You are on page 1of 11

Phytomedicine 65 (2019) 153090

Contents lists available at ScienceDirect

Phytomedicine
journal homepage: www.elsevier.com/locate/phymed

Original Article

Combined extracts of Echinacea angustifolia DC. and Zingiber officinale Roscoe T


in softgel capsules: Pharmacokinetics and immunomodulatory effects
assessed by gene expression profiling
Stefano Dall'Acquaa, Iztok Grabnarb, Roberto Verardoc, Enio Klaricc, Luigi Marchionnid,
Eddie Luidy-Imadad,e, Stefania Sutf, Chiara Agostinisg, Roberta Bullah, Beatrice Perissuttii,

Dario Voinovichi,
a
Department of Pharmaceutical Sciences, University of Padova, Via F. Marzolo 5, 35131, Padova, Italy
b
Faculty of Pharmacy, University of Ljubljana, Askerceva 7, SI-1000 Ljubljana, Slovenia
c
National Laboratory of the Interuniversity Consortium for Biotechnology, Area Science Park – Padriciano 99, 34149, Trieste, Italy
d
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
e
Department of Biochemistry and Immunology, ICB, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
f
Department of Agronomy, Food, Natural Resources, Animals and the Environment, Viale dell'Università 16 - 35020 Legnaro, Pd, Italy
g
Institute for Maternal and Child Health, IRCCS Burlo Garofolo, via dell'Istria 65/1, 34143, Trieste, Italy
h
Department of Life Sciences, University of Trieste, via Valerio, 28, 34127, Trieste, Italy
i
Department of Chemical and Pharmaceutical Sciences, University of Trieste, P.le Europa 1, 34127 Trieste, Italy

A R T I C LE I N FO A B S T R A C T

Keywords: Background and objective: Echinacea angustifolia DC. and Zingiber officinale Roscoe are two natural products with
Combination documented immunomodulatory activity, both able to modulate the expression of important immune-related
Echinacea angustifolia DC genes. Thus, their use in combination seems to be particularly promising. In this context, we have considered the
Zingiber officinale Roscoe oral supplementation of a highly standardized lipophilic extract combining both above-mentioned phytocom-
Lipophilic extract
plexes, formulated in attractive softgel capsules, with two objectives: on the one hand to study oral pharma-
Pharmacokinetics
cokinetic of main active extracts’ components and on the other hand to examine the immunomodulation and
Gene expression analysis
anti-inflammatory properties by gene expression profiling.
Methods: Softgel capsules containing a combination of E. angustifolia DC. and Z. officinale Roscoe (5 mg and
25 mg, respectively) were given by oral administration to 10 healthy volunteers. The plasma concentrations of
dodeca-2E,4E,8Z,10E/Z-tetraenoic isobutylamide (tetraene) for E. angustifolia DC., 6-gingerol and 6-shogaol
(free and glucuronide) for Z. officinale Roscoe were determined by LC-MS analysis, and the pharmacokinetic
analysis was performed. To understand the functional mechanisms responsible for the documented health
benefits, we also examined the overall transcriptional remodeling induced in the peripheral blood mononuclear
cells and performed an integrative functional analysis on the generated gene expression.
Results: All bioactive components were absorbed very rapidly, and their tmax were detected in plasma from
30 min to 1.40 h. The peak concentrations of tetraene, 6-gingerol, 6-shogaol and their glucuronide metabolites
were 14.74, 5.66, 9.25, 29.2 and 22.24 ng/ml, respectively. Integrated analysis performed on the generated gene
expression data highlighted immunomodulatory and anti-inflammatory effects similar to those exerted by

Abbreviations: ALDOA, Aldolase A; CAT, Correspondence-At-Top; CD, cluster of differentiation; cDNA, complementary DNA; CHD, coronary heart disease; CL/F,
apparent clearance; cRNA, complementary RNA; DEFA, alpha-defensin; FDR, False Discovery Rate; FGS, Functional Gene Sets; GEO, Gene Expression Omnibus;
GSEA, Gene Set Enrichment Analysis; GSE, Gene Expression Series; HNP, human neutrophil peptides; IFN-γ, interferon gamma; IL, interleukin; KEEG, Kyoto
Encyclopedia of Genes and Genomes; LEx, combined lipophilic extracts of Echinacea angustifolia DC. and Zingiber officinale Roscoe; MA-plot, application of a
Bland–Altman plot for visual representation of genomic data; MDA, melanoma differentiation associated gene; MsigDB, Broad Institute Molecular Signature
Database; NF-kB, nuclear factor kappa-light-chain-enhancer of activated B cells; PBMCs, peripheral blood mononuclear cells; PCA, principal components’ analysis;
PDZ, acronym of the first letters of the first three proteins discovered to share the domain: Post synaptic density protein, Drosophila disc large tumor suppressor and
Zonula occludens-1 protein; PMN, polymorphonuclear neutrophils; PPM1B, Protein phosphatase 1B; PRDX, Peroxiredoxin; qRT-PCR, quantitative Real Time poly-
merase chain reaction; RORA, Retinoic Acid Receptor-Related Orphan Receptor Alpha; RV, reservoir volume; SDCBP, syndecan-binding protein; Th, T helper; TNF-α,
tumor necrosis factor

Corresponding author.
E-mail address: vojnovic@units.it (D. Voinovich).

https://doi.org/10.1016/j.phymed.2019.153090
Received 14 December 2018; Received in revised form 10 September 2019; Accepted 15 September 2019
0944-7113/ © 2019 Elsevier GmbH. All rights reserved.
S. Dall'Acqua, et al. Phytomedicine 65 (2019) 153090

hydrocortisone.
Conclusion: These data demonstrated that the bioactive ingredients are highly and rapidly absorbed from softgel
capsules containing the combination of the above-mentioned lipophilic extracts, providing evidence to support
their immunomodulatory and anti-inflammatory properties. These data also help in defining the mechanistic
pathways underlying the health benefits of these plant-derived bioactive compounds.

Introduction PBMCs transcriptional profiling is a useful tool to define the im-


munomodulatory effects of different dietary interventions, we in-
Combination therapy, a treatment modality that combines two or vestigated the transcriptional effects on PBMCs gene expression in
more therapeutic agents, is rational and fashionable; lot of drugs are healthy volunteers after oral administration of combined E. angustifolia
coadministered to enhance efficacy and tolerability, avoiding multiple and Z. officinale lipophilic extracts in softgel capsules. In order to de-
administrations and improving patient compliance (EMA/CHMP/ termine the underlying functional mechanisms that correlate to re-
158268/2017). This is a marketing trend also in the field of food sup- ported health benefits of the combined lipophilic extracts, an in-
plements (Länger and Kubelka, 2001). Even though several papers re- tegrative functional analysis was performed on the generated gene
port simultaneous administration of plant extracts/compounds and expression matrices.
conventional drugs (e.g. antibiotics, chemotherapics, etc.)
(Hemalswarya and Doble, 2006; Samoilova et al., 2014; Trill et al., Materials and methods
2017), quite surprisingly in the face of the market demand, only a few
of those deal with combination therapies of plants extracts/compounds Softgel capsules (Indena, Milan, Italy), weighing 380 mg and con-
(Wagner and Ulrich-Merzenich, 2009; Mazumder et al., 2018; taining LEx. Each capsule contained 25 mg of Z. officinale lipophilic
Rondanelli et al., 2017). A further steadily growing market need in the extract (2,75 mg 6-gingerol, 0,75 mg 6-shogaol) and 5 mg of E. angu-
nutraceutical field is the quality of the plant extracts/compounds, to- stifolia lipophilic extract enriched in alkamides (0,5 mg of dodeca-
gether with the need of innovative formulations (Kaur, 2016). 2E,4E,8Z,10E/Z-tetraenoic isobutylamide). LC-MS-MS analysis was
In this scenario, to address the above-mentioned trends, we have performed to determine the content of above mentioned actives; the
considered a highly standardized combined lipophilic extract of results are reported in paragraph “LC-MS-MS analysis”.
Echinacea angustifolia DC. and Zingiber officinale Roscoe (below indicated
E. Angustifolia and Z. officinale) formulated in attractive softgel capsules. Reagents
These two natural products have documented immunomodulatory
activities that are able to modulate the expression of important im- Acetonitrile (HPLC grade, Rotisolv) and Tris buffer (Pufferan) were
mune-related genes (Altamirano-Dimas et al., 2007; Dall'Acqua et al., from Carl Roth GmbH+Co. (Karlsruhe, Germany). To obtain the pur-
2015; ified water for HPLC analysis was used a Barnstead (easy pure RF)
Grzanna et al., 2005; Randolph et al., 2003; Wang et al., 2006; compact ultrapure water purification system. Acetonitrile, formic acid,
Yu et al., 2011); hence their use in combination seems to be particularly hexane, ethyl acetate and methanol were from BDH (Liverpool, UK).
promising. Indeed, a recent clinical trial provided evidence of the fea- Tris buffer (Tris-d11, 99% D), benzanilide (98%), 6-gingerol, 6-shogaol,
sibility of their use in combination for the treatment of inflammation pelargonic acid vanillylamide and β-glucuronidase (Type IX-A from
and chronic pain: patients testified improvement of the symptoms and Escherichia coli) were purchased from Sigma-Aldrich (Milan, Italy),
functions (Rondanelli et al., 2017). dodeca-2E,4E,8Z,10E/Z-tetraenoic isobutylamide was obtained from
As for the formulation, since we already demonstrated the effect of Phytolab gmb (Vestenbergsgreuth, Germany).
the drug delivery system on the pharmacokinetics of natural products,
and in particular on Echinacea alkamides (Dall'Acqua et al., 2015), Study design
softgel capsules were chosen for this study since they had proved to be
the most bioavailable in our previous study. 10 healthy volunteers, 6 males and 4 females, between 30 and 55
Given that the content of active ingredients of both extracts was years of age, participated in this single blind study. All the participants
precisely known, we were able to monitor simultaneously their plasma were in good health, with normal liver function and without diagnosed
profiles after oral administration in healthy volunteers, and to carry out allergy to either Zingiberaceae or Compositae plants. In addition, vo-
the pharmacokinetic analysis, also in comparison to available phar- lunteers were not taking any chronic medications, must avoid smoke
macokinetic literature data of E. angustifolia lipophilic extract alone and be fasted from all food and drink except water, for 12 h prior to
(Dall'Acqua et al., 2015). As for the Z. officinale, in most clinical trials administration. All subjects gave their written consent to participate in
dry extracts have been considered with a non-measured content of ac- this study. Two softgel capsules were orally administered, equivalent to
tive ingredients, as already pointed out by Yu and coworkers (Yu et al., 5,5 mg of 6-gingerol, 1,5 mg of 6-shogaol and 1 mg of dodeca-
2011), and very limited studies have been dedicated to its pharmaco- 2E,4E,8Z,10E/Z-tetraenoic isobutylamides. Blood samples (2 × 5 ml)
kinetics as yet (Yu et al., 2011; Zick et al., 2008). Thus, this study re- were drawn from participants at baseline and at 15, 30, 45, 60, 90, 120,
presents a very remarkable contribution to the nutraceutical field. 180, 240, and 300 min. Plasma was separated immediately from blood
A second purpose of the paper was the study of the im- samples by centrifugation and stored frozen at −80 °C for analysis. The
munomodulation of this combined supplementation, once more fa- participants in the study refrained from smoking, eating and drinking
cilitated by the highly standardized combined extract and carried out until the last blood sample was taken. The study protocol was approved
by means of gene expression profiling. In this context, the rise of by the University of Trieste Human Research Ethics Committee.
genomics techniques such as whole genome transcriptome analyses
enabled the measurements of whole genome effects in immune cells Extraction of dodeca-2E,4E,8Z,10E/Z-tetraenoic isobutylamides
(Kussmann et al., 2008; Ovesná et al., 2008). Many studies have focused The plasma samples were extracted using a solid-phase extraction
on the immunomodulatory activities that such bioactive molecules or technique as previously described (Woelkart et al., 2005; Guiotto et al.,
extracts exert on specific cell types, including lymphocytes and per- 2008). Briefly, to 2 ml of plasma, 2 ml of Tris buffer and 100 μl of
ipheral blood mononuclear cells (PBMCs) isolated and in vitro cultured benzanilide solution (10 μg/ml), used as internal standard, were added
(Denzler et al., 2010; van Breda et al. 2014; Yan et al., 2013). Since and vortexed for 1 min. Subsequently, samples were centrifuged for

2
S. Dall'Acqua, et al. Phytomedicine 65 (2019) 153090

10 min at 3200 rpm using Eppendorf centrifuge 5810R (Hamburg, elution was used using acetonitrile with 0.1% formic acid and water
Germany). The supernatant was applied onto C18, 100 mg Isolute col- with 0.1% formic acid 40/60 (v/v) to 80/20 (v/v) in 13 min. As sta-
umns from International Sorbent Technology (Mid Glamorgan, UK), tionary phase a Zorbax SB-C18 Narrow Bore 2.1 × 150 mm 3.5 μm
pre-treated with 1 reservoir volume (RV) acetonitrile followed by 1 RV column from Agilent Technologies was used at a flow rate of 200 µl/
water. The C18 cartridges were placed on a VacMaster sample proces- min. For MS detection ESI interface operating in positive Ion mode was
sing station and subsequently washed with 1 RV of water under va- used. The source voltage was set at 4.8 kV, vaporizer temperature at
cuum. The dodeca-2E,4E,8Z,10E/Z-tetraenoic acid isobutylamides were 300 °C, nebulizer gas pressure was at 15 psi and helium gas was used for
eluted with 2 ml acetonitrile. The eluents were evaporated under a collision. For SRM (single reaction monitoring) mass transition m/z
stream of nitrogen at 40 °C using TurboVab LV vaporator (Zymark, 248 → m/z 149 was used for quantification of dodeca-2E,4E,8Z,10E/Z-
Hopkinton, MA, USA), the dry residue was dissolved in 100 µl acet- tetraenoic isobutylamides. The analytical procedure was validated by
onitrile: water (6:4) and 20 μl were used for Liquid chromato- analysis of quality control samples. Calibration curves were obtained by
graphy–mass spectrometry (LC/MS). plotting the dodeca-2E,4E,8Z,10E/Z-tetraenoic isobutylamides/I.S.
(Benzanilide) peak area versus the known spiked plasma concentration
Extraction of 6-gingerol and 6-shogaol of the analyte. The column was a Agilent XDB C-18 2.1 × 150 mm, the
Plasma samples (1 ml) were transferred to microcentrifuge tubes, mobile phases were water 1% Formic acid (A) and Acetonitrile (B).
then spiked with 10 μl of internal standard pelargonic acid vanillyla- Gradient started with 70% A and kept isocratic for 1 min. It then
mide working solution (10 μg/ml) and extracted with 4 ml ethyl reached 100% B after 10 min and kept isocratic for 14 min. Varian 320
acetate/hexane (1:1, v/v). Tubes were vortexed for 20 min at high TQD MS spectrometer was used as detector. The following transitions
speed and centrifuged for 15 min at 3000 rpm. The upper organic layer were used for quantification purposes, namely 6-glucuronyl-sogaol
was removed from the tubes, streamed into glass vials, and dried under (375.5→177.5), 6-glucuronyl gingerol (471.5→177.5), 6-diglcucyronyl
a stream of argon. The samples were re-suspended in 50 μl of methanol shogaol (471.5→177.5), 6-shogaol (277.1→137.5), nonivamide
for HPLC, vortexed at high speed and placed into autosampler vials for (294.4→137.3), 6-gingerol (295.5→177.5), 10-gingerol (351→177.1).
LC-ESI-TQ-MS/MS analysis, liquid chromatography triple quadrupole Spectrometer was working in positive ion mode, capillary was set at
tandem mass spectrometry (Yu et al., 2011). 5400 V, frying gas at 285 °C, nebulizer pressure at 50 psi and drying gas
pressure at 25 psi. Limit of detection was at 0.1 ng ml−1. Linearity was
Enzymatic hydrolysis of 6-gingerol and 6-shogaol conjugates obtained in the range of 0.1–110 ng ml−1.The amounts of 6-gingerol, 6-
Plasma samples (1 ml) were incubated with 200 μl of β-glucur- shogaol and dodeca-2E,4E,8Z,10E/Z-tetraenoic isobutylamide in softgel
onidase solution (5 mg/ml) at 37 °C for 3 h, according to the method of capsules were determined using the above mentioned LC-DAD-ESI-MS
Yu and coworkers (Yu et al., 2011). The samples were then mixed with method. A representative chromatogram is reported in Fig. 1. The dose
10 μl of internal standard PAV working solution (10 μg/ml) and ex- (corresponding to 2 softgel capsules) contained 5.51 ± 0.02 mg of 6-
tracted through the procedure given above. gingerol, 1.53 ± 0.02 mg of 6-shogaol and 1.00 ± 0.02 mg of dodeca-
2E,4E,8Z,10E/Z-tetraenoic isobutylamide (mean ± S.D., n = 3)
LC-MS-MS analysis
Pharmacokinetics analysis
Alkamides concentrations in samples were determined by LC/MS
analysis as previously described by Woelkart et al., 2005, using LC-ESI- Non-compartmental pharmacokinetic analysis of dodeca-
IT-MS/MS (liquid chromatography/electrospray ionization tandem 2E,4E,8Z,10E/Z-tetraenoic isobutylamide, 6-gingerol, 6-shogaol, 6-
mass spectrometry) composed of Varian 212 binary chromatograph, gingerol glucuronide, and 6-shogaol glucuronide was performed using
equipped with a Prostar 430 thermostatic autosampler and Varian 500 WinNonlin Version 2.1 software from Pharsight Corporation (Mountain
ion trap mass spectrometer with Varian Workstation software. Gradient View, CA). Maximum plasma concentration (Cmax) and time (tmax) were

Fig. 1. Representative HPLC-DAD chromatogram of the used product; peaks assigned to the active constituents highlighted in the study are reported.

3
S. Dall'Acqua, et al. Phytomedicine 65 (2019) 153090

reported as observed, when obtained. Terminal half-life (t1/2) was Molecular Signature Database (MsigDB) (Liberzon et al., 2011). In the
calculated as t1/2 = ln2 / λz, where λz is the terminal slope of the present study we focused on: a) chemical and genetic perturbations,
plasma concentration profile on the semi-log plot calculated by linear signaling pathways from KEGG (Kanehisa et al., 2006), Reactome
regression. The area under the concentration-time curve (AUC) was (Croft et al., 2014), and Biocarta (http://www.genecarta.com); b) bio-
calculated using the linear trapezoidal method and extrapolated to in- logical processes from Gene Ontology (Ashburner et al., 2000); c) cis-
finity (AUCinf), as AUC + Clast/λz, where Clast is the predicted con- regulatory motifs – corresponding to genes sharing microRNA seed
centration at the last quantified time point. Mean residence time (MRT) sequences or transcription factor binding sites (Xie et al., 2005) and d)
was calculated as AUMCinf/AUCinf, where AUMCinf is the area under the manually curated Functional Gene Sets (FGS) corresponding to funda-
first moment curve. Since fractions of absorbed dose (F) cannot be es- mental biological processes (‘Hallmarks’ FGS), immunological expres-
timated based exclusively on post-extravascular administration data, sion profiles, and oncogenic signatures derived from peer reviewed
apparent clearance (CL/F), calculated as Dose/AUCinf, and apparent manuscripts. Enrichment across contrasts and studies was compared
volume of distribution based on terminal phase (Vz/F), calculated as using Correspondence At Top (CAT) plots and the log10-transformed
CL/F/λz, are reported. FDR as previously described (Marchionni et al., 2017).
Dose normalized AUClast (AUC till the last quantified time point)
values from the current study were compared to the AUClast values from Gene expression validation by qRT-PCR
our previous pharmacokinetic study with Echinacea lipophilic extract Total RNA was extracted using RNeasy Mini Kit (QIAGEN) ac-
formulated as softgel capsules (Dall'Acqua et al., 2015) and the phar- cording to manufacturer's instructions. The purity of RNA samples was
macokinetic parameters of active ingredients of dry Ginger extract from estimated with the Nanodrop spectrophotometer (ThermoFisher
a study by Yu and co-workers (Yu et al., 2011) using independent Scientific) using the ratio of absorbance values at 260 nm and 280 nm.
samples t-test. 500 ng of total RNA were retrotranscribed with the QuantiTect Reverse
Transcription Kits (QIAGEN). For each sample SYBR® green real-time
Microarray data generation qPCR was performed in triplicate using the SYBR® Green PCR Master
Total RNA was extracted using the RNeasy Mini Kit (Qiagen), ac- Mix (ThermoFisher Scientific) on the StepOnePlus Real-Time PCR
cording to the manufacturer's protocol. Synthesis of cDNA and bioti- SYstem (ThermoFisher Scientific). The amplification protocol included
nylated cRNA (from 500 ng total RNA) was performed using the a melting curve dissociation step to confirm the inexistence of non-
Illumina TotalPrep RNA Amplification Kit (Ambion), according to the specific amplification products. Gene expression data of the selected
manufacturer's protocol. Quality assessment and quantitation of total target genes were normalized to the housekeeping gene β-actin. The
RNA and cRNAs were performed with Agilent RNA kits on a analysis was performed using the comparative Ct method (2−ΔΔCt) and
Bioanalyzer 2100 System (Agilent). Hybridization of cRNAs (750 ng) the fold change was calculated from normalized Ct values.
was carried out using Illumina Human 48 K gene chips (Human HT-12
v4 Expression BeadChip). Array chip washing was performed in High Results
Temp Wash Buffer (Illumina) at 55 °C for 10 min, followed by staining
using streptavidin-Cy3 dyes (Amersham Biosciences). Hybridized ar- Pharmacokinetic study
rays were stained and scanned in a BeadStation 500 System (Illumina).
The first part of the study aimed to assess the pharmacokinetics of
Microarray data pre-processing and analysis of differential expression the active ingredients of a highly standardized product combining 2
Raw expression data were extracted using the Illumina lipophilic extracts (E. angustifolia and Z. officinale) (hereafter also re-
GenomeStudio software (GenomeStudio V2011.1) and all further ana- ferred to as LEx) in softgel capsules. The major components, including
lyses were performed using state-of-the-art ‘lumi’ (Lin et al., 2008) and dodeca-2E,4E,8Z,10E/Z-tetraenoic isobutylamides, 6-gingerol, 6-sho-
‘limma’ software packages available through the R/Bioconductor pro- gaol and two conjugate metabolites, 6-gingerol glucuronide, and 6-
ject. Briefly, after routine quality assessment (i.e., PCA plots, hier- shogaol glucuronide, were quantified in all plasma samples at pre-
archical clustering plots, MA-plots, and box plots), raw expression data determined time points up to 4 h after administration. Mean plasma
were run through quantile normalization without background correc- concentration profiles are shown in Fig. 2. Results of the non-com-
tion. A generalized linear model approach, coupled with empirical partmental pharmacokinetic analysis are summarized in Table 1.
Bayes moderation of standard errors (Smyth, 2004) was used for
identifying differentially expressed genes upon treatment taking into Transcriptional remodeling of PBMCs following lipophilic extracts of E.
account sample matching status. Correction for multiple testing was angustifolia and Z. officinale administration
performed using the Benjamini–Hochberg method (Benjamini and Differentially expressed genes. Peripheral blood mononuclear cells
Hochberg 1995). (PBMCs), primarily composed of T-lymphocytes, B-lymphocytes, NK
To compare the oral administration effects of the softgel capsules cells, monocytes, macrophages and dendritic cells, are often considered
containing LEx to other studies of interest, we analyzed, using the same a standard in studying many aspects of immunological responses,
approach described above, the following publicly available datasets: including the inflammatory response. To investigate the
GSE14368, GSE34145 (Kolehmainen et al., 2012) GSE40838 transcriptional modulatory effect of oral administration of softgel
(Boisson et al., 2012) and GSE67255 (Olnes et al., 2016). Normalized capsules containing LEx, we performed genome-wide transcriptional
gene expression data were obtained from the National Center for Bio- profiling study on PBMCs isolated from 3 individuals involved in the
technology Information (NCBI) Gene Expression Omnibus (GEO) data- study. Based on plasma profiles (AUClast), three healthy volunteers were
base and treatment groups were compared as described in the original picked, having the lowest, highest and middle AUClast values. Blood was
manuscripts or the GEO record as available. Correspondence-At-Top drawn at baseline (T0) and 24 h after softgel capsule administration
plots (CAT-plots) were used to compare the agreement between ranked (T24), and total RNA from the isolated PMBCs was used in the DNA
vectors of differential gene expression across studies (Ross et al., 2011). microarray-based differential gene expression analysis study. Timing of
blood collection was chosen based on previous results (Dall'Acqua et al.,
Gene set enrichment analysis (GSEA) 2015). Based on the gene expression matrices generated, we compared
For the identification of enriched pathways and biological pro- the matched variation of gene expression between PBMCs at T0 and
cesses, Gene Set Enrichment Analysis (GSEA) (Liberzon et al., 2011) T24. The normalized expression values included for differential
was performed with 1000 permutations and weighted scoring. The expression analysis were filtered by the thresholds of FDR ≤ 0.05.
enrichment were analyzed in several collections from Broad Institute Using this criterion, 542 unique transcripts were found to be

4
S. Dall'Acqua, et al. Phytomedicine 65 (2019) 153090

Table 1
Plasma pharmacokinetic parameters of active ingredients of combined lipo-
philic extracts of Echinacea angustifolia DC. and Zingiber officinale Roscoe in 10
healthy volunteers following oral administration of lipophilic Echinacea extract
formulated as soft gelatin capsules (dose 5.51 mg of 6-gingerol, 1.53 mg of 6-
shogaol and 1.00 mg of dodeca-2E,4E,8Z,10E/Z-tetraenoic isobutylamide).
dodeca-2E,4E,8Z,10E/ 6-gingerol 6-shogaol
Z-tetraenoic (5.5 mg dose) (1.5 mg dose)
isobutylamides (1 mg
dose)

Mean SD Mean SD Mean SD

Free
Tmax (h) 0.53 0.34 0.58 0.24 1.35 0.70
Cmax (ng/ml) 14.74 10.55 5.66 1.75 9.25 1.76
AUClast (ng h/ml) 21.59 12.35 8.40 3.82 18.6 5.3
t1/2_Lambda_z (h) 0.841 0.406 3.32 2.40 2.65 1.11
AUCinf (ng h/ml) 22.42 12.01 10.75 4.77 31.6 9.0
AUClast/D (ng h/ 21.59 12.01 1.527 0.867 12.40 5.976
ml/mg)
Vz/F (l) 79.7 58.43 3438 4056 190.9 92.3
Cl/F (l/h) 61.17 38.37 627.2 352.2 50.9 13.9
MRTinf (h) 2.1 0.59 3.30 1.65 4.47 1.36
Glucuronide
Tmax (h) / / 1.40 0.80 0.875 0.680
Cmax (ng/ml) / / 29.2 10.6 22.24 2.78
AUClast (ng h/ml) / / 67.9 27.3 55.3 10.7
t1/2_Lambda_z (h) / / 1.84 0.89 3.49 1.68
AUCinf (ng h/ml) / / 83.3 29.0 112 39
AUClast/D (ng h/ / / 67.9 5.28 74.82 36.86
ml/mg)
Vz/F (l) / / 202.3 124.6 67.0 24.6
Cl/F (l/h) / / 76.9 37.6 15.3 6.6
MRTinf (h) / / 3.30 1.17 5.59 2.24

Validation of microarray results by Rt-qPCR analysis. Seven of the most


up-regulated (PPM1B, RORA) and down-regulated genes (ALDOA,
DEFA1/DEFA3, PRDX2, PRDX3, SDCBP) were chosen from the
generated list of statistically significant differentially expressed genes
(Supplementary Table S2) for validation by RT-qPCR. Fig. 3 shows the
overall agreement between the expression analysis results obtained by
the microarray technology and the RT-qPCR (shown at a single-patient
level).

Integrative functional analysis. In our analysis, we identified 542


differentially expressed genes upon oral administration of the softgel
capsules. Barring study GSE67255 (Olnes et al., 2016), which evaluated
the effects of hydrocortisone treatment in a time-course, none of the
remaining analyzed public datasets yielded significantly expressed
genes (data not shown). We therefore benchmarked our differential
expression analysis results against this study alone. First, we assessed
the overall agreement between studies for the contrasts included in our
linear model analyses. To this end, we used CAT plots, an approach
developed for comparing lists of differentially expressed genes by
Fig. 2. Mean ( ± SD) plasma concentration profiles of the active ingredients of
focusing at the top and the bottom of ranked lists. Overall, CAT-plots
Echinacea angustifolia DC. and Zingiber officinale Roscoe combined lipophilic revealed that softgel capsules' administration effects resemble those of
extracts in 10 volunteers following oral administration as softgel capsules (dose early hydrocortisone response (4 h), especially at the highest dosage
5.51 mg of 6-gingerol, 1.53 mg of 6-shogaol and 1.00 mg of dodeca- (250 mg). Indeed, the down-regulated genes were highly similar and
2E,4E,8Z,10E/Z-tetraenoic isobutylamide). the observed agreement exceeded what would be expected by chance
alone (see Supplementary Figure S1, light blue curve A). On the
differentially expressed in PBMCs after the lipophilic extracts contrary, the similarity between genes down-regulated by softgel
administration, subdivided in one transcriptional module of 293 up- capsule administration with low dosage hydrocortisone (50 mg) at a
regulated genes and one of 249 down-regulated genes (see late time point (8 h) did not exceed what would be expected by chance
Supplementary Table S1-A and B for a selected list of the most alone (see Supplementary Figure S1, red curve D). Similar results were
differentially expressed genes). Interestingly, among the most down- also obtained for the up-regulated genes (See Supplementary Figure
regulated genes in PBMCs isolated from the subjects, the bioinformatics S2).
analysis highlighted the α-defensins, DEFA1, DEFA1B and DEFA3. Among genes differentially expressed across both the administration
of the lipophilic extracts and early 250 mg hydrocortisone treatments,
there were genes related to immune response, anti-inflammatory

5
S. Dall'Acqua, et al. Phytomedicine 65 (2019) 153090

Fig. 3. Validation of selected genes differentially expressed following administration of the lipophilic extracts at two distinct time points (T0 and T24). Boxplots show
expression levels change between paired samples (3 distinct donors) analyzed by microarray (Upper Panel) and by RT-qPCR (Lower Panel). Expression levels are
expressed as normalized log2 expression intensities for microarrays and as normalized (to ACTB housekeeping gene) Ct values for RT-qPCR. Individual matching T0
and T24 measurements for each donor are connected. Our validation experiments confirmed that ALDOA, DEFA1/DEFA3, PRDX2, PRDX3, and SDCBP are down-
regulated upon treatment, while PPM1B and RORA are up-regulated. Primers pairs used for RT-qPCR are listed in Table S2).

activity, response to growth factors and hormones, and non-sense populations. Nevertheless, our findings suggest that the oral adminis-
mediated RNA decay (see heatmap gene intersection depicted in Fig. 4). tration of softgel capsules containing a LEx might have an anti-in-
Next, we compared the agreement between studies at the levels of flammatory effect, similar at a transcriptional level, to hydrocortisone's.
Functional Gene Sets (FGS). To this end, we first performed GSEA and
then we identified the FGS significantly enriched in both studies. GSEA
was performed to capture biological processes relevant in the in- Discussion
vestigated contrasts and to test whether differential gene expression
programs associated with LEx administration were similar to other In this study a highly standardized combined extract of E. angusti-
pharmacological treatments. Specifically, the GSEA approach was used folia and Z. officinale, in a modern formulation such as softgel capsules
to compute a p-value to test the hypothesis that genes contained in a was administered in healthy subjects with the aim of studying both the
Functional Gene Set (FGS), defined by a functional annotation, tend to pharmacokinetics of the main active components (and their conjugate
be more enriched in the extremes of a list (i.e. highly ranked in a list metabolites) of the two extracts and the immunomodulatory effects by
ordered by a functional measurement). In the present study we tested gene expression profiling.
whether the collection of FGS from MSigDB were significantly enriched In the first part of the research, plasma concentration of dodeca-
in the selected treatments. To achieve this, we ranked individual genes 2E,4E,8Z,10E/Z-tetraenoic isobutylamides, 6-gingerol, 6-shogaol, 6-
by their moderate t-statistics derived from the differential expression gingerol glucuronide, and 6-shogaol glucuronide were measured up to
analysis and corrected the resulting enrichment p-values for multiple 4 h after administration. In agreement with the high lipophilicity, do-
hypothesis testing using the Benjamini and Hochberg method. This deca-2E,4E,8Z,10E/Z-tetraenoic isobutylamides, 6-gingerol and 6-sho-
analysis reveals that both these treatments modulate immune related gaol are absorbed very rapidly and are detected in plasma as early as
processes, such as the regulation of interleukin-8 biosynthesis, the re- the first sampling time, i.e., after 15 min.
sponse to interferon alpha and gamma, and the response of CD4 T- Plasma concentration profile of dodeca-2E,4E,8Z,10E/Z-tetraenoic
lymphocytes to Interleukin-4 (see GSEA heatmap in Fig. 5, and Tables isobutylamides observed in the current study after supplementation of
S3-11 and related Figures S3-9 in Supplementary information). LEx resulted very similar to the plasma concentration profile of dodeca-
It is worth noting that, although PBMCs represent a heterogeneous 2E,4E,8Z,10E/Z-tetraenoic isobutylamides described in our previous
mixture of cell types, we were able to highlight an overall transcrip- study when pure Echinacea extract has been orally administered in
tional remodeling induced by the administered lipophilic extract, sug- softgel capsules (Dall'Acqua et al., 2015). These data indicated the ir-
gesting that the application of transcriptome analyses in a human relevance of Ginger extract on pharmacokinetic parameters of iso-
dietary intervention study represents a promising approach to unveil butylamides, also confirmed by comparing AUClast and AUCinf para-
mechanistic pathways linked to phytochemical-induced health effects. meters across the two studies (p = 0.316 and p = 0.490, respectively).
However, further studies are needed to better understand the exerted Since maximum concentration of 6-gingerol and 6-gingerol glucur-
effects by using more homogeneous samples, i.e. specific PBMCs sub- onide in plasma were observed after 30 min and after 2 h, respectively,
we hypothesized that the 6-gingerol reaches plasma mainly unmodified

6
S. Dall'Acqua, et al. Phytomedicine 65 (2019) 153090

Fig. 4. (Heatmap gene intersection) – Differential gene expression levels of lipophilic extracts administration (LEx) and 250 mg hydrocortisone after 4 h treatment in
contrast with control. Most genes found differentially expressed across both treatments were regulated in similar intensity and direction, indicating similar properties
of LEx and hydrocortisone (i.e. immune response modulation and anti-inflammatory activity). Color intensity and direction of differential expression are represented
by t-score derived from generalized linear models.

7
S. Dall'Acqua, et al. Phytomedicine 65 (2019) 153090

Fig. 5. (Gene Set Enrichment Analysis, GSEA) – Heat-maps visualizing significantly enriched functional gene sets (FGS) as determined by Analysis of Functional
Annotation (AFA) performed on expression profiles associated with LEx and Hydrocortisone treatment at different concentrations. Rows represent selected FGS
linked to immunomodulation, indicating that LEx treatment is most similar to the effects of Hydrocortisone treatment at 250 mg after 4 h. The color scales correspond
to the absolute adjusted p-values obtained from our analysis after negative base 10 logarithmic transformations and direction attribution (i.e., the number on the
color scale increases with decreasing FDR, negative values represent down regulated FGS and vice-versa). FGS were selected from different collections to capture
signaling pathways and biological themes modulated by treatment. The databases analyzed were collected from MSigDB. Complete tables and related figures with
results from enrichment test are reported in Table S3-11 and Figures S2-8.

and that 6-gingerol glucuronide is formed by systemic metabolism. The As for the pharmacokinetic analysis of Z. officinale, up to date lit-
conjugate metabolite, 6-shogaol glucuronide, appears more rapidly and erature data on pharmacokinetics of ginger active ingredients are very
its plasma concentration profile presents the same concentration profile scarce, therefore, no proper comparison with published papers can be
of free 6-shogaol, indicating that the conversion to the glucuronide is performed. The only paper worth of mention is by Yu and co-authors
very rapid and that 6-shogaol glucuronide is pre-systemically formed. (Yu et al., 2011), where the pharmacokinetic parameter for 6-gingerol
The secondary peak, at 2 h, is related to interconversion of 6-gingerol to glucuronide, 6-shogaol, and 6-shogaol glucuronide are reported. Ac-
6-shogaol. Such interconversion had been previously reported in si- cordingly, we compared those results with ours. The two studies display
mulated gastric fluid (Bhattarai et al., 2007). several differences, namely, a much higher dose level of ginger active
Systemic elimination of all active components was also rapid with a ingredients by Yu et al. compared to our study (52.8 mg vs. 5.5 mg for
terminal half-life ranging from 0.841 ± 0.406 h for dodeca- 6-gingerol and 45 mg vs. 1.5 mg for 6-shogaol). Likewise, Yu et al. used
2E,4E,8Z,10E/Z-tetraenoic isobutylamides to 3.49 ± 1.68 h for 6- dry ginger extracts, while - as mentioned before- we chose a lipophilic
shogaol glucuronide. These data indicate that there is no accumulation extract, which, in a previous study, had proved a promising choice to
of dodeca-2E,4E,8Z,10E/Z-tetraenoic isobutylamides, 6-gingerol, 6- improve bioavailability of natural active ingredients in
shogaol and their glucuronides with the recommended once daily (Dall'Acqua et al., 2015). Dose normalized AUClast for 6-shogaol and 6-
dosing. shogaol glucuronide determined in our study (12.40 ng h/ml/mg and

8
S. Dall'Acqua, et al. Phytomedicine 65 (2019) 153090

36.87 ng h/ml/mg, respectively) were significantly higher than the contains two PDZ domains and participates in diverse biological pro-
values reported by Yu and coauthors (Yu et al., 2011) (0.53 ng h/ml/ cesses (Jiao et al., 1998). Previous studies have identified SDCBP as a
mg, and 2.44 ng h/ml/mg, respectively). For 6-gingerol glucuronide, TNF-α- and IFN-γ inducible gene (Stier et al., 2000), and it was recently
however, there is no difference (14.02 ng h/ml/mg vs. 12.35 ng h/ml/ demonstrated that IL-6 induce SDCBP expression at both mRNA and
mg,). The reasons behind the difference in oral bioavailability may be protein levels (Cao et al., 2017). Intriguingly, the Fisher's group has
quite a few. The simultaneous presence of dodeca-2E,4E,8Z,10E/Z- highlighted a novel role of mda-9/syntenin in tumor-promoting in-
tetraenoic isobutylamides can improve the bioavailability of lipophilic flammation and immune suppression (Das et al., 2016).
compounds such as 6-shogaol and 6-gingerol through the formation of All these modulations, as summarized by Fig. 6, concurred to shift
micelles, as previously attested (Raduner et al., 2006). To verify this the leukocytes towards an anti-inflammatory phenotype.
hypothesis a pharmacokinetic study after oral administration of in- One of the most significant and interesting findings of this study has
dividual Z. officinale lipophilic extract is ongoing. emerged from the integrative functional analysis, which revealed that
A further explanation of the difference in AUClast may also be the the effect of LEx is similar to that of early hydrocortisone response,
nonlinear pharmacokinetics (less than proportional increase in AUC especially at the highest hydrocortisone dosage (250 mg). On the con-
with the increase of the dose). trary, down-regulated genes by softgel capsules or by the hydro-
In conclusion of the first part of the research, we can affirm that cortisone lowest dose (50 mg) administration were not significantly
these data testify the advantages in terms of pharmacokinetic perfor- similar at 8 h (last time point).
mance of the supplementation of softgel capsules containing the lipo- Previous studies demonstrated that corticosteroids exert several
philic standardized extracts of E. angustifolia and Z. officinale in com- effects on immunity cells, including depletion of monocytes, dendritic
bination. cells, and CD4+ CD8+ thymocytes by apoptosis, and mobilization of
A further goal of this study was the investigation of the transcrip- natural killer cells and regulatory T cells (Leung and Bloom, 2003). In
tional modulatory effect of oral administration of these softgel capsules vitro studies also showed that corticosteroids down-modulate several
(containing LEx). To this aim, we performed genome-wide transcrip- inflammatory cytokines (Da Silva, 1999) and influence intracellular
tional profiling study on PBMCs isolated from the blood of three healthy cytokine expression in T cells, resulting in an increased Th2/Th1 ratio
voluntaries. We analyzed only three of ten healthy volunteers treated (Segerer et al., 2009). In vitro and animal studies demonstrate that NF-
with softgel capsules, selecting subjects who proved poor, medium and κB exerts anti-apoptotic effects through signaling, by way of down-
high absorption (based on AUClast values). This rationale owes to pre- stream effectors. One putative mechanism by which corticosteroids
viously published results testifying that the immunomodulatory effect suppress immunity is down-regulation of the NF-κB signaling, an effect
of tetraene was not related to its content in the formulation. In fact, in that enhances apoptosis. Olnes and coworkers demonstrated in 2016
an earlier study, the effects of the three different tetraene doses on the (Olnes et al., 2016) that NF-κB signaling was down-regulated as early as
decrement of a given cytokine did not differ in a significant way, nei- one h in PBMCs after systemic hydrocortisone administration, with
ther were they related to AUC (Guiotto et al., 2008). peak inhibition occurring at 4–8 h and both doses exerting a similar
More than 500 transcripts were found to be differentially expressed magnitude of inhibition.
in PBMCs after the lipophilic extracts administration, 293 up-regulated As we previously have demonstrated, the anti-inflammatory effect
genes and one of 249 down-regulated genes. Thanks to the validation of of dodeca-2E, 4Edienoic acid isobutylamide is explicated by the down-
microarray results by RT-qPCR analysis, we confirmed the up-regula- regulation, in terms of both gene and protein expression, of IL-6 and
tion of mRNA expression of PPM1B and RORA and the down-regulation TNF-α as well as of IL-8 cytokines, a major player in systemic in-
of ALDOA, DEFA1/DEFA3, PRDX2, PRDX3 and SDCBP gene expression. flammation (Dall'Acqua et al., 2015). In this study, the regulation of
As described in Fig. 6, all these genes can cooperate for the reduction of biosynthesis of IL-8 also emerged from the selected immunologic sig-
inflammation. nature analysis, a confirmation of previous data.
Interestingly, our micro-array data, also confirmed by the RT-qPCR
validation, highlighted the α-defensins, DEFA1, DEFA1B and DEFA3 Conclusion
down-regulation. Human α-defensins, also called human neutrophil
peptides (HNP-1 to 4), are particularly abundant in polymorphonuclear We can conclude that the treatment of healthy volunteers with the
neutrophils (PMNs), certain macrophage populations, and Paneth cells combination of E. angustifolia and Z. officinale, titled in isobutylamides
of the small intestine (Cunliffe, 2003). These genes codify for cysteine- and gingerols, in softgel capsules, provides several advantages from the
rich cationic polypeptides, produced both constitutively and/or in re- pharmacokinetic and therapeutic points of view. Due to the individual
sponse to microbial products or pro-inflammatory cytokines, during diffused use of these extracts as health promoting products and con-
septic inflammation and acute coronary vascular disorders. Multiple sidering the actual lack of data supporting their effectiveness in com-
studies have reported that HNP-1–3 can produce both beneficial and bination, the data contained in the present study provide novel in-
pathologic effects. Interestingly, intraperitoneal administration of HNP- formation supporting the co-administration of these natural products.
1 to mice with dextran sulfate sodium-induced colitis begets more se- The experimental evidence showed that the combined extracts allows a
vere colitis with higher colonic cytokine expression compared to con-
trols, thus suggesting a potential pro-inflammatory role for HNP-1 in
ulcerative colitis (Hashimoto et al., 2012). Authors also suggested that
patients with ulcerative colitis are likely to benefit from reducing high
expression levels of HNP-1–3. In this context, it has been demonstrated
that HNPs utilize the signaling pathways downstream of P2 receptors to
stimulate human lung and intestinal epithelial cells to produce the pro-
inflammatory cytokine IL-8 (Ibusuki et al., 2015). It has been recently
suggested that increased α-defensin expression is a potential in-
flammatory biomarker that may predict the risk of coronary heart
disease (CHD) development in hyperlipidemia patients (Maneerat et al.,
2017). Among the most down-regulated gene transcripts, we also
highlighted the Syndecan-binding protein (SDCBP) or syntenin, also
known as MDA-9 (melanoma differentiation associated gene-9), first Fig. 6. Modulation concurring to shift the leukocytes towards an anti-in-
discovered in melanoma cells (Jiao et al., 1998). This scaffold protein flammatory phenotype.

9
S. Dall'Acqua, et al. Phytomedicine 65 (2019) 153090

high and rapid absorption of the bioactive ingredients and exerts im- Denzler, K.L., Waters, R., Jacobs, B.L., Rochon, Y., Langland, J.O., 2010. Regulation of
munomodulatory and anti-inflammatory effects, at least at the tran- inflammatory gene expression in PBMCs by immunostimulatory botanicals. PLoS One
5 (9), e12561. https://doi.org/10.1371/journal.pone.0012561.
scriptional level, similarly to those exerted by hydrocortisone. The aim EMA/CHMP/158268/2017, https://www.ema.europa.eu/documents/scientific-
of our study was to perform a pilot trial to assess the effects of si- guideline/guideline-clinical-development-fixed-combination-medicinal-products-
multaneous supplementation of both extracts. As a result, the research revision-2_en.pdf.
Grzanna, R., Lindmark, L., Frondoza, C.G., 2005. Ginger-an herbal medicinal product
has several limitations such as the small number of subjects considered with broad anti-inflammatory actions. J. Med. Food. Summer 8, 125–132.
in the PK study, the even smaller number of subjects in the genomic Guiotto, P., Woelkart, K., Grabnar, I., Voinovich, D., Perissutti, B., Invernizzi, S.,
study or the single individual tested dose of active components. Granzotto, M., Bauer, R., 2008. Pharmacokinetics and immunomodulatory effects of
phytotherapeutic lozenges (bonbons) with Echinacea purpurea extract. Phytomed 1,
Nevertheless, not only do our preliminary data support the previously 547–554.
published observations regarding the pharmacokinetic and im- Hashimoto, S., Uto, H., Kanmura, S., Sakiyama, T., Oku, M., Iwashita, Y., Ibusuki, R.,
munomodulatory effect of E. angustifolia, but also, thus far, this is the Sasaki, F., Ibusuki, K., Takami, Y., Moriuchi, A., Oketani, M., Ido, A., Tsubouchi, H.,
2012. Human neutrophil peptide-1 aggravates dextran sulfate sodium-induced colitis.
first study pertaining with the combination of E. angustifolia and Z.
Inflamm. Bowel Dis. 18, 667–675.
officinale. Inasmuch as the manuscript considers both pharmacokinetic Hemalswarya, S., Doble, M., 2006. Potential synergism of natural products in the treat-
issues and immunomodulatory effects assessed by gene expression ment of cancer. Phytother. Res. 20, 239–249.
profiling, it is arguably an important starting point for further in- Ibusuki, K., Sakiyama, T., Kanmura, S., Maeda, T., Iwashita, Y., Nasu, Y., Sasaki, F.,
Taguchi, H., Hashimoto, S., Numata, M., Uto, H., Tsubouchi, H., Ido, A., 2015.
vestigations. Human neutrophil peptides induce interleukin-8 in intestinal epithelial cells through
the P2 receptor and ERK1/2 signaling pathways. Int. J. Mol. Med. 35, 1603–1609.
Conflict of interest Jiao, J.L., Jiang, H., Fisher, P.B., 1998. Melanoma differentiation associated gene-9, mda
-9, is a human gamma interferon responsive gene. Gene 207, 105–110.
Kanehisa, M., Goto, S., Hattori, M., Aoki-Kinoshita, K.F., Itoh, M., Kawashima, S.,
The authors declare that there are no conflicts of interest associated Katayama, T., Araki, M., Hirakawa, M., 2006. From genomics to chemical genomics:
with this publication and there has been no significant financial support new developments in KEGG. Nucleic Acids Res. 34 (Database issue), D354–D357.
https://doi.org/10.1093/nar/gkj102. Jan 1.
for this work that could have influenced its outcome. Kaur, K., 2016. Functional nutraceuticals: past, present, and future. In: Grumezescu, A.M.
(Ed.), Nanotechnology in the Agri-Food Industry. Nutraceuticals, Academic Press, pp.
Acknowledgments 41–78.
Kolehmainen, M., Mykkänen, O., Kirjavainen, P.V., Leppänen, T., Moilanen, E., Adriaens,
M., Laaksonen, D.E., Hallikainen, M., Puupponen-Pimiä, R., Pulkkinen, L., Mykkänen,
We thank the Indena S.p.A. (Milan, Italy) for providing softgel H., Gylling, H., Poutanen, K., Törrönen, R., 2012. Bilberries reduce low-grade in-
capsules containing combined extracts of Echinacea angustifolia DC. and flammation in individuals with features of metabolic syndrome. Mol. Nutr. Food Res.
56, 1501–1510. https://doi.org/10.1002/mnfr.201200195.
Zingiber officinale Roscoe.
Kussmann, M., Rezzi, S., Daniel, H., 2008. Profiling techniques in nutrition and health
research. Curr. Opin. Biotechnol. 19, 83–99.
Supplementary materials Länger, R., Kubelka, W., 2001. Phytocodex,- Pflanziche Arneispexialitaten in Osterreich
2001/2002, ed. 2. Verlagshaus der Ärzte, Wien.
Leung, D.Y., Bloom, J.W., 2003. Update on glucocorticoid action and resistance. J.
Supplementary material associated with this article can be found, in Allergy Clin. Immunol. 111, 3–22.
the online version, at doi:10.1016/j.phymed.2019.153090. Liberzon, A., Subramanian, A., Pinchback, R., Thorvaldsdóttir, H., Tamayo, P., Mesirov,
J.P., 2011. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27,
1739–1740. https://doi.org/10.1093/bioinformatics/btr260.
References Lin, S.M., Du, P., Huber, W., Kibbe, W.A., 2008. Model-based variance-stabilizing trans-
formation for Illumina microarray data. Nucleic Acids Res. 36, e11. https://doi.org/
Altamirano-Dimas, M., Hudson, J.B., Cochrane, D., Nelson, C., Arnason, J.T., 2007. 10.1093/nar/gkm1075.
Modulation of immune response gene expression by echinacea extracts: results of a Maneerat, Y., Prasongsukarn, K., Benjathummarak, S., Dechkhajorn, W., 2017. PPBP and
gene array analysis. Can. J. Physiol. Pharmacol. 85, 1091–1098. DEFA1/DEFA3 genes in hyperlipidaemia as feasible synergistic inflammatory bio-
Ashburner, M., Ball, C.A., Blake, J.A., Botstein, D., Butler, H., Cherry, J.M., Davis, A.P., markers for coronary heart disease. Lipids Health Dis. 16, 80. https://doi.org/10.
Dolinski, K., Dwight, S.S., Eppig, J.T., Harris, M.A., Hill, D.P., Issel-Tarver, L., 1186/s12944-017-0471-0.
Kasarskis, A., Lewis, S., Matese, J.C., Richardson, J.E., Ringwald, M., Rubin, G.M., Marchionni, L., Hayashi, M., Guida, E., Ooki, A., Munari, E., Jabboure, F.J., Dinalankara,
Sherlock, G., 2000. Gene ontology: tool for the unification of biology. Gene Ontol. W., Raza, A., Netto, G.J., Hoque, M.O., Argani, P., 2017. MicroRNA expression pro-
Consort. Nat. Genet. 25, 25–29. filing of Xp11 renal cell carcinoma. Hum. Pathol. 67, 18–29. https://doi.org/10.
Benjamini, Y., Hochberg, Y., 1995. Controlling the false discovery rate: a practical and 1016/j.humpath.2017.03.011.
powerful approach to multiple testing. J. R. Stat. Soc. Ser. B Stat. Methodol. 57, Mazumder, A., Cerella, C., Diederich, M., 2018. Natural scaffolds in anticancer therapy
289–300. and precision medicine. Biotechnol. Adv. 36, 1563–1585.
Bhattarai, S., Tran, V.H., Duke, C.C., 2007. Stability of [6]-gingerol and [6]-shogaol in Olnes, M.J., Kotliarov, Y., Biancotto, A., Cheung, F., Chen, J., Shi, R., Zhou, H., Wang, E.,
simulated gastric and intestinal fluids. J. Pharm. Biomed. Anal. 45, 648–653. Tsang, J.S., Nussenblatt, R., Consortium, C.H.I., 2016. Effects of systemically ad-
Boisson, B., Laplantine, E., Prando, C., Giliani, S., Israelsson, E., Xu, Z., Abhyankar, A., ministered hydrocortisone on the human immunome. Sci. Rep. 6, 23002. https://doi.
Israël, L., Trevejo-Nunez, G., Bogunovic, D., Cepika, A.M., MacDuff, D., Chrabieh, M., org/10.1038/srep23002.
Hubeau, M., Bajolle, F., Debré, M., Mazzolari, E., Vairo, D., Agou, F., Virgin, H.W., Ovesná, J., Slabý, O., Toussaint, O., Kodícek, M., Marsík, P., Pouchová, V., Vanĕk, T.,
Bossuyt, X., Rambaud, C., Facchetti, F., Bonnet, D., Quartier, P., Fournet, J.C., 2008. High throughput 'omics' approaches to assess the effects of phytochemicals in
Pascual, V., Chaussabel, D., Notarangelo, L.D., Puel, A., Israël, A., Casanova, J.L., human health studies. Br. J. Nutr. 99 (Suppl 1). https://doi.org/10.1017/
Picard, C., 2012. Immunodeficiency, autoinflammation and amylopectinosis in hu- S0007114508965818. EES127-34.
mans with inherited HOIL-1 and LUBAC deficiency. Nat. Immunol. 13, 1178–1186. Raduner, S., Majewska, A., Chen, J.-.Z., Xie, X.-.Q., Hamon, J., Faller, B., Altmann, K.H.,
Cao, F., Zhang, Q., Chen, W., Han, C., He, Y., Ran, Q., Yao, S., 2017. IL-6 increases SDCBP Gertsch, J., 2006. Alkamides from Echinacea are a new class of cannabinomimetics,
expression, cell proliferation, and cell invasion by activating JAK2/STAT3 in human cannabinoid type 2 receptor-dependent and–independent immunomodulatory ef-
glioma cells. Am. J. Transl. Res. 9, 4617–4626 eCollection. fects. J. Biol. Chem. 281, 14192–14206.
Croft, D., Mundo, A.F., Haw, R., Milacic, M., Weiser, J., Wu, G., et al., 2014. The reactome Randolph, R.K., Gellenbeck, K., Stonebrook, K., Brovelli, E., Qian, Y., Bankaitis-Davis, D.,
pathway knowledgebase. Nucleic Acids Res. 42 (Database issue), D472–D477. Cheronis, J., 2003. Regulation of human immune gene expression as influenced by a
https://doi.org/10.1093/nar/gkt1102. JanEpub 2013 Nov 15. commercial blended Echinacea product: preliminary studies. Exp. Biol. Med.
Cunliffe, R.N., 2003. Alpha-defensins in the gastrointestinal tract. Mol. Immunol. 40, (Maywood) 228, 1051–1056.
463–467. Rondanelli, M., Riva, A., Morazzoni, P., Allegrini, P., Faliva, M.A., Naso, M., Miccono, A.,
Da Silva, J.A., 1999. Sex hormones and glucocorticoids: interactions with the immune Peroni, G., Degli Agosti, I., Perna, S., 2017. The effect and safety of highly standar-
system. Ann. NY Acad. Sci. 876, 102–117. dized Ginger (Zingiber officinale) and Echinacea (Echinacea angustifolia) extract
Dall'Acqua, S., Perissutti, B., Grabnar, I., Farra, R., Comar, M., Agostinis, C., Caristi, G., supplementation on inflammation and chronic pain in NSAIDs poor responders. A
Golob, S., Voinovich, D., 2015. Pharmacokinetics and immunomodulatory effect of pilot study in subjects with knee arthrosis. Nat. Prod. Res. 31, 1309–1313. https://
lipophilic Echinacea extract formulated in softgel capsules. Eur. J. Pharm. Biopharm. doi.org/10.1080/14786419.2016.1236097.
97, 8–14. Ross, A.E., Marchionni, L., Vuica-Ross, M., Cheadle, C., Fan, J., Berman, D.M., Schaeffer,
Das, S.K., Guo, C., Pradhan, A.K., Bhoopathi, P., Talukdar, S., Shen, X.N., Emdad, L., E.M., 2011. Gene expression pathways of high grade localized prostate cancer.
Subler, M.A., Windle, J.J., Sarkar, D., Wang, X.Y., Fisher, P.B., 2016. Knockout of Prostate 71, 1568–1577. https://doi.org/10.1002/pros.21373.
MDA-9/Syntenin (SDCBP) expression in the microenvironment dampens tumor- Samoilova, Z., Smirnova, G., Muzyka, N., Oktyabrsky, O., 2014. Medicinal plant extracts
supporting inflammation and inhibits melanoma metastasis. Oncotarget 7, variously modulate susceptibility of Escherichia coli to different antibiotics.
46848–46861. Microbiol. Res. 169, 307–313. https://doi.org/10.1016/j.micres.2013.06.013.

10
S. Dall'Acqua, et al. Phytomedicine 65 (2019) 153090

Segerer, S.E., Muller, N., van den Brandt, J., Kapp, M., Dietl, J., Reichardt, H.M., Rieger, of phytopharmaceuticals. Phytomedicine 16, 97–110. https://doi.org/10.1016/j.
L., Kämmerer, U., 2009. Impact of female sex hormones on the maturation and phymed.2008.12.018.
function of human dendritic cells. Am. J. Reprod. Immunol. 62, 165–173. https://doi. Wang, C.Y., Chiao, M.T., Yen, P.J., Huang, W.C., Hou, C.C., Chien, S.C., Yeh, K.C., Yang,
org/10.1111/j.1600-0897.2009.00726.x. W.C., Shyur, L.F., Yang, N.S., 2006. Modulatory effects of Echinacea purpurea ex-
Smyth, G.K., 2004. Linear models and empirical bayes methods for assessing differential tracts on human dendritic cells: a cell- and gene-based study. Genomics 88, 801–808.
expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3:Article 3. Woelkart, K., Koidl, C., Grisold, A., Gangemi, D., Turner, R.B., Marth, E., Bauer, R., 2005.
https://doi.org/10.2202/1544-6115.1027. Bioavailability and pharmacokinetics of alkamides from the roots of Echinacea an-
Stier, S., Totzke, G., Grunewald, E., Neuhaus, T., Fronhoffs, S., Sachinidis, A., Vetter, H., gustifolia in humans. J. Clin. Pharmacol. 45, 683–689.
Schulze-Osthoff, K., Ko, Y., 2000. Identification of syntenin and other TNF-inducible Xie, X., Lu, J., Kulbokas, E.J., Golub, T.R., Mootha, V., Lindblad-Toh, K., Lander, E.S.,
genes in human umbilical arterial endothelial cells by suppression subtractive hy- Kellis, M., 2005. Systematic discovery of regulatory motifs in human promoters and
bridization. FEBS Lett. 467, 299–304. 3′ UTRs by comparison of several mammals. Nature 434, 338–345.
Trill, J., Simpson, C., Webley, F., Radford, M., Stanton, L., Maishman, T., Galanopoulou, Yan, J., Ma, Y., Zhao, F., Gu, W., Jiao, Y., 2013. Identification of immunomodulatory
A., Flower, A., Eyles, C., Willcox, M., Hay, A., Griffiths, G., Little, P., Lewith, G., signatures induced by american ginseng in murine immune cells. Evid. Based
Moore, M., 2017. Uva-ursi extract and ibuprofen as alternative treatments of adult Complement. Alternat. Med. 2013, 972814. https://doi.org/10.1155/2013/972814.
female urinary tract infection (ATAFUTI): study protocol for a randomised controlled Yu, Y., Zick, S., Li, X., Zou, P., Wright, B., Sun, D., 2011. Examination of the pharma-
trial. Trials 18, 421. https://doi.org/10.1186/s13063-017-2145-7. cokinetics of active ingredients of ginger in humans. AAPS J. 13, 417–426.
van Breda, S.G., Wilms, L.C., Gaj, S., Jennen, D.G., Briedé, J.J., Helsper, J.P., Kleinjans, Zick, S.M., Djuric, Z., Ruffin, M.T., Litzinger, A.J., Normolle, D.P., Alrawi, S., Feng, M.R.,
J.C., de Kok, T.M., 2014. Can transcriptomics provide insight into the chemopre- Brenner, D.E., 2008. Pharmacokinetics of 6-gingerol, 8-gingerol, 10-gingerol, and 6
ventive mechanisms of complex mixtures of phytochemicals in humans? Antioxid. shogaol and conjugate metabolites in healthy human subjects. Cancer Epidemiol.
Redox Signal 20, 2107–2113. Biomarker Prev. 17, 1930–1936.
Wagner, H., Ulrich-Merzenich, G., 2009. Synergy research: approaching a new generation

11

You might also like