Next Article in Journal
Retinoic Acid Potentiates the Therapeutic Efficiency of Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSCs) against Cisplatin-Induced Hepatotoxicity in Rats
Previous Article in Journal
Evaluation of 5-[(Z)-(4-nitrobenzylidene)]-2-(thiazol-2-ylimino)-4-thiazolidinone (Les-6222) as Potential Anticonvulsant Agent
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Narrative Review on the Bioactivity and Health Benefits of Alpha-Phellandrene

by
Subramanian Thangaleela
1,
Bhagavathi Sundaram Sivamaruthi
2,
Periyanaina Kesika
1,2,
Tanawat Tiyajamorn
1,
Muruganantham Bharathi
1,* and
Chaiyavat Chaiyasut
1,*
1
Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
2
Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
*
Authors to whom correspondence should be addressed.
Sci. Pharm. 2022, 90(4), 57; https://doi.org/10.3390/scipharm90040057
Submission received: 9 September 2022 / Revised: 19 September 2022 / Accepted: 22 September 2022 / Published: 27 September 2022

Abstract

:
Aromatic essential oils play a significant role in pharmaceuticals, food additives, cosmetics, and perfumery. Essential oils mostly comprise aliphatic hydrocarbons, monoterpenoids, sesquiterpenoids and diterpenes. Plant extracts comprise a complex mixture of terpenes, terpenoids, aliphatic and phenol-derived aromatic components. Terpenes are a significant class of hydrocarbons with numerous health benefits. These biological functions of essential oil components are examined in vitro and in vivo studies. Some studies evaluated the properties and functions of α-phellandrene (α-PHE). Detailed evaluation to determine the functions of α-PHE over a spectrum of health care domains needs to be initiated. Its possible mechanism of action in a biological system could reveal the future opportunities and challenges in using α-PHE as a pharmaceutical candidate. The biological functions of α-PHE are reported, including anti-microbial, insecticidal, anti-inflammatory, anti-cancer, wound healing, analgesic, and neuronal responses. The present narrative review summarizes the synthesis, biotransformation, atmospheric emission, properties, and biological activities of α-PHE. The literature review suggests that extended pre-clinical studies are necessary to develop α-PHE-based adjuvant therapeutic approaches.

1. Introduction

Plant-based medicines are playing a vivid role in health care. Plant-derived essential oils are one of the prominent sources of bioactive molecules, including volatile lipophilic compounds with strong aromas. Essential oils are composed of hydrocarbons and their derivatives, such as alcohols, acids, esters, aldehydes, ketones, amines, nitrogen and sulfur compounds, oxygenated terpenes, terpenoid hydrocarbons and sesquiterpenes [1,2]. Essential oils are isolated from plants through various distillation processes, such as hydro and steam distillation, effleurage, maceration, supercritical, microwave-assisted, and solvent extractions [1]. Other chromatographic techniques, such as column, thin layer, high-pressure liquid chromatography, mass spectrometry (MS), nuclear magnetic resonance (NMR) spectroscopy, gas chromatography/mass spectrometry (GC/MS), liquid chromatography/mass spectrometry, and Fourier-transform infrared spectrophotometry were also used in the characterization and identification of compounds in the essential oils [1].
Monoterpenes are the most important constituents of essential oils. α-phellandrene (α-PHE) is a cyclic monoterpene with two double bonds in a heterocyclic ring (endocyclic) [3]. It was named after Eucalyptus phellandra (E. phellandra), the plant from where α-PHE was first isolated. α-PHE is a natural plant-derived compound with various medicinal properties, found useful in the pharmaceutical, food, cosmetic and perfume industries [4,5]. Phellandrene (PHE) compounds are prevalently used in fragrances [6]. PHE is found in plants, such as Angelica and Eucalyptus, and due to its pleasant scent, it has been used in fragrances [7]. E. microtheca, E. viminalis, and E. dives primarily emit α-PHE into the atmosphere, and the α-PHE produces particle nucleation due to monoterpene oxidation, observed over Eucalypt forests [8,9]. In the indoor environment, α-PHE is used in cleansing products, detergents, and room fresheners [10]. α-PHE interacts with the local environment when it is emitted. α-PHE is an important component for ozonolysis and produces a significant, blue-colored haze due to nocturnal nucleation over the Eucalypt forests [11].
Though several studies have detailed the pharmacological activities of α-PHE, the biological activities of α-PHE still need to be studied in detail. α-PHE can potentially be used to manage inflammatory diseases, such as osteoarthritis, rheumatoid arthritis, allergies, etc. Some adverse events may occur in the use of α-PHE. For example, any topical application of α-PHE can cause skin irritations, and oral administration of α-PHE may result in nausea, vomiting, diarrhea, and intestinal disturbances [12]. This review aimed to summarize the necessary details of α-PHE, including synthesis and biological functions.

2. Properties of Phellandrene

Phellandrenes comprise alpha-phellandrene (α-PHE) and beta-phellandrene (β-PHE), prominently found in eucalyptus plant species. PHEs are also present in other plants, such as mint, black pepper, parsley, cinnamon, lavender, pine, ginger grass, water fennel, and dill [13]. PHEs are herbaceous with different aromas, which have been known for their anti-fungal [14], anti-inflammatory, anti-hyperalgesic, anti-depressant [15], analgesic [16] and anti-cancer activity [17,18]. Plant-derived secondary metabolites comprise different active components, most prevalently, terpenes, alkaloids, and phenolic compounds [19]. The compounds are extracted from the medicinal plants via various methods, which yield a complex liquid mixture of chemicals, including terpenes, terpenoids, aliphatic and aromatic compounds and characteristic volatile properties, and are mostly lipophilic [4,16]. The monoterpene α-PHE is one of the predominant constituents of many plant species. The list of the representative plant species containing α-PHE is summarized in Table 1, and pictures of the few representative plant species are displayed in Figure 1.

3. Chemistry and Synthesis of α-PHE

Tea and lemon tree oils have high p-cymene and other monoterpenoids [39,40]. γ-Terpinene (1-isopropyl-4-methyl-1,4-cyclohexadiene) and α-PHE (5-isopropyl-2-methyl-1,3-cyclohexadiene) with the molecular formula C10H16 are grouped under the class of monoterpenes. In α-PHE, the cis-1, 3-diene chain is inserted into the 6-membered ring. Photooxidation of α-PHE revealed that the ring structure of α-PHE opens to produce a cyclohexatriene-type product [41]. Like γ-terpinene, α-PHE can also exhibit three structural conformations due to the internal rotation of the exocyclic isopropyl group. The three conformers can be identified using experimental IR spectra. The UV-induced photoreaction results in the reorganization of the π-system. At λ > 200 nm, new bands are produced in the spectrum of α-PHE; this shows that the α-PHE seems to be photolabile under UV-C light [41].
In solution, α-PHE exists in folded conformation with the left-handed diene helix [42]. The diene helix is left handed in the axial conformer, and in the equatorial conformer, it is right handed. In α-PHE, (R)-(−)-5-isopropyl-2-methyl-1,3-cyclohexadiene possesses the quasi-axial-isopropyl group to sustain in solution [43]. α-PHE exists as an axial-isopropyl conformer in conformational equilibrium, and its stability is due to the presence of the CH/π hydrogen bond [44]. The electron transfer catalysis reaction of (R)-α-PHE with 4-methoxystyrene produces mixed cycloaddition products [45].
Upon photoexcitation, the ground state and excited state dynamics of α-PHE were studied using broadband femtosecond UV absorption spectroscopy [46]. The ring-opening dynamics of α-PHE were examined using picosecond time-resolved UV resonance Raman spectroscopy [47]. In the matrix isolation experiment, α-PHE photochemistry and ground state conformational size were intermediate between 1,3-cyclohexadiene (CHD) and 7-dehydrocholesterol, provitamin D3 (DHC) [41].
Synthesis of α-PHE from (R)-carvone through the batch process was reported by Sen and Grosch [48] and, through the continuous-flow process, by [49]. In Sen and Grosch’s model of α-PHE synthesis, R-carvone converted to the unsaturated ketone with the help of Wilkinson’s catalyst (Rh (PPh3)3Cl) and toluene. Unsaturated ketone converted to N-tosyl hydrazone in the presence of tosyl hydrazide resin (TsNHNH2) and tetrahydrofuran (THF). With the help of methyllithium (2MeLi) and diethylether (Et2O), N-tosyl hydrazone converted into (S)-α-PHE, and the yield was 48% [48] (Figure 2). In the continuous-flow process model, R-carvone was subjected to reduction with the help of a metal nanoparticle as a catalyst in the first reactor. Then the condensation was carried out in the second reactor with TSNHNH2 and acid resin as the catalyst. The steam from the second reactor was linked with the third reactor, which contains a strong base, n-butyllithium (nBuLi). The organic phase was collected and dried in the presence of disodium sulphate (Na2SO4) and filtered with alumina, which yielded crude α-PHE. The product was then purified using flash chromatography and distilled to produce 96% pure α-PHE [49] (Figure 3).

4. Biotransformation of α-PHE

Generally, fragrance compounds exist as single enantiomers. The diversity in sensory properties of fragrance is associated with different types of enantiomers. The chemical synthesis or derivatizations of stereo-, enantio- and regioselective forms can be made by the biotransformation method. The stereo- and enantioselective chiral compound biotransformations were found useful in pharmaceutical and chemical industries in commercial-scale applications [50].
α-PHE exists in two enantiomer forms, such as (−)-(R) and (+)-(S), with varied physicochemical and olfactory properties [5]. İşcan et al. bio-transformed enantiomerically pure (−)-(R)-α-PHE using 16 microorganisms, including bacteria, yeasts, and fungi. Organisms, such as Escherichia coli (E. coli), Escherichia coli O157-H7, Staphylococcus aureus (S. aureus), Staphylococcus aureus MRSA, Staphylococcus epidermidis (S. epidermis), Pseudomonas aeruginosa (P. aeruginosa), Enterobacter aerogenes (E. aerogenes), Proteus vulgaris (P. vulgaris), Salmonella typhimurium (S. typhimrium), Bacillus cereus (B. cereus), Bacillus subtilis (B. subtilis), Agrobacterium tumefaciens (A. tumefaciens), Erwinia carotovora subsp. carotovora (E. carotovora subsp. carotovora), Peudomonas syringae pv. Phaseolicola (P. syringae pv. phaseolicola), and Pseudomonas syringae pv. tomato (P. syringae pv. tomato) and Serratia marcescens, were used in the biotransformation of (−)-(R)-α-PHE [5]. (−)-(R)-α-PHE biotransformation yielded six metabolites namely, 5-p-menthene-1,2-diol, 6-hydroxypiperitone, α-PHE epoxide, cis-p-menth-2-en-1-ol, p-mentha-1(7), 5-dien-2-ol and carvotanacetone. The resulting metabolites were screened by chromatographic techniques, and their biological activities were examined against pathogenic microorganisms. The anti-bacterial activity of α-PHE and the compound 5-p-menthene-1,2-diol were analyzed. Among these two metabolites, 5-p-menthene-1,2-diol showed significant anti-microbial and anti-candidal (anti-fungal) activity compared to α-PHE [5].
In other studies, α-PHE was bio-transformed into other metabolites using different organisms. Three different metabolites, 5-p-menthene-1,2-diol, p-mentha-1(7), 5-dien-2-ol and 5-p-menthen-2-one, were synthesized by Corynespora cassiicola (C. cassiicola) (DSM 62474) (Figure 4) [51]. Biotransformation of α-PHE by Alternaria alternata produces 5-p-menthen-1, 2-diol with anti-microbial activity [52].
The chemical transformation of α-PHE by photo-oxidation and electrochemical processes has been reported [53]. (−)-(R)-α-PHE was also bio-transformed by fungal species such as Fusarium heterosporium (F. heterosporium), Aspergillus alliaceus (A. alliaceus), Yarrowia lipolitica (Y. lipolitica), Alternaria alternata (A. alternata), Saccharomyces cerevisiae (S. cerevisiae), Kluyveromyces lactis (K. lactis), Neurospora crassa (N. crassa), Fusarium solani (G. solani), Fusarium culmorum (F. culmorum), Botrytis cinerea (B. cineria), Aspergillus flavus (A. flavus), Phanerochaete chrysosporium (P. chrysosporium), and Devosia riboflavina (D. riboflavina) strains [5].

5. α-PHE in Atmospheric Emission

Naturally, the monoterpenes play an important function in the atmosphere’s chemistry and influence the atmosphere’s oxidative capacity, which results in the formation and development of the tropospheric ozone and the secondary organic aerosol (SOA), which cause changes in climate and health [54,55]. The biogenic emission sources exceed the emission level of volatile compounds globally into the atmosphere; in this scenario, the monoterpenes produce a significant proportion of non-methane hydrocarbons [56]. α-PHE is the most reactive monoterpene, known for its tropospheric degradation and aerosol formation [57]. The ozonolysis changes of α-PHE and other associated mechanisms were studied. The results showed that when α-PHE reacts with ozone, one of the double bonds in α-PHE opens and releases SOA with 10 carbons. Then, the second addition of ozone causes fragmentation and produces SOA with three or seven carbons [57].

6. Bioactivities of α-PHE

Several studies recognized that medicinal plants are rich sources of secondary metabolites. Among the secondary metabolites, essential oils have been reported for several pharmacological activities and were also used in cosmetics and agricultural industries [4,58]. This section summarizes the reported bioactivities of α-PHE. The bioactivities of α-PHE are summarized in Table 2.

6.1. Insecticidal Property of α-PHE

The Curcuma longa leaves essential oil (CLLEO), rich in α-PHE, has insecticidal properties against the third instar stage larvae of the Australian sheep blowfly (Lucilia cuprina). The study by Chabaan et al. revealed the insecticidal activity of CLLEO by comparing it with commercially available α-PHE. The third instar larvae were placed over CLLEO spread over the filter paper with varying concentrations, such as 0.15 to 2.86 μL/cm2 and commercial α-PHE (0.29–1.47 μL/cm2). The insecticidal activities, such as progressive darkness in the body of larvae, reduced movement, cuticle color change, and the death of the larvae, were observed. The vacuolar degeneration, brain damage and neurotoxicity were observed. Henceforth, α-PHE can be considered an eco-friendly method of pest control [59,60].

6.2. Anti-Microbial Properties of α-PHE

Generally, essential oils possess anti-microbial activity against microbes, including bacteria and fungi such as Candida albicans, Bipolaris sp., Alternaria alternate, Cuvularia lunata and Fusarium oxysporium [72,73,74,75,76]. α-PHE, β-PHE, ocimene, myrcene, limonene and α-caryophyllene showed in vitro anti-microbial activities against Bacillus sp., Escherichia coli, Candida albicans, Pseudomonas aeruginosa and Staphylococcus aureus [77,78]. α-PHE inhibited the growth of Salmonella choleraesuis (S. choleraesius), E. coli and B. subtilis [71], Micrococcus luteus, Rhodococcus rhodochorus (R. rhodochorus), Arthobacter protophormiae (A. protophormiae) and S. aureus [79] and Aspergillus flavus [80].
Gram-positive bacteria are more highly prone to be affected by essential oils than Gram-negative bacteria [81]. Gram-negative bacteria’s outer hydrophilic polysaccharide membrane is a barrier against hydrophobic essential oils [82,83]. The anti-bacterial mechanism of essential oils could be the lipophilicity of essential oils causing expansion of cell membrane, which increases membrane fluidity and permeability, and disturbs membrane proteins, which indulge in respiration ion transport in microbes, subsequently causing cell death because of the leakage of ions and cellular contents [84,85,86,87].
In the case of anti-fungal activity, α-PHE decreases the lipid content and alters the fungal hyphae, thus disturbing the membrane integrity [88,89]. α-PHE and nonanal showed inhibition of mycelial growth of Penicillium cyclopium in a dose-dependent way. The inhibitory action is directly proportional to the concentration of α-PHE. Complete mycelial growth inhibition was observed at the concentration of ≥1.35 mL/L of α-PHE. The minimum inhibitory (MIC) and minimum fungicidal concentration (MFC) of 1.7 and 1.8 mL/L of α-PHE were observed, respectively. Nonanal was relatively more efficient than α-PHE in inhibiting fungal growth (0.10 mL/L for complete inhibition). The MIC and MFC of nonanal against P. cyclopium were 0.3 and 0.4 mL/L, respectively.
The effect of α-PHE on the release of cell components was evaluated by measuring the optical density (OD260) values. The OD values of α-PHE treated P. cyclopium suspension were profoundly higher than the nonanal treated samples. α-PHE also disturbs the shape, structure and appearance of hyphae and conidia of P. cyclopium and causes the leakage of potassium ion (K+) after 30 min of α-PHE treatment (1.520 μg/mL) [14].

6.3. Analgesic Property of α-PHE

Pain is one of the common unpleasant experiences among people having physical injuries and mental illnesses, such as emotional, cognitive, and social issues [90]. α-PHE exerts antinociceptive effects in acute nociception models. The oral administration of α-PHE was found to be more effective. α-PHE exerts its anti-nociceptive activity through glutamatergic, nitrergic, opioid, cholinergic and adrenergic systems [16].
The anti-nociceptive activity of α-PHE in the nociception mice model was studied. Oral administration of α-PHE (3.125, 6.25 and 12.5 mg/kg) significantly inhibited the acetic acid-induced abdominal writhing in mice. The injection of 50 mg/kg α-PHE reduced the paw-licking time in mice injected with 2% formalin compared to the control. Nociception was also drastically reduced with α-PHE (3.125, 6.25, 12.5 mg/kg) injection in capsaicin (2 µg/paw) treated mice, which confirms the anti-nociceptive effect of α-PHE. Declines in the licking and biting behavior were observed in the mice with glutamate-induced nociception. The inhibitory effects of α-PHE may be due to its regulatory effects on the histamine, serotonin, and prostaglandin E2 (PGE2) [16].
The essential oil extracted from Schinus terebinthifolius Raddi fruits is comprised of α-pinene, β-pinene, sabinene, z-salven, α-funebrene, (R)-(+)-limonene, myrcene and α-PHE [24]. The terpene α-PHE from S. terebinthifolius showed anti-hyperalgesic and anti-depressant effects in spared nerve injury (SNI)-induced experimental hyperalgesia and depression rat models. S. terebinthifolius essential oil reduced the hypernociception, depression-like behavior and immobility induced by SNI. This study could open new possibilities for developing novel anti-hyperalgesic and anti-depressive drugs for treating chronic pain and depression due to neuropathy and infections [15].
The anti-nociceptive ability of α-PHE was studied in a Sarcoma-180 (S-180) hypernociception mice model. Nociception was induced by implanted S-180 into the axillary region of mice for 8 days. α-PHE (6.25, 12.5, 25 and 50 mg/kg) was orally administrated to mice on the first day of S-180 inoculation. Acute α-PHE treatment reduced peritumoral mechanical sensitivity for up to 180 min and antinociception in a dose-dependent way [64]. Another study reported that the essential oil from the Eucalyptus genus reduced the S-180-induced nociception without affecting rodent tumor growth. This showed that the analgesia does not depend on tumor reduction [91].

6.4. Immune Responses of α-PHE

The role of α-PHE on the immune responses was analyzed in the murine cells. BALB/c mice were treated with α-PHE (1, 5 and 25 mg/kg) or olive oil (vehicle control) for 27 days with the standard laboratory diet. The results showed that α-PHE did not significantly affect the body weight compared to the control group. α-PHE supplementations increased the cluster of differentiation (CD) 3 (at 1 mg/kg), CD19 (at 5 and 25 mg/kg), and Mac-3 (Macrophages) (at 25 mg/kg) levels and repressed the CD11b level (at 5 and 25 mg/kg). α-PHE promoted the phagocytotic activity of macrophages, NK cell activity, and B- and T-cell proliferation. The results indicated that α-PHE did not induce any toxic effects in murine cells but impacted the activities of immune cells [62].
The impact of α-PHE on infections, susceptibility, and other non-specific immune responses, such as the levels of total hemocyte count (THC), phenoloxidase (PO) activity, respiratory bursts (RB), superoxide dismutase (SOD), and the phagocytic and lysozyme activity of white shrimp (Litopenaeus vannamei) were reported. In detail, white shrimps were individually treated with α-PHE (4, 8 and 12 µg/g on day 1). On the second day, they were administered 10 µL of Vibrio alginolyticus suspension (1 × 107 CFU) or saline (control). Shrimp survival or sustainability was observed after 12, 24, 36, 48, 60 and 72 h. The survival rate of white shrimp was increased in the α-PHE treated group. The PO and SOD activity of shrimp was significantly higher in the α-PHE treatment than in the control group. No significant changes were observed in THCs, and the shrimp’s RB activity was higher than in the control. The expression of prophenoloxidase, LPS- and β-1,3-glucan binding protein and peroxinectin were also significantly increased. Shrimp treated with V. alginolyticus showed affected hemocytes that displayed flattened, shrinkage and pseudopodia. α-PHE (8 and 12 µg/g) treated shrimp hemocytes were less flattened and showed pseudopodia [63].

6.5. Anti-Cancer Property of α-PHE

α-PHE exhibits potent anti-cancer properties against various cancer types, such as lung cancer [92], human breast and prostate cancer [27], and human liver tumor [17,64]. Lin et al. (2014) demonstrated the role of α-PHE in the activation of immune responses under leukemic conditions, this would potentially initiate additional studies concerning α-PHE treatment for leukemia [68]. α-PHE in the essential oil of Solanum erianthum possesses anti-cancer activities against human breast cancer cells (Hs 578T) and human prostate cancer cells (PC-3) [26].
WEHI-3 murine leukemia cells treated BALB/c mice were co-treated with α-PHE (25 mg/kg and 50 mg/kg), and the changes were measured. The results showed that α-PHE did not significantly reduce the body weight of animals. The weight of the spleen was reduced in the α-PHE group compared to the control groups. A high dose of α-PHE (50 mg/kg) stimulated the B-cell and T-cell proliferation in WEHI-3 treated group, which indicated that α-PHE enhanced the immune responses, increased phagocytosis and NK cell cytotoxic activity in the leukemia WEHI3 cells of treated mice [67]. The macrophages exhibit inflammatory responses during tissue damage or pathogens invasion, which could maintain tissue homeostasis [93]. The NK cells are involved in recognizing and clearing off the pathogen-infected cells [94].
The cytotoxic and autophagy-inducing functions of α-PHE in human liver cancer cells were reported. Human liver cancer (J5) cells were treated with 0, 10, 30 or 50 µM of α-PHE for 24 h. J5 cells treated with 30 or 50 µM α-PHE showed cell 61% and 51% of viability, respectively. α-PHE treated cells showed about 21–46% cell viability after 48 and 72 h of treatment, which was significantly decreased compared to the control group [64].
The α-PHE effect on pre-autophagosome and autophagosome formation was also examined in J5 cells using monodansylcadaverine (MDC) staining. Additionally, 30 or 50 µM α-PHE for 24 h showed more MDC-stained J5 cells than the control. The involvement of α-PHE in the autophagy-associated regulatory protein signal transduction pathway was studied by evaluating proteins such as PI3K-I, III, Akt, mTOR, phosphorylated BCl-2 (pBCl-2), Beclin-1, LC3-II, p53, TIGAR, pIκB, IκB, NF-κB also nuclear p53, DARM, and NF-κB. The predominant autophagy-inducing proteins PI3K-I, Akt and mTOR were suppressed by α-PHE. Autophagosome-formation modulating proteins PI3K-III, pBCl-2, Beclin-1, and LC3-II were significantly increased in the α-PHE treated cells compared to control. The p53, DRAM, NF-κB, Beclin-1 and pIκB were also increased after 24 h of α-PHE treatment. α-PHE increases the translocation of NF-κB into the nucleus from the cytoplasm. The cytoplasmic p53, NF-κB, TIGAR and IκB were significantly reduced. Thus, the α-PHE induced the autophagy of human liver tumor cells, thereby reducing its viability [64].
Schinus molle L. derived α-PHE induced the DNA damage and repair associated protein expression was evaluated in WEHI-3 murine leukemia cells. Among different concentrations of α-PHE, 50 µM of α-PHE treated WEHI-3 cells showed reduced cell viability. Comet assay described the dose-dependent effect on DNA damage, condensation, and fragmentation. α-PHE also induced DNA damage by suppressing the expression of DNA repair-associated proteins, such as p53, O6-methylguanine DNA methyltransferase (MGMT), DNA-dependent protein kinase (DNA-PK) and BRCA-1. α-PHE inhibited the translocation of pH2A.X and MDC-1 from the cytosol to nuclei in WEHI-3 cells. α-PHE induced caspase 3,8 and 9 activities in a time-dependent way and induced cytochrome c release from mitochondria of WEHI-3 cells, which shows that α-PHE can induce apoptosis by mitochondrial-dependent pathways. α-PHE upregulated the apoptosis regulating proteins such as Bax, Bad, tBid, Endo G, PARP, Caspase 3, 9, 12, ATF-6a, GRP78 and GADD153 and reduced the Bcl2 and Mcl 1, indicating that α-PHE could promote cell death through mitochondrial and endoplasmic reticulum pathways [65].
The functional role of α-PHE in DNA damage, cell cycle and apoptotic regulatory genes in WEHI-3 cells were reported. WEHI-3 cells were treated with 10 µM α-PHE for 24 h and then subjected to cDNA microarray analysis. The results indicated that α-PHE treatment upregulated the DNA damage-inducer transcript 4 and DNA fragmentation factor. The cell cycle checkpoint genes, such as cyclin G2 and cyclin-dependent kinases inhibitor 2D and p21, were upregulated. Apoptotic genes, such as BCL2/adenovirus EIB interacting protein 3, XIAP-associated factor 1, BCL2 modifying factor, caspase 8, and FADD-like apoptosis regulator genes, were upregulated. The genes TATA box-binding protein, cyclin E2, growth arrest-specific 5, Gm5426, and death box polypeptide were downregulated [67].

6.6. Anti-Inflammatory Properties of α-PHE

The anti-inflammatory property of α-PHE was studied using the carrageenan-induced peritonitis mice model. Mice were treated with commercial α-PHE at various concentrations (50, 100 or 200 mg/kg). After one hour of α-PHE treatment, the animals were injected with diluted carrageenan. After 4 h of carrageenan injection, the mice were sacrificed, and the levels of proinflammatory cytokines IL-6 (Interleukin-6) and TNF-α (tumor necrosis factor-α) were measured by ELISA. The leukocyte rolling and adhesion were examined with the help of the real-time intravital microscopy method. Mast cell degranulation in the mesenteric tissue was also determined. The examined results showed that the carrageenan-injected air pouch or peritoneum produced leukocyte influx, which inhibited the α-PHE treated group. Reduced TNF-α and IL-6 could exert the leukocyte influx control by α-PHE. α-PHE and dexamethasone reduced the influx of total leukocytes, thereby reducing the neutrophils rolling or endothelial adherence [67].
The migration of leukocytes into the joints of rheumatoid arthritis patients can induce the secretion of ROS, proteases, cytokines, and prostaglandins, which could be harmful to the synovial tissues, bone, and cartilage [95]. In addition to the influx of pro-inflammatory cytokines, carrageenan also induces the expression of cyclo-oxygenase-2 (COX-2) and PGE2 [96,97]. Moreover, in addition to the inhibition of TNF-α and IL-6 in carrageenan-induced animal models, α-PHE also modulated the other inflammatory mediators, such as leukotriene B4, IL-8 and C5a [98,99,100,101]. The inhibitory effect of α-PHE might be associated with the reduced TNF-α and IL-6 release and regulation of the adhesion molecules in the endothelial cells and polymorphonuclear cells [102,103].
α-PHE, because of its anti-inflammatory property, could be used to inhibit hemorrhagic cystitis (HC), one of the side effects of the clinical usage of ifosfamide (IFOS). The neuroprotective role of α-PHE was evaluated in the IFOS-induced HC mouse model. HC can lead to bladder constriction, anemia, urinary tract infections, renal failure, and hydronephrosis, which might lead to death [104]. α-PHE prevents IFOS-induced HC by suppressing oxidative stress and inflammation. Mice that received α-PHE (25 and 50 mg/kg) showed a reduction in cell loss, hemorrhage and edema in bladder tissue compared to the control group. A dose of 25 mg/kg of α-PHE increased the antioxidant score, restored IL-1β and TNF-α, and reduced bladder damage. α-PHE at 12.5, 25 and 50 mg/kg doses reduced vascular protein leakage. The lower concentration of α-PHE could prevent the damage caused by IFOS. These findings furthermore support the use of α-PHE as a potential therapeutic compound. [70]. α-PHE also proved to be an effective cytoprotective agent that can potentially reduce the proinflammatory cytokines [68], controlling inflammation and protecting from free radicals [105]. α-PHE proved to have anti-dematogenic properties in acute inflammation models [69].

6.7. Wound-Healing Activities of α-PHE

Wound healing requires complex processes that involve growth factors, cytokines and different types of cells and their interactions [106,107]. α-PHE possesses promising efficiency for treating skin wounds by reducing inflammation and oxidative stress through the regulation of nitric oxide (NO), superoxide anion (O2−), TNF-α and IL-6 [71]. The first level of defense in wound healing is the migration of dermal fibroblasts to the site of injury, which is an important step in cutaneous wound repair [106]. The inflammatory phase is a critical period in wound healing, producing pro-inflammatory cytokines by macrophages regulates the healing process. Both in vitro and in vivo studies demonstrated that α-PHE stimulates fibroblast proliferation and migration to the wound site, suppresses the overproduction of pro-inflammatory cytokines and reduces intracellular NO and superoxide anion, thereby reducing oxidative stress [71]. α-PHE also attenuates neutrophil migration and mast cell degranulation and contributes to wound healing [69].
The synthesis, maturation and deposition of collagen is an important characteristic feature of the wound-healing process [108]. The migration of fibroblasts into the wound bed and the formation of an extracellular matrix of granulation tissue is another feature of wound healing [109]. The essential oil of Bursera morelensis promoted fibroblast migration to the wound site and initiated collagen synthesis [110]. α-pinene and α-PHE treatments showed wound-healing activity, with improved scar tissue and collagen deposition [34].

6.8. Neuronal Properties of Terpenes

The plant-derived secondary metabolites, such as alkaloids, flavonoids, and triterpenes, can avert the breakdown of acetylcholine and improve cognitive performance [111]. Cognitive and memory processes are well coordinated by the cholinergic system of the central nervous system (CNS). Plants belonging to the genera Mentha produce high monoterpenes and sesquiterpenes [5]. Essential oil from the peppermint plant (Mentha piperita) showed cholinergic inhibitory activity and GABAA/nicotinic receptor binding properties, which increases cognition in healthy adults. Monoterpene-rich plant extracts, such as Melissa and Rosmarinus genera, involve cholinergic and GABAergic neurotransmission that affects mood and cognitive performance [112]. Borneol, a bicyclic monoterpene, possesses radical scavenging activity [113] and is reported to have neuroprotective functions in both in vivo and in vitro models of AD [114].
α-PHE inhibited the nociception induced by formalin. The formalin-induced nociception involves the first phase-neurogenic pain due to the chemical stimuli of both the myelinated and unmyelinated afferent fibers [115]. The second phase occurs because of inflammatory mediators, such as serotonin, histamine, prostaglandins, bradykinin, and changes in the spinal dorsal horn’s neurons [116].
The intraplantar injection of glutamate activates N-methyl D-aspartate (NMDA) and α-amino-3-hydroxyl-5-methyl-4-isoxazolepropionate (non-NMDA) receptors by NO release and intracellular Ca2+ concentration. Calcium initiates NO synthase [117], which releases the pro-inflammatory cytokines, ROS, that enhance inflammatory reactions [118]. α-PHE could inhibit NO production by inhibiting the interaction of glutamate with its receptors [16]. Although α-PHE has not been involved in any study related to neuroprotective effects, other terpenes have found applications in neuronal functions. However, more studies on α-PHE and its role in CNS functions are required. The bioactivities of α-PHE are represented in Figure 5.

7. Possible Mechanism of Action of α-PHE

The anti-microbial activities of phellandrene or other terpenes were more effective against Gram-positive bacteria. The anti-fungal activity of α-PHE involves alteration in the mycelial morphology, disturbed membrane stability, and increased ions and other cell materials leakage [84]. α-PHE can induce changes in lipid content and fatty acid content of the cells and cause K+ leakage, increase in extracellular pH and further destroy fungal or microbial cells [14]. α-PHE might exert anti-inflammation activity by preventing NF-kB activity, reducing the production of TNF-α and IL-6 from LPS-stimulated macrophages [68,119], pro-inflammatory enzymes [120] and through inhibition of neutrophil infiltration and mast cell degranulation [68].
The possible analgesic effect of α-PHE could involve glutamatergic, opioid, nitrergic, cholinergic and adrenergic systems [16]. The nociceptive receptors, such as the transient receptor potential (TRP) family, including TRPM8 and TRPA1, mediate the analgesic effect [121,122]. α-PHE might have antihyperalgesic activity against cold sensitivity, SNI-induced hyperalgesia, and depression-like behavior in rats. This enables α-PHE to be used in treating pain resulting from neuropathy and infections and is clinically involved in developing new antinociceptive drugs [15]. The antinociceptive effect of α-PHE was reverted by an opioid receptor antagonist, naloxone, which indicates that antinociceptive activities depend on the opioid system [61].
The pain modulation pathways are associated with GABA in the mammalian brain [123]. GABA is the inhibitory neurotransmitter, so GABAergic neurons inhibit the release of excitatory neurotransmitters, such as serotonin, dopamine, glutamate, and acetylcholine [124]. α-PHE positively contributes to the GABAergic system by enhancing GABA’s inhibitory activity and reducing neuronal excitability [124], which might provide the antinociceptive effects for α-PHE. During oncogenic pain, the activation of immune responses leads to TNF-α release, which later releases prostaglandins and pro-inflammatory amines that cause hyperalgesia [125]. Thus, α-PHE, through the GABAergic and opioid pathways, reduces the cytokines TNF-α, IL-1β, IL-4 and IL-6, consequently strengthening effective antinociception.
α-PHE exhibits wound-healing efficiency because the terpenes possess an adhesive effect on the skin, which initiates the healing effect [126]. Terpenes, such as α-PHE and α-pinene, act as primary agents in repairing wounds by providing good tensile strength for scar and accelerating wound closure [127]. α-PHE favors wound-healing activities by inducing the proliferation and migration of fibroblasts, which stimulate wound closure. α-PHE decreases intracellular NO levels and superoxide anion, thereby inhibiting oxidative stress and boosting cutaneous wound healing [71].
The enzymes lipoxygenase-5 (LOX-5) and cyclooxygenase II (COX II) regulate the arachidonic acid pathway of inflammation [12]. LOX-5 is important for leukotriene production by neutrophils and other related inflammatory mediators responsible for NF-kB activation, which is a sign of early-stage joint inflammation in gout patients [120,128]. α-PHE inhibits COX-II and LOX-5 enzyme activity in a dose-dependent manner, comparable to the standard anti-inflammatory drug diclofenac sodium [12]. The possible mechanisms of action of α-PHE are presented in Figure 6.

8. Conclusions and Future Prospectus

Essential oils and their components have functional attributes in various fields, including the food, cosmetic and pharmacology industries. α-PHE-containing essential oils have been used in conventional treatments of certain diseases and infections. α-PHE has been reported for its several biological activities. Notably, α-PHE exerts anti-microbial, antioxidant, anti-nociception, anti-tumor, and anti-inflammatory activities. In vitro and in vivo studies have proven the pharmacological importance of α-PHE. However, more studies are needed to acknowledge the bio-properties and the pharmacokinetics of α-PHE. Similarly, though the role of α-PHE in several activities is explained, the existing evidence is insubstantial and not conclusive in confirming the mechanism of action of α-PHE. Apart from the beneficial effects, any topical application of α-PHE can cause skin irritations, and oral administration may result in nausea, vomiting, diarrhea, and intestinal disturbances, so intensive studies on the adverse effects of α-PHE are required. Collectively, the current literature review suggests that extended pre-clinical studies are vital to developing safe α-PHE-based adjuvant therapeutic approaches.

Author Contributions

Conceptualization, C.C., B.S.S. and P.K.; methodology, S.T. and B.S.S.; validation, C.C., B.S.S. and P.K.; formal analysis, S.T. and T.T.; investigation, S.T. and B.S.S.; resources, C.C.; data curation, S.T., B.S.S. and T.T.; writing—original draft preparation, S.T., B.S.S., P.K., M.B. and C.C.; writing—review and editing, S.T., B.S.S., P.K., M.B. and C.C.; supervision, C.C. and B.S.S.; project administration, C.C.; funding acquisition, C.C. All authors have read and agreed to the published version of the manuscript.

Funding

This project was supported by Chiang Mai University, Chiang Mai, Thailand.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available within the article.

Acknowledgments

S.T. and M.B. gratefully acknowledge the CMU Post-Doctoral Fellowship for their support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Baser, K.H.C. Analysis, and Assessment of Essential Oils. In A Manual on the Essential Oil Industry; De Silva, K.T., Ed.; United Nations Industrial Development Organization: Vienna, Austria, 1995; pp. 155–178. [Google Scholar]
  2. Guan, X.; Ge, D.; Li, S.; Huang, K.; Liu, J.; Li, F. Chemical composition and antimicrobial activities of Artemisia argyi Levl. et Vant essential oils extracted by simultaneous distillation-extraction, subcritical extraction and hydrodistillation. Molecules 2019, 24, 483. [Google Scholar] [CrossRef] [PubMed]
  3. Kim, H.; Park, J. A density functional theory study on the reaction mechanism of α-Phellandrene with NO3. Bull. Korean Chem. Soc. 2015, 36, 2139–2142. [Google Scholar] [CrossRef]
  4. Bizzo, R.H.; Hovell, A.M.C.; Rezende, C.M. Brazilian essential oil: General view, developments, and perspectives. Quim. Nova 2009, 3, 588–594. [Google Scholar] [CrossRef]
  5. İşcan, G.; Kirimer, N.; Demirci, F.; Demirci, B.; Noma, Y.; Başer, K.H. Biotransformation of (−)-(R)-α-phellandrene: Antimicrobial activity of its major metabolite. Chem. Biodivers. 2012, 9, 1525–1532. [Google Scholar] [CrossRef]
  6. Gaich, T.; Mulzer, J. Chiral Pool Synthesis: Starting from Terpenes. In Comprehensive Chirality, 1st ed.; Carreira, E.M., Yamamoto, H., Eds.; Elsevier: Amsterdam, The Netherland, 2012; Volume 2, pp. 163–206. [Google Scholar]
  7. New Directions Aromatics. Available online: https://www.newdirectionsaromatics.com/tags/phellandrene/essential-oils/ (accessed on 13 June 2022).
  8. Lee, S.H.; Young, L.H.; Benson, D.R.; Suni, T.; Kulmala, M.; Junninen, H.; Campos, T.L.; Rogers, D.C.; Jensen, J. Observations of nighttime new particle formation in the troposphere. J. Geophys. Res. 2008, 113, D10210. [Google Scholar] [CrossRef]
  9. Suni, T.; Kulmala, M.; Hirsikko, A.; Bergman, T.; Laakso, L.; Aalto, P.P.; Leuning, R.; Cleugh, H.; Zegelin, S.; Hughes, D.; et al. Formation and characteristics of ions and charged aerosol particles in a native Australian Eucalypt Forest. Atmos. Chem. Phys. 2008, 8, 129–139. [Google Scholar] [CrossRef]
  10. Clarke, S. Essential Chemistry for Aromatherapy, 2nd ed.; Churchill Livingstone: London, UK, 2009; pp. 123–229. [Google Scholar]
  11. Mackenzie-Rae, F.A.; Liu, T.; Deng, W.; Saunders, S.M.; Fang, Z.; Zhang, Y.; Wang, X. Ozonolysis of α-phellandrene—Part 1: Gas- and particle-phase characterisation. Atmos. Chem. Phys 2017, 17, 6583–6609. [Google Scholar] [CrossRef]
  12. Valsalan Soba, S.; Babu, M.; Panonnummal, R. Ethosomal gel formulation of alpha phellandrene for the transdermal delivery in gout. Adv. Pharm. Bull. 2021, 11, 137–149. [Google Scholar] [CrossRef]
  13. Trulieve. Available online: https://www.trulieve.com/discover/blog/everything-you-need-to-know-about-a-phellandrene-and-its-benefits (accessed on 20 July 2022).
  14. Zhang, J.H.; Sun, H.L.; Chen, S.Y.; Wang, T.T. Anti-fungal activity, mechanism studies on α-Phellandrene and nonanal against Penicillium cyclopium. Bot. Stud. 2017, 58, 13. [Google Scholar] [CrossRef]
  15. Piccinelli, A.C.; Santos, J.A.; Konkiewitz, E.C.; Oesterreich, S.A.; Formagio, A.S.; Croda, J.H.; Ziff, E.B.; Kassuya, C.A. Antihyperalgesic and antidepressive actions of (R)-(+)-limonene, α-phellandrene, and essential oil from Schinus terebinthifolius fruits in a neuropathic pain model. Nutr. Neurosci. 2015, 18, 217–224. [Google Scholar] [CrossRef]
  16. Lima, D.F.; Brandão, M.S.; Moura, J.B.; Leitão, J.M.; Carvalho, F.A.; Miúra, L.M.; Leite, J.R.; Sousa, D.P.; Almeida, F.R. Antinociceptive activity of the monoterpene α-phellandrene in rodents: Possible mechanisms of action. J. Pharm. Pharmacol. 2012, 64, 283–292. [Google Scholar] [CrossRef] [PubMed]
  17. Hsieh, S.L.; Li, Y.C.; Chang, W.C.; Chung, J.G.; Hsieh, L.C.; Wu, C.C. Induction of necrosis in human liver tumor cells by α-phellandrene. Nutr. Cancer 2014, 66, 970–979. [Google Scholar] [CrossRef] [PubMed]
  18. Lin, J.J.; Hsu, S.C.; Lu, K.W.; Ma, Y.S.; Wu, C.C.; Lu, H.F.; Chen, J.C.; Lin, J.G.; Wu, P.P.; Chung, J.G. Alpha-phellandrene-induced apoptosis in mice leukemia WEHI-3 cells in vitro. Environ. Toxicol. 2016, 31, 1640–1651. [Google Scholar] [CrossRef] [PubMed]
  19. Kennedy, D.O.; Wightman, E.L. Herbal Extracts and Phytochemicals: Plant Secondary Metabolites and the Enhancement of Human Brain function. Adv. Nutr. 2011, 2, 32–50. [Google Scholar] [CrossRef] [PubMed]
  20. Singh, G.; Singh, O.P.; Maurya, S. Chemical and biocidal investigations on essential oils of some Indian Curcuma species. Prog. Cryst. Growth Charact. Mater. 2002, 45, 75–81. [Google Scholar] [CrossRef]
  21. Stashenko, E.E.; Jaramillo, B.E.; Martínez, J.R. Analysis of volatile secondary metabolites from Colombian Xylopia aromatica (Lamarck) by different extraction and headspace methods and gas chromatography. J. Chromatogr. A 2004, 1025, 105–113. [Google Scholar] [CrossRef]
  22. Napoli, E.M.; Curcuruto, G.; Ruberto, G. Screening of the essential oil composition of wild Sicilian rosemary. Biochem. Syst. Ecol. 2010, 38, 659–670. [Google Scholar] [CrossRef]
  23. Gilles, M.; Zhao, J.; An, M.; Agboola, S. Chemical composition and antimicrobial properties of essential oil of three Australian Eucalyptus species. Food Chem. 2010, 119, 731–737. [Google Scholar] [CrossRef]
  24. Nascimento, A.F.; Camara, C.A.; Moraes, M.M.; Ramos, C.S. Essential oil composition and acaricidal activity of Schinus terebinthifolius from Atlantic Forest of Pernambuco, Brazil against Tetranychusurticae. Nat. Prod. Commun. 2012, 7, 129–132. [Google Scholar]
  25. Bendaoud, H.; Romdhane, M.; Souchard, J.P.; Cazaux, S.; Bouajila, J. Chemical composition and anticancer and antioxidant activities of Schinus molle L. and Schinus terebinthifolius Raddi berries essential oils. J. Food Sci. 2010, 75, C466–C472. [Google Scholar] [CrossRef]
  26. Essien, E.E.; Ogunwande, I.A.; Setzer, W.N.; Ekundayo, O. Chemical composition, antimicrobial, and cytotoxicity studies on S. erianthum and S. macranthum essential oils. Pharm. Biol. 2012, 50, 474–480. [Google Scholar] [CrossRef] [PubMed]
  27. Asbaghian, S.; Shafaghat, A.; Zarea, K.; Kasimov, F.; Salimi, F. Comparison of volatile constituents, and antioxidant and antibacterial activities of the essential oils of Thymus caucasicus, T. kotschyanus and T. vulgaris. Nat. Prod. Commun. 2011, 6, 137–140. [Google Scholar] [CrossRef]
  28. Arjouni, M.Y.; Bahri, F.; Romane, A.; El Fels, M.A. Chemical composition and antimicrobial activity of essential oil of Cupressus atlantica. Nat. Prod. Commun. 2011, 6, 1519–1522. [Google Scholar] [CrossRef]
  29. Hajhashemi, V.; Abbasi, N. Hypolipidemic activity of Anethum graveolens in rats. Phytother. Res. 2008, 22, 372–375. [Google Scholar] [CrossRef] [PubMed]
  30. Popovici, J.; Bertrand, C.; Bagnarol, E.; Fernandez, M.P.; Comte, G. Chemical composition of essential oil and headspace-solid micro extracts from fruits of Myrica gale L. and antifungal activity. Nat. Prod. Res. 2008, 22, 1024–1032. [Google Scholar] [CrossRef]
  31. Brandt, J.J.; Wulf, S. Composition of the Essential Oils of Ligusticum mutellina (L.) Crantz (Apiaceae). J. Essen. Oil. Res. 1995, 7, 231–235. [Google Scholar] [CrossRef]
  32. Thakuri, B.C.; Padalia, R.C.; Tiwari, A.; Chanotiya, C.S.; Mathela, C.S. α-Phellandrene rich essential oil from Ligusticum marginatum C.B. Clarke. J. Essent. Oil Bear. Plants 2007, 10, 1–4. [Google Scholar] [CrossRef]
  33. Cha, J.D.; Jung, E.K.; Kil, B.S.; Lee, K.Y. Chemical composition, and antibacterial activity of essential oil from Artemisia feddei. J. Microbiol. Biotechnol. 2007, 17, 2061–2065. [Google Scholar]
  34. Salas-Oropeza, J.; Jimenez-Estrada, M.; Perez-Torres, A.; Castell-Rodriguez, A.E.; Becerril-Millan, R.; Rodriguez-Monroy, M.A.; Jarquin-Yañez, K.; Canales-Martinez, M.M. Wound healing activity of α-Pinene and α-Phellandrene. Molecules 2021, 26, 2488. [Google Scholar] [CrossRef]
  35. Koudou, J.; Etou, A.W.; Ossibi, K.; Abena, A.A.; Gbeassor, M.; Bessiere, J.M. Chemical composition and hypotensive effects of essential oil Monodora myristica Gaertn. J. Biol. Sci. 2007, 7, 937–942. [Google Scholar] [CrossRef]
  36. Onyenekwe, P.C.; Stahl, M.; Adejo, G. Post-irradiation changes of the volatile oil constituents of Monodora myristica (Gaertn) Dunal. Nat. Prod. Res. 2012, 26, 2030–2034. [Google Scholar] [CrossRef]
  37. Jirovetz, L.; Buchbauer, G.; Ngassoum, M.B.; Geissler, M. Aroma compound analysis of Piper nigrum and Piper guineense essential oils from Cameroon using solid-phase microextraction-gas chromatography, solid-phase microextraction-gas chromatography-mass spectrometry and olfactometry. J. Chromatogr. A 2002, 976, 265–275. [Google Scholar] [CrossRef]
  38. POWO Plants of the World Online. Facilitated by the Royal Botanic Gardens, Kew. 2022. Available online: http://www.plantsoftheworldonline.org/ (accessed on 17 September 2022).
  39. Hausen, B.M.; Reichling, J.; Harkenthal, M. Degradation products of monoterpenes are the sensitizing agents in tea tree oil. Am. J. Contact Dermat. 1999, 10, 68–77. [Google Scholar] [CrossRef]
  40. Iwanami, Y.; Tateba, H.; Kodama, N.; Kishino, K. Changes of lemon flavor components in an aqueous solution during UV irradiation. J. Agric. Food Chem. 1997, 45, 463–466. [Google Scholar] [CrossRef]
  41. Marzec, K.M.; Reva, I.; Fausto, R.; Proniewicz, L.M. Comparative matrix isolation infrared spectroscopy study of 1,3- and 1,4-diene monoterpenes (α-phellandrene and γ-terpinene). J. Phys. Chem. A 2011, 115, 4342–4353. [Google Scholar] [CrossRef]
  42. Burgstahler, A.W.; Ziffer, H.; Weiss, U. The configurations of levopimaric acid and α-phellandrene; Interpretation of their rotatory dispersions. J. Am. Chem. Soc. 1961, 83, 4660–4661. [Google Scholar] [CrossRef]
  43. Horsman, G.A.; Emeis, C.A. Low-temperature optical rotatory dispersion. Tetrahedron 1965, 22, 167–173. [Google Scholar] [CrossRef]
  44. Takahashi, O.; Yamasaki, K.; Kohno, Y.; Ueda, K.; Suezawa, H.; Nishio, M. Origin of the axial-alkyl preference of (R)-α-phellandrene and related compounds investigated by high-level ab initio MO calculations. Importance of the CH/π hydrogen bond. Tetrahedron 2008, 64, 5773–5778. [Google Scholar] [CrossRef]
  45. Sevov, C.S.; Wiest, O. Selectivity in the electron transfer catalyzed Diels-Alder reaction of (R)-alpha-phellandrene and 4-methoxystyrene. J. Org. Chem. 2008, 73, 7909–7915. [Google Scholar] [CrossRef]
  46. Arruda, B.C.; Smith, B.; Spears, K.G.; Sension, R.J. Ultrafast ring-opening reactions: A comparison of alpha-terpinene, alpha-phellandrene, and 7-dehydrocholesterol with 1,3-cyclohexadiene. Faraday Discuss. 2013, 163, 159–275. [Google Scholar] [CrossRef]
  47. Reid, P.J.; Doig, S.J.; Wickham, S.D.; Mathies, R.A. Photochemical ring-opening reactions are complete in picoseconds: A time resolved UV resonance Raman study of 1,3 cyclohexadiene. J. Am. Chem. Soc. 1993, 115, 4754–4763. [Google Scholar] [CrossRef]
  48. Sen, A.; Grosch, W. The synthesis of enantiomerically pure (S)(+)-a-phellandrene from (R)(−)-carvone. Flavour Fragr. J. 1990, 5, 233–234. [Google Scholar] [CrossRef]
  49. Miller, S.J.; Ishitani, H.; Furiya, Y.; Shu, K. High-Throughput Synthesis of (S)-α-Phellandrene through Three-Step Sequential Continuous-Flow Reactions. Org. Process Res. Dev. 2021, 25, 192–198. [Google Scholar] [CrossRef]
  50. Straathof, A.J.; Panke, S.; Schmid, A. The production of fine chemicals by biotransformations. Curr. Opin. Biotechnol. 2002, 13, 548–556. [Google Scholar] [CrossRef]
  51. Abraham, W.R.; Stumpf, B.; Kieslich, K. Microbial Transformations of Terpenoids with 1-p-Menthene Skeleton. Appl. Microbiol. Biotechnol. 1986, 24, 24–30. [Google Scholar] [CrossRef]
  52. Lai-King, S.Y.; Saunders, R.M.K.; Brown, G.D. Phytochemistry of Illicium dunnianum and the systematic position of the illiciaceae. Phytochemistry 1997, 44, 1099–1108. [Google Scholar]
  53. Montiel, V.; Lopez-Segura, M.; Aldaz, A.; Grande, M.; Barba, F. Electrooxidation of terpenes-I. Synthesis of dihydrocarvone and 1-hydroxyneodihydrocarveol by anodic oxidation of limonene. Electrochim. Acta 1984, 29, 1123–1126. [Google Scholar] [CrossRef]
  54. Hoffmann, T.; Odum, J.R.; Bowman, F.; Collins, D.; Klockow, D.; Flagan, R.C.; Seinfeld, J.H. Formation of organic aerosols from the oxidation of biogenic hydrocarbons. J. Atmos. Chem. 1997, 26, 189–222. [Google Scholar] [CrossRef]
  55. Griffin, R.J.; Cocker, D.R.; Flagan, R.C.; Seinfeld, J.H. Organic aerosol formation from the oxidation of biogenic hydrocarbons. J. Geophys. Res. 1999, 104, 3555–3567. [Google Scholar] [CrossRef]
  56. Schurgers, G.; Arneth, A.; Holzinger, R.; Goldstein, A.H. Process-based modelling of biogenic monoterpene emissions combining production and release from storage. Atmos. Chem. Phys. 2009, 9, 3409–3423. [Google Scholar] [CrossRef]
  57. Mackenzie-Rae, F.A.; Wallis, H.J.; Rickard, A.R.; Pereira, K.L.; Saunders, S.M.; Wang, X.; Hamilton, J.F. Ozonolysis of α-phellandrene—Part 2: Compositional analysis of secondary organic aerosol highlights the role of stabilised Criegee intermediates. Atmos. Chem. Phys. 2017, 18, 4673–4693. [Google Scholar] [CrossRef]
  58. Bakkali, F.; Averbeck, S.; Averbeck, D.; Idaomar, M. Biological effects of essential oils—A review. Food Chem. Toxicol. 2008, 46, 446–475. [Google Scholar] [CrossRef] [PubMed]
  59. Chaaban, A.; Richardi, V.S.; Carrer, A.R.; Brum, J.S.; Cipriano, R.R.; Martins, C.; Navarro-Silva, M.A.; Deschamps, C.; Molento, M.B. Cuticular damage of Lucilia cuprina larvae exposed to Curcuma longa leaves essential oil and its major compound α-phellandrene. Data Brief 2020, 21, 1776–1778. [Google Scholar] [CrossRef]
  60. Chaaban, A.; Richardi, V.S.; Carrer, A.R.; Brum, J.S.; Cipriano, R.R.; Martins, C.E.; Silva, M.A.; Deschamps, C.; Molento, M.B. Insecticide activity of Curcuma longa (leaves) essential oil and its major compound a-phellandrene against Lucilia cuprina larvae (Diptera: Calliphoridae): Histological and ultrastructural biomarkers assessment. Pestic. Biochem. Physiol. 2019, 153, 17–27. [Google Scholar] [CrossRef] [PubMed]
  61. Pinheiro-Neto, F.R.; Lopes, E.M.; Acha, B.T.; Gomes, L.; Dias, W.A.; Reis Filho, A.; Leal, B.S.; Rodrigues, D.; Silva, J.; Dittz, D.; et al. α-Phellandrene exhibits antinociceptive and tumor-reducing effects in a mouse model of oncologic pain. Toxicol. Appl. Pharmacol. 2021, 418, 115497. [Google Scholar] [CrossRef] [PubMed]
  62. Lin, J.J.; Lin, J.H.; Hsu, S.C.; Weng, S.W.; Huang, Y.P.; Tang, N.Y.; Lin, J.G.; Chung, J.G. Alpha-phellandrene promotes immune responses in normal mice through enhancing macrophage phagocytosis and natural killer cell activities. Vivo 2013, 27, 809–814. [Google Scholar]
  63. Wu, C.C.; Lin, C.L.; Huang, C.Y.; Hsieh, S.; Liu, C.H.; Hsieh, S.L. α-Phellandrene enhances the immune response and resistance against Vibrio alginolyticus in white shrimp (Litopenaeus vannamei). Fish Shellfish Immunol. 2019, 84, 1108–1114. [Google Scholar] [CrossRef]
  64. Hsieh, L.C.; Hsieh, S.L.; Chen, C.T.; Chung, J.G.; Wang, J.J.; Wu, C.C. Induction of α-phellandrene on autophagy in human liver tumor cells. Am. J. Chin. Med. 2015, 43, 121–136. [Google Scholar] [CrossRef] [PubMed]
  65. Lin, J.J.; Wu, C.C.; Hsu, S.C.; Weng, S.W.; Ma, Y.S.; Huang, Y.P.; Lin, J.G.; Chung, J.G. Alpha-phellandrene-induced DNA damage and affect DNA repair protein expression in WEHI-3 murine leukemia cells in vitro. Environ. Toxicol. 2015, 30, 1322–1330. [Google Scholar] [CrossRef] [PubMed]
  66. Lin, J.J.; Lu, K.W.; Ma, Y.S.; Tang, N.Y.; Wu, P.P.; Wu, C.C.; Lu, H.F.; Lin, J.G.; Chung, J.G. Alpha-phellandrene, a natural active monoterpene, influences a murine WEHI-3 leukemia model in vivo by enhancing macrophage phagocytosis and natural killer cell activity. Vivo 2014, 28, 583–588. [Google Scholar]
  67. Lin, J.J.; Yu, C.C.; Lu, K.W.; Chang, S.J.; Yu, F.S.; Liao, C.L.; Lin, J.G.; Chung, J.G. α-Phellandrene alters expression of genes associated with DNA damage, cell cycle, and apoptosis in murine leukemia WEHI-3 cells. Anticancer Res. 2014, 34, 4161–4180. [Google Scholar] [PubMed]
  68. Siqueira, H.D.S.; Neto, B.S.; Sousa, D.P.; Gomes, B.S.; da Silva, F.V.; Cunha, F.V.M.; Wanderley, C.; Pinheiro, G.; Cândido, A.; Wong, D.; et al. α-Phellandrene, a cyclic monoterpene, attenuates inflammatory response through neutrophil migration inhibition and mast cell degranulation. Life Sci. 2016, 160, 27–33. [Google Scholar] [CrossRef] [PubMed]
  69. Siqueira, H.D.S.; Neto, B.S.; Sousa, D.P.; Gomes, B.S.; Cunha, F.V.M.; Oliveira, F. Antiedematogenic effect of alpha-phellandrene in animal models. Curr. Bioact. Compd. 2016, 12, 107–113. [Google Scholar] [CrossRef]
  70. Gonçalves, R.; Cunha, F.; Sousa-Neto, B.; Oliveira, L.; Lopes, M.E.; Rezende, D.C.; Sousa, I.; Nogueira, K.M.; Souza, L.; Medeiros, J.; et al. α-Phellandrene attenuates tissular damage, oxidative stress, and TNF-α levels on acute model ifosfamide-induced hemorrhagic cystitis in mice. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2020, 393, 1835–1848. [Google Scholar] [CrossRef]
  71. De Christo Scherer, M.M.; Marques, F.M.; Figueira, M.M.; Peisino, M.; Schmitt, E.; Kondratyuk, T.P.; Endringer, D.C.; Scherer, R.; Fronza, M. Wound healing activity of terpinolene and α-phellandrene by attenuating inflammation and oxidative stress in vitro. J. Tissue. Viability 2019, 28, 94–99. [Google Scholar] [CrossRef]
  72. Al-Burtamani, S.K.S.; Fatope, M.O.; Marwah, R.G.; Onifade, A.K.; Al-Saidi, S.H. Chemical composition, antibacterial and antifungal activities of the essential oil of Haplophyllum tuberculatum from Oman. J. Ethnopharmacol. 2005, 96, 107–112. [Google Scholar] [CrossRef]
  73. Inouye, S.; Takizawa, T.; Yamaguchi, H. Antibacterial activity of essential oils and their major constituents against respiratory tract pathogens by gaseous contact. J. Antimicrob. Chemother. 2001, 47, 565–573. [Google Scholar] [CrossRef]
  74. Hernández, V.; Mora, F.; Araque, M.; Montijo, S.D.; Rojas, L.; Meléndez, P.; Tommasi, N.D. Chemical composition and antibacterial activity of Astronium graveolens JACQ essential oil. Rev. Latinoamer. Quím. 2013, 41, 89–94. [Google Scholar]
  75. Nurettin, Y.; Canan, G.; Osman, Ü.; Ahmet, Y.; Serdar, Ü.; Kamil, C.; Salih, T. Composition and antimicrobial activities of volatile components of Minuartia meyeri. Turk. J. Chem. 2006, 30, 71–76. [Google Scholar]
  76. Sharma, P.; Shah, G.C.; Dhami, D.S.; Chauhan, P.K.; Singh, V. Chemical composition, antibacterial and antioxidant activities of senecio laetus edgew. From cold desert of western Himalaya. Int. J. Pharm. Res. Bio. Sci. 2014, 3, 188–199. [Google Scholar]
  77. Costa, T.R.; Fernandes, O.F.L.; Santos, S.C.; Oliveira, C.M.A.; Liao, L.M.; Ferri, P.H.; Paula, J.R.; Ferreira, H.D.; Sales, B.H.N.; Silva, M.R.R. Antifungal activity of volatile constituents of Eugenia dysenterica leaf oil. J. Ethnopharmacol. 2000, 72, 111–117. [Google Scholar] [CrossRef]
  78. Perez, C.; Agnese, A.M.; Cabrera, J.L. The essential oil of Scenecio graveolans (compositae)-chemical composition and antimicrobial activity tests. J. Ethnopharmacol. 1999, 66, 91–96. [Google Scholar] [CrossRef]
  79. Kurade, N.P.; Jaitak, V.; Kaul, V.K.; Sharma, O.P. Chemical composition and antibacterial activity of essential oils of Lantana camara, Ageratum houstonianum and Eupatorium adenophorum. Pharm. Biol. 2010, 48, 539–544. [Google Scholar] [CrossRef] [PubMed]
  80. Sindhu, S.; Chempakam, B.; Leela, N.K.; Suseela Bhai, R. Chemoprevention by essential oil of turmeric leaves (Curcuma longa L.) on the growth of Aspergillus flavus and aflatoxin production. Food Chem. Toxicol. 2011, 49, 1188–1192. [Google Scholar] [CrossRef]
  81. Smith-Palmer, A.; Stewart, J.; Fyfe, L. Antimicrobial properties of plant essential oils and essences against five important food-borne pathogens. Lett. Appl. Microbiol. 1998, 26, 118–122. [Google Scholar] [CrossRef]
  82. Tassou, C.C.; Nychas, G.J.E. Antimicrobial activity of the essential oil of Mastic gum (Pistacia lentiscus var chia) on gram-positive and gram-negative bacteria in broth and model food systems. Int. Biodeterior. Biodegrad. 1995, 36, 411–420. [Google Scholar] [CrossRef]
  83. Mann, C.M.; Cox, S.D.; Markham, J.L. The outer membrane of Pseudomonas aeruginosa NCTC6749 contributes to its tolerance to the essential oil of Melaleuca alternifolia (tea tree oil). Lett. Appl. Microbiol. 2000, 30, 294–297. [Google Scholar] [CrossRef]
  84. Oonmetta-aree, J.; Suzuki, T.; Gasaluck, P.; Eumkeb, G. Antimicrobial properties and action of galangal (Alpinia galangal Linn.) on Staphylococcus aureus. LWT-Food Sci. Technol. 2006, 39, 1214–1220. [Google Scholar] [CrossRef]
  85. Burt, S. Essential oils: Their antibacterial properties and potential applications in foods-A review. Int. J. Food Microbiol. 2004, 94, 223–253. [Google Scholar] [CrossRef]
  86. Fadli, M.; Saad, A.; Sayadi, S.; Chevalier, J.; Mezriouia, N.E.; Pagèsc, J.M.; Hassani, L. Antibacterial activity of Thymus maroccanus and Thymus broussonetii essential oils against nosocomial infection-bacteria and their synergistic potential with antibiotics. Phytomedicine 2012, 19, 464–471. [Google Scholar] [CrossRef]
  87. Khan, A.; Ahmad, A.; Akhtar, F.; Yousuf, S.; Xess, I.; Khan, L.A.; Manzoor, N. Ocimum sanctum essential oil and its active principles exert their antifungal activity by disrupting ergosterol biosynthesis and membrane integrity. Res. Microbiol. 2010, 161, 816–823. [Google Scholar] [CrossRef] [PubMed]
  88. Tyagi, A.K.; Malik, A. Antimicrobial potential and chemical composition of Mentha piperita oil in liquid and vapour phase against food spoiling microorganisms. Food Control 2011, 22, 1707–1714. [Google Scholar] [CrossRef]
  89. Yahyazadeh, M.; Omidbaigi, R.; Zare, R.; Taheri, H. Effect of some essential oils on mycelial growth of Penicillium digitatum Sacc. World J. Microbiol. Biotechnol. 2008, 24, 1445–1450. [Google Scholar] [CrossRef]
  90. Silva, L.J.; Mendanha, D.M.; Gomes, P.P. The use of opioids in the treatment of oncologic pain in the elderly. Braz. J. Pain 2020, 3, 63–72. [Google Scholar] [CrossRef]
  91. Gouveia, D.N.; Costa, J.S.; Oliveira, M.A.; Rabelo, T.K.; Silva, A.M.O.; Carvalho, A.A.; Miguel-Dos-Santos, R.; Lauton-Santos, S.; Scotti, L.; Scotti, M.T.; et al. α-Terpineol reduces cancer pain via modulation of oxidative stress and inhibition of iNOS. Biomed. Pharmacother. 2018, 105, 652–661. [Google Scholar] [CrossRef]
  92. Sylvestre, M.; Legault, J.; Dufour, D.; Pichette, A. Chemical composition and anticancer activity of leaf essential oil of Myrica gale L. Phytomedicine 2005, 12, 299–304. [Google Scholar] [CrossRef]
  93. Cobos Jimenez, V.; Bradley, E.J.; Willemsen, A.M.; van Kampen, A.H.; Baas, F.; Kootstra, N.A. Next-generation sequencing of microRNAs uncovers expression signatures in polarized macrophages. Physiol. Genom. 2014, 46, 91–103. [Google Scholar] [CrossRef]
  94. Adib-Conquy, M.; Scott-Algara, D.; Cavaillon, J.M.; Souza Fonseca-Guimaraes, F. TLR-mediated activation of NK cells and their role in bacterial/viral immune responses in mammals. Immunol. Cell Biol. 2014, 92, 256–262. [Google Scholar] [CrossRef]
  95. Wright, H.L.; Moots, R.J.; Bucknall, R.C.; Edwards, S.W. Neutrophil function in inflammation and inflammatory diseases. Rheumatology 2010, 49, 1618–1631. [Google Scholar] [CrossRef]
  96. Waldleigh, D.J.; Reddy, S.T.; Kopp, E.; Ghosh, S.; Herschman, H.R. Transcriptional activation of the cyclooxygenase-2 gene in endotoxin-treated RAW 264.7 macrophages. J. Biol. Chem. 2000, 275, 6259–6266. [Google Scholar] [CrossRef]
  97. Romano, M.; Faggioni, R.; Sironi, M.; Sacco, S.; Echtenacher, B.; Di Santo, E.; Salmona, M.; Ghezzi, P. Carrageenan-induced acute inflammation in the mouse air pouch synovival model. Role of tumour necrosis factor. Mediat. Inflamm. 1997, 6, 32–38. [Google Scholar] [CrossRef] [PubMed]
  98. Leahy, K.M.; Koki, A.T.; Masferrer, J.L. Role of cyclooxygenases in angiogenesis. Curr. Med. Chem. 2000, 7, 1163–1170. [Google Scholar] [CrossRef] [PubMed]
  99. Damas, J.; Bourdon, V.; Remacle-Volon, G.; Adam, A. Kinins and peritoneal exudates induced by carrageenin and zymosan in rats. Br. J. Pharmacol. 1990, 101, 418–422. [Google Scholar] [CrossRef] [PubMed]
  100. Ribeiro, R.A.; Souza-Filho, M.V.; Souza, M.H.; Oliveira, S.H.; Costa, C.H.; Cunha, F.Q.; Ferreira, H.S. Role of resident mast cells and macrophages in the neutrophil migration induced by LTB4, fMLP and C5a des arg. Int. Arch. Allergy Immunol. 1997, 112, 27–35. [Google Scholar] [CrossRef] [PubMed]
  101. Oliveira, S.H.; Canetti, C.; Ribeiro, R.A.; Cunha, F.Q. Neutrophil migration induced by IL1beta depends upon LTB4 released by macrophages and upon TNF-alpha and IL1beta released by mast cells. Inflammation 2008, 31, 36–46. [Google Scholar] [CrossRef]
  102. Perretti, M. Endogenous mediators that inhibit the leukocyte–endothelium interaction. Trends Pharmacol. Sci. 1997, 18, 418–425. [Google Scholar]
  103. Carvalho, A.M.R.; Rocha, N.F.M.; Vasconcelos, L.F.; RiosERV, D.L.M.; Silva, M.I.G.; Fonteles, M.M.F.; Barbosa Filho, J.M.; Gutierrez, S.J.C.; Sousa, F.C.F. Evaluation of the anti-inflammatory activity of riparin II (O-methil-N-2-hidroxi-benzoyl tyramine) in animal models. Chem. Biol. Interact. 2013, 205, 165–172. [Google Scholar] [CrossRef]
  104. Sencer, S.F.; Haake, R.J.; Weisdorf, D.J. Hemorrhagic cystitis after bone marrow transplantation. Transplantation 1993, 56, 875–879. [Google Scholar] [CrossRef]
  105. Dobrek, Ł.; Thor, P.J. Bladder urotoxicity pathophysiology induced by the oxazaphosphorine alkylating agents and its chemoprevention. Postepy Hig. Med. Dosw. (Online) 2012, 66, 592–602. [Google Scholar] [CrossRef]
  106. Gurtner, G.; Werner, S.; Barrandon, Y.; Longaker, M. Wound repair and regeneration. Nature 2008, 453, 314–321. [Google Scholar] [CrossRef]
  107. Barrientos, S.; Stojadinovic, O.; Golinko, M.S.; Brem, H.; Tomic-Canic, M. Growth factors and cytokines in wound healing. Wound Repair Regen. 2008, 16, 585–601. [Google Scholar] [CrossRef] [PubMed]
  108. Sawatdee, S.; Choochuay, K.; Chanthorn, W.; Srichana, T. Evaluation of the topical spray containing Centella asiatica extract and its efficacy on excision wounds in rats. Acta Pharm. 2016, 66, 233–244. [Google Scholar] [CrossRef] [PubMed]
  109. Tracy, L.E.; Minasian, R.A.; Caterson, E.J. Extracellular Matrix and Dermal Fibroblast Function in the Healing Wound. Adv. Wound Care 2016, 5, 119–136. [Google Scholar] [CrossRef]
  110. Salas-Oropeza, J.; Jimenez-Estrada, M.; Perez-Torres, A.; Castell-Rodriguez, A.E.; Becerril-Millan, R.; Rodriguez-Monroy, M.A.; Canales-Martinez, M.M. Wound healing activity of the essential oil of Bursera morelensis, in mice. Molecules 2020, 25, 1795. [Google Scholar] [CrossRef] [PubMed]
  111. Babawale, O.P.; Taiwo, F.; Adetunj, O.S. Ethnobotanical survey of plants used as memory enhancers in three states of southwestern Nigeria. J. Appl. Pharm. Sci. 2016, 6, 209–214. [Google Scholar] [CrossRef]
  112. Kennedy, D.; Okello, E.; Chazot, P.; Howes, M.J.; Ohiomokhare, S.; Jackson, P.; Haskell-Ramsay, C.; Khan, J.; Forster, J.; Wightman, E. Volatile Terpenes and Brain Function: Investigation of the Cognitive and Mood Effects of Mentha × Piperita L. Essential Oil with In Vitro Properties Relevant to Central Nervous System Function. Nutrients 2018, 10, 1029. [Google Scholar] [CrossRef]
  113. Mkaddem, M.; Bouajila, J.; Ennajar, M.; Lebrihi, A.; Mathieu, F.; Romdhane, M. Chemical composition and antimicrobial and antioxidant activities of Mentha (Longifolia L. and viridis) essential oils. J. Food Sci. 2009, 74, M358–M363. [Google Scholar] [CrossRef]
  114. Hong, Y.K.; Park, S.H.; Lee, S.; Hwang, S.; Lee, M.J.; Kim, D.; Lee, J.H.; Han, S.Y.; Kim, S.T.; Kim, Y.K.; et al. Neuroprotective effect of SuHeXiang Wan in Drosophila models of Alzheimer’s disease. J. Ethnopharmacol. 2011, 134, 1028–1032. [Google Scholar] [CrossRef]
  115. Gonçalves, J.C.; Oliveira, F.; Benedito, R.B.; de Sousa, D.P.; de Almeida, R.N.; de Araújo, D.A. Antinociceptive activity of (−)-carvone: Evidence of association with decreased peripheral nerve excitability. Biol. Pharm. Bull. 2008, 31, 1017–1020. [Google Scholar] [CrossRef] [Green Version]
  116. França, D.S.; Souza, A.L.; Almeida, K.R.; Dolabella, S.S.; Martinelli, C.; Coelho, M.M. B vitamins induce an antinociceptive effect in the acetic acid and formaldehyde models of nociception in mice. Eur. J. Pharmacol. 2001, 421, 157–164. [Google Scholar] [CrossRef]
  117. Millan, M.J. The induction of pain: An integrative review. Prog. Neurobiol. 1999, 57, 161–164. [Google Scholar]
  118. Beirith, A.; Santos, A.R.; Calixto, J.B. Mechanisms underlying the nociception and paw oedema caused by injection of glutamate into the mouse paw. Brain Res. 2002, 924, 219–228. [Google Scholar] [CrossRef]
  119. Oliveira, M.G.; Marques, R.B.; Santana, M.F.; Santos, A.B.; Brito, F.A.; Barreto, E.O.; Sousa, D.P.; Almeida, F.R.; Badauê-Passos, D.; Antoniolli, A.R.; et al. α-Terpineol reduces mechanical hypernociception and inflammatory response. Basic Clin. Pharmacol. Toxicol. 2012, 111, 120–125. [Google Scholar] [CrossRef] [PubMed]
  120. Kawata, J.; Kameda, M.; Miyazawa, M. Cyclooxygenase-2 inhibitory effects of monoterpenoids with a p-methane skeleton. Int. J. Essent. Oil Ther. 2008, 2, 145–148. [Google Scholar]
  121. Yamamoto, S.; Ohsawa, M.; Ono, H. Contribution of TRPV1 receptor-expressing fibers to spinal ventral root after-discharges and mechanical hyperalgesia in a spared nerve injury (SNI) rat model. J. Pharmacol. Sci. 2013, 121, 9–16. [Google Scholar] [CrossRef]
  122. Dhaka, A.; Murray, A.N.; Mathur, J.; Earley, T.J.; Petrus, M.J.; Patapoutian, A. TRPM8 is required for cold sensation in mice. Neuron 2007, 54, 371–378. [Google Scholar] [CrossRef]
  123. Thiaucourt, M.; Shabes, P.; Schloss, N.; Sack, M.; Baumgartner, U.; Schmahl, C.; Ende, G. Posterior insular GABA levels inversely correlate with the intensity of experimental mechanical pain in healthy subjects. Neuroscience 2018, 387, 116–122. [Google Scholar] [CrossRef]
  124. Sigel, E.; Steinmann, M.E. Structure, function, and modulation of GABAA receptors. J. Biol. Chem. 2012, 287, 40224–40231. [Google Scholar] [CrossRef]
  125. Ren, K.; Torres, R. Role of interleukin-1beta during pain and inflammation. Brain Res. Rev. 2009, 60, 57–64. [Google Scholar] [CrossRef]
  126. Heal, C.F.; Banks, J.L.; Lepper, P.D.; Kontopantelis, E.; van Driel, M.L. Topical antibiotics for preventing surgical site infection in wounds healing by primary intention. Cochrane Database Syst. Rev. 2016, 11, Cd011426. [Google Scholar] [CrossRef]
  127. Nedelec, B.; Ghahary, A.; Scott, P.G.; Tredget, E.E. Control of wound contraction. Basic and clinical features. Hand Clin. 2000, 16, 289–302. [Google Scholar] [CrossRef]
  128. Held, S.; Schieberle, P.; Somoza, V. Characterization of alpha terpineol as an anti-inflammatory component of orange juice by in vitro studies using oral buccal cells. J. Agric. Food Chem. 2007, 55, 8040–8046. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The pictures of the few representative plant species reported to have α-phellandrene [38].
Figure 1. The pictures of the few representative plant species reported to have α-phellandrene [38].
Scipharm 90 00057 g001
Figure 2. The batch process model of the synthesis of (S)—phellandrene [48]. Chemical structures were drawn using free online ChemDoodle Web software (https://web.chemdoodle.com/demos/2d-sketcher, accessed on 22 August 2022).
Figure 2. The batch process model of the synthesis of (S)—phellandrene [48]. Chemical structures were drawn using free online ChemDoodle Web software (https://web.chemdoodle.com/demos/2d-sketcher, accessed on 22 August 2022).
Scipharm 90 00057 g002
Figure 3. The illustration describes the process of (S)—phellandrene synthesis using a continuous-flow process [49]. Chemical structures were drawn using free online ChemDoodle Web software (https://web.chemdoodle.com/demos/2d-sketcher, accessed on 22 August 2022).
Figure 3. The illustration describes the process of (S)—phellandrene synthesis using a continuous-flow process [49]. Chemical structures were drawn using free online ChemDoodle Web software (https://web.chemdoodle.com/demos/2d-sketcher, accessed on 22 August 2022).
Scipharm 90 00057 g003
Figure 4. The microbial biotransformation of (−)-(R)-(α)-phellandrene produces different products namely (A) 5-p-menthene-1,2-diol, (B) 6-hydroxypiperitone, (C) α-PHE epoxide, (D) cis-p-menth-2-en-1-ol, (E) p-mentha-1(7), 5-dien-2-ol, (F) carvotanacetone, (G) 5-p-menthen-2-one [5]. Chemical structures were drawn using free online ChemDoodle Web software (https://web.chemdoodle.com/demos/2d-sketcher, accessed on 22 August 2022).
Figure 4. The microbial biotransformation of (−)-(R)-(α)-phellandrene produces different products namely (A) 5-p-menthene-1,2-diol, (B) 6-hydroxypiperitone, (C) α-PHE epoxide, (D) cis-p-menth-2-en-1-ol, (E) p-mentha-1(7), 5-dien-2-ol, (F) carvotanacetone, (G) 5-p-menthen-2-one [5]. Chemical structures were drawn using free online ChemDoodle Web software (https://web.chemdoodle.com/demos/2d-sketcher, accessed on 22 August 2022).
Scipharm 90 00057 g004
Figure 5. The biological functions of α-PHE involve insecticidal, anti-microbial, analgesic, immune, inflammatory, neural-response, anti-cancer, and wound-healing effects. (Figure created using BioRender.com accessed on 29 August 2022).
Figure 5. The biological functions of α-PHE involve insecticidal, anti-microbial, analgesic, immune, inflammatory, neural-response, anti-cancer, and wound-healing effects. (Figure created using BioRender.com accessed on 29 August 2022).
Scipharm 90 00057 g005
Figure 6. The possible mechanism of α-PHE: (A) α-PHE exhibit insecticidal activity by inducing neurotoxicity and death of insect in the larval stage. (B) Anti-microbial activity of α-PHE occurs by increased membrane permeability, fluidity, and leakage of ions from the microbial cells. (C) α-PHE showed anti-nociception activity by regulating neurotransmitters through various signaling pathways such as opioid, GABA, glutamatergic, nitrergic, cholinergic and adrenergic systems. (D) The immune response of α-PHE is exhibited through macrophages and NK cell activation, T-cell and B-cell proliferation. (E) Anti-cancer activity of α-PHE is associated with inducing autophagy-associated protein pathways and DNA damage. (F) The anti-inflammatory properties of α-PHE are carried out by reducing TNF-α, and IL6, regulating other inflammatory mediators, and preventing mast cell degranulation. (G) α-PHE wound healing property works by inhibiting COX II, LOX 5, TNF-α, IL6, and superoxide anions and inducing activated fibroblasts’ migration to the wound site. (Figure created using BioRender.com, accessed on 29 August 2022).
Figure 6. The possible mechanism of α-PHE: (A) α-PHE exhibit insecticidal activity by inducing neurotoxicity and death of insect in the larval stage. (B) Anti-microbial activity of α-PHE occurs by increased membrane permeability, fluidity, and leakage of ions from the microbial cells. (C) α-PHE showed anti-nociception activity by regulating neurotransmitters through various signaling pathways such as opioid, GABA, glutamatergic, nitrergic, cholinergic and adrenergic systems. (D) The immune response of α-PHE is exhibited through macrophages and NK cell activation, T-cell and B-cell proliferation. (E) Anti-cancer activity of α-PHE is associated with inducing autophagy-associated protein pathways and DNA damage. (F) The anti-inflammatory properties of α-PHE are carried out by reducing TNF-α, and IL6, regulating other inflammatory mediators, and preventing mast cell degranulation. (G) α-PHE wound healing property works by inhibiting COX II, LOX 5, TNF-α, IL6, and superoxide anions and inducing activated fibroblasts’ migration to the wound site. (Figure created using BioRender.com, accessed on 29 August 2022).
Scipharm 90 00057 g006
Table 1. The list of representative plant species that are reported to have α-Phellandrene.
Table 1. The list of representative plant species that are reported to have α-Phellandrene.
S. No.Plant Speciesα-Phellandrene Content (%) #References
1Curcuma zedoaria (Christm)14.93[20]
2Xylopia aromatica L.2.2–6.4[21]
3Rosmarinus officinalis L.0.1–0.4[22]
4Eucalyptus dives17.4[23]
5Eucalyptus staigeriana8.8[23]
7Schinus terebinthifolius Raddi15.7[24]
34.38[25]
6Schinus molle L.46.52[25]
8Solanum erianthum D. Don.17.5[26]
9Thymus kotschyanus Boiss. and Hohen.10.8[27]
10Cupressus atlantica Gaussen5.5[28]
11Anethum graveolens L.32[29]
12Myrica gale L.8[30]
13Ligusticum mutellina L. Crantz23.4[31]
14Ligusticum marginatum50.2[32]
15Artemisia feddei5.78[33]
16Bursera morelensis1[34]
17Monodora myristica (Gaertn.)34.4[35]
Monodora myristica (Gaertn.) Dunal53[36]
18Piper nigrum8.56[37]
# α-PHE content (%) in the essential oil of the respective plants.
Table 2. The reported bioactivities of α-phellandrene.
Table 2. The reported bioactivities of α-phellandrene.
Study ModelDetails of α-PHE and EO UsedExperimental ConditionStudy ResultsReferences
Insecticidal activity
Third instar stage larvae (L3) of the Australian blowfly Lucilia cuprinaCommercial α-PHE and Curcuma longa leaves EO (CLLEO).L3 larvae were kept on tissue paper infused with 0.15–2.86 mL/cm2 of CLLEO and 0.29–1.47 mL/cm2 of α-PHE dissolved in ethanol. Toxicity was observed at 6, 24 and 48 h.
  • Intensive darkening of the L3 body.
  • Reduced movement and color changes in the cuticle of the larva and dead after 6 and 24 h of α-PHE and CLLEO treatment.
[59]
Third instar stage larvae (L3) of the Australian blowfly Lucilia cuprinaCommercial α-PHE and Curcuma longa leaves EO (CLLEO).L3 larvae were kept on tissue paper infused with 0.15–2.86 mL/cm2 of CLLEO and 0.29–1.47 mL/cm2 of α-PHE s dissolved in ethanol. Toxicity was observed at 6, 24 and 48 h.
  • CLLEO and α-PHE exposure cause damage to the cuticle, brain, and fat body of L3.
  • Vacuolar degeneration and pyknotic profiles detected in L3 brain.
  • Reduced neurotoxicity.
[60]
Anti-microbial activity
Penicillium cyclopiumCommercial α-PHE and Nonanal 6mm diameter discs of P. cyclopium inoculum were taken out from culture and placed at the center of Petri dishes containing α-PHE (0, 0.25, 0.50, 0.75, 1.00, 1.25, 1.50, 1.75 and 2.00 mL/L) and Nonanal (0, 50, 100, 150, 200, 250, 300, 350 and 400 μL/L)
  • Mycelial growth of P. cyclopium was affected depends on the dosage.
  • The cell constituents were released.
  • Shrunken mycelium, depressed conidia and distorted hyphae were observed.
[14]
Swiss male mice and Wistar male rats. Macrophages were isolated from the peritoneal cavity of the mice.Commercial α-PHEMice were treated with α-PHE and saline before (1 h) the treatment with 0.75% acetic acid, 2% formalin, capsaicin (2 mg/paw), glutamate (20 mmol/paw), 1% carrageenan.
The anti-nociceptive effect of α-PHE and mechanical hypernociception were analyzed.
  • Oral administration of α-PHE significantly reduced abdominal writhing induced by acetic-acid and capsaicin-induced licking response.
  • 50 mg/kg α-PHE reduced licking response in the formalin test.
  • α-PHE reduced licking and biting response in glutamate test.
  • α-PHE 50 mg/kg significantly reduced withdrawal response in carrageenan-induced hypernociception test.
[16]
Spared Nerve Injury (SNI) induced hyperalgesia in Wistar rats.Commercial (R)-(+)-phellandrene, α-limonene and Schinus terebinthifolius fruits EOCommercial (R)-(+)-phellandrene (10 mg/kg) and α-limonene (10 mg/kg) and S. terebinthifolius EO (3, 30 and 100 mg/kg) were treated with SNI-induced hyperalgesia rats.
  • Reduced mechanical induced hypernociception and depression-like behavior.
  • (R)-(+)-limonene and α-PHE reduced mechanical hyperalgesia and depressive-like activity.
  • α-PHE inhibited mechanical and cold sensitivity and the Forced swim test.
[15]
S-180 inoculated Swiss mice female. Melanoma B-16/F-10 and S-180 murine cells.Commercial α-PHEAnimals were separated into seven groups as follows: Sham treated, vehicle-treated (pregabalin 10 mg/kg), positive control (25 mg/kg of 5-fluorouracil), and α-PHE in doses of 6.25, 12.5, 25 and 50 mg/kg orally by gavage once a day.
  • Mechanical sensitivity in mice increased significantly.
  • α-PHE treatment showed antinociceptive responses.
  • α-PHE treatment showed anti-proliferative activity.
  • α-PHE treatment reduced malondialdehyde, and TNF-α and IL-1β.
[61]
Immune responses
BALB/c miceCommercial α-PHEBALB/c mice were divided into five groups; I: normal diet; II: olive oil vehicle as positive control; other groups III, IV and V: α-PHE at 1, 5 and 25 mg/kg dissolved in olive oil for 27 days.
  • α-PHE treatment did not significantly affect body weight.
  • α-PHE treatment increased CD3, CD 11b, CD19, and Mac-3 levels.
  • α-PHE significantly enhanced the phagocytic activity.
  • Significantly increased the NK cell activity and B-cell and T-cell proliferation.
[62]
Litopenaeus vannamei White shrimpCommercial α-PHEL. vannamei was administered with α-PHE (4,8 and 12 μg/g) and challenged against V. alginolyticus (2 × 105 CFU/shrimp).
Non-pre-treated shrimps as controls.
Shrimp survival was observed.
The immune responses, gene expression and hemocytes morphology, were studied.
  • Survival of shrimp increased.
  • PO, RB, and SOD activities, Phagocytic and lysozyme activities were higher in the α-PHE treated group.
  • The expression of immune genes was increased.
  • α-PHE treatment reduced the flattening of hemocytes and pseudopodia.
[63]
Anti-cancer activity
Human liver cancer (J5) cellsCommercial α-PHEJ5 cells were treated with 0, 10, 30, or 50 mM of α-PHE for 0, 6, 12, 24, or 48 h. The cytotoxic effects and expressions of Bax, Bcl-2, PARP, and caspase-3 proteins were studied. The necrotic cells level, ROS, MMP, ATP and LDH levels in J5 cells were investigated.
  • α-PHE treatment reduced the viability of J5 cells.
  • Bax, Bcl-2, PARP and caspase-3 protein levels were not changed.
  • ROS level was increased, and MMPs were decreased.
  • ATP level was decreased.
  • LDH leakage, NO production and J5 necrotic cell death were increased.
[17]
Human liver cancer (J5) cellsCommercial α-PHEJ5 cells were treated with 0, 10, 30, or 50 mM of α-PHE. The expression levels of cytoplasmic proteins PI3K-I, PI3K-III, Akt, mTOR, phosphorylated Bcl-2 (p-Bcl-2), Beclin-1, LC3-II, p53, TIGAR, phosphorylated IκB (p-IκB), IκB, and NF-κB (p65) and nuclear p53, DARM, and NF-B were evaluated.
  • α-PHE induced apoptosis and autophagy in J5 cells.
  • PI3K-I, Akt proteins reduced, mTOR proteins increased the phosphorylation of Bcl-2, Beclin-1, PI3K-III, and LC3-II proteins.
  • α-PHE induces pre-autophagosome formation.
  • Increased Nuclear p53 and DRAM proteins. Decreased cytoplasmic p53, and TIGAR proteins
  • α-PHE was involved in autophagosome formation.
[64]
WEHI-3 murine leukemia cellsCommercial α-PHEWEHI-3 cells were treated with α-PHE (0, 10, 20, 30, and 50 µM) for 48 h. DNA damage and condensation and DNA repair associated-proteins expression were analyzed.
  • α-PHE significantly reduced cell viability and induced DNA damage, condensation, and fragmentation in a dose-dependent way.
  • α-PHE decreased p53, MGMT, DNA-PK, and BRCA-1 expression.
  • Increased p-p53, p-H2A.X, 14-3-3σ, and MDC1 expression.
[65]
Male BALB/c mice injected with WEHI-3 murine leukemia cellsCommercial α-PHEWEHI-3 injected mice were treated with 25 and 50 mg/kg of α-PHE. After 2 weeks, immune responses and macrophage phagocytic and NK cell activities were studied.
  • α-PHE treatment reduced the spleen weight of WEHI-3 treated mice.
  • α-PHE treatment increased CD3 and CD11b and reduced the CD19.
  • α-PHE treatment increased the Mac-3 cell population.
  • α-PHE treatment affects phagocytic activity and NK cell activity.
  • α-PHE treatment increased the T-cell and B-cell proliferation.
[66]
WEHI-3 murine leukemia cell lineCommercial α-PHEWEHI-3 cells were treated with 10 μM of α-PHE or vehicle for 48 h. The cells were analyzed for expression of DNA damage-related genes, cell cycle and apoptotic cell death.
  • α-PHE treatment upregulated the DNA damage inducer transcript-4, DNA fragmentation factor, cyclin G2, cyclin-dependent kinase inhibitor 2D and IA (p21) and apoptosis genes such as BCL2/adenovirus EIB interacting protein 3, associated factor 3, BCL2-modifying factor, caspase-8.
  • DNA damage-associated genes TATA box BP, cyclin F2 and death box polypeptide 33, apoptosis genes growth arrest-specific 5 and ATP synthase were down-regulated.
[67]
WEHI-3 murine leukemia cell lineCommercial α-PHEWEHI-3 cells were treated with 0, 5, 10, 30, 40, and 50 µM of α-PHE.
The cytotoxicity effect of α-PHE was assessed.
  • α-PHE causes damage to leukemia cells in a dose-depended manner.
  • α-PHE treatment affected ROS production.
  • α-PHE induces Ca2+ production and caspase 8, 9 and 3 in a time-dependent manner.
  • α-PHE induces mitochondria-dependent apoptosis.
  • The level of Fas, Fas-L, Caspase-8, Bax, Bad, tBid, AIF, Endo G, cytochrome c, Caspases-9,3 and 12, PARP, ATF-6a, GRP78 and GADD153 were increased after α-PHE treatment.
  • α-PHE decreased Bcl-2 and Mcl-1 expressions in WEHI-3 cells.
[18]
Anti-inflammatory activity
Wistar male rats and Swiss miceCommercial α-PHEBefore carrageenan injection, rats or mice were treated with vehicle or α-PHE (50, 100, or 200 mg/kg) or dexamethasone (0.5 mg/kg).
  • Carrageenan injection induced neutrophil migration.
  • Pre-treatment with α-PHE (50, 100, and 200 mg/kg) significantly prevented inflammatory cell accumulation in the cavities.
  • α-PHE attenuated TNF-α, IL-6 expression.
  • α-PHE inhibited leukocyte rolling and prevented mast cell degranulation.
[68]
Wistar male rats and Swiss miceCommercial α-PHEBefore carrageenan injection, rats or mice were treated with vehicle or α-PHE (50, 100, or 200 mg/kg) or dexamethasone (0.5 mg/kg).
  • α-PHE treatment reduced histamine and Substance P-induced paw edema.
  • α-PHE treatment reduced ear edema induced by croton oil, TPA and arachidonic acid.
[69]
Male Mus musculus mice of Swiss albino lineage.Commercial α-PHEMice were treated with tween (vehicle control), ifosfamide (IFOS; negative control), Mesna (positive control), and α-PHE.
  • IFOS-induced urinary bladder toxicity was prevented by α-PHE treatment.
  • α-PHE treatment decreased the activity of myeloperoxidase, malondialdehyde, nitrite/nitrate, and prevented the depletion of superoxide dismutase and reduced glutathione in bladder tissues.
  • α-PHE significantly reduced TNF-α but did not affect the IL-1β level.
  • α-PHE treatment showed anti-inflammatory properties against pathological conditions.
[70]
Wound healing activities
Fibroblasts (L929), macrophages and human embryonic renal cells (HEK 293) transfected with the luciferase-expressing gene Commercial α-PHE and TerpinoleneFor the cytotoxicity study: Fibroblasts and macrophages were treated with terpinolene and α-PHE.
For wound healing study: Fibroblasts were exposed to 10, 100 and 200 μM of terpinolene and α-PHE.
  • α-PHE showed better antioxidant activity than terpinolene.
  • Terpinolene and α-PHE did not exhibit a cytotoxic effect against macrophages but enhanced proliferation and migration of fibroblasts in a dose-dependent way.
  • Suppressed the NO production and inhibited the superoxide anion in LPS-stimulated macrophages. Terpinolene and α-PHE reduced the release of IL-6, TNF-α, and NF-kB.
[71]
Male CD-1 mice and fibroblastsCommercial α-PHE and α-PineneThe mice’s back skin was shaved, and an incision was made.
Mice were divided into groups I: untreated skin without wounds, group II: untreated wounds (negative control), group III: wounds treated with Recoverón NC® (positive control), group IV: cosmetic grade mineral oil (vehicle), and the next four groups treated with α-pinene and α-PHE at 0.1 and 0.01 mg/mL concentrations for 10 days.
  • α-pinene and α-PHE treatment did not affect the cell viability and did not stimulate fibroblast proliferation.
  • No alteration in morphology of fibroblasts.
  • α-pinene and α-PHE showed the highest wound closing speeds.
  • Histopathological study showed that α-pinene and α-PHE-treated skin presented some dermal collagen fiber bundles and blood microvasculature vessels inside the mononuclear infiltrate.
[34]
CLLEO, Curcuma longa leaves essential oil; SNI, spared nerve injury; EO, essential oil; TNF-α, tumor necrosis factor-α; IL-1β, interleukin 1β; CD3, CD 11b, and CD19, cluster of differentiation 3, 11b, and 19; Mac-3, macrophage; NK cell, natural killer cell; B cell, B lymphocyte; T cell, T lymphocyte; CFU, colony forming unit; PO, phenoloxidase, RB, respiratory bursts, SOD, superoxide dismutase; THC, total hemocyte count; ROS, reactive oxygen species; MMP, mitochondrial membrane potential; ATP, adenosine tri-phosphate; LDH, lactate dehydrogenase; PI3k, phosphatidylinositol 3 kinases; Akt, protein kinase B; mTOR, mammalian target of rapamycin; TIGAR, Tp53-induced glycolysis and apoptosis regulator; DRAM, damage-regulated autophagy modulator; NF-κB, nuclear factor-κB; MGMT, O6-methylguanine-DNA methyltransferase; DNA-PK, DNA-dependent protein kinase; BRCA-1, breast cancer protein; p-H2A.X, phosphorylated histone 2A X variant protein; MDC-1, mediator of DNA damage check point 1 protein; PARP, poly ADP ribose polymerase; ATF, activating transcription factor; GRB, glucose regulated protein; GADD, growth arrest and DNA damage inducible protein; TPA, 12O-tetradecanoylphorbol-13-acetate; IFOS, ifosfamide; LPS, lipopolysaccharide.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Thangaleela, S.; Sivamaruthi, B.S.; Kesika, P.; Tiyajamorn, T.; Bharathi, M.; Chaiyasut, C. A Narrative Review on the Bioactivity and Health Benefits of Alpha-Phellandrene. Sci. Pharm. 2022, 90, 57. https://doi.org/10.3390/scipharm90040057

AMA Style

Thangaleela S, Sivamaruthi BS, Kesika P, Tiyajamorn T, Bharathi M, Chaiyasut C. A Narrative Review on the Bioactivity and Health Benefits of Alpha-Phellandrene. Scientia Pharmaceutica. 2022; 90(4):57. https://doi.org/10.3390/scipharm90040057

Chicago/Turabian Style

Thangaleela, Subramanian, Bhagavathi Sundaram Sivamaruthi, Periyanaina Kesika, Tanawat Tiyajamorn, Muruganantham Bharathi, and Chaiyavat Chaiyasut. 2022. "A Narrative Review on the Bioactivity and Health Benefits of Alpha-Phellandrene" Scientia Pharmaceutica 90, no. 4: 57. https://doi.org/10.3390/scipharm90040057

Article Metrics

Back to TopTop