Skip to main content
  • Research article
  • Open access
  • Published:

Natural product HTP screening for antibacterial (E.coli 0157:H7) and anti-inflammatory agents in (LPS from E. coli O111:B4) activated macrophages and microglial cells; focus on sepsis

Abstract

Background

Acute systemic inflammatory response syndrome arising from infection can lead to multiple organ failure and death, with greater susceptibility occurring in immunocompromised individuals. Moreover, sub-acute chronic inflammation is a contributor to the pathology of diverse degenerative diseases (Parkinson’s disease, Alzheimer’s disease and arthritis). Given the known limitations in Western medicine to treat a broad range of inflammatory related illness as well as the emergence of antibiotic resistance, there is a renewed interest in complementary and alternative medicines (CAMs) to achieve these means.

Methods

A high throughput (HTP) screening of >1400 commonly sold natural products (bulk herbs, cooking spices, teas, leaves, supplement components, nutraceutical food components, fruit and vegetables, rinds, seeds, polyphenolics etc.) was conducted to elucidate anti-inflammatory substances in lipopolysaccharide (LPS) (E. coli serotype O111:B4) monocytes: RAW 264.7 macrophages [peripheral], BV-2 microglia [brain]) relative to hydrocortisone, dexamethasone and L-N6-(1Iminoethyl)lysine (L-NIL). HTP evaluation was also carried out for lethal kill curves against E.coli 0157:H7 1x106 CFU/mL relative to penicillin. Validation studies were performed to assess cytokine profiling using antibody arrays. Findings were corroborated by independent ELISAs and NO2–/iNOS expression quantified using the Griess Reagent and immunocytochemistry, respectively. For robust screening, we developed an in-vitro efficacy paradigm to ensure anti-inflammatory parameters were observed independent of cytotoxicity. This caution was taken given that many plants exert tumoricidal and anti-inflammatory effects at close range through similar signaling pathways, which could lead to false positives.

Results

The data show that activated BV-2 microglia cells (+ LPS 1μg/ml) release >10-fold greater IL-6, MIP1/2, RANTES and nitric oxide (NO2–), where RAW 264.7 macrophages (+ LPS 1μg/ml) produced > 10-fold rise in sTNFR2, MCP-1, IL-6, GCSF, RANTES and NO2–. Data validation studies establish hydrocortisone and dexamethasone as suppressing multiple pro-inflammatory processes, where L-NIL suppressed NO2–, but had no effect on iNOS expression or IL-6. The screening results demonstrate relative few valid hits with anti-inflammatory effects at < 250μg/ml for the following: Bay Leaf (Laurus nobilis), Elecampagne Root (Inula helenium), Tansy (Tanacetum vulgare),Yerba (Eriodictyon californicum) and Centipeda (Centipeda minima), Ashwagandha (Withania somnifera), Feverfew (Tanacetum parthenium), Rosemary (Rosmarinus officinalis), Turmeric Root (Curcuma Longa), Osha Root (Ligusticum porteri), Green Tea (Camellia sinensis) and constituents: cardamonin, apigenin, quercetin, biochanin A, eupatorin, (-)-epigallocatechin gallate (EGCG) and butein. Natural products lethal against [E. coli 0157:H7] where the LC50 < 100 μg/ml included bioactive silver hydrosol-Argentyn 23, green tea (its constituents EGCG > Polyphenon 60 > (-)-Gallocatechin > Epicatechin > (+)-Catechin), Grapeseed Extract (Vitis vinifera), Chinese Gallnut (its constituents gallic acid > caffeic acid) and gallic acid containing plants such as Babul Chall Bark (Acacia Arabica), Arjun (Terminalia Arjuna) and Bayberry Root Bark (Morella Cerifera).

Conclusions

These findings emphasize and validate the previous work of others and identify the most effective CAM anti-inflammatory, antibacterial compounds using these models. Future work will be required to evaluate potential combination strategies for long-term use to prevent chronic inflammation and possibly lower the risk of sepsis in immunocompromised at risk populations.

Peer Review reports

Background

Global health initiatives are encumbered by a vast majority of the population suffering from non-communicable inflammatory diseases such as cardiovascular disease, neurodegeneration, diabetes, arthritis, ulcerative colitis/bowel disease and cancer. Also, with increased incidence of antibiotic resistance, acute inflammation from sepsis plays a major role in mortalities arising from diverse infectious agents [1, 2]. Given limitations in Western medicine to treat/prevent a broad range of inflammatory related illness, there is a renewed interest in complementary and alternative medicines (CAMs) to achieve these means [39].

While there exist a plethora of scientific publications on the efficacy of individual CAMs in specific inflammatory models, there lacks a relative comparative potency rank of the most commonly marketed CAMs in a single study, conducted under uniform conditions. Our high throughput (HTP) screening library houses over 1400 products most which are available and sold to consumers throughout the world in the form of bulk herbs, cooking spices, teas, leaves, supplement components, nutraceutical food components, fruit and vegetables, roots, rinds, seeds, polyphenolics etc. The purpose of the current study is to screen commonly utilized CAMs for anti-inflammatory efficacy under uniform standard conditions to elucidate the most potent at non-toxic/low therapeutic concentrations (<250 μg/mL), and further to compare these to steroidal and NSAID drugs.

The in-vitro model employed was that of monocytes (peripheral and central nervous system) stimulated by lipopolysaccharide (LPS) derived from E. coli O111:B4. LPS is a cell wall endotoxic component from gram negative bacteria which evokes a deadly cytokine storm associated with septicemia, septic shock and multi organ failure. Known biologic consequences of LPS include the colossal release of chemotactic cytokines, IL-3 IL-12, TNF-alpha, IL-6, IL-1 beta, inducible nitric oxide (iNOS) NO3 -/NO2 -, P-selectin, CD 11b/CD18 (Mac-1) ICAM-1, PGE2 which enable massive neutrophil infiltration and hemolytic [1013]. While many of these inflammatory molecules at high concentrations are lethal, sub-chronic rises of the same are associated with age related inflammatory degenerative diseases such as Parkinson’s disease, Alzheimer’s disease and arthritis [1417]. Therefore, the use of LPS in this model and subsequent elucidation of the most effective CAMs against inflammatory parameters, can provide information on potential therapeutics for both chronic and acute inflammatory processes.

In this study, we conduct a HTP screening of CAMs to assess both capacity to kill a pathogenic strain of E.coli 0157:H7 as well as to mitigate the pro-inflammatory effects from E.Coli derived endotoxin cell wall component; LPS.

Methods

Hanks Balanced Salt Solution, (4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid) (HEPES), ethanol, sulfanilamide, 96 well plates, general reagents and supplies, were all purchased from Sigma-Aldrich, (St Louis, MO, USA) or VWR (Radnor, PA, USA). Imaging probes were purchased from Life Technologies (Grand Island, NY, USA). Natural products were purchased from Frontier Natural Products Co-op (Norway, IA, USA), Monterey Bay Spice Company (Watsonville, CA, USA), Mountain Rose Herbs (Eugene, OR, USA), Mayway Traditional Chinese Herbs (Oakland, CA, USA), Kalyx Natural Marketplace (Camden, NY, USA), Futureceuticals (Momence, IL, USA), organic fruit vegetable market: New Leaf (Tallahassee, FL, USA), Florida Food Products Inc. (Eustis, FL, USA), Patel Brothers Indian Grocery (Tampa, FL, USA), Opil Gold from Aging Sciences LLC (Wayland, MA, USA) and Colloidal Silver - Argentyn 23® Natural Immunogenics (Sarasota, FL, USA). Elisa kits and cytokine antibody arrays were purchased from Assay Biotech (Sunnyvale, CA) and Raybiotech (Norcross, GA, USA).

Cell culture

BV-2 microglia (BV-2) cells were provided by Elizabeta Blasi [18], and RAW 264.7 cells were purchased from American Type Culture Collection (Manassas, VA, USA). Cells were cultured in DMEM high glucose media [glucose 4500 mg/L] containing 5% FBS, 4 mM L-glutamine, and penicillin/streptomycin (100 U/0.1 mg/mL). Culture conditions were maintained at 37 °C in 5% CO2/atmosphere and every 2–3 days, the media was replaced and cells sub-cultured. For experiments, plating media consisted of DMEM (minus phenol red) [glucose 4500 mg/L], 2.5% FBS and penicillin/streptomycin (100 U/0.1 mg/mL). LPS O111:B4 was prepared in HBSS at 1 mg/mL and stored at –20 °C. For experiments, LPS was added to the culture media at a working concentration of 1μg/mL.

Bacterial culture

A single colony of E. coli O157:H7 was grown on an agar plate. E. coli was then inoculated into a 20 mL of Luria-Bertani (LB) in a flask, grown at 37 °C with moderate shaking (180 rpm), to an OD 600 nm = 0.6. One mL of the culture suspension was moved into a 1.5 mL Eppendorf tube and centrifuged for 1 min at 10,000 g (4 °C). After discarding the supernatant, the bacterial pellet was re- suspended in 1 mL sterilized water. This centrifugation was repeated twice. The bacteria were stored at 4 °C. The bacterial cell numbers were then determined using colony forming units (CFU) through serial dilution plating on LB plate at 37 °C. The experimental concentration of E. coli was 1 x 106 CFU/mL.

Sample preparation

All natural chemicals and reference drugs were dissolved in DMSO [5–20mg/mL] and crude herbs in absolute ethanol [50 mg/mL] after being diced, macerated and powdered then stored at –20 °C. All plants were cataloged by manufacturer, botanical and common names. All dilutions were prepared in sterile HBSS + 5 mM HEPES, adjusted to a pH of 7.4, ensuring solvent concentration of DMSO or absolute ethanol at less than 0.5%.

Cell and microbial -viability

Cell and microbial viability were assessed using resazurin [7-Hydroxy-3H-phenoxazin-3-one 10-oxide] (Alamar Blue) indicator dye [19]. A working solution of resazurin was prepared in sterile HBSS minus phenol red (0.5 mg/mL), then added (15% v/v) to each sample. Samples were returned to the incubator for 2–4 h, and reduction of the dye by viable cells (to resorufin, a fluorescent compound) was quantitatively analyzed using a Synergy HTX multi-mode reader Bio-Tek Inc. (Winooski, VT, USA) with settings at [550 nm/580 nm], [excitation/emission].

In-Vitro efficacy index

Several methodological concerns were addressed regarding HTP screenings. These included basic controls for pH (neutralized with buffered HBSS) and cell viability. In-vitro, immortal (malignant) immunocompetent cell lines such as glioma cells, macrophages, microglia, lymphocytes or granulocytes are of tumor origin, and many natural compounds simultaneously induce apoptosis in malignant cells and attenuate inflammation via the same pathways (i.e. phosphorylation of extracellular signal-regulated kinase (ERK), c-jun NH2-terminal kinase (JNK) phosphorylation and mitogen-activated protein kinases (MAPK)/NF-κB) [2026]. For this reason, we constructed and utilized an in-vitro efficacy index (iEI) paradigm, to ensure that anti-inflammatory effects are occurring at non-cytotoxic concentrations. The iEI is defined as the LC50 (toxic concentration)/IC50 (anti-inflammatory concentration) ratio, with higher values reflecting a greater confidence in the anti-inflammatory effects occurring independently of cell death.

Nitrite (NO2–)/iNOS expression

Quantification of nitrite (NO2−) was determined using the Greiss reagent [27]. The Greiss reagent was prepared by mixing an equal volume of 1.0% sulfanilamide in 10% phosphoric acid and 0.1% N-(1-naphthyl)-ethylenediamine in deionized water, which was added directly to the cell supernatant (experimental media consisting of DMEM - phenol red) and incubated at room temperature for 10 min. Controls and blanks were run simultaneously, and subtracted from the final value to eliminate interference. Samples were analyzed at 540 nm on a Synergy HTX multi-mode reader; Bio-Tek (Winooski, VT, USA).

iNOS protein expression was determined using immunocytochemistry. Cells were fixed in 4% paraformaldehyde/permeabilized in 0.2% Triton X 100 in phosphate buffered saline (PBS) and incubated with anti-iNOS, an N-Terminal antibody produced in rabbit for 24 h at 4 °C in a casein blocking buffer. Samples were washed in PBS, then incubated with anti-rabbit Alexa Fluor® 488 conjugate for two hours at RT. Samples were counterstained with propidium iodide and imaged using a fluorescent/inverted microscope, CCD camera and data acquisition using ToupTek View ; ToupTek Photonics Co (Zhejiang, P.R.China).

Mouse cytokine antibody array

Mouse Cytokine Antibody Arrays (Product Code: AAM-CYT-1000) Ray Biotech; (Norcross, GA, USA) were used to profile the effects of LPS (1μg/mL) on BV-2 and RAW 234.7 cell lines. Each experiment was carried out according to the manufacturer’s instructions, and in triplicate. Briefly, antibody-coated array membranes were first incubated for 30 min with 1 mL of blocking buffer. After 30 min, blocking buffer was decanted and replaced with 1 mL supernatant from control (untreated) samples, cells treated with (1ug/mL LPS only) and a media blank. Membranes were incubated overnight at 4 °C with mild shaking. The next day, the medium was decanted; membranes were washed, and subsequently incubated with 1 mL biotin-conjugated antibodies for 6 h. Lastly, biotin-conjugated antibodies were removed and membranes were incubated with HRP-conjugated streptavidin (2h), then evaluated for densitometry using a chemiluminescence substrate monitored on a VersaDoc Imager/Quantity One software from Bio-Rad: (Hercules, CA, USA).

IL-6 (Interleukin-6) ELISA

After experimental treatment, cells supernatants were directly evaluated for concentration of IL-6 using a Murine OmniKine™ IL-6 ELISA (Product Code # OK-0187), Assay Biotechnology Inc. (Sunnyvale, CA, USA), performed according to the manufacturer’s guidelines. Data was quantified by optical density at 450 nm using a Synergy HTX multi-mode reader from Bio-Tek (Winooski, VT, USA).

Data analysis

Statistical analysis was performed using Graph Pad Prism (version 3.0; Graph Pad Software Inc. San Diego, CA, USA) with significance of difference between the groups assessed using a one-way ANOVA then followed by Tukey post hoc means comparison test, or a Student’s t test. IC50s were determined by regression analysis using Origin Software (Origin Lab, Northampton, MA).

Results

Validation

Validation studies were conducted to determine profiled cytokine differentials in LPS activated RAW 264.7 (Fig. 1) and BV-2 cells (Fig. 2), respectively - using semi quantitative antibody microarrays, which were run in triplicate. The representative panel shows both cell lines exposed to LPS prompted the greater release of MCP-1, GCSF, MIP1a, MIP1g and MIP-2, sTNFR1/11, RANTES and IL-6. Quantitative analysis of IL-6 was corroborated by ELISA (Fig. 3), and iNOS protein expression was evaluated by ICC (Fig. 4b) and NO2- release using the Griess Reagent (Fig. 4a), the latter of which was reduced in the presence of iNOS inhibitor (L-NIL).

Fig. 1
figure 1

Cytokine release profile in LPS activated RAW 264.7 cells. The data are displayed as the cytokine array blot image and array grid layout with leading changes presented in highlighted boxes

Fig. 2
figure 2

Cytokine release profile in LPS activated BV-2 cells. The data are displayed as the cytokine array blot image and array grid layout with leading changes presented in highlighted boxes

Fig. 3
figure 3

ELISA: Quantification of IL-6 in the supernatant of resting and LPS activated cells (BV-2 cells, RAW 264.7 cells). The data represent IL-6 (pg/μl) and are expressed as the Mean ± S. E. M., n = 3. Differences between resting and LPS activated cells were determined by a student’s T test (*) P < 0.001

Fig. 4
figure 4

NO2- production in resting and LPS activated cells (BV-2 cells, RAW 264.7 cells) ± selective iNOS inhibitor: L-NIL (12 μg/mL). The data represent NO2- produced (μM) and are expressed as the Mean ± S. E. M., n = 3. Differences between resting and LPS activated cells were determined by a student’s T test (*) P < 0.001. Differences between LPS vs. LPS/L-NIL treated cells were determined by a student’s T test [*] P < 0.001. iNOS expression was analyzed by ICC using rabbit anti-mouse iNOS/goat anti-rabbit Alexafluor 488, in fixed permeabilized, propidium iodide nuclear counterstained cells (a) resting BV-2 cells (b) resting RAW 264.7 cells (c) LPS activated BV-2 cells (d) and RAW 264.7 cells

Screening

The initial HTP screening was conducted using a natural plant library housing over 1400 extracts including: 1) Plants: seeds, fruits, vegetables and herbs (of diverse ethnic nature including Chinese, Egyptian, Indian etc.) 2) Natural derived chemicals/polyphenolics 3) Metabolic Substrates: amino acids, vitamins and energy intermediates such as organic acids, glycolytic intermediates and 4) Reference NSAID and steroidal anti-inflammatory drugs. The preliminary screen was conducted to assess reduction of NO2- in LPS activated cells [equal to or less than 230 μg/mL] for all compounds (Fig. 5a). Substances that attenuated NO2– at less than 50% of the 1st tier starting concentrations, were re-evaluated over a dose range where LC50s (cytotoxicity) and IC50s (NO2-) were simultaneously evaluated (Fig. 5b, c). From the linear regression, LC50, IC50s concentrations were determined and in-vitro efficacy index (iEI) was calculated by the ratio value : LC50/IC50 . The higher the ratio, the greater confidence of true anti-inflammatory effects, not attributable to cell death. All iEI values are presented in Table 1 for RAW 264.7 cells/Table 2 for BV-2 cells, with matching logarithmic scatter-plots (Figs.6 and 7). Figure 8 shows a sample of NO2-/viability dose response data, with corresponding immunochemical imaging for iNOS in RAW 264.7 cells, where supernatant was evaluated for IL–6. The data from these experiments show that L-NIL, while capable of inhibiting the catalytic function of iNOS, was not an anti-inflammatory in the true sense. L-NIL suppressed NO2- but had no effects on cytokine release or expression of iNOS. Most lead compounds that reduced NO2- in both cell lines at sub-lethal concentrations (2 x IC50 for NO2-inhibition) which corresponded to a reduction of IL-6 in sample supernatant (Fig. 9). The antimicrobial effects of natural products on the survival of E.coli 0157:H7 (1x106 CFU/mL) were then evaluated. The data show only a select few have therapeutic potency relative to penicillin/streptomycin (Table 3), colloidal silver being the most effective (Fig. 10). The findings from this study delineate the most potent anti-inflammatory/and antibacterial natural compounds, when conducted in a uniform controlled fashion in these particular models.

Fig. 5
figure 5

High-through-put study design. The basic study layout consisted of a primary first level tier 1 screening by which all CAMs were tested to reduce NO2- in LPS treated BV-2 and RAW 264.7 cells [maximum working concentrations were : 230μg/mL (plant based) and 92μg/ml (metabolites, drugs and polyphenolics)]. a, b Compounds displaying an IC50 below the 1st tier concentrations were further evaluated as per the template (c). All compounds were simultaneously evaluated for toxicity/anti-inflammatory effects and an iEI differential was established (LC50/IC50) to prevent false positives incurred by cytotoxic effects

Table 1 Efficacy of anti-inflammatory CAMS relative to cellular toxicity in LPS activated RAW 264.7 cells
Table 2 Efficacy of natural anti-inflammatory compounds relative to cellular toxicity in LPS activated BV-2 cells
Fig. 6
figure 6

In-vitro efficacy scatter-plot for NO2–, inhibition vs. cell toxicity in RAW 264.7 cells. The data are presented as LC50 (toxicity) vs. IC50 (iNOS inhibition) determined from a regression analysis on a minimum of 6 concentrations

Fig. 7
figure 7

In-vitro efficacy scatter-plot for NO2–, inhibition vs. cell toxicity in BV-2 cells. The data are presented as LC50 (toxicity) vs. IC50 (iNOS inhibition) determined from a regression analysis on a minimum of 6 concentrations

Fig. 8
figure 8

Linear regression profiles for NO2- inhibition and toxicity in RAW 264.7 cells, by which LC50s and IC50s were calculated for all compounds, presented along with corresponding iNOS ICC images and IL-6 release measured at concentrations reflected by a square (). The data represent NO2- and viability (% LPS Control), presented as the Mean ± S.E.M, n = 4. Statistical difference from the Controls were determined by a one-way ANOVA, followed by a Tukey post – hoc test * P < 0.05 and IL-6 from controls by a student’s t-test* P < 0.05

Fig. 9
figure 9

Effects of lead anti-inflammatory products on IL-6 release in LPS treated BV-2 and RAW 264.7 cells. The data represents IL-6 (as % LPS Control) and expressed as the Mean ± S.E.M., n = 3. Differences between activated cells ± natural compounds at sub-lethal dose were determined by a student’s T test (*) P < 0.001

Table 3 Efficacy of CAM antibacterial compounds on survival of E.coli 0157:H7 (1x106 CFU/ml) @ 8 h in 31 °C
Fig. 10
figure 10

The effect of colloidal Silver-Argentyn 23® on the survival of E.coli 0157:H7 (1x106 CFU/ml) @ 8 h in 31°C. The data represent viability (% Control), presented as the Mean ± S.E.M, n = 4. Statistical differences from the Control were determined by a one-way ANOVA, using the Tukey post – hoc test. * P < 0.05

Discussion

The data from this study establish several findings including [A] uncorroborated anti-inflammatory/antimicrobial effects for over a thousand natural compounds at concentrations less than 230 μg/mL using this model; [B] corroborating data of existing work by other research groups regarding anti-inflammatory effects of green tea, curcumin, turmeric and rosemary ; and antimicrobial properties of green tea, its catechins, Chinese gallnut, gallic acid plant derived anti-fungal agents (cotton/gossypol) or silver nanoparticle dispersions [2837]. Lastly; [C] this work provides new evidence on some lesser acknowledged herbs to which historical medicinal value has been attributed, but little research has been documented. Some of these include the following.

Elecampane (Inula helenium) (IH) has extensive historical medicinal value, where its use dates back to the Iron Age (c. 800–450 B.C.) throughout the third century B.C. to 79 A.D. also mentioned by Pliny and further corroborated in the Chilandar Medical Codex (13th or 14th centuries A.D.) [38]. Within the last century, scientific documentation is somewhat sparse on this herb having primarily focused on its ability to cause allergic dermatitis or act as an anti-cancer agent attributable to the content of alantolactone and isoalantolactone [3944]. Meager work has been performed investigating the effect of IH on sepsis or age relate chronic inflammatory conditions. Although meager research has been conducted in IH, the findings presented here are in alignment with existing researchers who have reported its capacity to attenuate iNOS/NO2-, COX-2/PGE2, HMGB1 release and NF-κB in LPS-activated RAW 264.7 cells or phorbol activated T cells [4548]. Interestingly, although we did not find IH to have significant antimicrobial effects on E.coli 0157:H7 (1x106 CFU/mL) at the low concentrations criteria used in this study, others have reported its capacity to destroy invasive pathogens such as Staphylococcus aureus/methicillin-resistant (MRSA) gram-positive bacteria, yeasts parasites and Mycobacterium tuberculosis [42, 4953]. These studies suggest IH as being somewhat promising for attenuating inflammation arising from diverse infective or inflammatory insults.

The data from this work also show that fresh dried ethanol extracts of Bay leaf (BL) (Laurus nobilis) contains anti-inflammatory properties [54, 55]. Previous work by others demonstrates the oil (not aqueous) extracts to contain antimicrobial/food preserving properties due to cineole, eugenol, pinene, eucalyptol, linalool, carvacrol and α-terpinenyl acetate all evidentially toxic to Gram-positive bacteria (Staphylococcus aureus/pyogenes) and fungi (Candida albicans, Aspergillus fumigatus) [5659]. Again, regarding the aqueous extract of BL, our work corroborates the work of others having reported the capacity to attenuate LPS mediated microglia/macrophage activation thought to be attributable to its sesquiterpene content [60, 61]. These type of substances are thought to be beneficial in chronic age related degeneration, by not only reducing inflammation but also blocking neurotoxicity of AD pathological Aβ (25-35)-component fragments [62].

Another herbal extract used in the current study to which little data exists is Centipeda minima (CM). CM has previously been reported to contain high levels of helenalin with the capacity to LPS mediated elevation of NO2–,TNF-α, IL-1b, iNOS and cyclooxygenase-2 in macrophages [63]. CM also demonstrates the capacity to attenuate tissue injury in-vivo involving inflammation such as carrageen paw edema and liver fibrosis [6365]. Although we did not find CM to have significant antimicrobial effects on E.coli 0157:H7 (1 x 106 CFU/mL) at the low concentration criteria used in this study, others have reported its capacity to kill Enterobacter aerogenes, Staphylococcus aureus, Yersinia enterocolitica and Bacillus subtilis [66, 67].

Feverfew (Tanacetum parthenium) (TP) is another rarely evaluated herb which long been reported to treat inflammatory conditions including psoriasis, allergies, arthritis, asthma and particularly migraines [68]. TP derived sesquiterpene lactones such as parthenolide are believed to be responsible for observed anti-inflammatory effects in animal models of carrageenan-induced edema, osteoarthritis, colitis cystic fibrosis and phorbol triggered mouse-ear edema [6973]. TP constituents also antagonize toll-like receptors, Akt/mTOR and NF-κB pathways and block the downstream release of cytokines [74, 75]. Like the present study, previous research reports also corroborate capacity to reduce LPS activation of BV-2 cells and RAW 264.7 cells alike [76, 77]. There is also an antinociceptive aspect to feverfew commonly reported, having benefit to ameliorate pain associated with diabetic peripheral neuropathy [78].

The use of tansy (Tanacetum vulgare) (TV) as a medicinal plant was reported dating back to the 8th century A.D., when the Benedictine monks used it to treat intestinal worms, ameliorate digestive problems, fevers and sores. Interestingly, both feverfew and tansy have in common hyper allergenic potential due to parthenolide [79, 80]. Tansy is also rich in flavonoid glycosides, 7-O-glucosides of apigenin, luteolin, scutellarein and 6- hydroxyluteolin, chrysoeriol and eriodictyol as well as aglycones, hispidulin, nepetin, eupatilin, jaceosidin, pectolinarigenin and axillarin [81]. The oil contains 1,8-cineole and β-thujone as a major constituent along with carveol, umbellulone, davanone, dihydrocarvone, chrysanthenol, borneol and myrtenol [8285].

Ashwagandha (Withania somnifera) (WS) is a highly studied herb with a plethora of known health benefits, in particular for prevention of cardiovascular disease. Its primary bioactive compound (Withaferin A) directly inhibits β1-adrenergic receptors, HMG-CoA, angiotensinogen-converting enzyme, total cholesterol, triglycerides, low density lipoprotein and elevation of protective high density lipoproteins and endogenous antioxidant systems [86, 87]. In animal models, WS prevents isoproterenol induced myocardial infarction, stroke distal middle cerebral artery occlusion and monocrotaline induced pulmonary hypertension in rats [8891]. With respect to the immune system, WS can attenuate mitogen induced T/B-cell activation, secretion of Th1 and Th2 cytokines and inhibit NF-κB nuclear translocation in lymphocytes [92]. These immunomodulating effects of WS are also reported in-vitro for systemic LPS or E. coli administration in mammals, where there is a reduction in neutrophil tissue infiltration [93, 94] as well as tissue damage and pain associated with rheumatoid arthritis [9597]. Ashwagandha is also an anti-infective agent lethal to gram-positive bacteria/cocci such as methicillin resistant Staphylococcus aureus and Enterococcus, respectively [98]. The data in this study ranks, validates and confirms pre-existing research showing significant antimicrobial effects of green tea EGCG polyphenon-60 (PP-60) Acacia arabica,grapeseed extract, caffeic, gallic acid, chapparal (Larrea tridentata) [99109], where little has been investigated on antimicrobial herbs such as balm of Gilead Bud (Populus candicans), an herb of great historical significance with observed antibacterial and anti-inflammatory properties.

Conclusion

In conclusion, the data obtained in this work affords general information on validated CAM anti-inflammatory and antimicrobial compounds and relative potency at sub lethal concentrations in LPS activated BV-2 and RAW 264.7 cells. Moreover, the data obtained also provide relative lethal potency of CAMs against the growth of E.coli 0157:H7. These findings can serve as a guide for future examination of specific CAM based herbal/nutraceutical anti-inflammatory/antimicrobial modalities for use in prevention or treatment of disease.

Abbreviations

ANOVA:

One-way analysis of variance

CAMs:

Complementary and alternative medicines

CM:

Centipeda minima

CNS:

Central nervous system

COX2:

Cyclooxygenase-2

DMEM:

Dulbecco’s modified eagle medium

EGCG:

(-)-epigallocatechin gallate

ELISA:

Enzyme-linked immunosorbent assay

GCSF:

Granulocyte-colony stimulating factor

HMGB1:

High mobility group box 1 protein

HTP:

High throughput

iEI:

in-vitro efficacy index

IH:

Inula helenium

INOS:

Inducible nitric oxide

LPS:

Lipopolysaccharide

MSRA:

Methicillin-resistant

NO2-:

Nitrite

NSAID:

Non-steroidal anti-inflammatory drugs

PBS:

phosphate buffered saline

RANTES:

Regulated on activation, normal T cell expressed and secreted

TP:

Tanacetum parthenium (Feverfew)

TV:

Tanacetum vulgare (Tansy)

WS:

Withania somnifera (Ashwagandha)

References

  1. Hostinar CE, Ross KM, Chen E, Miller GE. Modeling the association between lifecourse socioeconomic disadvantage and systemic inflammation in healthy adults: The role of self-control. Health Psychol. 2015;34(6):580–90.

    PubMed  Google Scholar 

  2. Hebert JR, Braun KL, Kaholokula JK, Armstead CA, Burch JB, Thompson B. Considering the Role of Stress in Populations of High-Risk, Underserved Community Networks Program Centers. Prog Community Health Partnersh. 2015;9(Suppl):71–82.

    PubMed  PubMed Central  Google Scholar 

  3. Yang YH, Rajaiah R, Lee DY, Ma Z, Yu H, Fong HH, Lao L, Berman BM, Moudgil KD. Suppression of ongoing experimental arthritis by a chinese herbal formula (huo-luo-xiao-ling dan) involves changes in antigen-induced immunological and biochemical mediators of inflammation. Evid Based Complement Alternat Med. 2011;2011:642027.

    PubMed  Google Scholar 

  4. Vojdani A, Lambert J, Kellermann G. The Role of Th17 in Neuroimmune Disorders: A Target for CAM Therapy. Part III. Evid Based Complement Alternat Med. 2011;2011:548086.

    PubMed  PubMed Central  Google Scholar 

  5. Kes VB, Cesarik M, Matovina LZ, Zavoreo I, Coric L, Drnasin S, Demarin V. The role of complementary and alternative medicine in therapy of multiple sclerosis. Acta Clin Croat. 2013;52(4):464–71.

    PubMed  Google Scholar 

  6. Lissat A, Joerschke M, Shinde DA, Braunschweig T, Meier A, Makowska A, Bortnick R, Henneke P, Herget G, Gorr TA, et al. IL6 secreted by Ewing sarcoma tumor microenvironment confers anti-apoptotic and cell-disseminating paracrine responses in Ewing sarcoma cells. BMC Cancer. 2015;15:552.

    PubMed  PubMed Central  Google Scholar 

  7. Yao H, Chen Y, Zhang L, He X, Lian L, Wu X, Lan P. Carnosol inhibits cell adhesion molecules and chemokine expression by tumor necrosis factor-alpha in human umbilical vein endothelial cells through the nuclear factor-kappaB and mitogen-activated protein kinase pathways. Mol Med Rep. 2014;9(2):476–80.

    CAS  PubMed  Google Scholar 

  8. Salaga M, Zatorski H, Sobczak M, Chen C, Fichna J. Chinese herbal medicines in the treatment of IBD and colorectal cancer: a review. Curr Treat Options Oncol. 2014;15(3):405–20.

    PubMed  PubMed Central  Google Scholar 

  9. Kwak S, Ku SK, Bae JS. Fisetin inhibits high-glucose-induced vascular inflammation in vitro and in vivo. Inflamm Res. 2014;63(9):779–87.

    CAS  PubMed  Google Scholar 

  10. Weber GF, Chousterman BG, He S, Fenn AM, Nairz M, Anzai A, Brenner T, Uhle F, Iwamoto Y, Robbins CS, et al. Interleukin-3 amplifies acute inflammation and is a potential therapeutic target in sepsis. Science. 2015;347(6227):1260–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Martin I, Caban-Hernandez K, Figueroa-Santiago O, Espino AM. Fasciola hepatica fatty acid binding protein inhibits TLR4 activation and suppresses the inflammatory cytokines induced by lipopolysaccharide in vitro and in vivo. J Immunol. 2015;194(8):3924–36.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Lee SK, Park YJ, Ko MJ, Wang Z, Lee HY, Choi YW, Bae YS. A novel natural compound from garlic (Allium sativum L.) with therapeutic effects against experimental polymicrobial sepsis. Biochem Biophys Res Commun. 2015;464(3):774–9.

    CAS  PubMed  Google Scholar 

  13. Barichello T, Generoso JS, Silvestre C, Costa CS, Carrodore MM, Cipriano AL, Michelon CM, Petronilho F, Dal-Pizzol F, Vilela MC, et al. Circulating concentrations, cerebral output of the CINC-1 and blood-brain barrier disruption in Wistar rats after pneumococcal meningitis induction. Eur J Clin Microbiol Infect Dis. 2012;31(8):2005–9.

    CAS  PubMed  Google Scholar 

  14. Bessler H, Djaldetti R, Salman H, Bergman M, Djaldetti M. IL-1 beta, IL-2, IL-6 and TNF-alpha production by peripheral blood mononuclear cells from patients with Parkinson’s disease. Biomed Pharmacother. 1999;53(3):141–5.

    CAS  PubMed  Google Scholar 

  15. Blum-Degen D, Muller T, Kuhn W, Gerlach M, Przuntek H, Riederer P. Interleukin-1 beta and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer’s and de novo Parkinson’s disease patients. Neurosci Lett. 1995;202(1-2):17–20.

    CAS  PubMed  Google Scholar 

  16. Brodacki B, Staszewski J, Toczylowska B, Kozlowska E, Drela N, Chalimoniuk M, Stepien A. Serum interleukin (IL-2, IL-10, IL-6, IL-4), TNFalpha, and INFgamma concentrations are elevated in patients with atypical and idiopathic parkinsonism. Neurosci Lett. 2008;441(2):158–62.

    CAS  PubMed  Google Scholar 

  17. Zhu L, Wang J, Wei T, Gao J, He H, Chang X, Yan T. Effects of Naringenin on inflammation in complete freund’s adjuvant-induced arthritis by regulating Bax/Bcl-2 balance. Inflammation. 2015;38(1):245–51.

    CAS  PubMed  Google Scholar 

  18. Blasi E, Barluzzi R, Bocchini V, Mazzolla R, Bistoni F. Immortalization of murine microglial cells by a v-raf/v-myc carrying retrovirus. J Neuroimmunol. 1990;27(2-3):229–37.

    CAS  PubMed  Google Scholar 

  19. Evans SM, Casartelli A, Herreros E, Minnick DT, Day C, George E, Westmoreland C. Development of a high throughput in vitro toxicity screen predictive of high acute in vivo toxic potential. Toxicol In Vitro. 2001;15(4-5):579–84.

    CAS  PubMed  Google Scholar 

  20. Zhang X, Luo W, Zhao W, Lu J, Chen X. Isocryptotanshinone Induced Apoptosis and Activated MAPK Signaling in Human Breast Cancer MCF-7 Cells. J Breast Cancer. 2015;18(2):112–8.

    PubMed  PubMed Central  Google Scholar 

  21. Huang WC, Hsu RM, Chi LM, Leu YL, Chang YS, Yu JS. Selective downregulation of EGF receptor and downstream MAPK pathway in human cancer cell lines by active components partially purified from the seeds of Livistona chinensis R. Brown Cancer Lett. 2007;248(1):137–46.

    CAS  PubMed  Google Scholar 

  22. Amirghofran Z. Herbal medicines for immunosuppression. Iran J Allergy Asthma Immunol. 2012;11(2):111–9.

    PubMed  Google Scholar 

  23. Pan HC, Jiang Q, Yu Y, Mei JP, Cui YK, Zhao WJ. Quercetin promotes cell apoptosis and inhibits the expression of MMP-9 and fibronectin via the AKT and ERK signalling pathways in human glioma cells. Neurochem Int. 2015;80:60–71.

    CAS  PubMed  Google Scholar 

  24. Cheng YW, Chang CY, Lin KL, Hu CM, Lin CH, Kang JJ. Shikonin derivatives inhibited LPS-induced NOS in RAW 264.7 cells via downregulation of MAPK/NF-kappaB signaling. J Ethnopharmacol. 2008;120(2):264–71.

    CAS  PubMed  Google Scholar 

  25. Chen CC, Tsai PC, Wei BL, Chiou WF. 8-Prenylkaempferol suppresses inducible nitric oxide synthase expression through interfering with JNK-mediated AP-1 pathway in murine macrophages. Eur J Pharmacol. 2008;590(1-3):430–6.

    CAS  PubMed  Google Scholar 

  26. Ci X, Liang X, Luo G, Yu Q, Li H, Wang D, Li R, Deng X. Regulation of inflammatory mediators in lipopolysaccharide-stimulated RAW 264.7 cells by 2”-hydroxy-3”-en-anhydroicaritin involves down-regulation of NF-kappaB and MAPK expression. Int Immunopharmacol. 2010;10(9):995–1002.

    CAS  PubMed  Google Scholar 

  27. Cendan JC, Topping DL, Pruitt J, Snowdy S, Copeland 3rd EM, Lind DS. Inflammatory mediators stimulate arginine transport and arginine-derived nitric oxide production in a murine breast cancer cell line. J Surg Res. 1996;60(2):284–8.

    CAS  PubMed  Google Scholar 

  28. Yang F, de Villiers WJ, McClain CJ, Varilek GW. Green tea polyphenols block endotoxin-induced tumor necrosis factor-production and lethality in a murine model. J Nutr. 1998;128(12):2334–40.

    CAS  PubMed  Google Scholar 

  29. Chan MM, Fong D, Ho CT, Huang HI. Inhibition of inducible nitric oxide synthase gene expression and enzyme activity by epigallocatechin gallate, a natural product from green tea. Biochem Pharmacol. 1997;54(12):1281–6.

    CAS  PubMed  Google Scholar 

  30. Senthil Kumaran V, Arulmathi K, Sundarapandiyan R, Kalaiselvi P. Attenuation of the inflammatory changes and lipid anomalies by epigallocatechin-3-gallate in hypercholesterolemic diet fed aged rats. Exp Gerontol. 2009;44(12):745–51.

    CAS  PubMed  Google Scholar 

  31. Bengmark S, Mesa MD, Gil A. Plant-derived health: the effects of turmeric and curcuminoids. Nutr Hosp. 2009;24(3):273–81.

    CAS  PubMed  Google Scholar 

  32. Al-Okbi SY. Nutraceuticals of anti-inflammatory activity as complementary therapy for rheumatoid arthritis. Toxicol Ind Health. 2014;30(8):738–49.

    CAS  PubMed  Google Scholar 

  33. Liang W, Fernandes AP, Holmgren A, Li X, Zhong L. Bacterial thioredoxin and thioredoxin reductase as mediators for epigallocatechin 3-gallate-induced antimicrobial action. FEBS J. 2016;283(3):446–58.

    CAS  PubMed  Google Scholar 

  34. Noormandi A, Dabaghzadeh F. Effects of green tea on Escherichia coli as a uropathogen. J Tradit Complement Med. 2015;5(1):15–20.

    PubMed  Google Scholar 

  35. Shao D, Li J, Li J, Tang R, Liu L, Shi J, Huang Q, Yang H. Inhibition of Gallic Acid on the Growth and Biofilm Formation of Escherichia coli and Streptococcus mutans. J Food Sci. 2015;80(6):M1299–305.

    CAS  PubMed  Google Scholar 

  36. Chen XY, Chen Y, Heinstein P, Davisson VJ. Cloning, expression, and characterization of (+)-delta-cadinene synthase: a catalyst for cotton phytoalexin biosynthesis. Arch Biochem Biophys. 1995;324(2):255–66.

    CAS  PubMed  Google Scholar 

  37. He M, Lu L, Zhang J, Li D. Immobilized Silver Nanoparticles on Chitosan with Special Surface State-Enhanced Antimicrobial Efficacy and Reduced Cytotoxicity. J Nanosci Nanotechnol. 2015;15(9):6435–43.

    CAS  PubMed  Google Scholar 

  38. Al-Gammal SY. Elecampane and Job’s disease. Bull Indian Inst Hist Med Hyderabad. 1998;28(1):7–11.

    CAS  PubMed  Google Scholar 

  39. P’Iankova ZP. Nugmanova ML: [Dermatitis caused by elecampane]. Vestn Dermatol Venerol. 1975;12:52–4.

    Google Scholar 

  40. Wang J, Zhao YM, Zhang B, Guo CY. Protective Effect of Total Phenolic Compounds from Inula helenium on Hydrogen Peroxide-induced Oxidative Stress in SH-SY5Y Cells. Indian J Pharm Sci. 2015;77(2):163–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhao YM, Wang J, Liu HB, Guo CY, Zhang WM. Microwave-assisted Extraction of Alantolactone and Isoalantolactone from Inula helenium. Indian J Pharm Sci. 2015;77(1):116–20.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Cantrell CL, Abate L, Fronczek FR, Franzblau SG, Quijano L, Fischer NH. Antimycobacterial eudesmanolides from Inula helenium and Rudbeckia subtomentosa. Planta Med. 1999;65(4):351–5.

    CAS  PubMed  Google Scholar 

  43. Rasul A, Khan M, Ali M, Li J, Li X. Targeting apoptosis pathways in cancer with alantolactone and isoalantolactone. ScientificWorldJournal. 2013;2013:248532.

    PubMed  PubMed Central  Google Scholar 

  44. Yang C, Yang J, Sun M, Yan J, Meng X, Ma T. Alantolactone inhibits growth of K562/adriamycin cells by downregulating Bcr/Abl and P-glycoprotein expression. IUBMB Life. 2013;65(5):435–44.

    CAS  PubMed  Google Scholar 

  45. Cabronero MC, Campos E, Picazo JJ, Romero J. Evaluation of an agglutination method of latex particles sensitized for the detection of vaginal candidiasis. Med Clin (Barc). 1990;94(9):329–32.

    CAS  Google Scholar 

  46. Nesterova Iu V, Zelenskaia KL, Vetoshkina TV, Aksinenko SG, Gorbacheva AV, Gorbatykh NA. Mechanisms of antistressor activity of Inula helenium preparations. Eksp Klin Farmakol. 2003;66(4):63–5.

    CAS  PubMed  Google Scholar 

  47. Danilets MG, Belska NV, Bel’sky YP, Uchasova EG, Trophimova ES, Ligatcheva AA, Guriev AM, Belousov MV, Ahmedganov RR, Usubov MS, et al. Effects of plant water-soluble polysaccharides on the production of immunoglobulins E and G1 by lymphocytes of mice sensitized with ovalbumin. Bull Exp Biol Med. 2008;146(5):585–7.

    CAS  PubMed  Google Scholar 

  48. Na BR, Kim HR, Kwon MS, Lee HS, Piragyte I, Choi EJ, Choi HK, Han WC, Lee SH, Jun CD. Aplotaxene blocks T cell activation by modulation of protein kinase C-theta-dependent pathway. Food Chem Toxicol. 2013;62:23–31.

    CAS  PubMed  Google Scholar 

  49. Radulovic NS, Denic MS, Stojanovic-Radic ZZ. Synthesis of small combinatorial libraries of natural products: identification and quantification of new long-chain 3-methyl-2-alkanones from the root essential oil of Inula helenium L. (Asteraceae). Phytochem Anal. 2014;25(1):75–80.

    CAS  PubMed  Google Scholar 

  50. Gokbulut A, Ozhan O, Satilmis B, Batcioglu K, Gunal S, Sarer E. Antioxidant and antimicrobial activities, and phenolic compounds of selected Inula species from Turkey. Nat Prod Commun. 2013;8(4):475–8.

    CAS  PubMed  Google Scholar 

  51. O’Shea S, Lucey B, Cotter L. In vitro activity of Inula helenium against clinical Staphylococcus aureus strains including MRSA. Br J Biomed Sci. 2009;66(4):186–9.

    PubMed  Google Scholar 

  52. El Garhy MF, Mahmoud LH. Anthelminthic efficacy of traditional herbs on Ascaris lumbricoides. J Egypt Soc Parasitol. 2002;32(3):893–900.

    PubMed  Google Scholar 

  53. Cantrell CL, Fischer NH, Urbatsch L, McGuire MS, Franzblau SG. Antimycobacterial crude plant extracts from South, Central, and North America. Phytomedicine. 1998;5(2):137–45.

    CAS  PubMed  Google Scholar 

  54. Kilic A, Hafizoglu H, Kollmannsberger H, Nitz S. Volatile constituents and key odorants in leaves, buds, flowers, and fruits of Laurus nobilis L. J Agric Food Chem. 2004;52(6):1601–6.

    CAS  PubMed  Google Scholar 

  55. Yahyaa M, Matsuba Y, Brandt W, Doron-Faigenboim A, Bar E, McClain A, Davidovich-Rikanati R, Lewinsohn E, Pichersky E, Ibdah M. Identification, Functional Characterization, and Evolution of Terpene Synthases from a Basal Dicot. Plant Physiol. 2015;169(3):1683–97.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Marzouki H, Khaldi A, Marongiu B, Piras A, Harzallah-Skhiri F. Chemical polymorphism of essential oils from populations of Laurus nobilis grown on Tunisia. Algeria and France Nat Prod Commun. 2011;6(10):1483–6.

    CAS  PubMed  Google Scholar 

  57. Ramos C, Teixeira B, Batista I, Matos O, Serrano C, Neng NR, Nogueira JM, Nunes ML, Marques A. Antioxidant and antibacterial activity of essential oil and extracts of bay laurel Laurus nobilis Linnaeus (Lauraceae) from Portugal. Nat Prod Res. 2012;26(6):518–29.

    CAS  PubMed  Google Scholar 

  58. Fukuyama N, Ino C, Suzuki Y, Kobayashi N, Hamamoto H, Sekimizu K, Orihara Y. Antimicrobial sesquiterpenoids from Laurus nobilis L. Nat Prod Res. 2011;25(14):1295–303.

    CAS  PubMed  Google Scholar 

  59. Liu MH, Otsuka N, Noyori K, Shiota S, Ogawa W, Kuroda T, Hatano T, Tsuchiya T. Synergistic effect of kaempferol glycosides purified from Laurus nobilis and fluoroquinolones on methicillin-resistant Staphylococcus aureus. Biol Pharm Bull. 2009;32(3):489–92.

    CAS  PubMed  Google Scholar 

  60. Chen H, Xie C, Wang H, Jin DQ, Li S, Wang M, Ren Q, Xu J, Ohizumi Y, Guo Y. Sesquiterpenes inhibiting the microglial activation from Laurus nobilis. J Agric Food Chem. 2014;62(20):4784–8.

    CAS  PubMed  Google Scholar 

  61. De Marino S, Borbone N, Zollo F, Ianaro A, Di Meglio P, Iorizzi M. New sesquiterpene lactones from Laurus nobilis leaves as inhibitors of nitric oxide production. Planta Med. 2005;71(8):706–10.

    PubMed  Google Scholar 

  62. Pacifico S, Gallicchio M, Lorenz P, Duckstein SM, Potenza N, Galasso S, Marciano S, Fiorentino A, Stintzing FC, Monaco P. Neuroprotective potential of Laurus nobilis antioxidant polyphenol-enriched leaf extracts. Chem Res Toxicol. 2014;27(4):611–26.

    CAS  PubMed  Google Scholar 

  63. Huang SS, Chiu CS, Lin TH, Lee MM, Lee CY, Chang SJ, Hou WC, Huang GJ, Deng JS. Antioxidant and anti-inflammatory activities of aqueous extract of Centipeda minima. J Ethnopharmacol. 2013;147(2):395–405.

    CAS  PubMed  Google Scholar 

  64. Qin RA, Mei X, Wan L, Shi JL, Shen YJ. Effects of the volatile oil of Centipeda minima on acute pleural effusion in rats induced by an intrapleural injection of car. Zhongguo Zhong Yao Za Zhi. 2005;30(15):1192–4.

    PubMed  Google Scholar 

  65. Lin X, Zhang S, Huang R, Wei L, Tan S, Liang S, Tian Y, Wu X, Lu Z, Huang Q. Helenalin attenuates alcohol-induced hepatic fibrosis by enhancing ethanol metabolism, inhibiting oxidative stress and suppressing HSC activation. Fitoterapia. 2014;95:203–13.

    CAS  PubMed  Google Scholar 

  66. Soetardjo Jr S, Jong PC, Ahmad MN, Lachimanan YL, Sreenivasan S. Chemical Composition and Biological Activity of the Centipeda minima (Asteraceae). Malays J Nutr. 2007;13(1):81–7.

    PubMed  Google Scholar 

  67. Taylor RS, Towers GH. Antibacterial constituents of the Nepalese medicinal herb, Centipeda minima. Phytochemistry. 1998;47(4):631–4.

    CAS  PubMed  Google Scholar 

  68. Murphy JJ, Heptinstall S, Mitchell JR. Randomised double-blind placebo-controlled trial of feverfew in migraine prevention. Lancet. 1988;2(8604):189–92.

    CAS  PubMed  Google Scholar 

  69. Zhao Y, Chen SJ, Wang JC, Niu HX, Jia QQ, Chen XW, Du XY, Lu L, Huang B, Zhang Q, et al. Sesquiterpene lactones inhibit advanced oxidation protein product-induced MCP-1 expression in podocytes via an IKK/NF-kappaB-dependent mechanism. Oxid Med Cell Longev. 2015;2015:934058.

    PubMed  PubMed Central  Google Scholar 

  70. Di Cesare ML, Tenci B, Zanardelli M, Maidecchi A, Lugli A, Mattoli L, Ghelardini C. Widespread pain reliever profile of a flower extract of Tanacetum parthenium. Phytomedicine. 2015;22(7-8):752–8.

    Google Scholar 

  71. Zhao ZJ, Xiang JY, Liu L, Huang XL, Gan HT. Parthenolide, an inhibitor of the nuclear factor-kappaB pathway, ameliorates dextran sulfate sodium-induced colitis in mice. Int Immunopharmacol. 2012;12(1):169–74.

    CAS  PubMed  Google Scholar 

  72. Saadane A, Masters S, DiDonato J, Li J, Berger M. Parthenolide inhibits IkappaB kinase, NF-kappaB activation, and inflammatory response in cystic fibrosis cells and mice. Am J Respir Cell Mol Biol. 2007;36(6):728–36.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Schinella GR, Giner RM, Recio MC. Mordujovich de Buschiazzo P, Rios JL, Manez S: Anti-inflammatory effects of South American Tanacetum vulgare. J Pharm Pharmacol. 1998;50(9):1069–74.

    CAS  PubMed  Google Scholar 

  74. Nam YJ, da Lee H, Lee MS, Lee CS. Sesquiterpene lactone parthenolide attenuates production of inflammatory mediators by suppressing the Toll-like receptor-4-mediated activation of the Akt, mTOR, and NF-kappaB pathways. Naunyn Schmiedebergs Arch Pharmacol. 2015;388(9):921–30.

    CAS  PubMed  Google Scholar 

  75. Park SJ, Shin HJ, Youn HS. Parthenolide inhibits TRIF-dependent signaling pathway of Toll-like receptors in RAW264.7 macrophages. Mol Cells. 2011;31(3):261–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Magni P, Ruscica M, Dozio E, Rizzi E, Beretta G, Maffei Facino R. Parthenolide inhibits the LPS-induced secretion of IL-6 and TNF-alpha and NF-kappaB nuclear translocation in BV-2 microglia. Phytother Res. 2012;26(9):1405–9.

    CAS  PubMed  Google Scholar 

  77. Hwang D, Fischer NH, Jang BC, Tak H, Kim JK, Lee W. Inhibition of the expression of inducible cyclooxygenase and proinflammatory cytokines by sesquiterpene lactones in macrophages correlates with the inhibition of MAP kinases. Biochem Biophys Res Commun. 1996;226(3):810–8.

    CAS  PubMed  Google Scholar 

  78. Galeotti N, Maidecchi A, Mattoli L, Burico M, Ghelardini C. St. John’s Wort seed and feverfew flower extracts relieve painful diabetic neuropathy in a rat model of diabetes. Fitoterapia. 2014;92:23–33.

    CAS  PubMed  Google Scholar 

  79. Salapovic H, Geier J, Reznicek G. Quantification of Sesquiterpene Lactones in Asteraceae Plant Extracts: Evaluation of their Allergenic Potential. Sci Pharm. 2013;81(3):807–18.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Jovanovic M, Poljacki M, Duran V, Vujanovic L, Sente R, Stojanovic S. Contact allergy to Compositae plants in patients with atopic dermatitis. Med Pregl. 2004;57(5-6):209–18.

    PubMed  Google Scholar 

  81. Uehara A, Akiyama S, Iwashina T. Foliar flavonoids from Tanacetum vulgare var. boreale and their geographical variation. Nat Prod Commun. 2015;10(3):403–5.

    PubMed  Google Scholar 

  82. Piras A, Falconieri D, Bagdonaite E, Maxia A, Goncalves MJ, Cavaleiro C, Salgueiro L, Porcedda S. Chemical composition and antifungal activity of supercritical extract and essential oil of Tanacetum vulgare growing wild in Lithuania. Nat Prod Res. 2014;28(21):1906–9.

    CAS  PubMed  Google Scholar 

  83. Godinho LS, de Carvalho LS A, de Castro CC B, Dias MM, Pinto Pde F, Crotti AE, Pinto PL, de Moraes J, Da Silva Filho AA. Anthelmintic activity of crude extract and essential oil of Tanacetum vulgare (Asteraceae) against adult worms of Schistosoma mansoni. Sci World J. 2014;2014:460342.

    Google Scholar 

  84. Formisano C, Senatore F, Bruno M, Rosselli S, Bellone G, Spadaro V. Essential oil composition of Tanacetum vulgare subsp. siculum (Guss.) Raimondo et Spadaro (Asteraceae) from Sicily. Nat Prod Commun. 2009;4(4):567–70.

    CAS  PubMed  Google Scholar 

  85. Hethelyi E, Tetenyi P, Dabi E, Danos B. The role of mass spectrometry in medicinal plant research. Biomed Environ Mass Spectrom. 1987;14(11):627–32.

    CAS  PubMed  Google Scholar 

  86. Ravindran R, Sharma N, Roy S, Thakur AR, Subhadra G, Sriram K, Devi J, Rajkumar J. Interaction studies of Withania somnifera’s key metabolite Withaferin A with different receptors associated with cardiovascular disease. Curr Comput Aided Drug Des. 2015.

  87. Shukla K, Dikshit P, Shukla R, Sharma S, Gambhir JK. Hypolipidemic and antioxidant activity of aqueous extract of fruit of Withania coagulans (Stocks) Dunal in cholesterol-fed hyperlipidemic rabbit model. Indian J Exp Biol. 2014;52(9):870–5.

    PubMed  Google Scholar 

  88. Khalil MI, Ahmmed I, Ahmed R, Tanvir EM, Afroz R, Paul S, Gan SH, Alam N. Amelioration of Isoproterenol-Induced Oxidative Damage in Rat Myocardium by Withania somnifera Leaf Extract. Biomed Res Int. 2015;2015:624159.

    PubMed  PubMed Central  Google Scholar 

  89. Ahmad H, Khandelwal K, Samuel SS, Tripathi S, Mitra K, Sangwan RS, Shukla R, Dwivedi AK. Neuro-protective potential of a vesicular system of a standardized extract of a new chemotype of Withania somnifera Dunal (NMITLI118RT+) against cerebral stroke in rats. Drug Deliv. 2015;1–12.

  90. Raghavan A, Shah ZA. Withania somnifera Improves Ischemic Stroke Outcomes by Attenuating PARP1-AIF-Mediated Caspase-Independent Apoptosis. Mol Neurobiol. 2015;52(3):1093–105.

    CAS  PubMed  Google Scholar 

  91. Kaur G, Singh N, Samuel SS, Bora HK, Sharma S, Pachauri SD, Dwivedi AK, Siddiqui HH, Hanif K. Withania somnifera shows a protective effect in monocrotaline-induced pulmonary hypertension. Pharm Biol. 2015;53(1):147–57.

    PubMed  Google Scholar 

  92. Gambhir L, Checker R, Sharma D, Thoh M, Patil A, Degani M, Gota V, Sandur SK. Thiol dependent NF-kappaB suppression and inhibition of T-cell mediated adaptive immune responses by a naturally occurring steroidal lactone Withaferin A. Toxicol Appl Pharmacol. 2015;289(2):297–312.

    CAS  PubMed  Google Scholar 

  93. Gao S, Li H, Zhou XQ, You JB, Tu DN, Xia G, Jiang JX, Xin C. Withaferin A attenuates lipopolysaccharide-induced acute lung injury in neonatal rats. Cell Mol Biol (Noisy-le-grand). 2015;61(3):102–6.

    CAS  Google Scholar 

  94. El-Boshy Mel S, Abdalla OM, Risha A, Moustafa F. Effect of Withania somnifera Extracts on Some Selective Biochemical, Hematological, and Immunological Parameters in Guinea Pigs Experimental Infected with E. coli. ISRN Vet Sci. 2013;2013:153427.

    Google Scholar 

  95. Kumar G, Srivastava A, Sharma SK, Rao TD, Gupta YK. Efficacy & safety evaluation of Ayurvedic treatment (Ashwagandha powder & Sidh Makardhwaj) in rheumatoid arthritis patients: a pilot prospective study. Indian J Med Res. 2015;141(1):100–6.

    PubMed  PubMed Central  Google Scholar 

  96. Khan MA, Subramaneyaan M, Arora VK, Banerjee BD, Ahmed RS. Effect of Withania somnifera (Ashwagandha) root extract on amelioration of oxidative stress and autoantibodies production in collagen-induced arthritic rats. J Complement Integr Med. 2015;12(2):117–25.

    PubMed  Google Scholar 

  97. Gupta A, Singh S. Evaluation of anti-inflammatory effect of Withania somnifera root on collagen-induced arthritis in rats. Pharm Biol. 2014;52(3):308–20.

    CAS  PubMed  Google Scholar 

  98. Bisht P, Rawat V. Antibacterial activity of Withania somnifera against Gram-positive isolates from pus samples. Ayu. 2014;35(3):330–2.

    PubMed  PubMed Central  Google Scholar 

  99. Zakir M, Sultan KB, Khan H, Ihsaanullah, Khan MA, Fazal H, Rauf A. Antimicrobial activity of different tea varieties available in Pakistan. Pak J Pharm Sci. 2015;28(6):2091–4.

    PubMed  Google Scholar 

  100. Klancnik A, Piskernik S, Bucar F, Vuckovic D, Mozina SS, Jersek B. Reduction of microbiological risk in minced meat by a combination of natural antimicrobials. J Sci Food Agric. 2014;94(13):2758–65.

    CAS  PubMed  Google Scholar 

  101. Gupta S, Bansal R, Ali J, Gabrani R, Dang S. Development and characterization of polyphenon 60 and caffeine microemulsion for enhanced antibacterial activity. Biomed Res Int. 2014;2014:932017.

    PubMed  PubMed Central  Google Scholar 

  102. Tambekar DH, Khante BS, Chandak BR, Titare AS, Boralkar SS, Aghadte SN. Screening of antibacterial potentials of some medicinal plants from Melghat forest in India. Afr J Tradit Complement Altern Med. 2009;6(3):228–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Ahn J, Grun IU, Mustapha A. Antimicrobial and antioxidant activities of natural extracts in vitro and in ground beef. J Food Prot. 2004;67(1):148–55.

    CAS  PubMed  Google Scholar 

  104. Vandal J, Abou-Zaid MM, Ferroni G, Leduc LG. Antimicrobial activity of natural products from the flora of Northern Ontario. Canada Pharm Biol. 2015;53(6):800–6.

    CAS  PubMed  Google Scholar 

  105. Favela-Hernandez JM, Garcia A, Garza-Gonzalez E, Rivas-Galindo VM, Camacho-Corona MR. Antibacterial and antimycobacterial lignans and flavonoids from Larrea tridentata. Phytother Res. 2012;26(12):1957–60.

    CAS  PubMed  Google Scholar 

  106. Pinho E, Ferreira IC, Barros L, Carvalho AM, Soares G, Henriques M. Antibacterial potential of northeastern Portugal wild plant extracts and respective phenolic compounds. Biomed Res Int. 2014;2014:814590.

    PubMed  PubMed Central  Google Scholar 

  107. Lopez-Nicolas R, Gonzalez-Bermudez CA, Ros-Berruezo G, Frontela-Saseta C. Influence of in vitro gastrointestinal digestion of fruit juices enriched with pine bark extract on intestinal microflora. Food Chem. 2014;157:14–9.

    CAS  PubMed  Google Scholar 

  108. Li Z, Summanen PH, Komoriya T, Henning SM, Lee RP, Carlson E, Heber D, Finegold SM. Pomegranate ellagitannins stimulate growth of gut bacteria in vitro: Implications for prebiotic and metabolic effects. Anaerobe. 2015;34:164–8.

    PubMed  Google Scholar 

  109. Valéria NL, Oliveira-Tintino CD, Santos ES, et al. Antimicrobial and enhancement of the antibiotic activity by phenolic compounds: Gallic acid, caffeic acid and pyrogallol. Microb Pathog. 2016;99:56–61.

Download references

Acknowledgments

We wish to thank undergraduate students for sample prep and diverse project tasks: Ms. Swan and Marquis Cromartie, as well as the STEM Summer Students- Leandria Harvey and Gabrielle Bradley and their mentor Dr. Tiffany Ardley.

Funding

This research was supported by the National Institute of Minority Health and Health Disparities of the National Institutes of Health through Grant Number 8 G12MD007582-28 and Grant Number 1P20 MD006738-01.

Availability of data and materials

Materials and data of this study are available to other researchers upon request.

Authors’ contributions

EAM was responsible for overview, planning, carrying out basic natural product screenings, confirmation of ELISAS, imaging and manuscript preparation. NL, MD, HW and LT were responsible for planning, overseeing and carrying out the microbial HTP screenings, DB, PM and ET were responsible for cytokine profiling and KFAS was in charge of project planning, overview and manuscript preparation. All authors read and approved the final manuscript.

Competing interest

The authors declare that they have no competing interests.

Consent for publication

NA.

Ethics approval and consent to participate

NA.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Karam F. A. Soliman.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mazzio, E.A., Li, N., Bauer, D. et al. Natural product HTP screening for antibacterial (E.coli 0157:H7) and anti-inflammatory agents in (LPS from E. coli O111:B4) activated macrophages and microglial cells; focus on sepsis. BMC Complement Altern Med 16, 467 (2016). https://doi.org/10.1186/s12906-016-1429-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12906-016-1429-x

Keywords