 28.0% – 52.1% (the most abundant palmitic acid 15.3% –32.0%), oxidated monoterpenes 4.3% – 13.2% with linalool 2.7% – 3.5%, ketones and aldehydes 6.9%–10.0% with pentyl vinyl ketone 2.0% –3.4%, and alcohols 3.8%–9.2% with 1-octen-3-ol 2.4%–8.2%. They pointed out that apocarotenoids (1.5%–2.3%) are the important constituents because of their intense fragrance and they were identified in a relatively high amount. The importance is in its potential manufacture control of raw material to supply food supplements [1]. The high content of 1-octen-3-ol (up to 8.2%) has been observed in the Bajer et al., 2016 study [1] in accordance with Fons [76]. This compound in the present study was about 1.27%.
Other studies showed that P. major essential oil has anti-tumor and anti-cancer activities because octodrine [28] and 1-[α-(1-adamantyl) benzylidene] thiosemicarbazide [54] were present in P. major essential oil. The anti-microbial components, i.e., 2-dodecen-1-yl(-) succinic anhydride [12]; 2-chloroacetamide [17]; isoborneol [23]; octodrine [30]; actinobolin [49]; 1-[α-(1-adamantyl) benzylidene] thiosemicarbazide [54]; cyclobutanol, 2-ethyl- [64]; antiviral compounds, including isoborneol [22]; 1-[α-(1-adamantyl) benzylidene] thiosemicarbazide [54]; antioxidant and anti-inflammatory compounds, such as 2-dodecen-1-yl(-)succinic anhydride [12]; desmethyldoxepin [56] and 1-[α-(1-adamantyl) benzylidene] thiosemicarbazide [54] were observed in the analysis of P. major essential oil. Some of the compounds identified in the analysis of the P. major essential oil showed important characteristics, such as cycloserine [71] and actinobolin [49] which are antibiotic drugs (0.75% and 0.13%) and isoborneol is anti-infective (1.17%) [22] (Table 4). The percentage and differences in the amount of these compounds depend on many factors, such as climatic conditions, type of region, plant growth conditions, and harvesting methods.
The present study indicated that a very low IC50 value of 5-FU was required to inhibit HCT-116 cell viability compared to the essential oil of P. lanceolata and P. major. However, the IC50 obtained for the essential oil of P.lanceolata and P.major were valuable and has increasingly important medical applications. Our previous studies reported the cytotoxic effects of alcoholic and acetonic extracts of P.major leaf and root on HCT-116 and HEK-293. The P. major root extract was more effective than the aerial parts, and IC50 values for ethanolic, methanolic, and acetonic root extracts were 405.59, 470.16, and 82.26 μg/mL, respectively on HCT-116 at 72 h [77]. In a study by Velasco-Lezama (2006), the cytotoxic activity of P. major methanolic extract has been reported on HCT-15 [78].
For the lethality of nauplii, if LC50, detected for each sample, is more than 1000 µg/mL, it will be non-toxic [79]. At 400 µg/mL of P. lanceolata and P. major, the percentage of the lethality of nauplii was 8% and 12%, respectively. Thus, the essential oils were not toxic. 
Other researchers have also evaluated the toxicity effect of P. major methanolic extract on A. salina and A. uramiana with LC50 of 303.7 μg/mL [80]. The LC50 values of Plantago squarrosa Murray extracts were more than 1000 μg/mL; therefore, the extracts were non-toxic in the Artemia franciscana bioassay [81]. Our previous study showed that at all concentrations of ethanolic extracts of P.major aerial parts and roots, no toxicity was observed [77]. 

Conclusions
Given the non-aromatic nature of P. lanceolata and P. major and the very small amount of essential oil in these plants, most phytochemical studies are usually performed on their extracts. Therefore, in the present study, the essential oils analysis of two well-known species of Plantago was conducted to discover the valuable compositions. The hydrodistillation method enabled us to gain a great number of volatile constituents, which is evident from the number of peaks that occurred in chromatograms. The most abundant family of compounds was amines. There were also identified acids, alcohols, alkaloids, alkanes, alkenes, amides, amino acids, esters, ketones, phenols, and terpenes that most of the terpenes were oxidated as monoterpenes. On the other hand, nitriles, oximes, and organic compounds were found in a relatively small amount. 
Regarding the chemical compounds identified in the P. lanceolata and P. major essential oils, these components could be employed as an important economical source in the pharmaceutical and chemical industries. We intend to study their biological activities in the future.

Ethical Considerations
Compliance with ethical guidelines

There were no ethical considerations to be considered in this research.

Funding
The paper was extracted from the PhD. Dissertation of the first author at Department of Plant production and Genetics, Faculty of Agriculture, Zanjan University of Medical Sciences (Grant number: A-12-848-35).

Authors' contributions
project administration, investigation, formal analysis, and writing-original draft: Samaneh Rahamouz-Haghighi; formal analysis, methodology, and validation: Alireza Yazdinezhad; Funding and supervision: Khadijeh Bagheri; Funding, supervision, conceptualization, and editing of the English version of the manuscript: Ali Sharafi. 

Conflict of interest
The authors declare that there are no conflicts of interest regarding the publication of this article.

Acknowledgments
This work was supported by Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran (Grant number: A-12-848-35). In addition, the authors would like to thank the authority of the School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran.

References
  1. Bajer T, Janda V, Bajerová P, Kremr D, Eisner A, Ventura K. Chemical composition of essential oils from Plantago lanceolata L. leaves extracted by hydrodistillation. J Food Sci Technol 2016; 53(3):1576-84. [DOI:10.1007/s13197-015-2083-x] [PMID] [PMCID]
  2. Ji X, Hou C, Guo X. Physicochemical properties, structures, bioactivities and future prospective for polysaccharides from Plantago L.(Plantaginaceae): A review. Int J Biol Macromol. 2019; 135:637-46. [DOI:10.1016/j.ijbiomac.2019.05.211] [PMID]
  3. Najafian Y, Hamedi SS, Farshchi MK, Feyzabadi Z. Plantago major in Traditional Persian Medicine and modern phytotherapy: A narrative review. Electron Physician. 2018; 10(2):6390-9. [PMID] [PMCID]
  4. Gálvez M, Martín-Cordero C, Houghton PJ, Ayuso MJ. Antioxidant activity of methanol extracts obtained from Plantago species. J Agric Food Chem. 2005; 53(6):1927-33. [PMID]
  1. Samuelsen AB. The traditional uses, chemical constituents and biological activities of Plantago major L. A review. J Ethnopharmacol. 2000; 71(1-2):1-21. [DOI:10.1016/S0378-8741(00)00212-9]
  2. Weryszko-Chmielewska E, Matysik-Wozniak A, Sulborska A, Rejdak R. Commercially important properties of plants of the genus Plantago. Acta Agrobot. 2012; 65(1):11-20. [DOI:10.5586/aa.2012.038]
  3. Akbari J, Saeedi M, Morteza-Semnani K, Zarrabi B, Rostamkalaei SS, Kelidari HR. The effect of Plantago major seed mucilage combined with carbopol on the release profile and bioadhesive properties of propranolol HCl buccoadhesive tablets. Pharm Biomed Res. 2016; 2(2):84-100. [DOI:10.18869/acadpub.pbr.2.2.84]
  4. Khalaf HAA, Mahdi MF, Abaas IS. Preliminary phytochemical and GC-MS analysis of chemical constituents of Iraqi Plantago lanceoleta L. Al-Mustansiriyah J Pharm Sci. 2018; 18(2):114-21. [DOI:10.32947/ajps.18.02.0381]
  5. Plumb JA. Cell sensitivity assays: Clonogenic assay. Methods Mol Med. 2004; 88:159-64. [PMID]
  6. Rajabi S, Ramazani A, Hamidi M, Naji T. Artemia salina as a model organism in toxicity assessment of nanoparticles. Daru. 2015; 23(1):20. [PMID] [PMCID]
  7. Al-Marzoqi AH, Hameed IH, Idan SA. Analysis of bioactive chemical components of two medicinal plants (Coriandrum sativum and Melia azedarach) leaves using gas chromatography-mass spectrometry (GC-MS). Afr J Biotechnol. 2015; 14(40):2812-30. [DOI:10.5897/AJB2015.14956]
  8. Jatin RR, Priya R S. Determination of bioactive components of cynodon dactylon by GC-MS analysis & it’s in vitro antimicrobial activity. Int J Pharm Life Sci. 2016; 7(1):4880-5.[Link]
  9. Xiong C, Li Q, Li S, Chen C, Chen Z, Huang W. In vitro antimicrobial activities and mechanism of 1-octen-3-ol against food-related bacteria and pathogenic fungi. J Oleo Sci. 2017; 66(9):1041-9. [PMID]
  10. Chitarra GS, Abee T, Rombouts FM, Posthumus MA, Dijksterhuis J. Germination of penicillium paneum conidia is regulated by 1-octen-3-ol, a volatile self-inhibitor. Appl Environ Microbiol. 2004; 70(5):2823-9. [PMID]
  11. Zin T, Sundaram CS, Rao UM. In silico analysis of the phenylephrine from the sand crab (emerita asiatica) for its antimicrobial activities. Res J Pharm Technol. 2019; 12(5):01-4. [DOI:10.5958/0974-360X.2019.00356.1]
  12. Hameed IH, Altameme HJ, Idan SA. Artemisia annua: Biochemical products analysis of methanolic aerial parts extract and anti-microbial capacity. Res J Pharm Biol Chem Sci. 2016; 7(2):1843-68. [Link]
  13. Dabbagh A, Tajbakhsh A, Talebi Z, Rajaei S. Cardiovascular pharmacology in adult patients undergoing cardiac surgery. In: Dabbagh A, Esmailian F, Aranki S. Postoperative critical care for adult cardiac surgical patients. Cham: Springer; 2018. [DOI:10.1007/978-3-319-75747-6_4]
  14. Katke SA, Amrutkar SV, Bhor RJ, Khairnar MV. Synthesis of biologically active 2-chloro-N-alkyl/aryl acetamide derivatives. Int J Pharm Sci Res. 2011; 2(7):148-56. [Link]
  15. Devi RB, Barkath T, Vijayaraghavan P, Rejiniemon TS. GC-MS analysis of phytochemical from Psidium guajava Linn leaf extract and their in-vitro antimicrobial activities. Int J Pharma Bio Sci. 2019; 8:583-9. [Link]
  16. Sisler EC, Blankenship SM, Guest M. Competition of cyclooctenes and cyclooctadienes for ethylene binding and activity in plants. Plant Growth Regul. 1990; 9:157-64. [DOI:10.1007/BF00027443]
  17. Martinez ML, Campagna MN, Ratti MS, Nocito I, Serra E, Gattuso S, et al. Trypanocide activity of Castela coccinea Griseb. extracts. Boletín Latinoamericano y del Caribe de Plantas Medicinales y Aromáticas. 2009; 8(3):211-8. [Link]
  18. Armaka M, Papanikolaou E, Sivropoulou A, Arsenakis M. Antiviral properties of isoborneol, a potent inhibitor of Herpes simplex virus type 1. Antivir Res. 1999; 43(2):79-92. [DOI:10.1016/S0166-3542(99)00036-4]
  19. Tabanca N, Kırımer N, Demirci B, Demirci F, Başer KH. Composition and antimicrobial activity of the essential oils of Micromeria cristata subsp. phrygia and the enantiomeric distribution of borneol. J Agric Food Chem. 2001; 49(9):4300-3. [PMID]
  20. Asressu KH, Tesema TK. Chemical and antimicrobial investigations on essential oil of Rosmarinus officinalis leaves grown in Ethiopia and comparison with other countries. J Appl Pharm Sci. 2014; 6:132-42. [DOI:10.21065/19204159.6.3.112]
  21. Wessolowski A, Bienert M, Dathe M. Antimicrobial activity of arginineand tryptophanrich hexapeptides: The effects of aromatic clusters, damino acid substitution and cyclization. J Pept Res. 2004; 64(4):159-69. [PMID]
  22. Kenaga EE, Allison WE. Commercial and experimental organic insecticides (1969 revision). Bulletin Entomol Soc Am. 1969; 15(2):85-148. [DOI:10.1093/besa/15.2.85]
  23. Dean L. Atomoxetine therapy and CYP2D6 genotype. In: Pratt VM, Scott SA, Pirmohamed M, Esquivel B, Kane MS, Kattman BL,et al, editors. Medical genetics summaries. Bethesda: National Center for Biotechnology Information (US); 2020. [PMID]
  24. Catalani V, Prilutskaya M, Al-Imam A, Marrinan S, Elgharably Y, Zloh M, et al. Octodrine: New questions and challenges in sport supplements. Brain sci. 2018; 8(2):34. [PMID]
  25. Kim K, Zilbermintz L, Martchenko M. Repurposing FDA approved drugs against the human fungal pathogen, Candida albicans. Ann Clin Microbiol J Antimicrob. 2015; 14:32. [PMID] [PMCID]
  26. Niu H, Cui P, Shi W, Zhang S, Feng J, Wang Y, et al. Identification of anti-persister activity against uropathogenic Escherichia coli from a clinical drug library. Antibiotics Basel. 2015; 4(2):179-87. [PMID] [PMCID]
  27. Pellati F, Benvenuti S, Melegari M. Enantioselective LC analysis of synephrine in natural products on a protein-based chiral stationary phase. J Pharm Biomed Anal. 2005; 37(5):839-49. [PMID]
  28. Hartling L, Bialy LM, Vandermeer B, Tjosvold L, Johnson DW, Plint AC, et al. Epinephrine for bronchiolitis. Cochrane Database Syst Rev. 2011; 6:1-138. [DOI:10.1002/14651858.CD003123.pub3]
  29. Parish HG, Morton JR, Brown JC. A systematic review of epinephrine stability and sterility with storage in a syringe. Allergy Asthma Clin Immunol. 2019; 15:7. [PMID]
  30. Kamal SA, Hamza LF, Ibraheam IA. Characterization of antifungal metabolites produced by Aeromonas hydrophila and analysis of its chemical compounds using GC-MS. Res J Pharm Technol. 2017; 10(11):3845-51. [DOI:10.5958/0974-360X.2017.00697.7]
  31. Crean RD, Davis SA, Von Huben SN, Lay CC, Katner SN, Taffe MA. Effects of (±) 3, 4-methylenedioxymethamphetamine,(±) 3, 4-methylenedioxyamphetamine and methamphetamine on temperature and activity in Rhesus macaques. Neuroscience. 2006; 142(2):515-25. [PMID] [PMCID]
  32. Adeoya-Osiguwa S, Fraser LR. Cathine, an amphetamine-related compound, acts on mammalian spermatozoa via β1-and α2A-adrenergic receptors in a capacitation state-dependent manner. Hum Reprod. 2007; 22(3):756-65. [DOI:10.1093/humrep/del454] [PMID]
  33. Dal Cason TA. A re-examination of the mono-methoxy positional ring isomers of amphetamine, methamphetamine and phenyl-2-propanone. Forensic Sci Int. 2001; 119(2):168-94. [DOI:10.1016/S0379-0738(00)00425-4]
  34. Allam YA, Swellem RH, Nawwar GA. Cyanoacetylurea in heterocyclic synthesis: A simple synthesis of heterocyclic condensed uracils. J Chem Res. 2001; 2001(8):346-8. [DOI:10.3184/030823401103170034]
  35. Ohwada S, Ikeya T, Yokomori T, Kusaba T, Roppongi T, Takahashi T, et al. Adjuvant immunochemotherapy with oral Tegafur/Uracil plus PSK in patients with stage II or III colorectal cancer: A randomised controlled study. Br J cancer. 2004; 90(5):1003-10. [PMID] [PMCID]
  36. Peters D, Hörnfeldt AB, Gronowitz S, Johansson NG. Synthesis and antiviral activity of various 5-substituted 2′-deoxyuridines and-cytidines. Nucleosides Nucleotides.1992; 11(6):1151-73. [DOI:10.1080/07328319208018333]
  37. Dooley M, Goa KL.Urapidil. A reappraisal of its use in the management of hypertension. Drugs. 1998; 56(5):929-55. [PMID]
  38. Yagi K, Akimoto K, Mimori N, Miyake T, Kudo M, Arai K, et al. Synthesis and insecticidal/acaricidal activity of novel 3(2, 4, 6trisubstituted phenyl) uracil derivatives. Pest Manag Sci. (Formerly Pestic Sci). 2000; 56(1):65-73. [DOI:10.1002/(SICI)1526-4998(200001)56:13.0.CO;2-S]
  39. Lenders JW, Pacak K, Walther MM, Linehan WM, Mannelli M, Friberg P, et al. Biochemical diagnosis of pheochromocytoma: Which test is best? JAMA. 2002; 287(11):1427-34. [PMID]
  40. Badenhorst D, Sutherland F, De Jager A, Scanes T, Hundt H, Swart K, et al. Determination of doxepin and desmethyldoxepin in human plasma using liquid chromatography-tandem mass spectrometry. J Chromatogr B: Biome Sci Appl. 2000; 742(1):91-8. [DOI:10.1016/S0378-4347(00)00136-5]
  41. Canavero S. Central pain syndrome: Pathophysiology, diagnosis and management. Cambridge: Cambridge University Press; 2011. [DOI:10.1017/CBO9780511845673]
  42. Ashraf SA, Al-Shammari E, Hussain T, Tajuddin S, Panda BP. In-vitro antimicrobial activity and identification of bioactive components using GC-MS of commercially available essential oils in Saudi Arabia. J Food Sci Technol. 2017; 54(12):3948-58. [PMID]
  43. Sun S, Sun D, Yang L, Han J, Liu R, Wang L. Dose-dependent effects of intravenous methoxamine infusion during hip-joint replacement surgery on postoperative cognitive dysfunction and blood TNF-α level in elderly patients: A randomized controlled trial. BMC Anesthesiol. 2017; 17(1):75. [PMID]
  44. Sundaram S, Prabhakaran J. Effect of 1-methylcyclopropene (1-MCP) on volatile compound production in papaya (Carica papaya L.) fruit. Pharm Innovat J. 2017; 6:532-6. [Link]
  45. Imuta S, Tanimoto H, Momose MK, Chida N. Total synthesis of actinobolin from d-glucose by way of the stereoselective three-component coupling reaction. Tetrahedron. 2006; 62(29):6926-44. [DOI:10.1016/j.tet.2006.04.079]
  46. Joshi R, Adhikari S, Patro BS, Chattopadhyay S, Mukherjee T. Free radical scavenging behavior of folic acid: Evidence for possible antioxidant activity. Free Radic Biol Med. 2001; 30(12):1390-9. [DOI:10.1016/S0891-5849(01)00543-3]
  47. Atkinson HJ, Slater MA, Williams RA, Zisook S, Patterson TL, Grant I, et al. A placebo-controlled randomized clinical trial of nortriptyline for chronic low back pain. Pain. 1998; 76(3):287-96. [DOI:10.1016/S0304-3959(98)00064-5]
  48. Kadhim MJ, Mohammed GJ, Hameed IH. In vitro antibacterial, antifungal and phytochemical analysis of methanolic extract of fruit Cassia fistula. Orient J Chem. 2016; 32(3):1329-46. [DOI:10.13005/ojc/320307]
  49. Gajjala K, Anisetti R, Dodla JP, Rathod AK. Phytochemical investigation and cytotoxic activity of hydro alcoholic fraction of Trianthema decandra. Indian J Biotechnol. 2019; 18:193-203. https://scholar.google.com/scholar?hl=en&as_sdt=0%2C5&q
  50. Sardari S, Feizi S, Rezayan AH, Azerang P, Mohammad Shahcheragh S, Ghavami G, et al. Synthesis and biological evaluation of thiosemicarbazide derivatives endowed with high activity toward Mycobacterium bovis. Iran J Pharm Res. 2017; 16(3):1128-40. [DOI:10.22037/IJPR.2017.2069]
  51. Kamstra RL, Freywald A, Floriano WB. N(2, 4)dinitrophenylLarginine interacts with EphB4 and functions as an EphB4 Kinase modulator. Chem Biol Drug Des. 2015; 86(4):476-86. [PMID]
  52. Palchoudhuri S, Mukhopadhyay D, Roy DS, Ghosh B, Das S, Dastidar SG. The antidepressant drug doxepin: A promising antioxidant. Asian J Pharm Clin Res. 2017; 10(3):97-102. [DOI:10.22159/ajpcr.2017.v10i3.15149]
  53. Kirchheiner J, Henckel HB, Franke L, Meineke I, Tzvetkov M, Uebelhack R, et al. Impact of the CYP2D6 ultra-rapid metabolizer genotype on doxepin pharmacokinetics and serotonin in platelets. Pharmacogenet Genomics. 2005; 15(8):579-87. [PMID]
  54. Williams DA. Lemke TL. Foye’s principles of medicinal chemistry. Philadelphia: Lippincott Williams & Wilkins; 2012. [Link]
  55. Swiss Pharmaceutical Society. Index nominum 2000: International drug directory. Guildford: Medpharm Scientific Publishers; 2000. [Link]
  56. Elks J. The dictionary of drugs: Chemical data: Chemical data, structures and bibliographies. New York: Springer; 2014. [Link]
  57. Morton IK, Hall JM. Concise dictionary of pharmacological agents: Properties and synonyms. Berlin: Springer Science & Business Media; 1999. [DOI:10.1007/978-94-011-4439-1]
  58. Gupta RC. Veterinary toxicology: Basic and clinical principles. Amsterdam: Elsevier Science; 2012. [Link]
  59. Adams HR, Papich MG, Riviere JE. Veterinary pharmacology and therapeutics, 9th ed. Ames, Iowa: Wiley-Blackwell, 2009. https://lib.ugent.be/catalog/rug01:001318502
  60. Ara I, Shinwari M, Rashed S, Bakir M. Evaluation of antimicrobial properties of two different extracts of Juglans regia tree bark and search for their compounds using gas chromatography-mass spectrum. Int J Biol. 2013; 5(2):92-102. [DOI:10.5539/ijb.v5n2p92]
  61. Al-Rubaye AF, Kaizal AF, Hameed IH. Phytochemical screening of methanolic leaves extract of Malva sylvestris. Int J Pharmacog Phytochem Res. 2017; 9(4):537-52. [DOI:10.25258/phyto.v9i4.8127]
  62. Hussein AO, Mohammed GJ, Hadi MY, Hameed IH. Phytochemical screening of methanolic dried galls extract of Quercus infectoria using gas chromatography-mass spectrometry (GC-MS) and Fourier transform-infrared (FT-IR). J Pharmacogn Phytotherapy. 2016; 8:49-59. [DOI:10.5897/JPP2015.0368]
  63. Kadhim MJ. In vitro antifungal potential of Acinetobacter baumannii and determination of its chemical composition by gas chromatography-mass spectrometry. Der Pharma Chemica. 2016; 8(19):657-65. [Link]
  64. Perumal R, Albertmanoharan S, Pemiah B. GC-MS evidence based herbocure from indian system of medicine for stomach disorders in vets. Asian J Anim Vet Adv. 2018; 13(1): 73-84. [DOI:10.3923/ajava.2018.73.84]
  65. Suo C, Sun Z, Wang Y, Dong S, Lv C, Wang T, et al. Active components in leaves of Rhus chinensis Mill. Therm Sci. 2020; 24(3A):1729-35. [DOI:10.2298/TSCI190529045S]
  66. Jain S, Chandra V, Jain PK, Pathak K, Pathak D, Vaidya A. Comprehensive review on current developments of quinoline-based anticancer agents. Arab J Chem. 2019; 12(8):4920-46. [DOI:10.1016/j.arabjc.2016.10.009]
  67. Hwang TJ, Wares DF, Jafarov A, Jakubowiak W, Nunn P, Keshavjee S. Safety of cycloserine and terizidone for the treatment of drug-resistant tuberculosis: A meta-analysis. Int J Tuberc Lung Dis. 2013; 17(10):1257-66. [PMID]
  68. Nguyen LA, He H, Pham-Huy C. Chiral drugs: An overview. Int J Biomed Sci. 2006; 2(2):85-100. [PMID]
  69. Ayodele OO, Onajobi FD, Osoniyi OR. Phytochemical profiling of the hexane fraction of crassocephalum crepidioides benth S. Moore leaves by GC-MS. Afr J Pure Appl Chem. 2020; 14(1):1-8. [DOI:10.5897/AJPAC2019.0815]
  70. Malekzadeh H, Fatemi MH. Analysis of flavor volatiles of some Iranian rice cultivars by optimized static headspace gas chromatography-mass spectrometry. J Iran Chem Soc. 2015; 12:2245-51. [DOI:10.1007/s13738-015-0703-z]
  71. Clarke S. Essential chemistry for aromatherapyk. ChurchillLivingstone, London: Elsevier Health Sciences; 2009. https://www.sciencedirect.com/book/9780443104039/essential-chemistry-for-aromatherapy#book-info
  72. Fons F, Rapior S, Gargadennec A, Andary C, Bessière JM. Volatile components of Plantago lanceolata (Plantaginaceae). Acta Bot. Gall. 1998; 145(4):265-9. [DOI:10.1080/12538078.1998.10516306]
  73. Rahamooz-Haghighi S, Bagheri K, Danafar H, Sharafi A. Anti-proliferative properties, biocompatibility, and chemical composition of different extracts of plantago major medicinal plant. Iran Biomed J. 2021; 25(2):106-16. [PMID]
  74. Velasco-Lezama R, Tapia-Aguilar R, Román-Ramos R, Vega-Avila E, Pérez-Gutiérrez MS. Effect of Plantago major on cell proliferation in vitro. J Ethnopharmacol. 2006; 103(1):36-42. [PMID]
  75. Ruebhart DR, Wickramasinghe W, Cock IE. Protective efficacy of the antioxidants vitamin E and Trolox against Microcystis aeruginosa and microcystin-LR in Artemia franciscana nauplii. J Toxicol Environ Health Part A. 2009; 72(24):1567-75. [PMID]
  76. Mirzaei M, Mirzaei A. Comparison of the Artemia salina and Artemia uramiana bioassays for toxicity of 4 Iranian medicinal plants. Int J Biol Sci. 2013; 2(3):49-54. [Link]
  77. Omer E, Elshamy AI, Nassar M, Shalom J, White A, Cock IE. Plantago squarrosa Murray extracts inhibit the growth of some bacterial triggers of autoimmune diseases: GC-MS analysis of an inhibitory extract. Inflammopharmacology 2019; 27(2):373-85. [PMID]
Type of Study: Original Research | Subject: Phyochemistry

Add your comments about this article : Your username or Email:
CAPTCHA

Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

© 2024 CC BY-NC 4.0 | Pharmaceutical and Biomedical Research

Designed & Developed by : Yektaweb