Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Screening of herbal extracts for TLR2- and TLR4-dependent anti-inflammatory effects

  • Anne Schink,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Validation, Visualization, Writing – original draft, Writing – review & editing

    Affiliation Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany

  • Jan Neumann,

    Roles Formal analysis, Investigation, Methodology, Validation, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany, Institute of Molecular Biology, Mainz, Germany

  • Anna Lena Leifke,

    Roles Investigation, Writing – review & editing

    Affiliation Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany

  • Kira Ziegler,

    Roles Writing – review & editing

    Affiliation Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany

  • Janine Fröhlich-Nowoisky,

    Roles Writing – review & editing

    Affiliation Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany

  • Christoph Cremer,

    Roles Writing – review & editing

    Affiliations Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany, Institute of Molecular Biology, Mainz, Germany

  • Eckhard Thines,

    Roles Conceptualization, Writing – review & editing

    Affiliations Institut für Biotechnologie und Wirkstoff-Forschung gGmbH, Kaiserslautern, Germany, Institute of Molecular Physiology, Johannes Gutenberg University Mainz, Mainz, Germany

  • Bettina Weber,

    Roles Resources, Visualization, Writing – review & editing

    Affiliation Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany

  • Ulrich Pöschl,

    Roles Conceptualization, Methodology, Project administration, Resources, Supervision, Visualization, Writing – review & editing

    Affiliation Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany

  • Detlef Schuppan ,

    Roles Conceptualization, Methodology, Project administration, Resources, Supervision, Writing – review & editing

    ‡ These authors are joint senior authors on this work.

    Affiliations Institute of Translational Immunology, University of Mainz Medical Center, Mainz, Germany, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America

  • Kurt Lucas

    Roles Conceptualization, Methodology, Project administration, Resources, Supervision, Validation, Visualization, Writing – review & editing

    k.lucas@mpic.de

    ‡ These authors are joint senior authors on this work.

    Affiliation Multiphase Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany

Abstract

Herbal extracts represent an ample source of natural compounds, with potential to be used in improving human health. There is a growing interest in using natural extracts as possible new treatment strategies for inflammatory diseases. We therefore aimed at identifying herbal extracts that affect inflammatory signaling pathways through toll-like receptors (TLRs), TLR2 and TLR4. Ninety-nine ethanolic extracts were screened in THP-1 monocytes and HeLa-TLR4 transfected reporter cells for their effects on stimulated TLR2 and TLR4 signaling pathways. The 28 identified anti-inflammatory extracts were tested in comparative assays of stimulated HEK-TLR2 and HEK-TLR4 transfected reporter cells to differentiate between direct TLR4 antagonistic effects and interference with downstream signaling cascades. Furthermore, the ten most effective anti-inflammatory extracts were tested on their ability to inhibit nuclear factor-κB (NF-κB) translocation in HeLa-TLR4 transfected reporter cell lines and for their ability to repolarize M1-type macrophages. Ethanolic extracts which showed the highest anti-inflammatory potential, up to a complete inhibition of pro-inflammatory cytokine production were Castanea sativa leaves, Cinchona pubescens bark, Cinnamomum verum bark, Salix alba bark, Rheum palmatum root, Alchemilla vulgaris plant, Humulus lupulus cones, Vaccinium myrtillus berries, Curcuma longa root and Arctostaphylos uva-ursi leaves. Moreover, all tested extracts mitigated not only TLR4, but also TLR2 signaling pathways. Seven of them additionally inhibited translocation of NF-κB into the nucleus. Two of the extracts showed impact on repolarization of pro-inflammatory M1-type to anti-inflammatory M2-type macrophages. Several promising anti-inflammatory herbal extracts were identified in this study, including extracts with previously unknown influence on key TLR signaling pathways and macrophage repolarization, serving as a basis for novel lead compound identification.

Introduction

Herbs, algae, fungi and cyanobacteria have been used in traditional medicine for centuries. During the last decades, plant extracts and natural compounds became a focal point of interest again as novel lead compounds for the treatment of inflammatory diseases are needed [1]. Several diseases progression and development are influenced by acute and chronic inflammation, such as: autoimmune diseases, allergies, obesity, diabetes, organ fibrosis and dysfunction. Plant extracts that contain largely orally available compounds which attenuate inflammatory processes may be highly attractive as potential therapies [28]. Regardless of the origin, inflammation is often associated with a self-enhancing, cyclic process, involving stimulation of innate immunity, prominently of TLRs, production of reactive oxygen and nitrogen species (ROS/RNS), pro-inflammatory cytokine/chemokine secretion, as well as the release of host-derived damage associated molecular patterns (DAMPs) [9,10]. In healthy individuals the initial immune response to an acute stimulus, e.g. a microbial infection, is mitigated over time by downregulation of TLR stimulation, leading to a diminished cytokine production and interruption of the vicious inflammatory circle. In diseases associated with chronic inflammation, the appropriate regulation of TLRs and their downstream signaling pathways is often absent [1, 11]. Antagonists for TLR signaling play an important role in counter-regulating such overwhelming reactions, especially for TLR4 which is a central danger-sensing innate immune receptor. Different from all other TLRs, stimulation of TLR4, leads to activation of two major pathways: 1) the myeloid differentiation 88-dependent (MyD88) or canonical pathway of NF-κB activation, and 2) the MyD88-independent or Toll/interleukin-1 receptor (TIR)-domain-containing adaptor molecule (TRAM) pathway. The canonical pathway can also be activated via TLR2 stimulation [12,13]. Some synthetic small molecules (e.g. Eritoran and TAK-242), but also natural compounds (e.g. epigallocatechin-3-gallate and 6-shogaol) inhibit TLR4 signaling [1418]. Nevertheless, to date, no effective orally active TLR4 antagonist is available for experimental or clinical application.

Due to their easy oral application and minor adverse effects, herbal extracts compromising of TLR4 antagonistic activity would be highly interesting as new oral treatment strategies for inflammatory diseases. Nevertheless, identification of the active compounds and their targets are often complex. Furthermore, also metabolization products and not only the applied compounds themselves might interact with the TLR signaling pathways. This further complicates the identification of the responsible mechanism(s). Recently, numerous studies have focused on Chinese herbal medicines and their impact on several diseases [1922], however, their anti-inflammatory effects remain largely unknown. Thus, in the current study we analyzed ethanolic extracts of medicinal plants, which may have anti-inflammatory properties (see S1 Table in the supplementary data).

Materials and methods

Ethanolic extracts

Most of the ethanolic extracts were purchased directly from Maros Arznei GmbH. All other samples listed in Table 1 were freshly prepared by grinding 10 g plants or algae in a mortar, if necessary under liquid nitrogen. Afterwards, the powder was resuspended in 50 ml of 70% ethanol (VWR International GmbH, Darmstadt, Germany). Subsequently, the samples were incubated for ten days in the dark at room temperature (RT), being inverted once a day. Then, the extracts were filtered through Rotilabo-folded filters (type 113P; Carl Roth, Karlsruhe, Germany) to remove unresolved residues.

Cell cultures and treatments

HeLa-TLR4 cell line.

The HeLa-TLR4 transfected reporter cell line (Novusbio, Wiesbaden Nordenstadt, Germany) was cultured in Dulbecco's Modified Eagle Medium (DMEM, Thermo Fisher Scientific, Darmstadt, Germany) supplemented with 10% fetal bovine serum (FBS, Biochrom, Berlin, Germany), 1% penicillin-streptomycin (Thermo Fisher Scientific), 5 μg/ml blasticidine (Sigma-Aldrich, Darmstadt, Germany), 1 μg/ml puromycin (Sigma-Aldrich), and 1 mg/ml geneticin (G418, Sigma-Aldrich) at 37°C in humidified atmospheric air supplemented with 5% CO2. In black 96-well microplates with clear bottom (Greiner Bio-One, Solingen, Germany), 2×105 cells/ml were seeded in 100 μl medium and allowed to settle overnight. Experiments were conducted with different concentrations of the extracts or the same amount of vehicle (70% ethanol). Concentrations ranging from 0.01% to 3% in cell culture medium were used to receive a dose-response curve and to cover a broad spectrum of concentrations. Due to toxic effects, higher concentrations than 3% were not used in the performed assay systems. Cells were pre-incubated with extracts or vehicle (70% ethanol) for 2 h at 37°C. Afterwards, lipopolysaccharide (LPS-EB, from E. coli O111:B4, Invivogen, Toulouse, France) in a final concentration of 25 ng/ml was added to stimulate TLR4 (incubation for 8 h at 37°C). Alamar Blue assay was used to determine cell viability and TLR4 stimulation was measured using luciferase assay (Novusbio).

NF-κB translocation experiments were conducted on a monoclonal HeLa-TLR4 cell line (Novusbio) stably transfected with a plasmid expressing constitutive active firefly luciferase, as a reporter for cell viability (HeLa-TLR4 dual reporter cell line). Cells were cultured as described for HeLa-TLR4 reporter cell line, but selective antibiotics were replaced by 140 μg/ml Hygromycin B Gold (Invivogen). HeLa-TLR4 dual reporter cells were seeded on 12-well glass bottom plates (Cellvis, Mountain View, USA) at a concentration of 2.0×105 cells/ml in 1 ml complete medium per well and incubated for 48 h at 37°C. Cells were pre-incubated with 0.6% extracts in final cell culture medium or with the same amount of vehicle control for 2 h at 37°C. After pre-incubation, LPS-EB in a final concentration of 50 ng/ml was added for 1 h at 37°C. Next, cells were washed 1x with phosphate buffered saline (PBS) and fixed with 4% formaldehyde in PBS buffer (Thermo Fisher Scientific) for 10 min at 37°C. Subsequently, cells were washed 3x with PBS before proceeding with immunostaining for NF-κB.

THP-1 cell line.

The myeloid THP-1 cell line TIB-202 (ATCC; LGC Standards, Wesel, Germany) was cultured in Roswell Park Memorial Institute (RPMI) 1640 medium (Thermo Fisher Scientific) supplemented with 10% FBS (heat-inactivated), 1% penicillin/streptomycin and 0.05 mM β-mercaptoethanol (Sigma-Aldrich) at 37°C in humidified atmospheric air supplemented with 5% CO2. In a 96-well microplate (Greiner Bio-One), 4×105 cells/ml were seeded in 80 μl complete medium and were allowed to settle for 1 h. Experiments were conducted at the same extract concentration range used for HeLa-TLR4 cells. Cells were incubated with extracts or vehicle for 2 h at 37°C. Afterwards, LPS-EB in a final concentration of 50 ng/ml was added to stimulate TLR4 (4 h incubation at 37°C). Alamar Blue assay was used to determine cell viability. TLR4 stimulation was measured using enzyme-linked immunosorbent assay (ELISA, BD Biosciences, Heidelberg, Germany).

Polarization experiments were conducted with THP-1 monocytes differentiated to macrophages. In a 96-well microplate (Greiner Bio-One), 1.5x105 cells/ml were seeded in 200 μl complete growth medium containing 10 ng/ml phorbol 12-myristate 13-acetate (PMA, Sigma-Aldrich). Cells were incubated for three days at 37°C to differentiate to M0 macrophages. Afterwards, the medium was exchanged to 200 μl complete growth medium without PMA and cells were incubated overnight at 37°C. Cells were polarized to M1-type macrophages with 20 ng/ml interferon-gamma (IFNγ, Thermo Fisher Scientific) for 24 h. All further steps were performed as described for THP-1 monocytes.

HEK-TLR2 and HEK-TLR4 cell lines.

The HEK-Blue hTLR2 (HEK-TLR2) and HEK-Blue hTLR4 cell lines (HEK-TLR4) (both Invivogen) were cultured in DMEM high glucose medium supplemented with 10% FBS (heat-inactivated), 1% penicillin/streptomycin and 1x HEK-Blue Selection (Invivogen) at 37°C in humidified atmospheric air supplemented with 5% CO2. In experiments examining cell viability, medium without HEK-Blue Selection was used. In experiments analyzing TLR2 and TLR4 stimulation, cells were cultured in HEK-Blue detection medium (Invivogen). Extracts with concentrations between 0.01% and 3% in final cell culture medium or vehicle (70% ethanol) were added in empty 96-well microplates. Directly afterwards, cells were seeded with 2.8×105 cells/ml in 100 μl complete growth medium and were incubated with the extracts for 2 h at 37°C. Subsequently, receptor activity was stimulated by adding 1 ng/ml S-(2,3-bis(palmitoyloxy)-(2RS)-propyl)-(R)-cysteinyl-(S)-seryl-(S)-lysyl-(S)-lysyl-(S)-lysyl-(S)-lysine (Pam2CSK4, HEK-TLR2 cell line, Invivogen) or 100 ng/ml LPS-EB Ultrapure (HEK-TLR4 cell line, LPS from E. coli O111:B4, Invivogen). Cells were incubated overnight at 37°C. Alamar Blue assay was used to determine cell viability and TLR2/TLR4 stimulation was measured using HEK-Blue detection assay (Invivogen).

Determination of cell viability

Cell viability was measured by the Alamar Blue assay (Thermo Fisher Scientific) according to the manufacturer’s protocol (10% final concentration of Alamar Blue solution in cell culture medium). Cells were incubated with Alamar Blue solution at 37°C for 4 h (HeLa-TLR4 cell line) or overnight (all other cell lines). Fluorescence intensity was measured with microplate reader Synergy Neo (Biotek, Bad Friedrichshall, Germany) at an excitation wavelength of 560 nm and an emission wavelength of 590 nm.

IL-8 transcriptional activity in HeLa-TLR4 cells

LightSwitch Luciferase assay (Novusbio) was used to determine production of renilla luciferase under transcriptional control of interleukin-8 (IL-8) promoter. HeLa-TLR4 cells were washed with PBS (Thermo Fisher Scientific) after performing the cell viability assay to remove the Alamar Blue staining and were frozen overnight at -80°C for better cell lysis. After thawing the cells, LightSwitch Luciferase assay was performed according to manufacturer’s protocol and luminescence was measured with microplate reader Synergy Neo (Biotek).

IL-8, IL-10 and TNF-α secretion

Supernatants of pre-treated THP-1 monocytes and macrophages were examined for concentration of IL-8, IL-10 or tumor necrosis factor alpha (TNF-α) using ELISAs (BD Biosciences). Sandwich ELISA was carried out according to manufacturer’s protocol with optimized washing buffer volume. Supernatant was diluted up to 6-fold (total used volume: 100 μl/well). Absorbance was measured using a Synergy Neo plate reader at a wavelength of 450 nm and a reference wavelength of 570 nm. Based on pipetted standard values, a four-parameter logarithmic standard curve was determined with the Gen5 software on SynergyNeo (Biotek). Cytokine production of THP-1 monocytes and macrophages were calculated according to standard curves and dilution factors.

Determination of transcription activity in HEK-Blue cells

HEK-Blue Detection assay (Invivogen) was used to determine production of inducible secreted embryonic alkaline phosphatase (SEAP), based on activation of NF-κB and activator protein 1 (AP-1). HEK-Blue cells were cultured in HEK-Blue Detection medium during incubation with extracts and stimulation of receptor activity. Afterwards, stimulation of TLR2 (HEK-TLR2 cell line) and TLR4 (HEK-TLR4 cell line) were determined using a Synergy Neo plate reader (Biotek) at a wavelength of 640 nm.

Determination of NF-κB p65 translocation by fluorescence microscopy

For immunostaining of NF-κB, HeLa-TLR4 dual reporter cells were incubated in blocking solution consisting of 5% bovine serum albumin (BSA, Cell Signaling Technology, Cambridge, UK) and 0.3% Triton X-100 (Merck, Darmstadt, Germany) in PBS for 1 h at RT. Next, cells were incubated with primary anti-NF-κB p65 antibody (D14E12, Cell Signaling Technology) overnight at 4°C. After washing, secondary anti-rabbit Alexa Fluor 568 antibody was applied (Thermo Fisher Scientific) for 1 h at RT. Both antibodies were diluted 1:400 in PBS containing 1% BSA and 0.3% Triton X-100. Cell nuclei were counterstained using 4’,6-diamidino-2-phenylindole (DAPI, Thermo Fisher Scientific).

Stained cells were imaged using the Opera Phenix High-Content Screening System (PerkinElmer, Waltham, USA) with laser lines for Alexa Fluor 568 at 568 nm excitation and 570 nm—630 nm emission and laser lines for DAPI at 405 nm excitation and 435 nm—480 nm emission. Image processing was done using Harmony software (PerkinElmer). Nuclear masks (DAPI stained cell nuclei) and surrounding ring-like masks (cytoplasm) were identified and the mean fluorescence intensity ratio of nuclear to cytoplasmic NF-κB was determined.

Statistical analyses

Statistical analyses were accomplished in GraphPad Prism 5.01 (GraphPad Software, San Diego, California, USA, www.graphpad.com) conducting one-way ANOVA followed by Dunnett’s post hoc test. Values of p<0.05 were considered as significant. For statistical analyses of data from macrophage polarization, ANOVA was accomplished, followed by an unpaired t-test. Values of p<0.05 were considered significant.

Results

Screening of ethanolic herbal extracts for TLR-dependent anti-inflammatory effects

During initial screening, 99 ethanolic extracts (96 herbal extracts, one cyanobacterial, one green algae and one lichen extract) were tested for their anti-inflammatory activity. Ten extracts displayed major anti-inflammatory activity combined with a high cell viability (Fig 1, Fig 2 and Fig 3). The results for the other 89 extracts are graphically displayed in the supplementary data (S1 Fig). Comparing the effects in both cell lines used for efficacy and toxicity readout, HeLa-TLR4 transfected reporter cells were more susceptible to toxic effects of most extracts. THP-1 monocytes showed a viability above 85% after treatment with the ten most effective anti-inflammatory extracts in concentrations up to 1%, except for Humulus lupulus and Arctostaphylos uva-ursi (Fig 1, Fig 2 and Fig 3). Some extracts such as Castanea sativa, Cinnamomum verum, Humulus lupulus and Curcuma longa revealed comparable anti-inflammatory efficacy in both cell lines, whereas other extracts (e.g. Cinchona pubescens and Rheum palmatum) were more effective in THP-1 monocytes than in HeLa-TLR4 cells. Castanea sativa was the only extract with a dose-dependent anti-inflammatory activity already at low concentrations, whereas most of the other extracts possessed an inhibition threshold (e.g. between 0.3% and 0.6% for cinnamon extract). Ethanolic extracts with the highest anti-inflammatory potential were sweet chestnut (Castanea sativa), cinchona (Cinchona pubescens), cinnamon (Cinnamomum verum), white willow (Salix alba), rhubarb (Rheum palmatum), common lady’s mantle (Alchemilla vulgaris), hops (Humulus lupulus), bilberries (Vaccinium myrtillus), turmeric (Curcuma longa) and bearberry (Arctostaphylos uva-ursi) (Fig 1, Fig 2 and Fig 3). Three bark extracts were within the top five anti-inflammatory extracts (number 2: Cinchona pubescens, number 3: Cinnamomum verum and number 4: Salix alba). Fig 4 shows the 25 most effective anti-inflammatory extracts and S2 Fig displays all 99 extracts with a ranking according to their anti-inflammatory potential. For visualization, heat maps display TLR4 stimulation normalized to viability shown in green for anti-inflammatory and in red for pro-inflammatory effects. THP-1 monocytes naturally express TLR4 and several other innate immune receptors, in contrast to HeLa-TLR4 cells, which were transfected with TLR4. THP-1 monocytes more reflect the physiological mechanisms in the human body, therefore effects on THP-1 monocytes vs. HeLa-TLR4 cells were weighted in a ratio of 2:1 for ranking of the extracts. In addition, TLR4 inhibition was calculated with a higher impact than toxic effects. Extracts resulting in anti-inflammatory activity but with a viability below 75% should be viewed critically, since the anti-inflammatory effects have to be rather attributed to toxicity. In the heat maps these values are displayed in grey.

thumbnail
Fig 1. Cell viability and concentration-dependent anti-inflammatory effects of selected herbal extracts (part 1).

HeLa-TLR4 cells (red) and THP-1 monocytes (blue) were incubated with extracts in different concentrations or vehicle (70% ethanol), followed by stimulation with LPS-EB. Viability was measured using the Alamar Blue Assay and was normalized to the negative control (untreated cells). TLR4 receptor stimulation was measured using Renilla luciferase expression for the HeLa-TLR4 cell line and IL-8 ELISA (pg/ml) for the THP-1 monocytes and was normalized to ethanol-treated cells. Data are displayed as viability (%) in the left graphs and TLR4 stimulation divided by normalized viability (%) in the right graphs. Data represents means (n≥2). For graphical display of further extracts, see Fig 2, Fig 3 and supplementary data S1 Fig.

https://doi.org/10.1371/journal.pone.0203907.g001

thumbnail
Fig 2. Cell viability and concentration-dependent anti-inflammatory effects of selected herbal extracts (part 2).

HeLa-TLR4 cells (red) and THP-1 monocytes (blue) were incubated with extracts in different concentrations or vehicle (70% ethanol), followed by stimulation with LPS-EB. Viability was measured using the Alamar Blue Assay and was normalized to the negative control (untreated cells). TLR4 receptor stimulation was measured using Renilla luciferase expression for the HeLa-TLR4 cell line and IL-8 ELISA (pg/ml) for the THP-1 monocytes and was normalized to ethanol-treated cells. Data are displayed as viability (%) in the left graphs and TLR4 stimulation divided by normalized viability (%) in the right graphs. Data represents means (n≥2). For graphical display of further extracts, see Fig 1, Fig 3 and supplementary data S1 Fig.

https://doi.org/10.1371/journal.pone.0203907.g002

thumbnail
Fig 3. Cell viability and concentration-dependent anti-inflammatory effects of selected herbal extracts (part 3).

HeLa-TLR4 cells (red) and THP-1 monocytes (blue) were incubated with extracts in different concentrations or vehicle (70% ethanol), followed by stimulation with LPS-EB. Viability was measured using the Alamar Blue Assay and was normalized to the negative control (untreated cells). TLR4 receptor stimulation was measured using Renilla luciferase expression for the HeLa-TLR4 cell line and IL-8 ELISA (pg/ml) for the THP-1 monocytes and was normalized to ethanol-treated cells. Data are displayed as viability (%) in the left graphs and TLR4 stimulation divided by normalized viability (%) in the right graphs. Data represents means (n≥2). For graphical display of further extracts, see Fig 1, Fig 2 and supplementary data S1 Fig.

https://doi.org/10.1371/journal.pone.0203907.g003

thumbnail
Fig 4. Ethanolic extracts with highest anti-inflammatory activity.

HeLa-TLR4 cells or THP-1 monocytes were incubated with extracts in different concentrations or vehicle (70% ethanol), followed by stimulation with LPS-EB. Viability was measured using the Alamar Blue Assay and was normalized to the negative control (untreated cells) (Viability (%)). TLR4 receptor activity was measured using Renilla luciferase expression for the HeLa-TLR4 cell line or IL-8 ELISA for the THP-1 monocytes and was normalized to ethanol-treated cells (TLR4-Activity). Data are displayed as TLR4 stimulation divided by viability and ranked ascending by the following formula: (150—Viability (%)) * (2 * TLR4-Activity + 100) weighted in a ratio of 2:1 for THP-1 monocytes vs. HeLa-TLR4 cells. The 25 extracts with the highest mitigation of LPS-induced inflammatory signal are displayed here (for comparison of all extracts see S2 Fig). Data represents means (n≥2).

https://doi.org/10.1371/journal.pone.0203907.g004

Screening for exclusive TLR4 antagonistic effects

Besides the identification of anti-inflammatory extracts, we were also interested in specific antagonists for TLR4. Twenty-eight ethanolic extracts with strong anti-inflammatory effects were additionally tested in a comparative assay with Pam2CSK4-stimulated HEK-TLR2 and LPS-EB Ultrapure-stimulated HEK-TLR4 cells to discriminate a direct TLR4 antagonistic effect from interference with downstream signaling pathways shared by TLR2 and TLR4 signaling. All tested extracts exhibited mitigation of TLR2- and TLR4-dependent responses (see Figs 5 and 6 for the five most effective anti-inflammatory extracts and S3 Fig for all other extracts). For most of the extracts, especially the most effective anti-inflammatory extracts, HEK-TLR2 and HEK-TLR4 cell lines showed comparable dose-dependent anti-inflammatory effects. Interestingly, Castanea sativa leaves inhibited TLR2- and TLR4-dependent inflammatory responses already at 0.01% extract dilution, accompanied by a cell viability of 98%. An increase of inflammatory activity was observed for most of the extracts with concentrations between 1% and 3% in cell culture medium. This may be due to toxic effects and should therefore be viewed critically. Heat maps displaying the TLR2- and TLR4 stimulation are shown in Fig 7 (the five most promising extracts) and S4 Fig (all 28 extracts), with anti-inflammatory activity shown in green to pro-inflammatory activity shown in red. The ranking of the extracts was taken from the THP-1 and HeLa-TLR4 screening shown in Fig 4 and S2 Fig. Values with viability below 75% are marked in grey.

thumbnail
Fig 5. Extracts with TLR2 and TLR4 antagonistic effects (part 1).

HEK-TLR2 cells (purple) and HEK-TLR4 cells (orange) were incubated with extracts in different concentrations or vehicle (70% ethanol), followed by stimulation of HEK-TLR2 cells with Pam2CSK4 or HEK-TLR4 cells with LPS-EB Ultrapure. Viability was measured using the Alamar Blue Assay and was normalized to the negative control (untreated cells). TLR2 and TLR4 receptor stimulation were measured using SEAP production and were normalized to ethanol-treated cells. Data are displayed as viability (%) in the left graphs and TLR4 stimulation divided by viability (%) in the right graphs. Data represents means (n≥4). For graphical display of further extracts, see Fig 6 and supplementary data S3 Fig.

https://doi.org/10.1371/journal.pone.0203907.g005

thumbnail
Fig 6. Extracts with TLR2 and TLR4 antagonistic effects (part 2).

HEK-TLR2 cells (purple) and HEK-TLR4 cells (orange) were incubated with extracts in different concentrations or vehicle (70% ethanol), followed by stimulation of HEK-TLR2 cells with Pam2CSK4 or HEK-TLR4 cells with LPS-EB Ultrapure. Viability was measured using the Alamar Blue Assay and was normalized to the negative control (untreated cells). TLR2 and TLR4 receptor stimulation were measured using SEAP production and were normalized to ethanol-treated cells. Data are displayed as viability (%) in the left graphs and TLR4 stimulation divided by viability (%) in the right graphs. Data represents means (n≥4). For graphical display of further extracts, see Fig 5 and supplementary data S3 Fig.

https://doi.org/10.1371/journal.pone.0203907.g006

thumbnail
Fig 7. Ethanolic extracts with TLR2 and TLR4 antagonistic activities in HEK-TLR2 and HEK-TLR4 cell lines.

HEK-TLR2 or HEK-TLR4 cells were incubated with extracts in different concentrations or vehicle (70% ethanol), followed by stimulation of HEK-TLR2 cells with Pam2CSK4 or HEK-TLR4 cells with LPS-EB Ultrapure. Viability was measured using the Alamar Blue Assay and was normalized to the negative control (untreated cells). TLR2 and TLR4 receptor activity were measured using SEAP production and were normalized to ethanol-treated cells. Data are displayed as receptor stimulation divided by normalized viability. The five extracts with the highest mitigation of LPS-induced inflammatory signal from Fig 4 are displayed here (for comparison of further extracts, see S4 Fig). Data represents means (n≥4).

https://doi.org/10.1371/journal.pone.0203907.g007

Inhibition of NF-κB p65 translocation by select extracts

Several pro-inflammatory signaling pathways cumulate in the nuclear translocation of the transcription factor NF-κB. Therefore, the ten extracts with the highest anti-inflammatory activity, combined with no or low toxicity in THP-1 and HeLa-TLR4 cells (Fig 1, Fig 2 and Fig 3) were tested regarding their influence on NF-κB p65 translocation in LPS-stimulated HeLa-TLR4 dual reporter cells. Here Castanea sativa, Cinnamomum verum, Salix alba, Rheum palmatum, Humulus lupulus, Curcuma longa as well as Arctostaphylos uva-ursi significantly inhibited NF-κB translocation, compared to the LPS-stimulated vehicle control (Fig 8). Interestingly, Cinchona pubescens, Alchemilla vulgaris and Vaccinium myrtillus, which demonstrated strong anti-inflammatory activity in the THP-1 and HeLa-TLR4 as well as in the comparative HEK-TLR2/HEK-TLR4 assays, did not prevent the LPS-induced NF-κB translocation into the nucleus, indicating interference with another, alternative pro-inflammatory mechanism.

thumbnail
Fig 8. NF-κB translocation of select anti-inflammatory extracts.

A: Fluorescence microscopy images of NF-κB stained HeLa-TLR4 dual reporter cells incubated with extracts or vehicle (70% ethanol), followed by stimulation with LPS-EB. For better visibility, images were cropped and adjusted in brightness and contrast. Scale bar = 50 μm. B: Quantitative evaluation of NF-κB p65 translocation. Mean fluorescence ratios of nuclear to cytoplasmic NF-κB p65 were calculated and compared to ethanol control. Data represents means ± SD (n = 3, 72 images (fields) per experiment and per treatment condition). Dunnett’s post hoc test with **p<0.01; ***p<0.001 compared to ethanol control.

https://doi.org/10.1371/journal.pone.0203907.g008

Repolarization of THP-1 macrophages by select extracts

The same ten extracts were additionally tested for their influence on repolarization of pro-inflammatory M1-type to anti-inflammatory M2-type macrophages. Here, enhanced secretion of TNF-α is an indicator for M1-type polarization, whereas IL-10 secretion suggests M2-type polarization. All tested M1-polarized, LPS and extract treated THP-1 macrophages showed a viability above 85%, except for Humulus lupulus extract, which resulted in 64% viability compared to the vehicle control (Fig 9A). A significant mitigation of TNF-α secretion was observed after incubation with all extracts, except for Salix alba and Vaccinium myrtillus (Fig 9B). Furthermore, IL-10 production was significantly induced by several extracts, especially Rheum palmatum and Arctostaphylos uva-ursi (Fig 9C).

thumbnail
Fig 9. Effect of ten most effective anti-inflammatory extracts on macrophage repolarization.

THP-1 M1 macrophages were incubated with extracts or vehicle (70% ethanol) control, followed by stimulation with LPS-EB. Negative control: untreated M1-type macrophages. A: Viability (Alamar Blue assay) was normalized to viability of untreated cells. B: TNF-α secretion (pg/ml) measured by ELISA. C: IL-10 secretion (pg/ml) measured by ELISA. Data represents means ± SD of 2 independent experiments (each with n = 3); unpaired t-test with ***p<0.001, **p<0.005 compared to respective ethanol control.

https://doi.org/10.1371/journal.pone.0203907.g009

Discussion

Potent anti-inflammatory effects were observed in THP-1 monocytes and HeLa-TLR4 cells after treatment with various ethanolic extracts (Fig 1, Fig 2 and Fig 3). In addition, the most effective extracts were tested in a comparative assay of HEK-TLR2 and HEK-TLR4 cells (Figs 5 and 6). Comparable dose-dependent anti-inflammatory activities in both transfected HEK cell lines suggested that all tested extracts interfere with the NF-κB/AP-1 signaling pathways of TLR2 and TLR4 (and some other innate immune receptors). Thus, it is reasonable that no exclusive TLR4 antagonist was found among the tested extracts. Furthermore, the ten most effective extracts were tested for their ability to inhibit translocation of NF-κB p65 into the cell nucleus (Fig 8). Seven extracts mitigated NF-κB p65 translocation. Despite being anti-inflammatory in THP-1 and HeLa-TLR4 screening as well as in the comparative HEK-TLR2/HEK-TLR4 assay, three extracts (Cinchona pubescens, Alchemilla vulgaris and Vaccinium myrtillus) showed no effect on NF-κB p65 translocation. This is remarkable, since nuclear translocation of NF-κB is an essential step in inflammation. Thus, other signaling pathways, such as those triggered by TNF-α receptor (TNFαR) and interleukin-1 receptor (IL-1R) lead to the observed translocation of NF-κB, however, without IL-8 cytokine release. Those three extracts that lack inhibition of NF-κB translocation may inhibit other molecules downstream of NF-κB. Ethanolic and organic extracts were shown to mainly comprise of anti-inflammatory compounds, whereas water extracts primarily comprise of compounds stimulating the immune system [23, 24]. A mixture of anti-inflammatory and pro-inflammatory compounds within our tested 70% ethanolic extracts might therefore serve as a partial explanation for the contradictory results between the anti-inflammatory effects in HeLa-TLR4 and THP-1 screening vs. the lack of inhibition of LPS-induced NF-κB translocation observed after incubation with Cinchona pubescens, Alchemilla vulgaris and Vaccinium myrtillus. Furthermore, incubation with two out of the ten most effective extracts (Rheum palmatum and Arctostaphylos uva-ursi, Fig 9) suggest an activity to repolarize pro-inflammatory and tissue destructive M1-type towards M2-type macrophages, which are involved in tissue repair and angiogenesis [25]. Comparing both cell lines, THP-1 monocytes were more susceptible to TLR4-dependent anti-inflammatory effects than HeLa-TLR4 cells. Since HeLa-TLR4 cells were co-transfected with the functional human TLR4, MD-2 and CD-14 complex they do exhibit high TLR4 canonical pathway specificity compared to other TLR4-related pathways. Extracts with stronger anti-inflammatory effects in THP-1 monocytes could possibly influence molecular mechanisms absent in HeLa-TLR4 cells. Therefore, the results observed in THP-1 monocytes may be better suited to reflect physiological TLR4 signaling.

In the NF-κB translocation experiments, the incubation time differs from those in THP-1 and HeLa-TLR4 based screening assays (1 h vs. 4–8 h incubation with LPS), which might also explain the differing results. These LPS incubation times were chosen, as influences of the extracts on THP-1 and HeLa TLR4 screening were best seen after 4 h and 8 h, whereas NF-κB showed highest translocation into the nucleus at 1-2 h.

Due to the high number of extracts tested, we focused on the ten most effective anti-inflammatory extracts identified on screening. These included Castanea sativa leaves, Cinchona pubescens bark, Cinnamomum verum bark, Salix alba bark, Rheum palmatum root, Alchemilla vulgaris plant, Humulus lupulus cones, Vaccinium myrtillus berries, Curcuma longa root and Arctostaphylos uva-ursi leaves. To the best of our knowledge, Castanea sativa leaves and Alchemilla vulgaris plant extracts had not been reported to modulate the NF-κB, TLR2 or TLR4 signaling pathways, thus representing new promising candidates for further investigations. However, spiny burrs of sweet chestnuts inhibited NF-κB activation in vivo [26] and ellagitannins present in Alchemilla vulgaris were shown to possess NF-κB dependent anti-inflammatory effects [27].

Sweet chestnut (Castanea sativa)—leaves

Strong dose-dependent anti-inflammatory effects, represented by decreased IL-8 concentrations, were observed after sweet chestnut extract incubation. In addition, high anti-inflammatory activity in the comparative HEK-Blue assay, even at low extract concentrations, were found. Apparently, sweet chestnut leaf extracts affect the signaling pathway of both TLR2 and TLR4. Sweet chestnut extract is used in traditional medicines for treatment of skin and soft tissue infections [28]. In modern studies several medicinal properties have been reported, e.g. anti-oxidant and anti-microbial effects [2830]. Spiny burrs of sweet chestnuts applied for hepatorenal injury in diabetic rats inhibit NF-κB activation [27]. The carotenoid content in fruit and leaves has been suggested to possess direct anti-oxidant effects as well as to influence redox-sensitive and vitamin A signaling pathways [29,31]. Nevertheless, for sweet chestnut leaves only limited information has been available concerning its anti-inflammatory potential, including its ability to suppress TLR2 or TLR4 signaling pathways.

Common lady’s mantle (Alchemilla vulgaris)–whole plant

In our experiments, common lady’s mantle plant extract led to a decrease of LPS-induced IL-8 release and showed inhibitory effects on stimulated signaling pathways of both TLR2 and TLR4, but it displayed no inhibitory effects on nuclear NF-κB translocation. Thus, these results offer a promising starting point for identification of novel pro-inflammatory pathways and anti-inflammatory lead compounds. Common lady’s mantle has been used traditionally in Europe to treat several disorders like urogenital diseases, eczema, inflammation, diarrhea and sepsis [32, 33]. But also, in modern science, beneficial health effects like anti-viral, anti-oxidant and wound-healing properties have been reported [3235]. Again, its properties to mitigate TLR2 or TLR4 stimulation and to influence NF-κB signaling has not yet been described. However, ellagitannins, which are known to be present in Alchemilla vulgaris, were reported to mitigate inflammatory effects via the NF-κB pathway [27].

Besides these two extracts, with newly TLR2 and TLR4 dependent anti-inflammatory effects, the anti-inflammatory activities of our remaining herbal extracts are supported by existing descriptions in the literature. For several of these extracts, anti-inflammatory effects or the influence on TLR2 and TLR4 signaling pathways were solely described for single compounds but not for the complex mixtures present within the whole extract. Therefore, it is possible that further compounds, not investigated so far, contribute to their beneficial health effects.

Bearberry (Arctostaphylos uva-ursi)–leaves

In our assays, bearberry leaves showed strong dose-dependent anti-inflammatory activities in several cell lines and inhibition of LPS-induced NF-κB translocation. In addition, this extract mitigated the secretion of TNF-α accompanied with an increased secretion of IL-10 in macrophages, indicating a switch from pro-inflammatory M1-type to anti-inflammatory M2-type macrophages [26]. Bearberry leaves are traditionally used to treat symptoms of lower urinary tract infections and have been proven in modern medicine to possess anti-septic and anti-adhesion properties [36]. The most representative constituent of bearberry, the phenolic glycoside arbutin, is an anti-oxidant and anti-inflammatory agent [37]. Among others, arbutin significantly reduced production of pro-inflammatory cytokines including IL-1β and TNF-α, and other inflammation-related genes such as monocyte chemoattractant protein-1 (MCP-1) and IL-6. Furthermore, it inhibits the nuclear translocation of NF-κB [37]. These data support our data. Although arbutin is the major active ingredient, whole bearberry extracts are necessary to reveal the complete pharmaceutical activity [38]. The complex mixture of bearberry extract has not yet been reported to influence TLR2/TLR4 signaling pathways or to induce macrophage repolarization, making it a suitable candidate for further investigations.

Cinchona (Cinchona pubescens)–bark

Cinchona bark extract diminished LPS-induced inflammatory signals, especially in THP-1 monocytes and the comparative HEK-TLR2/TLR4 assay system, whereas LPS-induced NF-κB translocation was not affected. In vivo inhibition of TLR2- and TLR4-mediated signaling cascades by cinchonine from cinchona bark are demonstrated in literature, supporting our observed inhibition of pro-inflammatory cytokine production in stimulated TLR2 and TLR4 signaling pathways [39]. Besides cinchonine, further quinine derivatives in cinchona bark, e.g. chloroquine inhibited different TLR signaling pathways [40,41]. However, whole cinchona bark extract has, based on our present knowledge, not yet been described to influence TLR2 and/or TLR4 signaling pathways. Further analyses need to be performed to explore the underlying active constituents, besides cinchonine, especially those that affect TLR2 and TLR4 signaling.

Hops (Humulus lupulus)–cones

A strong decrease of pro-inflammatory cytokine production and a mitigation of LPS-induced NF-κB translocation were observed after hops extract treatment in our cell culture-based assays. Flavonoids from hops cones possess pharmaceutically important properties, e.g. anti-oxidant, anti-microbial, anti-carcinogenic, anti-inflammatory and estrogenic effects [42]. In particular, flavonoids like xanthohumol are effective inhibitors of arachidonic acid metabolism through inhibition of cyclooxygenase 1 and 2 (COX-1, COX-2) [43]. Furthermore, LPS-induced production of nitric oxide (NO), IL-1β, and TNF-α, and the activation of NF-κB signaling were inhibited by xanthohumol isolated from hops [44,45]. Again, it is likely that further compounds present in hops extract contribute to its strong anti-inflammatory effects demonstrated in our experiments.

White willow (Salix alba)–bark

Strong dose-dependent anti-inflammatory effects, as shown by decreased IL-8 production, were observed in TLR2- and TLR4-stimulated cell lines, coupled with a significant decrease in LPS-induced NF-κB translocation. White willow extract is commonly known for its beneficial health effects. Its active compound, the alcoholic β-glucoside salicin, was the basis for the discovery of Aspirin, a COX inhibitor, which still is one of the most widely used medicines in the world [46]. During absorption, salicin is metabolized into several salicylate derivatives resulting in pharmaceutical activities based on various components. This leads to a different mode of action compared to Aspirin and its pure compound acetylsalicylic acid, with less severe side effects [47]. In numerous in vitro and in vivo studies, anti-inflammatory effects have been shown for white willow extract, e.g. downregulation of several pro-inflammatory cytokines like TNF-α and inhibition of NF-κB translocation, which supports the data obtained by us [4850]. The anti-inflammatory effects have been credited to salicin [48]. However, salicin alone cannot satisfactorily explain the anti-inflammatory effects, and other compounds within the complex mixture, especially polyphenols such as flavonoids and proanthocyanidins, are suggested to contribute to its overall activity and thereby broaden the mechanisms of action [51].

Turmeric (Curcuma longa)–root

Our results suggest that TLR2/TLR4 signaling pathways are molecular targets of turmeric extract. This is supported by reports that show that turmeric and some of its compounds, especially curcumin and aromatic-turmerone, inhibit LPS-induced NF-κB activation, and expression of TLR4 and the downstream genes IL-1 receptor-associated kinase 1 (IRAK-1) and tumor necrosis factor receptor-associated factor 6 (TRAF-6) [52]. Treatment with curcumin inhibits both MyD88- and TRAM/TRIF-dependent pathways in LPS-induced TLR4 signaling. Production of TNF-α, IL-6 and ROS as well as levels of TLR4, MyD88 and the downstream effectors including NF-κB, interferon regulatory factor 3 (IRF3), MyD88, and TIRF were attenuated [5355]. Incubation with aromatic-turmerone, another compound of turmeric extract, blocked TLR4-mediated downstream signaling and the release of pro-inflammatory mediators [56]. Furthermore, Curcuma longa induced a shift from M1-type to M2-type macrophages in the murine RAW 264.7 macrophage cell line [57], which is in line with our data showing a decreased TNF-α secretion.

Rhubarb (Rheum palmatum)–root

Rhubarb root extract led to a decrease of LPS-induced cytokine production, inhibited both TLR2 and TLR4 signaling, nuclear NF-κB translocation, and induced M1-type to M2-type macrophage polarization. Treatment of inflammatory diseases with rhubarb roots has been described to significantly decrease TLR2, TLR4 and NF-κB mRNA, and protein expression in vivo [58,59], which is in line with our results. Emodin, which has been isolated from rhizomes of Rheum palmatum, inhibits several target molecules in inflammation and cancer, such as NF-κB and the serine/threonine kinase Akt, both important molecules in TLR2 and TLR4 signaling pathways [60]. Furthermore, emodin suppressed LPS-induced pro-inflammatory cytokines and chemokines as well as NF-κB inhibitor alpha (IκBα) degradation and thus NF-κB activation by disruption of lipid rafts [61]. Further research has to be conducted on whether the inhibition of TLR2- and TLR4 mediated signaling is only related to the reported effects of emodin or if further compounds present in the complex mixture of rhubarb root extract play a role in the observed anti-inflammatory effects.

Cinnamon (Cinnamomum verum)–bark

Cinnamon bark extract strongly mitigated pro-inflammatory cytokine production in stimulated TLR2 and TLR4 signaling pathways, and in LPS-induced nuclear NF-κB translocation. For cinnamon extract, several beneficial health effects are reported in literature, e.g. anti-inflammatory effects on murine alcohol-induced steatosis and colitis [6264]. Ethanolic cinnamon bark extracts have been shown to suppress release of TNF-α, IL-1β and IL-6 and NF-κB activation, in line with our data [65,66]. The major constituent present in cinnamon bark extract, trans-cinnamaldehyde, showed several anti-inflammatory effects [6770]. Still, more active compounds may be present in cinnamon extract.

Bilberries (Vaccinium myrtillus)–berries

Bilberry extract mitigated LPS-induced inflammatory signals, whereas LPS-induced NF-κB translocation was not affected. Bilberry extract, which is rich in antioxidant anthocyanins, showed anti-inflammatory effects in vivo, e.g. suppression of LPS-induced inducible nitric oxide synthase (iNOS), TNF-α, IL-1β and IL-6 transcripts, and iNOS, TNF-α and NF-κB protein levels in liver inflammation [71], compatible with our results. In a randomized, controlled dietary intervention for patients with the metabolic syndrome, bilberry supplementation reduced serum high-sensitivity C-reactive protein (CRP), IL-6, IL-12 and LPS levels, and downregulated genes associated with the TLR pathway [72]. Consumption of bilberry juice by patients with an increased risk for cardiovascular disease led to decreased plasma concentrations of CRP, IL-6 and IL-15 [73]. Overall, bilberry consumption may reduce low-grade metabolic inflammation, decreasing the risk of cardiometabolic diseases [72]. In our study, bilberries showed no inhibitory effects on nuclear translocation of NF-κB. In contrast, quercetin, resveratrol and epicatechin, all polyphenols present in bilberry, were shown to inhibit LPS-induced NF-κB activation in vitro, which suggests that conflicting effects of compounds in whole bilberry extract may play a significant role [73]. Taken together, both clinical studies demonstrate not only the anti-inflammatory potential of bilberries, but also their oral effectiveness in humans.

In conclusion, we showed that numerous ethanolic herbal extracts mitigate TLR2- and TLR4-dependent signaling and downstream inflammation. Several of them are known as officinal plants, but the underlying molecular mechanisms of their anti-inflammatory effects are often ill-defined or unknown. TLR pathways are of fundamental relevance in diverse inflammatory diseases. With the identification of orally active TLR-and/or NF-κB antagonizing herbal extracts and their active compounds, new promising treatment strategies might be developed. Moreover, a previously unknown attenuation of TLR-induced inflammatory responses by Castanea sativa leaves and Alchemilla vulgaris plants could be shown in our study. Interestingly, three out of the ten most effective anti-inflammatory extracts showed no inhibition of NF-κB translocation. Further identification of the active compounds of different extracts, investigations concerning their anti-inflammatory activity and their molecular targets within the signaling cascades are necessary. Furthermore, the anti-inflammatory activity demonstrated with our in vitro assays might differ in animals and humans due to alterations by digestion, absorption and metabolism of different compounds, as well as excretion of the extracts. Often, the overall efficacy in complex herbal mixtures is not only based on single compound activity, but on the combination of several different compounds. Hereby, not only additive but also synergistic effects were observed [7476]. Therefore, experiments with combinations of defined compounds can help to elucidate these interactions.

Supporting information

S1 Table. Overview of herbal extracts.

Scientific and common names, taxonomic information, active compounds and classification and reported beneficial health effects. ↓: downregulation, ↑: upregulation, AChE: acetylcholinesterase, ALP: alkaline phosphatase, ALT: alanine transaminase, AP-1: activator protein 1, AST: aspartate transaminase, CRP: C-reactive protein, CXCL1: chemokine (C-X-C motif) ligand 1, EGCG: epigallocatechin-3-gallate, ERK: extracellular signal-regulated kinase, Fg: Fibrinogen, GSH: glutathione, Hmox1: heme oxygenase (decycling) 1, IBD: inflammatory bowel disease, IFN: interferon, IL: interleukin, iNOS: inducible nitric oxide synthase, IRAK1: IL-1 receptor-associated kinase, IRF: interferon regulatory factor, IκBα: NF-κB inhibitor alpha, JNK: c-Jun N-terminal kinase, LDH: lactate dehydrogenase, LOX: lipoxygenase, MCP-1: monocyte chemoattractant protein-1, MDA: malondialdehyde, MPO: myeloperoxidase, MyD88: myeloid differentiation primary response 88, NF-κB: Nuclear factor-κB, NLRP3: NOD-like receptor family pyrin domain-containing 3, NO: nitric oxide, Nrf2: nuclear factor erythroid 2-related factor 2, PGE2: prostaglandin E2, RAGE: receptor for advanced glycation end products, RANKL: receptor activator of nuclear factor kappa-B ligand, ROS: reactive oxygen species, SOD: superoxide dismutase, TLR4: Toll-like receptor 4, TGFβ1: Transforming growth factor beta 1, TRAF6: TNF receptor-associated factor 6.

https://doi.org/10.1371/journal.pone.0203907.s001

(PDF)

S1 Fig. Cell viability and anti-inflammatory effects of ethanolic herbal extracts.

HeLa-TLR4 cells (red) and THP-1 monocytes (blue) were incubated with extracts (the ten extracts with highest anti-inflammatory potential are displayed in Fig 1, Fig 2 and Fig 3) or vehicle (70% ethanol), followed by stimulation with LPS-EB. Viability was measured using the Alamar Blue Assay was normalized to the negative control (untreated cells). TLR4 receptor activity was measured using Renilla luciferase expression for the HeLa-TLR4 cell line or IL-8 ELISA (pg/ml) for the THP-1 monocytes and was normalized to ethanol-treated cells. Data are displayed as viability (%) in the left graphs and TLR4 activity divided by normalized viability (%) in the right graphs. Data represents means (n≥2).

https://doi.org/10.1371/journal.pone.0203907.s002

(PDF)

S2 Fig. Anti- and pro-inflammatory effect of ethanolic extracts in HeLa-TLR4 reporter cells and THP-1 monocytes.

HeLa-TLR4 reporter cells or THP-1 monocytes were incubated with extracts in different concentrations or vehicle (70% ethanol), followed by stimulation with LPS-EB. Viability was measured using Alamar Blue Assay and was normalized to the negative control (Viability (%)). TLR4 receptor activity was measured using Renilla luciferase expression for the HeLa-TLR4 cell line or IL-8 ELISA (pg/ml) for THP-1 monocytes and was normalized to ethanol-treated cells (TLR4-Activity). Data are displayed as TLR4 stimulation divided by viability and sorted ascending by the following formula: (150—Viability (%)) * (2 * TLR4-Activity + 100) weighted in a ratio of 2:1 for THP-1 monocytes vs. HeLa-TLR4 cells. Data represents means (n≥2).

https://doi.org/10.1371/journal.pone.0203907.s003

(PDF)

S3 Fig. TLR2 and TLR4 antagonistic effects of ethanolic herbal extracts.

HEK-TLR2 cells (purple) and HEK-TLR4 cells (orange) were incubated with extracts (the five extracts with highest anti-inflammatory potential are displayed in Figs 5 and 6) or vehicle (70% ethanol), followed by stimulation of HEK-TLR2 cells with Pam2CSK4 or HEK-TLR4 cells with LPS-EB Ultrapure. Viability was measured using the Alamar Blue Assay was normalized to the respective negative control. TLR2 and TLR4 receptor activity were measured using SEAP production was normalized to ethanol-treated cells. Data are displayed as viability (%) in the left graphs and receptor activity divided by viability (%) in the right graphs. Data represents means (n≥2).

https://doi.org/10.1371/journal.pone.0203907.s004

(PDF)

S4 Fig. TLR2 and TLR4 antagonistic effects of select ethanolic extracts in HEK-TLR2 and HEK-TLR4 cell lines.

HEK-TLR2 or HEK-TLR4 cells were incubated with extracts in different concentrations or vehicle (70% ethanol), followed by stimulation of HEK-TLR2 cells with Pam2CSK4 or HEK-TLR4 cells with LPS-EB Ultrapure. Viability was measured using Alamar Blue Assay was normalized to the negative control. TLR2 and TLR4 receptor activity were measured using SEAP production and were normalized to ethanol-treated cells. Data are displayed as receptor activity divided by normalized viability. Data represents means (n≥2).

https://doi.org/10.1371/journal.pone.0203907.s005

(PDF)

Acknowledgments

The authors thank Fangxia Shen for providing samples from mung beans. The authors acknowledge the financial support by the Max Planck Graduate Center with the Johannes Gutenberg University Mainz (MPGC). They also acknowledge the stimulating exchange and discussions with the members of the Mainz Program for Chemical Allergology (MPCA), and numerous colleagues in the scientific communities in life sciences, pharmaceutics and medicine. Support by the IMB Microscopy and Histology Core Facility is gratefully acknowledged, as well as the German Research Foundation (DFG) for funding the Opera Phenix High Content Spinning Disk Microscope (Project 402386039).

References

  1. 1. Chahal DS, Sivamani RK, Isseroff RR, Dasu MR. Plant-based modulation of Toll-like receptors: an emerging therapeutic model. Phytother Res. 2013;27(10):1423–38. pmid:23147906
  2. 2. Liu X, Wu WY, Jiang BH, Yang M, Guo DA. Pharmacological tools for the development of traditional Chinese medicine. Trends Pharmacol Sci. 2013;34(11):620–8. pmid:24139610
  3. 3. Buenz EJ, Verpoorte R, Bauer BA. The Ethnopharmacologic Contribution to Bioprospecting Natural Products. Annu Rev Pharmacol Toxicol. 2018;6;58:509–530. pmid:29077533
  4. 4. McGrath KW, Icitovic N, Boushey HA, Lazarus SC, Sutherland ER, Chinchilli VM et al. A large subgroup of mild-to-moderate asthma is persistently noneosinophilic. Am J Respir Crit Care Med. 2012;185(6):612–9. pmid:22268133
  5. 5. Schuppan D, Kim YO. Evolving therapies for liver fibrosis. J Clin Invest. 2013;123(5):1887–901. pmid:23635787
  6. 6. Holmdahl R, Malmström V, Burkhardt H. Autoimmune priming, tissue attack and chronic inflammation—the three stages of rheumatoid arthritis. Eur J Immunol. 2014;44(6):1593–9. pmid:24737176
  7. 7. McAlees JW, Whitehead GS, Harley IT, Cappelletti M, Rewerts CL, Holdcroft AM et al. Distinct Tlr4-expressing cell compartments control neutrophilic and eosinophilic airway inflammation. Mucosal Immunol. 2015;8(4):863–73. pmid:25465099
  8. 8. Martel J, Ojcius DM, Chang CJ, Lin CS, Lu CC, Ko YF et al. Anti-obesogenic and antidiabetic effects of plants and mushrooms. Nat Rev Endocrinol. 2017;13(3):149–160. pmid:27636731
  9. 9. Lucas K, Maes M. Role of the Toll Like receptor (TLR) radical cycle in chronic inflammation: possible treatments targeting the TLR4 pathway. Mol Neurobiol., 2013;48(1):190–204. pmid:23436141
  10. 10. Walsh D, McCarthy J, O'Driscoll C, Melgar S. Pattern recognition receptors—molecular orchestrators of inflammation in inflammatory bowel disease. Cytokine Growth Factor Rev. 2013;24(2):91–104. pmid:23102645
  11. 11. Kramer CD, Genco CA. Microbiota, Immune Subversion, and Chronic Inflammation. Front Immunol. 2017;8:255. pmid:28348558
  12. 12. Takeuchi O, Akira S. Toll-like receptors; their physiological role and signal transduction system. Int Immunopharmacol. 2001;1(4):625–35. pmid:11357875
  13. 13. Ghosh S, Dass JF. Study of pathway cross-talk interactions with NF-κB leading to its activation via ubiquitination or phosphorylation: A brief review. Gene. 2016;584(1):97–109. pmid:26968890
  14. 14. Yamada M, Ichikawa T, Ii M, Sunamoto M, Itoh K, Tamura N et al. Discovery of novel and potent small-molecule inhibitors of NO and cytokine production as antisepsis agents: synthesis and biological activity of alkyl 6-(N-substituted sulfamoyl)cyclohex-1-ene-1-carboxylate. J Med Chem. 2005;48(23):7457–67. pmid:16279805
  15. 15. Youn HS, Lee JY, Saitoh SI, Miyake K, Kang KW, Choi YJ et al. Suppression of MyD88- and TRIF-dependent signaling pathways of Toll-like receptor by (-)-epigallocatechin-3-gallate, a polyphenol component of green tea. Biochem Pharmacol. 2006;72(7):850–9. pmid:16890209
  16. 16. Park SJ, Lee MY, Son BS, Youn HS. TBK1-targeted suppression of TRIF-dependent signaling pathway of Toll-like receptors by 6-shogaol, an active component of ginger. Biosci Biotechnol Biochem. 2009;73(7):1474–1478. pmid:19584560
  17. 17. Tidswell M, Tillis W, Larosa SP, Lynn M, Wittek AE, Kao R et al. Phase 2 trial of eritoran tetrasodium (E5564), a toll-like receptor 4 antagonist, in patients with severe sepsis. Crit Care Med. 2010;38(1):72–83. pmid:19661804
  18. 18. Peri F, Calabrese V. Toll-like receptor 4 (TLR4) modulation by synthetic and natural compounds: an update. J Med Chem. 2014;57(9):3612–3622. pmid:24188011
  19. 19. Tian J, Zhao Y, Liu Y, Liu Y, Chen K, Lyu S. Roles and Mechanisms of Herbal Medicine for Diabetic Cardiomyopathy: Current Status and Perspective. Oxid Med Cell Longev. 2017;2017:8214541. pmid:29204251
  20. 20. Zhao Y, Liu Y. A mechanistic overview of herbal medicine and botanical compounds to target transcriptional factors in Breast cancer. Pharmacol Res. 2017;pii: S1043-6618(17)30838-1.
  21. 21. Jiao L, Bi L, Lu Y, Wang Q, Gong Y, Shi J et al. Cancer chemoprevention and therapy using chinese herbal medicine. Biol Proced Online. 2018;20:1. pmid:29321719
  22. 22. Pan Y, Kong LD. High fructose diet-induced metabolic syndrome: pathophysiological mechanism and treatment by traditional Chinese medicine. Pharmacol Res. 2018;pii: S1043-6618(17)31549-9.
  23. 23. Lu CC, Hsu YJ, Chang CJ, Lin CS, Martel J, Ojcius DM et al. Immunomodulatory properties of medicinal mushrooms: differential effects of water and ethanol extracts on NK cell-mediated cytotoxicity. Innate Immun. 2016;22(7):522–33. pmid:27469258
  24. 24. Martel J, Ko YF, Ojcius DM, Lu CC, Chang CJ, Lin CS et al. Immunomodulatory Properties of Plants and Mushrooms. Trends Pharmacol Sci. 2017;38(11):967–981. pmid:28863984
  25. 25. Fraternale A, Brundu S, Magnani M. Polarization and Repolarization of Macrophages. J Clin Cell Immunol. 2015;6: 319.
  26. 26. Jovanović JA, Mihailović M, Uskoković AS, Grdović N, Dinić S, Poznanović G et al. Evaluation of the Antioxidant and Antiglycation Effects of Lactarius deterrimus and Castanea sativa Extracts on Hepatorenal Injury in Streptozotocin-Induced Diabetic Rats. Front Pharmacol. 2017;8:793. pmid:29163175
  27. 27. Landete JM. Ellagitannins, ellagic acid and their derived metabolites: a review about source, metabolism, functions and health. Food Research International. 2011;44(5):1150–1160.
  28. 28. Quave CL, Lyles JT, Kavanaugh JS, Nelson K, Parlet CP, Crosby HA et al. Castanea sativa (European Chestnut) Leaf Extracts Rich in Ursene and Oleanene Derivatives Block Staphylococcus aureus Virulence and Pathogenesis without Detectable Resistance. PLoS One. 2015;10(8):e0136486. pmid:26295163
  29. 29. De Vasconcelos MC, Bennett RN, Rosa EA, Ferreira-Cardoso JV. Composition of European chestnut (Castanea sativa Mill.) and association with health effects: fresh and processed products. J Sci Food Agric. 2010;90(10):1578–89. pmid:20564434
  30. 30. Pinto D, Rodrigues F, Braga N, Santos J, Pimentel FB, Palmeira-de-Oliveira A et al. The Castanea sativa bur as a new potential ingredient for nutraceutical and cosmetic outcomes: preliminary studies. Food Funct. 2017;8(1):201–208. pmid:27990543
  31. 31. Elliott R. Mechanisms of genomic and non-genomic actions of carotenoids. Biochim Biophys Acta. 2005;1740(2):147–54. pmid:15949681
  32. 32. Kiselova Y, Ivanova D, Chervenkov T, Gerova D, Galunska B, Yankova T. Correlation between the in vitro antioxidant activity and polyphenol content of aqueous extracts from Bulgarian herbs. Phytother Res. 2006;20(11):961–5. pmid:16906640
  33. 33. Takır S, Altun IH, Sezgi B, Süzgeç-Selçuk S, Mat A, Uydeş-Doǧan BS. Vasorelaxant and blood pressure lowering effects of alchemilla vulgaris: A comparative study of methanol and aqueous extracts. Pharmacogn Mag. 2015;11(41):163–9. pmid:25709228
  34. 34. Shrivastava R, Cucuat N, John GW. Effects of Alchemilla vulgaris and glycerine on epithelial and myofibroblast cell growth and cutaneous lesion healing in rats. Phytother Res. 2007;21(4):369–73. pmid:17236169
  35. 35. Filippova EI. Antiviral Activity of Lady's Mantle (Alchemilla vulgaris L.) Extracts against Orthopoxviruses. Bull Exp Biol Med. 2017;163(3):374–377. pmid:28744637
  36. 36. Yarnell E. Botanical medicines for the urinary tract. World J Urol. 2002;20(5):285–93. pmid:12522584
  37. 37. Lee HJ, Kim KW. Anti-inflammatory effects of arbutin in lipopolysaccharide-stimulated BV2 microglial cells. Inflamm Res. 2012;61(8):817–25. pmid:22487852
  38. 38. de Arriba SG, Naser B, Nolte KU. Risk assessment of free hydroquinone derived from Arctostaphylos Uva-ursi folium herbal preparations. Int J Toxicol. 2013;32(6):442–53. pmid:24296864
  39. 39. Jung SA, Choi M, Kim S, Yu R, Park T. Cinchonine Prevents High-Fat-Diet-Induced Obesity through Downregulation of Adipogenesis and Adipose Inflammation. PPAR Res. 2012;2012:541204. pmid:22675336
  40. 40. Kuznik A, Bencina M, Svajger U, Jeras M, Rozman B, Jerala R. Mechanism of endosomal TLR inhibition by antimalarial drugs and imidazoquinolines. J Immunol. 2011;186(8):4794–804. pmid:21398612
  41. 41. Kalia S, Dutz JP. New concepts in antimalarial use and mode of action in dermatology. Dermatol Ther. 2007;20(4):160–74. pmid:17970883
  42. 42. Karabin M, Hudcova T, Jelinek L, Dostalek P. Biotransformations and biological activities of hop flavonoids. Biotechnol Adv. 2015;33(6 Pt 2):1063–90.
  43. 43. Gerhäuser C Beer constituents as potential cancer chemopreventive agents. Eur J Cancer. 2005;41(13):1941–54. pmid:15953717
  44. 44. Albini A, Dell'Eva R, Vené R, Ferrari N, Buhler DR, Noonan DM et al. Mechanisms of the antiangiogenic activity by the hop flavonoid xanthohumol: NF-kappaB and Akt as targets. FASEB J. 2006;20(3):527–9. pmid:16403733
  45. 45. Lee IS, Lim J, Gal J, Kang JC, Kim HJ, Kang BY et al. Anti-inflammatory activity of xanthohumol involves heme oxygenase-1 induction via NRF2-ARE signaling in microglial BV2 cells. Neurochem Int. 2011;58(2):153–60. pmid:21093515
  46. 46. Desborough MJR, Keeling DM. The aspirin story—from willow to wonder drug. Br J Haematol. 2017;177(5):674–683. pmid:28106908
  47. 47. Vlachojannis J, Magora F, Chrubasik S. Willow species and aspirin: different mechanism of actions. Phytother Res. 2011;25(7):1102–4. pmid:21226125
  48. 48. Bonaterra GA, Heinrich EU, Kelber O, Weiser D, Metz J, Kinscherf R. Anti-inflammatory effects of the willow bark extract STW 33-I (Proaktiv) in LPS-activated human monocytes and differentiated macrophages. Phytomedicine. 2010;17(14):1106–13. pmid:20570123
  49. 49. Shakibaei M, Allaway D, Nebrich S, Mobasheri A. Botanical Extracts from Rosehip (Rosa canina), Willow Bark (Salix alba), and Nettle Leaf (Urtica dioica) Suppress IL-1β-Induced NF-κB Activation in Canine Articular Chondrocytes. Evid Based Complement Alternat Med. 2012;2012:509383. pmid:22474508
  50. 50. Shara M, Stohs SJ. Efficacy and Safety of White Willow Bark (Salix alba) Extracts. Phytother Res. 2015;29(8):1112–6. pmid:25997859
  51. 51. Nahrstedt A, Schmidt M, Jäggi R, Metz J, Khayyal MT. Willow bark extract: the contribution of polyphenols to the overall effect. Wien Med Wochenschr. 2007;157(13–14):348–51. pmid:17704985
  52. 52. Angel-Morales G, Noratto G, Mertens-Talcott SU. Standardized curcuminoid extract (Curcuma longa l.) decreases gene expression related to inflammation and interacts with associated microRNAs in human umbilical vein endothelial cells (HUVEC). Food Funct. 2012;3(12):1286–93. pmid:22972459
  53. 53. Youn HS, Saitoh SI, Miyake K, Hwang DH. Inhibition of homodimerization of Toll-like receptor 4 by curcumin. Biochem Pharmacol. 2006;72(1):62–9. pmid:16678799
  54. 54. Zhu HT, Bian C, Yuan JC, Chu WH, Xiang X, Chen F et al. Curcumin attenuates acute inflammatory injury by inhibiting the TLR4/MyD88/NF-κB signaling pathway in experimental traumatic brain injury. J Neuroinflammation. 2014;11:59. pmid:24669820
  55. 55. Yu S, Wang X, He X, Wang Y, Gao S, Ren L et al. Curcumin exerts anti-inflammatory and antioxidative properties in 1-methyl-4-phenylpyridinium ion (MPP(+))-stimulated mesencephalic astrocytes by interference with TLR4 and downstream signaling pathway. Cell Stress Chaperones. 2016;21(4):697–705. pmid:27164829
  56. 56. Chen M, Chang YY, Huang S, Xiao LH, Zhou W, Zhang LY et al. Aromatic-turmerone Attenuates LPS-Induced Neuroinflammation and Consequent Memory Impairment by Targeting TLR4-Dependent Signaling Pathway. Mol Nutr Food Res. 2018;62(2).
  57. 57. Li B, Hu Y, Zhao Y, Cheng M, Qin H, Cheng T et al. Curcumin Attenuates Titanium Particle-Induced Inflammation by Regulating Macrophage Polarization In Vitro and In Vivo. Front Immunol. 2017;8:55. pmid:28197150
  58. 58. Liu Y, Yan F, Liu Y, Zhang C, Yu H, Zhang Y et al. Aqueous extract of rhubarb stabilizes vulnerable atherosclerotic plaques due to depression of inflammation and lipid accumulation. Phytother Res. 2008;22(7):935–42. pmid:18384190
  59. 59. Yao P, Cui M, Li Y, Deng Y, Wu H. Effects of rhubarb on intestinal flora and toll-like receptors of intestinal mucosa in rats with severe acute pancreatitis. Pancreas. 2015;44(5):799–804. pmid:25931256
  60. 60. Shrimali D, Shanmugam MK, Kumar AP, Zhang J, Tan BK, Ahn KS et al. Targeted abrogation of diverse signal transduction cascades by emodin for the treatment of inflammatory disorders and cancer. Cancer Lett. 2013;341(2):139–49. pmid:23962559
  61. 61. Meng G, Liu Y, Lou C, Yang H. Emodin suppresses lipopolysaccharide-induced pro-inflammatory responses and NF-κB activation by disrupting lipid rafts in CD14-negative endothelial cells. Br J Pharmacol. 2010;161(7):1628–44. pmid:20726986
  62. 62. Kanuri G, Weber S, Volynets V, Spruss A, Bischoff SC, Bergheim I. Cinnamon extract protects against acute alcohol-induced liver steatosis in mice. J Nutr. 2009;139(3):482–7. pmid:19126670
  63. 63. Hagenlocher Y, Hösel A, Bischoff SC, Lorentz A. Cinnamon extract reduces symptoms, inflammatory mediators and mast cell markers in murine IL-10(-/-) colitis. 2016;J Nutr Biochem. 30:85–92. pmid:27012624
  64. 64. Hagenlocher Y, Satzinger S, Civelek M, Feilhauer K, Köninger J, Bischoff SC et al. Cinnamon reduces inflammatory response in intestinal fibroblasts in vitro and in colitis in vivo leading to decreased fibrosis. Mol Nutr Food Res. 2017;61(9).
  65. 65. Ho SC, Chang KS, Chang PW. Inhibition of neuroinflammation by cinnamon and its main components. Food Chem. 2013;138(4):2275–82. pmid:23497886
  66. 66. Gunawardena D, Govindaraghavan S, Münch G. Anti-inflammatory properties of cinnamon polyphenols and their monomeric precursors. In: Watson RR, Preedy VR, Zibadi S, editors. Polyphenols in Human Health and Disease, volume 1. Elsevier, Amsterdam, Boston, Heidelberg, London, New York, Oxford, Paris, San Diego, San Francisco, Singapore, Sydney, Tokyo, 2014. pp 30:409–425.
  67. 67. Chao LK, Hua KF, Hsu HY, Cheng SS, Lin IF, Chen CJ et al. Cinnamaldehyde inhibits pro-inflammatory cytokines secretion from monocytes/macrophages through suppression of intracellular signaling. Food Chem Toxicol. 2008;46(1):220–231. pmid:17868967
  68. 68. Youn HS, Lee JK, Choi YJ, Saitoh SI, Miyake K, Hwang DH et al. Cinnamaldehyde suppresses toll-like receptor 4 activation mediated through the inhibition of receptor oligomerization. Biochem Pharmacol. 2008;75(2):494–502. pmid:17920563
  69. 69. Gunawardena D, Karunaweera N, Lee S, van Der Kooy F, Harman DG, Raju R et al. Anti-inflammatory activity of cinnamon (C. zeylanicum and C. cassia) extracts–identification of E-cinnamaldehyde and o-methoxy cinnamaldehyde as the most potent bioactive compounds. Food Funct. 2015;6(3):910–9. pmid:25629927
  70. 70. Hagenlocher Y, Kiessling K, Schäffer M, Bischoff SC, Lorentz A. Cinnamaldehyde is the main mediator of cinnamon extract in mast cell inhibition. Eur J Nutr. 2015;54(8):1297–309. pmid:25504111
  71. 71. Luo H, Lv XD, Wang GE, Li YF, Kurihara H, He RR. Anti-inflammatory effects of anthocyanins-rich extract from bilberry (Vaccinium myrtillus L.) on croton oil-induced ear edema and Propionibacterium acnes plus LPS-induced liver damage in mice. Int J Food Sci Nutr. 2014;65(5):594–601. pmid:24548119
  72. 72. Kolehmainen M, Mykkänen O, Kirjavainen PV, Leppänen T, Moilanen E, Adriaens M et al. Bilberries reduce low-grade inflammation in individuals with features of metabolic syndrome. Mol Nutr Food Res. 2012;56(10):1501–10. pmid:22961907
  73. 73. Karlsen A, Paur I, Bøhn SK, Sakhi AK, Borge GI, Serafini M et al. Bilberry juice modulates plasma concentration of NF-kappaB related inflammatory markers in subjects at increased risk of CVD. Eur J Nutr. 2010;49(6):345–55. pmid:20119859
  74. 74. Stickel F, Schuppan D. Herbal medicine in the treatment of liver diseases. Dig Liver Dis. 2007;39:293–304. pmid:17331820
  75. 75. Wink M. Evolutionary advantage and molecular modes of action of multi-component mixtures used in phytomedicine. Curr. Drug Metab. 2008;9(10):996–1009. pmid:19075616
  76. 76. Wink . Modes of Action of Herbal Medicines and Plant Secondary Metabolites. Medicines. 2015;2(3):251–286. pmid:28930211