Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Aug 1.
Published in final edited form as: Phytother Res. 2016 May 24;30(8):1287–1297. doi: 10.1002/ptr.5648

Therapeutic Potential of Polyphenols from Epilobium angustifolium (Fireweed)

Igor A Schepetkin 1, Andrew G Ramstead 1, Liliya N Kirpotina 1, Jovanka M Voyich 1, Mark A Jutila 1, Mark T Quinn 1
PMCID: PMC5045895  NIHMSID: NIHMS818633  PMID: 27215200

Abstract

Epilobium angustifolium is a medicinal plant used around the world in traditional medicine for the treatment of many disorders and ailments. Experimental studies have demonstrated that Epilobium extracts possess a broad range of pharmacological and therapeutic effects, including antioxidant, anti-proliferative, anti-inflammatory, antibacterial, and anti-aging properties. Flavonoids and ellagitannins, such as oenothein B, are among the compounds considered to be the primary biologically active components in Epilobium extracts. In this review, we focus on the biological properties and the potential clinical usefulness of oenothein B, flavonoids, and other polyphenols derived from E. angustifolium. Understanding the biochemical properties and therapeutic effects of polyphenols present in E. angustifolium extracts will benefit further development of therapeutic treatments from this plant.

Keywords: Epilobium angustifolium, polyphenol, ellagitannin, oenothein B

INTRODUCTION

Polyphenols are a structural class of organic chemicals characterized by the presence of more than one phenolic ring. The number and characteristics of these phenol structures influences the physical, chemical, and biological properties of various classes of these compounds, which include flavonoids, phenolic acids, lignans, coumarins, stilbenes, and tannins (Quideau et al., 2011). Polyphenols are mainly natural but can also be also synthetic or semisynthetic. Plant-derived polyphenols exhibit beneficial effects on human health because of their anti-inflammatory, anti-allergic, anti-atherogenic, anti-microbial, anti-viral, anti-proliferative, and immunomodulatory properties (Feldman, 2005; Okuda, 2005; Holderness et al., 2008; Stagos et al., 2012; Gollucke et al., 2013; Korkina et al., 2013; Chirumbolo, 2014; Ratz-Lyko et al., 2015). The ability of natural polyphenols to modulate certain immune responses may explain, in part, some of beneficial effects of various medicinal plants. In addition, polyphenols exhibit antioxidant properties because of their ability to scavenge reactive oxygen species (ROS) and chelate metal ions (Rice-Evans et al., 1995; Rice-evans et al., 1996). Polyphenols isolated from various medicinal plants play an important role in the prevention and therapy of a variety of ailments and chronic diseases, and the study of polyphenols has become an increasingly important area of human nutrition research (Landete, 2012; Kishimoto et al., 2013). For example, some dietary polyphenols have been reported to influence the colonic flora via prebiotic effects (Landete, 2012). Polyphenols have also been shown to modulate the immune system by rapidly inducing lymphocyte gene transcription, leading to cytokine production and increased responsiveness to secondary signals (Holderness et al., 2007; Holderness et al., 2008). In vivo studies have demonstrated the lifespan-extending properties of polyphenol-containing plants (Uysal et al., 2013), and certain polyphenols may protect against Alzheimer's disease-type cognitive deterioration and neurodegeneration during brain aging and dementia (Pasinetti, 2012; Schaffer et al., 2012). Recent reports also indicate strong epigenetic effects of polyphenols (Joven et al., 2013; Ayissi et al., 2014).

Among the Epilobium species, E. (Chamerion) angustifolium is one of the best known medicinal plants and has been used worldwide in traditional medicine. It is also commonly known as fireweed or rosebay willow-herb. Extracts obtained from fireweed are known in folk medicine to exhibit a variety of pharmacological effects (Vitalone et al., 2001; Vitalone et al., 2003a; Vitalone et al., 2003b). Based on the importance of E. angustifolium in traditional medicine and the potential for therapeutic development of its constituents in modern medicine, it is important to understand the composition and pharmacological properties of E. angustifolium extracts. Since polyphenols are among the most abundant medically active constituents, we have focused this review on the contribution of these compounds to the pharmacological properties of E. angustifolium extracts and medicinal preparations.

PHARMACOLOGICAL EFFECTS OF EPILOBIUM EXTRACTS

Therapeutic properties of Epilobium extracts have been described in various pharmacological studies. Traditional use of fireweed includes an infusion or tea, which has been reported as a treatment for migraine headaches, insomnia, anemia, delirium tremens, infections, and colds. E. angustifolium extracts have been reported to be effective treatments for gastric ulcer; duodenal ulcer; gastritis; colitis; various gastrointestinal disorders, such as dysentery and diarrhea; and prostate or urinary problems, such as urethral inflammation, micturition disorders, prostatic adenoma, and benign prostatic hyperplasia (BPH) (Vitalone et al., 2001; Vitalone et al., 2003a; Vitalone et al., 2003b). E. angustifolium has also been used topically as a cleansing, soothing, antiseptic, and healing agent to treat minor burns, skin rashes, ulcers, and infections, and for treatment of inflammation of the ear, nose, and throat (Vogl et al., 2013). Experimentally, fireweed extracts have been reported to exhibit analgesic properties using hot plate and writhing tests (Tita et al., 2001). Aqueous extracts of the herb have also been reported to have anti-inflammatory properties and reduced carrageenan-induced paw edema (Hiermann et al., 1986; Juan et al., 1988). Extracts of E. angustifolium also have been shown to have bactericidal and antifungal effects (Moskalenko, 1986; Jones et al., 2000; Rauha et al., 2000; Battinelli et al., 2001; Webster et al., 2008; Bartfay et al., 2012; Kosalec et al., 2013). Moreover, administration of E. angustifolium extracts prior to influenza virus exposure reduced mortality and increased survival mean time (SMT). These effects were even more striking when infection occurred seven days after the last administration of the extract, where mortality rate was reduced by 50% and SMT was increased ~5 fold (Vila et al., 1989).

E. angustifolium extracts have also been reported to exhibit anti-tumor properties, including inhibition of human prostate epithelial cell PZ-HPV-7 growth (Vitalone et al., 2001). Likewise, treatment of androgen-sensitive human prostate adenocarcinoma cells LNCaP with Epilobium extracts (20–70 μg/ml) resulted in a significant increase in the number of apoptotic cells (Stolarczyk et al., 2013a). Various Epilobium extracts, including extracts from E. angustifolium, caused a similar inhibitory effect on the proliferation of human cancer cell lines and inhibited DNA synthesis in human astrocytoma cells 1321N1 (Vitalone et al., 2003b). In addition, E. angustifolium aqueous extracts (Kiss et al., 2006b; Kiss et al., 2006a) demonstrated higher anti-proliferative activity than ethanol extracts (Vitalone et al., 2003a; Vitalone et al., 2003b). E. hirsutum extracts also exhibited antitumor properties in a mouse model of leukemia (P388 cells) and ascites tumor (Voynova et al., 1991), and small doses of this extract (1–3 mg/kg) prolonged the lifespan of mice with tumors over 150% (Voynova et al., 1991).

Epilobium extracts may also exhibit anti-aging properties, and treatment of human dermal fibroblasts with 10 μg/ml extract down-regulated UV-induced release of matrix metalloproteinase-1 and -3, tissue inhibitor of matrix metalloproteinases-1 and -2, and hyaluronidase 2 gene expression (Ruszova et al., 2013). The authors suggested that polyphenols might account for these benefits. In addition, Epilobium extracts have high antioxidant activity, which is comparable to that of well-known antioxidants and flavonoids (Hevesi Tóth et al., 2009). Indeed, aqueous extracts of E. angustifolium are able to scavenge superoxide anion (O2−.) and hydroxyl radicals, as well as inhibit ROS production by stimulated neutrophils (Myagmar,Aniya, 2000).

POLYPHENOLIC COMPOUNDS

Over 100 compounds have been identified in different materials from the Epilobium genus [reviewed in (Granica et al., 2014)], with polyphenols being the predominant constituent. Indeed, it is thought that polyphenols of E. angustifolium could explain, at least in part, many of the therapeutic and beneficial properties of this plant because of the known immunomodulatory properties of polyphenols (Holderness et al., 2007; Ramiro-Puig et al., 2008; Schepetkin et al., 2009; Daughenbaugh et al., 2011; Holderness et al., 2011; Skyberg et al., 2011; Ramstead et al., 2012; Ramstead et al., 2015).

High concentrations of polyphenols are present in members of the genus Epilobium L. (Onagraceae), which consists of over 200 species found worldwide. Secondary metabolites have been characterized in approximately 25% of the species from this genus, and flavonoids and tannins have been found to be the principal bioactive constituents in E. hirsutum L., E. parviflorum Schreb., E. montanum L., E. tetragonum L., E. roseum L., E. adenocaulon Hausskn., E. palustre L., and E. angustifolium L. (Ivancheva et al., 1992; Lesuisse et al., 1996; Kiss et al., 2006a; Hevesi Tóth et al., 2009; Schepetkin et al., 2009; Kiss et al., 2011; Jurgenson et al., 2012; Remmel et al., 2012; Ruszova et al., 2013). In fact, the content of oenothein B and quercetin-3-O-glucuronide has been suggested as a basis for the standardization of commercially available Epilobium products (Bazylko et al., 2007; Hevesi Tóth et al., 2009; Granica et al., 2012; Monschein et al., 2015).

E. angustifolium contains a variety of polyphenols (Jurgenson et al., 2012). Phytochemical analyses of E. angustifolium extracts have identified three major polyphenol groups: flavonoids, phenolic acids, and ellagitannins (Ducrey et al., 1997; Shikov et al., 2006; Remmel et al., 2012; Ruszova et al., 2013). Flavonoids include flavonol aglycones (quercetin, kaempferol, myricetin) and flavonoid glycosides, such as afzelin (kaempferol-3-O-rhamnoside), juglalin (kaempferol-3-O-arabinofuranoside), avicularin (quercetin-3-O-α-arabinofuranoside), hyperoside (quercetin-3-O-galactoside), isoquercetin (quercetin-3-O-glucoside), quercitrin (quercetin-3-O-rhamnoside), and miquelianin (quercetin-3-O-glucuronide) (Table 1). Among the flavonoid glycosides that have been identified in Epilobium species, miquelianin is a main flavonoid in E. angustifolium, whereas myricitrin (myricetin-3-O-rhamnoside) was found to be the main flavonoid in other species (Hevesi Tóth et al., 2009). Some of these compounds are active constituents of many medicinal plants that are used in traditional medicines for their neuroprotective, anti-inflammatory, antioxidant, anti-proliferative, and other pharmacological properties (Table 2). Miquelianin is a major flavonoid glycoside from E. angustifolium. The activity of miquelianin seems to be primarily due to its bioactive metabolites (Jimenez et al., 2015; Messer et al., 2015) (Table 2).

Table 1.

Chemical structures of selected flavonoids and their glycosides found in E. angustifolium (Hiermann et al., 1991; Ducrey et al., 1995; Hevesi Tóth et al., 2009).

graphic file with name nihms818633u1.jpg
Compound Name R1 R2 R3
1 Kaempferol H H H
2 Quercetin H OH H
3 Myricetin H OH OH
4 Afzelin (kaempferol-3-O-rhamnoside) Rha H H
5 Quercitrin (quercetin-3-O-rhamnoside) Rha OH H
6 Myricetin-3-O-rhamnoside Rha OH OH
7 Juglalin (kaempferol-3-O-arabinofuranoside) Ara H H
8 Guajaverin (quercetin 3-O-arabinopyranside) Ara OH H
9 Hyperoside (quercetin-3-O-galactoside) Gal OH H
10 Isoquercetin (quercetin-3-O-glucoside) Glc OH H
11 Isomyricitrin (myricetin-3-O-glucoside) Glc OH OH
12 Quercetin-3-O-(6”-galloyl)-galactoside (6”-Gall)Gal OH H
13 Miquelianin (quercetin-3-O-glucuronide) GlcA OH H
14 Kaempferol-3-O-β-glucuronide GlcA H H
15 Myricetin-3-O-glucuronide GlcA OH OH
16 Myricetin-3-O-galactoside Gal OH OH

Table 2.

Biological properties of selected flavonoids found in E. angustifolium.

Compound Biological properties References
Afzelin (kaempferol-3-O-rhamnoside) Antibacterial, DNA-protective, antioxidant, anti- inflammatory, anti-complement activity, inhibitor of angiotensin converting enzyme (ACE) (Hansen et al., 1996; Min et al., 2003; Shin et al., 2013; Lee et al., 2014)
Hyperoside (quercetin-3-O-galactoside) Suppresses vascular inflammatory, anti-apoptotic and antithrombotic activity, relieves pain and improves cardiovascular functions, neuroprotective, inhibits cytochrome P450 activity (Zeng et al., 2011; Liu et al., 2012; Ku et al., 2013; Song et al., 2013; Zhang et al., 2013)
Isoquercetin (quercetin-3-O-glucoside) Antidiabetic, anti-inflammatory, antiviral, neuroprotective, antioxidative, inhibitor of α-glucosidase (Li et al., 2009; Nguyen et al., 2009; Kim et al., 2010; Zhang et al., 2011; Thapa et al., 2012; Wang et al., 2013)
Kaempferol Antioxidative, anti-inflammatory, anti-proliferative, antimicrobial, cardioprotective, neuroprotective (Khlebnikov et al., 2007; Calderon-Montano et al., 2011; Chen,Chen, 2013; Rajendran et al., 2014)
Miquelianin (quercetin-3-O-glucuronide) Immunostimulatory and anti-inflammatory; ameliorates insulin resistance in skeletal cells under inflammatory conditions; suppresses plasmin-mediated mechanisms of cancer cell migration (Al-Shalmani et al., 2011; Cuccioloni et al., 2012; Liao,Lin, 2014; Liu et al., 2016)
Myricetin Quercetin Antioxidant, anti-inflammatory, antimicrobial, anti- proliferative, anti-aging Anti-proliferative, antioxidative, neuroprotective, and anti-inflammatory, pleiotropic kinase inhibitor, inhibitor of α-glucosidase (Cushnie,Lamb, 2005; Leonarduzzi et al., 2010; Ruzic et al., 2010) (Li et al., 2009; Dajas, 2012; Bruning, 2013; Furst,Zundorf, 2014; Russo et al., 2014; Sak, 2014)
Quercitrin (quercetin-3-O-rhamnoside) Promotes osteoblast differentiation and inhibits osteoclastogenesis, antioxidative, antileishmanial activity, inhibitor of aldose reductase, p90S6 ribosomal kinase (RSK), AP-1 and MAPK signaling, protects mice against fatal anaphylactic shock (Khlebnikov et al., 2007; Cruz et al., 2008; Ding et al., 2010; da Silva et al., 2012; Derewenda et al., 2013; Kim et al., 2013; Satue et al., 2013; Yin et al., 2013)

Phenolic acids identified in Epilobium species are gallic acid (3,4,5-trihydroxybenzoic acid) and its methyl ester, protocatechuic acid (3,4-dihydroxybenzoic acid), ellagic acid, octyl gallate, 5-O-caffeoylquinic acid, 6-O-galloyl-glucose, 1,2,6-O-trigalloyl glucose, and 1,2,3,4,6-O-pentagalloyl glucose (Hiermann et al., 1991; Ducrey et al., 1995; Kiss et al., 2004; Shikov et al., 2006; Bazylko et al., 2007; Hevesi Tóth et al., 2009; Kiss et al., 2011; Stolarczyk et al., 2013b; Karakurt et al., 2016).

Among the relatively high-molecular weight polyphenols identified are tellimagrandin I-based oligomeric ellagitannins (Ducrey et al., 1997; Shikov et al., 2006; Bazylko et al., 2007; Yoshida et al., 2010; Baert et al., 2015; Kaskoniene et al., 2015b). Oenothein B is comprised of two tellimagrandin I monomers linked between the hexahydroxydiphenoyl groups and the galloyl groups on the glucopyranose ring (Figure 1). Several other oligomeric tannins have also been isolated from E. angustifolium extracts, including oenothein A (trimer), and tellimagrandin I-based heptameric ellagitannins (Sasov et al., 2010; Baert et al., 2015). Overall, oenothein B is the predominant polyphenol in this plant (14–23%), while flavonoid content was < 2% (Ducrey et al., 1997; Kiss et al., 2011).

Figure 1.

Figure 1

Chemical structure of oenothein B.

The majority of plant polyphenols occur in the form of biologically unavailable polymers or glycosides, which are degraded to low molecular weight compounds by intestinal enzymes originating from the host organism or secreted by colonic microflora (D'Archivio et al., 2010). Since the biological activity of many of these polyphenols has been established using cell cultures in vitro, interpretation of their potential pharmacological effects in vivo could be problematic, and further evaluation using in vivo studies is warranted. In addition, the activity of polyphenolic compounds can even be influenced by the routes of in vivo administration, as the active metabolites generated in vivo can differ based on route of administration. For example, neither hyperoside nor its metabolites were detected in rat brain after intragastric administration, whereas both compounds were detected after intraperitoneal administration (Guo et al., 2012).

OENOTHIEN B

Oenothein B is a macrocyclic ellagitannin (Hatano et al., 1990). It is a major contributor to the biological activity of Epilobium extracts and is present at high concentration in these species (Lesuisse et al., 1996; Kiss et al., 2006b; Kiss et al., 2006a; Kiss et al., 2011). Biological activities of oenothein B include antioxidant, immunomodulatory, tumor cell cytotoxicity, enzyme inhibition, and enzyme induction (Miyamoto et al., 1993a; Miyamoto et al., 1993b; Aoki et al., 1995; Lesuisse et al., 1996; Sakagami et al., 2000; Kiss et al., 2006b; Kiss et al., 2006a; Kiss et al., 2011; Stolarczyk et al., 2013b) (Table 3). Much of the research on oenothein B has focused on its effects on abnormal prostate cells, where it inhibits cell proliferation, prostate specific antigen (PSA) secretion, and arginase activity (Stolarczyk et al., 2013b). Oenothein B is an inhibitor of the enzyme 5-α-reductase, which is an important target enzyme in certain prostate disorders (Lesuisse et al., 1996). In addition, oenothein B induced neutral endopeptidase activity in a prostate tumor cell line, thereby inhibiting cell proliferation (Kiss et al., 2006b; Kiss et al., 2006a). Therefore, supplements containing oenothein B may benefit for individuals with prostate disorders, including prostate cancer, through the modulation of prostate enzyme activity. Oenothein B was also recently reported to be an α-glucosidase inhibitor (Kawakami et al., 2014).

Table 3.

Biological properties of oenothein B

Assay Biological properties References
In vitro Anti-proliferative in human neuroblastoma SK-N-SK cells, human prostate tumor cell line PC-3, and human prostate adenocarcinoma LNCaP cells (Kiss et al., 2006b; Kiss et al., 2006a; Stolarczyk et al., 2013b)
Reduces prostate specific antigen (PSA) secretion in LNCaP cells (Stolarczyk et al., 2013b)
Inhibits arginase activity in LNCaP cells (Stolarczyk et al., 2013b)
Inhibits α-glucosidase (Kawakami et al., 2014)
Stimulates release of IL-1β from macrophages (Miyamoto et al., 1993a; Miyamoto et al., 1993b)
Stimulate Ca2+ flux and ROS production in neutrophils (Schepetkin et al., 2009)
Stimulated NF-κB activation and production of TNF and IL-6 in human monocytic THP-1 cells (Schepetkin et al., 2009)
Activates NK cells, αβ T cells, and γδ T cells, resulting in increased expression of the activation marker CD69 and IFNγ production (Ramstead et al., 2012)
Induces more IFNγ production by CD45RO+ compared to naïve T cells memory T cells (Ramstead et al., 2015)
Scavenger of ROS (O2 and H2O2) (Schepetkin et al., 2009; Kiss et al., 2011; Granica et al., 2015)
Inhibits mouse mammary tumor virus transcription in 34I cells (Aoki et al., 1995)
Antibacterial activity against Helicobacter pylori (Funatogawa et al., 2004).
In vivo Inhibits growth of MM2 ascites tumors and Meth-A solid type tumor in mice (Miyamoto et al., 1993a; Miyamoto et al., 1993b)
Induces recruitment of neutrophils in peritoneal cavity, which is associated with induction of KC production (Schepetkin et al., 2009)
Inhibits IL-1β and IL-6 production by activated dendritic cells and attenuates neuroinflammation in response to LPS treatment (Okuyama et al., 2013)

In addition to its effects on prostate cells, oenothein B was also found to inhibit tumor growth in vivo (Miyamoto et al., 1993a; Miyamoto et al., 1993b). Although oenothein B can induce apoptosis in tumor cell lines (Sakagami et al., 2000), the previously observed inhibition of tumor growth was not believed to be caused by direct cytotoxicity to the tumor cells. Instead, oenothein B was found to stimulate macrophages and promote the production of interleukin (IL) 1, and this was proposed to contribute to the observed antitumor effects of oenothein B in murine models (Miyamoto et al., 1993a; Miyamoto et al., 1993b). In support of this idea, we found that oenothein B activated both mouse and human neutrophils and monocytes/macrophages. Among the neutrophil responses enhanced by oenothein B were intracellular calcium flux and ROS production (Schepetkin et al., 2009). In addition, oenothein B stimulated monocyte nuclear factor NF-κB activation and pro-inflammatory cytokine production, including tumor necrosis factor (TNF) and IL-6 (Schepetkin et al., 2009), which may contribute to the antitumor effects. Note, however, that additional compounds besides oenothein B may contribute to the antitumor properties of Epilobium extracts (Vitalone et al., 2003b), possibly through synergistic mechanisms. While oenothein B was found to enhance neutrophil ROS production, this compound can also directly scavenge O2 and H2O2 (Schepetkin et al., 2009; Kiss et al., 2011; Granica et al., 2015). Indeed, oenothein B had the highest radical scavenging activity among other polyphenols in methanol extracts from E. angustifolium (Kaskoniene et al., 2015a). Antioxidant activity is one of main properties of tannins and has been reported for many other ellagitannins, such as agrimoniin, corilagin, punicalagin, punicalin, and pedunculagin (Lin et al., 2001; Chung et al., 2003; Marzouk et al., 2007; Bazylko et al., 2013). In addition, the metabolites of various ellagitannins may have even more potent antioxidant activity than their respective parent compounds (Ishimoto et al., 2012). Thus, combined enhancement of innate immune defenses and protection of host tissues through antioxidant effects could allow oenothein B to optimally provide therapeutic benefits.

In addition to effects on myeloid cells and neutrophils, we have also shown that oenothein B activates lymphocytes, including NK cells, αβ T cells, and γδ T cells, resulting in increased expression of the activation marker CD69 (Ramstead et al., 2012). It should be noted that the effect of oenothein B on CD69 expression in γδ T cells was similar to the immunomodulatory properties of condensed tannins isolated from Uncaria tomentosa (Cat’s Claw) and Malus domestica (apple) (Holderness et al., 2007). Treatment with oenothein B also enhanced interferon γ (IFNγ) production by αβ T cells, γδ T cells, and NK cells in response to secondary stimuli, including IL-18 and a tumor cell line (Ramstead et al., 2012). Oenothein B activated T cells from both young and adult individuals, although higher levels of IFNγ were produced by T cells from adults compared to those from young individuals after oenothein B treatment (Ramstead et al., 2015). Furthermore, oenothein B induced more IFNγ production by CD45RO+ memory T cells compared to naïve T cells (Ramstead et al., 2015). Thus, it is clear that oenothein B is a potent immune cell agonist and can enhance the activity of various types of immune cells.

In contrast to our studies, others have reported anti-inflammatory effects of oenothein B. For example, Kiss et al. (Kiss et al., 2011) reported that myeloperoxidase release and production of ROS by activated neutrophils were inhibited by oenothein B. Likewise, oenothein B has been reported to inhibit nitric oxide production, NF-κB activity, and the production of IL-1β, IL-6, and TNF by a murine macrophage cell line pretreated with Toll-like receptor (TLR) 2 and TLR4 agonists (Schmid et al., 2012). A related ellagitannin, punicalagin, was also found to inhibit TLR4-mediated NF-κB signaling pathways (Peng et al., 2015). Moreover, oenothein B inhibited IL-1β and IL-6 production by activated dendritic cells and inhibited neuroinflammation in response to systemic lipopolysaccharide treatment (Okuyama et al., 2013; Yoshimura et al., 2013). Therefore, it appears that oenothein B has a complex influence on innate immune cells, which is similar to what has been observed for other ellagitannins. One possible explanation for the discrepancies in these data is the activation state of the cells at the time of treatment with oenothein B. For much of our research demonstrating activation of immune cells, unstimulated, resting cells were used (Schepetkin et al., 2009; Ramstead et al., 2012). However, in the studies demonstrating immune suppression by oenothein B, the authors used pre-stimulated, activated cells (Kiss et al., 2011; Schmid et al., 2012; Yoshimura et al., 2013). These data suggest that oenothein B may have differential effects on activated and resting immune cells, suppressing activated cells and stimulating resting ones. Additional research is needed to better understand the complex effects of oenothein B on immunity and how these effects contribute to the proposed health benefits associated with oenothein B and E. angustifolium extracts.

One factor that appears to be important for stimulation of T cell cytokine production by polyphenols is the size of the polyphenol molecule. Indeed, molecular subunits of oenothein B with smaller molecular weights do not have the same leukocyte immunomodulatory capacity (Schepetkin et al., 2009; Granica et al., 2015). Similar observations were made by Yamanaka et al. (Yamanaka et al., 2012), who found that stimulation of murine splenocytes by polymerized polyphenols with large molecular weights, but not their corresponding monomers, enhanced T cell cytokine production. Furthermore, our research has found that procyanidin oligomers, but not monomers, stimulate innate lymphocytes (Holderness et al., 2008). The importance of molecular size is consistent with the activity of oenothein B, as it is a large polyphenol (Schepetkin et al., 2009).

In addition to immunomodulatory effects, oenothein B has been reported to exhibit direct antimicrobial and antiviral activity. For example, this compound has been reported to have antibacterial activity against Helicobacter pylori (Funatogawa et al., 2004). Likewise, we found that oenothein B directly inhibited Staphylococcus aureus growth in vitro with an IC50 of ~0.7 μM (Figure 2A) and also enhanced S. aureus killing by human neutrophils (Figure 2B). The ability of S. aureus to survive after neutrophil phagocytosis is thought to contribute significantly to the relative virulence of this pathogen (Voyich et al., 2005; Voyich et al., 2006; Bubeck Wardenburg et al., 2007; Wang et al., 2007; Voyich et al., 2009). This is exemplified by the observed increase in susceptibility to S. aureus infections of individuals suffering from defects that alter normal neutrophil function, such as chronic granulomatous disease, leukocyte adhesion deficiency, and neutropenia (Bodey et al., 1966; Dale et al., 1979; Lekstrom-Himes,Gallin, 2000). Therefore, our finding that oenothein B significantly enhanced staphylocidal activity of human neutrophils is promising, and future studies will investigate the therapeutic potential of oenothein B in vivo and in combination with antibiotics to see if bacterial clearance can be improved by this compound.

Figure 2.

Figure 2

Effect of oenothein B on Staphylococcus aureus growth and human neutrophil staphylocidal activity. Panel A. Direct effect of oenothein B on growth of Staphylococcus aureus. S. aureus USA300 (2 x 106 in 200 μl of RPMI) were mixed with the indicated concentrations of oenothein B in microplate wells and incubated at 37°C for 3 hours. Oenothein B was isolated from E. angustifolium, as described previously (Schepetkin et al., 2009). Bacterial growth was monitored spectrophotometrically at 600 nm using a SpectraMax Plus microplate reader. Background measurements of the oenothein B dilutions in RPMI alone were subtracted from each OD600 reading, and % growth was calculated as: (OD600 of oenothein B-treated S. aureus/OD600 of control, untreated S. aureus in RPMI)× 100. We subtracted these values from 100 to plot % growth inhibition resulting from the oenothein B concentrations tested. Values are the mean ± S.D. of triplicate samples from one experiment, which is representative of three independent experiments. Panel B. Effect of oenothein B on human neutrophil bactericidal activity. Human neutrophils were purified from the blood of healthy donors (in accordance with a protocol approved by the Institutional Review Board at Montana State University) using dextran sedimentation, followed by Histopaque 1077 (Sigma-Aldrich) gradient separation and hypotonic lysis of red blood cells, as previously described (Schepetkin et al., 2011). The neutrophils were pre-incubated with the indicated concentrations of oenothein B or control Hank’s balanced-salt solution (HBSS) for 15 min, washed to remove remaining oenothein B, and incubated with opsonized S. aureus USA300 at a 1:10 ratio of neutrophils to bacteria (106 neutrophils and 107 bacteria in 200 μl of RPMI). After 3 hours incubation, neutrophils were lysed with 0.1% saponin (Sigma-Aldrich) for 5 min at 4°C, and the total bacterial CFUs were determined by plating the mixture on tryptic soy agar for 18 hours at 37°C and then counting colonies. CFU values are presented as the % of bacteria remaining after incubation with control HBSS pre-treated neutrophils (mean ± S.D. of duplicate samples from one experiment, which is representative of three independent experiments). Statistically significant differences (* p<0.05; ** p<0.01) between HBBS-treated neutrophils and cells treated with oenothein B are indicated.

Oenothein B has also been reported to inhibit mouse mammary tumor virus (MMTV) transcription (Aoki et al., 1995), which was believed to be due to inhibition of poly(ADP-ribose) glycohydrolase. In addition to MMTV, oenothein B also inhibited herpes simplex virus, which is similar to other tannins, including coriariin A, rugosin D, cornusiin A, tellimagrandin I, casuarictin, and 1,2,3,4,6-penta-O-galloyl-β-D-glucopyranose (Fukuchi et al., 1989; Kim et al., 2001). Since tellimagrandin I also has direct antibacterial activity (Funatogawa et al., 2004; Shiota et al., 2004), it is possible that some antiviral and bactericidal activities of oenothein B could be related to its tellimagrandin I substructures. Finally, oenothein B has been suggested to have antifungal activity based on its inhibition of 1,3-β-glucan synthase transcription in Paracoccidioides brasiliensis (Santos et al., 2007; Zambuzzi-Carvalho et al., 2013). Together, these reports suggest that oenothein B may be beneficial during certain bacterial, viral, and fungal infections.

Not much is known regarding the cellular binding properties of oenothein B. We found that serum levels of IL-6 in TLR4 knockout mice after intraperitoneal injection of oenothein B were similar to those in TLR2 knockout and wild-type mice. These data suggest that oenothein B binding could be mediated via TLR2/TLR4-independent signaling pathways. Recent studies have demonstrated that the binding of ellagitannins, including oenothein B, to albumin increases in strength and affinity for the larger tannins (dimers) compared to their monomer forms and that bond rotational flexibility of oenothein B also plays a role by increasing the strength of interaction and number of stronger (possibly hydrogen bonding) binding sites on the protein surface (Dobreva et al., 2014). Clearly, further studies are necessary to elucidate the cellular targets of oenothein B, especially in relation to immune cell regulation.

Similar to tannins and other polyphenols (Yoshioka et al., 2001; Mira et al., 2002; Andrade et al., 2005), oenothein B has strong metal-chelating properties. Recently Tahara et al. (Tahara et al., 2014) reported that oenothein B binds Al(III) ions and suggested that formation of Al(III) complexes with oenothein B in roots could contribute to high aluminum resistance of E. camaldulensis (Tahara et al., 2014). We found that oenothein B was also able to chelate Cu(II) ions, with a 1:1 stoichiometry for the soluble complex, whereas addition of excess Cu(II) initiated the formation of insoluble precipitates (Figure 3). Previously, it has been reported that Cu(II) complexes of several other polyphenols altered their biological properties (Yoshioka et al., 2001; Mira et al., 2002; Yu et al., 2005). Thus, it is possible that oenothein B-copper complexes could have altered antioxidant or other biological properties compared with native oenothein B.

Figure 3.

Figure 3

Binding of Cu(II) to oenothein B. Oenothein B (20 μM), isolated from E. angustifolium as described previously (Schepetkin et al., 2009), was mixed with increasing concentrations of CuCl2 (Sigma-Aldrich) in 20 mM phosphate buffer (pH 7.5) and spectra of the Cu(II)-oenothein complex were obtained using a SpectraMax Plus microplate reader. The spectra of the complex revealed absorbance peaks at 244 and 322 nm and two isobestic points at 235 and 280 nm. Inset: The increase in Cu(II) concentration caused a linear increase in the absorbance at 244 nm, and saturation occurred at 20 μM Cu(II), suggesting that oenothein B is able to chelate Cu(II) ions, with a 1:1 stoichiometry for the soluble complex. The spectra shown are representative of three independent determinations.

Clear data regarding the bioavailability of ingested oenothein B are still missing. Although this dimeric ellagitannin is effective after oral administration (Okuda et al., 1989), it is still not clear if the active molecule(s) is the same as the parent. Indeed, most polyphenolic compounds undergo metabolic transformations, which significantly change their biological activities (Lewandowska et al., 2013; Tarko et al., 2013). For example, the primary products of acid degradation of oenothein B are gallic and ellagic acids, and recent in vitro and in vivo experiments have revealed that ellagic acid elicits, for example, antitumor effects by inhibiting tumor cell proliferation, inducing apoptosis, blocking virus infection, and disturbing inflammation, angiogenesis, and drug-resistance processes required for tumor growth and metastasis (Zhang et al., 2014). Gallic acid is one of most well-absorbed polyphenols (Manach et al., 2005) and has neuroprotective properties in different models of neurotoxicity, neurodegeneration, and oxidative stress (Daglia et al., 2014). Likewise, gallic acid has been shown to inhibit carcinogenesis in vitro by cancerous cell lines and in animal models (Carpentier et al., 1984). Since ellagic acid-derived metabolites produced by human colonic microflora are urolithins, biological effects of dibenzo[b,d]pyran-6-one should be also be considered (Larrosa et al., 2006; Piwowarski et al., 2014).

CLINICAL STUDIES

Various fireweed preparations have been developed for clinical use. For example, Chanerol is a complex polyphenolic medicinal drug prepared from the blossoms of fireweed (Pukhal'skaia et al., 1970; Petrova et al., 1974; Pukhalskaya et al., 1975). It is likely that oenothein B is one of main bioactive constituents of Chanerol and could be responsible for its pharmacological activities (Spiridonov et al., 1997; Sasov et al., 2010), including antitumor activity (Syrkin,Konyaeva, 1984). An aqueous extract of E. angustifolium was patented for use as an oral anti-inflammatory treatment (German Patent No. 3,605,250 of Jul. 16, 1987). In addition, skin care treatments containing E. angustifolium extract have been patented (WO2011007183) by a Canadian company, Fytokem (Saskatoon, Saskatchewan), that markets several different Willowherb™ extracts with anti-irritant effects. Fytokem claims that the principle bioactive molecules found in Willowherb™ extract are oenothein B and flavonols.

CONCLUSION

E. angustifolium (a.k.a. fireweed) is a medicinal plant widely used in traditional medicine. Extracts from this plant represent a rich source of biologically active polyphenols, such as oenothein B and its metabolites. These polyphenols are responsible for many of the biological responses that contribute to the therapeutic potential of fireweed extracts in a variety of diseases. The therapeutic effects of fireweed polyphenols are mediated by multiple mechanisms, including direct killing of cancer cells and microbes, antioxidant activity, metal chelation, and both pro- and anti-inflammatory immunomodulation. Although oenothein B is the predominant polyphenol in E. angustifolium responsible for many of its therapeutic properties, its putative receptor and downstream signal transduction pathways are not well understood and will require further research to elucidate. This is essential, as polyphenolic compounds can react with many protein targets. Certainly, a better understanding of fireweed's active molecules and their mechanisms of action is essential for maximizing the therapeutic potential of this interesting plant and ensuring safe use of these compounds as a therapeutics. Although fireweed extract and its components appear to be relatively safe, further clinical studies are also clearly necessary to assess potential adverse effects and interactions with other drugs, as is normally performed for conventional medicines (Izzo et al., 2016).

Acknowledgments

This research was supported in part by National Institutes of Health IDeA Program COBRE grant GM110732, a USDA National Institute of Food and Agriculture Hatch project, Montana University System Research Initiative: 51040-MUSRI2015-03, and the Montana State University Agricultural Experiment Station.

Footnotes

Conflict of Interest

There are no actual conflicts of interest for the authors.

References

  1. Al-Shalmani S, Suri S, Hughes DA, Kroon PA, Needs PW, Taylor MA, Tribolo S, Wilson VG. Quercetin and its principal metabolites, but not myricetin, oppose lipopolysaccharide-induced hyporesponsiveness of the porcine isolated coronary artery. Br J Pharmacol. 2011;162:1485–1497. doi: 10.1111/j.1476-5381.2010.00919.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Andrade RG, Jr, Dalvi LT, Silva JM, Jr, Lopes GK, Alonso A, Hermes-Lima M. The antioxidant effect of tannic acid on the in vitro copper-mediated formation of free radicals. Arch Biochem Biophys. 2005;437:1–9. doi: 10.1016/j.abb.2005.02.016. [DOI] [PubMed] [Google Scholar]
  3. Aoki K, Maruta H, Uchiumi F, Hatano T, Yoshida T, Tanuma S. A macrocircular ellagitannin, oenothein B, suppresses mouse mammary tumor gene expression via inhibition of poly(ADP-ribose) glycohydrolase. Biochem Biophys Res Commun. 1995;210:329–337. doi: 10.1006/bbrc.1995.1665. [DOI] [PubMed] [Google Scholar]
  4. Ayissi VBO, Ebrahimi A, Schluesenner H. Epigenetic effects of natural polyphenols: A focus on SIRT1-mediated mechanisms. Mol Nutr Food Res. 2014;58:22–32. doi: 10.1002/mnfr.201300195. [DOI] [PubMed] [Google Scholar]
  5. Baert N, Karonen M, Salminen JP. Isolation, characterisation and quantification of the main oligomeric macrocyclic ellagitannins in Epilobium angustifolium by ultra-high performance chromatography with diode array detection and electrospray tandem mass spectrometry. J Chromatogr A. 2015;1419:26–36. doi: 10.1016/j.chroma.2015.09.050. [DOI] [PubMed] [Google Scholar]
  6. Bartfay WJ, Bartfay E, Johnson JG. Gram-negative and gram-positive antibacterial properties of the whole plant extract of willow herb (Epilobium angustifolium) Biol Res Nurs. 2012;14:85–89. doi: 10.1177/1099800410393947. [DOI] [PubMed] [Google Scholar]
  7. Battinelli L, Tita B, Evandri MG, Mazzanti G. Antimicrobial activity of Epilobium spp. extracts. Farmaco. 2001;56:345–348. doi: 10.1016/s0014-827x(01)01047-3. [DOI] [PubMed] [Google Scholar]
  8. Bazylko A, Kiss AK, Kowalski J. High-performance thin-layer chromatography method for quantitative determination of oenothein B and quercetin glucuronide in aqueous extract of Epilobii angustifolii herba. J Chromatogr A. 2007;1173:146–150. doi: 10.1016/j.chroma.2007.10.019. [DOI] [PubMed] [Google Scholar]
  9. Bazylko A, Piwowarski JP, Filipek A, Bonarewicz J, Tomczyk M. In vitro antioxidant and anti-inflammatory activities of extracts from Potentilla recta and its main ellagitannin, agrimoniin. J Ethnopharmacol. 2013;149:222–227. doi: 10.1016/j.jep.2013.06.026. [DOI] [PubMed] [Google Scholar]
  10. Bodey GP, Buckley M, Sathe YS, Freireich EJ. Quantitative relationships between circulating leukocytes and infection in patients with acute leukemia. Ann Intern Med. 1966;64:328–340. doi: 10.7326/0003-4819-64-2-328. [DOI] [PubMed] [Google Scholar]
  11. Bruning A. Inhibition of mTOR Signaling by Quercetin in Cancer Treatment and Prevention. Anti-Cancer Agents in Med Chem. 2013;13:1025–1031. doi: 10.2174/18715206113139990114. [DOI] [PubMed] [Google Scholar]
  12. Bubeck Wardenburg J, Bae T, Otto M, Deleo FR, Schneewind O. Poring over pores: alpha-hemolysin and Panton-Valentine leukocidin in Staphylococcus aureus pneumonia. Nat Med. 2007;13:1405–1406. doi: 10.1038/nm1207-1405. [DOI] [PubMed] [Google Scholar]
  13. Calderon-Montano JM, Burgos-Moron E, Perez-Guerrero C, Lopez-Lazaro M. A review on the dietary flavonoid kaempferol. Mini Rev Med Chem. 2011;11:298–344. doi: 10.2174/138955711795305335. [DOI] [PubMed] [Google Scholar]
  14. Carpentier JL, Dayer J-M, Lang V, silvermann R, Orchi L, Gorden P. Down regulation and recycling of insulin receptors: Effect of monensin in IM-9 lymphocytes and U937 monocyte-like cells. J Biol Chem. 1984;259:14190–14195. [PubMed] [Google Scholar]
  15. Chen AY, Chen YC. A review of the dietary flavonoid, kaempferol on human health and cancer chemoprevention. Food Chem. 2013;138:2099–2107. doi: 10.1016/j.foodchem.2012.11.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Chirumbolo S. Dietary assumption of plant polyphenols and prevention of allergy. Curr Pharm Des. 2014;20:811–839. doi: 10.2174/13816128113199990042. [DOI] [PubMed] [Google Scholar]
  17. Chung SK, Nam JA, Jeon SY, Kim SI, Lee HJ, Chung TH, Song KS. A prolyl endopeptidase-inhibiting antioxidant from Phyllanthus ussurensis. Arch of Pharmacal Res. 2003;26:1024–1028. doi: 10.1007/BF02994753. [DOI] [PubMed] [Google Scholar]
  18. Cruz EA, Da-Silva SAG, Muzitano MF, Silva PMR, Costa SS, Rossi-Bergmann B. Immunomodulatory pretreatment with Kalanchoe pinnata extract and its quercitrin flavonoid effectively protects mice against fatal anaphylactic shock. Int Immunopharmacol. 2008;8:1616–1621. doi: 10.1016/j.intimp.2008.07.006. [DOI] [PubMed] [Google Scholar]
  19. Cuccioloni M, Bonfili L, Mozzicafreddo M, Cecarini V, Eleuteri AM, Angeletti M. Sanguisorba minor extract suppresses plasmin-mediated mechanisms of cancer cell migration. Biochim Biophys Acta. 2012;1820:1027–1034. doi: 10.1016/j.bbagen.2012.02.002. [DOI] [PubMed] [Google Scholar]
  20. Cushnie TP, Lamb AJ. Antimicrobial activity of flavonoids. Int J Antimicrob Agents. 2005;26:343–356. doi: 10.1016/j.ijantimicag.2005.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. D'Archivio M, Filesi C, Vari R, Scazzocchio B, Masella R. Bioavailability of the polyphenols: status and controversies. Int J Mol Sci. 2010;11:1321–1342. doi: 10.3390/ijms11041321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. da Silva ER, Maquiaveli CD, Magalhaes PP. The leishmanicidal flavonols quercetin and quercitrin target Leishmania (Leishmania) amazonensis arginase. Exp Parasitol. 2012;130:183–188. doi: 10.1016/j.exppara.2012.01.015. [DOI] [PubMed] [Google Scholar]
  23. Daglia M, Di Lorenzo A, Nabavi SF, Talas ZS, Nabavi SM. Polyphenols: well beyond the antioxidant capacity: gallic acid and related compounds as neuroprotective agents: you are what you eat! Curr Pharm Biotechnol. 2014;15:362–372. doi: 10.2174/138920101504140825120737. [DOI] [PubMed] [Google Scholar]
  24. Dajas F. Life or death: neuroprotective and anticancer effects of quercetin. J Ethnopharmacol. 2012;143:383–396. doi: 10.1016/j.jep.2012.07.005. [DOI] [PubMed] [Google Scholar]
  25. Dale DC, Guerry Dt, Wewerka JR, Bull JM, Chusid MJ. Chronic neutropenia. Medicine (Baltimore) 1979;58:128–144. doi: 10.1097/00005792-197903000-00002. [DOI] [PubMed] [Google Scholar]
  26. Daughenbaugh KF, Holderness J, Graff JC, Hedges JF, Freedman B, Graff JW, Jutila MA. Contribution of transcript stability to a conserved procyanidin-induced cytokine response in gd T cells. Genes Immun. 2011;12:378–389. doi: 10.1038/gene.2011.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Derewenda U, Artamonov M, Szukalska G, Utepbergenov D, Olekhnovich N, Parikh HI, Kellogg GE, Somlyo AV, Derewenda ZS. Identification of quercitrin as an inhibitor of the p90 S6 ribosomal kinase (RSK): structure of its complex with the N-terminal domain of RSK2 at 1.8 angstrom resolution. Acta Crystallographica Section D-Biological Crystallography. 2013;69:266–275. doi: 10.1107/S0907444912045520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Ding M, Zhao JS, Bowman L, Lu YJ, Shi XL. Inhibition of AP-1 and MAPK signaling and activation of Nrf2/ARE pathway by quercitrin. International Journal of Oncology. 2010;36:59–67. [PubMed] [Google Scholar]
  29. Dobreva MA, Green RJ, Mueller-Harvey I, Salminen JP, Howlin BJ, Frazier RA. Size and molecular flexibility affect the binding of ellagitannins to bovine serum albumin. J Agric Food Chem. 2014;62:9186–9194. doi: 10.1021/jf502174r. [DOI] [PubMed] [Google Scholar]
  30. Ducrey B, Marston A, Gohring S, Hartmann RW, Hostettmann K. Inhibition of 5 a-reductase and aromatase by the ellagitannins oenothein A and oenothein B from Epilobium species. Planta Medica. 1997;63:111–114. doi: 10.1055/s-2006-957624. [DOI] [PubMed] [Google Scholar]
  31. Ducrey B, Wolfender JL, Marston A, Hostettmann K. Analysis of Flavonol Glycosides of 13 Epilobium Species (Onagraceae) by Lc-Uv and Thermospray Lc-Ms. Phytochemistry. 1995;38:129–137. [Google Scholar]
  32. Feldman KS. Recent progress in ellagitannin chemistry. Phytochemistry. 2005;66:1984–2000. doi: 10.1016/j.phytochem.2004.11.015. [DOI] [PubMed] [Google Scholar]
  33. Fukuchi K, Sakagami H, Okuda T, Hatano T, Tanuma S, Kitajima K, Inoue Y, Inoue S, Ichikawa S, Nonoyama M. Inhibition of herpes simplex virus infection by tannins and related compounds. Antiviral Res. 1989;11:285–297. doi: 10.1016/0166-3542(89)90038-7. [DOI] [PubMed] [Google Scholar]
  34. Funatogawa K, Hayashi S, Shimomura H, Yoshida T, Hatano T, Ito H, Hirai Y. Antibacterial activity of hydrolyzable tannins derived from medicinal plants against Helicobacter pylori. Microbiol Immunol. 2004;48:251–261. doi: 10.1111/j.1348-0421.2004.tb03521.x. [DOI] [PubMed] [Google Scholar]
  35. Furst R, Zundorf I. Plant-derived anti-Inflammatory compounds: Hopes and disappointments regarding the translation of preclinical knowledge into clinical progress. Mediat Inflamm. 2014;2014:146832. doi: 10.1155/2014/146832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Gollucke APB, Aguiar O, Barbisan LF, Ribeiro DA. Use of Grape Polyphenols Against Carcinogenesis: Putative Molecular Mechanisms of Action Using In Vitro and In Vivo Test Systems. J Med Food. 2013;16:199–205. doi: 10.1089/jmf.2012.0170. [DOI] [PubMed] [Google Scholar]
  37. Granica S, Bazylko A, Kiss AK. Determination of Macrocyclic Ellagitannin Oenothein B in Plant Materials by HPLC-DAD-MS: Method Development and Validation. Phytochem Analysis. 2012;23:582–587. doi: 10.1002/pca.2358. [DOI] [PubMed] [Google Scholar]
  38. Granica S, Piwowarski JP, Czerwinska ME, Kiss AK. Phytochemistry, pharmacology and traditional uses of different Epilobium species (Onagraceae): A review. J Ethnopharmacol. 2014;156:316–346. doi: 10.1016/j.jep.2014.08.036. [DOI] [PubMed] [Google Scholar]
  39. Granica S, Piwowarski JP, Kiss AK. Ellagitannins modulate the inflammatory response of human neutrophils ex vivo. Phytomedicine. 2015;22:1215–1222. doi: 10.1016/j.phymed.2015.10.004. [DOI] [PubMed] [Google Scholar]
  40. Guo JM, Lin P, Duan JA, Shang EX, Qian DW, Tang YP. Application of microdialysis for elucidating the existing form of hyperoside in rat brain: Comparison between intragastric and intraperitoneal administration. J Ethnopharmacol. 2012;144:664–670. doi: 10.1016/j.jep.2012.10.008. [DOI] [PubMed] [Google Scholar]
  41. Hansen K, Adsersen A, Smitt UW, Nyman U, Christensen SB, Schwartner C, Wagner H. Angiotensin converting enzyme (ACE) inhibitory flavonoids from Erythroxylum laurifolium. Phytomedicine. 1996;2:313–317. doi: 10.1016/S0944-7113(96)80075-4. [DOI] [PubMed] [Google Scholar]
  42. Hatano T, Yasuhara T, Matsuda M, Yazaki K, Yoshida T, Okuda T. Oenothein B, a dimeric, hydrolyzable tannin with macrocyclic structure, and accompanying tannins from Oenothera erythrosepala. J Chem Soc-Perkin Trans 1. 1990;10:2735–2743. [Google Scholar]
  43. Hevesi Tóth B, Blazics B, Kéry A. Polyphenol composition and antioxidant capacity of Epilobium species. JPharmBiomedAnal. 2009;49:26–31. doi: 10.1016/j.jpba.2008.09.047. [DOI] [PubMed] [Google Scholar]
  44. Hiermann A, Juan H, Sametz W. Influence of Epilobium extracts on prostaglandin biosynthesis and carrageenin induced oedema of the rat paw. J Ethnopharmacol. 1986;17:161–169. doi: 10.1016/0378-8741(86)90055-3. [DOI] [PubMed] [Google Scholar]
  45. Hiermann A, Reidlinger M, Juan H, Sametz W. Isolation of the antiphlogistic principle from Epilobium angustifolium. Planta Medica. 1991;57:357–360. doi: 10.1055/s-2006-960117. [DOI] [PubMed] [Google Scholar]
  46. Holderness J, Hedges JF, Daughenbaugh K, Kimmel E, Graff J, Freedman B, Jutila MA. Response of gd T cells to plant-derived tannins. Crit Rev Immunol. 2008;28:377–402. doi: 10.1615/critrevimmunol.v28.i5.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Holderness J, Jackiw L, Kimmel E, Kerns H, Radke M, Hedges JF, Petrie C, McCurley P, Glee PM, Palecanda A, Jutila MA. Select plant tannins induce IL-2Ra up-regulation and augment cell division in gd T cells. J Immunol. 2007;179:6468–6478. doi: 10.4049/jimmunol.179.10.6468. [DOI] [PubMed] [Google Scholar]
  48. Holderness J, Schepetkin IA, Freedman B, Kirpotina LN, Quinn MT, Hedges JF, Jutila MA. Polysaccharides isolated from Acai fruit induce innate immune responses. PLoS One. 2011;6:e17301. doi: 10.1371/journal.pone.0017301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Ishimoto H, Tai A, Yoshimura M, Amakura Y, Yoshida T, Hatano T, Ito H. Antioxidative properties of functional polyphenols and their metabolites assessed by an ORAC assay. Biosci Biotechnol Biochem. 2012;76:395–399. doi: 10.1271/bbb.110717. [DOI] [PubMed] [Google Scholar]
  50. Ivancheva S, Manolova N, Serkedjieva J, Dimov V, Ivanovska N. Polyphenols from Bulgarian medicinal plants with anti-infectious activity. Basic Life Sci. 1992;59:717–728. doi: 10.1007/978-1-4615-3476-1_43. [DOI] [PubMed] [Google Scholar]
  51. Izzo AA, Hoon-Kim S, Radhakrishnan R, Williamson EM. A critical approach to evaluating clinical efficacy, adverse events and drug interactions of herbal remedies. Phytother Res. 2016 doi: 10.1002/ptr.5591. [DOI] [PubMed] [Google Scholar]
  52. Jimenez R, Lopez-Sepulveda R, Romero M, Toral M, Cogolludo A, Perez-Vizcaino F, Duarte J. Quercetin and its metabolites inhibit the membrane NADPH oxidase activity in vascular smooth muscle cells from normotensive and spontaneously hypertensive rats. Food Funct. 2015;6:409–414. doi: 10.1039/c4fo00818a. [DOI] [PubMed] [Google Scholar]
  53. Jones NP, Arnason JT, Abou-Zaid M, Akpagana K, Sanchez-Vindas P, Smith ML. Antifungal activity of extracts from medicinal plants used by First Nations Peoples of eastern Canada. J Ethnopharmacol. 2000;73:191–198. doi: 10.1016/s0378-8741(00)00306-8. [DOI] [PubMed] [Google Scholar]
  54. Joven J, Rull A, Rodriguez-Gallego E, Camps J, Riera-Borrull M, Hernandez-Aguilera A, Martin-Paredero V, Segura-Carretero A, Micol V, Alonso-Villaverde C, Menendez JA. Multifunctional targets of dietary polyphenols in disease: A case for the chemokine network and energy metabolism. Food Chem Toxicol. 2013;51:267–279. doi: 10.1016/j.fct.2012.10.004. [DOI] [PubMed] [Google Scholar]
  55. Juan H, Sametz W, Hiermann A. Anti-inflammatory effects of a substance extracted from Epilobium angustifolium. Agents Actions. 1988;23:106–107. doi: 10.1007/BF01967206. [DOI] [PubMed] [Google Scholar]
  56. Jurgenson S, Matto V, Raal A. Vegetational variation of phenolic compounds in Epilobium angustifolium. Nat Prod Res. 2012;26:1951–1953. doi: 10.1080/14786419.2011.643310. [DOI] [PubMed] [Google Scholar]
  57. Karakurt S, Semiz A, Celik G, Gencler-Ozkan AM, Sen A, Adali O. Contribution of ellagic acid on the antioxidant potential of medicinal plant Epilobium hirsutum. Nutr Cancer. 2016;68:173–183. doi: 10.1080/01635581.2016.1115092. [DOI] [PubMed] [Google Scholar]
  58. Kaskoniene V, Maruska A, Akuneca I, Stankevicius M, Ragazinskiene O, Bartkuviene V, Kornysova O, Briedis V, Ugenskiene R. Screening of antioxidant activity and volatile compounds composition of Chamerion angustifolium (L.) Holub ecotypes grown in Lithuania. Nat Prod Res. 2015a;29:1–9. doi: 10.1080/14786419.2015.1058792. [DOI] [PubMed] [Google Scholar]
  59. Kaskoniene V, Stankevicius M, Drevinskas T, Akuneca I, Kaskonas P, Bimbiraite-Surviliene K, Maruska A, Ragazinskiene O, Kornysova O, Briedis V, Ugenskiene R. Evaluation of phytochemical composition of fresh and dried raw material of introduced Chamerion angustifolium L. using chromatographic, spectrophotometric and chemometric techniques. Phytochemistry. 2015b;115:184–193. doi: 10.1016/j.phytochem.2015.02.005. [DOI] [PubMed] [Google Scholar]
  60. Kawakami K, Li P, Uraji M, Hatanaka T, Ito H. Inhibitory effects of pomegranate extracts on recombinant human maltase-glucoamylase. J Food Sci. 2014;79:H1848–H1853. doi: 10.1111/1750-3841.12568. [DOI] [PubMed] [Google Scholar]
  61. Khlebnikov AI, Schepetkin IA, Domina NG, Kirpotina LN, Quinn MT. Improved quantitative structure-activity relationship models to predict antioxidant activity of flavonoids in chemical, enzymatic, and cellular systems. Bioorg Med Chem. 2007;15:1749–1770. doi: 10.1016/j.bmc.2006.11.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Kim HJ, Lee JS, Woo ER, Kim MK, Yang BS, Yu YG, Park H, Lee YS. Isolation of virus-cell fusion inhibitory components from Eugenia caryophyllata. Planta Med. 2001;67:277–279. doi: 10.1055/s-2001-11993. [DOI] [PubMed] [Google Scholar]
  63. Kim Y, Narayanan S, Chang KO. Inhibition of influenza virus replication by plant-derived isoquercetin. Antiviral Res. 2010;88:227–235. doi: 10.1016/j.antiviral.2010.08.016. [DOI] [PubMed] [Google Scholar]
  64. Kim YS, Jung DH, Lee IS, Choi SJ, Yu SY, Ku SK, Kim MH, Kim JS. Effects of Allium victorialis leaf extracts and its single compounds on aldose reductase, advanced glycation end products and TGF-beta 1 expression in mesangial cells. BMC Complementary and Alternative Medicine. 2013;13:251. doi: 10.1186/1472-6882-13-251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Kishimoto Y, Tani M, Kondo K. Pleiotropic preventive effects of dietary polyphenols in cardiovascular diseases. European Journal of Clinical Nutrition. 2013;67:532–535. doi: 10.1038/ejcn.2013.29. [DOI] [PubMed] [Google Scholar]
  66. Kiss A, Kowalski J, Melzig MF. Compounds from Epilobium angustifolium inhibit the specific metallopeptidases ACE, NEP and APN. Planta Medica. 2004;70:919–923. doi: 10.1055/s-2004-832617. [DOI] [PubMed] [Google Scholar]
  67. Kiss A, Kowalski J, Melzig MF. Effect of Epilobium angustifolium L. extracts and polyphenols on cell proliferation and neutral endopeptidase activity in selected cell lines. Pharmazie. 2006a;61:66–69. [PubMed] [Google Scholar]
  68. Kiss A, Kowalski J, Melzig MF. Induction of neutral endopeptidase activity in PC-3 cells by an aqueous extract of Epilobium angustifolium L. and oenothein B. Phytomedicine. 2006b;13:284–289. doi: 10.1016/j.phymed.2004.08.002. [DOI] [PubMed] [Google Scholar]
  69. Kiss AK, Bazylko A, Filipek A, Granica S, Jaszewska E, Kiarszys U, Kosmider A, Piwowarski J. Oenothein B's contribution to the anti-inflammatory and antioxidant activity of Epilobium sp. Phytomedicine. 2011;18:557–560. doi: 10.1016/j.phymed.2010.10.016. [DOI] [PubMed] [Google Scholar]
  70. Korkina LG, Pastore S, Dellambra E, De Luca C. New molecular and cellular targets for chemoprevention and treatment of skin tumors by plant polyphenols: a critical review. Curr Med Chem. 2013;20:852–868. [PubMed] [Google Scholar]
  71. Kosalec I, Kopjar N, Kremer D. Antimicrobial activity of Willowherb (Epilobium angustifolium L.) leaves and flowers. Curr Drug Targets. 2013;14:986–991. doi: 10.2174/13894501113149990177. [DOI] [PubMed] [Google Scholar]
  72. Ku SK, Kim TH, Lee S, Kim SM, Bae JS. Antithrombotic and profibrinolytic activities of isorhamnetin-3-O-galactoside and hyperoside. Food and Chemical Toxicology. 2013;53:197–204. doi: 10.1016/j.fct.2012.11.040. [DOI] [PubMed] [Google Scholar]
  73. Landete JM. Updated knowledge about polyphenols: Functions, bioavailability, metabolism, and health. Crit Rev Food Sci Nutr. 2012;52:936–948. doi: 10.1080/10408398.2010.513779. [DOI] [PubMed] [Google Scholar]
  74. Larrosa M, Gonzalez-Sarrias A, Garcia-Conesa MT, Tomas-Barberan FA, Espin JC. Urolithins, ellagic acid-derived metabolites produced by human colonic microflora, exhibit estrogenic and antiestrogenic activities. J Agric Food Chem. 2006;54:1611–1620. doi: 10.1021/jf0527403. [DOI] [PubMed] [Google Scholar]
  75. Lee SY, So YJ, Shin MS, Cho JY, Lee J. Antibacterial effects of afzelin isolated from Cornus macrophylla on Pseudomonas aeruginosa, a leading cause of illness in immunocompromised individuals. Molecules. 2014;19:3173–3180. doi: 10.3390/molecules19033173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Lekstrom-Himes JA, Gallin JI. Immunodeficiency diseases caused by defects in phagocytes. N Engl J Med. 2000;343:1703–1714. doi: 10.1056/NEJM200012073432307. [DOI] [PubMed] [Google Scholar]
  77. Leonarduzzi G, Testa G, Sottero B, Gamba P, Poli G. Design and development of nanovehicle-based delivery systems for preventive or therapeutic supplementation with flavonoids. Curr Med Chem. 2010;17:74–95. doi: 10.2174/092986710789957760. [DOI] [PubMed] [Google Scholar]
  78. Lesuisse D, Berjonneau J, Ciot C, Devaux P, Doucet B, Gourvest JF, Khemis B, Lang C, Legrand R, Lowinski M, Maquin P, Parent A, Schoot B, Teutsch G. Determination of oenothein B as the active 5-a-reductase-inhibiting principle of the folk medicine Epilobium parviflorum. J Nat Prod. 1996;59:490–492. doi: 10.1021/np960231c. [DOI] [PubMed] [Google Scholar]
  79. Lewandowska U, Szewczyk K, Hrabec E, Janecka A, Gorlach S. Overview of metabolism and bioavailability enhancement of polyphenols. J Agric Food Chem. 2013;61:12183–12199. doi: 10.1021/jf404439b. [DOI] [PubMed] [Google Scholar]
  80. Li YQ, Zhou FC, Gao F, Bian JS, Shan F. Comparative evaluation of quercetin, isoquercetin and rutin as inhibitors of alpha-glucosidase. J Agric Food Chem. 2009;57:11463–11468. doi: 10.1021/jf903083h. [DOI] [PubMed] [Google Scholar]
  81. Liao YR, Lin JY. Quercetin, but not its metabolite quercetin-3-glucuronide, exerts prophylactic immunostimulatory activity and therapeutic antiinflammatory effects on lipopolysaccharide-treated mouse peritoneal macrophages ex vivo. J Agric Food Chem. 2014;62:2872–2880. doi: 10.1021/jf405630h. [DOI] [PubMed] [Google Scholar]
  82. Lin CC, Hsu YF, Lin TC. Antioxidant and free radical scavenging effects of the tannins of Terminalia catappa L. Anticancer Res. 2001;21:237–243. [PubMed] [Google Scholar]
  83. Liu K, Mei F, Wang Y, Xiao N, Yang L, Wang Y, Li J, Huang F, Kou J, Liu B, Qi LW. Quercetin oppositely regulates insulin-mediated glucose disposal in skeletal muscle under normal and inflammatory conditions: The dual roles of AMPK activation. Mol Nutr Food Res. 2016;60:551–565. doi: 10.1002/mnfr.201500509. [DOI] [PubMed] [Google Scholar]
  84. Liu RL, Xiong QJ, Shu Q, Wu WN, Cheng J, Fu H, Wang F, Chen JG, Hu ZL. Hyperoside protects cortical neurons from oxygen-glucose deprivation-reperfusion induced injury via nitric oxide signal pathway. Brain Res. 2012;1469:164–173. doi: 10.1016/j.brainres.2012.06.044. [DOI] [PubMed] [Google Scholar]
  85. Manach C, Williamson G, Morand C, Scalbert A, Remesy C. Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am J Clin Nutr. 2005;81:230S–242S. doi: 10.1093/ajcn/81.1.230S. [DOI] [PubMed] [Google Scholar]
  86. Marzouk MSA, Moharram FA, Mohamed MA, Gamal-Eldeen AM, Aboutabl EA. Anticancer and antioxidant tannins from Pimenta dioica leaves. Zeitschrift Fur Naturforschung C-a Journal of Biosciences. 2007;62:526–536. doi: 10.1515/znc-2007-7-811. [DOI] [PubMed] [Google Scholar]
  87. Messer JG, Hopkins RG, Kipp DE. Quercetin metabolites up-regulate the antioxidant response in osteoblasts isolated from fetal rat calvaria. J Cell Biochem. 2015;116:1857–1866. doi: 10.1002/jcb.25141. [DOI] [PubMed] [Google Scholar]
  88. Min BS, Lee SY, Kim JH, Lee JK, Kim TJ, Kim DH, Kim YH, Joung H, Lee HK, Nakamura N, Miyashiro H, Hattori M. Anti-complement activity of constituents from the stem-bark of Juglans mandshurica. Biol Pharmaceut Bull. 2003;26:1042–1044. doi: 10.1248/bpb.26.1042. [DOI] [PubMed] [Google Scholar]
  89. Mira L, Fernandez MT, Santos M, Rocha R, Florencio MH, Jennings KR. Interactions of flavonoids with iron and copper ions: a mechanism for their antioxidant activity. Free Radic Res. 2002;36:1199–1208. doi: 10.1080/1071576021000016463. [DOI] [PubMed] [Google Scholar]
  90. Miyamoto K, Murayama T, Nomura M, Hatano T, Yoshida T, Furukawa T, Koshiura R, Okuda T. Antitumor activity and interleukin-1 induction by tannins. Anticancer Res. 1993a;13:37–42. [PubMed] [Google Scholar]
  91. Miyamoto K, Nomura M, Sasakura M, Matsui E, Koshiura R, Murayama T, Furukawa T, Hatano T, Yoshida T, Okuda T. Antitumor activity of oenothein B, a unique macrocyclic ellagitannin. Jpn J Cancer Res. 1993b;84:99–103. doi: 10.1111/j.1349-7006.1993.tb02790.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Monschein M, Jaindl K, Buzimkic S, Bucar F. Content of phenolic compounds in wild populations of Epilobium angustifolium growing at different altitudes. Pharm Biol. 2015;53:1576–1582. doi: 10.3109/13880209.2014.993039. [DOI] [PubMed] [Google Scholar]
  93. Moskalenko SA. Preliminary screening of far-eastern ethnomedicinal plants for antibacterial activity. J Ethnopharmacol. 1986;15:231–259. doi: 10.1016/0378-8741(86)90163-7. [DOI] [PubMed] [Google Scholar]
  94. Myagmar BE, Aniya Y. Free radical scavenging action of medicinal herbs from Mongolia. Phytomedicine. 2000;7:221–229. doi: 10.1016/S0944-7113(00)80007-0. [DOI] [PubMed] [Google Scholar]
  95. Nguyen XN, Phan VK, Chau VM, Ninh KB, Nguyen XC, Tai BH, Kim YH. Phenylpropanoid glycosides from Heterosmilax erythrantha and their antioxidant activity. Arch Pharmacal Res. 2009;32:1373–1377. doi: 10.1007/s12272-009-2005-4. [DOI] [PubMed] [Google Scholar]
  96. Okuda T. Systematics and health effects of chemically distinct tannins in medicinal plants. Phytochemistry. 2005;66:2012–2031. doi: 10.1016/j.phytochem.2005.04.023. [DOI] [PubMed] [Google Scholar]
  97. Okuda T, Yoshida T, Hatano T. Ellagitannins as active constituents of medicinal plants. Planta Medica. 1989;117–122:117–122. doi: 10.1055/s-2006-961902. [DOI] [PubMed] [Google Scholar]
  98. Okuyama S, Makihata N, Yoshimura M, Amakura Y, Yoshida T, Nakajima M, Furukawa Y. Oenothein B suppresses lipopolysaccharide (LPS)-induced inflammation in the mouse brain. Int J Mol Sci. 2013;14:9767–9778. doi: 10.3390/ijms14059767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Pasinetti GM. Novel role of red wine-derived polyphenols in the prevention of Alzheimer's Disease dementia and brain pathology: Experimental approaches and clinical implications. Planta Medica. 2012;78:1614–1619. doi: 10.1055/s-0032-1315377. [DOI] [PubMed] [Google Scholar]
  100. Peng J, Wei D, Fu Z, Li D, Tan Y, Xu T, Zhou J, Zhang T. Punicalagin ameliorates lipopolysaccharide-induced acute respiratory distress syndrome in mice. Inflammation. 2015;38:493–499. doi: 10.1007/s10753-014-9955-5. [DOI] [PubMed] [Google Scholar]
  101. Petrova MF, Pukhal'skaia EC, Gotinskaya LG. A study for structure of Chanerol and characterization of its antitumor activity. Prob Chemother Tumors Moscow-Kiev. 1974;1:49. [Google Scholar]
  102. Piwowarski JP, Granica S, Kiss AK. Influence of gut microbiota-derived ellagitannins' metabolites urolithins on pro-inflammatory activities of human neutrophils. Planta Med. 2014;80:887–895. doi: 10.1055/s-0034-1368615. [DOI] [PubMed] [Google Scholar]
  103. Pukhal'skaia E, Petrova MF, Kibal'chich PN, Alieva TA, Denisova SI. Isolation of a polymer from Chamaenerium angustifolium and study of its antineoplastic action. Antibiotiki. 1970;15:782–785. [PubMed] [Google Scholar]
  104. Pukhalskaya EC, Chernyakhovskaya I, Petrova MF, Denisova SI, Alieva TA. Macromolecular antitumor agents from Chamaenerium angustifolium. Neoplasma. 1975;22:29–37. [PubMed] [Google Scholar]
  105. Quideau S, Deffieux D, Douat-Casassus C, Pouysegu L. Plant polyphenols: chemical properties, biological activities, and synthesis. Angew Chem Int Ed Engl. 2011;50:586–621. doi: 10.1002/anie.201000044. [DOI] [PubMed] [Google Scholar]
  106. Rajendran P, Rengarajan T, Nandakumar N, Palaniswami R, Nishigaki Y, Nishigaki I. Kaempferol, a potential cytostatic and cure for inflammatory disorders. Eur J Med Chem. 2014;86C:103–112. doi: 10.1016/j.ejmech.2014.08.011. [DOI] [PubMed] [Google Scholar]
  107. Ramiro-Puig E, Perez-Cano FJ, Ramos-Romero S, Perez-Berezo T, Castellote C, Permanyer J, Franch A, Izquierdo-Pulido M, Castell M. Intestinal immune system of young rats influenced by cocoa-enriched diet. J Nutr Biochem. 2008;19:555–565. doi: 10.1016/j.jnutbio.2007.07.002. [DOI] [PubMed] [Google Scholar]
  108. Ramstead AG, Schepetkin IA, Quinn MT, Jutila MA. Oenothein B, a Cyclic Dimeric Ellagitannin Isolated from Epilobium angustifolium, Enhances IFNgamma Production by Lymphocytes. PLoS One. 2012;7:e50546. doi: 10.1371/journal.pone.0050546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Ramstead AG, Schepetkin IA, Todd K, Loeffelholz J, Quinn MT, Jutila MA. Aging influences the response of T cells to stimulation by the ellagitannin, oenothein B. Int Immunopharmacol. 2015;26:367–377. doi: 10.1016/j.intimp.2015.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Ratz-Lyko A, Arct J, Majewski S, Pytkowska K. Influence of polyphenols on the physiological processes in the skin. Phytother Res. 2015;29:509–517. doi: 10.1002/ptr.5289. [DOI] [PubMed] [Google Scholar]
  111. Rauha JP, Remes S, Heinonen M, Hopia A, Kahkonen M, Kujala T, Pihlaja K, Vuorela H, Vuorela P. Antimicrobial effects of Finnish plant extracts containing flavonoids and other phenolic compounds. Int J Food Microbiol. 2000;56:3–12. doi: 10.1016/s0168-1605(00)00218-x. [DOI] [PubMed] [Google Scholar]
  112. Remmel I, Vares L, Toom L, Matto V, Raal A. Phenolic compounds in five Epilobium species collected from Estonia. Nat Prod Commun. 2012;7:1323–1324. [PubMed] [Google Scholar]
  113. Rice-Evans CA, Miller NJ, Bolwell PG, Bramley PM, Pridham JB. The relative antioxidant activities of plant-derived polyphenolic flavonoids. Free Radic Res. 1995;22:375–383. doi: 10.3109/10715769509145649. [DOI] [PubMed] [Google Scholar]
  114. Rice-evans CA, Miller NJ, Paganga G. Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radic Biol Med. 1996;20:933–956. doi: 10.1016/0891-5849(95)02227-9. [DOI] [PubMed] [Google Scholar]
  115. Russo GL, Russo M, Spagnuolo C, Tedesco I, Bilotto S, Iannitti R, Palumbo R. Quercetin: A pleiotropic kinase inhibitor against cancer. Adv Nutr Cancer. 2014;159:185–205. doi: 10.1007/978-3-642-38007-5_11. [DOI] [PubMed] [Google Scholar]
  116. Ruszova E, Cheel J, Pavek S, Moravcova M, Hermannova M, Matejkova I, Spilkova J, Velebny V, Kubala L. Epilobium angustifolium extract demonstrates multiple effects on dermal fibroblasts in vitro and skin photo-protection in vivo. Gen Physiol Biophys. 2013;32:347–359. doi: 10.4149/gpb_2013031. [DOI] [PubMed] [Google Scholar]
  117. Ruzic I, Skerget M, Knez Z. Potential of phenolic antioxidants. Acta Chim Slov. 2010;57:263–271. [PubMed] [Google Scholar]
  118. Sak K. Site-specific anticancer effects of dietary flavonoid quercetin. Nutr Cancer. 2014;66:177–193. doi: 10.1080/01635581.2014.864418. [DOI] [PubMed] [Google Scholar]
  119. Sakagami H, Jiang Y, Kusama K, Atsumi T, Ueha T, Toguchi M, Iwakura I, Satoh K, Ito H, Hatano T, Yoshida T. Cytotoxic activity of hydrolyzable tannins against human oral tumor cell lines--a possible mechanism. Phytomedicine. 2000;7:39–47. doi: 10.1016/S0944-7113(00)80020-3. [DOI] [PubMed] [Google Scholar]
  120. Santos GD, Ferri PH, Santos SC, Bao SN, Soares CMA, Pereira M. Oenothein B inhibits the expression of PbFKS1 transcript and induces morphological changes in Paracoccidioides brasiliensis. Med Mycol. 2007;45:609–618. doi: 10.1080/13693780701502108. [DOI] [PubMed] [Google Scholar]
  121. Sasov SA, Tolkachev VN, Yartseva IV, Tolkachev ON. Macrocyclic tannins of Chamerion Angustifolium. Problems of Biological, Medical and Pharmaceutical Chemistry. 2010;10:24–27. [Google Scholar]
  122. Satue M, Arriero MD, Monjo M, Ramis JM. Quercitrin and Taxifolin stimulate osteoblast differentiation in MC3T3-E1 cells and inhibit osteoclastogenesis in RAW 264.7 cells. Biochem Pharmacol. 2013;86:1476–1486. doi: 10.1016/j.bcp.2013.09.009. [DOI] [PubMed] [Google Scholar]
  123. Schaffer S, Asseburg H, Kuntz S, Muller WE, Eckert GP. Effects of polyphenols on brain ageing and Alzheimer's Disease: Focus on mitochondria. Mol Neurobiol. 2012;46:161–178. doi: 10.1007/s12035-012-8282-9. [DOI] [PubMed] [Google Scholar]
  124. Schepetkin IA, Kirpotina LN, Jakiw L, Khlebnikov AI, Blaskovich CL, Jutila MA, Quinn MT. Immunomodulatory activity of oenothein B isolated from Epilobium angustifolium. J Immunol. 2009;183:6754–6766. doi: 10.4049/jimmunol.0901827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Schepetkin IA, Kirpotina LN, Khlebnikov AI, Jutila MA, Quinn MT. Gastrin-releasing peptide/neuromedin B receptor antagonists PD176252, PD168368, and related analogs are potent agonists of human formyl-peptide receptors. Mol Pharmacol. 2011;79:77–90. doi: 10.1124/mol.110.068288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Schmid D, Gruber M, Piskaty C, Woehs F, Renner A, Nagy Z, Kaltenboeck A, Wasserscheid T, Bazylko A, Kiss AK, Moeslinger T. Inhibition of NF-kappa B-dependent cytokine and inducible nitric oxide synthesis by the macrocyclic ellagitannin oenothein B in TLR-stimulated RAW 264.7 macrophages. J Nat Prod. 2012;75:870–875. doi: 10.1021/np200756f. [DOI] [PubMed] [Google Scholar]
  127. Shikov AN, Poltanov EA, Dorman HJ, Makarov VG, Tikhonov VP, Hiltunen R. Chemical composition and in vitro antioxidant evaluation of commercial water-soluble willow herb (Epilobium angustifolium L.) extracts. J Agric Food Chem. 2006;54:3617–3624. doi: 10.1021/jf052606i. [DOI] [PubMed] [Google Scholar]
  128. Shin SW, Jung E, Kim S, Kim JH, Kim EG, Lee J, Park D. Antagonizing effects and mechanisms of Afzelin against UVB-induced cell damage. PLoS One. 2013;8:e61971. doi: 10.1371/journal.pone.0061971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Shiota S, Shimizu M, Sugiyama J, Morita Y, Mizushima T, Tsuchiya T. Mechanisms of action of corilagin and tellimagrandin I that remarkably potentiate the activity of beta-lactams against methicillin-resistant Staphylococcus aureus. Microbiol Immunol. 2004;48:67–73. doi: 10.1111/j.1348-0421.2004.tb03489.x. [DOI] [PubMed] [Google Scholar]
  130. Skyberg JA, Robison A, Golden S, Rollins MF, Callis G, Huarte E, Kochetkova I, Jutila MA, Pascual DW. Apple polyphenols require T cells to ameliorate dextran sulfate sodium-induced colitis and dampen proinflammatory cytokine expression. J Leukoc Biol. 2011;90:1043–1054. doi: 10.1189/jlb.0311168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Song M, Hong M, Lee MY, Jee JG, Lee YM, Bae JS, Jeong TC, Lee S. Selective inhibition of the cytochrome P450 isoform by hyperoside and its potent inhibition of CYP2D6. Food Chem Toxicol. 2013;59:549–553. doi: 10.1016/j.fct.2013.06.055. [DOI] [PubMed] [Google Scholar]
  132. Spiridonov NA, Arkhipov VV, Foĭgel AG, Tolkachev ON, Sasov SA, Syrkin AB, Tolkachev VN. The cytotoxicity of Chamaenerium angustifolium (L.) Scop. and Hippophae rhamnoides L. tannins and their effect on mitochondrial respiration. Eksp Klin Farmakol. 1997;60(4):60–63. [PubMed] [Google Scholar]
  133. Stagos D, Amoutzias GD, Matakos A, Spyrou A, Tsatsakis AM, Kouretas D. Chemoprevention of liver cancer by plant polyphenols. Food Chem Toxicol. 2012;50:2155–2170. doi: 10.1016/j.fct.2012.04.002. [DOI] [PubMed] [Google Scholar]
  134. Stolarczyk M, Naruszewicz M, Kiss AK. Extracts from Epilobium sp. herbs induce apoptosis in human hormone-dependent prostate cancer cells by activating the mitochondrial pathway. J Pharm Pharmacol. 2013a;65:1044–1054. doi: 10.1111/jphp.12063. [DOI] [PubMed] [Google Scholar]
  135. Stolarczyk M, Piwowarski JP, Granica S, Stefanska J, Naruszewicz M, Kiss AK. Extracts from Epilobium sp herbs, their components and gut microbiota metabolites of Epilobium Ellagitannins, urolithins, inhibit hormone-dependent prostate cancer cells-(LNCaP) proliferation and PSA secretion. Phytother Res. 2013b;27:1842–1848. doi: 10.1002/ptr.4941. [DOI] [PubMed] [Google Scholar]
  136. Syrkin AB, Konyaeva OI. Pharmacological investigations of some new antineoplastic compounds. Khimiko-Farmatsevticheskii Zhurnal. 1984;18:1172–1180. [Google Scholar]
  137. Tahara K, Hashida K, Otsuka Y, Ohara S, Kojima K, Shinohara K. Identification of a hydrolyzable tannin, oenothein B, as an aluminum-detoxifying ligand in a highly aluminum-resistant tree, Eucalyptus camaldulensis. Plant Physiology. 2014;164:683–693. doi: 10.1104/pp.113.222885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Tarko T, Duda-Chodak A, Zajac N. Digestion and absorption of phenolic compounds assessed by in vitro simulation methods. A review. Rocz Panstw Zakl Hig. 2013;64:79–84. [PubMed] [Google Scholar]
  139. Thapa M, Kim Y, Desper J, Chang KO, Hua DH. Synthesis and antiviral activity of substituted quercetins. Bioorg Med Chem Lett. 2012;22:353–356. doi: 10.1016/j.bmcl.2011.10.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Tita B, Abdel-Haq H, Vitalone A, Mazzanti G, Saso L. Analgesic properties of Epilobium angustifolium, evaluated by the hot plate test and the writhing test. Farmaco. 2001;56:341–343. doi: 10.1016/s0014-827x(01)01046-1. [DOI] [PubMed] [Google Scholar]
  141. Uysal U, Seremet S, Lamping JW, Adams JM, Liu DY, Swerdlow RH, Aires DJ. Consumption of polyphenol plants may slow aging and associated diseases. Current Pharmaceutical Design. 2013;19:6094–6111. doi: 10.2174/1381612811319340004. [DOI] [PubMed] [Google Scholar]
  142. Vila S, Prahoveanu E, Esanu V, Mihaila V, Pintilie G, Niculescu M, Teoteoi N, Colceru S. The action of a Chamenerion angustifolium extract in experimental influenza virus infection in mice. Virologie. 1989;40:129–132. [PubMed] [Google Scholar]
  143. Vitalone A, Bordi F, Baldazzi C, Mazzanti G, Saso L, Tita B. Anti-proliferative effect on a prostatic epithelial cell line (PZ-HPV-7) by Epilobium angustifolium L. Farmaco. 2001;56:483–489. doi: 10.1016/s0014-827x(01)01067-9. [DOI] [PubMed] [Google Scholar]
  144. Vitalone A, Guizzetti M, Costa LG, Tita B. Extracts of various species of Epilobium inhibit proliferation of human prostate cells. J Pharm Pharmacol. 2003a;55:683–690. doi: 10.1211/002235703765344603. [DOI] [PubMed] [Google Scholar]
  145. Vitalone A, McColl J, Thome D, Costa LG, Tita B. Characterization of the effect of Epilobium extracts on human cell proliferation. Pharmacology. 2003b;69:79–87. doi: 10.1159/000072360. [DOI] [PubMed] [Google Scholar]
  146. Vogl S, Picker P, Mihaly-Bison J, Fakhrudin N, Atanasov AG, Heiss EH, Wawrosch C, Reznicek G, Dirsch VM, Saukel J, Kopp B. Ethnopharmacological in vitro studies on Austria's folk medicine--an unexplored lore in vitro anti-inflammatory activities of 71 Austrian traditional herbal drugs. J Ethnopharmacol. 2013;149:750–771. doi: 10.1016/j.jep.2013.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Voyich JM, Braughton KR, Sturdevant DE, Whitney AR, Said-Salim B, Porcella SF, Long RD, Dorward DW, Gardner DJ, Kreiswirth BN, Musser JM, DeLeo FR. Insights into mechanisms used by Staphylococcus aureus to avoid destruction by human neutrophils. J Immunol. 2005;175:3907–3919. doi: 10.4049/jimmunol.175.6.3907. [DOI] [PubMed] [Google Scholar]
  148. Voyich JM, Otto M, Mathema B, Braughton KR, Whitney AR, Welty D, Long RD, Dorward DW, Gardner DJ, Lina G, Kreiswirth BN, DeLeo FR. Is Panton-Valentine leukocidin the major virulence determinant in community-associated methicillin-resistant Staphylococcus aureus disease? J Infect Dis. 2006;194:1761–1770. doi: 10.1086/509506. [DOI] [PubMed] [Google Scholar]
  149. Voyich JM, Vuong C, DeWald M, Nygaard TK, Kocianova S, Griffith S, Jones J, Iverson C, Sturdevant DE, Braughton KR, Whitney AR, Otto M, DeLeo FR. The SaeR/S gene regulatory system is essential for innate immune evasion by Staphylococcus aureus. J Infect Dis. 2009;199:1698–1706. doi: 10.1086/598967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Voynova E, Dimitrova S, Naydenova E, Karadjov P. Inhibitory action of extracts of Maclura aurantiaca and Epilobium hirsutum on tumour models in mice. Acta Physiol Pharmacol Bulg. 1991;17:50–52. [PubMed] [Google Scholar]
  151. Wang CP, Li JL, Zhang LZ, Zhang XC, Yu S, Liang XM, Ding F, Wang ZW. Isoquercetin protects cortical neurons from oxygen-glucose deprivation-reperfusion induced injury via suppression of TLR4-NF-kappa B signal pathway. Neurochem Int. 2013;63:741–749. doi: 10.1016/j.neuint.2013.09.018. [DOI] [PubMed] [Google Scholar]
  152. Wang R, Braughton KR, Kretschmer D, Bach TH, Queck SY, Li M, Kennedy AD, Dorward DW, Klebanoff SJ, Peschel A, DeLeo FR, Otto M. Identification of novel cytolytic peptides as key virulence determinants for community-associated MRSA. Nat Med. 2007;13:1510–1514. doi: 10.1038/nm1656. [DOI] [PubMed] [Google Scholar]
  153. Webster D, Taschereau P, Belland RJ, Sand C, Rennie RP. Antifungal activity of medicinal plant extracts; preliminary screening studies. Journal of Ethnopharmacology. 2008;115:140–146. doi: 10.1016/j.jep.2007.09.014. [DOI] [PubMed] [Google Scholar]
  154. Yamanaka D, Tamiya Y, Motoi M, Ishibashi K, Miura NN, Adachi Y, Ohno N. The effect of enzymatically polymerised polyphenols on CD4 binding and cytokine production in murine splenocytes. PLoS One. 2012;7:e36025. doi: 10.1371/journal.pone.0036025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Yin YQ, Li WQ, Son YO, Sun LJ, Lu J, Kim D, Wang X, Yao H, Wang L, Pratheeshkumar P, Hitron AJ, Luo J, Gao N, Shi XL, Zhang Z. Quercitrin protects skin from UVB-induced oxidative damage. Toxicol Appl Pharmacol. 2013;269:89–99. doi: 10.1016/j.taap.2013.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Yoshida T, Amakura Y, Yoshimura M. Structural features and biological properties of ellagitannins in some plant families of the order Myrtales. Int J Mol Sci. 2010;11:79–106. doi: 10.3390/ijms11010079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Yoshimura M, Akiyama H, Kondo K, Sakata K, Matsuoka H, Amakura Y, Teshima R, Yoshida T. Immunological effects of oenothein B, an ellagitannin dimer, on dendritic cells. International Journal of Molecular Sciences. 2013;14:46–56. doi: 10.3390/ijms14010046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Yoshioka H, Senba Y, Saito K, Kimura T, Hayakawa F. Spin-trapping study on the hydroxyl radical formed from a tea catechin-Cu(II) system. Biosci Biotechnol Biochem. 2001;65:1697–1706. doi: 10.1271/bbb.65.1697. [DOI] [PubMed] [Google Scholar]
  159. Yu HN, Shen SR, Xiong YK. Cytotoxicity of epigallocatechin-3-gallate to LNCaP cells in the presence of Cu2+ J Zhejiang Univ Sci B. 2005;6:125–131. doi: 10.1631/jzus.2005.B0125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Zambuzzi-Carvalho PF, Tomazett PK, Santos SC, Ferri PH, Borges CL, Martins WS, Soares CMD, Pereira M. Transcriptional profile of Paracoccidioides induced by oenothein B, a potential antifungal agent from the Brazilian Cerrado plant Eugenia uniflora. BMC Microbiology. 2013;13:227. doi: 10.1186/1471-2180-13-227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Zeng KW, Wang XM, Ko H, Kwon HC, Cha JW, Yang HO. Hyperoside protects primary rat cortical neurons from neurotoxicity induced by amyloid beta-protein via the PI3K/Akt/Bad/Bcl(XL)-regulated mitochondrial apoptotic pathway. Eur J Pharmacol. 2011;672:45–55. doi: 10.1016/j.ejphar.2011.09.177. [DOI] [PubMed] [Google Scholar]
  162. Zhang HM, Zhao L, Li H, Xu H, Chen WW, Tao L. Research progress on the anticarcinogenic actions and mechanisms of ellagic acid. Cancer Biol Med. 2014;11:92–100. doi: 10.7497/j.issn.2095-3941.2014.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Zhang R, Yao Y, Wang Y, Ren G. Antidiabetic activity of isoquercetin in diabetic KK -Ay mice. Nutr Metab (Lond) 2011;8:85. doi: 10.1186/1743-7075-8-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Zhang ZY, Sethiel MS, Shen WZ, Liao ST, Zou YX. Hyperoside downregulates the receptor for advanced glycation end products (RAGE) and promotes proliferation in ECV304 cells via the c-Jun N-terminal kinase (JNK) pathway following stimulation by advanced glycation end-products in vitro. Int J Mol Sci. 2013;14:22697–22707. doi: 10.3390/ijms141122697. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES