The Natural Flavonoid Pinocembrin: Molecular Targets and Potential Therapeutic Applications.
Journal: 2016/December - Molecular Neurobiology
ISSN: 1559-1182
Abstract:
Pinocembrin is a natural flavonoid compound extracted from honey, propolis, ginger roots, wild marjoram, and other plants. In preclinical studies, it has shown anti-inflammatory and neuroprotective effects as well as the ability to reduce reactive oxygen species, protect the blood-brain barrier, modulate mitochondrial function, and regulate apoptosis. Considering these pharmaceutical characteristics, pinocembrin has potential as a drug to treat ischemic stroke and other clinical conditions. In this review, we summarize its pharmacologic characteristics and discuss its mechanisms of action and potential therapeutic applications.
Relations:
Content
Citations
(18)
References
(72)
Grants
(5)
Drugs
(4)
Chemicals
(3)
Organisms
(2)
Affiliates
(2)
Similar articles
Articles by the same authors
Discussion board
Mol Neurobiol 53(3): 1794-1801

The natural flavonoid pinocembrin: molecular targets and potential therapeutic applications

Introduction

Pinocembrin (5,7-dihydroxyflavanone, Figure 1) is a natural flavonoid compound that has been identified in honey, propolis, and several plants, such as ginger roots and wild marjoram [16]. Apart from natural extraction, pinocembrin has been successfully biosynthesized [714] and chemosynthesized [15]. In pharmacological studies, it has shown a variety of properties that could hold promise for treating diseases such as endotoxin shock, cancer, and cardiovascular diseases [16]. It is interesting to note that after oral administration, this compound is well metabolized and absorbed [1719]. In vitro, it has been shown to pass through the blood-brain barrier (BBB) in a passive transport process, which is partly conducted by p-glycoprotein [20]. This finding indicates that pinocembrin might be useful for treatment of diseases in the central nervous system (CNS). In fact, previous studies have shown that pinocembrin has anti-inflammatory and neuroprotective effects and the ability to reduce reactive oxygen species (ROS), protect the BBB, modulate mitochondrial function, and regulate apoptosis. As discussed in detail below, in vitro and in vivo studies have provided evidence that pinocembrin can protect the brain against damage from ischemic stroke. Therefore, in 2008, pinocembrin was approved by the State Food and Drug Administration of China for clinical trials in patients with ischemic stroke [21], and is currently enrolling patients for Phase II clinical trials [22].

An external file that holds a picture, illustration, etc.
Object name is nihms670117f1.jpg

Chemical structure of pinocembrin.

In this review, we summarize recent preclinical studies of pinocembrin and focus on its effects in CNS diseases and related indications. We discuss information regarding the potential targets of pinocembrin, its possible mechanisms of action, existing problems, and potential therapeutic applications.

Pharmacokinetics of pinocembrin

In preclinical studies, two methods have been used to detect pinocembrin in rat plasma. In the first, investigators administered 22.5 mg/kg or 67.5 mg/kg pinocembrin to rats via intravenous injection (i.v.) and then used reversed-phase high-performance liquid chromatography with ultraviolet detection [23]. In the second study, rats were injected i.v. with 10 mg/kg pinocembrin. Then high-performance liquid chromatographic–electrospray ionization–mass spectrometry was used to detect S-pinocembrin and R-pinocembrin in plasma [17]. In clinical trials, high-performance liquid chromatography–mass spectrometry-mass spectrometry has been used to measure pinocembrin in the plasma of healthy volunteers [21] (Table 1).

Table 1

Pharmacokinetic characteristics of pinocembrin

SubjectDose (route)AUCT1/2 (min)CLVd
SD rat3[23]22.5 mg/kg (i.v. injection)0–2 h: 340.37 ± 2.67 mg/L min14.61 ± 3.740.07 ± 0.02 L/min/kg
SD rat [23]67.5 mg/kg (i.v. injection)0–2 h: 1698.55 ± 335.95 mg/L min13.93 ± 5.020.04 ± 0.01 L/min/kg
SD rat [17]10 mg/kg (i.v. injection)S-Pino: 0–∞ 1.821 ± 0.211 h mg/mL12.72 ± 8.40.092 ± 0.011 L/min/kg1.758 ± 1.313 L/kg
SD rat [17]10 mg/kg (i.v. injection)R-Pino: 0–∞ 1.876 ± 0.427 h mg/mL13.38 ± 4.980.092 ± 0.020 L/min/kg1.793 ± 0.805 L/kg
Healthy human [21]20 mg (i.v. drip)0–8 h: 10,236.0 ± 1547.4 ng/mL min
0–∞: 10,338.1 ± 1539.4 ng/mL min
47.4 ± 14.02.0 ± 0.3 L/min136.6 ± 52.8 L

AUC, area under the curve; CL, clearance; i.v., intravenous; R-Pino, R-Pinocembrin; SD, Sprague-Dawley; S-Pino, S-Pinocembrin; T1/2, half-life; Vd, volume of distribution

Biosynthesis and synthesis of pinocembrin analogs

Pinocembrin was first isolated from Eriodictyon californicum in 1992 [24]. In plants, pinocembrin is synthesized from phenylalanine by the action of three enzymes: phenylalanine ammonia lyase, 4-coumarate: CoA ligase, and chalcone synthase [25]. Based on this finding, large amounts of pinocembrin have been biosynthesized from recombinant Escherichia coli [26,9] and Saccharomyces cerevisiae [27] containing an artificial gene cluster. Recently, Kim et al. [28] successfully synthesized pinocembrin from glucose using engineered E. coli. To achieve industrial production of pinocembrin, Yuan et al. [15] developed a convenient method to synthesize (+/−)-pinocembrin by using a chiral primary amine or a chiral sulfinamide as resolving agent.

Potential therapeutic applications and mechanism of action

Pinocembrin has been studied in stroke, Alzheimer’s disease, cardiovascular diseases, and atherosclerosis in vitro and in vivo (Table 2). Its potential application for cancer treatment is not covered by this review.

Table 2

Main published applications of pinocemrin in vitro and in vivo

DiseasesModel/organismEffective doseResults
Ischemic strokepMCAO/rat [34,5]10 mg/kg (i.v)+
MCAO-reperfusion/rat [33,47]10 mg/kg (i.v)+
4-VO/rat [48,30]5 mg/kg (i.v)+
OGD/R-induced primary neurons [33]10 μM+
Glutamate-induced SH-Sy5y [32]10 μM+
OGD/R-induced RCMECs [48]10 μM+
Alzheimer’s diseaseAPP/PS1 transgenic mouse [51]40 mg/kg (i.g.)+
APPsw-overexpressing SH-Sy5y cell line [50]10 μM+
25-35 induced primary neurons [50]10 μM+
AtherosclerosisApoE mice [56]20 mg/kg (i.g.)+
Ang II-induced rat thoracic aortic rings [54]100 μM+
Endotoxin shockLPS-induced endotoxin shock mice [57]50 mg/kg (i.p)+

4-VO, 4-vessel occlusion; Ang II, angiotensin II; i.g., intragastric; i.v., intravascular; LPS, lipopolysaccharide; MCAO, middle cerebral artery occlusion; OGD/R, oxygen–glucose deprivation/reoxygenation; pMCAO, permanent MCAO; RCMECs, rat cerebral microvascular endothelial cells.

Stroke

Stroke is the fourth leading cause of death in the United States and one of the leading causes of adult disability. It can also have tremendous economic and social impacts [29]. Many survivors are left with lifelong, devastating neurologic deficits. In the past decade, various studies have demonstrated that pinocembrin can protect against cerebral ischemic injury with a wide therapeutic time window [30]. It has shown neuroprotective [31,32], anti-oxidative [33], and anti-inflammatory effects [5] both in vitro and in vivo. Pinocembrin exerts potent protective effects in rats with ischemic stroke [3033]. Such findings indicate that it is a promising compound for the development of novel, multiple-action drug therapies.

Effects of pinocembrin in animal models of middle cerebral artery occlusion (MCAO)

The first stroke model used to evaluate the efficacy of pinocembrin was MCAO. In rats with permanent cerebral ischemia (pMCAO), Gao et al. [34] showed that pinocembrin (10 mg/kg, i.v.) could reduce brain swelling; improve behavioral deficits; and alleviate neuronal apoptosis, edema of astrocytic end-feet, and the deformation of endothelial cells and capillaries. Moreover, pinocembrin also protected the BBB by decreasing matrix metalloproteinase-9 protein expression and reducing mRNA level of the tight junction protein zonula occludens-1. Acting on the neurovascular unit, pinocembrin reduces the level of proinflammatory cytokines [tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1β)], chemokines [intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1)], inducible nitric oxide (NO) synthase (iNOS), and aquaporin-4 [5], suggesting that pinocembrin has an anti-inflammatory effect. In a recent study, Wang et al. [35] co-administered pinocembrin and antagonists of epoxyeicosatrienoic acids (EETs) to pMCAO rats. Results showed that pinocembrin suppressed soluble epoxide hydrolase (sEH) activity and that the EET antagonist weakened the beneficial effects of pinocembrin in pMCAO rats. These results indicate that sEH and the EETs might be targets of pinocembrin. sEH metabolizes EETs into less-active dihydroxyeicosatrienoic acids (DHETs) [3638]. Because of multiple protective properties [39,40], increasing the level of EETs would be beneficial for injured brain. In mouse and human brain, sEH is expressed in multiple brain cell types, such as neurons, astrocytes, and endothelial cells [39,41]. sEH deletion or inhibition has shown protective effects in both ischemic stroke models [4245] and neurons subjected to oxygen-glucose deprivation [44,46].

Pinocembrin (10 mg/kg) was also shown to improve ischemic outcomes in the MCAO-reperfusion animal model [33]. It was suggested that pinocembrin reduces endoplasmic reticulum (ER) stress and apoptosis by decreasing C/EBP homologous protein (CHOP)/GADD153 and caspase-12 expression via the PERK-elF2α-ATF4 signaling pathway in MCAO rats [47].

Effects of pinocembrin on 4-vessel occlusion (4-VO) animal model

In a 4-VO rat model, 5 mg/kg pinocembrin protected the BBB by regulating the pathological changes of ultrastructure in microvessels, neurons, and glial cells [48]. In addition, Shi et al. [30] further reported that pinocembrin had an antioxidant effect in 4-VO rats. Pinocembrin reduces the compensatory increase of superoxide dismutase activity, malondialdehyde content, and myeloperoxidase activity; modulates the concentration of excitatory and inhibitory amino acids; and lessens the neuronal loss in rats after global cerebral 4-VO ischemia in a dose-dependent manner (1, 5, 10 mg/kg). In terms of the therapeutic time window, it is notable that pinocembrin was still protective even if administered up to 6 hours after 4-VO induction. Thus, it has a wider application window than tissue plasminogen activator (tPA, 3 h) [49], the only approved drug for ischemic stroke.

In vitro studies

Pinocembrin has been shown to increase neuronal viability, decrease lactate dehydrogenase release, inhibit the production of NO and ROS, increase glutathione levels, and downregulate the expression of neuronal NO synthase (nNOS) and iNOS in primary cortical neurons subjected to oxygen–glucose deprivation/reoxygenation (OGD/R) [33]. These results indicate that pinocembrin is neuroprotective in vitro.

Pinocembrin is also able to regulate mitochondrial function and apoptosis. In primary neurons subjected to OGD/R, pinocembrin decreased caspase-3 expression and increased PARP degradation [33]. The decrease in caspase-3 activity has also been demonstrated in tunicamycin-induced SH-Sy5y cells [31]. Chinese propolis, which contains pinocembrin, protects against neuronal toxicity induced by ER stress [31]. In the glutamate-induced SH-Sy5y cell line, it has been shown that pinocembrin decreases the release of cytochrome c from mitochondria into cytoplasm and reduces the synthesis of pro-apoptotic Bax.

Pinocembrin also protects the BBB in vitro. Meng et al. [48] found that pinocembrin protected cultured rat cerebral microvascular endothelial cells from OGD/R damage and increased mitochondrial membrane potential. These results suggest that pinocembrin exerts not only neuroprotection but also vascular protection, further supporting its therapeutic application in stroke.

Taken together, increasing evidence from in vivo and in vitro studies supports the idea that pinocembrin could be a candidate drug to treat ischemic stroke. Although the neuroprotective effects of pinocembrin have not been compared directly with other herbal products, acute toxicity testing showed that the LD50 of i.v. pinocembrin in mice is greater than 700 mg/kg (unpublished work by Drs. Song Wu and Guanhua Du), suggesting that pinocembrin is an exceptionally safe drug candidate. In addition, pinocembrin may have a wider therapeutic window (up to 6 hours) than the current 3-hour therapeutic window of tPA.

Effects of pinocembrin in animal models of middle cerebral artery occlusion (MCAO)

The first stroke model used to evaluate the efficacy of pinocembrin was MCAO. In rats with permanent cerebral ischemia (pMCAO), Gao et al. [34] showed that pinocembrin (10 mg/kg, i.v.) could reduce brain swelling; improve behavioral deficits; and alleviate neuronal apoptosis, edema of astrocytic end-feet, and the deformation of endothelial cells and capillaries. Moreover, pinocembrin also protected the BBB by decreasing matrix metalloproteinase-9 protein expression and reducing mRNA level of the tight junction protein zonula occludens-1. Acting on the neurovascular unit, pinocembrin reduces the level of proinflammatory cytokines [tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1β)], chemokines [intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1)], inducible nitric oxide (NO) synthase (iNOS), and aquaporin-4 [5], suggesting that pinocembrin has an anti-inflammatory effect. In a recent study, Wang et al. [35] co-administered pinocembrin and antagonists of epoxyeicosatrienoic acids (EETs) to pMCAO rats. Results showed that pinocembrin suppressed soluble epoxide hydrolase (sEH) activity and that the EET antagonist weakened the beneficial effects of pinocembrin in pMCAO rats. These results indicate that sEH and the EETs might be targets of pinocembrin. sEH metabolizes EETs into less-active dihydroxyeicosatrienoic acids (DHETs) [3638]. Because of multiple protective properties [39,40], increasing the level of EETs would be beneficial for injured brain. In mouse and human brain, sEH is expressed in multiple brain cell types, such as neurons, astrocytes, and endothelial cells [39,41]. sEH deletion or inhibition has shown protective effects in both ischemic stroke models [4245] and neurons subjected to oxygen-glucose deprivation [44,46].

Pinocembrin (10 mg/kg) was also shown to improve ischemic outcomes in the MCAO-reperfusion animal model [33]. It was suggested that pinocembrin reduces endoplasmic reticulum (ER) stress and apoptosis by decreasing C/EBP homologous protein (CHOP)/GADD153 and caspase-12 expression via the PERK-elF2α-ATF4 signaling pathway in MCAO rats [47].

Effects of pinocembrin on 4-vessel occlusion (4-VO) animal model

In a 4-VO rat model, 5 mg/kg pinocembrin protected the BBB by regulating the pathological changes of ultrastructure in microvessels, neurons, and glial cells [48]. In addition, Shi et al. [30] further reported that pinocembrin had an antioxidant effect in 4-VO rats. Pinocembrin reduces the compensatory increase of superoxide dismutase activity, malondialdehyde content, and myeloperoxidase activity; modulates the concentration of excitatory and inhibitory amino acids; and lessens the neuronal loss in rats after global cerebral 4-VO ischemia in a dose-dependent manner (1, 5, 10 mg/kg). In terms of the therapeutic time window, it is notable that pinocembrin was still protective even if administered up to 6 hours after 4-VO induction. Thus, it has a wider application window than tissue plasminogen activator (tPA, 3 h) [49], the only approved drug for ischemic stroke.

In vitro studies

Pinocembrin has been shown to increase neuronal viability, decrease lactate dehydrogenase release, inhibit the production of NO and ROS, increase glutathione levels, and downregulate the expression of neuronal NO synthase (nNOS) and iNOS in primary cortical neurons subjected to oxygen–glucose deprivation/reoxygenation (OGD/R) [33]. These results indicate that pinocembrin is neuroprotective in vitro.

Pinocembrin is also able to regulate mitochondrial function and apoptosis. In primary neurons subjected to OGD/R, pinocembrin decreased caspase-3 expression and increased PARP degradation [33]. The decrease in caspase-3 activity has also been demonstrated in tunicamycin-induced SH-Sy5y cells [31]. Chinese propolis, which contains pinocembrin, protects against neuronal toxicity induced by ER stress [31]. In the glutamate-induced SH-Sy5y cell line, it has been shown that pinocembrin decreases the release of cytochrome c from mitochondria into cytoplasm and reduces the synthesis of pro-apoptotic Bax.

Pinocembrin also protects the BBB in vitro. Meng et al. [48] found that pinocembrin protected cultured rat cerebral microvascular endothelial cells from OGD/R damage and increased mitochondrial membrane potential. These results suggest that pinocembrin exerts not only neuroprotection but also vascular protection, further supporting its therapeutic application in stroke.

Taken together, increasing evidence from in vivo and in vitro studies supports the idea that pinocembrin could be a candidate drug to treat ischemic stroke. Although the neuroprotective effects of pinocembrin have not been compared directly with other herbal products, acute toxicity testing showed that the LD50 of i.v. pinocembrin in mice is greater than 700 mg/kg (unpublished work by Drs. Song Wu and Guanhua Du), suggesting that pinocembrin is an exceptionally safe drug candidate. In addition, pinocembrin may have a wider therapeutic window (up to 6 hours) than the current 3-hour therapeutic window of tPA.

Neurodegenerative diseases

Investigators have also explored the effect of pinocembrin on Alzheimer’s disease in vitro and in vivo. Liu et al. [50] used three types of cells for in vitro studies: cells transfected with the receptor for advanced glycation end products (RAGE); an SH-Sy5y cell line that overexpresses amyloid precursor protein with the Swedish mutation (APPsw); and neurons induced with amyloid-b peptide (Aβ)25-35. They demonstrated that pinocembrin (10 μM) inhibits RAGE and its downstream signaling pathways, indicating that the RAGE protein might be a target of pinocembrin, and that pinocembrin also regulates mitochondrion-mediated apoptosis in the APPsw SH-Sy5y cell line. Liu et al. [51] further showed that, in APP/PS1 transgenic mice, oral administration of pinocembrin significantly reduces learning and memory deficits, mainly by inhibiting glial activation and downregulating RAGE-induced p38 mitogen-activated protein kinase (MAPK) expression in APP/PS1 transgenic mice. These data provide strong evidence that pinocembrin might protect the brain by targeting neuroinflammation. Finally, pinocembrin was reported to decrease neurotoxicity and inhibit mitochondrial dysfunction in 1-methyl-4-phenylpyridinium (MPP(+))–treated SH-Sy5y cells [52] and to attenuate neuronal death through the Nrf2/ARE pathway in 6-OHDA-treated SH-Sy5y cells [53], suggesting a potential application in Parkinson’s disease.

Cardiovascular diseases and atherosclerosis

Pinocembrin is thought to affect cardiovascular diseases based on its ability to regulate ApoE and reduce rho kinase (ROCK). Li et al. [54] demonstrated that pinocembrin inhibits the angiotensin II-induced increase in phosphorylation of MYPT1/Thr696 and ROCK1. The inhibition of vascular contraction caused by pinocembrin is mediated, at least in part, by reduction of MYPT1/Thr696 phosphorylation and ROCK1 expression. Moreover, pinocembrin has been shown to inhibit angiotensin II-induced vasoconstriction by suppressing the increase in [Ca] and ERK1/2 activation and blocking angiotensin II type I receptor (AT1R) in the rat aorta [54]. Sang et al. [55] showed that combined treatment with simvastatin and pinocembrin for 14 weeks significantly decreased serum lipid levels, improved endothelial function, and reduced atherosclerosis symptoms in ApoE mice. The effect of the combination therapy was better than that with simvastatin alone. These findings suggest that pinocembrin might be used to treat atherosclerosis when combined with other drugs. Yang et al. [56] reported that pinocembrin improved the biological functions of bone marrow-derived endothelial progenitor cells (EPCs) via the PI3K/AKT/eNOS pathway. However, more evidence is needed to confirm the effects of pinocembrin on cardiovascular diseases, including atherosclerosis, both in vitro and in vivo.

Inflammatory and infectious diseases

As mentioned above, anti-inflammation is one of the main mechanisms of pinocembrin. Therefore, many experiments have explored possible applications of pinocembrin in the treatment of inflammatory diseases. In an in vitro study, Soromou et al. [57] demonstrated that pinocembrin inhibits proinflammatory cytokines in the murine macrophage and endotoxin-induced acute lung injury model, partly by decreasing the levels of MAPK and NF-κB activation. In vivo, pretreatment with pinocembrin (intraperitoneal, 50 mg/kg) attenuated inflammation and reduced lung injury in a murine model of lipopolysaccharide (LPS)-induced inflammation. Soromou et al. [58] also found that pretreatment with pinocembrin (intraperitoneal, 20 mg/kg or 50 mg/kg) reduced mortality from LPS-induced endotoxin shock in mice; however, posttreatment with pinocembrin failed to exert any therapeutic effects.

Pinocembrin was first discovered to have anti-fungal properties in 1977 [59]. In the decades since, studies have shown that pinocembrin can significantly inhibit the activity of Penicillium italicum [60], Candida albicans [6165], Staphylococcus aureus [66,67,61,68,69,65,11], E. coli [70,61], Bacillus subtilis, Trichophyton mentagrophytes, Streptococcus mutans [71,65,19], and Neisseria gonorrhoeae [72]. Recently, Soromou et al. [66] reported that pinocembrin reduced α-haemolysin production and attenuated α-haemolysin-mediated cell injury at low concentrations and protected mice from S. aureus-induced pneumonia.

Together, these findings provide evidence regarding pinocembrin’s anti-inflammatory mechanisms and highlight its anti-infectious effect, suggesting that pinocembrin could be a drug candidate for treating post-stroke infections.

Discussion and prospect analysis

As a natural compound with good pharmacological and pharmaceutical properties, pinocembrin has wide applications, including ischemic stroke, neurodegenerative diseases, and atherosclerosis (Fig. 2).

An external file that holds a picture, illustration, etc.
Object name is nihms670117f2.jpg

Potential therapeutic applications of pinocembrin. In the central nervous system (CNS), pinocembrin is a drug candidate for ischemic stroke, but it also exerts protection against Alzheimer’s disease (AD) and Parkinson’s disease (PD). Pinocembrin has been shown to exert anti-inflammatory and anti-infectious effects (including against bacterial and fungal infections), indicating that it might be useful for treating post-stroke infections. In addition, pinocembrin might have potential application in the treatment of hemorrhagic stroke. In the circulatory system, pinocembrin reduces atherosclerosis (AS) symptoms in animals when combined with simvastatin.

As discussed above, pinocembrin has multiple protective properties (Fig.3). 1) In the CNS, pinocembrin primarily exerts anti-inflammatory activity by inhibiting NF-κB transcription, MAPK signaling pathways, and their downstream gene transcription (pro-inflammatory cytokines). Among these pathways, RAGE might be an important target of pinocembrin to treat neurodegenerative diseases such as Alzheimer’s disease or Parkinson’s disease. Second, pinocembrin induces antioxidative effects by decreasing superoxide dismutase, malondialdehyde, myeloperoxidase, and ROS. Pinocembrin also inhibits apoptosis by decreasing the synthesis of pro-apoptotic Bax, reducing caspase-3 activity, and reducing ER stress. These actions are mediated by decreasing CHOP/GADD153 and caspase-12 expression via the PERK-elF2α-ATF4 signaling pathway. Third, pinocembrin might inhibit sEH expression/activity in astrocytes that release high levels of EETs to modulate microglial phenotype and function. 2) In the circulatory system, pinocembrin primarily inhibits vasoconstriction by downregulating the phosphorylation of MYPT and ERK1/2, the expression of ROCK1, and calcium levels. It also improves endothelial function via the PI3K/AKT/eNOS pathway. 3) In other diseases, pinocembrin exhibits anti-inflammatory and anti-infectious effects and might be used to treat microbial-induced diseases and post-stroke infection.

An external file that holds a picture, illustration, etc.
Object name is nihms670117f3.jpg

Main mechanisms of pinocembrin. Clockwise from top: Based on their multiple protective properties, high levels of epoxyeicosatrienoic acids (EETs) released from astrocytes could modulate microglial, neuronal, and oligodendrocyte activity to benefit the injured brain. However, EETs can be metabolized by sEH into less-active dihydroxyeicosatrienoic acids (DHETs). Pinocembrin might inhibit sEH expression/activity and thereby increase EET level in the brain. The effects of pinocembrin on the activity/function of different cell types (neurons, astrocytes, microglia, or oligodendrocytes) and the role of sEH in these cells or cell-cell interactions needs further study. To modulate mitochondrial function, pinocembrin reduces endoplasmic reticulum stress via C/EBP homologous protein (CHOP) and caspase-12, and decreases apoptosis of neurons by suppressing caspase-3 expression/activity. Pinocembrin decreases oxidation, as evidenced by the inhibition of superoxide dismutase (SOD), malondialdehyde (MDA), myeloperoxidase (MPO), and reactive oxygen species (ROS). Pinocembrin decreases expression of the receptor for advanced glycation end products (RAGE) and regulates its downstream targets, including NF-κB and MAPK pathways, to exert an anti-inflammatory effect in macrophages. In the circulatory system, pinocembrin inhibits vasoconstriction via extracellular-signal-related kinase (ERK), Rho-activated kinases (ROCK), and PI3K pathways.

Although pinocembrin has many potential applications, it is currently being tested for ischemic stroke in Phase II clinical trials. Thus, it is closest to clinical application for this condition. However, many questions remain concerning potential targets and applications of pinocembrin. 1) Although several signaling pathways have been shown to mediate the effects of pinocembrin, the cellular and molecular targets of pinocembrin in stroke still need further study, in particular, the role of the EET-sEH pathway. 2) As we reviewed above, one of the most important mechanisms of pinocembrin is anti-inflammation, but the exact effect of pinocembrin on microglial and macrophage function is still unknown. This uncertainty limits the further application of pinocembrin in CNS diseases. 3) With anti-inflammatory, anti-oxidant, and neuroprotective properties, pinocembrin might have additional applications, such as in hemorrhagic stroke. Inflammation plays an important role in the development of secondary brain injury after hemorrhagic stroke [73]. We suggest that future studies should focus on the cellular and molecular targets of pinocembrin and explore other possible applications for CNS diseases.

Acknowledgments

This work was supported by an AHA Mid-Atlantic Affiliate Grant-in-Aid 13GRNT15730001 and NIH grants K01AG031926, R01NS078026, and R01AT007317. We thank Claire Levine, MS, ELS, for editorial assistance and the Wang lab members for insightful discussions.

Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
Address correspondence to: Jian Wang, MD, PhD, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross Bldg 370B, Baltimore, MD 21205. ude.imhj@97gnawj; Tel: +1-443-287-5490

Abstract

Pinocembrin is a natural flavonoid compound extracted from honey, propolis, ginger roots, wild marjoram, and other plants. In preclinical studies, it has shown anti-inflammatory and neuroprotective effects as well as the ability to reduce reactive oxygen species, protect the blood-brain barrier, modulate mitochondrial function, and regulate apoptosis. Considering these pharmaceutical characteristics, pinocembrin has potential as a drug to treat ischemic stroke and other clinical conditions. In this review, we summarize its pharmacologic characteristics and discuss its mechanisms of action and potential therapeutic applications.

Keywords: neuroinflammation, neuroprotection, pinocembrin, stroke
Abstract

Footnotes

Compliance with ethical standards

The authors declare no conflict of interest.

Footnotes

References

  • 1. Massaro CF, Katouli M, Grkovic T, Vu H, Quinn RJ, Heard TA, Carvalho C, Manley-Harris M, Wallace HM, Brooks PAnti-staphylococcal activity of C-methyl flavanones from propolis of Australian stingless bees (Tetragonula carbonaria) and fruit resins of Corymbia torelliana (Myrtaceae) Fitoterapia. 2014 doi: 10.1016/j.fitote.2014.03.024.] [[PubMed][Google Scholar]
  • 2. Danert FC, Zampini C, Ordonez R, Maldonado L, Bedascarrasbure E, Isla MINutritional and functional properties of aqueous and hydroalcoholic extracts from Argentinean propolis. Nat Prod Commun. 2014;9 (2):167–170.[PubMed][Google Scholar]
  • 3. Bertelli D, Papotti G, Bortolotti L, Marcazzan GL, Plessi M(1)H-NMR simultaneous identification of health-relevant compounds in propolis extracts. Phytochem Anal. 2012;23 (3):260–266. doi: 10.1002/pca.1352.] [[PubMed][Google Scholar]
  • 4. Tuchinda P, Reutrakul V, Claeson P, Pongprayoon U, Sematong T, Santisuk T, Taylor WCAnti-inflammatory cyclohexenyl chalcone derivatives in Boesenbergia pandurata. Phytochemistry. 2002;59 (2):169–173.[PubMed][Google Scholar]
  • 5. Gao M, Zhu SY, Tan CB, Xu B, Zhang WC, Du GHPinocembrin protects the neurovascular unit by reducing inflammation and extracellular proteolysis in MCAO rats. J Asian Nat Prod Res. 2010;12 (5):407–418. doi: 10.1080/10286020.2010.485129.] [[PubMed][Google Scholar]
  • 6. Stashenko EE, Martinez JR, Ruiz CA, Arias G, Duran C, Salgar W, Cala MLippia origanoides chemotype differentiation based on essential oil GC-MS and principal component analysis. J Sep Sci. 2010;33 (1):93–103. doi: 10.1002/jssc.200900452.] [[PubMed][Google Scholar]
  • 7. Diaz Napal GN, Palacios SMPhytotoxicity of secondary metabolites isolated from Flourensia oolepis S.F.Blake. Chem Biodivers. 2013;10 (7):1295–1304. doi: 10.1002/cbdv.201200204.] [[PubMed][Google Scholar]
  • 8. Leonard E, Lim KH, Saw PN, Koffas MAEngineering central metabolic pathways for high-level flavonoid production in Escherichia coli. Appl Environ Microbiol. 2007;73 (12):3877–3886. doi: 10.1128/AEM.00200-07.] [[Google Scholar]
  • 9. Miyahisa I, Funa N, Ohnishi Y, Martens S, Moriguchi T, Horinouchi SCombinatorial biosynthesis of flavones and flavonols in Escherichia coli. Appl Microbiol Biotechnol. 2006;71 (1):53–58. doi: 10.1007/s00253-005-0116-5.] [[PubMed][Google Scholar]
  • 10. Park SR, Yoon JA, Paik JH, Park JW, Jung WS, Ban YH, Kim EJ, Yoo YJ, Han AR, Yoon YJEngineering of plant-specific phenylpropanoids biosynthesis in Streptomyces venezuelae. J Biotechnol. 2009;141 (3–4):181–188. doi: 10.1016/j.jbiotec.2009.03.013.] [[PubMed][Google Scholar]
  • 11. Bremner PD, Meyer JJPinocembrin chalcone: an antibacterial compound from Helichrysum trilineatum. Planta Med. 1998;64 (8):777. doi: 10.1055/s-2006-957585.] [[PubMed][Google Scholar]
  • 12. Chang LS, Li CB, Qin N, Jin MN, Duan HQSynthesis and antidiabetic activity of 5,7-dihydroxyflavonoids and analogs. Chem Biodivers. 2012;9 (1):162–169. doi: 10.1002/cbdv.201100049.] [[PubMed][Google Scholar]
  • 13. Miyahisa I, Kaneko M, Funa N, Kawasaki H, Kojima H, Ohnishi Y, Horinouchi SEfficient production of (2S)-flavanones by Escherichia coli containing an artificial biosynthetic gene cluster. Appl Microbiol Biotechnol. 2005;68 (4):498–504. doi: 10.1007/s00253-005-1916-3.] [[PubMed][Google Scholar]
  • 14. Wu J, Du G, Zhou J, Chen JMetabolic engineering of Escherichia coli for (2S)-pinocembrin production from glucose by a modular metabolic strategy. Metab Eng. 2013;16:48–55. doi: 10.1016/j.ymben.2012.11.009.] [[PubMed][Google Scholar]
  • 15. Yuan Y, Yang QY, Tong YF, Chen F, Qi Y, Duan YB, Wu SSynthesis and enantiomeric resolution of (+/−)-pinocembrin. J Asian Nat Prod Res. 2008;10 (9–10):999–1002. doi: 10.1080/10286020802240418.] [[PubMed][Google Scholar]
  • 16. Rasul A, Millimouno FM, Ali Eltayb W, Ali M, Li J, Li XPinocembrin: a novel natural compound with versatile pharmacological and biological activities. Biomed Res Int. 2013;2013:379850. doi: 10.1155/2013/379850.] [[Google Scholar]
  • 17. Sayre CL, Takemoto JK, Martinez SE, Davies NMChiral analytical method development and application to pre-clinical pharmacokinetics of pinocembrin. Biomed Chromatogr. 2013;27 (6):681–684. doi: 10.1002/bmc.2853.] [[PubMed][Google Scholar]
  • 18. Metzner J, Bekemeier H, Schneidewind EM, Wenzel UPharmacokinetic studies of the propolis constituent pinocembrin in the rat (author’s transl) Pharmazie. 1979;34 (3):185–187.[PubMed][Google Scholar]
  • 19. Park YK, Koo MH, Abreu JA, Ikegaki M, Cury JA, Rosalen PLAntimicrobial activity of propolis on oral microorganisms. Curr Microbiol. 1998;36 (1):24–28.[PubMed][Google Scholar]
  • 20. Yang ZH, Sun X, Qi Y, Mei C, Sun XB, Du GHUptake characteristics of pinocembrin and its effect on p-glycoprotein at the blood-brain barrier in in vitro cell experiments. J Asian Nat Prod Res. 2012;14 (1):14–21. doi: 10.1080/10286020.2011.620393.] [[PubMed][Google Scholar]
  • 21. Yan B, Cao G, Sun T, Zhao X, Hu X, Yan J, Peng Y, Shi A, Li Y, Xue W, Li M, Li K, Liu YDetermination of pinocembrin in human plasma by solid-phase extraction and LC/MS/MS: application to pharmacokinetic studies. Biomed Chromatogr. 2014 doi: 10.1002/bmc.3186.] [[PubMed][Google Scholar]
  • 22. National Institutes of Health. Phase II study of pinocembrin injection to treat ischemic stroke. 2014 http://www.clinicaltrials.gov/ct2/show/?term=pinocembrin&rank=1.[PubMed]
  • 23. Yang Z, Liu R, Li X, Tian S, Liu Q, Du GDevelopment and validation of a high-performance liquid chromatographic method for determination of pinocembrin in rat plasma: application to pharmacokinetic study. J Pharm Biomed Anal. 2009;49 (5):1277–1281. doi: 10.1016/j.jpba.2009.02.030.] [[PubMed][Google Scholar]
  • 24. Liu YL, Ho DK, Cassady JM, Cook VM, Baird WMIsolation of potential cancer chemopreventive agents from Eriodictyon californicum. J Nat Prod. 1992;55 (3):357–363.[PubMed][Google Scholar]
  • 25. Weisshaar B, Jenkins GIPhenylpropanoid biosynthesis and its regulation. Curr Opin Plant Biol. 1998;1 (3):251–257.[PubMed][Google Scholar]
  • 26. Kaneko M, Hwang EI, Ohnishi Y, Horinouchi SHeterologous production of flavanones in Escherichia coli: potential for combinatorial biosynthesis of flavonoids in bacteria. J Ind Microbiol Biotechnol. 2003;30 (8):456–461. doi: 10.1007/s10295-003-0061-1.] [[PubMed][Google Scholar]
  • 27. Yan Y, Kohli A, Koffas MABiosynthesis of natural flavanones in Saccharomyces cerevisiae. Appl Environ Microbiol. 2005;71 (9):5610–5613. doi: 10.1128/AEM.71.9.5610-5613.2005.] [[Google Scholar]
  • 28. Kim BG, Lee H, Ahn JHBiosynthesis of Pinocembrin from Glucose Using Engineered Escherichia coli. J Microbiol Biotechnol. 2014;24 (11):1536–1541.[PubMed][Google Scholar]
  • 29. Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Judd SE, Kissela BM, Lackland DT, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Matchar DB, McGuire DK, Mohler ER, 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Willey JZ, Woo D, Yeh RW, Turner MB American Heart Association Statistics C, Stroke Statistics SExecutive summary: heart disease and stroke statistics-2015 update: a report from the american heart association. Circulation. 2015;131 (4):434–441. doi: 10.1161/CIR.0000000000000157.] [[PubMed][Google Scholar]
  • 30. Shi LL, Chen BN, Gao M, Zhang HA, Li YJ, Wang L, Du GHThe characteristics of therapeutic effect of pinocembrin in transient global brain ischemia/reperfusion rats. Life Sci. 2011;88 (11–12):521–528. doi: 10.1016/j.lfs.2011.01.011.] [[PubMed][Google Scholar]
  • 31. Izuta H, Shimazawa M, Tazawa S, Araki Y, Mishima S, Hara HProtective effects of Chinese propolis and its component, chrysin, against neuronal cell death via inhibition of mitochondrial apoptosis pathway in SH-SY5Y cells. J Agric Food Chem. 2008;56 (19):8944–8953. doi: 10.1021/jf8014206.] [[PubMed][Google Scholar]
  • 32. Gao M, Zhang WC, Liu QS, Hu JJ, Liu GT, Du GHPinocembrin prevents glutamate-induced apoptosis in SH-SY5Y neuronal cells via decrease of bax/bcl-2 ratio. Eur J Pharmacol. 2008;591 (1–3):73–79. doi: 10.1016/j.ejphar.2008.06.071.] [[PubMed][Google Scholar]
  • 33. Liu R, Gao M, Yang ZH, Du GHPinocembrin protects rat brain against oxidation and apoptosis induced by ischemia-reperfusion both in vivo and in vitro. Brain Res. 2008;1216:104–115. doi: 10.1016/j.brainres.2008.03.049.] [[PubMed][Google Scholar]
  • 34. Gao M, Liu R, Zhu SY, Du GHAcute neurovascular unit protective action of pinocembrin against permanent cerebral ischemia in rats. J Asian Nat Prod Res. 2008;10 (5–6):551–558. doi: 10.1080/10286020801966955.] [[PubMed][Google Scholar]
  • 35. Wang SB, Pang XB, Gao M, Fang LH, Du GHPinocembrin protects rats against cerebral ischemic damage through soluble epoxide hydrolase and epoxyeicosatrienoic acids. Chin J Nat Med. 2013;11 (3):207–213. doi: 10.1016/S1875-5364(13)60018-7.] [[PubMed][Google Scholar]
  • 36. Iliff JJ, Alkayed NJSoluble Epoxide Hydrolase Inhibition: Targeting Multiple Mechanisms of Ischemic Brain Injury with a Single Agent. Future Neurol. 2009;4 (2):179–199.[Google Scholar]
  • 37. Node K, Huo Y, Ruan X, Yang B, Spiecker M, Ley K, Zeldin DC, Liao JKAnti-inflammatory properties of cytochrome P450 epoxygenase-derived eicosanoids. Science. 1999;285 (5431):1276–1279.[Google Scholar]
  • 38. Capdevila JH, Falck JRThe CYP P450 arachidonic acid monooxygenases: from cell signaling to blood pressure regulation. Biochem Biophys Res Commun. 2001;285 (3):571–576. doi: 10.1006/bbrc.2001.5167.] [[PubMed][Google Scholar]
  • 39. Imig JD, Simpkins AN, Renic M, Harder DRCytochrome P450 eicosanoids and cerebral vascular function. Expert Rev Mol Med. 2011;13:e7. doi: 10.1017/S1462399411001773.] [[Google Scholar]
  • 40. Bellien J, Joannides REpoxyeicosatrienoic acid pathway in human health and diseases. J Cardiovasc Pharmacol. 2013;61 (3):188–196. doi: 10.1097/FJC.0b013e318273b007.] [[PubMed][Google Scholar]
  • 41. Sura P, Sura R, Enayetallah AE, Grant DFDistribution and expression of soluble epoxide hydrolase in human brain. J Histochem Cytochem. 2008;56 (6):551–559. doi: 10.1369/jhc.2008.950659.] [[Google Scholar]
  • 42. Jouihan SA, Zuloaga KL, Zhang W, Shangraw RE, Krasnow SM, Marks DL, Alkayed NJRole of soluble epoxide hydrolase in exacerbation of stroke by streptozotocin-induced type 1 diabetes mellitus. J Cereb Blood Flow Metab. 2013;33 (10):1650–1656. doi: 10.1038/jcbfm.2013.130.] [[Google Scholar]
  • 43. Zhang W, Koerner IP, Noppens R, Grafe M, Tsai HJ, Morisseau C, Luria A, Hammock BD, Falck JR, Alkayed NJSoluble epoxide hydrolase: a novel therapeutic target in stroke. J Cereb Blood Flow Metab. 2007;27 (12):1931–1940. doi: 10.1038/sj.jcbfm.9600494.] [[Google Scholar]
  • 44. Zhang W, Otsuka T, Sugo N, Ardeshiri A, Alhadid YK, Iliff JJ, DeBarber AE, Koop DR, Alkayed NJSoluble epoxide hydrolase gene deletion is protective against experimental cerebral ischemia. Stroke. 2008;39 (7):2073–2078. doi: 10.1161/STROKEAHA.107.508325.] [[Google Scholar]
  • 45. Shaik JS, Ahmad M, Li W, Rose ME, Foley LM, Hitchens TK, Graham SH, Hwang SH, Hammock BD, Poloyac SMSoluble epoxide hydrolase inhibitor trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid is neuroprotective in rat model of ischemic stroke. Am J Physiol Heart Circ Physiol. 2013;305 (11):H1605–1613. doi: 10.1152/ajpheart.00471.2013.] [[Google Scholar]
  • 46. Brenneis C, Sisignano M, Coste O, Altenrath K, Fischer MJ, Angioni C, Fleming I, Brandes RP, Reeh PW, Woolf CJ, Geisslinger G, Scholich KSoluble epoxide hydrolase limits mechanical hyperalgesia during inflammation. Mol Pain. 2011;7:78. doi: 10.1186/1744-8069-7-78.] [[Google Scholar]
  • 47. Wu CX, Liu R, Gao M, Zhao G, Wu S, Wu CF, Du GHPinocembrin protects brain against ischemia/reperfusion injury by attenuating endoplasmic reticulum stress induced apoptosis. Neurosci Lett. 2013;546:57–62. doi: 10.1016/j.neulet.2013.04.060.] [[PubMed][Google Scholar]
  • 48. Meng F, Liu R, Gao M, Wang Y, Yu X, Xuan Z, Sun J, Yang F, Wu C, Du GPinocembrin attenuates blood-brain barrier injury induced by global cerebral ischemia-reperfusion in rats. Brain Res. 2011;1391:93–101. doi: 10.1016/j.brainres.2011.03.010.] [[PubMed][Google Scholar]
  • 49. Boudreau DM, Guzauskas GF, Chen E, Lalla D, Tayama D, Fagan SC, Veenstra DLCost-effectiveness of recombinant tissue-type plasminogen activator within 3 hours of acute ischemic stroke: current evidence. Stroke. 2014;45 (10):3032–3039. doi: 10.1161/STROKEAHA.114.005852.] [[PubMed][Google Scholar]
  • 50. Liu R, Wu CX, Zhou D, Yang F, Tian S, Zhang L, Zhang TT, Du GHPinocembrin protects against beta-amyloid-induced toxicity in neurons through inhibiting receptor for advanced glycation end products (RAGE)-independent signaling pathways and regulating mitochondrion-mediated apoptosis. BMC Med. 2012;10:105. doi: 10.1186/1741-7015-10-105.] [[Google Scholar]
  • 51. Liu R, Li JZ, Song JK, Zhou D, Huang C, Bai XY, Xie T, Zhang X, Li YJ, Wu CX, Zhang L, Li L, Zhang TT, Du GHPinocembrin improves cognition and protects the neurovascular unit in Alzheimer related deficits. Neurobiol Aging. 2014;35 (6):1275–1285. doi: 10.1016/j.neurobiolaging.2013.12.031.] [[PubMed][Google Scholar]
  • 52. Wang Y, Gao J, Miao Y, Cui Q, Zhao W, Zhang J, Wang HPinocembrin Protects SH-SY5Y Cells Against MPP(+)-Induced Neurotoxicity Through the Mitochondrial Apoptotic Pathway. J Mol Neurosci. 2014;53 (4):537–545. doi: 10.1007/s12031-013-0219-x.] [[PubMed][Google Scholar]
  • 53. Jin X, Liu Q, Jia L, Li M, Wang XPinocembrin Attenuates 6-OHDA-induced Neuronal Cell Death Through Nrf2/ARE Pathway in SH-SY5Y Cells. Cell Mol Neurobiol. 2014 doi: 10.1007/s10571-014-0128-8.] [[PubMed][Google Scholar]
  • 54. Li L, Yang HG, Yuan TY, Zhao Y, Du GHRho kinase inhibition activity of pinocembrin in rat aortic rings contracted by angiotensin II. Chin J Nat Med. 2013;11 (3):258–263. doi: 10.1016/S1875-5364(13)60025-4.] [[PubMed][Google Scholar]
  • 55. Sang H, Yuan N, Yao S, Li F, Wang J, Fang Y, Qin SInhibitory effect of the combination therapy of simvastatin and pinocembrin on atherosclerosis in ApoE-deficient mice. Lipids Health Dis. 2012;11:166. doi: 10.1186/1476-511X-11-166.] [[Google Scholar]
  • 56. Yang N, Qin S, Wang M, Chen B, Yuan N, Fang Y, Yao S, Jiao P, Yu Y, Zhang Y, Wang JPinocembrin, a major flavonoid in propolis, improves the biological functions of EPCs derived from rat bone marrow through the PI3K-eNOS-NO signaling pathway. Cytotechnology. 2013;65 (4):541–551. doi: 10.1007/s10616-012-9502-x.] [[Google Scholar]
  • 57. Soromou LW, Chu X, Jiang L, Wei M, Huo M, Chen N, Guan S, Yang X, Chen C, Feng H, Deng XIn vitro and in vivo protection provided by pinocembrin against lipopolysaccharide-induced inflammatory responses. Int Immunopharmacol. 2012;14 (1):66–74. doi: 10.1016/j.intimp.2012.06.009.] [[PubMed][Google Scholar]
  • 58. Soromou LW, Jiang L, Wei M, Chen N, Huo M, Chu X, Zhong W, Wu Q, Balde A, Deng X, Feng HProtection of mice against lipopolysaccharide-induced endotoxic shock by pinocembrin is correlated with regulation of cytokine secretion. J Immunotoxicol. 2014;11 (1):56–61. doi: 10.3109/1547691X.2013.792886.] [[PubMed][Google Scholar]
  • 59. Metzner J, Schneidewind EM, Friedrich EEffect of propolis and pinocembrin on fungi. Pharmazie. 1977;32 (11):730.[PubMed][Google Scholar]
  • 60. Sala A, Recio MC, Schinella GR, Manez S, Giner RM, Cerda-Nicolas M, Rosi JLAssessment of the anti-inflammatory activity and free radical scavenger activity of tiliroside. Eur J Pharmacol. 2003;461 (1):53–61.[PubMed][Google Scholar]
  • 61. Katerere DR, Gray AI, Nash RJ, Waigh RDPhytochemical and antimicrobial investigations of stilbenoids and flavonoids isolated from three species of Combretaceae. Fitoterapia. 2012;83 (5):932–940. doi: 10.1016/j.fitote.2012.04.011.] [[PubMed][Google Scholar]
  • 62. Metzner J, Schneidewind EMEffect of pinocembrin on the course of experimental candida infections in mice. Mykosen. 1978;21 (8):257–262.[PubMed][Google Scholar]
  • 63. Metzner J, Bekemeier H, Paintz M, Schneidewind EOn the antimicrobial activity of propolis and propolis constituents (author’s transl) Pharmazie. 1979;34 (2):97–102.[PubMed][Google Scholar]
  • 64. Lopez A, Ming DS, Towers GHAntifungal activity of benzoic acid derivatives from Piper lanceaefolium. J Nat Prod. 2002;65 (1):62–64.[PubMed][Google Scholar]
  • 65. Uzel A, Sorkun K, Oncag O, Cogulu D, Gencay O, Salih BChemical compositions and antimicrobial activities of four different Anatolian propolis samples. Microbiol Res. 2005;160 (2):189–195.[PubMed][Google Scholar]
  • 66. Soromou LW, Zhang Y, Cui Y, Wei M, Chen N, Yang X, Huo M, Balde A, Guan S, Deng X, Wang DSubinhibitory concentrations of pinocembrin exert anti-Staphylococcus aureus activity by reducing alpha-toxin expression. J Appl Microbiol. 2013;115 (1):41–49. doi: 10.1111/jam.12221.] [[PubMed][Google Scholar]
  • 67. Peralta MA, Calise M, Fornari MC, Ortega MG, Diez RA, Cabrera JL, Perez CA prenylated flavanone from Dalea elegans inhibits rhodamine 6 G efflux and reverses fluconazole-resistance in Candida albicans. Planta Med. 2012;78 (10):981–987. doi: 10.1055/s-0031-1298627.] [[PubMed][Google Scholar]
  • 68. Estevinho L, Pereira AP, Moreira L, Dias LG, Pereira EAntioxidant and antimicrobial effects of phenolic compounds extracts of Northeast Portugal honey. Food Chem Toxicol. 2008;46 (12):3774–3779. doi: 10.1016/j.fct.2008.09.062.] [[PubMed][Google Scholar]
  • 69. Drewes SE, van Vuuren SFAntimicrobial acylphloroglucinols and dibenzyloxy flavonoids from flowers of Helichrysum gymnocomum. Phytochemistry. 2008;69 (8):1745–1749. doi: 10.1016/j.phytochem.2008.02.022.] [[PubMed][Google Scholar]
  • 70. Hegazi AG, Abd El Hady FK, Abd Allah FAChemical composition and antimicrobial activity of European propolis. Z Naturforsch C. 2000;55 (1–2):70–75.[PubMed][Google Scholar]
  • 71. Barrientos L, Herrera CL, Montenegro G, Ortega X, Veloz J, Alvear M, Cuevas A, Saavedra N, Salazar LAChemical and botanical characterization of Chilean propolis and biological activity on cariogenic bacteria Streptococcus mutans and Streptococcus sobrinus. Braz J Microbiol. 2013;44 (2):577–585. doi: 10.1590/S1517-83822013000200038.] [[Google Scholar]
  • 72. Ruddock PS, Charland M, Ramirez S, Lopez A, Neil Towers GH, Arnason JT, Liao M, Dillon JAAntimicrobial activity of flavonoids from Piper lanceaefolium and other Colombian medicinal plants against antibiotic susceptible and resistant strains of Neisseria gonorrhoeae. Sex Transm Dis. 2011;38 (2):82–88. doi: 10.1097/OLQ.0b013e3181f0bdbd.] [[PubMed][Google Scholar]
  • 73. Wang JPreclinical and clinical research on inflammation after intracerebral hemorrhage. Prog Neurobiol. 2010;92 (4):463–477.[Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.