Next Article in Journal
Old and Young Actors Playing Novel Roles in the Drama of Multiple Myeloma Bone Marrow Microenvironment Dependent Drug Resistance
Next Article in Special Issue
The Extracts and Major Compounds Derived from Astragali Radix Alter Mitochondrial Bioenergetics in Cultured Cardiomyocytes: Comparison of Various Polar Solvents and Compounds
Previous Article in Journal
Identification of Novel Somatic TP53 Mutations in Patients with High-Grade Serous Ovarian Cancer (HGSOC) Using Next-Generation Sequencing (NGS)
Previous Article in Special Issue
Antimalarial Activity of Plant Metabolites
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Copaifera of the Neotropics: A Review of the Phytochemistry and Pharmacology

1
Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Pará, 66075-900 Belém, Brazil
2
Programa de Pós-Graduação em Química, Universidade Federal do Pará, 66075-900 Belém, Brazil
3
Department of Chemistry, University of Alabama in Huntsville, Huntsville, AL 35899, USA
4
Aromatic Plant Research Center, 615 St. George Square Court, Suite 300, Winston-Salem, NC 27103, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2018, 19(5), 1511; https://doi.org/10.3390/ijms19051511
Submission received: 25 April 2018 / Revised: 15 May 2018 / Accepted: 15 May 2018 / Published: 18 May 2018
(This article belongs to the Special Issue Plant Natural Products for Human Health)

Abstract

:
The oleoresin of Copaifera trees has been widely used as a traditional medicine in Neotropical regions for thousands of years and remains a popular treatment for a variety of ailments. The copaiba resins are generally composed of a volatile oil made up largely of sesquiterpene hydrocarbons, such as β-caryophyllene, α-copaene, β-elemene, α-humulene, and germacrene D. In addition, the oleoresin is also made up of several biologically active diterpene acids, including copalic acid, kaurenoic acid, alepterolic acid, and polyalthic acid. This review presents a summary of the ecology and distribution of Copaifera species, the traditional uses, the biological activities, and the phytochemistry of copaiba oleoresins. In addition, several biomolecular targets relevant to the bioactivities have been implicated by molecular docking methods.

Graphical Abstract

1. Introduction to the Genus Copaifera

The copaiba trees belong to the genus Copaifera, family Fabaceae, and subfamily Caesalpinoideae. The genus was described the first time by Marcgraf and Piso in 1638, who employed the name “Copaiba” without designating the species [1]. In 1760, Nicolaus Joseph Von Jacquin described the species Copaiva officinalis in the work Enumeratio Systematica Plantarum [2]. Afterwards, in the year 1764, Carl von Linnaeus did a more detailed study of the genus in the work Species Plantarum, in which he described the type species Copaifera officinalis (Jacq.) L. [3]. There are more than 70 Copaifera species distributed throughout the world, with widespread occurrence in Central and South America; there are also four species found in Africa and one species found on the island of Borneo, situated in the Pacific Ocean [4]. Brazil is the country with the greatest biodiversity of Copaifera with 26 species and 8 varieties [5].
The vernacular name copaíba probably originated from the Tupi-Guarani and alludes to the names used by indigenous peoples, copaíva and copahu (kupa’iwa and kupa’u, respectively), which refers to the tree exudate, in reference to the oil stored in its interior [6]. Sixteenth-century records produced by chroniclers during the Brazilian colonization report the widespread use of copaiba oil among the natives as anti-inflammatory and healing agents, and also for esoteric purposes, such as aphrodisiac and contraceptive [4,6,7]. This natural product is known and valued to the present day, mainly in the Amazon region, where the rural population has little access to industrialized pharmaceutical products and public health care [6,8].
The copaiba trees have shrub or arboreal habits, can reach up to 40 m height and 4 m diameter at breast height (dbh), have slow growth, and can live up to 400 years [6]. Their cylindrical trunks contain intercellular secretory channels arranged in bands of marginal axial parenchyma, the lumen from secretory cells is formed schizogenously, and the oleoresin is synthesized in parenchyma cells of the canal. The species have alternate leaves, which are pinnate with 2–12 pairs of leaflets (opposite, alternate, or subopposite), usually glabrous, and may have translucent points and glands at the base of the marginal vein; they have small and interpetiolar stipules and are generally deciduous. The inflorescences are alternate panicles and the flower buds are protected by small bracts; they have small flowers, numerous and sessile, which are monoclamids with a tetramer chalice that forms short tubes and contains internally hirsute sepals. The androecium holds 10 free stamens, glabrous fillets, and oblong and rimose anther; and the gynoecium presents a sessile ovary with two elongate ovules, filiform style, and globular and papillary stigma. The fruits are bivalved, dehiscent, laterally compressed, and monospermic. The seed is a pendulum, oblong-globose, covered by abundant white or yellow aril, and lacking endosperm [1,9,10,11].
Although the Copaifera genus has been extensively studied taxonomically, there are still difficulties in identifying some species, mainly due to their intricate floral morphology and absence of reproductive structures in the samples studied. With regard to the Amazonian species, the scarcity of field information and illustrations of specimens comprise the main limitations for botanical descriptions of the group. These taxonomic problems have restricted the advance of chemical and pharmacological research, limited the industrial and rational uses of resin oils and wood, and have also hampered the development of projects, plans for sustainable management, and conservation of commercially targeted species [9,12].
The main economic contributions of Copaifera species have been wood and oleoresins. Among Copaifera species that are used in the production of oleoresins, C. reticulata is the most frequent, representing 70% of the production [6]. Copaiba oleoresin is one of the most important renewable natural remedies for the indigenous people from the Amazon region and its use is widely diffused due its various pharmacological properties [13]. The oleoresin is a transparent, colored liquid with variable viscosity, and is constituted by a nonvolatile fraction composed of diterpenes and a volatile fraction composed of sesquiterpenes [14,15]. Its chemical profile may vary according to species, seasonal and climatic characteristics of the environment, soil type and composition, and rainfall index. Biotic pressures, such as insect predation and pathogen infection, also cause differences in oleoresin composition [16,17]. The extraction of copaiba oil is done through the perforation of the trunk with a punch, and the resin is collected with the help of a polyvinyl chloride (PVC) pipe, through which the oil flows and is then stored. This practice is mainly done by plant extraction; therefore, the product of several trees is often mixed, resulting in an additional obstacle to the botanical identity of the copaiba trees. In addition, the lack of parameters to characterize the oil and to perform quality control of the botanical drug also constitutes an obstacle for the registration and exportation of herbal products containing copaiba [18,19].

2. Ecology and Distribution of Copaifera

The genus Copaifera is native to tropical regions of Latin America, an area of great species diversity [1]. Distributed widely in the Americas, stretching from Mexico to northern Argentina, the genus also occurs in West Africa and Asia [20]. The greatest richness of species occurs in Brazil, where they are distributed from the north to the south of the country. The most common species are C. multijuga Hayne, which is found in the Amazonas, Pará and Rondônia states; C. reticulata Ducke that occurs in Amapá, Pará and Roraima;, and C. langsdorffii Desf., which can occur from the northern to southern regions of Brazil [5]. Other species have more restricted distribution, such as C. guyanensis Desf. (Amazonas), C. majorina Dwyer (Bahia), C. cearensis Huber ex Ducke (Ceará, Bahia, Piauí and Rio de Janeiro), C. elliptica Mart. (Goias and Mato Grosso), C. paupera (Herzog) Dwyer (Acre), and C. lucens Dwyer (Bahia, Espírito Santo, Minas Gerais, Rio de Janeiro, São Paulo) [5]. Although many species of Copaifera have wide occurrence within the Brazilian territory, and may occur in different phytogeographic domains (e.g., C. langsdorffii), some feature endemism, such as C. trapezifolia Hayne, which occurs in an extremely disturbed region of the Atlantic rainforest, of which only 11.6% of the natural vegetation cover remains [21]. Thus, morphological, physiological, and ecological studies are highly relevant for the preservation of species and their natural environment [11]. A study conducted in the Minas Gerais state on the geographical distribution and environmental characteristics of arboreal species showed that C. langsdorffii has wide occurrence throughout the whole state, where latosol type soil predominates, but additionally has a preference for ustic soils (62%) [22].
In relation to the ecological group, copaíba are classified as long-living, late secondary, and climax tree species, demanding of light but tolerant to shade [23]. They are considered generalists because they are adapted to a wide variety of environments. They can occur in floodplains, riparian forest, and streams of the Amazon basin and the forests of the Cerrado in the center of Brazil [24]. C. langsdorffii, for example, has great ecological plasticity, occurring in several biomes, such as Cerrado, Atlantic Forest, Caatinga, and Amazon rainforest [23]. Copaifera species have great plasticity in relation to edaphic conditions; they occur in areas with fertile soil and well-drained soil and in areas with very poor acidic soils, such as Cerrado fields. They grow well on sandy and clayey soils and generally occupy the forest canopy [25,26].
Phenological studies on Copaifera are important for the rational use of the species and for the preparation of management plans [27]. The reproduction of copaibas occurs from the fifth year of growth after planting in a climax forest ecosystem [6]. C. multijuga, commonly found in the Amazon, blooms in the rainy season—between the months of December–April—and fructifies between April and July [27]. Blooming of C. reticulata occurs from January to March, with fruiting from March to August, lasting into October [26,28]. C. langsdorffii, observed in the Tijuca Forest, Rio de Janeiro, blooms between March and April and fructifies between August and September. Another survey carried out near Campinas, São Paulo state, showed that flowering of C. langsdorffii occurs in the middle of the rainy season (December–February), with development of fruit during the dry season (April–September) [29,30]. The phenophases of C. officinalis were monitored in the municipality of Boa Vista (Roraima state, Brazil), and showed that the flowering of the species occurs between the months of September and November and the fruiting from November to March. Depending on the stage of fruit ripening, the dehiscence can begin in January, in which the seeds enveloped by the aril are exposed, allowing for their dispersal [28].
Copaifera is a hermaphrodite plant of mixed reproduction with a predominance of allogamy. The trees are generally bee pollinated (melittophily), and Apis mellifera and Trigona spp. are its main pollinating agents [25]. C. langsdorffii has high fecundity, producing large quantity of fruits in a short period of time. Its seeds have low nutritional value, mainly composed of carbohydrates, but can attract a wide variety of animals with a general diet [30]. The dispersion of the copaiba seeds occurs mainly in zoocoric and barocoric forms [27,28]. Some vertebrates, such as birds and mammals, have been observed visiting the fruits of Copaifera [31]. Its seeds have morphological characteristics that fit the ornithocoria syndrome, mainly because they are black with colored, fleshy arils, which, after being swallowed, can be regurgitated intact and remain viable for germination [32]. A study revealed that 10 species of Passeriformes, such as Ramphastos toco, Cyanocorax cristatellus, and Turdus rufiventris, visited the fruits of C. langsdorffii. Likewise, monkeys of the species Eriodes arachnoides and Cebus paella also eat the fruits of C. langsdorffii [31]. Copaiba seeds may also present hydrocoric dispersion due to their frequent occurrence near waterways [25]. Copaiba seeds are of conventional behavior and may be conserved in the long term ex situ, with dormancy due to the deposition of coumarin in the tegument, and its germination is of the epigene type [25]. A tree can produce from 2 to 3 kg of seeds [33].
The population density of copaiba trees in an area is usually very low. It is possible to find only one tree every 5 ha, but they may occur in densities of one to two trees per hectare. The production of oleoresin by species is fairly variable and can be influenced by genetic differences among species, habitat, soil, and intensity of exploitation [34]. The production of oleoresin per tree ranges from 100 mL to 60 L per year. In addition, not all trees produce oil [24]. Therefore, detailed investigations regarding extraction methods and equipment that do not harm the plant, correlation of genetic data to botanical identification of species, floristic inventory of copaiba populations, and ecological studies on its ecosystems are indispensable for the sustainable and rational use of this resource [35,36].

3. Traditional Uses of Copaifera

3.1. Medicinal Uses

In Pará state (Amazon region, Brazil), people of all ages and social classes consider copaiba one of the most important natural remedies from the Amazon region. Several parts and preparations of the plant are used in folk medicine [24]. The oleoresin or bark decoction is used as an anti-inflammatory and contraceptive by native people from the Brazilian Amazon. The topical application of oil on the skin serves to heal wounds. It is used in massages on the head to cure paralysis, pains, and convulsions. In Amapá state, it is recommended to soak a cotton ball in oil and place on tumors, ulcers, or hives. The daily intake of two drops of oil mixed with one tablespoon of honey is indicated for inflammation, syphilis, bronchitis, and cough [6,37,38]. In Venezuela, the oil is used to prepare a patch that is applied to heal ulcers and wounds, and the decoction of the bark in the form of a bath is used to combat rheumatism, to wash infected wounds such as dog bites, and to use as an anti-tetanus [37,38]. A tea from the seeds is also used as a purgative and for treatment of asthma. In northern Brazil, the practice of “embrocation” (applying oil directly to the throat) is common to treat throat infections [39]. In Belém, the “garrafada”—an infusion of the bark sold in bottles—is currently used as a substitute for the oleoresin due to the difficulty in obtaining the oil in the city [38].
Copaiba has a wide range of ethnopharmacological indications, including for the treatment of: cystitis, urinary incontinence, gonorrhea, and syphilis; respiratory ailments, including bronchitis, strep throat, hemoptysis, pneumonia, and sinusitis; infections in the skin and mucosa, such as dermatitis, eczema, psoriasis, and wounds; ulcers and lesions of the uterus; leishmaniasis and leucorrhea; anemia; headaches; and snake bites. It is also used for its aphrodisiac, stimulant, anti-inflammatory, antiseptic, anti-tetanus, antirheumatic, antiherpetic, anthelminthic, anticancer, antitumor (prostate tumors), and antiparalytic properties [4,6,26,38,40]. Copaifera species are used by people of Igarapé Miri (Pará state) for healing wounds [41].
Studies have shown that the ingestion of high doses of copaiba oil can cause adverse side effects, such as gastrointestinal irritation, sialorrhea, and central nervous system depression. A dose of 10 g may cause symptoms of intolerance, nausea, vomiting, colic and diarrhea, and exanthema. Prolonged use may cause kidney damage and topical reactions in susceptible individuals [39,42]. Thus, the advance in pharmacological and quality control studies of copaiba formulations sold at herbal markets is indispensable for the safe use of this plant drug.

3.2. Human Nutrition

Copaiba oil was approved in the United States as a food additive and is used in small amounts as a flavoring agent in foods and beverages [43].

3.3. Cosmetic Uses

The species of Copaifera are intensively pursued for inclusion in the cosmetics market due to their therapeutic properties and fragrant value of their oils [44]. Copaiba oil is currently used in the cosmetic industry as a fixative for perfumes and perfuming soaps [38]. As an emollient, bactericidal, and anti-inflammatory agent, copaiba oil is used in the production of soaps, lotions, creams and moisturizers, bath foams, shampoos, and hair conditioners [6,24]. In addition, it aids in the treatment of dandruff and acne [38,45]. Despite its fragrant value, little information regarding its odorant potential is available in the literature [44].

3.4. Fuel

As a renewable source of hydrocarbons, the use of copaiba oil as an ecologically clean fuel has been evaluated. Experimental plantations were started in the early 1980s near Manaus, Brazil to test its viability as an alternative energy source to fossil fuels [7]. For potential use as fuel, a combination with diesel oil in a ratio of 9:1 (diesel oil to copaiba) has been recommended [6]. Various reports indicate that the liquid can be poured directly into the fuel tank of a diesel-powered car and the vehicle will run normally, with a bluish exhaust smoke being the only noticeable difference [46]. Traditionally, the oil is used in lamps as fuel for lighting [24].

3.5. Wood

The copaiba trees are considered hardwoods with high demand due to their properties of strength, as well as insect and xylophagous fungi repellency. The wood is saturated with oil and resin and has been used in both shipbuilding and civil construction, especially in the manufacture of steam caves, pool cues, and decorative and furniture coverings. It is also used in the preparation of lumbers, rafters, door and window frames, and boards in general, including for agricultural implements, general carpentry, flooring furniture, coatings, lamination, plywood sheets. The wood has a high content of lignin and is very good for the production of alcohol and charcoal. C. langsdorffii has traditionally been exploited extensively for charcoal in the Cariri Region, south of Ceará [24,47].

3.6. Veterinary Uses

In southern Pará state, farmers have used copaiba oil to prevent foot-and-mouth infection in cattle. The oil is poured on the floor next to the salt lick so that when cattle approach to eat salt, they step in oil soaking their feet [24]. When wounded, some animals lick and rub their bodies in the oil that flows from the trees [24].

3.7. Other Uses

Hunters often hunt under the copaiba tree during fruiting because the seeds and oil attract animals [24]. The oleoresin is used in the photographic industry to improve image clarity in areas of low contrast and resolution. The resin has also been used in paper making, as an additive for butadiene in the production of synthetic rubber, as a source of a chiral substrate in the synthesis of biomarkers of sediment and oil residues, and as fixative in the manufacture of varnish, perfume, and paints used in the painting of porcelain, fabrics, and for dying cotton yarn [6,24,38].

4. Essential Oil Chemistry of Copaifera

The major components of the essential oils from Copaifera species are summarized in Table 1. In general, copaiba oils derived from Copaifera oleoresins are rich in sesquiterpene hydrocarbons and often dominated by β-caryophyllene [15]. Some copaiba oils, however, also show significant concentrations of diterpene acids, which are generally analyzed as their methyl esters [15]. A perusal of internet sources of copaiba oil suggests that the most important commercial sources of copaiba oil are C. langsdorffii, C. officinalis, and C. reticulata, and the most prized copaiba oils are rich in β-caryophyllene. The oleoresin essential oils from these three Copaifera species can have as much as 33% (C. langsdorffii), 87% (C. officinalis), and 68% (C. reticulata) β-caryophyllene (see Table 1).

5. Nonvolatile Chemistry of Copaifera

The oleoresins of several Copaifera species have been shown to be rich sources of clerodane, kaurane, and labdane triterpenoids (Figure 1, Figure 2 and Figure 3, Table 2). In particular, C. langsdorffii resin is composed of biologically active copalic acid [70,71] and kaurenoic acid [72,73,74]. C. multijuga [74] and C. paupera [75] resins are also good sources of copalic acid.

6. Biological Activities of Copaifera

Copaifera oleoresins have shown remarkable biological activities, many of which have been attributed to diterpenoid acids (see Table 3). Generally, Copaifera oleoresins and their diterpenoid constituents have shown antibacterial, anti-inflammatory, antileishmanial, antiproliferative, antitrypanosomal, and wound-healing activities.

6.1. Antiparasitic Activity of Copaiba

Several Copaifera oleoresin oils have shown in vitro antiparasitic activity against Leishmania amazonensis promastigotes, including C. cearensis, C. langsdorffii, C. lucens, C. martii, C. multijuga, C. officinalis, C. paupera, and C. reticulata [48]. The resin oil of C. martii showed in vivo antileishmanial activity in a mouse model [97] and C. reticulata resin oil showed activity against L. amazonensis axenic amastigotes (IC50 = 15.0 μg/mL) and intracellular amastigotes (IC50 = 20 μg/mL) [109]. Diterpenoids isolated from C. officinalis—agathic acid, alepterolic acid, kaurenoic acid, methyl copalate, pinifolic acid, and ent-polyalthic acid—showed antileishmanial activity against L. amazonensis promastigotes [104].
Copaifera oleoresins and diterpene acids have also shown antitrypanosomal activities. C. duckei and C. reticulata resins showed in vitro activity against T. evansi trypomastigotes [86]. The diterpene acids—agathic acid, copalic acid, alepterolic acid, kaurenoic acid, methyl copalate, pinifolic acid, and ent-polyalthic acid—all showed antitrypanosomal activity against T. cruzi, including in the epimastigote, trypomastigote, and amastigote forms of the protozoan [111].
A number of parasitic protozoal proteins have been identified as potential targets for antiparasitic chemotherapy [113]. In conjunction with this review, we have examined the potential parasitic targets of Copaifera diterpenoids using molecular docking. It is currently not known what biomolecular targets from Leishmania or Trypanosoma may be responsible for the antiprotozoal activities of copaiba. The Copaifera diterpenoids (Figure 1, Figure 2 and Figure 3) were screened, in silico, against Leishmania drug targets [114,115,116] and Trypanosoma cruzi protein targets [117] using Molegro Virtual Docker v. 6.0.1 as previously described [114,115,116,117]. The docking energies are summarized in Table 4 and Table 5.
The Leishmania protein target with the best overall docking properties with Copaifera diterpenoids was L. major dihydroorotate dehydrogenase (average Edock = −109.2 kJ/mol). These docking energies were better than the docking energy for the normal substrate, dihydroorotate (Edock = −72.1 kJ/mol) and comparable to the co-crystallized ligand for this protein, nitroorotate (Edock = −104.2 kJ/mol). Docking energies for Copaifera diterpenoids with TcDHODH (average −92.5 kJ/mol) were also better than the normal substrate (dihydroorotate, Edock = −64.2 kJ/mol), but worse than the synthetic TcDHODH inhibitor, 5-[2-(5-carboxynaphthalen-2-yl)ethyl]-2,6-dioxo-1,2,3,6-tetrahydro-pyrimidine-4-carboxylic acid (TT2-2-199, Edock = −140.7 kJ/mol). Similarly, Copaifera diterpenoids docked with L. donovani DHODH (average Edock = −89.9 kJ/mol) better than dihydroorotate (Edock = −60.9 kJ/mol). Based on these docking energies, protozoal dihydroorotate dehydrogenases are likely targets for Copaifera diterpenoids.
Leishmania major methionyl-tRNA synthetase was another Leishmania protein target with good docking energies. Although the docking energies with this protein were excellent (average Edock = −106.9 kJ/mol), they are much poorer than the docking energy of the normal substrate, methionyl adenylate (Edock = −168.1 kJ/mol). Similarly, the T. cruzi target protein with the best docking was UDP-galactose mutase (average Edock = −104.5 kJ/mol), but the normal substrate and co-crystallized ligand, uridine diphosphate (UDP), had a much superior docking energy (Edock = −232.8 kJ/mol). Likewise, L. major UDP-glucose pyrophosphorylase showed an average docking energy of −99.9 kJ/mol, which was much worse than UDP itself (Edock = −145.9 kJ/mol). The diterpenoids showed good docking to T. cruzi spermidine synthase, with an average docking energy of −96.8 kJ/mol; however, these are much worse than the docking energy of the co-crystallized ligand, S-adenosyl methionine, with a docking energy of −133.0 kJ/mol. Thus, although they exhibited good docking properties, it is unlikely that Copaifera diterpenoids can compete with the normal substrate ligands for these proteins.
Copaifera diterpenoids showed excellent docking to L. mexicana pyruvate kinase (average Edock = −103.4 kJ/mol), much better than the normal substrate, phosphoenolpyruvate (Edock = −59.8 kJ/mol). Docking energies with T. cruzi pyruvate kinase were not as impressive (average −80.3 kJ/mol), but still better than phosphoenolpyruvate (Edock = −48.6 kJ/mol) and comparable to the TcPYK inhibitor, ponceau S (Edock = −83.6 kJ/mol). Parasite pyruvate kinases can be expected to be target proteins for Copaifera diterpenoids.
Protozoal triosephosphate isomerases (LmexTIM and TcTIM) are expected to be targeted by Copaifera diterpenoids. The average docking energy with LmexTIM (−90.7 kJ/mol) was much better than either the normal substrate (dihydroxyacetone phosphate, Edock = −52.4 kJ/mol) or the co-crystallized ligand, phosphoglycolohydroxamic acid (Edock = −61.1 kJ/mol). Likewise, docking energies with TcTIM (average −88.2 kJ/mol) were better than the dihydroxyacetone phosphate (Edock = −59.7 kJ/mol) and comparable to the TcTIM inhibitor, 3-(2-benzothiazolylthio)-1-propanesulfonic acid (Edock = −85.5 kJ/mol).
Both L. major pteridine reductase and T. cruzi pteridine reductase had docking properties with Copaifera diterpenoids with comparable energies (average Edock = −93.8 and −96.8 kJ/mol, respectively) with the normal substrate dihydrobiopterin (Edock = −96.9 and −100.1 kJ/mol, respectively). Thus, Copaifera diterpenoids may compete with dihydrobiopterin for pteridine reductase.
Sterols are the normal substrates for sterol 14α-demethylase (CYP51), and triterpenoids are expected to also target this protein as inhibitors [118]. Nevertheless, Copaifera diterpenoids showed docking energies that may compete with normal sterols for these protein targets. L. infantum CYP51 had an average docking energy with the diterpenoids of −90.2 kJ/mol, which was generally not as good as a normal sterol substrate (obtusifoliol, Edock = −104.4 kJ/mol), but comparable to the known LinfCYP51 inhibitor fluconazole (Edock = −87.5 kJ/mol). Likewise, T. cruzi CYP51 had an average diterpenoid docking energy of −89.5 kJ/mol, but substrate (obtusifoliol) docking of −105.6 kJ/mol, and fluconazole docking energy of −90.9 kJ/mol.
Copaifera diterpenoids generally showed weak docking energies against the parasite cysteine proteases, L. donovani cathepsin B, L. major cathepsin B, or cruzain. This docking behavior of diterpenoids with Leishmania cathepsin B [114] and cruzain [117] was previously observed. Leishmania donovani and T. cruzi cyclophilins also showed weak docking energies.
Although Copaifera diterpenoids showed only weak docking to parasite glyceraldehyde-3-phosphate dehydrogenases, they may still target these proteins. LmexGAPDH had an average Edock of −73.0 kJ/mol and TcGAPDH had an average Edock of −70.3 kJ/mol, but these docking energies are better than the docking energies of the normal substrate, glyceraldehyde-3-phosphate (Edock = −58.9 and −52.6 kJ/mol, respectively).
Additional Leishmania proteins expected to be targeted by Copaifera diterpenoids include glycerol-3-phosphate dehydrogenase, which showed excellent docking energies (average −100.4 kJ/mol) to LmexGPDH, better than the normal substrate, glycerol-3-phosphate (Edock = −62.5 kJ/mol). Also targeted with a weak docking energy are: glucose-6-phosphate isomerase (Lmex GPI Edock average = −73.0 kJ/mol), though better than the docking energy of the normal substrate (glucose-6-phosphate, Edock = −62.0 kJ/mol); and phosphomannomutase (LmexPMM Edock average = −94.2 kJ/mol), which is better compared to the normal substrate (mannose-6-phosphate, Edock = −72.5 kJ/mol).
Additional T. cruzi protein targets may be dihydrofolate reductase—thymidylate synthase (TcDHFR–TS), which showed an average docking energy with Copaifera diterpenoids of −93.2 kJ/mol, comparable to the docking energy of the normal substrate (dihydrofolate, −99.3 kJ/mol), as well as the TcDHFR–TS inhibitor cycloguanil (Edock = −83.1 kJ/mol); farnesyl diphosphate synthase (TcFPPS), with docking energies that averaged −96.2 kJ/mol, which is comparable to the docking energy of the normal substrate, isopentenyl diphosphate (Edock = −98.9 kJ/mol); and hypoxanthine phosphoribosyltransferase (TcHPRT), having an average Edock = −82.1 kJ/mol, compared to the normal substrate, hypoxanthine, with Edock of −65.9 kJ/mol.

6.2. Antibacterial Activity of Copaiba

Copaiba oleoresin has shown antibacterial activity against several strains, in particular, Gram-positive Bacillus subtilis and Staphylococcus aureus with minimum inhibitory concentration (MIC) values of 5 μg/mL for both organisms [110]. Copalic acid, isolated from C. langsdorffii, showed excellent antibacterial activity against Bacillus cereus (MIC 8.0 μg/mL), B. subtilis (MIC 5.0 μg/mL), Kocuria rhizophila (MIC 5.0 μg/mL), Streptococcus pyogenes (MIC 3.0 μg/mL), S. pneumoniae (MIC 3.0 μg/mL), S. agalactiae (MIC 2.0 μg/mL), S. dysgalactiae (MIC 1.0 μg/mL), S. epidermidis (MIC 0.5 μg/mL) [71], S. salivarius (MIC 2.0 μg/mL), S. mutans (MIC 3.0 μg/mL), S. mitis (MIC 5.0 μg/mL), S. sobrinus (MIC 3.0 μg/mL), and S. sanguinis (MIC 6.0 μg/mL) [70]. Likewise, kaurenoic acid showed remarkable activity against S. pyogenes (MIC 5.0 μg/mL), S. pneumoniae (MIC 5.0 μg/mL), S. dysgalactiae (MIC 8.0 μg/mL) [71], S. epidermidis (MIC 4–5 μg/mL), B. subtilis (MIC 3.1–6.3 μg/mL), and S. aureus (MIC 8–10 μg/mL) [75]. 3α-Alepterolic acid acetate (acetoxycopalic acid) showed moderate antibacterial activity against cariogenic Streptococcus bacteria, with MIC values ranging from 12.0 to 60.0 μg/mL [70]. ent-Polyalthic acid also showed moderate antibacterial activity against B. subtilis (MIC 20–30 μg/mL), S. aureus (MIC 40–50 μg/mL), and S. epidermidis (MIC 40 μg/mL) [75].
In order to provide some insight into the mechanisms of activity, a virtual screening of copaiba diterpenoids has been carried out against several bacterial protein targets, including peptide deformylase, DNA gyrase, topoisomerase IV, UDP-galactopyranose mutase, protein tyrosine phosphatase, cytochrome P450 CYP 121, and nicotinamide adenine dinucleotide (NAD+)-dependent DNA ligase [119] (see Table 6). The best bacterial target for copalic acid was Mycobacterium tuberculosis DNA gyrase B (PDB 3ZKD) with a docking energy (Edock) of −105.7 kJ/mol. The protein with the best docking energy with kaurenoic acid was S. pneumoniae peptide deformylase (PDB 2AIE, Edock = −89.7 kJ/mol). 3α-Alepterolic acid acetate was the best docking ligand to Escherichia coli topoisomerase IV (PDB 1S16) and M. tuberculosis DNAGyrB (PDB 3ZKD) with docking energies of −118.8 and −118.3 kJ/mol, respectively. 3β-Alepterolic acid acetate also showed excellent docking to these two proteins with docking energies of −117.1 and −117.3 kJ/mol, respectively. The best bacterial target for ent-polyalthic acid was M. tuberculosis protein tyrosine phosphatase (PDB 2OZ5, Edock = −107.2 kJ/mol). The copaiba diterpenoid ligand with the best docking properties was 7α-acetoxyhardwickiic acid with S. aureus peptide deformylase (PDB 3U7M, Edock = −120.6 kJ/mol).

6.3. Antiproliferative Activity of Copaiba

Copaiba oleoresins have exhibited both in vitro and in vivo antiproliferative activities (Table 3). Copaifera reticulata oleoresin, for example, has shown in vitro cytotoxic activity against GM07492-A human lung fibroblast cells with an IC50 of 51.85 μg/mL [68]. The oleoresin of C. multijuga has shown in vitro cytotoxic activity against B16F10 murine melanoma cells with an IC50 of 457 μg/mL [57]. Furthermore, in a mouse model of lung metastasis and tumor growth, oral administration of C. multijuga oleoresin reduced tumor growth, tumor mass, and number of lung nodules after inoculation of B16F10 tumor cells [57]. Likewise, C. multijuga oleoresin, in doses varying between 100 and 200 mg/kg, showed antineoplastic properties against Ehrlich ascetic tumors and solid tumors in an in vivo mouse model [98]. On the other hand, C. officinalis oleoresin actually stimulated growth of Walker 256 carcinoma by 70% in an in vivo rat model [101].
Diterpenoids isolated from Copaifera species have shown cytotoxic activities (Table 3). Copalic acid, isolated from C. langsdorffii, showed in vitro cytotoxicity on MO59J human glioblastoma cells and HeLa human cervical adenocarcinoma cells with IC50 of 68.3 and 44.0 μg/mL, respectively [71]. Kaurenoic acid has demonstrated cytotoxicity against several human tumor cell lines, including CEM leukemia, MCF-7 breast tumor, HCT-8 colon tumor [73], AGP01 gastric tumor, and SF-295 glioblastoma [74]. Growth inhibition of AGP01 and SF-295 cells was also demonstrated by 3β-alepterolic acid and 3β-alepterolic acid acetate [73]. Methyl copalate showed remarkable cytotoxic activity on P-388 murine lymphoma (IC50 = 2.5 μg/mL), A-549 human lung carcinoma (IC50 = 5 μg/mL), HT-29 human colon carcinoma (IC50 = 5 μg/mL), and MEL-28 human melanoma (IC50 = 10 μg/mL) cells [75].
Molecular docking (Molegro Virtual Docker, Aarhus, Denmark) has been carried out with Copaifera diterpenoids on cancer molecular targets, including androgen receptor, aromatase, caseine kinase II, cyclin-dependent kinases 2, 4, and 6, cyclooxygenase 2, DNA (cytosine-5)-methyltransferase-1 and -3A, epidermal growth factor receptor, estrogen receptor α, estrogen receptor β, heat shock protein 90, insulin-like growth factor 1 receptor, 5-lipoxygenase, mitogen-activated protein kinase 1, NF-κB, p90 ribosomal protein S6 kinase, P-glycoprotein, phosphatidylinositol-4,5-bisphosphate 3-kinase, topoisomerase I, topoisomerase IIα, topoisomerase IIβ, tubulin, and vascular endothelial growth factor receptor (Table 7). The best overall cancer targets for the copaiba diterpenoids were human DNA (cytosine-5)-methyltransferase-1 (HsDNMT1), human estrogen receptor β (HsERβ), and human mitogen-activated protein kinase 1 (HsMEK1), with average MolDock docking energies of −102.7, −99.2, and −101.5 kJ/mol, respectively. DNA (cytosine-5)-methyltransferase-1 (DNMT1) is the enzyme responsible for DNA methylation of carbon-5 of cytosine within CpG dinucleotides [120]. The enzyme is required for embryonic development [121], but is overexpressed in lung, liver, colorectal, gastric, breast, and lung tumors [122]. Thus, DNMT1 has emerged as an attractive target for cancer chemotherapy [123,124]. The mitogen-activated protein kinase (MAPK) signaling cascade is one of the most important pathways involved in cellular proliferation and differentiation [125] and, therefore, inhibition of components of this pathway, such as MEK1, can potentially target tumors that depend on MAPK signaling [126]. Agonism of estrogen receptor α (ERα) stimulates proliferation of breast, uterus, and prostate tissues, whereas ERβ agonism inhibits proliferation of these tissues [127]. Thus, compounds that can selectively bind and activate ERβ, but not ERα, could represent effective antitumor agents for treatment of prostate and breast cancer [128]. Copalic acid and methyl copalate both targeted HsMEK1, with docking energies of −108.2 and −111.0 kJ/mol, respectively, while 3β-alepterolic acid and 3β-alepterolic acid acetate showed excellent docking with HsDNMT1 (Edock = −107.2 and −121.7 kJ/mol, respectively). Kaurenoic acid was a relatively weakly docking ligand but did show selective docking to aromatase (Edock = −93.7 kJ/mol). The best-docking ligand was patagonic acid, which had a docking energy of −121.8 kJ/mol with HsDNMT1.

6.4. Anti-Inflammatory Activity of Copaiba

Inflammation is the biological response of body tissues to detrimental stimuli, such as pathogenic microorganisms, chemical or physical irritants, or injury. Inflammation is manifested by redness, swelling, heat, and sometimes pain. While acute inflammation is a normal part of the healing process, chronic inflammation often plays a role in chronic diseases such as osteoarthritis, lupus, and inflammatory bowel disease, and can be problematic. Several copaiba oleoresins have shown anti-inflammatory activity, including C. cearensis [13], C. duckei [84], C. langsdorffii [50,91], C. multijuga [13,58,61,100], C. officinalis [62], and C. reticulata [13] (Table 3).
The immune response is a complex cascade of interacting cytokines and reactions, and there are several biomolecular targets important in treating chronic inflammation. We have carried out virtual screening of copaiba diterpenoids against soluble epoxide hydrolase (EPHX2), fibroblast collagenase, phospholipase A2 (PLA2), 5-lipoxygenase, inducible nitric oxide synthase, phosphoinositide 3-kinase, interleukin-1 receptor-associated kinase 4, glutathione S-transferase ω-1, cyclooxygenase-1, cyclooxygenase-2, c-Jun N-terminal kinase, nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), inhibitor of κB kinase β, NF-κB essential modulator, lipid binding protein MD-2, myeloperoxidase, p38 mitogen-activated protein kinase, peroxisome proliferator-activated receptor γ, and cAMP-specific 3′,5′-cyclic phosphodiesterase 4D (Table 8). The overall best target proteins were murine soluble epoxide hydrolase and murine phospholipase A2, with average docking energies of −108.3 and −100.0 kJ/mol. Secretory phospholipase A2 and cytosolic phospholipase A2 are both targets for anti-inflammatory drug development [129]. Soluble epoxide hydrolase has been identified as a molecular target not only for inflammatory diseases, but also as a target for neurodegenerative diseases and for treatment of pain [130]. Thus, targeting EPHX2 and/or PLA2 by copaiba diterpenoids may explain the anti-inflammatory activities of copaiba oleoresins.

7. Computational Methods—Molecular Docking

Molecular docking analyses were carried out using Molegro Virtual Docker (v. 6.0.1, Molegro ApS, Aarhus, Denmark) against known bacterial [119], Leishmania [114,115,116], Trypanosoma cruzi [117], and cancer-relevant and inflammation-relevant protein targets [131], as previously described [114,115,116,117,119,132].

8. Conclusions

The oleoresins from Copaifera species (copaiba) have been used by native peoples of the Amazon region for thousands of years. These materials have shown remarkable biological activities, including antibacterial, antiparasitic, antineoplastic, and anti-inflammatory activities. Copaiba resins have been distilled to give essential oils that are largely composed of sesquiterpenoids, particularly β-caryophyllene. The resins are also composed of diterpene acids, which are responsible for many of the observed biological activities. Molecular docking of copaiba diterpene acids with documented protein targets has revealed potential mechanisms of activity for these bioactive constituents. Future research to validate the molecular mechanisms of copaiba diterpenoids is encouraged.

Author Contributions

R.d.T., J.K.d.S. and W.N.S. authors contributed equally to literature searching and manuscript writing, review and editing; W.N.S. carried out the molecular docking.

Funding

W.N.S. is grateful to dōTERRA International (Available online: https://www.doterra.com/US/en) for financial support of the Aromatic Plant Research Centers (APRC, Available online: https://aromaticplant.org/).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dwyer, J.D. The Central American, West Indian, and South American species of Copaifera (Caesalpiniaceae). Brittonia 1951, 7, 143–172. [Google Scholar] [CrossRef]
  2. Jacquin, N.J. Enumeratio Systematica Plantarum; Apud Theodorum Haak: Leiden, The Netherlands, 1760. [Google Scholar]
  3. Linné, C. Species Plantarum; Joannis Thomae de Trattner: Vienna, Austria, 1764. [Google Scholar]
  4. Joyce, B.L.; Al-Ahmad, H.; Chen, F.; Stewart, C.N. Diesel Trees. In Handbook of Bioenergy Crop Plants; Kole, C., Joshi, C.P., Shonnard, D.R., Eds.; CRC Press: Boca Raton, FL, USA, 2012; pp. 619–629. [Google Scholar]
  5. Costa, J.A.S.; Copaifera. In Flora do Brasil 2020, em Construção. Available online: http://floradobrasil.jbrj.gov.br/reflora/floradobrasil/FB22895 (accessed on 6 March 2018).
  6. Veiga Junior, V.F.; Pinto, A.C. O gênero Copaifera L. Quim. Nova 2002, 25, 273–286. [Google Scholar] [CrossRef]
  7. Plowden, C. The ethnobotany of copaiba (Copaifera) oleoresin in the Amazon. Econ. Bot. 2004, 58, 729–733. [Google Scholar] [CrossRef]
  8. Cascon, V.; Gilbert, B. Characterization of the chemical composition of oleoresins of Copaifera guianensis Desf., Copaifera duckei Dwyer and Copaifera multijuga Hayne. Phytochemistry 2000, 55, 773–778. [Google Scholar] [CrossRef]
  9. Martins-da-Silva, R.C.V.; Pereira, J.F.; de Lima, H.C. O gênero Copaifera (Leguminosae—Caesalpinioideae) na Amazônia Brasileira. Rodriguésia 2008, 59, 455–476. [Google Scholar]
  10. Souza, V.C.; Lorenzi, H. Botânica Sistemática: Guia Ilustrado para Identificação das Famílias de Angiospermas da Flora Brasileira, Baseado na APG III, 3rd ed.; Instituto Plantarum: Nova Odessa, Brazil, 2012. [Google Scholar]
  11. Milani, J.F.; Rocha, J.F.; de Pádua Teixeira, S. Oleoresin glands in copaíba (Copaifera trapezifolia Hayne: Leguminosae), a Brazilian rainforest tree. Trees 2012, 26, 769–775. [Google Scholar] [CrossRef] [Green Version]
  12. Soares, N.S.; Goncalves, C.A.; Araujo, G.M.; Lomonaco, C. Floristic composition and abundance in forest fragments: A case study from southern Goias, Brazil. Biosci. J. 2015, 31, 1238–1252. [Google Scholar] [CrossRef]
  13. Veiga Junior, V.F.; Rosas, E.C.; Carvalho, M.V.; Henriques, M.G.M.O.; Pinto, A.C. Chemical composition and anti-inflammatory activity of copaiba oils from Copaifera cearensis Huber ex Ducke, Copaifera reticulata Ducke and Copaifera multijuga Hayne—A comparative study. J. Ethnopharmacol. 2007, 112, 248–254. [Google Scholar] [CrossRef] [PubMed]
  14. Langenheim, J.H. Plant Resins: Chemistry, Evolution, Ecology, and Ethnobotany; Timber Press, Inc.: Portland, OR, USA, 2003. [Google Scholar]
  15. Leandro, L.M.; de Sousa Vargas, F.; Barbosa, P.C.S.; Neves, J.K.O.; da Silva, J.A.; da Veiga-Junior, V.F. Chemistry and biological activities of terpenoids from copaiba (Copaifera spp.) oleoresins. Molecules 2012, 17, 3866–3889. [Google Scholar] [CrossRef] [PubMed]
  16. Langenheim, J.H. Higher plant terpenoids: A phytcentric overview of their ecological roles. J. Chem. Ecol. 1994, 20, 1223–1280. [Google Scholar] [CrossRef] [PubMed]
  17. Barbosa, P.C.S.; Medeiros, R.S.; Sampaio, P.T.B.; Vieira, G.; Wiedemann, L.S.M.; Veiga-Junior, V.F. Influence of abiotic factors on the chemical composition of copaiba oil (Copaifera multijuga Hayne): Soil composition, seasonality and diameter at breast height. J. Braz. Chem. Soc. 2012, 23, 1823–1833. [Google Scholar] [CrossRef]
  18. Biavatti, M.W.; Dossin, D.; Deschamps, F.C.; Lima, M.D.P. Análise de óleos-resinas de copaíba: Contribuição para o seu controle de qualidade. Rev. Bras. Farmacogn. 2006, 16, 230–235. [Google Scholar] [CrossRef]
  19. Pieri, F.A.; Mussi, M.C.; Moreira, M.A.S. Óleo de copaíba (Copaifera sp.): Histórico, extração, aplicações industriais e propriedades medicinais. Rev. Bras. Plantas Med. 2009, 11, 465–472. [Google Scholar] [CrossRef]
  20. Rosa, J.C.; da Silva Gomes, A.M. Os aspectos etnobotânicos da copaíba. Rev. Geogr. 2009, 4, 59–77. [Google Scholar] [CrossRef]
  21. Ribeiro, R.D.; de Lima, H.C. Riqueza e distribuição geográfica de espécies arbóreas da família Leguminosae e implicações para conservação no Centro de Diversidade Vegetal de Cabo Frio, Rio de Janeiro, Brasil. Rodriguésia 2009, 60, 111–127. [Google Scholar] [CrossRef]
  22. Carvalho, M.C.; Gomide, L.R.; Santos, R.M.D.; Scolforo, J.R.S.; Carvalho, L.M.T.D.; Mello, J.M.D. Modeling ecological niche of tree species in Brazilian tropical area. Cerne 2017, 23, 229–240. [Google Scholar] [CrossRef]
  23. Carvalho, P.E.R. Espécies Arbóreas Brasileiras, Vol. 1; EMBRAPA Informação Tecnológica: Brasilia, Brazil, 2003. [Google Scholar]
  24. Shanley, P.; Leite, A.; Alechandre, A.; Azevedo, C. Copaíba: Copaifera spp. In Frutíferas e Plantas Úteis na Vida Amazônica; Shanley, P., Medina, G., Eds.; CIFOR and IMAZON: Belém, Brazil, 2005; pp. 85–94. [Google Scholar]
  25. Carvalho, P.E.R. Espécies Florestais Brasileiras: Recomendações Silviculturais, Potencialidades e Uso da Madeira; EMBRAPA-CNPF: Colombo, Brazil, 1994. [Google Scholar]
  26. Rigamonte-Azevedo, O.C.; Wadt, P.G.S.; Wadt, L.D.O. Copaíba: Ecologia e Produção de Óleo-Resina; Embrapa Acre: Rio Branco, Brazil, 2004. [Google Scholar]
  27. Alencar, J.D.C. Estudos silviculturais de uma população natural de Copaifera multijuga Hayne—Leguminosae, na Amazônia central. IV. Interpretação de dados fenológicos em relação a elementos climáticos. Acta Amaz. 1988, 18, 199–209. [Google Scholar] [CrossRef]
  28. Muniz, F.H. Padrões de floração e frutificação de árvores da Amazônia Maranhense. Acta Amaz. 2008, 38, 617–626. [Google Scholar] [CrossRef]
  29. Dos Santos, N. Fenologia. Rodriguésia 1979, 31, 223–226. [Google Scholar]
  30. Pedroni, F.; Sanchez, M.; Santos, F.A.M. Fenologia da copaíba (Copaifera langsdorffii Desf.—Leguminosae, Caesalpinioideae) em uma floresta semidecídua no sudeste do Brasil. Rev. Bras. Botânica 2002, 25, 183–194. [Google Scholar] [CrossRef]
  31. Pedroni, F. Ecologia da Copaíba (Copaifera langsdorffii Desf. Caelsapiniaceae) na Reserva Municipal de Santa Genebra, Campinas, SP; Universidade Estadual de Campinas: Barão Geraldo, Brazil, 1993. [Google Scholar]
  32. Motta Junior, C.J.; Lombardi, J.A. Aves como agentes dispersores da copaiba (Copaifera langsdorffii, Caesalpiniaceae) em São Carlos, estado de São Paulo. Rev. Bras. Ornitol. 1990, 1, 105–106. [Google Scholar]
  33. Brum, H.D.; Camargo, J.L.C.; Ferraz, I.D.K. Copaíba-roxa, Copaifera mutlijuga Hayne. In Manual de Sementes da Amazônia; Ferraz, I.D.K., Camargo, J.L.C., Eds.; INPA: Manaus, Brazil, 2009; p. 12. [Google Scholar]
  34. Pinto, A.; Amaral, P.; Gaia, C.; de Oliveira, W. Boas Práticas para Manejo Florestal e Agroindustrial de Produtos Florestais Não Madeireiros: Açaí, Andiroba, Babaçu, Castanha-do-Brasil, Copaíba e Unha-de-Gato; Sebrae-AM: Belém, Brazil, 2010. [Google Scholar]
  35. Dos Santos, J.C.; Leite, A.C.P.; Wadt, L.H.O.; Borges, K.H.; Andrade, F.G.; Menezes, R.S.; Muniz, P.S.B. Demandas Tecnologicas para o Sistema Produtivo de Óleo de Copaíba (Copaifera spp.) no Estado do Acre; Embrapa Acre: Rio Branco, Brazil, 2001. [Google Scholar]
  36. Newton, P.; Watkinson, A.R.; Peres, C.A. Determinants of yield in a non-timber forest product: Copaifera oleoresin in Amazonian extractive reserves. For. Ecol. Manag. 2011, 261, 255–264. [Google Scholar] [CrossRef]
  37. Estrella, E. Plantas Medicinales Amazónicas: Realidad y Perspectivas; Tratado de Cooperación Amazónica: Lima, Peru, 1995. [Google Scholar]
  38. Rios, M.N.D.S.; Pastore Junior, F. Plantas da Amazônia: 450 Espécies de Uso Geral; Universidade de Brasília: Brasilia, Brazil, 2011. [Google Scholar]
  39. Maciel, M.A.M.; Pinto, A.C.; Veiga Junior, V.F. Plantas medicinais: A necessidade de estudos multidisciplinares. Quim. Nova 2002, 25, 429–438. [Google Scholar] [CrossRef]
  40. Luz, F.J.F. Plantas medicinais de uso popular em Boa Vista, Roraima, Brasil. Hortic. Bras. 2001, 19, 88–96. [Google Scholar] [CrossRef]
  41. De Albuquerque, K.C.O.; da Veiga, A.D.S.S.; da Silva e Silva, J.V.; Brigido, H.P.C.; Ferreira, E.P.D.R.; Costa, E.V.S.; Marinho, A.M.D.R.; Percário, S.; Dolabela, M.F. Brazilian Amazon traditional medicine and the treatment of difficult to heal leishmaniasis wounds with Copaifera. Evid.-Based Complement. Altern. Med. 2017, 2017. [Google Scholar] [CrossRef] [PubMed]
  42. Basile, A.C.; Sertié, J.A.; Freitas, P.C.; Zanini, A.C. Anti-inflammatory activity of oleoresin from Brazilian Copaifera. J. Ethnopharmacol. 1988, 22, 101–109. [Google Scholar] [CrossRef]
  43. Taylor, L. The Healing Power of Rainforest Herbs: A Guide to Understanding and Using Herbal Medicinals; Square One Publishers: Garden City Park, NY, USA, 2005. [Google Scholar]
  44. Sant’Anna, B.M.P.; Fontes, S.P.; Pinto, A.C.; Rezende, C.M. Characterization of woody odorant contributors in copaiba oil (Copaifera multijuga Hayne). J. Braz. Chem. Soc. 2007, 18, 984–989. [Google Scholar] [CrossRef]
  45. Revilla, J. Apontamentos para a Cosmética Amazônica; SEBRAE-INPA: Manaus, Brazil, 2002. [Google Scholar]
  46. Shanley, P.; Pierce, A.R.; Laird, S.A.; Guillén, A. Tapping the Green Market: Certification & Management of Non-Timber Forest Products; Earthscan: London, UK, 2002. [Google Scholar]
  47. Gonzaga, A.L. Madeira: Uso e Conservação; IPHAN/MONUMENTA: Brasilia, Brazil, 2006. [Google Scholar]
  48. Santos, A.O.; Ueda-Nakamura, T.; Dias Filho, B.P.; Veiga Junior, V.F.; Pinto, A.C.; Nakamura, C.V. Effect of Brazilian copaiba oils on Leishmania amazonensis. J. Ethnopharmacol. 2008, 120, 204–208. [Google Scholar] [CrossRef] [PubMed]
  49. Lameira, O.A.; Martins-da-Silva, R.C.V.; Zoghbi, M.D.G.B.; Oliveira, E.C.P. Seasonal variation in the volatiles of Copaifera duckei Dwyer growing wild in the state of Pará—Brazil. J. Essent. Oil Res. 2009, 21, 105–108. [Google Scholar] [CrossRef]
  50. Gelmini, F.; Beretta, G.; Anselmi, C.; Centini, M.; Magni, P.; Ruscica, M.; Cavalchini, A.; Maffei Facino, R. GC-MS profiling of the phytochemical constituents of the oleoresin from Copaifera langsdorffii Desf. and a preliminary in vivo evaluation of its antipsoriatic effect. Int. J. Pharm. 2013, 440, 170–178. [Google Scholar] [CrossRef] [PubMed]
  51. Estevão, L.R.M.; de Medeiros, J.P.; Baratella-Evêncio, L.; Simões, R.S.; Mendonça, F.D.S.; Evêncio-Neto, J. Effects of the topical administration of copaiba oil ointment (Copaifera langsdorffii) in skin flaps viability of rats. Acta Cirúrgica Bras. 2013, 28, 863–869. [Google Scholar] [CrossRef]
  52. Zimmermam-Franco, D.C.; Bolutari, E.B.; Polonini, H.C.; do Carmo, A.M.R.; Chaves, M.D.G.A.M.; Raposo, N.R.B. Antifungal activity of Copaifera langsdorffii Desf oleoresin against dermatophytes. Molecules 2013, 18, 12561–12570. [Google Scholar] [CrossRef] [PubMed]
  53. De Almeida, L.F.R.; Portella, R.D.O.; Bufalo, J.; Marques, M.O.M.; Facanali, R.; Frei, F. Non-oxygenated sesquiterpenes in the essential oil of Copaifera langsdorffii Desf. increase during the day in the dry season. PLoS ONE 2016, 11, e0149332. [Google Scholar] [CrossRef] [PubMed]
  54. Do Nascimento, M.E.; Zoghbi, M.D.G.B.; Pinto, J.E.B.P.; Bertolucci, S.K.V. Chemical variability of the volatiles of Copaifera langsdorffii growing wild in the southeastern part of Brazil. Biochem. Syst. Ecol. 2012, 43, 1–6. [Google Scholar] [CrossRef]
  55. De Almeida, L.F.R.; Portella, R.D.O.; Facanali, R.; Marques, M.O.M.; Frei, F. Dry and wet seasons set the phytochemical profile of the Copaifera langsdorffii Desf. essential oils. J. Essent. Oil Res. 2014, 26, 292–300. [Google Scholar] [CrossRef]
  56. Zoghbi, M.D.G.B.; Lameira, O.A.; Oliveira, E.C.P. Seasonal variation of oleoresin and volatiles from Copaifera martii Hayne growing wild in the State of Pará, Brazil. J. Essent. Oil Res. 2007, 19, 504–506. [Google Scholar] [CrossRef]
  57. Lima, S.R.M.; Veiga Junior, V.F.; Christo, H.B.; Pinto, A.C.; Fernandes, P.D. In vivo and in vitro studies on the anticancer activity of Copaifera multijuga Hayne and its fractions. Phyther. Res. 2003, 17, 1048–1053. [Google Scholar] [CrossRef] [PubMed]
  58. Kobayashi, C.; Fontanive, T.O.; Enzweiler, B.G.; de Bona, L.R.; Massoni, T.; Apel, M.A.; Henriques, A.T.; Richter, M.F.; Ardenghi, P.; Suyenaga, E.S. Pharmacological evaluation of Copaifera multijuga oil in rats. Pharm. Biol. 2011, 49, 306–313. [Google Scholar] [CrossRef] [PubMed]
  59. Trindade, F.T.T.; Stabeli, R.G.; Pereira, A.A.; Facundo, V.A.; Silva, A.D.A. Copaifera multijuga ethanolic extracts, oil-resin, and its derivatives display larvicidal activity against Anopheles darlingi and Aedes aegypti (Diptera: Culicidae). Braz. J. Pharmacogn. 2013, 23, 464–470. [Google Scholar] [CrossRef]
  60. Souza Barbosa, P.C.; Moreira Wiedemann, L.S.; da Silva Medeiros, R.; Barbosa Sampaio, P.D.T.; Vieira, G.; da Veiga-Junior, V.F. Phytochemical fingerprints of copaiba oils (Copaifera multijuga Hayne) determined by multivariate analysis. Chem. Biodivers. 2013, 10, 1350–1360. [Google Scholar] [CrossRef] [PubMed]
  61. Veiga Junior, V.F.; Luciano, Z.; Patitucci, M.L.; Pinto, A.C.; Calixto, J.B. The inhibition of paw oedema formation caused by the oil of Copaifera multijuga Hayne and its fractions. J. Pharm. Pharmacol. 2006, 58, 1405–1410. [Google Scholar] [CrossRef] [PubMed]
  62. Dias, D.S.; Fontes, L.B.A.; Crotti, A.E.M.; Aarestrup, B.J.V.; Aarestrup, F.M.; da Silva Filho, A.A.; Corrêa, J.O.A. Copaiba oil suppresses inflammatory cytokines in splenocytes of C57Bl/6 mice induced with experimental autoimmune encephalomyelitis (EAE). Molecules 2014, 19, 12814–12826. [Google Scholar] [CrossRef] [PubMed]
  63. Zoghbi, M.D.G.B.; Martins-da-Silva, R.C.V.; Trigo, J.R. Volatiles of oleoresins of Copaifera paupera (Herzog) Dwyer, C. piresii Dwyer and C. pubiflora Benth. (Leguminosae). J. Essent. Oil Res. 2009, 21, 403–405. [Google Scholar] [CrossRef]
  64. Zoghbi, M.D.G.B.; Andrade, E.H.A.; Martins-Da-Silva, R.C.V.; Trigo, J.R. Chemical variation in the volatiles of Copaifera reticulata Ducke (Leguminosae) growing wild in the states of Pará and Amapá, Brazil. J. Essent. Oil Res. 2009, 21, 501–503. [Google Scholar] [CrossRef]
  65. Herrero-Jáuregui, C.; Casado, M.A.; Zoghbi, M.D.G.B.; Martins-Da-Silva, R.C. Chemical variability of Copaifera reticulata Ducke oleoresin. Chem. Biodivers. 2011, 8, 674–685. [Google Scholar] [CrossRef] [PubMed]
  66. Sachetti, C.G.; de Carvalho, R.R.; Paumgartten, F.J.R.; Lameira, O.A.; Caldas, E.D. Developmental toxicity of copaiba tree (Copaifera reticulata Ducke, Fabaceae) oleoresin in rat. Food Chem. Toxicol. 2011, 49, 1080–1085. [Google Scholar] [CrossRef] [PubMed]
  67. Guimarães-Santos, A.; Santos, D.S.; Santos, I.R.; Lima, R.R.; Pereira, A.; de Moura, L.S.; Carvalho, R.N.; Lameira, O.; Gomes-Leal, W. Copaiba oil-resin treatment is neuroprotective and reduces neutrophil recruitment and microglia activation after motor cortex excitotoxic injury. Evid.-Based Complement. Altern. Med. 2012, 2012. [Google Scholar] [CrossRef] [PubMed]
  68. Bardají, D.K.R.; da Silva, J.J.M.; Bianchi, T.C.; de Souza Eugênio, D.; de Oliveira, P.F.; Leandro, L.F.; Rogez, H.L.G.; Venezianni, R.C.S.; Ambrosio, S.R.; Tavares, D.C.; et al. Copaifera reticulata oleoresin: Chemical characterization and antibacterial properties against oral pathogens. Anaerobe 2016, 40, 18–27. [Google Scholar] [CrossRef] [PubMed]
  69. Veiga, V.F.; Pinto, A.C.; de Lima, H.C. The essential oil composition of Copaifera trapezifolia Hayne leaves. J. Essent. Oil Res. 2006, 18, 430–431. [Google Scholar] [CrossRef]
  70. Souza, A.B.; Martins, C.H.G.; Souza, M.G.M.; Furtado, N.A.J.C.; Heleno, V.C.G.; de Sousa, J.P.B.; Rocha, E.M.P.; Bastos, J.K.; Cunha, W.R.; Veneziani, R.C.S.; et al. Antimicrobial activity of terpenoids from Copaifera langsdorffii Desf. against cariogenic bacteria. Phyther. Res. 2011, 25, 215–220. [Google Scholar] [CrossRef] [PubMed]
  71. Abrão, F.; de Araújo Costa, L.D.; Alves, J.M.; Senedese, J.M.; de Castro, P.T.; Ambrósio, S.R.; Veneziani, R.C.S.; Bastos, J.K.; Tavares, D.C.; Martins, C.H.G. Copaifera langsdorffii oleoresin and its isolated compounds: Antibacterial effect and antiproliferative activity in cancer cell lines. BMC Complement. Altern. Med. 2015, 15, 1–10. [Google Scholar] [CrossRef] [PubMed]
  72. Paiva, L.A.F.; Gurgel, L.A.; Silva, R.M.; Tomé, A.R.; Gramosa, N.V.; Silveira, E.R.; Santos, F.A.; Rao, V.S.N. Anti-inflammatory effect of kaurenoic acid, a diterpene from Copaifera langsdorffii on acetic acid-induced colitis in rats. Vascul. Pharmacol. 2003, 39, 303–307. [Google Scholar] [CrossRef]
  73. Costa-Lotufo, L.V.; Cunha, G.M.A.; Farias, P.A.M.; Viana, G.S.B.; Cunha, K.M.A.; Pessoa, C.; Moraes, M.O.; Silveira, E.R.; Gramosa, N.V.; Rao, V.S.N. The cytotoxic and embryotoxic effects of kaurenoic acid, a diterpene isolated from Copaifera langsdorffii oleo-resin. Toxicon 2002, 40, 1231–1234. [Google Scholar] [CrossRef]
  74. Vargas, F.D.S.; de Almeida, P.D.O.; Aranha, E.S.P.; Boleti, A.P.D.A.; Newton, P.; de Vasconcellos, M.C.; Veiga Junior, V.F.; Lima, E.S. Biological activities and cytotoxicity of diterpenes from Copaifera spp. oleoresins. Molecules 2015, 20, 6194–6210. [Google Scholar] [CrossRef] [PubMed]
  75. Tincusi, B.M.; Jiménez, I.A.; Bazzocchi, I.L.; Moujir, L.M.; Mamani, Z.A.; Barroso, J.P.; Ravelo, A.G.; Hernández, B.V. Antimicrobial terpenoids from the oleoresin of the Peruvian medicinal plant Copaifera paupera. Planta Med. 2002, 68, 808–812. [Google Scholar] [CrossRef] [PubMed]
  76. Pinto, A.C.; Braga, W.F.; Rezende, C.M.; Garrido, F.M.S.; Veiga, V.F.; Bergter, L.; Patitucci, M.L.; Antunes, O.A.C. Separation of acid diterpenes of Copaifera cearensis Huber ex Ducke by flash chromatography using potassium hydroxide impregnated silica gel. J. Braz. Chem. Soc. 2000, 11, 355–360. [Google Scholar] [CrossRef]
  77. Souza, A.B.; de Souza, M.G.M.; Moreira, M.A.; Moreira, M.R.; Furtado, N.A.J.C.; Martins, C.H.G.; Bastos, J.K.; dos Santos, R.A.; Heleno, V.C.G.; Ambrosio, S.R.; et al. Antimicrobial evaluation of diterpenes from Copaifera langsdorffii oleoresin against periodontal anaerobic bacteria. Molecules 2011, 16, 9611–9619. [Google Scholar] [CrossRef] [PubMed]
  78. Lemos, M.; Santin, J.R.; Mizuno, C.S.; Boeing, T.; de Sousa, J.P.B.; Nanayakkara, D.; Bastos, J.K.; de Andrade, S.F. Copaifera langsdorffii: Evaluation of potential gastroprotective of extract and isolated compounds obtained from leaves. Braz. J. Pharmacogn. 2015, 25, 238–245. [Google Scholar] [CrossRef]
  79. De Sousa, J.P.B.; Brancalion, A.P.S.; Junior, M.G.; Bastos, J.K. A validated chromatographic method for the determination of flavonoids in Copaifera langsdorffii by HPLC. Nat. Prod. Commun. 2012, 7, 25–28. [Google Scholar] [PubMed]
  80. Batista, Â.G.; Ferrari, A.S.; da Cunha, D.C.; da Silva, J.K.; Cazarin, C.B.B.; Correa, L.C.; Prado, M.A.; de Carvalho-Silva, L.B.; Esteves, E.A.; Maróstica Júnior, M.R. Polyphenols, antioxidants, and antimutagenic effects of Copaifera langsdorffii fruit. Food Chem. 2016, 197, 1153–1159. [Google Scholar] [CrossRef] [PubMed]
  81. Costa, A.R.M.; Freitas, L.A.P.; Mendiola, J.; Ibáñez, E. Copaifera langsdorffii supercritical fluid extraction: Chemical and functional characterization by LC/MS and in vitro assays. J. Supercrit. Fluids 2015, 100, 86–96. [Google Scholar] [CrossRef]
  82. Geris, R.; da Silva, I.G.; da Silva, H.H.G.; Barison, A.; Rodrigues-Filho, E.; Ferreira, A.G. Diterpenoids from Copaifera reticulata Ducke with larvicidal activity against Aedes aegypti (L.) (Diptera, Culicidae). Rev. Inst. Med. Trop. Sao Paulo 2008, 50, 25–28. [Google Scholar] [PubMed]
  83. Dos Santos, A.O.; Ueda-Nakamura, T.; Prado Dias Filho, B.; da Veiga Junior, V.F.; Pinto, A.C.; Nakamura, C.V. Antimicrobial activity of Brazilian copaiba oils obtained from different species of Copaifera. Mem. Inst. Oswaldo Cruz 2008, 103, 277–281. [Google Scholar] [CrossRef]
  84. Carvalho, J.C.T.; Cascon, V.; Possebon, L.S.; Morimoto, M.S.S.; Cardoso, L.G.V.; Kaplan, M.A.C.; Gilbert, B. Topical antiinflammatory and analgesic activities of Copaifera duckei Dwyer. Phyther. Res. 2005, 19, 946–950. [Google Scholar] [CrossRef] [PubMed]
  85. Castro-e-Silva, O., Jr.; Zucoloto, S.; Ramalho, F.S.; Ramalho, L.N.Z.; Reis, J.M.C.; Bastos, A.A.C.; Brito, M.V.H. Anti-proliferative activity of oleoresin from Brazilian Copaifera duckei oleoresin on liver regeneration in rats. Phyther. Res. 2004, 18, 92–94. [Google Scholar] [CrossRef] [PubMed]
  86. Dorneles, F.D.S.; Da Silva, A.S.; Oliveira, C.B.; Zimmermann, C.E.P.; Rosa, L.D.; Tonin, A.A.; de Oliveira, E.C.P.; Santurio, J.M.; Monteiro, S.G. Susceptibility of Trypanosoma evansi in the Copaiba oil: In vitro test and in mice experimentally infected with the parasite. Acta Sci. Vet. 2013, 41, 1136. [Google Scholar]
  87. Paiva, L.A.F.; Rao, V.S.N.; Gramosa, N.V.; Silveira, E.R. Gastroprotective effect of Copaifera langsdorffii oleo-resin on experimental gastric ulcer models in rats. J. Ethnopharmacol. 1998, 62, 73–78. [Google Scholar] [CrossRef]
  88. Paiva, L.A.F.; Gurgel, L.A.; Campos, A.R.; Silveira, E.R.; Rao, V.S.N. Attenuation of ischemia/reperfusion-induced intestinal injury by oleo-resin from Copaifera langsdorffii in rats. Life Sci. 2004, 75, 1979–1987. [Google Scholar] [CrossRef] [PubMed]
  89. Paiva, L.A.F.; de Alencar Cunha, K.M.; Santos, F.A.; Gramosa, N.V.; Silveira, E.R.; Rao, V.S.N. Investigation on the wound healing activity of oleo-resin from Copaifera langsdorffii in rats. Phyther. Res. 2002, 16, 737–739. [Google Scholar] [CrossRef] [PubMed]
  90. De Lima Silva, J.J.; Guimarães, S.B.; da Silveira, E.R.; de Vasconcelos, P.R.L.; Lima, G.G.; Torres, S.M.; de Vasconcelos, R.C. Effects of Copaifera langsdorffii Desf. on ischemia-reperfusion of randomized skin flaps in rats. Aesthet. Plast. Surg. 2009, 33, 104–109. [Google Scholar] [CrossRef] [PubMed]
  91. Neto, J.N.; Lindoso, M.J.D.S.; Coelho, L.F.; Antonio, R.; Carvalho, R.A.F.; Rodrigues, T.G.P.D.M.; de Araújo, A.G.P.; Girão, M.J.B.C.; Schor, E. Changes in the volume and histology of endometriosis foci in rats treated with copaiba oil (Copaifera langsdorffii). Acta Cirúrgica Bras. 2011, 26, 20–24. [Google Scholar] [CrossRef]
  92. Masson-Meyers, D.S.; Enwemeka, C.S.; Bumah, V.; Andrade, T.A.M.; Cashin, S.E.; Frade, M.A.C. Antimicrobial effects of Copaifera langsdorffii oleoresin in infected rat wounds. Int. J. Appl. Microbiol. Sci. 2013, 2, 9–20. [Google Scholar]
  93. Masson-Meyers, D.S.; Andrade, T.A.M.; Leite, S.N.; Frade, M.A.C. Cytotoxicity and wound healing properties of Copaifera langsdorffii oleoresin in rabbits. Int. J. Nat. Prod. Sci. 2013, 3, 10–20. [Google Scholar]
  94. Masson-Meyers, D.; Enweme, C.S.; Bumah, V.; Andrade, T.; Frade, M.A. Topical treatment with Copaifera langsdorffii oleoresin improves wound healing in rats. Int. J. Phytomed. 2013, 5, 378–386. [Google Scholar]
  95. Barbosa, F.S.; Leite, G.L.D.; Alves, S.M.; Nascimento, A.F.; D’Ávila, V.D.A.; da Costa, C.A. Insecticide effects of Ruta graveolens, Copaifera langsdorffii and Chenopodium ambrosioides against pests and natural enemies in commercial tomato plantation. Acta Sci. Agron. 2011, 33, 37–43. [Google Scholar]
  96. De Alencar Cunha, K.M.; Paiva, L.A.; Santos, F.A.; Gramosa, N.V.; Silveira, E.R.; Rao, V.S. Smooth muscle relaxant effect of kaurenoic acid, a diterpene from Copaifera langsdorffii on rat uterus in vitro. Phyther. Res. 2003, 17, 320–324. [Google Scholar] [CrossRef] [PubMed]
  97. Dos Santos, A.O.; Costa, M.A.; Ueda-Nakamura, T.; Dias-Filho, B.P.; da Veiga-Júnior, V.F.; de Souza Lima, M.M.; Nakamura, C.V. Leishmania amazonensis: Effects of oral treatment with copaiba oil in mice. Exp. Parasitol. 2011, 129, 145–151. [Google Scholar] [CrossRef] [PubMed]
  98. Gomes, N.D.M.; Rezende, C.D.M.; Fontes, S.P.; Hovell, A.M.C.; Landgraf, R.G.; Matheus, M.E.; Pinto, A.D.C.; Fernandes, P.D. Antineoplasic activity of Copaifera multijuga oil and fractions against ascitic and solid Ehrlich tumor. J. Ethnopharmacol. 2008, 119, 179–184. [Google Scholar] [CrossRef] [PubMed]
  99. Gomes, N.M.; Rezende, C.M.; Fontes, S.P.; Matheus, M.E.; Fernandes, P.D. Antinociceptive activity of Amazonian copaiba oils. J. Ethnopharmacol. 2007, 109, 486–492. [Google Scholar] [CrossRef] [PubMed]
  100. Gomes, N.D.M.; de Rezende, C.M.; Fontes, S.P.; Matheus, M.E.; Pinto, A.D.C.; Fernandes, P.D. Characterization of the antinociceptive and anti-inflammatory activities of fractions obtained from Copaifera multijuga Hayne. J. Ethnopharmacol. 2010, 128, 177–183. [Google Scholar] [CrossRef] [PubMed]
  101. Brito, N.M.B.; Brito, M.V.H.; Carvalho, R.D.K.V.; Matos, L.T.D.M.B.; Lobato, R.C.; Correa, S.C.; Brito, R.B. The effect of copaiba balsam on Walker 256 carcinoma inoculated into the vagina and uterine cervix of female rats. Acta Cir. Bras. 2010, 25, 176–180. [Google Scholar] [CrossRef] [PubMed]
  102. Pieri, F.A.; Mussi, M.C.M.; Fiorini, J.E.; Moreira, M.A.S.; Schneedorf, J.M. Bacteriostatic effect of copaiba oil (Copaifera officinalis) against Streptococcus mutans. Braz. Dent. J. 2012, 23, 36–38. [Google Scholar] [CrossRef] [PubMed]
  103. Guimarães, A.L.; Cunha, E.A.; Matias, F.O.; Garcia, P.G.; Danopoulos, P.; Swikidisa, R.; Pinheiro, V.A.; Nogueira, R.J. Antimicrobial activity of copaiba (Copaifera officinalis) and pracaxi (Pentaclethra macroloba) oils against Staphylococcus aureus: Importance in compounding for wound care. Int. J. Pharm. Compd. 2016, 20, 58–62. [Google Scholar] [PubMed]
  104. Dos Santos, A.O.; Izumi, E.; Ueda-Nakamura, T.; Dias-Filho, B.P.; da Veiga-Júnior, V.F.; Vataru Nakamura, C. Antileishmanial activity of diterpene acids in copaiba oil. Mem. Inst. Oswaldo Cruz 2013, 108, 59–64. [Google Scholar] [CrossRef]
  105. Curio, M.; Jacone, H.; Perrut, J.; Pinto, Â.C.; Veiga Filho, V.F.; Silva, R.C.B. Acute effect of Copaifera reticulata Ducke copaiba oil in rats tested in the elevated plus-maze: An ethological analysis. J. Pharm. Pharmacol. 2009, 61, 1105–1110. [Google Scholar] [CrossRef] [PubMed]
  106. Silva, I.G.; Zanon, V.O.M.; Silva, H.H.G. Larvicidal activity of Copaifera reticulata Ducke oil-resin against Culex quinquefasciatus Say (Diptera: Culicidae). Neotrop. Entomol. 2003, 32, 729–732. [Google Scholar] [CrossRef]
  107. Da Silva, H.H.G.; Geris, R.; Rodrigues Filho, E.; Rocha, C.; da Silva, I.G. Larvicidal activity of oil-resin fractions from the Brazilian medicinal plant Copaifera reticulata Ducke (Leguminosae-Caesalpinoideae) against Aedes aegypti (Diptera, Culicidae). Rev. Soc. Bras. Med. Trop. 2007, 40, 264–267. [Google Scholar] [CrossRef] [PubMed]
  108. Fernandes, F.D.F.; Freitas, E.D.P.S. Acaricidal activity of an oleoresinous extract from Copaifera reticulata (Leguminosae: Caesalpinioideae) against larvae of the southern cattle tick, Rhipicephalus (Boophilus) microplus (Acari: Ixodidae). Vet. Parasitol. 2007, 147, 150–154. [Google Scholar] [CrossRef] [PubMed]
  109. Dos Santos, A.O.; Ueda-Nakamura, T.; Dias Filho, B.P.; da Veiga Junior, V.F.; Nakamura, C.V. Copaiba oil: An alternative to development of new drugs against leishmaniasis. Evid.-Based Complement. Altern. Med. 2012, 2012. [Google Scholar] [CrossRef] [PubMed]
  110. Pacheco, T.A.R.C.; Barata, L.E.S.; Duarte, M.C.T. A ntimicrobial activity of copaiba (Copaifera spp.) balsams. Rev. Bras. Plantas Med. 2006, 8, 123–124. [Google Scholar]
  111. Izumi, E.; Ueda-Nakamura, T.; Veiga, V.F.; Pinto, A.C.; Nakamura, C.V. Terpenes from Copaifera demonstrated in vitro antiparasitic and synergic activity. J. Med. Chem. 2012, 55, 2994–3001. [Google Scholar] [CrossRef] [PubMed]
  112. Soares, D.C.; Portella, N.A.; Ramos, M.F.D.S.; Siani, A.C.; Saraiva, E.M. trans-β-Caryophyllene: An effective antileishmanial compound found in commercial copaiba oil (Copaifera spp.). Evid.-Based Complement. Altern. Med. 2013, 2013. [Google Scholar] [CrossRef] [PubMed]
  113. Ogungbe, I.V.; Setzer, W.N. The potential of secondary metabolites from plants as drugs or leads against protozoan neglected diseases-Part III: In-silico molecular docking investigations. Molecules 2016, 21, 1389. [Google Scholar] [CrossRef] [PubMed]
  114. Ogungbe, I.V.; Setzer, W.N. In-silico Leishmania target selectivity of antiparasitic terpenoids. Molecules 2013, 18, 7761–7847. [Google Scholar] [CrossRef] [PubMed]
  115. Ogungbe, I.V.; Ng, J.D.; Setzer, W.N. Interactions of antiparasitic alkaloids with Leishmania protein targets: A molecular docking analysis. Future Med. Chem. 2013, 5, 1777–1799. [Google Scholar] [CrossRef] [PubMed]
  116. Ogungbe, I.V.; Erwin, W.R.; Setzer, W.N. Antileishmanial phytochemical phenolics: Molecular docking to potential protein targets. J. Mol. Graph. Model. 2014, 48, 105–117. [Google Scholar] [CrossRef] [PubMed]
  117. Mcculley, S.F.; Setzer, W.N. An in-silico investigation of anti-Chagas phytochemicals. Curr. Clin. Pharmacol. 2014, 9, 205–257. [Google Scholar] [CrossRef] [PubMed]
  118. Warfield, J.; Setzer, W.N.; Ogungbe, I.V. Interactions of antiparasitic sterols with sterol 14α-demethylase (CYP51) of human pathogens. SpringerPlus 2014, 3, 679. [Google Scholar] [CrossRef] [PubMed]
  119. Setzer, M.S.; Sharifi-Rad, J.; Setzer, W.N. The search for herbal antibiotics: An in-silico investigation of antibacterial phytochemicals. Antibiotics 2016, 5, 30. [Google Scholar] [CrossRef] [PubMed]
  120. Veeck, J.; Esteller, M. Breast cancer epigenetics: From DNA methylation to microRNAs. J. Mammary Gland Biol. Neoplasia 2010, 15, 5–17. [Google Scholar] [CrossRef] [PubMed]
  121. Chen, T.; Hevi, S.; Gay, F.; Tsujimoto, N.; He, T.; Zhang, B.; Ueda, Y.; Li, E. Complete inactivation of DNMT1 leads to mitotic catastrophe in human cancer cells. Nat. Genet. 2007, 39, 391–396. [Google Scholar] [CrossRef] [PubMed]
  122. Lin, R.K.; Wu, C.Y.; Chang, J.W.; Juan, L.J.; Hsu, H.S.; Chen, C.Y.; Lu, Y.Y.; Tang, Y.A.; Yang, Y.C.; Yang, P.C.; et al. Dysregulation of p53/Sp1 control leads to DNA methyltransferase-1 overexpression in lung cancer. Cancer Res. 2010, 70, 5807–5817. [Google Scholar] [CrossRef] [PubMed]
  123. Medina-Franco, J.L.; Caulfield, T. Advances in the computational development of DNA methyltransferase inhibitors. Drug Discov. Today 2011, 16, 418–425. [Google Scholar] [CrossRef] [PubMed]
  124. Maldonado-Rojas, W.; Olivero-Verbel, J.; Marrero-Ponce, Y. Computational fishing of new DNA methyltransferase inhibitors from natural products. J. Mol. Graph. Model. 2015, 60, 43–54. [Google Scholar] [CrossRef] [PubMed]
  125. Isshiki, Y.; Kohchi, Y.; Iikura, H.; Matsubara, Y.; Asoh, K.; Murata, T.; Kohchi, M.; Mizuguchi, E.; Tsujii, S.; Hattori, K.; et al. Design and synthesis of novel allosteric MEK inhibitor CH4987655 as an orally available anticancer agent. Bioorg. Med. Chem. Lett. 2011, 21, 1795–1801. [Google Scholar] [CrossRef] [PubMed]
  126. Weekes, C.D.; Von Hoff, D.D.; Adjei, A.A.; Leffingwell, D.P.; Eckhardt, S.G.; Gore, L.; Lewis, K.D.; Weiss, G.J.; Ramanathan, R.K.; Dy, G.K.; et al. Multicenter phase I trial of the mitogen-activated protein kinase 1/2 inhibitor BAY 86-9766 in patients with advanced cancer. Clin. Cancer Res. 2013, 19, 1232–1243. [Google Scholar] [CrossRef] [PubMed]
  127. Warner, M.; Gustafsson, J.Å. The role of estrogen receptor β (ERβ) in malignant diseases—A new potential target for antiproliferative drugs in prevention and treatment of cancer. Biochem. Biophys. Res. Commun. 2010, 396, 63–66. [Google Scholar] [CrossRef] [PubMed]
  128. Warner, M.; Huang, B.; Gustafsson, J.A. Estrogen receptor β as a pharmaceutical target. Trends Pharmacol. Sci. 2017, 38, 92–99. [Google Scholar] [CrossRef] [PubMed]
  129. Tibes, U. Phospholipase A2 inhibitors in development. Expert Opin. Investig. Drugs 1997, 6, 279–298. [Google Scholar] [CrossRef] [PubMed]
  130. Wagner, K.M.; McReynolds, C.B.; Schmidt, W.K.; Hammock, B.D. Soluble epoxide hydrolase as a therapeutic target for pain, inflammatory and neurodegenerative diseases. Pharmacol. Ther. 2017, 180, 62–76. [Google Scholar] [CrossRef] [PubMed]
  131. Setzer, W.N.; Byler, K.G. In-silico approaches to natural products drug discovery: A review of the recent literature. In Natural Products and Drug Discovery: From Pharmacochemistry to Pharmacological Approaches; Diniz, M.F.F.M., Scotti, M.T., Scotti, L., Alves, M.F.E., Eds.; Editora UFPB: João Pessoa, Brazil, 2018. [Google Scholar]
  132. Setzer, M.S.; Byler, K.G.; Ogungbe, I.V.; Setzer, W.N. Natural products as new treatment options for trichomoniasis: A molecular docking investigation. Sci. Pharm. 2017, 85, 5. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Clerodane diterpenoids found in Copaifera species.
Figure 1. Clerodane diterpenoids found in Copaifera species.
Ijms 19 01511 g001
Figure 2. Kaurane diterpenoids found in Copaifera species.
Figure 2. Kaurane diterpenoids found in Copaifera species.
Ijms 19 01511 g002
Figure 3. Labdane diterpenoids found in Copaifera species.
Figure 3. Labdane diterpenoids found in Copaifera species.
Ijms 19 01511 g003
Table 1. Major components of the essential oils of Copaifera species.
Table 1. Major components of the essential oils of Copaifera species.
Copaifera spp.EO SourceMajor Components (>5%)Ref.
C. cearensis Huber ex Duckeoleoresinα-copaene (8.2%),
β-caryophyllene (19.7%),
δ-cadinene (7.2%),
β-bisabolol (8.2%),
hardwickiic acid (6.2%),
clorechinic acid (11.3%)
[13]
C. cearensis Huber ex Duckeoleoresinα-copaene (8.2%),
β-caryophyllene (19.7%),
hardwickiic acid (6.2%)
[48]
C. duckei Dwyeroleoresinβ-caryophyllene (0.7–6.2%),
trans-α-bergamotene (3.4–7.9%),
β-selinene (5.5–7.3%),
β-bisabolene (8.9–12.1%),
kaur-16-en-19-oic acid (19.8–24.5%),
polyalthic acid (17.1–27.7%),
hardwickiic acid (0–24.3%)
[8]
C. duckei Dwyeroleoresinβ-elemene (8.3–9.4%),
β-caryophyllene (13.0–15.5%),
trans-α-bergamotene (8.3–10.6%),
β-selinene (13.8–15.4%),
α-selinene (8.8–9.9%),
β-bisabolene (15.7–17.6%)
[49]
C. duckei Dwyeroleoresinβ-caryophyllene (25.1–50.2%),
trans-α-bergamotene (6.4–12.0%),
(E)-β-farnesene (2.9–5.8%),
β-selinene (1.8–6.7%),
β-bisabolene (5.2–33.6%)
[49]
C. guianensis Desf.oleoresintrans-α-bergamotene (7.2%),
caryophyllene oxide (19.1%),
kaur-16-en-19-oic acid (17.5%),
polyalthic acid (10.6%),
hardwickiic acid (11.0%)
[8]
C. langsdorffii Desf.oleoresinβ-caryophyllene (32.8%),
kaurenoic acid (44.3%),
copalic acid (5.6%),
hardwickiic acid (8.2%)
[48]
C. langsdorffii Desf.oleoresincyclosativene (5.0%),
β-elemene (5.1%),
β-caryophyllene (5.5%),
trans-α-bergamotene (48.4%),
β-selinene (5.0%),
α-himachalene (11.2%)
[50]
C. langsdorffii Desf.oleoresinα-copaene (5.8%),
γ-muurolene (22.7%),
eremophilone (6.8%),
kaurene (6.8%),
methyl oleate (26.5%)
[51]
C. langsdorffii Desf.oleoresinβ-elemene (8.0%),
β-caryophyllene (31.4%),
trans-α-bergamotene (10.2%),
γ-muurolene (16.1%)
[52]
C. langsdorffii Desf.oleoresinβ-caryophyllene (1.1–9.0%),
germacrene D (4.0–18.0%),
bicyclogermacrene (1.5–5.7%),
spathulenol (12.6–35.7%),
caryophyllene oxide (7.4–16.6%),
α-cadinol (3.2–7.9%)
[53]
C. langsdorffii Desf.pericarpα-copaene (3.2–14.4%),
β-elemene (0–11.1%),
β-caryophyllene (2.7–10.5%),
germacrene D (1.9–10.7%),
bicyclogermacrene (0–6.3%),
spathulenol (2.2–16.2%),
caryophyllene oxide (4.0–5.1%),
iso-spathulenol (5.6–21.6%)
[54]
C. langsdorffii Desf.leafα-copaene (1.8–6.9%),
β-elemene (0–8.4%),
β-caryophyllene (5.7–17.5%),
germacrene D (0–17.3%),
bicyclogermacrene (0–11.5%),
δ-cadinene (1.6–6.1%),
spathulenol (3.8–12.4%),
caryophyllene oxide (0–15.6%),
α-muurolol (4.8–6.2%),
α-cadinol (4.9–6.8%)
[54]
C. langsdorffii Desf.leafβ-caryophyllene (10.1–16.8%),
germacrene D (9.1–45.2%),
bicyclogermacrene (4.8–21.1%),
spathulenol (4.9–29.4%),
caryophyllene oxide (3.8–18.8%)
[55]
C. langsdorffii Desf.seedcoumarin (0–12.6%),
spathulenol (19.4–38.9%),
caryophyllene oxide (0–21.8%),
humulene epoxide II (0–5.1%),
iso-spathulenol (6.9–25.8%),
τ-muurolol (1.3–5.0%),
α-cadinol (2.0–10.4%)
[54]
C. langsdorffii Desf.stemβ-caryophyllene (2.4–13.9%),
germacrene D (0–19.1%),
bicyclogermacrene (0–8.0%),
δ-cadinene (0–5.7%),
spathulenol (3.6–13.7%),
caryophyllene oxide (4.9–13.3%),
iso-spathulenol (0–7.9%),
τ-muurolol (3.4–7.9%),
α-cadinol (4.9–11.5%)
[54]
C. lucens Dwyeroleoresinpolyalthic acid (69.8%),
copalic acid (11.1%)
[48]
C. martii Hayneoleoresinα-copaene (36.4–51.2%),
β-elemene (4.1–6.2%),
allo-aromadendrene (4.2–5.0%),
δ-cadinene (13.7–17.2%)
[56]
C. martii Hayneoleoresinβ-bisabolene (10.7%),
zingiberene (7.2%),
kaurenoic acid (7.9%),
kovalenic acid (29.0%)
[48]
C. multijuga Hayneoleoresinα-copaene (2.1–5.2%),
β-caryophyllene (42.9–60.3%),
trans-β-bergamotene (2.0–7.0%),
caryophyllene oxide (tr–8.8%),
copalic acid (1.9–11.0%),
3-acetoxycopalic acid (0.8–6.2%)
[8]
C. multijuga Hayneoleoresinβ-caryophyllene (57.5%),
α-humulene (8.3%),
copalic acid (6.2%)
[57]
C. multijuga Hayneoleoresinβ-caryophyllene (57.5%),
α-humulene (8.3%),
copalic acid (6.2%)
[13]
C. multijuga Hayneoleoresinβ-caryophyllene (60.2%),
trans-α-bergamotene (6.4%),
α-humulene (8.6%),
copalic acid (9.5%)
[44]
C. multijuga Hayneoleoresinβ-caryophyllene (57.5%),
copalic acid (6.2%)
[48]
C. multijuga Hayneoleoresinα-copaene (18.8%),
β-caryophyllene (36.0%),
trans-α-bergamotene (7.0%),
β-bisabolene (8.5%),
δ-cadinene (6.1%)
[58]
C. multijuga Hayneoleoresinα-copaene (2.0–15.0%),
β-caryophyllene (5.1–64.0%),
α-humulene (0–8.9%),
germacrene D (0–16.7%),
δ-cadinene (0–5.4%),
caryophyllene oxide (0.2–31.5%),
copalic acid (1.7–7.1%)
[17]
C. multijuga Hayneoleoresinβ-caryophyllene (57.1%),
α-humulene (10.2%),
β-sesquiphellandrene (9.9%)
[59]
C. multijuga Hayneoleoresinα-copaene (2.5–14.9%),
β-caryophyllene (10.6–62.7%),
α-humulene (2.4–8.7%),
germacrene D (0–18.9%),
caryophyllene oxide (0.2–32.5%),
copalic acid (1.1–5.2%)
[60]
C. multijuga Hayneoleoresinα-copaene (5.0%),
β-gurjunene (5.3%),
β-caryophyllene (29.6%),
α-humulene (5.7%),
caryophyllene alcohol (5.8%),
caryophyllene oxide (13.0%)
[61]
C. multijuga Hayneoleoresinβ-caryophyllene (58.4%),
α-humulene (8.4%),
copalic acid (6.3%)
[61]
C. officinalis (Jacq.) L.oleoresinβ-caryophyllene (8.5%),
copalic acid (13.9%),
hardwickiic acid (30.7%)
[48]
C. officinalis (Jacq.) L.oleoresinβ-caryophyllene (24.9%),
allo-aromadendrene (7.5%),
germacrene B (5.1%),
β-bisabolene (6.3%),
δ-cadinene (15.3%),
α-cadinene (5.6%)
[62]
C. officinalis (Jacq.) L.oleoresinβ-caryophyllene (62.7%),
trans-α-bergamotene (7.6%),
α-humulene (8.1%)
Setzer a
C. officinalis (Jacq.) L.oleoresinβ-caryophyllene (87.3%)Setzer b
C. paupera (Herzog) Dwyeroleoresinβ-bisabolene (20.2%),
zingiberene (19.4%),
kaurenoic acid (13.3%),
copalic acid (6.1%)
[48]
C. paupera (Herzog) Dwyeroleoresinα-cubebene (5.5%),
α-copaene (42.5%),
β-caryophyllene (14.1%),
δ-cadinene (10.4%)
[63]
C. piresii Duckeoleoresinα-copaene (45.5%),
β-caryophyllene (10.3%),
δ-cadinene (13.7%)
[63]
C. publflora Benth.oleoresinβ-caryophyllene (65.9%),
α-humulene (7.3%),
β-selinene (10.2%),
α-selinene (5.5%)
[63]
C. reticulata Duckeoleoresinβ-caryophyllene (40.9%),
α-humulene (6.0%),
germacrene D (5.0%)
[13]
C. reticulata Duckeoleoresin (Pará)β-caryophyllene (40.9%)[48]
C. reticulata Duckeoleoresin (Acre)α-copaene (25.1%),
β-caryophyllene (13.1%),
copalic acid (7.7%),
kaurenoic acid (7.5%),
hardwickiic acid (6.9%)
[48]
C. reticulata Duckeoleoresinβ-elemene (0.5–5.6%),
β-caryophyllene (1.4–68.0%),
trans-α-bergamotene (2.4–29.6%),
α-humulene (1.1–9.7%),
β-selinene (0–20.6%),
α-selinene (0–13.2%),
β-bisabolene (3.7–42.4%),
caryophyllene oxide (0.1–15.2%)
[64]
C. reticulata Duckeoleoresinβ-elemene (0–6.0%),
β-caryophyllene (0–43.4%),
trans-α-bergamotene (12.0–32.8%),
α-guaiene (0–9.5%),
α-humulene (0–7.0%),
β-selinene (0–17.1%),
α-selinene (0–10.4%),
trans-β-guaiene (0–5.8%),
β-bisabolene (24.2–50.3%)
[65]
C. reticulata Duckeoleoresinβ-caryophyllene (25.1–50.2%),
trans-α-bergamotene (6.4–12.0%),
α-humulene (4.1–5.8%),
β-selinene (1.8–6.7%),
β-bisabolene (5.2–17.4%)
[66]
C. reticulata Duckeoleoresinβ-caryophyllene (37.3%),
trans-α-bergamotene (9.0%),
α-humulene (5.4%),
β-bisabolene (14.5%)
[67]
C. reticulata Duckeoleoresinβ-caryophyllene (7.7%),
trans-α-bergamotene (22.0%),
β-selinene (12.2%),
α-selinene (11.4%),
β-bisabolene (24.9%)
[68]
C. trapezifolia Hayneleafβ-caryophyllene (33.5%),
α-humulene (6.2%),
germacrene D (11.0%),
spathulenol (7.6%)
[69]
a Unpublished analysis of a commercial essential oil from New Directions Aromatics (Sydney, Australia). b Unpublished analysis of a commercial essential oil from Améo Essential Oils/Zija International (Lehi, Utah).
Table 2. Nonvolatile components isolated and/or identified from Copaifera species.
Table 2. Nonvolatile components isolated and/or identified from Copaifera species.
Copaifera spp.Plant PartCompounds Isolated and/or IdentifiedRef.
C. cearensis Huber ex Duckeoleoresinepuric acid, cativic acid, copalic acid, kolavenic acid, crolechinic acid, hardwickiic acid, haplociliatic acid, labdanolic acid, patagonic acid.[76]
C. guianensis Desf.oleoresinmethyl copalate[74]
C. langsdorffii Desf.oleoresincopalic acid, kaurenoic acid, acetoxycopalic acid (=3α-alepterolic acid acetate), ent-agathic acid, hydroxycopalic acid (=3α-alepterolic acid)[71]
C. langsdorffii Desf.oleoresincopalic acid, acetoxycopalic acid (=3α-alepterolic acid acetate), 3-hydroxy-14,15-dinorlabd-8(17)-en-13-one, ent-agathic acid, hydroxycopalic acid
(=3α-alepterolic acid)
[70]
C. langsdorffii Desf.oleoresinkaurenoic acid[72]
C. langsdorffii Desf.oleoresinkaurenoic acid[73]
C. langsdorffii Desf.oleoresinkaurenoic acid[74]
C. langsdorffii Desf.oleoresinsclareol, manool, copalic acid, acetoxycopalic acid (=3α-alepterolic acid acetate), hydroxycopalic acid (=3α-alepterolic acid), ent-agathic acid[77]
C. langsdorffii Desf.leaveskaurenoic acid, quercitrin, afzelin[78]
C. langsdorffii Desf.leavesrutin, quercetin-3-O-α-l-rhamnopyranoside, kaempferol-3-O-α-l-rhamnopyranoside, quercetin, kaempferol[79]
C. langsdorffii Desf.fruitgallic acid, epicatechin gallate, catechin, epicatechin, isoquercitrin[80]
C. langsdorffii Desf.leaveskaurenoic acid, quercitrin, afzelin, eupatorin, galloyl quinic acid, gallic acid 4-O-glucoside[81]
C. multijuga Hayneoleoresincopalic acid, 3-hydroxycopalic acid
(=3α-alepterolic acid), 3-acetoxycopalic acid
(=3α-alepterolic acid acetate)
[74]
C. paupera (Herzog) Dwyeroleoresincopalic acid, methyl copalate, agathic acid 15-methyl ester, agathic acid 15,19-dimethyl ester, ent-polyalthic acid, methyl ent-polyalthate, ent-pinifolic acid, methyl 3β-hydroxy-labda-8(17),13-dien-15-ate, methyl 18-hydroxy-copaiferolate, 14,15-bisnorlabd-8(17)-en-13-one, ent-kaurenic acid, 16β-kauran-19-oic acid, 3-methyl-5-(2,2,6-trimethyl-6-hydroxycyclohexyl)- pentanoic acid, pauperol[75]
C. reticulata Duckeoleoresin3β-alepterolic acid, 3β-alepterolic acid acetate, 3β-hydroxylabdan-8(17)-en-15-oic acid, ent-agathic acid[82]
Table 3. Biological activities of Copaifera oleoresins, essential oils, and isolated components.
Table 3. Biological activities of Copaifera oleoresins, essential oils, and isolated components.
Copaifera spp.MaterialBiological ActivityRef.
C. cearensis Huber ex DuckeoleoresinAnti-inflammatory: At a concentration of 50 μg/mL, in vitro NO production in mouse peritoneal macrophages was significantly reduced by C. cearensis oil.[13]
C. cearensis Huber ex DuckeoleoresinAntileishmanial: L. amazonensis promastigotes (IC50 = 18.0 μg/mL).[48]
C. cearensis Huber ex DuckeoleoresinAntibacterial: Methicillin-resistant Staphylococcus aureus (MRSA, MIC = 125 μg/mL),
B. subtilis (MIC = 62.5 μg/mL), Enterococcus faecalis (MIC = 62.5 μg/mL)
[83]
C. duckei DwyeroleoresinAnti-inflammatory: Carrageenin-induced paw edema test (rats, 18% edema inhibition with dose of 1802 mg/kg; granuloma test, 42% inhibition with dose of 1802 mg/kg);
croton oil-induced dermatitis test (mice, IC50 = 663 mg/kg)
[84]
C. duckei DwyeroleoresinAntinociceptive: Mouse model (acetic acid-induced writhing test, IC50 = 704 mg/kg)[84]
C. duckei DwyeroleoresinAntiproliferative: In vivo hepatocellular proliferation, partial hepatectomy, rats. Hepatocellular proliferation and liver mitochondrial respiration were significantly lower in C. duckei treated rats compared to control (saline solution).[85]
C. duckei DwyeroleoresinAntitrypanosomal: T. evansi, in vivo mouse model, doses of 0.63 mL/kg/day over 5 days showed no curative effects. T. evansi, in vitro trypomastigotes, 0.5% solution of C. duckei oil showed 100% killing after 6 h.[86]
C. langsdorffii Desf.oleoresinAnti-inflammatory: Preincubation of LPS-stimulated human THP-1 monocytes with diterpenoid-rich oleoresin reduced the release of proinflammatory cytokines
(IL-1β, IL-6, TNFα).
[50]
C. langsdorffii Desf.oleoresinAntifungal: Tricophyton mentagrophytes (MIC = 170 μg/mL). Scanning electron microscopic (SEM) analysis revealed physical damage and morphological alterations of the fungi upon exposure to copaiba oleoresin.[52]
C. langsdorffii Desf.oleoresinAntipsoriatic: Human clinical trial, topical application of copaiba resin exhibited a significant improvement of the typical signs of psoriasis.[50]
C. langsdorffii Desf.oleoresinGastroprotective: Ethanol or indomethacin-induced ulcer in rats, oral administration of copaiba resin at doses of 400 mg/kg provided dose-dependent significant protection against gastric damage caused by ethanol or indomethacin.[87]
C. langsdorffii Desf.oleoresinGastroprotective: Mesenteric ischemia/reperfusion (I/R) in rats: Copaiba resin treatment caused significant attenuations in I/R-associated increases of myeloperoxidase, malondialdehyde, and catalase, and effectively prevented the I/R-associated depletion of glutathione.[88]
C. langsdorffii Desf.oleoresinWound-healing: Rat incision wounds, 4% oleoresin topically applied showed significant wound contraction and tensile strength compared to controls.[89]
C. langsdorffii Desf.oleoresinWound healing: Rat dorsal skin flaps, oral administration of copaiba oleoresin
(400 mg/kg), copaiba-treatment presented discrete anti-lipoperoxidation action, intense antioxidant action, and anti-inflammatory activity during the ischemia and reperfusion of randomized cutaneous flaps.
[90]
C. langsdorffii Desf.oleoresinAnti-inflammatory: Rat model of experimental endometriosis. Copaiba oil caused a marked reduction in endometrial growth.[91]
C. langsdorffii Desf.oleoresinAntileishmanial: L. amazonensis promastigotes (IC50 = 20.0 μg/mL).[48]
C. langsdorffii Desf.oleoresinAntibacterial: B. subtilis (MIC = 62.5 μg/mL)[83]
C. langsdorffii Desf.10% copaiba oil ointmentWound-healing: Rat dorsal skin flaps, topical copaiba oil ointment favors angiogenesis and accelerates the viability of random skin flaps in rats.[51]
C. langsdorffii Desf.10% oleoresin creamAntibacterial: Open wounds on rats inoculated with Streptococcus pyogenes or Staphylococcus aureus. Treatment with 10% copaiba cream reduced bacterial populations to 0.02% (S. pyogenes) and 0.3% (S. aureus) after 14 days.[92]
C. langsdorffii Desf.10% oleoresin creamWound-healing: Rabbit ears, 10% oleoresin cream-treated wounds presented better clinical outcomes, confirmed by histology with evidence of fibroblastic activity by day 7 and organized collagen fibers observed from day 14.[93]
C. langsdorffii Desf.10% oleoresin creamWound-healing: Rat skin biopsy punch on dorsal surface, 10% oleoresin cream-treated wounds showed a faster wound-healing rate compared to saline or cream only controls, by regulating matrix metalloproteinase, (MMP)-2 and MMP-9 activities, stimulating collagen synthesis, and promoting tissue remodeling and re-epithelialization.[94]
C. langsdorffii Desf.3α-alepterolic acid acetateAntibacterial: Cariogenic Streptococcus spp.; MIC range 12.0–60.0 μg/mL[70]
C. langsdorffii Desf.aqueous leaf extractInsecticidal: 5% Extract inhibited Bemisia tabaci infestation of tomato plants in the field.[95]
C. langsdorffii Desf.copalic acidAntibacterial: Gram-positive bacteria (MIC range 0.5 μg/mL to 15.0 μg/mL)[71]
C. langsdorffii Desf.copalic acidAntibacterial: Cariogenic Streptococcus spp.; MIC range 2.0–6.0 μg/mL[70]
C. langsdorffii Desf.copalic acidAntibacterial: Periodontal anaerobic bacteria: Actinomyces naeslundii (MIC 6.2 μg/mL), Bacteroides fragilis (MIC 25.0 μg/mL), Peptostreptococcus anaerobius (MIC 3.1 μg/mL), Porphyromonas gingivalis (MIC 3.1 μg/mL).[77]
C. langsdorffii Desf.copalic acidAntiproliferative: In vitro cytotoxicity on MO59J (human glioblastoma cells,
IC50 = 68.3 μg/mL) and HeLa (human cervical adenocarcinoma cells, IC50 = 44.0 μg/mL).
[71]
C. langsdorffii Desf.EtOH/H2O leaf extractGastroprotective: Ethanol/HCl-induced ulcer in mice, the extract (500 mg/kg) showed a significant decrease in the total gastric juice acidity and an increase in mucus production; isolated compounds (30 mg/kg) α-humulene, β-caryophyllene and caryophyllene oxide showed greater gastroprotective activity in the ethanol/HCl induced ulcer model.[78]
C. langsdorffii Desf.kaurenoic acidAnti-inflammatory: Rat model of acetic acid-induced colitis. A marked reduction in gross damage score (52% and 42%) and wet weight of damaged colon tissue (39% and 32%) were observed in rats that received 100 mg/kg kaurenoic acid, respectively,
by rectal and oral routes.
[72]
C. langsdorffii Desf.kaurenoic acidAntibacterial: Gram-positive bacteria (MIC range 5.0 μg/mL to 100.0 μg/mL)[71]
C. langsdorffii Desf.kaurenoic acidAntiproliferative: In vitro cytotoxicity, 78 μM concentration, on CEM (human leukemia, 95% growth inhibition), MCF-7 (human breast tumor, 45% growth inhibition), and HCT-8 (human colon tumor, 45% growth inhibition) cells.[73]
C. langsdorffii Desf.kaurenoic acidAntiproliferative: In vitro cytotoxicity, 20 μM concentration, on AGP01
(human gastric cancer, 28% growth inhibition) and SF-295 (human glioblastoma, 28% growth inhibition) cells.
[74]
C. langsdorffii Desf.kaurenoic acidSmooth muscle relaxant: Rat uterine muscle ex vivo: kaurenoic acid, exerts a uterine relaxant effect acting principally through calcium blockade and in part, by the opening of ATP-sensitive potassium channels.[96]
C. lucens DwyeroleoresinAntileishmanial: L. amazonensis promastigotes (IC50 = 20.0 μg/mL).[48]
C. lucens DwyeroleoresinAntibacterial: S. aureus (MIC = 125 μg/mL), B. subtilis (MIC = 125 μg/mL)[83]
C. martii HayneoleoresinAntileishmanial: L. amazonensis promastigotes (IC50 = 14.0 μg/mL).[48]
C. martii HayneoleoresinAntileishmanial: In vivo mouse model, copaiba oil oral treatment (100 mg/kg/day) caused a significant reduction in the average lesion size (1.1 ± 0.4 mm) against Leishmania amazonensis lesions compared with untreated mice (4.4 ± 1.3 mm).[97]
C. martii HayneoleoresinAntibacterial: S. aureus (MIC = 62.5 μg/mL), MRSA (MIC = 62.5 μg/mL), B. subtilis
(MIC = 15.6 μg/mL), S. epidermidis (MIC = 62.5 μg/mL), Enterococcus faecalis
(MIC = 62.5 μg/mL)
[83]
C. multijuga HayneoleoresinAntiproliferative: In vitro cytotoxicity, B16F10 (murine melanoma cells, IC50 = 457 μg/mL).[57]
C. multijuga HayneoleoresinAntiproliferative: In vivo lung metastasis and tumor growth, mouse model: Oral administration of C. multijuga oleoresin (at 2 g/Kg in the days 3, 5, 7, 10, 12, and 14 after inoculation of tumoral cells) reduced tumor growth by 58% and tumor weight by 76% and reduced the number of lung nodules by 47.1%.[57]
C. multijuga HayneoleoresinAntiproliferative: In vivo Ehrlich ascitic and solid tumor, mouse model: C. multijuga oleoresin (doses varying between 100 and 200 mg/kg) showed antineoplastic properties against Ehrlich ascitic tumor (EAT) and solid tumor during 10 consecutive
days of treatment.
[98]
C. multijuga HayneoleoresinInsecticidal: Mosquito larvicidal activity (Anopheles darlingi, LC50 = 31 μg/mL; Aedes aegypti, LC50 = 93 μg/mL)[59]
C. multijuga HayneoleoresinAnti-inflammatory: The β-caryophyllene-rich oleoresin oil of C. multijuga, at a dose of
100 mg/kg, inhibited zymosan-induced pleurisy in a mouse model, reducing total leukocytes by 45% and neutrophil accumulation by 73%. C. multijuga oil also showed in vitro reduction of NO production in mouse peritoneal macrophages at a concentration of 5 μg/mL.
[13]
C. multijuga HayneoleoresinAnti-inflammatory: Rat pleurisy model, doses of 100 mg/kg and 200 mg/kg presented in vivo anti-inflammatory effects.[58]
C. multijuga HayneoleoresinAntileishmanial: L. amazonensis promastigotes (IC50 = 10.0 μg/mL).[48]
C. multijuga HayneoleoresinAntibacterial: MRSA (MIC = 125 μg/mL), B. subtilis (MIC = 125 μg/mL)[83]
C. multijuga HayneoleoresinAntinociceptive: Mouse model (acetic acid-induced writhing, tail flick, hot plate), oral administration of C. multijuga oil with doses of 30–150 mg/kg significantly showed antinociception, which was reversed with naloxone.[99]
C. multijuga HayneoleoresinInsecticidal: Mosquito larvicidal activity (Anopheles darlingi, LC50 = 128 μg/mL;
Aedes aegypti, LC50 = 18 μg/mL)
[59]
C. multijuga Hayneoleoresin extractsAnti-inflammatory: The CH2Cl2 and MeOH fractions obtained from C. multijuga oleoresin, given by the intraperitoneal route, caused a significant inhibition of carrageenan-induced rat paw edema with inhibition of 49 ± 13% and 64 ± 9%, respectively.[61]
C. multijuga Hayneoleoresin extractsAnti-inflammatory: The hexane, chloroform and methanol fractions obtained from
C. multijuga oleoresin, given by the oral gavage, caused a significant inhibition of carrageenan-induced rat paw edema.
[100]
C. multijuga Hayneoleoresin extractsAntinociceptive: The hexane, chloroform and methanol fractions obtained from
C. multijuga oleoresin, given by the oral gavage, caused a significant inhibition (in a concentration-dependent way) the number of contortions induced by acetic acid and the second phase of formalin-induced licking response. Similar results were observed in the tail flick model; administration of naloxone inhibited the antinociceptive effect of the fractions indicating that copaiba resin maybe acting on opioid receptors.
[100]
C. multijuga Hayne3β-alepterolic acidAntiproliferative: In vitro cytotoxicity, 20 μM concentration, on AGP01 (human gastric cancer, 8.5% growth inhibition) and SF-295 (human glioblastoma, 21% growth inhibition) cells.[74]
C. multijuga Hayne3β-alepterolic acid acetateAntiproliferative: In vitro cytotoxicity, 20 μM concentration, on AGP01 (human gastric cancer, 13% growth inhibition) and SF-295 (human glioblastoma, 18% growth inhibition) cells.[74]
C. officinalis (Jacq.) L.oleoresinAntiproliferative: In vivo Walker 256 carcinoma inoculated into the vagina and uterine cervix of female rats, C. officinalis oleoresin stimulated tumor growth by 70%.[101]
C. officinalis (Jacq.) L.oleoresin EOAnti-inflammatory: Dias and coworkers investigated the immunomodulatory effects of C. officinalis oleoresin essential oil on inflammatory cytokines (NO, H2O2, TNF-α, IFN-γ, and IL-17) in a murine model of experimental autoimmune encephalomyelitis. At a concentration of 100 μg/mL, C. officinalis oil inhibited the in vitro production of the inflammatory cytokines, modulating the immune response.[62]
C. officinalis (Jacq.) L.oleoresinAntileishmanial: L. amazonensis promastigotes (IC50 = 20.0 μg/mL).[48]
C. officinalis (Jacq.) L.oleoresinAntibacterial: S. aureus (MIC = 62.5 μg/mL), MRSA (MIC = 125 μg/mL), B. subtilis
(MIC = 31.3 μg/mL), S. epidermidis (MIC = 31.3 μg/mL), Enterococcus faecalis
(MIC = 31.3 μg/mL)
[83]
C. officinalis (Jacq.) L.oleoresinAntibacterial: Streptococcus mutans (MIC = 780 μg/mL)[102]
C. officinalis (Jacq.) L.oleoresinAntibacterial: Staphylococcus aureus (MIC = 312.5 μg/mL)[103]
C. officinalis (Jacq.) L.agathic acidAntileishmanial: L. amazonensis promastigotes (IC50 = 28.0 μg/mL), amastigotes
(IC50 = 17.0 μg/mL)
[104]
C. officinalis (Jacq.) L.alepterolic acid
(=hydroxycopalic acid)
Antileishmanial: L. amazonensis promastigotes (IC50 = 2.5 μg/mL), amastigotes
(IC50 = 18.0 μg/mL)
[104]
C. officinalis (Jacq.) L.kaurenoic acidAntileishmanial: L. amazonensis promastigotes (IC50 = 28.0 μg/mL), amastigotes
(IC50 = 3.5 μg/mL)
[104]
C. officinalis (Jacq.) L.methyl copalateAntileishmanial: L. amazonensis promastigotes (IC50 = 6.0 μg/mL), amastigotes
(IC50 = 14.0 μg/mL)
[104]
C. officinalis (Jacq.) L.pinifolic acidAntileishmanial: L. amazonensis promastigotes (IC50 = 70.0 μg/mL), amastigotes
(IC50 = 4.0 μg/mL)
[104]
C. officinalis (Jacq.) L.ent-polyalthic acidAntileishmanial: L. amazonensis promastigotes (IC50 = 35.0 μg/mL), amastigotes
(IC50 = 15.0 μg/mL)
[104]
C. paupera (Herzog) DwyeroleoresinAntileishmanial: L. amazonensis promastigotes (IC50 = 11.0 μg/mL).[48]
C. paupera (Herzog) DwyeroleoresinAntibacterial: B. subtilis (MIC = 62.5 μg/mL), Enterococcus faecalis (MIC = 62.5 μg/mL)[83]
C. paupera (Herzog) Dwyercopalic acidAntibacterial: Bacillus subtilis (MIC = 3.1–6.3 μg/mL), Staphylococcus aureus
(MIC = 8–10 μg/mL), Staphylococcus epidermidis (MIC = 4–5 μg/mL).
[75]
C. paupera (Herzog) Dwyerent-polyalthic acidAntibacterial: Bacillus subtilis (MIC = 20–30 μg/mL), Staphylococcus aureus
(MIC = 40–50 μg/mL), Staphylococcus epidermidis (MIC = 40 μg/mL).
[75]
C. paupera (Herzog) Dwyerkaurenoic acidAntibacterial: Bacillus subtilis (MIC = 2.5–5 μg/mL), Staphylococcus aureus
(MIC = 6–8 μg/mL), Staphylococcus epidermidis (MIC = 4–6 μg/mL).
[75]
C. paupera (Herzog) Dwyermethyl copalateAntiproliferative: In vitro cytotoxicity, P-388 (murine lymphoma, IC50 = 2.5 μg/mL), A-549 (human lung carcinoma, IC50 = 5 μg/mL), HT-29 (human colon carcinoma, IC50 = 5 μg/mL), MEL-28 (human melanoma, IC50 = 10 μg/mL).[75]
C. reticulata DuckeoleoresinAntibacterial: Porphyromonas gingivalis (MIC = 6.25 μg/mL), Streptococcus spp.
(MIC 25–50 μg/mL)
[68]
C. reticulata DuckeoleoresinAntiproliferative: In vitro cytotoxicity, GM07492-A (human lung fibroblast cells,
IC50 = 51.85 μg/mL)
[68]
C. reticulata DuckeoleoresinAnxiolytic: elevated plus-maze test with rats: Oral doses of 100, 400, and 800 mg/kg produced a dose-dependent anxiolytic-like effect over the dose range tested.[105]
C. reticulata DuckeoleoresinInsecticidal: Mosquito larvicidal activity (Culex quinquefasciatus, LC50 = 0.4, 0.9, 39, and
90 μg/mL against the 1st, 2nd, 3rd, and 4th larval instars, respectively)
[106]
C. reticulata DuckeoleoresinInsecticidal: Mosquito larvicidal activity (Aedes aegypti, LC50 = 8.9 μg/mL against the 3rd larval instar)[107]
C. reticulata DuckeoleoresinNeuroprotective: Rat model of motor cortex excitotoxic injury, C. reticulata resin treatment induces neuroprotection by modulating inflammatory response following an acute damage to the central nervous system.[67]
C. reticulata DuckeoleoresinAcaricidal: Rhipicephalus (Boophilus) microplus (LC50 = 1579 μg/mL)[108]
C. reticulata DuckeoleoresinAnti-inflammatory: At a concentration of 500 μg/mL, C. reticulata oleoresin oil showed 85% inhibition of NO production in mouse peritoneal macrophages in vitro.[13]
C. reticulata DuckeoleoresinAntileishmanial: A β-caryophyllene-rich C. reticulata (from Pará state) oleoresin oil showed remarkable activity against L. amazonensis promastigotes with IC50 of 5.0 μg/mL. Another sample of C. reticulata oil (from Acre) with lower concentration of β-caryophyllene was less active (IC50 = 22.0 μg/mL).[48]
C. reticulata Duckeoleoresin EOAntileishmanial: C. reticulata oleoresin essential oil showed significant antileishmanial activity against axenic amastigotes (IC50 = 15.0 μg/mL) and intracellular amastigotes
(IC50 = 20 μg/mL) of L. amazonensis. Note that the major component of the oil,
β-caryophyllene, was inactive against the amastigotes. Interference with the mitochondrial membrane was suggested as the mechanism for antileishmanial activity.
[109]
C. reticulata DuckeoleoresinAntinociceptive: Mouse model (acetic acid-induced writhing, tail flick, hot plate), oral administration of C. reticulata oil with doses of 30–150 mg/kg significantly showed antinociception, which was reversed with naloxone.[99]
C. reticulata DuckeoleoresinAntitrypanosomal: T. evansi, in vivo mouse model, doses of 0.63 mL/kg/day over 5 days showed no curative effects. T. evansi, in vitro trypomastigotes, 0.5% solution of C. reticulata oil showed 100% killing after 6 h.[86]
C. reticulata Duckeoleoresin EO (Acre)Antibacterial: S. aureus (MIC = 62.5 μg/mL), MRSA (MIC = 125 μg/mL), B. subtilis
(MIC = 31.3 μg/mL), S. epidermidis (MIC = 62.5 μg/mL), Enterococcus faecalis
(MIC = 62.5 μg/mL)
[83]
C. reticulata Ducke3β-alepterolic acidInsecticidal: Mosquito larvicidal activity (Aedes aegypti, LC50 = 87.3 μg/mL against the 3rd larval instar)[82]
C. reticulata Ducke3β-alepterolic acid acetateInsecticidal: Mosquito larvicidal activity (Aedes aegypti, LC50 = 0.8 μg/mL against the 3rd larval instar)[82]
Copaifera spp.oleoresinAntibacterial: Oleoresin oils from unidentified species of Copaifera showed remarkable antibacterial activity against the Gram-positive Bacillus subtilis and Staphylococcus aureus (MIC = 5 μg/mL). The oils were inactive against Gram-negative organisms.[110]
Copaifera spp.agathic acidAntitrypanosomal: T. cruzi epimastigotes (IC50 = 86.8 μM), trypomastigotes
(IC50 = 823 μM), amastigotes (IC50 = 14.9 μM)
[111]
Copaifera spp.copalic acidAntitrypanosomal: T. cruzi epimastigotes (IC50 = 47.2 μM), trypomastigotes
(IC50 = 444 μM), amastigotes (IC50 = 1.3 μM). Note: β-caryophyllene + copalic acid showed a significant synergistic effect against T. cruzi trypomastigotes.
[111]
Copaifera spp.alepterolic acid
(=hydroxycopalic acid)
Antitrypanosomal: T. cruzi epimastigotes (IC50 = 41.2 μM), trypomastigotes
(IC50 = 453 μM), amastigotes (IC50 = 1.8 μM)
[111]
Copaifera spp.kaurenoic acidAntitrypanosomal: T. cruzi epimastigotes (IC50 = 167 μM), trypomastigotes
(IC50 = 596 μM), amastigotes (IC50 = 16.5 μM)
[111]
Copaifera spp.methyl copalateAntitrypanosomal: T. cruzi epimastigotes (IC50 = 83.3 μM), trypomastigotes
(IC50 = 377 μM), amastigotes (IC50 = 2.5 μM)
[111]
Copaifera spp.pinifolic acidAntitrypanosomal: T. cruzi epimastigotes (IC50 = 854 μM), trypomastigotes
(IC50 = 1630 μM), amastigotes (IC50 = 18.6 μM)
[111]
Copaifera spp.ent-polyalthic acidAntitrypanosomal: T. cruzi epimastigotes (IC50 = 168 μM), trypomastigotes
(IC50 = 965 μM), amastigotes (IC50 = 28.4 μM)
[111]
Copaifera spp.β-caryophylleneAntileishmanial: L. amazonensis amastigotes (IC50 = 1.3 μg/mL)[112]
Copaifera spp.β-caryophylleneAntitrypanosomal: T. cruzi epimastigotes (IC50 = 78.4 μM), trypomastigotes
(IC50 = 1593 μM), amastigotes (IC50 = 63.7 μM)
[111]
Table 4. MolDock docking energies (kJ/mol) of Copaifera diterpenoids with Leishmania protein targets.
Table 4. MolDock docking energies (kJ/mol) of Copaifera diterpenoids with Leishmania protein targets.
Leishmania TargetsPDBaEdock (ave)Edock (min)Best-Docking Diterpenoid Ligand
Cathepsin B (LdonCatB)homology−84.6−100.63α-Alepterolic acid acetate
Cathepsin B (LmajCatB)homology−80.8−93.77α-Acetoxyhardwickiic acid
Cyclophilin A (LdonCypA)3EOV−83.3−102.6ent-Pinifolic acid
Deoxyuridine triphosphate nucleotidohydrolase (LmajdUTPase)2YAY−85.3−103.819-O-Acetyl-1,2-dehydrokautriwaic acid
Dihydroorotate dehydrogenase (LdonDHODH)3GYE−89.9−102.77α-Acetoxyhardwickiic acid
Dihydroorotate dehydrogenase (LmajDHODH)3MHU−109.2−126.77α-Acetoxyhardwickiic acid
Glucose-6-phosphate isomerase (LmexGPI)1Q50−73.0−85.319-O-Acetyl-1,2-dehydrokautriwaic acid
Glyceraldehyde-3-phosphate dehydrogenase (LmexGAPDH)1A7K−74.2−83.019-O-Acetyl-1,2-dehydrokautriwaic acid
Glycerol-3-phosphate dehydrogenase (LmexGPDH)1N1E−100.4−114.33α-Alepterolic acid acetate
Methionyl-tRNA synthetase (LmajMetRS)3KFL−106.9−123.03α-Alepterolic acid acetate
Nicotinamidase (LinfPnC1)3R2J−61.3−75.93β-Alepterolic acid
N-Myristoyl transferase (LmajNMT)4A30−92.3−104.019-O-Acetyl-1,2-dehydrokautriwaic acid
Nucleoside diphosphate kinase b (LmajNDKb)3NGS−83.9−105.87α-Acetoxyhardwickiic acid
Nucleoside hydrolase (LmajNH)1EZR−80.5−90.57-Acetylbacchotricuneatin D
Oligopeptidase B (LmajOPB)2XE4−97.8−106.17α-Acetoxyhardwickiic acid
Phosphodiesterase B1 (LmajPDEB1)2R8Q−89.5−105.83β-Alepterolic acid acetate
Phosphomannumutase (LmexPMM)2I55−94.2−117.519-O-Acetyl-1,2-dehydrokautriwaic acid
Pteridine reductase 1 (LmajPTR1)1E7W−93.8−110.7Copaiferolic acid
Pyruvate kinase (LmexPYK)1PKL−103.4−113.57α-Acetoxyhardwickiic acid
Sterol 14α-demethylase (LinfCYP51)3L4D−90.2−111.319-O-Acetyl-1,2-dehydrokautriwaic acid
Thiol-dependent reductase I (LinfTDR1)4AGS−78.7−88.819-O-Acetyl-1,2-dehydrokautriwaic acid
Triosephosphate isomerase (LmexTIM)2VXN−90.7−101.53α-Alepterolic acid acetate
Trypanothione reductase (LinfTR)4APN−92.5−109.019-O-Acetyl-1,2-dehydrokautriwaic acid
Tyrosyl-tRNA synthetase (LmajTyrRS)3P0J−92.4−102.9Patagonic acid
Uridine diphosphate-glucose pyrophosphorylase (LmajUGPase)2OEG−99.9−113.9Patagonic acid
a PDB = Protein Data Bank.
Table 5. MolDock docking energies (kJ/mol) of Copaifera diterpenoids with Trypanosoma cruzi protein targets.
Table 5. MolDock docking energies (kJ/mol) of Copaifera diterpenoids with Trypanosoma cruzi protein targets.
Trypanosoma cruzi TargetsPDBEdock (ave)Edock (min)Best-Docking Diterpenoid Ligand
Cruzain3IUT−80.2−92.6Patagonic acid
Cyclophilin (TcCyp19)1XQ7−78.9−92.03β-Alepterolic acid acetate
Deoxyuridine triphosphatase (TcUTPase)1OGK−83.4−101.03β-Alepterolic acid acetate
Dihydrofolate reductase—thymidylate synthase (TcDHFR-TS)3IRN−93.2−110.77α-Acetoxyhardwickiic acid
Dihydroorotate dehydrogenase (TcDHODH)3W6Y−92.5−109.77α-Acetoxyhardwickiic acid
Farnesyl diphosphate synthase (TcFPPS)3ICZ−96.2−109.87α-Acetoxyhardwickiic acid
Glyceraldehyde-3-phosphate dehydrogenase (TcGAPDH)1QXS−70.3−85.3Copaiferolic acid
Hypoxanthine phosphoribosyltransferase (TcHPRT)1P19−82.1−94.47α-Hydroxyhardwickiic acid
Nucleoside diphosphate kinase B (TcNDKb)3PRV−71.6−88.4Crolechinic acid
Old yellow enzyme (=Prostaglandin F2α synthase) (TcPGFS)3ATY−85.6−97.3Patagonic acid
Pteridine reductase 2 (TcPTR2)1MXH−96.8−118.4(+)-Hardwickiic acid
Pyruvate kinase (TcPYK)3QV9−80.3−87.4(−)-Hardwickiic acid
Spermidine synthase (TcSpdSyn)3BWC−96.8−106.819-O-Acetyl-1,2-dehydrokautriwaic acid
Sterol 14α-demethylase (TcCYP51)3KLO−89.5−101.819-O-Acetyl-1,2-dehydrokautriwaic acid
Triosephosphate isomerase (TcTIM)1SUX−88.2−100.7ent-Polyalthic acid
Trypanothione reductase (TcTR)1BZL−81.9−95.8Copaiferolic acid
UDP-galactose mutase (TcUGM)4DSH−104.5−115.7Copaiferolic acid
Table 6. MolDock docking energies (kJ/mol) of Copaifera diterpenoids with bacterial protein targets.
Table 6. MolDock docking energies (kJ/mol) of Copaifera diterpenoids with bacterial protein targets.
Bacterial Protein TargetsPDBEdock (ave)Edock (min)Best Docking Diterpenoid Ligand
Pseudomonas aeruginosa peptide deformylase (PaPDF)1LRY−96.3−113.5(+)-Hardwickiic acid
Streptococcus pneumoniae peptide deformylase (SpPDF)2AIE−100.2−115.47α-Acetoxyhardwickiic acid
Mycobacterium tuberculosis peptide deformylase (MtPDF)3E3U−95.7−107.3(+)-Hardwickiic acid
Escherichia coli topoisomerase IV (EcTopoIV)1S16−100.5−118.87α-Acetoxyhardwickiic acid
Mycobacterium tuberculosis DNA gyrase B (MtDNAGyrB)3ZKD−101.3−118.33α-Alepterolic acid acetate
Mycobacterium tuberculosis protein tyrosine phosphatase (MtPTPB)2OZ5−89.2−107.2ent-Polyalthic acid
Mycobacterium tuberculosis UDP-galactopyranose mutase (MtUGM)4RPL−92.2−104.419-O-Acetyl-1,2-dehydrokautriwaic acid
Mycobacterium tuberculosis mycocyclosin synthase (MtCYP121)5IBE−87.0−108.219-O-Acetyl-1,2-dehydrokautriwaic acid
Escherichia coli DNA ligase (EcLigA)2OWO−97.8−108.519-O-Acetyl-1,2-dehydrokautriwaic acid
Mycobacterium tuberculosis DNA ligase (MtLigA)1ZAU−89.0−107.219-O-Acetyl-1,2-dehydrokautriwaic acid
Staphylococcus aureus DNA ligase (SaLigA)4CC6−85.2−97.8Methyl copalate
Streptococcus pneumoniae DNA ligase (SpLigA)4GLW−88.1−109.5Cativic acid
Table 7. MolDock docking energies (kJ/mol) of Copaifera diterpenoids with cancer-relevant protein targets.
Table 7. MolDock docking energies (kJ/mol) of Copaifera diterpenoids with cancer-relevant protein targets.
Cancer-Relevant Protein TargetsPDBEdock (ave)Edock (min)Best Docking Ligand
Human androgen receptor (HsAR)5VO4−67.6−103.2Cativic acid
Human aromatase (HsCYP19A1)5JKW−97.1−112.7ent-Pinifolic acid
Human casein kinase II (HsCK2)5N9K−83.3−101.03α-Alepterolic acid acetate
Human cyclin-dependent kinase 2 (HsCDK2)5JQ8−91.1−104.17α-Acetoxyhardwickiic acid
Human cyclin-dependent kinase 4 (HsCDK4)2W96−95.2−117.63α-Alepterolic acid acetate
Human cyclin-dependent kinase 6 (HsCDK6)5L2S−87.3−99.0(+)-Hardwickiic acid
Murine cyclooxygenase 2 (MmCOX-2)6COX−93.7−106.9Kolavenic acid
Human DNA (cytosine-5)-methyltransferase 1 (HsDNMT1)3SWR−102.7−121.8Patagonic acid
Human DNA (cytosine-5)-methyltransferase 3A (HsDNMT3A)2QRV−94.3−113.47α-Hydroxyhardwickiic acid
Human epidermal growth factor receptor (HsEGFR)1XKK−81.2−98.819-O-Acetyl-1,2-dehydrokautriwaic acid
Human estrogen receptor α (HsERα)1X7E−96.5−107.519-O-Acetyl-1,2-dehydrokautriwaic acid
Human estrogen receptor β (HsERβ)1U3S−99.6−120.119-O-Acetyl-1,2-dehydrokautriwaic acid
Human heat shock protein HSP 90-α5J2X−83.8−92.6Cativic acid
Human insulin-like growth factor 1 receptor (HsIGF1R)3LW0−86.1−94.2Copaiferolic acid
Human 5-lipoxygenase (Hs5-LOX)3V99−89.9−106.23α-Alepterolic acid acetate
Human mitogen-activated protein kinase kinase 1 (HsMEK1)3OS3−101.5−113.53β-Alepterolic acid acetate
Murine nuclear factor κ-light-chain-enhancer of activated B cells (MmNF-κB)1VKX−74.2−87.73α-Alepterolic acid acetate
Human p90 ribosomal protein S6 kinase (HsRSK2) C-terminal domain4D9U−75.3−90.73β-Alepterolic acid acetate
Human p90 ribosomal protein S6 kinase (HsRSK2) N-terminal domain4NW6−86.4−99.77α-Acetoxyhardwickiic acid
Murine P-glycoprotein3G60−97.6−116.1Patagonic acid
Human phosphatidylinositol-4,5-bisphosphate 3-kinase (HsPI3K)2A5U−84.5−97.67α-Acetoxyhardwickiic acid
Human topoisomerase I (HsTOPO-I)1NH3−83.7−99.6Copaiferolic acid
Human topoisomerase IIα (HsTOPO-IIα)4FM9−94.2−105.219-O-Acetyl-1,2-dehydrokautriwaic acid
Human topoisomerase IIβ (HsTOPO-IIβ)4J3N−85.6−98.93α-Alepterolic acid acetate
Bovine tubulin (colchicine binding site)1SA1−94.2−103.83α-Alepterolic acid acetate
Bovine tubulin (paclitaxel binding site)1JFF−79.1−88.67α-Acetoxyhardwickiic acid
Bovine tubulin (vinblastine binding site)1Z2B−89.1−101.0(+)-Hardwickiic acid
Human vascular endothelial growth factor receptor (HsVEGFR)4ASE−92.9−105.57-acetylbacchotricuneatin D
Table 8. MolDock docking energies (kJ/mol) of Copaifera diterpenoids with cancer-relevant protein targets.
Table 8. MolDock docking energies (kJ/mol) of Copaifera diterpenoids with cancer-relevant protein targets.
Inflammation-Relevant Protein TargetsPDBEdock (ave)Edock (min)Best Docking Ligand
Murine soluble epoxide hydrolase (MmEPHX2)1CR6−108.3−125.619-O-Acetyl-1,2-dehydrokautriwaic acid
Human soluble epoxide hydrolase (HsEPHX2)4HAI−95.1−104.5ent-Agathic_acid
Human fibroblast collagenase (HsMMP-1)1CGL−96.5−109.019-O-Acetyl-1,2-dehydrokautriwaic acid
Porcine phospholipase A2 (SsPLA2)2B03−100.0−112.4(+)-Hardwickiic acid
Human phospholipase A2 (HsPLA2)1J1A−95.4−109.57α-Acetoxyhardwickiic acid
Human 5-lipoxygenase (Hs5-LOX)3V99−89.9−106.23α-Alepterolic acid acetate
Murine inducible nitric oxide synthase (MmiNOS)1M8D−87.5−110.4Copaiferolic acid
Human phosphatidylinositol-4,5-bisphosphate 3-kinase γ (HsPI3Kγ)2A5U−84.5−97.67α-Acetoxyhardwickiic acid
Human interleukin-1 receptor-associated kinase 4 (HsIRAK4)5T1S−90.7−100.67α-Acetoxyhardwickiic acid
Human glutathione S-transferase ω-1 (HsGSTO1)5V3Q−82.7−94.219-O-Acetyl-1,2-dehydrokautriwaic acid
Ovine cyclooxygenase-1 (OaCOX-1)3N8Z−62.1−92.0Crolechinic acid
Murine cyclooxygenase-2 (MmCOX-2)6COX−93.7−106.9Kolavenic acid
Human c-Jun N-terminal kinase (HsJNK)4Y46−83.7−97.719-O-Acetyl-1,2-dehydrokautriwaic acid
Xenopus laevis inhibitor of κB kinase β (XlIKKB)3RZF−88.1−100.9Patagonic acid
Human NF-κB essential modulator (HsNEMO)3BRT−85.9−105.33α-Alepterolic acid acetate
Human lipid binding protein MD-2 (HsMD-2)2E59−71.2−84.319-O-Acetyl-1,2-dehydrokautriwaic acid
Human myeloperoxidase (HsMPO)4C1M−84.3−98.8(+)-Hardwickiic acid
Murine nuclear factor κ-light-chain-enhancer of activated B cells (MmNF-κB)3DO7−72.7−85.9(+)-Hardwickiic acid
Human p38 mitogen-activated protein kinase (Hsp38MAPK)1OZ1−91.3−116.419-O-Acetyl-1,2-dehydrokautriwaic acid
Human peroxisome proliferator-activated receptor γ (HsPPARγ)3ADV−91.9−107.6Copaiferolic acid
Human cAMP-specific 3′,5′-cyclic phosphodiesterase 4D (HsPDE4D)5K32−90.2−107.37α-Acetoxyhardwickiic acid

Share and Cite

MDPI and ACS Style

Da Trindade, R.; Da Silva, J.K.; Setzer, W.N. Copaifera of the Neotropics: A Review of the Phytochemistry and Pharmacology. Int. J. Mol. Sci. 2018, 19, 1511. https://doi.org/10.3390/ijms19051511

AMA Style

Da Trindade R, Da Silva JK, Setzer WN. Copaifera of the Neotropics: A Review of the Phytochemistry and Pharmacology. International Journal of Molecular Sciences. 2018; 19(5):1511. https://doi.org/10.3390/ijms19051511

Chicago/Turabian Style

Da Trindade, Rafaela, Joyce Kelly Da Silva, and William N. Setzer. 2018. "Copaifera of the Neotropics: A Review of the Phytochemistry and Pharmacology" International Journal of Molecular Sciences 19, no. 5: 1511. https://doi.org/10.3390/ijms19051511

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop