Next Article in Journal
Methotrexate Gold Nanocarriers: Loading and Release Study: Its Activity in Colon and Lung Cancer Cells
Next Article in Special Issue
Bioinformatic Analysis of Genome-Predicted Bat Cathelicidins
Previous Article in Journal
FT-IR/ATR Solid Film Formation: Qualitative and Quantitative Analysis of a Piperacillin-Tazobactam Formulation
Previous Article in Special Issue
Withanolide-Type Steroids from Withania aristata as Potential Anti-Leukemic Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Phytochemicals with Added Value from Morella and Myrica Species

by
Gonçalo P. Rosa
1,2,
Bruno J. C. Silva
3,
Ana M. L. Seca
1,2,3,
Laila M. Moujir
4 and
Maria Carmo Barreto
2,3,*
1
LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
2
cE3c—Centre for Ecology, Evolution and Environmental Changes/Azorean Biodiversity Group, Rua Mãe de Deus, 9501-321 Ponta Delgada, Portugal
3
Faculty of Sciences and Technology, University of Azores, Rua Mãe de Deus, 9501-321 Ponta Delgada, Portugal
4
Departamento de Bioquímica, Microbiología, Biología Celular y Genética, Facultad de Farmacia, Universidad de La Laguna, Avda Astrofísico Francisco Sánchez, s/n., 38200 La Laguna, Spain
*
Author to whom correspondence should be addressed.
Molecules 2020, 25(24), 6052; https://doi.org/10.3390/molecules25246052
Submission received: 27 November 2020 / Revised: 14 December 2020 / Accepted: 17 December 2020 / Published: 21 December 2020

Abstract

:
Terrestrial plants, due to their sessile nature, are highly exposed to environmental pressure and therefore need to produce very effective molecules that enable them to survive all the threats. Myrica and Morella (Myricaceae) are taxonomically close genera, which include species of trees or shrubs with edible fruits that exhibit relevant uses in traditional medicine. For instance, in Chinese or Japanese folk medicine, they are used to treat diarrhea, digestive problems, headache, burns, and skin diseases. A wide array of compounds isolated from different parts of Myrica and/or Morella species possess several biological activities, like anticancer, antidiabetic, anti-obesity, and cardio-/neuro-/hepatoprotective activities, both in vitro and in vivo, with myricanol, myricitrin, quercitrin, and betulin being the most promising. There are still many other compounds isolated from both genera whose biological activities have not been evaluated, which represents an excellent opportunity to discover new applications for those compounds and valorize Morella/Myrica species.

Graphical Abstract

1. Introduction

Nature is an important source of new biologically active compounds and molecules with very diverse and unique biological properties and chemical structures, several of them being commercialized as medicines [1]. This diversity is the result of a long and selective evolutionary pressure [2], that led to an array of different biosynthetic pathways producing primary and, in particular, secondary metabolites with a large variety of basic skeletons and functional groups, which have been huge contributors to the improvement of human health [3].
That evolutionary pressure is more intense in plants and other sessile organisms because, since they are unable to move, they are more exposed to the action of herbivores, pathogens. and/or variable sunlight conditions, and therefore need to produce very effective molecules that enable them to survive all of these threats [4]. That fact, allied to the extraordinary biological and chemical diversity within plants, offers a particularly rich potential in biologically active compounds that can be used to provide lead compounds for the production of medications for treating various diseases from migraine to cancer. Twenty-five percent of all prescribed drugs are derived from plants, and of the 252 drugs considered as basic and essential by the World Health Organization, 11% are exclusively of plant origin and a significant number are drugs obtained from natural precursors by semi-synthesis. Thus, medicinal plants and their derived natural compounds are still an increasing topic of investigation and interest [5,6].
The members of the Myrica and Morella genera are woody shrubs or tree pioneers in nitrogen-poor soils such as sandy soil or gravelly sites, because they are actinorhizal plants able to fix nitrogen through nitrogen-fixing root nodules induced by soil actinomycetes of the genus Frankia, with which they establish a symbiotic relationship [7]. In addition to the economic interest of these species as sources of paper and rope from the bark, as fuel wood, for biomass production, and land reclamation, they are also appreciated because their fruits that can be eaten raw and are used in the production of jams, syrups, and juices [8], and their applications in traditional medicine are also noteworthy.
The Myrica genus comprised, before 2002, circa 97 species with a wide distribution in both temperate and sub-tropical regions [8,9]. Macdonald [10] presented various reasons for splitting this genus in two, Myrica and Morella, with his arguments only being accepted in 2002 [11]. A taxonomic key was published to allow the simple discrimination between species of the two genera [12]. The splitting of the Myrica genus led to many of the species previously belonging to the Myrica genus being reclassified and included in the Morella genus, which can cause problems when trying to correlate the discovery of secondary metabolites with the plant of origin. Many studies published before 2005 report the isolation of secondary metabolites from Myrica species which are now classified as Morella species, causing misleading reports on secondary metabolites found for the first time in the genus. Another issue is the fact that more recent publications use the previous scientific name, that consequently can lead to a compound not being properly detected in a literature survey. To prevent this from happening, the present work covers the secondary metabolites isolated from both genera and their respective biological activities. The botanical names of the species used in the present work are the ones currently accepted by the International Plant Names Index (IPNI) database, even if in the original publication the former name was used. In this case, the former name will appear in brackets.

2. Biological Activities Exhibited by Secondary Metabolites from Morella and Myrica Species

Several of the secondary metabolites isolated from Morella and Myrica species have been studied to evaluate their potential application in human health. Silva et al. [13] reviewed compounds isolated from Morella and Myrica species exhibiting antioxidant and anti-inflammatory activities, reporting the antioxidant potential of various compounds as well as pertinent structure/activity relationships that could direct future research in order to obtain new, more active, and safer molecules. Therefore, this section is focused on other biological activities exhibited by compounds isolated from species of the Morella and Myrica genera. There are two bioactive compounds, myricetin and arjunolic acid, isolated, among others, from Myrica esculenta Buch.-Ham. Ex D. Don [14], whose biological activities have already been revised by Gupta et al. [15] and Gosh et al. [16], ranging from antidiabetic to antibacterial, anticancer, or anti-inflammatory. For that reason, those compounds will not be addressed in this work.

2.1. In Vitro Activities

In vitro studies represent the first step in the evaluation of the pharmacological effects of compounds. They are a simpler and cheaper way to assess the bioactivities of the tested compounds and yield very important and relevant information to direct further investigations on the full pharmacological potential of a compound. The results of in vitro tests performed with compounds isolated from Morella and Myrica are summarized in Table 1, where the compounds are organized by family.
As seen in Table 1, bioactive compounds are found in every part of the plant, from roots to the leaves, and the same compound can be found in members of both genera, which shows a close chemotaxonomic relationship, resulting from the taxonomic reorganization of the previous genus Myrica [12].
A variety of compounds are present, including diarylheptanoids, dihydrochalcones, triterpenoids and flavonoids, but the majority of the bioactive compounds obtained from Morella and Myrica genera are cyclic diarylheptanoids, a class of compounds known by their wide array of biological activities [63]. From the analysis of Table 1 it is also clear that the compounds which have been more widely studied are myricanone (1), myricanol (4), quercitrin (9), myricitrin (10), and betulin (16), so details about the most interesting works and compounds from Table 1 will be discussed in the following paragraphs.
Myricanone (1) presents cytotoxic activity against A549 and HepG2 cell-lines with EC50 of 3.22 µg/mL [25], and 32.46 µM [27], respectively. Myricanone (1) increases the apoptotic rate of A549 tumor cells from 4.13% in untreated cells to 34.9% (after treatment) in a dose-dependent manner, and significantly inhibits colony formation in A-549, by inducing cell-cycle arrest in G1 phase [25]. Although the authors indicate that fluorouracil was used as positive control, and that myricanone (1) is more active than the positive control [25], the EC50 of fluorouracil is not presented, which hinders a more realistic assessment of the potency of myricanone effects against A549 cells.
Myricanol (4) showed cytotoxic activity against HL60 (leukemia), A549 (lung), and SK-BR-3 (breast) cell-lines, with IC50 similar or lower (Table 1) than the positive control cisplatin [18]. The author also demonstrated that induced apoptosis cell via mitochondrial (caspase 3, 8, and 9 were activated) and death receptor pathway in HL-60 cell, suggesting its potential anticancer activity. Similar results were obtained by Dai et al. [33] against A-549 cells.
Myricanol (4) also presents a very interesting activity by inhibiting muscle atrophy and dysfunction caused by dexamethasone in C2C12 myotubes [33]. As seen in Table 1, pretreatment with 4 induces a variety of processes that lead to or are a reflex of the direct activation of Sirtuin-1 [30], a protein that regulates skeletal muscle remodeling, which directly deacetylates and therefore activates PGC-1α to regulate mitochondrial biogenesis [64], induces autophagy to enhance degraded protein clearance and modulates FoxOs transcriptional activity to reduce atrogin-1 and MuRF1 expression, which are responsible for protein degradation, and consequently control loss of muscle mass and function [65]. Since there are only a few molecules approved for the treatment of muscle atrophy, and the majority come with very undesirable side effects like insulin resistance, sodium retention, or possibility of thromboembolisms [66], there is a need to find alternative molecules capable of treating this condition. The results shown by myricanol (4) turn it into a good candidate for that alternative.
Additionally, myricanol shows neuroprotective effects in N2a neuronal cells exposed to H2O2 [32]. Pretreatment with 0.84 mM of 4 increased 80% of cell viability, decreased 40% of intracellular reactive oxygen species (ROS) formation and reduced intracellular calcium concentrations, compared with the H2O2 group.
Quercitrin (9) is one of the most studied among the compounds from Morella and Myrica genera. Despite most of the studies being performed with quercitrin isolated from other sources, the fact that this compound has also been isolated in Morella and Myrica genera, namely Morella adenophora (Hance) J. Herb. [19], is in itself an added value to the biological importance of these genera. Compound 9 is a potent α-glucosidase inhibitor, an enzymatic target used to evaluate antidiabetic effect. It has an IC50 four times lower than acarbose, a commercial inhibitor, which indicates the great potential of quercitrin as a hypoglycemic agent for diabetes treatment [36]. Mercury and lead are considered as threats to human organisms, due to their low elimination rate and high accumulation, which have nefarious effects in several physiological functions, especially of liver and nervous system [67,68]. A study carried out by Aldana et al. [39] demonstrated the capacity of quercitrin (9) as cytoprotective agent for the HepG2 cell line against both metals. However, on PC12 cells, quercitrin was only able to protect against the effects of lead [39]. Further investigations are necessary to explain the cytoprotective mechanism of the compound.
Quercitrin (9) also presents a moderate antiviral activity by inhibition of neuraminidase (NA) [37], a viral enzyme common in human influenza viruses, which is essential for the virus release from infected cells to the neighboring cells of the respiratory tract [69]. It has also been reported that quercitrin (9) inhibits the activity of myeloperoxidase, with an IC50 of 2.0 µM [38], a result that hints at its possible role in reducing endothelial dysfunction and consequently that the protective effect of 9 should be evaluated in an atherosclerosis model.
A great number of the studies reviewed about the pharmacological activities of myricitrin (10) focused on its effects against cardiovascular problems [43,44,45]. Qin et al. [45] suggest that myricitrin treatment protected human umbilical vein endothelial cells (HUVECs) against the effects of oxidized low-density lipoprotein (ox-LDL), which results in reduced atherosclerotic plaque formation.
It was also found that 40 µM myricitrin protects H9c2 cardiomyocytes against hypoxia/reoxygenation injury, increasing cell-survival from 65.48% in untreated cells to 87.48% (Table 1) [43]. In addition, there was a reduction in the expression of pro-apoptotic factors, like caspase-3 or Bax, and an increase in the expression of anti-apoptotic factors, like Bcl-2. Likewise, the authors formulated that myricitrin (10) might exert its cardioprotective effects via stimulation of the expression of heat shock protein 90, whose activation promotes the anti-apoptotic PI3-K/Akt signaling pathway [43].
In a model of diabetic cardiomyopathy injury, Zhang et al. [44] found that pretreatment with 53.8 µM of myricitrin significantly decreased advanced glycation end products (AGEs)-induced inflammatory cytokine expression, limited an increase in ROS levels, and reduced cell apoptosis, fibrosis, and hypertrophy in H9c2 cells (Table 1). Overall, the protective effects of myricitrin could be attributed to its antioxidant and anti-inflammatory effects, as well as its ability to activate the Akt signaling pathway [43,44]. The anti-ROS activity of 10 is also responsible for the neuroprotective activities reported by Shen et al. [17] and Wang et al. [47] as observable by the effects described in Table 1.
Myricitrin (10) is also a potent α-glucosidase (IC50 = 1.1 µM) and β-amylase inhibitor (IC50 = 1.9 µM), being 20- and 10-fold more active than acarbose, respectively [48].
Myricanone (1), myricanol (4), and myricitrin (10) were found to inhibit melanogenesis in B16 mouse melanoma cells (Table 1) [26]. The melanin content found after exposure to the compounds was lower than the positive control, arbutin. However, the mechanism by which this occurred was different. For myricanone (1) and myricanol (4), it was due to their cytotoxic effects on the B16 cells, while 10 exerted its antimelanogenesis effect without killing the B16 cells, similar to the effect of arbutin [26].
Respiration uncoupling seems to be implicated in numerous physiological and pathological processes like autophagy, regulation of ROS production, protein secretion, capacity to carry out physical exercise, and adipose tissue biology [70]. As seen in Table 1, a series of dihydrochalcones, which have been isolated from various species of Morella and Myrica, possess the ability to uncouple the mitochondrial respiratory chain. Myrigalone A (11) and myrigalone G (15) were the best uncouplers, increasing the respiratory rate more than two-fold with respect to the positive control, 2,4-dinitrophenol (DNP) [53]. Although respiratory uncouplers may have some therapeutic interest, they must be evaluated for their safety, and there are no records of such study on the safety of myrigalones 1115 as respiratory chain uncoupling agents.
Myrigalone B (12) and myrigalone G (15) inhibited α-glucosidase activity with IC50 of 19 and 7 µM, respectively, which is lower than the positive control, acarbose (43 µM). For β-amylase inhibition, 12 was again the most active with an IC50 of 8.3 µM against 33 µM obtained with compound 15 (Table 1) [48]. In this case myrigalone B (12) was still more active than acarbose, but not myrigalone G (15), since the IC50 of acarbose for β-amylase inhibition was 19 µM. Despite their potential as antidiabetic compounds, 12 and 15 are still less active inhibitors than myricitrin (10).
Another very interesting compound found in Morella/Myrica genera is betulin (16), which has presented a wide array of antitumor activities (Table 1). Lin et al. [57] reported the anticancer activity against two lines of osteosarcoma, MG-63 and HOS cell lines, with an IC50 of 14.54 and 11.70 µM, respectively. Unfortunately, no positive control was used to compare with the potency of botulin (16). The authors found that the effect of 16 in osteosarcoma cell lines is through the induction of apoptosis and autophagy to suppress cell viability, as well as by the inhibition of mTOR signaling. Betulin (16) is also very active against the NCi-H460 (IC50 = 2.8 µM) and HT29-MTX (IC50 = 1.6 µM) cell lines, being much more active than the positive control podophyllotoxin (22 and 24 µM against these two cell lines, respectively) [58]. Compound 16 was also found to be active against A549 and SK-Br-3 cell lines, with a more potent effect than cisplatin (Table 1), but is less potent against the HL60 cell-line [18].
Betulin (16) also exerts hepatoprotective activity against continuous ethanol exposure [59]. Ethanol-induced HSC-T6 cells treated with 25 µM of 16 suffered a decrease in the expression levels of collagen-I and α-SMA, which indicates that betulin (16) reduces ethanol-induced hepatic fibrosis. The expression levels of TLR4 were decreased after betulin pretreatment and there was an increased activation of STAT3. This modulation of TLR4 and STAT3 pathways is proposed as the mechanism through which 16 protects the hepatocytes against fibrosis.
A good number of the compounds from Table 1 were tested for their anti-tuberculosis activity against Mycobacterium tuberculosis H37Rv, (+)-Galeon (8) being the most active compound with a minimum inhibitory concentration (MIC) of 15 µg/mL. However, this value was still higher than the one obtained with the positive control, ethambutol, which has an MIC of 6.25 µg/mL [19]. The second-best compound, 5-deoxymyricanone (2), has an MIC of 25.8 µg/mL, which is more than four times the MIC of the positive control, so it cannot be considered very active.
Morella/Myrica compounds show remarkable pharmacological potential, with a variety of interesting in vitro activities. For further understanding of their full potential and clarification of mechanisms of action, their activities must be confirmed in vivo. The following section focuses on what is already known about the effects of compounds isolated from Morella/Myrica in living organisms.

2.2. In Vivo Tests

In vivo studies are necessary to understand the actual potential of compounds as future therapeutic agents. A few compounds isolated from Morella/Myrica genera reached the in vivo stage of evaluation of their pharmacological effects, which means that research teams recognize their potential and want to prove their full pharmacological value. The main results of those studies are summarized in Table 2, and the most relevant aspects are discussed below.
As expected, the three compounds which have been more extensively studied in vitro and which were reported to be bioactive were the ones selected by researchers to be evaluated concerning their in vivo activity (Table 2). These compounds are myricanol (4), myricitrin (10), and betulin (16), and many of the in vivo studies found in the present literature survey aimed to confirm or to clarify the mechanisms of action found on the in vitro studies.
A dosage of 5 mg/kg of myricanol (4) administered to C57BL/6 mice with dexamethasone-induced muscle wasting led to a reduction of muscle loss in both quadriceps and gastrocnemius muscle (Table 2). In addition, the mice from the treated group showed an improvement of grip strength of about 50 g and almost doubled the forced swim time when compared with the untreated group [30]. Muscle atrophy was inhibited, with muscle fiber diameter increasing almost by 25% when compared with the untreated group. As observed in the in vitro tests, myricanol (4) also prevents dexamethasone-induced muscle atrophy and weakness by activating SIRT1 to reduce muscle protein degradation, enhance autophagy, and promote mitochondrial biogenesis and function in mice [30].
In another study, myricanol (4) at a dosage of 25 mg/kg, when fed to C57BL/6J mice simultaneously with a high fat diet (HFD), was able to reduce body weight and body fat accumulation, when compared with mice fed only with the HFD [71]. Furthermore, compound 4 administration led to a 33% decrease in lipids serum levels and a 50% reduction of fasting insulin levels, which can be explained by the 2-fold increase observed in the insulin sensitivity on the treated group (Table 2). Other effects of 4 in the HFD-fed mice were the activation of a series of pathways, like adenosine monophosphate-activated protein kinase (AMPK), leading to the suppression of adipogenesis and induction of lipolysis and lipid combustion in adipocytes, or insulin receptor substrate 1 (IRS-1), which leads to mitochondrial biogenesis, increases mitochondrial oxidative metabolism, and benefits adenosine triphosphate (ATP) synthesis. Myricanol (4) also increases irisin serum levels, which induces mitochondrial oxidative metabolism, mitochondrial uncoupling, fatty acid oxidation in skeletal muscle [76], and increases energy expenditure in human adipocytes, resulting in reduced lipid accumulation [77]. Altogether, these data show this compound could be developed as a candidate for the treatment of insulin resistance and obesity. The effect of myricanol on lipid accumulation was confirmed by Shen et al. [31], but in a zebrafish model, with their findings showing the inhibition of lipid accumulation by suppressing adipogenic factors, including peroxisome proliferator activated receptor γ (PPARγ) and CCAAT/enhancer binding protein α (C/EBPα).
Myricanol (4) also decreased tumor growth in xenografted BALB/c nude mice at a dosage of 40 mg/kg, by activating the expression levels of pro-apoptotic factors within the tumor cells and downregulating the protein expression of Bcl-2, VEGF, HIF-1α, and survivin, thus increasing the apoptotic rate of tumor cells [72]. These effects led to a reduction in tumor volume of 39.4% after 14 days when compared with the untreated mice, showing the great potential of myricanol to become an anticancer drug.
In a murine model of atherosclerosis, a dosage of 50 mg/kg of myricitrin prevented the formation of Ox-LDL, which represented a reduction of 25% in serum levels of that atherosclerosis promoter [45]. A similar reduction was found in the mice treated with the positive control, probucol, but at a 40-fold higher dose than that of myricitrin, which means the latter is much more effective. By reducing the levels of Ox-LDL, myricitrin (10) led to a reduction of 20% in the expression of caspase-3 in the aortic arch, thus reducing endothelial cell apoptosis. This resulted in a complete inhibition of calcification in the aortic arch of treated mice, and a reduction on the atherosclerotic plaque area (Table 2) [45].
The in vivo investigation of the effects of myricitrin in a model of diabetic cardiomyopathy demonstrated that oral administration of a dosage of 300 mg/kg/day for 8 weeks remarkably decreased the expression of enzymes associated with cardiomyopathy, as well as the expression of inflammatory cytokines and apoptotic proteins [44]. This led to 20–25% improvement of diastolic dysfunction and attenuated histological abnormalities, by inhibiting the destruction of the collagen network, which reduces the fibrotic alterations of the heart. Mechanistically, the authors found that myricitrin (10) attenuated diabetes-induced Nrf2 inhibition via the regulation of Akt and extracellular-signal-related kinase (ERK) phosphorylation in the diabetic heart.
Another in vivo study showed that a dosage of 50 mg/kg of myricitrin (10) improved neuron injury and increased the expressions levels of PSD-95 protein and tyrosine hydroxylase TH protein in lipopolysaccharide (LPS)-stimulated BALB/c mice [74]. Loss of these proteins is found in Parkinson’s disease [78], so the fact that myricitrin increases their expression hints for a possible role in the prevention of this disease. In addition, myricitrin (10) decreased the production of pro-inflammatory factors including IL-1β, IL-6, and TNFα, decreased the level of chemokine MCP-1, and suppressed the expressions of COX-2 and iNOS (Table 2). Meanwhile, myricitrin suppressed HMGB1, TLR4, and MyD88 expression in the nigrostriatum of LPS-stimulated mice, and inhibited NF-κB and mitogen-activated protein kinase (MAPK) signaling pathways activated by LPS [74]. These effects show that myricitrin (10) is efficient in protecting the brain against inflammation and all the nefarious effects it causes.
Myricitrin (10) at a dosage of 100 mg/kg significantly ameliorated CCl4-induced hepatotoxicity in BALB/cN mice [73]. Treatment with this compound caused an increase in serum aspartate transaminase (AST) and alanine transaminase (ALT) levels and prevented histopathological changes in the liver (Table 2). Hepatic oxidative stress was reduced by myricitrin (10), as evidenced by the decrease in lipid peroxidation, with concomitant increase in glutathione (GSH) level and cytochrome P450 2E1 (CYP2E1) expression. Furthermore, cyclooxygenase-2 (COX-2) and tumor necrosis factor-alpha (TNF-a) overexpression in the liver was reduced, suggesting the suppression of inflammation [73]. Myricitrin (10) also improved the regeneration of hepatic tissue after CCl4-intoxication, as evidenced by increased proliferating cell nuclear antigen (PCNA) expression. The results presented by Domitrovic et al. [73] suggest that the anti-inflammatory and antioxidant effects of myricitrin (10) are responsible for its hepatoprotective activity. In fact, all the myricitrin (10) in vivo studies presented in Table 2 strongly indicate that 10 exerts its protective effects through the blockage of inflammation, oxidative stress, and apoptosis [44,45,73,74], which confirms myricitrin (10) as a potential candidate for the treatment and prevention of a wide array of disorders related to oxidative stress and inflammation.
Betulin (16) in vivo studies focused on its hepatoprotective effects against both chronic alcohol consumption [75] or acute ethanol induced fatty liver [47]. In the model of chronic consumption, administration of 20 mg/kg of betulin (16) to alcoholic C57BL/6 mice attenuated the increases in serum aminotransferase and triglyceride levels, while significantly inhibiting SREBP-1 expression and activating LKB1-AMPK phosphorylation [75]. Additionally, betulin (16) enhanced the sirtuin 1 (SIRT1) expression mediated by ethanol. Taken together, betulin alleviates alcoholic liver injury possibly through blocking the regulation of SREBP-1 on fatty acid synthesis and activating SIRT1-LKB1-AMPK signaling pathway.
In the acute ethanol-induced fatty liver model, administration of 50 mg/kg of betulin (16) to C57BL/6 mice induced a decrease in serum levels of hepatic enzymes and triglycerides [47]. The expression of SREBP-1, a transcription factor that promotes fatty acid synthesis [79] and whose activation by ethanol leads to fat accumulation on the liver, is reduced by betulin (16) to 75% of the expression levels presented by the untreated groups [47]. Betulin (16) administration also significantly decreased the expression levels of CYP2E1 and TLR4 and increased the activation of STAT3 (Table 2), thus impairing the ethanol induced pro-inflammatory response and, consequently reducing liver steatosis and fibrosis [47].

3. Other Phytochemicals Identified in Morella and Myrica Species

Many other secondary metabolites whose therapeutic application was not evaluated yet have been isolated in the last decades from Morella and Myrica species, constituting a pool of natural compounds structurally diverse with scientific interest, in which a research gap exists and which it is important to evaluate. These compounds are compiled in Table 3.
The pharmacological potential of the compounds presented in Table 3 has not been evaluated so far by the researchers who have studied Morella and Myrica species. It should be noted that the isolation and structural elucidation are the first steps to describing the metabolomic profile of a species or genus, helping to assess their potential as a source of commercially valuable phytochemicals, adding value to the species, as well as finding alternative sources of pharmacologically active metabolites.
The compounds listed in Table 3 belong to several families, like chalcones, dihydrochalcones, flavonoids, diterpenoids, triterpenoids, and diarylheptanoids, the latter two being the families with the highest number of compounds isolated. A similar tendency had already been observed in Table 1, where it was observed that diarylheptanoids were the most represented family, which indicates that Morella and Myrica genera are a good source of this family of compounds.
Since many of the unstudied compounds compiled in Table 3 share many structural features with some of the most active compounds that have already been assessed for their biological activities, there is plenty of potential for finding new added value compounds to increase the value of species from Morella/Myrica genera.
As an example, the only structural difference between myricanol (4) and myricananin B (39), two compounds isolated from the roots of Morella nana (A. Chev.) J. Herb. [21,28], is that the latter has an additional OH group at C-12. However, the biological activities of myricanol are widely studied, as reviewed in the present work, whereas the activities of myricananin B are not, which is unfortunate, since such structural similarity indicates a good pharmacological potential for myricananin B. A possible explanation is the fact that myricananin B (39) is obtained in low yields. In fact, starting with 20 kg of dried Morella nana (A. Chev.) J. Herb., Wang et al. [28] only obtained 5 mg of myricananin B, which according to the authors, represents a yield of 0.0000025%. With such a reduced amount of compound, it is understandable that myricananin B is so understudied, but the similarities with the bioactive myricanol point to a high pharmacological potential of compound 39, so strategies to obtain higher amounts of this compound, like total or semi-synthesis should be employed.
Another example is the group of unstudied pentacyclic terpenes 5770. A compound from the same family, ursolic acid, was proven by in vivo and preclinical studies to possess multiple pharmacological activities, like anti-inflammatory, antitumor, cardioprotective, and antidiabetic, among others [85]. However, the poor water solubility and low intestinal absorption of ursolic acid, which leads to a rapid elimination by the gut wall/liver metabolism, resulting in low bioavailability [86,87], hinder the clinical applicability of ursolic acid. Some of the pentacyclic terpenes listed in Table 3, like arjunglucoside (68) and 3-O-(E)-caffeoyl-ursolic acid (70), present structural aspects that might overcome some of the limitations of the clinical use of ursolic acid, namely by being less lipophilic, which could increase their intestinal absorption and improve bioavailability.
These examples show that there are still many research opportunities in the study of Morella/Myrica and point out a great potential to increase the value of species belonging to these genera.

4. Conclusions

Compounds isolated from Morella and Myrica genera show great potential to become important pharmacological agents. Both in vitro and in vivo studies showed that compounds isolated from these genera, like myricanol (4), myricitrin (10), and betulin (16), are already seen as important candidates for the treatment of several diseases, namely due to their antitumor and cardio-/neuro-/hepatoprotective activities. Interestingly, in most cases their activity is related to their capacity to modulate oxidative stress and inflammation pathways, which increases their value even further, because of the myriad of pathologies related with those processes.
Although many of the studies presented their results with quality, there were some works where there was a lack of important information for a more correct interpretation of the validity of results. In some cases, authors present the IC50 without indicating the associated error, in others a positive control is not used, which hinders the perception of the true potency of the results. Future works should take these factors into consideration.
There are still many compounds isolated from Morella and Myrica genera whose biological activities have not been evaluated. However, since these compounds belong to chemical families with a high number of reported biological activities, a great potential for the chemical valorization of Morella/Myrica species can be foreseen, opening the door for future studies.

Author Contributions

A.M.L.S., L.M.M., and M.C.B. conceptualized and revised the paper; G.P.R. and B.J.C.S. conducted the research and wrote the first draft. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by FCT—Fundação para a Ciência e a Tecnologia, supporting G.P.R.’s grant (SFRH/BD/144446/2019) as well by FCT—Fundação para a Ciência e Tecnologia, the European Union, QREN, FEDER, and COMPETE, through funding the cE3c center (UIDB/00329/2020) and the LAQV-REQUIMTE (UIDB/50006/2020). Funded by RTI2018-094356-B-C21 Spanish MINECO project, co-funded by European Regional Development Fund (FEDER).

Acknowledgments

Thanks are due to the University of Azores and Universidad de La Laguna.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

6-OHDA
ACE-1
6-hydroxydopamine
Angiotensin-converting-enzyme 1
AGEAdvanced glycation end products
AICAR5-Aminoimidazole-4-carboxamide ribonucleotide
AktProtein kinase B
ALTAlanine transaminase
AMPK
aP2
Adenosine monophosphate-activated protein kinase
Adipocyte protein 2
ASTAspartate transaminase
ATPAdenosine tri-phosphate
α-SMAalpha smooth muscle actin
BaxBcl-2-associated X
Bcl-2B-cell lymphoma 2
C/EBPαCCAAT/enhancer-binding-protein-α
COX-2Cycloxygenase-2
CYP2E1Cytochrome P450 2E1
DNP2,4-dinitrophenol
EC50Half maximal effective concentration
ERKExtracellular-signal-regulated kinase
FoxOsForkhead box O3
γ-GCSGamma-glutamylcysteine synthetase
GSK-3βGlycogen synthase kinase 3 beta
HDLHigh-density lipoprotein
HFDHigh-fat diet
HIF-1αHypoxia-inducible factor 1-alpha
HMGB1High mobility group box 1 protein
HO-1Heme oxygenase 1
HUVECsHuman umbilical vein endothelial cells
IC50Half maximal inhibitory concentration
IL-1βInterleukin-1β
IL-6Interleukin-6
iNOSNitric oxide synthase
IRS-1Insulin receptor substrate 1
IKK-βI-kappa-B-kinase beta
JNKc-Jun NH2-terminal cinase
LDHLactate Dehydrogenase
LDLLow-density lipoprotein
LKB1Tumor suppressor serine/threonine-protein kinase
LPSLipopolysaccharide
MAPKMitogen-activated protein kinase
MCP-1Monocyte chemoattractant protein-1
MICMinimum inhibitory concentration
mTORmammalian target of rapamycin
MuRF1Muscle RING-finger protein-1
MyD88Myeloid differentiation primary response 88
NANeuraminidase
NF-KBNuclear factor kappa B
NQO-1NAD(P)H Quinone Dehydrogenase 1
Nrf2Nuclear factor erythroid 2-related factor 2
Ox-LDLOxidized low-density lipoprotein
ROSReactive oxygen species
PGC-1αPeroxisome proliferator-activated receptor-gamma coactivator
PI3-KPhosphoinositide 3-kinases
PPARγPeroxisome proliferator-activated receptor gamma
PSD-45Postsynaptic density protein-45
SIRT1Sirtuin 1
SREB-1Sterol regulatory element-binding transcription factor 1
STAT3Signal transducer and activator of transcription 3
TGF-β1Transforming growth factor beta 1
THTyrosine hydroxylase
TGTriglycerides
TLR4Toll-like receptor 4
TNF-αTumor necrosis factor α
VEGFVascular endothelial growth factor

References

  1. Seca, A.M.L.; Moujir, L.M. Natural compounds: A dynamic field of applications. Appl. Sci. 2020, 10, 4025. [Google Scholar] [CrossRef]
  2. Kroymann, J. Natural diversity and adaptation in plant secondary metabolism. Curr. Opin. Plant Biol. 2011, 14, 246–251. [Google Scholar] [CrossRef] [PubMed]
  3. Thirumurugan, D.; Cholarajan, A.; Raja, S.S.S.; Vijayakumar, R. An introductory chapter: Secondary metabolites. In Secondary Metabolites—Sources and Applications; Vijayakumar, R., Raja, S.S.S., Eds.; IntechOpen: London, UK, 2018; pp. 13–21. [Google Scholar] [CrossRef] [Green Version]
  4. Austen, N.; Walker, H.J.; Lake, J.A.; Phoenix, G.K.; Cameron, D.D. The regulation of plant secondary metabolism in response to abiotic stress: Interactions between heat shock and elevated CO2. Front. Plant Sci. 2019, 10, 1463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. World Health Organization. WHO Global Report on Traditional and Complementary Medicine 2019; WHO Press: Geneva, Switzerland, 2019; Available online: https://www.who.int/traditional-complementary-integrativemedicine/WhoGlobalReportOnTraditionalAndComplementaryMedicine2019.pdf?ua=1 (accessed on 1 September 2020).
  6. Atanasov, A.G.; Waltenberger, B.; Pferschy-Wenzig, E.-M.; Linder, T.; Wawrosch, C.; Uhrin, P.; Temml, V.; Wang, L.; Schwaiger, S.; Heiss, E.H.; et al. Discovery and resupply of pharmacologically active plant-derived natural products: A review. Biotechnol. Adv. 2015, 33, 1582–1614. [Google Scholar] [CrossRef] [Green Version]
  7. Gtari, M.; Dawson, J.O. An overview of actinorhizal plants in Africa. Funct. Plant Biol. 2011, 38, 653–661. [Google Scholar] [CrossRef]
  8. Yanthan, M.; Misra, A.K. Molecular approach to the classification of medicinally important actinorhizal genus Myrica. Indian J. Biotechnol. 2013, 12, 133–136. [Google Scholar]
  9. Huguet, V.; Gouy, M.; Normand, P.; Zimpfer, J.F.; Fernandez, M.P. Molecular phylogeny of Myricaceae: A reexamination of host-symbiont specificity. Mol. Phylogen. Evol. 2005, 34, 557–568. [Google Scholar] [CrossRef]
  10. Macdonald, A.D. The morphology and relationships of the Myricaceae. In Evolution, Systematics, and Fossil History of the Hamamelidae; Crane, P.R., Blackmore, S., Eds.; Oxford University Press: Oxford, UK, 1989; Volume 2, pp. 147–165. [Google Scholar]
  11. Staples, G.W.; Imada, C.T.; Herbst, D.R. New Hawaiian plant records for 2000. Bish. Mus. Occas. Pap. 2002, 68, 3–18. [Google Scholar]
  12. Herbert, J. Systematic and Biogeography of Myricaceae. Ph.D. Thesis, University of St Andrews, St Andrews, UK, 2005. [Google Scholar]
  13. Silva, B.J.C.; Seca, A.M.L.; Barreto, M.D.C.; Pinto, D.C.G.A. Recent breakthroughs in the antioxidant and anti-inflammatory effects of Morella and Myrica species. Int. J. Mol. Sci. 2015, 16, 17160–17180. [Google Scholar] [CrossRef] [Green Version]
  14. Nhiem, N.X.; van Kiem, P.; van Minh, C.; Tai, B.H.; Cuong, N.X.; Thu, V.K.; Anh, H.L.T.; Jo, S.-H.; Jang, H.-D.; Kwon, Y.-I.; et al. A new monoterpenoid glycoside from Myrica esculenta and the inhibition of angiotensin I-converting enzyme. Chem. Pharm. Bull. 2010, 58, 1408–1410. [Google Scholar] [CrossRef] [Green Version]
  15. Gupta, G.; Siddiqui, M.A.; Khan, M.M.; Ajmal, M.; Ahsan, R.; Rahaman, M.A.; Ahmad, M.A.; Arshad, M.; Khushtar, M. Current pharmacological trends on myricetin. Drug Res. 2020, 70, 448–454. [Google Scholar] [CrossRef] [PubMed]
  16. Ghosh, J.; Sil, P.C. Arjunolic acid: A new multifunctional therapeutic promise of alternative medicine. Biochimie 2013, 95, 1098–1109. [Google Scholar] [CrossRef] [PubMed]
  17. Shen, S.; Zhao, M.; Li, C.; Chang, Q.; Liu, X.; Liao, Y.; Pan, R. Study on the material basis of neuroprotection of Myrica rubra bark. Molecules 2019, 24, 2993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Zhang, J.; Yamada, S.; Ogihara, E.; Kurita, M.; Banno, N.; Qu, W.; Feng, F.; Akihisa, T. Biological activities of triterpenoids and phenolic compounds from Myrica cerifera bark. Chem. Biodivers. 2016, 13, 1601–1609. [Google Scholar] [CrossRef]
  19. Ting, Y.-C.; Ko, H.-H.; Wang, H.-C.; Peng, C.-F.; Chang, H.-S.; Hsieh, P.-C.; Chen, I.-S. Biological evaluation of secondary metabolites from the roots of Myrica adenophora. Phytochemistry 2014, 103, 89–98. [Google Scholar] [CrossRef]
  20. Tene, M.; Tane, P.; Connolly, J.D. Triterpenoids and diarylheptanoids from Myrica arborea. Biochem. Syst. Ecol. 2008, 36, 872–874. [Google Scholar] [CrossRef]
  21. Yu, Y.-F.; Lu, Q.; Guo, L.; Mei, R.-Q.; Liang, H.-X.; Luo, D.-Q.; Cheng, Y.-X. Myricananone and myricananadiol: Two new cyclic ‘diarylheptanoids’ from the roots of Myrica nana. Helv. Chim. Acta 2007, 90, 1691–1696. [Google Scholar] [CrossRef]
  22. Nagai, M.; Sakurai, N.; Yumoto, N.; Nagumo, S.; Seo, S. Oleanane acid from Myrica cerifera. Chem. Pharm. Bull. 2000, 48, 1427–1428. [Google Scholar] [CrossRef] [Green Version]
  23. Joshi, B.S.; Pelletier, S.W.; Newton, M.G.; Lee, D.; McGaughey, G.B.; Puar, M.S. Extensive 1D, 2D NMR spectra of some [7.0]metacyclophanes and X-ray analysis of (2)-myricanol. J. Nat. Prod. 1996, 59, 759–764. [Google Scholar] [CrossRef]
  24. Nagai, M.; Dohi, J.; Morihara, M.; Sakurai, N. Diarylheptanoids from Myrica gale var. tomentosa and revised structure of porson. Chem. Pharm. Bull. 1995, 43, 1674–1677. [Google Scholar] [CrossRef] [Green Version]
  25. Dai, G.; Tong, Y.; Chen, X.; Ren, Z.; Yang, F. In vitro anticancer activity of myricanone in human lung adenocarcinoma A549 cells. Chemotherapy 2014, 60, 81–87. [Google Scholar] [CrossRef] [PubMed]
  26. Akazawa, H.; Fujita, Y.; Banno, N.; Watanabe, K.; Kimura, Y.; Manosroi, A.; Manosroi, J.; Akihisa, T. Three new cyclic diarylheptanoids and other phenolic compounds from the bark of Myrica rubra and their melanogenesis inhibitory and radical scavenging activities. J. Oleo Sci. 2010, 59, 213–221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Paul, A.; Das, J.; Das, S.; Samadder, A.; Khuda-Bukhsh, A.R. Anticancer potential of myricanone, a major bioactive component of Myrica cerifera: Novel signaling cascade for accomplishing apoptosis. J. Acupunct. Meridian Stud. 2013, 6, 188–198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Wang, J.; Dong, S.; Wang, Y.; Lu, Q.; Zhong, H.; Du, G.; Zhang, L.; Cheng, Y. Cyclic diarylheptanoids from Myrica nana inhibiting nitric oxide release. Bioorg. Med. Chem. 2008, 16, 8510–8515. [Google Scholar] [CrossRef]
  29. Tene, M.; Wabo, H.K.; Kamnaing, P.; Tsopmo, A.; Tane, P.; Ayafor, J.F.; Sterner, O. Diarylheptanoids from Myrica arborea. Phytochemistry 2000, 54, 975–978. [Google Scholar] [CrossRef]
  30. Shen, S.; Liao, Q.; Liu, J.; Pan, R.; Lee, S.M.; Lin, L. Myricanol rescues dexamethasone-induced muscle dysfunction via a sirtuin 1-dependent mechanism. J. Cachexia Sarcopenia Muscle 2019, 10, 429–444. [Google Scholar] [CrossRef]
  31. Shen, S.; Liao, Q.; Feng, Y.; Liu, J.; Pan, R.; Lee, S.M.-Y.; Lin, L. Myricanol mitigates lipid accumulation in 3T3-L1 adipocytes and high fat diet-fed zebrafish via activating AMP-activated protein kinase. Food Chem. 2019, 270, 305–314. [Google Scholar] [CrossRef]
  32. Chen, P.; Lin, X.; Yang, C.-H.; Tang, X.; Chang, Y.-W.; Zheng, W.; Luo, L.; Xu, C.; Chen, Y.-H. Study on chemical profile and neuroprotective activity of Myrica rubra leaf extract. Molecules 2017, 22, 1226. [Google Scholar] [CrossRef] [Green Version]
  33. Dai, G.H.; Meng, G.M.; Tong, Y.L.; Chen, X.; Ren, Z.M.; Wang, K.; Yang, F. Growth-inhibiting and apoptosis-inducing activities of Myricanol from the bark of Myrica rubra in human lung adenocarcinoma A549 cells. Phytomedicine 2014, 21, 1490–1496. [Google Scholar] [CrossRef]
  34. Malterud, K.E.; Anthonsen, T.; Hjortas, J. 14-Oxa-[7.1]-metapara-cyclophanes from Myrica gale L., a new class of natural products. Tetrahedron Lett. 1976, 17, 3069–3072. [Google Scholar] [CrossRef]
  35. Morihara, M.; Sakurai, N.; Inoue, T.; Kawai, K.-I.; Nagai, M. Two novel diarylheptanoid glucosides from Myrica gale var. tomentosa and absolute structure of plane-chiral galeon. Chem. Pharm. Bull. 1997, 45, 820–823. [Google Scholar] [CrossRef] [Green Version]
  36. Zhang, X.; Cheng, B.; Liu, X.; Li, Y.; Hou, J.; Chen, S.; Chen, J.; Li, S. Screening of α-glucosidase inhibitors from Houttuynia cordata and evaluation of the binding mechanisms. ChemistrySelect 2020, 5, 8440–8446. [Google Scholar] [CrossRef]
  37. Sun, X.; Zhang, L.; Cao, Y.; Li, J.; Atanasov, A.G.; Huang, L. Anti-neuraminidase activity of chemical constituents of Balanophora involucrata. Biomed. Chromatogr. 2020, 34, 4949. [Google Scholar] [CrossRef] [PubMed]
  38. Calassara, L.L.; Pinto, S.C.; Condack, C.P.M.; Leite, B.F.; Nery, L.C.D.E.S.; Tinoco, L.W.; Aguiar, F.A.; Leal, I.C.R.; Martins, S.M.; Silva, L.L.D.; et al. Isolation and characterization of flavonoids from Tapirira guianensis leaves with vasodilatory and myeloperoxidase-inhibitory activities. Nat. Prod. Res. 2020, 25, 1–4. [Google Scholar] [CrossRef] [PubMed]
  39. Aldana, J.A.; De Grandis, R.A.; Nicolella, H.; Guissoni, A.P.P.; Squarisi, I.; Arruda, C.; Ribeiro, V.P.; Tavares, D.C.; Barcelos, G.R.M.; Antunes, L.M.G.; et al. Evaluation of cytoprotective effects of compounds isolated from Copaifera langsdorffii Desf. against induced cytotoxicity by exposure to methylmercury and lead. Nat. Prod. Res. 2020, 34, 2528–2532. [Google Scholar] [CrossRef] [PubMed]
  40. Jones, J.R.; Lebar, M.D.; Jinwal, U.K.; Abisambra, J.F.; Koren, J.; Blair, L.; O’Leary, J.C.; Davey, Z.; Trotter, J.; Johnson, A.G.; et al. The diarylheptanoid (+)−aR,11S-myricanol and two flavones from bayberry (Myrica cerifera) destabilize the microtubule-associated protein Tau. J. Nat. Prod. 2011, 74, 38–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Kim, H.H.; Kim, D.H.; Kim, M.H.; Oh, M.H.; Kim, S.R.; Park, K.J.; Lee, M.W. Flavonoid constituents in the leaves of Myrica rubra Sieb. et Zucc. with anti-inflammatory activity. Arch. Pharmacal. Res. 2013, 36, 1533–1540. [Google Scholar] [CrossRef] [PubMed]
  42. Paul, B.D.; Rao, G.S.; Kapadia, G.J. Isolation of myricadiol, myricitrin, taraxerol, and taraxerone from Myrica cerifera L. root bark. J. Pharm. Sci. 1974, 63, 958–959. [Google Scholar] [CrossRef]
  43. Wang, M.; Sun, G.-B.; Du, Y.-Y.; Tian, Y.; Liao, P.; Liu, X.-S.; Ye, J.-X.; Sun, X.-B. Myricitrin protects cardiomyocytes from hypoxia/reoxygenation injury: Involvement of heat shock protein 90. Front. Pharmacol. 2017, 8, 353. [Google Scholar] [CrossRef] [Green Version]
  44. Zhang, B.; Shen, Q.; Chen, Y.; Pan, R.; Kuang, S.; Liu, G.; Sun, G.; Sun, X. Myricitrin alleviates oxidative stress-induced inflammation and apoptosis and protects mice against diabetic cardiomyopathy. Sci. Rep. 2017, 7, 44239. [Google Scholar] [CrossRef]
  45. Qin, M.; Luo, Y.; Meng, X.-B.; Wang, M.; Wang, H.-W.; Song, S.-Y.; Ye, J.-X.; Pan, R.-L.; Yao, F.; Wu, P.; et al. Myricitrin attenuates endothelial cell apoptosis to prevent atherosclerosis: An insight into PI3K/Akt activation and STAT3 signaling pathways. Vasc. Pharmacol. 2015, 70, 23–34. [Google Scholar] [CrossRef] [PubMed]
  46. Chen, W.; Feng, L.; Shen, Y.; Su, H.; Li, Y.; Zhuang, J.; Zhang, L.; Zheng, X. Myricitrin inhibits acrylamide-mediated cytotoxicity in human Caco-2 cells by preventing oxidative stress. BioMed Res. Int. 2013, 2013, 1–7. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, Y.-H.; Xuan, Z.-H.; Tian, S.; He, G.-R.; Du, G.-H. Myricitrin attenuates 6-hydroxydopamine-induced mitochondrial damage and apoptosis in PC12 cells via inhibition of mitochondrial oxidation. J. Funct. Foods 2013, 5, 337–345. [Google Scholar] [CrossRef]
  48. Manaharan, T.; Appleton, D.; Cheng, H.M.; Palanisamy, U.D. Flavonoids isolated from Syzygium aqueum leaf extract as potential antihyperglycaemic agents. Food Chem. 2012, 132, 1802–1807. [Google Scholar] [CrossRef]
  49. Fang, J.; Paetz, C.; Schneider, B. C-methylated flavanones and dihydrochalcones from Myrica gale seeds. Biochem. Syst. Ecol. 2011, 39, 68–70. [Google Scholar] [CrossRef]
  50. Oracz, K.; Voegele, A.; Tarkowská, D.; Jacquemoud, D.; Turečková, V.; Urbanová, T.; Strnad, M.; Sliwinska, E.; Leubner-Metzger, G. Myrigalone A inhibits Lepidium sativum seed germination by interference with gibberellin metabolism and apoplastic superoxide production required for embryo extension growth and endosperm rupture. Plant. Cell Physiol. 2012, 53, 81–95. [Google Scholar] [CrossRef] [Green Version]
  51. Popovici, J.; Bertrand, C.; Jacquemoud, D.; Bellvert, F.; Fernandez, M.P.; Comte, G.; Piola, F. An allelochemical from Myrica gale with strong phytotoxic activity against highly invasive Fallopia x bohemica taxa. Molecules 2011, 16, 2323–2333. [Google Scholar] [CrossRef]
  52. Mathiesen, L.; Malterud, K.; Sund, R. Antioxidant activity of fruit exudate and C-methylated dihydrochalcones from Myrica gale. Planta Med. 1995, 61, 515–518. [Google Scholar] [CrossRef]
  53. Mathiesen, L.; Malterud, K.E.; Sund, R.B. Uncoupling of respiration and inhibition of ATP synthesis in mitochondria by C-methylated flavonoids from Myrica gale L. Eur. J. Pharm. Sci. 1996, 4, 373–379. [Google Scholar] [CrossRef]
  54. Popovici, J.; Comte, G.; Bagnarol, Ã.; Alloisio, N.; Fournier, P.; Bellvert, F.; Bertrand, C.; Fernandez, M.P. Differential effects of rare specific flavonoids on compatible and incompatible strains in the Myrica gale-Frankia actinorhizal symbiosis. Appl. Environ. Microbiol. 2010, 76, 2451–2460. [Google Scholar] [CrossRef] [Green Version]
  55. Gafner, S.; Wolfender, J.-L.; Mavi, S.; Hostettmann, K. Antifungal and antibacterial chalcones from Myrica serrata. Planta Med. 1996, 62, 67–69. [Google Scholar] [CrossRef] [PubMed]
  56. Malterud, K.E. C-Methylated dihydrochalcones from Myrica gale fruit exudate. Acta Pharm. Nordica 1992, 4, 65–68. [Google Scholar]
  57. Lin, Y.; Chen, H.; Hsieh, C.; Huang, Y.; Chang, I. Betulin inhibits mTOR and induces autophagy to promote apoptosis in human osteosarcoma cell lines. Environ. Toxicol. 2020, 35, 879–887. [Google Scholar] [CrossRef] [PubMed]
  58. Wold, C.W.; Gerwick, W.H.; Wangensteen, H.; Inngjerdingen, K.T. Bioactive triterpenoids and water-soluble melanin from Inonotus obliquus (Chaga) with immunomodulatory activity. J. Funct. Foods 2020, 71, 104025. [Google Scholar] [CrossRef]
  59. Wan, Y.; Jiang, S.; Lian, L.-H.; Bai, T.; Cui, P.-H.; Sun, X.-T.; Jin, X.-J.; Wu, Y.-L.; Nan, J.-X. Betulinic acid and betulin ameliorate acute ethanol-induced fatty liver via TLR4 and STAT3 in vivo and in vitro. Int. Immunopharmacol. 2013, 17, 184–190. [Google Scholar] [CrossRef]
  60. Sakurawi, K.; Yasuda, F.; Tozyo, T.; Nakamura, M.; Sato, T.; Kikuchi, J.; Terui, Y.; Ikenishi, Y.; Iwata, T.; Takahashi, K.; et al. Endothelin receptor antagonist triterpenoid, myriceric acid A, isolated from Myrica cerifera, and structure activity relationships of its derivatives. Chem. Pharm. Bull. 1996, 44, 343–351. [Google Scholar] [CrossRef] [Green Version]
  61. Fujimoto, M.; Mihara, S.-I.; Nakajima, S.; Ueda, M.; Nakamura, M.; Sakurai, K.-S. A novel non-peptide endothelin antagonist isolated from bayberry, Myrica cerifera. FEBS Lett. 1992, 305, 41–44. [Google Scholar] [CrossRef] [Green Version]
  62. Zhang, J.; Koike, R.; Yamamoto, A.; Ukiya, M.; Fukatsu, M.; Banno, N.; Miura, M.; Motohashi, S.; Tokuda, H.; Akihisa, T. Glycosidic inhibitors of melanogenesis from leaves of Passiflora edulis. Chem. Biodivers. 2013, 10, 1851–1865. [Google Scholar] [CrossRef]
  63. Ganapathy, G.; Preethi, R.; Moses, J.A.; Anandharamakrishnan, C. Diarylheptanoids as nutraceutical: A review. Biocatal. Agric. Biotechnol. 2019, 19, 101109. [Google Scholar] [CrossRef]
  64. Gurd, B.J. Deacetylation of PGC-1α by SIRT1: Importance for skeletal muscle function and exercise-induced mitochondrial biogenesis. Appl. Physiol. Nutr. Metab. 2011, 36, 589–597. [Google Scholar] [CrossRef]
  65. Lee, D.; Goldberg, A.L. SIRT1 protein, by blocking the activities of transcription factors FoxO1 and FoxO3, inhibits muscle atrophy and promotes muscle growth. J. Biol. Chem. 2013, 288, 30515–30526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Gullett, N.P.; Hebbar, G.; Ziegler, T.R. Update on clinical trials of growth factors and anabolic steroids in cachexia and wasting. Am. J. Clin. Nutr. 2010, 91, 1143–1147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Flora, G.; Gupta, D.; Tiwari, A. Toxicity of lead: A review with recent updates. Interdiscip. Toxicol. 2012, 5, 47–58. [Google Scholar] [CrossRef] [PubMed]
  68. Nersesyan, A.; Kundi, M.; Waldherr, M.; Setayesh, T.; Mišík, M.; Wultsch, G.; Filipic, M.; Mazzaron Barcelos, G.R.; Knasmueller, S. Results of micronucleus assays with individuals who are occupationally and environmentally exposed to mercury, lead and cadmium. Mutat. Res. Rev. Mutat. Res. 2016, 770, 119–139. [Google Scholar] [CrossRef]
  69. Chen, Y.-Q.; Wohlbold, T.J.; Zheng, N.-Y.; Huang, M.; Huang, Y.; Neu, K.E.; Lee, J.; Wan, H.; Rojas, K.T.; Kirkpatrick, E.; et al. Influenza infection in humans induces broadly cross-reactive and protective neuraminidase-reactive antibodies. Cell 2018, 173, 417–429. [Google Scholar] [CrossRef] [Green Version]
  70. Demine, S.; Renard, P.; Arnould, T. Mitochondrial uncoupling: A key controller of biological processes in physiology and diseases. Cells 2019, 8, 795. [Google Scholar] [CrossRef] [Green Version]
  71. Shen, S.; Liao, Q.; Zhang, T.; Pan, R.; Lin, L. Myricanol modulates skeletal muscle–adipose tissue crosstalk to alleviate high-fat diet-induced obesity and insulin resistance. Br. J. Pharmacol. 2019, 176, 3983–4001. [Google Scholar] [CrossRef] [Green Version]
  72. Dai, G.; Tong, Y.; Chen, X.; Ren, Z.; Ying, X.; Yang, F.; Chai, K. Myricanol induces apoptotic cell death and anti-tumor activity in non-small cell lung carcinoma in vivo. Int. J. Mol. Sci. 2015, 16, 2717–2731. [Google Scholar] [CrossRef]
  73. Domitrović, R.; Rashed, K.; Cvijanović, O.; Vladimir-Knežević, S.; Škoda, M.; Višnić, A. Myricitrin exhibits antioxidant, anti-inflammatory and antifibrotic activity in carbon tetrachloride-intoxicated mice. Chem. Biol. Interact. 2015, 230, 21–29. [Google Scholar] [CrossRef]
  74. Yang, Y.-L.; Liu, M.; Cheng, X.; Li, W.-H.; Zhang, S.-S.; Wang, Y.-H.; Du, G.-H. Myricitrin blocks activation of NF-κB and MAPK signaling pathways to protect nigrostriatum neuron in LPS-stimulated mice. J. Neuroimmunol. 2019, 337, 577049. [Google Scholar] [CrossRef] [Green Version]
  75. Bai, T.; Yang, Y.; Yao, Y.-L.; Sun, P.; Lian, L.-H.; Wu, Y.-L.; Nan, J.-X. Betulin alleviated ethanol-induced alcoholic liver injury via SIRT1/AMPK signaling pathway. Pharmacol. Res. 2016, 105, 1–12. [Google Scholar] [CrossRef] [PubMed]
  76. Xin, C.; Liu, J.; Zhang, J.; Zhu, D.; Wang, H.; Xiong, L.; Lee, Y.; Lian, K.; Xu, C.; Zhang, L.; et al. Irisin improves fatty acid oxidation and glucose utilization in type 2 diabetes by regulating the AMPK signaling pathway. Int. J. Obes. 2016, 40, 443–451. [Google Scholar] [CrossRef] [PubMed]
  77. Huh, J.Y.; Mougios, V.; Kabasakalis, A.; Fatouros, I.; Siopi, A.; Douroudos, I.I.; Filippaios, A.; Panagiotou, G.; Park, K.H.; Mantzoros, C.S. Exercise-induced irisin secretion is independent of age or fitness level and increased irisin may directly modulate muscle metabolism through AMPK activation. J. Clin. Endocrinol. Metab. 2014, 99, E2154–E2161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Fourie, C.; Kim, E.; Waldvogel, H.; Wong, J.M.; Mcgregor, A.; Faull, R.L.M.; Montgomery, J.M. Differential changes in postsynaptic density proteins in postmortem huntington’s disease and parkinson’s disease human brains. J. Neurodegener. Dis. 2014, 2014, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. He, L.; Simmen, F.A.; Ronis, M.J.J.; Badger, T.M. Post-transcriptional regulation of sterol regulatory element-binding protein-1 by ethanol induces class I alcohol dehydrogenase in rat liver. J. Biol. Chem. 2004, 279, 28113–28121. [Google Scholar] [CrossRef] [Green Version]
  80. Santos, S.C.; Waterman, P.G. Condensed tannins from Myrica gale. Fitoterapia 2000, 71, 610–612. [Google Scholar] [CrossRef]
  81. Wang, J.-F.; Zhang, C.-L.; Lu, Q.; Yu, Y.-F.; Zhong, H.-M.; Long, C.-L.; Cheng, Y.-X. Three new diarylheptanoids from Myrica nana. Helv. Chim. Acta 2009, 92, 1594–1599. [Google Scholar] [CrossRef]
  82. Sakurai, N.; Hosono, Y.; Morihara, M.; Ishidi, J.; Kawai, K.-I. A new triterpenoid myricalactone and others from Myrica gale var. tomentosa. Yakugaku Zasshi 1997, 117, 211–219. [Google Scholar] [CrossRef] [Green Version]
  83. Agarwal, K.P.; Roy, A.C.; Dhar, M.L. Triterpenes from the bark of Myrica esculenta Buch.-Ham. Indian J. Chem. 1963, 1, 28–30. [Google Scholar]
  84. Jahanban-Esfahlan, A.; Ostadrahimi, A.; Tabibiazar, M.; Amarowicz, R. A comprehensive review on the chemical constituents and functional uses of walnut (Juglans spp.) Husk. IJMS 2019, 20, 3920. [Google Scholar] [CrossRef] [Green Version]
  85. Seo, D.Y.; Lee, S.R.; Heo, J.-W.; No, M.-H.; Rhee, B.D.; Ko, K.S.; Kwak, H.-B.; Han, J. Ursolic acid in health and disease. Korean J. Physiol. Pharmacol. 2018, 22, 235–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Liao, Q.; Yang, W.; Jia, Y.; Chen, X.; Gao, Q.; Bi, K. LC-MS determination and pharmacokinetic studies of ursolic acid in rat plasma after administration of the traditional chinese medicinal preparation Lu-Ying extract. Yakugaku Zasshi 2005, 125, 509–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Chen, Q.; Luo, S.; Zhang, Y.; Chen, Z. Development of a liquid chromatography–mass spectrometry method for the determination of ursolic acid in rat plasma and tissue: Application to the pharmacokinetic and tissue distribution study. Anal. Bioanal. Chem. 2011, 399, 2877–2884. [Google Scholar] [CrossRef] [PubMed]
Table 1. In vitro biological activities exhibited by compounds from Morella and Myrica species.
Table 1. In vitro biological activities exhibited by compounds from Morella and Myrica species.
CompoundOriginBiological Activities
Myricanone (1)
Molecules 25 06052 i001
Methanol extract of Myrica rubra (Lour.) Siebold & Zucc. bark [17]
Hexane extract of Morella cerifera (L.) Small (Myrica cerífera) bark [18]
Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
Chloroform and methanol extract of Morella arborea (Hutch.) Cheek twigs [20]
95% EtOH extract of Morella nana (A. Chev.) J. Herb. roots [21]
Hexane extract of Morella cerifera (L.) Small (Myrica cerífera) twigs [22]
Ethanol 95% extract of Morella cerifera (L.) Small (Myrica cerífera) bark [23]
Methanol extract of Myrica gale L. (Myrica gale var tormentosa L.) branches [24]
Anti-tuberculosis [19]
Minimum inhibitory concentration (MIC) = >150 µg/mL (Ethambutol MIC = 6.25 µg/mL).
Cytotoxic activity [25]
A549 cell line EC50 = 3.22 µg/mL (Fluorouracil EC50 = not shown).
Increase in apoptotic rate to 34.9% at 5.0 µg/mL (4.13% on untreated cells).
Antimelanogenesis activity [26]
Melanin content in B16 mouse melanoma cells at 25 µg/mL: 23.5 ± 3.4% (arbutin 25 µg/mL: 77.4 ± 2.9%).
Cell viability at 25 µg/mL: 17.7 ± 1.7% (arbutin 25 µg/mL: 102.0 ± 1.5%).
Cytotoxic activity [27]
HepG2 cell line: EC50 = 32.46 µg/mL.
WRL-68 cell line: at 100 µg/mL cell viability was 89.27% (non-tumor cell line).
5-Deoxymyricanone (2)
Molecules 25 06052 i002
Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]Anti-tuberculosis [19]
MIC = 25.8 µg/mL (Ethambutol MIC = 6.25 µg/mL).
12-Hydroxymyricanone (3)
Molecules 25 06052 i003
Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
Ethanol 80% extracts of Morella nana (A. Chev.) J. Herb. roots [28]
Methanol extract of Myrica gale L. (Myrica gale var tormentosa L.) branches [24]
Anti-tuberculosis [19]
MIC = 35.8 µg/mL (Ethambutol MIC = 6.25 µg/mL).
Myricanol (4)
Molecules 25 06052 i004
Methanol extract of Myrica rubra (Lour.) Siebold and Zucc. bark [17]
Hexane extract of Morella cerifera (L.) Small (Myrica cerífera) bark [18]
Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
Methanol extract of Myrica esculenta Buch.-Ham. Ex D. Don leaves [14]
Chloroform and methanol extract of Morella arborea (Hutch.) Cheek twigs [20]
95% EtOH extract of Morella nana (A. Chev.) J. Herb. roots [21]
Dichloromethane: methanol (1:1) extract of Morella arborea (Hutch.) Cheek bark and stem [29]
Ethanol 95% extract of Morella cerifera (L.) Small bark [23]
Inhibition of muscle atrophy and dysfunction [30]
C2C12 myotubes treated with 10 µM myricanol and dexamethasone showed:
↑ myosin heavy chain expression (0.89 against 0.33 on cells treated only with dexamethasone),
↓ atrogin-1 expression (1.53 against 2.31 on cells treated only with dexamethasone),
↓ MuRF1 expression (0.99 against 1.55 on cells treated only with dexamethasone),
↑ ATP production (5.84 nM/mg protein against 3.83 nM/mg protein on cells treated only with dexamethasone),
↑ mitochondrial content (116.38% against 68.12% on cells treated only with dexamethasone),
↑ mitochondrial O2 consumption (223.77 pmol/min against 166.59 pmol/min on cells treated only with dexamethasone).
Mitigation of lipid accumulation in 3TS-L1 adipocytes [31]
Treatment with 5 µM myricanol increased AMPK activation in 50% when compared to the untreated cells.
Decrease of 28% in lipid accumulation when compared to the untreated cells.
Neuroprotective effects [32]
80% increase in N2a cells viability treated with 0.84 mM and 100 mM H2O2 (against cells treated only with H2O2).
40% reduction of intracellular ROS in N2a cells treated with 0.84 mM and 100 mM H2O2 when compared with the cells treated only with H2O2.
Intracellular calcium concentration of 777.81 nM in N2a cells treated with 0.84 mM and 100 mM H2O2 (3045.51 nM in cells treated only with H2O2).
Antitumor activity [18]
HL60 cell-line: IC50 = 5.3 ± 0.7 µM (Cisplatin EC50 = 4.2 ± 1.1 µM).
A549 cell-line: IC50 = 16.5 ± 0.8 µM (Cisplatin EC50 = 18.4 ± 1.9 µM).
SK-BR-3 cell-line: EC50 = 14.8 ± 5.5 µM (Cisplatin EC50 = 18.8 ± 0.6 µM).
Anti tuberculosis [19]
MIC = 30 µg/mL (Ethambutol MIC = 6.25 µg/mL).
Cytotoxic activity [33]
A549 cell line EC50 = 4.85 µg/mL (Fluorouracil EC50 = not shown).
Antimelanogenesis activity [26]
Melanin content in B16 mouse melanoma cells at 25 µg/mL: 3.8 ± 0.4% (arbutin 25 µg/mL: 77.4 ± 2.9%).
Cell viability at 25 µg/mL: 8.8 ± 0.2% (arbutin 25 µg/mL: 102.0 ± 1.5%).
Myricanol 11-sulphate (5)
Molecules 25 06052 i005
Methanol extract of Myrica rubra (Lour.) Siebold and Zucc. bark [17]
Protection against glutamate-induced damage [17]
PC12 cell line: 5 µM of myricanol 11-sulphate maintained 72.09 ± 2.09% of cell viability after 24 h exposure to glutamate. (Cells treated only with glutamate showed viability of 50%).
Porson (6)
Molecules 25 06052 i006
Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
95% EtOH extract of Morella nana (A. Chev.) J. Herb. roots [21]
Methanol extract of Myrica gale L. (Myrica gale var tormentosa L.) branches [24]
Ethyl acetate extract of Myrica gale L. stems [34]
Anti-tuberculosis [19]
MIC = 40 µg/mL (Ethambutol MIC = 6.25 µg/mL).
Myricananin C (7)
Molecules 25 06052 i007
Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
Ethanol 80% extract of Morella nana (A. Chev.) J. Herb. roots [28]
Anti-tuberculosis [19]
MIC = 55.5 µg/mL (Ethambutol MIC = 6.25 µg/mL).
(+)-Galeon (8)
Molecules 25 06052 i008
Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
Methanol extract of Myrica gale L. (Myrica gale var tormentosa L.) branches [35]
Ethyl acetate extract of Myrica gale L. stems [34]
Anti-tuberculosis [19]
MIC = 15.0 µg/mL (Ethambutol MIC = 6.25 µg/mL).
Quercitrin (9)
Molecules 25 06052 i009
Methanol extract of Myrica rubra (Lour.) Siebold and Zucc. bark [17]
Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
α-Glucosidase Inhibition [36]
IC50 = 0.231 ± 0.033 mg/mL (Acarbose IC50 = 1.457 ± 0.144 mg/mL).
Antiviral Activity [37]
Neuraminidase inhibition: IC50 = 311.76 μM (Oseltamivir acid IC50 = 280 μM).
Myeloperoxidase inhibition [38]
IC50 = 2.0 ± 0.2 μM
Prevention of metal toxicity effects [39]
Almost 100% HepG2 cell line viability when treated with 1 μM + 15 μM MeHg (50% viability when only treated with 15 μM MeHg).
Almost 100% HepG2 cell line viability when treated with 1 μM + 40 μM Pb(NO3)2 (50% viability when only treated with 40 μM Pb(NO3)2).
About 50% PC12 cell line viability when treated with 1 μM + 10 μM MeHg (65% viability when only treated with 10 μM MeHg).
Almost 90% PC12 cell line viability when treated with 1 μM + 350 μM Pb(NO3)2 (60% viability when only treated with 350 μM Pb(NO3)2).
Anti-tuberculosis [19]
MIC = >150 µg/mL (Ethambutol MIC = 6.25 µg/mL).
Myricitrin (10)
Molecules 25 06052 i010
Methanol extract of Myrica rubra (Lour.) Siebold and Zucc. bark [17]
Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
Toluene extract of the root-bark Morella cerifera (L.) Small (Myrica cerifera) [40]
Methanol extract of Myrica esculenta Buch.-Ham. Ex D. Don leaves [14]
80% Acetone extract of Myrica rubra (Lour.) Siebold and Zucc. leaves [41]
Benzene extract (after petroleum ether) of Morella cerifera (L.) Small (Myrica cerifera) root bark [42]
Protection against glutamate-induced damage [17]
PC12 cell line: 10 µM of myricitrin maintained 73.15 ± 3.23% of cell viability after 24 h exposure to glutamate. (Cells treated only with glutamate showed only viability of 50%).
Protection against hypoxia/reoxygenation (R/H) injury in cardiomyocyte [43]
H9c2 cells pretreated for 12 h with 40 µM had a survival rate of 87.48 ± 7.68% (survival rate of untreated cells = 65.48 ± 6.42%).
50% decrease of LDH levels secreted to culture medium in the cells pretreated for 12 h with 40 µM when compared with untreated cells.
Pretreatment with 40 µM inhibited intracellular ROS generation in 50% when compared to untreated cells.
Reduction of 50% in the apoptotic rate on cells treated with 40 µM when compared with the untreated cells.
Pretreatment with 40 µM decreased the caspase-3 activity in 25% when compared with the untreated cells.
Pretreatment with 40 µM increased the Bcl-2/Bax expression ratio to 1.28 ± 0.31 (0.46 ± 0.12 in untreated cells.
Protection against advanced glycation end products (AGEs) injury in cardiomyocyte [44]
Pretreatment with 25 µg/mL led to H9c2 cells viability of 78.94 ± 4.52% (60.34 ± 6.52% when treated only with 400 μg/mL AGEs).
Pretreatment with 25 µg/mL µM decreased phospho-IKK-β expression in about 40%, when compared with the untreated cells.
Pretreatment with 25 µg/mL decreased TNF-α expression in about 50%, when compared with the untreated cells.
Pretreatment with 25 µg/mL decreased intracellular ROS generation in about 50%, when compared with the untreated cells.
Pretreatment with 25 µg/mL markedly attenuated the inhibition of NQO-1, γ -GCS, and HO-1 expression induced by AGEs.
Pretreatment with 25 µg/mL decreased caspase-3 and caspase-9 activity in 3 and 2-fold, respectively, when compared with the untreated cells.
Pretreatment with 25 µg/mL decreased apoptotic rate in about 7%, when compared with the untreated cells.
Pretreatment with 25 µg/mL led to a 2-fold decrease in the expression of Bax compared with the untreated cells.
Pretreatment with 25 µg/mL led to a 2-fold increase in the expression of Bcl-2 compared with the untreated cells.
Pretreatment with 25 µg/mL led to a 33% decrease in the expression of collagen-1 compared with the untreated cells.
Prevention of atherosclerosis [45]
Pretreatment with 40 µM in oxidized low-density lipoprotein (ox-LDL) exposed human umbilical vein endothelial cells (HUVECs):
Increased cell viability (70.75 ± 8.44% against 50.25 ± 7.95% in the untreated cells);
Reduction of apoptotic cells (15.58 ± 4.65% against 23.89 ± 3.65% in the untreated cells);
3-fold reduction of intracellular ROS levels when compared with untreated cells;
~33% reduction on caspase-3 activity when compared with untreated cells.
Anti-tuberculosis [19]
MIC = >150 µg/mL (Ethambutol MIC = 6.25 µg/mL).
Protection against acrylamide induced oxidative stress [46]
Acrylamide induced Caco-2 cells treated with
40 µg/mL presented a viability of 80% (untreated cells had a viability of 50%).
Neuroprotective activity [47]
Pretreatment with 10µM prevents PC12 cell death induced by 6-hydroxydopamine
(6-OHDA) (untreated cells presented 25% of cell mortality).
Pretreatment with 10µM decreased cytochrome C release by almost 60% when compared with the untreated group.
Pretreatment with 10µM decreased caspase-3 activity release by almost 50% when compared with the untreated group.
Pretreatment with 10µM decreased the apoptotic rate activity by almost 50% when compared with the untreated group.
Antidiabetic activity [48]
α-glucosidase inhibition: IC50 = 1.1 ± 0.06 µM
(acarbose: IC50 = 43 ± 1.6 µM).
β-amylase inhibition: IC50 = 1.9 ± 0.02 µM.
(acarbose: IC50 = 19 ± 1.6 µM).
Antimelanogenesis activity [26]
Melanin content in B16 mouse melanoma cells at 25 µg/mL: 69.9 ± 2.3% (arbutin 25 µg/mL: 77.4 ± 2.9%).
Cell viability at 25 µg/mL: 102.9 ± 3.3% (arbutin 25 µg/mL: 102.0 ± 1.5%).
Myrigalone A (11)
Molecules 25 06052 i011
50% acetone extract of Myrica gale L. seeds [49]
Methanol extract of Myrica gale L. leaves and fruits [34], fruits [50], seeds [51]
Diethyl ether extract of the fruit exudate of Myrica gale L. [52]
Uncoupling activity [53]
Increases mitochondrial respiration rate by 87 ± 8 natoms O/min/mg (DNP 36 ± 3 natoms O/min/mg) at 45 µM.
Myrigalone B (12)
Molecules 25 06052 i012
50% Acetone extract of Myrica gale L. seeds [49]
Methanol extract of Myrica gale L. seeds [54]
Dichloromethane extract of Morella serrata (Lam.) Killick leaves [55]
Diethyl ether extract of the fruit exudate of Myrica gale L. [52]
Antidiabetic activity [48]
α-glucosidase inhibition: IC50 = 19 ± 1.0 µM (acarbose: IC50 = 43 ± 1.6 µM).
β-amylase inhibition: IC50 = 8.3 ± 1.3 µM (acarbose: IC50 = 19 ± 1.6 µM).
Uncoupling activity [53]
Increases mitochondrial respiration rate by 40 ± 10 natoms O/min/mg (DNP 36 ± 3 natoms O/min/mg) at 45µM.
Myrigalone D (13)
Molecules 25 06052 i013
50% acetone extract of Myrica gale L. seeds [49]
Methanol extract of Myrica gale L. seeds [54] and leaves [34]
From the Myrica gale L. fruits [53]
Uncoupling activity [53]
Increases mitochondrial respiration rate by 14 ± 2 natoms O/min/mg (DNP 36 ± 3 natoms O/min/mg) at 45 µM.
Myrigalone H (14)
Molecules 25 06052 i014
Diethyl ether extract of the fruit exudate of Myrica gale L. [52]
Myrica gale L. fruit exudate [56]
Uncoupling activity [53]
Increases mitochondrial respiration rate by 17 ± 3 natoms O/min/mg (DNP 36 ± 3 natoms O/min/mg) at 45 µM.
Myrigalone G (15)
Molecules 25 06052 i015
50% acetone extract of Myrica gale L. seeds [49]
Diethyl ether extract of the fruit exudate of Myrica gale L. [52]
Antidiabetic activity [48]
α-glucosidase inhibition: IC50 = 7 ± 1.4 µM (acarbose: IC50 = 43 ± 1.6 µM).
β-amylase inhibition: IC50 = 33 ± 6.6 µM (acarbose: IC50 = 19 ± 1.6 µM).
Uncoupling activity [53]
Increases mitochondrial respiration rate by 71 ± 5 natoms O/min/mg (DNP 36 ± 3 natoms O/min/mg) at 45 µM.
Betulin (16)
Molecules 25 06052 i016
Hexane extract of Morella cerifera (L.) Small (Myrica cerifera) bark [18]Anti-osteosarcoma activity [57]
MG-63 cell line: EC50 = 14.54 µM.
HOS cell line: EC50 = 11.70 µM.
Antitumor activity [58]
NCI-H460 cell line: EC50 = 2.8 ± 0.4 µM (Podophyllotoxin: EC50 = 22 ± 6 µM).
HT29-MTX cell line: EC50 = 1.6 ± 0.4 µM (Podophyllotoxin: EC50 = 24 ± 3 µM).
Antitumor activity [18]
HL60 cell-line: EC50 = 15.2 ± 2.3 µM (Cisplatin EC50 = 4.2 ± 1.1 µM).
A549 cell-line: EC50 = 5.2 ± 3.0 µM (Cisplatin EC50 = 18.4 ± 1.9 µM).
SK-BR-3 cell-line: EC50 = 3.1 ± 0.6 µM (Cisplatin EC50 = 18.8 ± 0.6 µM).
Hepatoprotective activity [59]
Pretreatment with 25 µM in ethanol-induced HSC-T6 cells:
Decrease in the expression levels of collagen-I (35%), α-SMA (25%) and TLR4 (60%), when compared with untreated cells.
~30% increase in phosphorylation of STAT3, when compared with the untreated group.
Myriceric acid A (17)
Molecules 25 06052 i017
Methanol extract of Morella cerifera (L.) Small (Myrica cerifera) twigs [60] and branches [61]Anti-hypertension activity [60]
Endothelin 1 receptor antagonist: IC50 = 11 ± 2 nM.
3β-trans-p-Coumaroyloxy-2α,23-dihydroxyolean-12-en-28-oic acid (18)
Molecules 25 06052 i018
Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]Anti-tuberculosis [19]
MIC = 45 µg/mL (Ethambutol MIC = 6.25 µg/mL).
(R)-4-(5-Hydroxy-7-(4-hydroxyphenyl)heptyl)-2-methoxyphenol (19)
Molecules 25 06052 i019
Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]Anti-tuberculosis [19]
MIC = 52 µg/mL (Ethambutol MIC = 6.25 µg/mL).
Corchoionoside C (20)
Molecules 25 06052 i020
Methanol extract of Myrica esculenta Buch.-Ham. Ex D. Don leaves [14]Angiotensin I-converting enzyme [14]
29.97 ± 4.77% ACE 1 inhibition at 100 µM (Captopril 88.64 ± 2.57 at 4.6 µM).
(6S,9R)-Roseoside (21)
Molecules 25 06052 i021
Methanol extract of Myrica esculenta Buch.-Ham. Ex D. Don leaves [14]Antimelanogenesis activity [62]
Inhibition of melanogenesis in B16 mouse melanoma cells at 100 µM: 62.7 ± 3.1% (arbutin 100 µM: 70.3 ± 5.5%).
Cell viability at 100 µM: 95.0 ± 2.2% (arbutin 100 µM: 87.5 ± 2.8%).
Angiotensin I-converting enzyme [14]
25.63 ± 1.35% ACE 1 inhibition at 100 µM (Captopril 88.64 ± 2.57 at 4.6 µM).
Myricadenin A (22)
Molecules 25 06052 i022
Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]Anti-tuberculosis [19]
MIC = 80.0 µg/mL (Ethambutol MIC = 6.25 µg/mL).
d-Glcpy = d-glucopyranoside.
Table 2. In vivo biological activities exhibited by compounds from Morella and Myrica species.
Table 2. In vivo biological activities exhibited by compounds from Morella and Myrica species.
CompoundModelDoseActivity
Myricanol (4)C57BL/6 mice5 mg/kgProtection against muscle atrophy [30]
Reduction of quadriceps muscle mass loss (1.36 ± 0.02% b/w against 1.18 ± 0.06% b/w on the untreated group).
Reduction of gastrocnemius muscle mass loss (0.87 ± 0.08% b/w against 0.78 ± 0.05% b/w on the untreated group).
Improvement of grip strength (120.58 ± 7.93 g against 70.90 ± 04.59 g on the untreated group).
Increase in forced swim time (83.75 ± 15.19 s against 48.80 ± 11.43 s on the untreated group).
Inhibition of muscle atrophy (~25% increase in muscle fiber diameter when compared with untreated group).
C57BL/6J25 mg/kgAnti-obesity and diabetic activity [71]
Reduction of body weight and body fat gain under high fat diet when compared with untreated group.
Decrease in serum total cholesterol, triglycerides, LDL-cholesterol, and LDL/HDL ratio (~33%).
50% reduction of fasting insulin levels when compared with untreated group.
~2-fold increase in insulin sensitivity when compared with untreated group.
Increased levels of phosphorylation of IRS-1 (2.5-fold), AKT (3-fold), and GSK-3β (1.5-fold) when compared with untreated group.
35% decrease in adipocyte diameter when compared with untreated group.
~33% increase in irisin serum levels when compared with untreated group.
Zebrafish1 µMAnti-obesity activity [31]
66% decrease in lipid accumulation under high-fat diet, when compared with the untreated group (positive control AICAR (5 µM): 75% decrease).
~70–80% reduction of PPAR-γ, C/EBPα, SREB-1 and aP2 expression when compared with the untreated group (similar values obtained with the positive control).
BALB/c nude mice40 mg/kgAntitumor activity [72]
39.4% reduction on A549 xenograft tumor volume after 14 days, when compared with untreated group.
~33% increase in the expression levels of Bax when compared with untreated group.
Decrease in the expression levels of Bcl-2 (25%), vascular endothelial growth factor (VEGF) (20%), and Survivin (33%) when compared with untreated group.
20% increase on the number of apoptotic tumor cells when compared with untreated group.
Myricitrin (10)ApoE -/- mice50 mg/kgPrevention of atherosclerosis [45]
~25% reduction of serum levels of Ox-LDL (similar reduction in the positive control group treated with 2 g/kg of probucol).
22% reduction on aortic wall thickness when compared to the untreated group.
Complete inhibition of calcification on aortic arch. (Calcification observed in the untreated group).
26% reduction on atherosclerotic plaque area when compared with the untreated group.
~20% reduction of caspase-3 expression in aortic arch.
BALB/cN mice100 mg/kgHepatoprotective activity [73]
Protection against CCl4 intoxication.
Reduction in body weight change (−12.9 ± 0.8% against 19.6 ± 2.3% on the untreated group.
Reduction of ALT serum levels (481 ± 53 U/L against 2126 ± 268 U/L on the untreated group).
Reduction of AST serum levels (592 ± 74 U/L against 2538 ± 322 U/L on the untreated group).
Reduction of liver lipids peroxidation.
Increase on glutathione levels (46.1 ± 3.4 µmol/g protein against 28.6 ± 2.8 µmol/g protein on the untreated group).
~25% decrease in necrotic areas on the liver when compared with untreated group.
5-fold increase on CYP2E1 expression when compared with the untreated group.
BALB/c mice300 mg/kgProtection against diabetic cardiomyopathy [44]
~20–25% improvement in cardiac function of diabetic mice when compared with untreated group.
Decrease on abnormalities in the arrangement of cardiac fibers and morphology of cardiomyocytes when compared with untreated group.
Inhibition of collagen network destruction and reduction of fibrotic alterations of the hearth.
2-fold reduction in the expression of TGF-β1 when compared with the untreated group.
8-fold reduction in the expression levels of collagen-1.
Reduction on serum levels of IL-6 (35.72 pg/mL against 112.41 pg/mL in the untreated group).
Reduction on serum levels of TNF-α (24.83 pg/mL against 56.21 pg/mL in the untreated group).
Decrease of Bax/Bcl-2 ratio (35-fold), caspase-3 (5-fold), and caspase-9 (4-fold) expression levels, when compared with the untreated group.
BALB/c mice50 mg/kgNeuroprotective activity [74]
In LPS-stimulated mice:
~2-fold increase on the expression levels of PSD-95 and TH, when compared with untreated group.
Reduction on expression levels of IL-1β (3-fold), IL-6 (2-fold), TNF-α (2-fold), and MCP-1 (2-fold), when compared with the untreated group.
Reduction on the expression levels of COX-2 (3-fold) and iNOS (2-fold), when compared with the untreated group.
Suppression of LPS-stimulated p38 (4-fold), ERK (4-fold), and JNK (2-fold) activation.
Betulin (16)C57BL/6 mice20 mg/kgHepatoprotective activity [75]
Decrease of liver/body weight ratio in alcoholic mice, when compared with untreated group.
Decrease in serum levels of ALT (66%), AST (33%), and TG (50%), when compared with the untreated group.
Decrease on expression levels of collagen-I (20%), SREBP-1 (25%), and α-SMA (50%), when compared with untreated group.
Increased phosphorylation of LKB1 (15%) and AMPK (25%) when compared with untreated group.
7-fold increase in SIRT-1 expression when compared with untreated group.
C57BL/6 mice50 mg/kgHepatoprotective activity [47]
Decrease in serum levels of ALT (50%), AST (50%), and TG (33%) in ethanol induced fatty-liver mice, when compared with the untreated group.
Decrease in expression levels of SREBP-1 (75%), CYP2E1 (60%), and TLR4 (60%), when compared with the untreated group.
~30% increase in phosphorylation of STAT3, when compared with the untreated group.
Table 3. Other secondary metabolites isolated from Morella/Myrica genera.
Table 3. Other secondary metabolites isolated from Morella/Myrica genera.
NameStructureExtract, Species, and Part of Plant
2′,4′-Dihydroxy-6′-methoxy-3′,5′-dimethylchalcone (23) Molecules 25 06052 i023Dichloromethane extract of Morella serrata (Lam.) Killick leaves [55]
Aurentiacin A (24) Molecules 25 06052 i024Dichloromethane extract of Morella serrata (Lam.) Killick leaves [55]
2′,6′-Dihydroxy-4′-methoxy-3′-methyl-dihydrochalcone (25) Molecules 25 06052 i025Dichloromethane extract of Morella serrata (Lam.) Killick leaves [55]
Myrigalone E (26) Molecules 25 06052 i026Methanol extract of Myrica gale L. seeds [54]
From the Myrica gale L. fruits [53]
Dichloromethane extract of Morella serrata (Lam.) Killick leaves [55]
Myrigalone P (27) Molecules 25 06052 i027Methanol extract of Myrica gale L. seeds [54]
Uvangoletin (28) Molecules 25 06052 i028Methanol extract of Myrica gale L. seeds [54]
Angoletin (29) Molecules 25 06052 i029Fruit exudate from the Myrica gale L. fruits [53,56]
Demethoxymatteucinol (30) Molecules 25 06052 i030Dichloromethane extract of Morella serrata (Lam.) Killick leaves [55]
Methanol extract of Myrica gale L. seeds [54]
Cryptostrobin (31) Molecules 25 06052 i031Dichloromethane extract of Morella serrata (Lam.) Killick leaves [55]
Demethoximatteucinol-7-methoxy (32) Molecules 25 06052 i032Methanol extract of Myrica gale L. seeds [54]
Myricetin 3-O-6″-galloyl-β-D-galactopyranoside (33) Molecules 25 06052 i033Methanol extract of Myrica gale L. (Myrica gale var tormentosa L.) branches [24]
Gallocatechin-4-α-8-epicatechin (34) Molecules 25 06052 i034Aerial parts of Myrica gale L. [80]
Gallocatechin-4-α-8-epigallocatechin (35) Molecules 25 06052 i035Aerial parts of Myrica gale L. [80]
Adenodimerin B (36) Molecules 25 06052 i036Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
Adenodimerin C (37) Molecules 25 06052 i037Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
Gallocatechin-(4-α-8)-gallocatechin-(4-α-8)-gallocatechin (38) Molecules 25 06052 i038Aerial parts of Myrica gale L. [80]
Myricananin B (39) Molecules 25 06052 i039Ethanol 80% extracts of Morella nana (A. Chev.) J. Herb. roots [28]
Myricananin E (40) Molecules 25 06052 i040Ethanol 80% extracts of Morella nana (A. Chev.) J. Herb. roots [28]
Myricananin F (41) * Molecules 25 06052 i04180% ethanol extracts of Morella nana (A. Chev.) J. Herb. roots [81]
Myricananin G (42) Molecules 25 06052 i04280% ethanol extracts of the of Morella nana (A. Chev.) J. Herb. roots [81]
Myricananin H (43) Molecules 25 06052 i04380% ethanol extracts of Morella nana (A. Chev.) J. Herb. roots [81]
Myricatomentogenin (44) Molecules 25 06052 i044Ethanol 80% extracts of Morella nana (A. Chev.) J. Herb. roots [81]
Myricanone 5-O-β-D-glucopyranoside (45) Molecules 25 06052 i045Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
80% ethanol extract of Morella nana (A. Chev.) J. Herb. roots [81]
Acerogenin 2-methyl ether (46) Molecules 25 06052 i04680% ethanol extract of Morella nana A. Chev.) J. Herb. roots [81]
Myricananone (47) Molecules 25 06052 i04795% EtOH extract of Morella nana A. Chev.) J. Herb. roots [21]
Myricananadiol (48) Molecules 25 06052 i04895% EtOH extract of Morella nana A. Chev.) J. Herb. roots [21]
Myricanol 11-O-β-D-xylopyranosyl (49) Molecules 25 06052 i049Methanol extract of Morella adenophora (Hance) J. Herb. roots [19] Dichloromethane: methanol (1:1) extract in of Morella arborea (Hutch.) Cheek bark and stem [29]
Myricatomentoside I (50) Molecules 25 06052 i050Methanol extract of Myrica gale L. (Myrica gale var tormentosa L.) branches [35]
Myricatomentoside II (51) Molecules 25 06052 i051Methanol extract of Myrica gale L. (Myrica gale var tormentosa L.) branches [35]
12-Dehydroporson (52) Molecules 25 06052 i052Methanol extract of Myrica gale L. (Myrica gale var tormentosa L.) branches [24]
Myricarborin (53) Molecules 25 06052 i053Dichloromethane: methanol (1:1) extract of Morella arborea (Hutch.) Cheek bark and stem [29]
Myricanene A 5-O-α-L-arabinofuranosyl (1→6)-β-D-glucopyranoside (54) Molecules 25 06052 i054Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
Myresculoside (55) Molecules 25 06052 i055Methanol extract of Myrica esculenta Buch.-Ham. ex D. Don leaves [14]
(1S,2S,4R)-2-Hydroxy-1,8-cineole β-D-glucopyranoside (56) Molecules 25 06052 i056Methanol extract of Myrica esculenta Buch.-Ham. ex D. Don leaves [14]
Taraxerol (57) Molecules 25 06052 i057Methanol extract of Morella adenophora (Hance) J. Herb. roots [19]
Chloroform and methanol extract of Morella arborea (Hutch.) Cheek twigs [20]
Hexane extract of Morella cerifera (L.) Small twigs [23]
Benzene extract of Myrica gale L. (Myrica gale var tormentosa L.) stems [82]
Benzene extract of Morella cerifera (L.) Small (Myrica cerifera) root bark [42]
Extract of the bark of Myrica esculenta Buch.-Ham. ex D. Don [83]
Taraxerone (58) Molecules 25 06052 i058Chloroform and methanol extract of Morella arborea (Hutch.) Cheek twigs [20]
Benzene extract of Morella cerifera (L.) Small (Myrica cerifera) root bark [42]
Myricadiol (59) Molecules 25 06052 i059Chloroform and methanol extract of Morella arborea (Hutch.) Cheek twigs [20]
Benzene extract of Myrica gale L. (Myrica gale var tormentosa L.) stems [82]
Hexane extract of Morella cerifera (L.) Small twigs [23]
Alphitolic acid (60) Molecules 25 06052 i060Chloroform and methanol extract of Morella arborea (Hutch.) Cheek twigs [20]
Maslinic acid (61) Molecules 25 06052 i061Chloroform and methanol extract of Morella arborea (Hutch.) Cheek twigs [20]
Myriceric acid C (62) Molecules 25 06052 i062Methanol extract of Morella cerifera (L.) Small (Myrica cerifera) twigs [60]
Myriceric acid B (63) Molecules 25 06052 i063Methanol extract of Morella cerifera (L.) Small (Myrica cerifera) twigs [60]
Myriceric acid D (64) Molecules 25 06052 i064Methanol extract of Morella cerifera (L.) Small (Myrica cerifera) twigs [60]
Serratenedione (65) Molecules 25 06052 i065Benzene extract of Myrica gale L. (Myrica gale var tormentosa L.) stems [82]
Serratenediol (66) Molecules 25 06052 i066Benzene extract of Myrica gale L. (Myrica gale var tormentosa L.) stems [82]
Myricolal (67) Molecules 25 06052 i067Benzene extract of Myrica gale L. (Myrica gale var tormentosa L.) stems [82]
Arjunglucoside (68) Molecules 25 06052 i068Methanol extract of Myrica esculenta Buch.-Ham. ex D. Don leaves [14]
3-epi-Ursolic acid (69) ** Molecules 25 06052 i069Methanol extract of Myrica esculenta Buch.-Ham. ex D. Don leaves [14]
3-O-(E)-Caffeoyl-ursolic acid (70) ** Molecules 25 06052 i070Methanol extract of Myrica esculenta Buch.-Ham. ex D. Don leaves [14]
* The stereochemistry was defined by Jahanban-Esfahlan et al. [84]; ** The name was corrected based in the structure presented in the original paper since ursonic acid has a ketone group at C-3; L-Araf = L-arabinofuranosyl; D-Glcpy = D-glucopyranoside; Xyl = D-xylopyranosyl; Galloyl = 3,4,5-trihydroxybenzoyl; Api = −β-D-Apiofuranosyl; Caffeoyl = 3-hydroxycoumaroyl.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rosa, G.P.; Silva, B.J.C.; Seca, A.M.L.; Moujir, L.M.; Barreto, M.C. Phytochemicals with Added Value from Morella and Myrica Species. Molecules 2020, 25, 6052. https://doi.org/10.3390/molecules25246052

AMA Style

Rosa GP, Silva BJC, Seca AML, Moujir LM, Barreto MC. Phytochemicals with Added Value from Morella and Myrica Species. Molecules. 2020; 25(24):6052. https://doi.org/10.3390/molecules25246052

Chicago/Turabian Style

Rosa, Gonçalo P., Bruno J. C. Silva, Ana M. L. Seca, Laila M. Moujir, and Maria Carmo Barreto. 2020. "Phytochemicals with Added Value from Morella and Myrica Species" Molecules 25, no. 24: 6052. https://doi.org/10.3390/molecules25246052

Article Metrics

Back to TopTop