Next Article in Journal
Simulation Study of Rice Cleaning Based on DEM-CFD Coupling Method
Previous Article in Journal
Influence of Nano-Silica/Chitosan Film Coating on the Quality of ‘Tommy Atkins’ Mango
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Association of Tanacetum parthenium and Salix alba Extracts Reduces Cortex Serotonin Turnover, in an Ex Vivo Experimental Model of Migraine

1
Department of Pharmacy, Botanic Garden “Giardino dei Semplici”, “Gabriele d’Annunzio” University, Via dei Vestini 31, 66100 Chieti, Italy
2
Cristalfarma S.R.L., Via San Giuseppe Cottolengo, 20143 Milan, Italy
3
Veridia Italia S.R.L., Via Raiale 285, 65100 Pescara, Italy
4
Biochemistry and Physiology Research Laboratory, Department of Biology, Faculty of Science, Selcuk University, Konya 42130, Turkey
5
Department of Life Sciences and Biotechnology (SVeB), UR7 Terra&Acqua Tech, University of Ferrara, 44121 Ferrara, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Processes 2022, 10(2), 280; https://doi.org/10.3390/pr10020280
Submission received: 28 December 2021 / Revised: 17 January 2022 / Accepted: 27 January 2022 / Published: 30 January 2022

Abstract

:
The mixture of water extracts from Tanacetum parthenium and Salix alba was studied in an ex vivo assessment of neurotoxicity constituted by isolated mouse cortex specimens challenged with K+ 60 mM Krebs–Ringer buffer (neurotoxicity stimulus). The effects of the mixture on lactate dehydrogenase (LDH), nitrite and serotonin levels were investigated. The phytochemical profile of the mixture was also evaluated. A docking approach was conducted to predict, albeit partially, the putative mechanism underlying the observed effects. The extracts displayed a good profile of polyphenolic compounds (22 chromatographic peaks detected), with caftaric acid and epicatechin being the prominent phenols. In isolated cortex, the association of T. parthenium and S. alba extracts was effective in reducing the K+ 60 mM-induced levels of LDH and nitrites, whereas the neurotoxicity stimulus-induced serotonin depletion was prevented by the treatment. Regarding the inhibition of serotonin catabolism, epicatechin (44.65 µg/mg) and caftaric acid (10.51 µg/mg) were putatively the main compounds involved in the inhibition of monoamineoxidase-A, which is known to play a master role in serotonin turnover. Collectively, the results of the present study point to the efficacy of the present extract mixture as an innovative pharmacological tool to prevent the onset of migraine.

1. Introduction

Migraine is one of the most widespread neurovascular disorders, with an incidence which increases during aging, especially in women [1]. Serotonin (5-HT) has long been involved in migraine etiopathogenesis, with clinical data pointing to tight relationships between neurotransmitter activity and migraine attacks [2]. In this context, the activation of 5-HT1B and 5-HT1D receptors could lead to the control of migraine attacks [3]; by contrast, the blockade of the 5-HT2A and 5-HT2C receptors was found effective in the prevention therapy [4]. The involvement of the trigemino-vascular system and neuroinflammation has also been demonstrated in the pathophysiology of migraine [5,6], although the site of origin of migraine is still unsettled. However, cortical spreading depression (CSD), a supraphysiological neurotoxicity stimulus, has been related to the migraine onset. In particular, CSD would represent a possible link between 5-HT depletion and trigeminal nociception [7]. Currently, the pharmacotherapy options for migraine include aborting attack treatments, namely analgesics, nonsteroidal anti-inflammatory drugs, and triptans [8], and preventive medications, including anti-epileptics, anti-depressants and anti-hypertensives [9]. More recently, monoclonal antibody therapy has been demonstrated to be effective and tolerated by migraineurs [10]. Nevertheless, despite there being different and efficacious pharmacological treatments for the management of migraine, the frequent occurrence of side effects is an impulse to search for innovative options with a minor impact, in terms of side effects [11,12]. Specifically, a new perspective is arising from the anti-migraine use of nutraceuticals and botanicals [12]. Among botanicals, a promising approach is represented by the use of extracts from T. parthenium and Salix alba, which are known to possess anti-inflammatory effects [13,14]. T. parthenium has been used with success for preventing migraine attacks in both adult and child patients [15,16], whereas S. alba improved 5-HT level in rat hippocampus after in vivo administration [17]. Both were found effective, as single treatments, to prevent 5-HT turnover, in a preclinical experimental model of CSD [18,19]. The scientific literature has suggested their pharmacological association for migraine prophylaxis [20]. The co-administration of magnesium and coenzyme Q10 would further improve the anti-migraine efficacy of these botanicals [15].
In this context, the aim of the present study was to investigate the potential use of the extract mixture of T. parthenium and S. alba water extracts in an ex vivo model of CSD constituted by isolated mouse cortex specimens exposed to an supraphysiological depolarizing stimulus (K+ 60 mM). The abovementioned ingredients have been reconstituted in water, and their effects on 5-HT turnover and markers of nitrosative stress and tissue damage, namely nitrites and lactate dehydrogenase, were assayed as well [17,18,21]. The tolerability of the formula was also evaluated through validated toxicological models, namely the Artemia salina and Daphnia magna toxicity tests [22]. Finally, phytochemical and in silico analyses were also conducted in order to shed light the mechanism of action underlying the observed bio-pharmacological effects.

2. Results and Discussion

2.1. Phytochemical Analysis

The extract mixture was formerly investigated via qualitative MS analysis (scan mode 100–1200 m/z), for confirming the presence of parthenolide and salicin, which are reference compounds in T. parthenium and S. alba, respectively. The m/z 249.3 and 271.1 were monitored in positive scan mode to identify the parthenolide, and the MS spectrum confirmed the presence of parthenolide, in the mixture (Figure 1). The m/z ratio 285.1 was scanned in negative scan mode for the identification of salicin, whose MS spectrum is shown (Figure 2). The abovementioned m/z ratios were selected according to the MS spectra reported in MassBankEurope databases. Regarding parthenolide, the selection of the m/z 249.3 agrees with the study by Zhao et al. (2016) [23]. In the case of salicin, the m/z ratio was also considered on the basis of the study by Kammerer and colleagues (2005) [24]. These authors observed that ionization-induced salicin derivatives tended partially to decompose to m/z 123, forming its aglycone form, namely saligenin.
The phytochemical composition was also studied through HPLC-DAD-MS, with the aim of elucidating the quali-quantitative profile in phenolic compounds (Table 1), deeply involved in the anti-inflammatory and anti-oxidant effects of herbal extracts, especially from plant materials extracted with polar solvents [25]. Specifically, the quantitative chromatographic analysis, whose experimental conditions are depicted in Table 1, indicated epicatechin (277.89 µg/mL) and caftaric acid (123.93 µg/mL) as the main phenolics (Figure 3). These compounds were also prominent in other plant materials with intriguing anti-oxidant properties, namely grape and pomace [26,27,28]. However, if catechins and caftaric acid were identified in plants belonging to the Tanaceutm taxa [29,30,31], Salix species were described to synthesize catechins and other polyphenolic compounds, including apigenin, luteolin, naringenin and quercetin [32]. A qualitative 1H-NMR analysis was also conducted in the water suppression signal mode [33], and confirmed the presence of phenolics, but also sugars (Figure 4). The presence of sugars is indicated by the chemical shift values, in the range 3–4 ppm. Considering the elevated level (15% w/w) of salicin in the S. alba extract, we cannot exclude that the signals in this chemical shift range could be related, albeit in part, to this glucoside, as also suggested by the 1H-NMR spectrum reported in the PubChem database. However, the content of phenolics is witnessed by the signals at chemical shift values in the range 7–8 [34,35,36]. Nevertheless, future studies with independent analytical methods, such as 1H-NMR over 400 MHz and LC-HR-MS/MS, will improve our knowledge about the phenolic composition of the extract mixture [37]. The phytochemical profile is also consistent with intrinsic antiradical and neuroprotective properties of T. parthenium and S. alba extracts [18,38,39,40]. Moreover, considering the ability of catechins to cross the blood brain barrier [41], it is rational to evaluate the efficacy of herbal extracts based on these plants in reversing the burden of oxidative stress and neuroinflammation which occurs in neurological diseases, such as migraine. Therefore, after the evaluation of the biocompatibility limits with ecotoxicological models, the extract mixture was administered to isolated mouse cortex specimens exposed to a neurotoxicity stimulus (K+ 60 mM), and selected biomarkers, including serotonin, were assayed for evaluating neuroprotective and antimigraine effects.

2.2. Toxicological and Pharmacological Studies

The toxicological properties of the mixture T. parthenium/S. alba (1:2, w/w) were initially assayed through the Artemia salina (brine shrimp) lethality assay, to define the biocompatibility of the product in the concentration range 0.1–20 mg/mL [22]. This test is considered, albeit partially, as a reliable alternative model for predicting cytotoxicity, in eukaryotic cells [42]. Specifically, the mixture yielded a LC50 > 2 mg/mL (Figure 5), which mirrored the toxicity of T. parthenium and S. alba, in the same experimental model [17]. Considering the results of the brine shrimp assay, the mixture was tested at the concentration of 2 mg/mL in the Daphnia magna toxicity test, for evaluating eventual cardiotoxicity effects, in respect to ethanol 10% solution, as representing the reference cardiotoxicity stimulus. At the concentration of 2 mg/mL, the mixture was able to prevent the ethanol-induced reduction of heart rate, in Daphnia magna (Figure 6); thus scaling back its toxicity impact in eukaryotic organisms. This is also consistent with the abovementioned phytochemical profile; the Toxicity Estimation Software Tool (T.E.S.T.) predicted LC50 values > 3 mg/mL for salicin, catechins and caftaric acid, in this experimental model [22]. Nevertheless, a concentration of the mixture at least ten-fold lower (200 µg/mL) was selected for the subsequent evaluation on cortex tissue. This choice is motivated by the high susceptibility of neurons to oxidative and nitrosative stress [42], and also by the intrinsic capability of phenolics to induce mild oxidative stress [43].
Specifically, the mixture (200 µg/mL) was tested in isolated mouse cortex specimens exposed to different concentrations (3, 15, 60 mM) of K+, in the Krebs–Ringer buffer. If K+ 3 and 15 mM are classical basal and depolarizing conditions in neuronal endings, respectively [44], K+ 60 mM represents a supraphysiological depolarizing stimulus, also called CSD and able to induce a massive calcium-independent release, with concomitant increase of neurotransmitter degradation [7]. The CSD is common to different neuroinflammatory conditions, including migraine, and it is characterized by increased oxidative stress and serotonin degradation in the cortex [21]. In the present study, the mixture was found effective in inducing protective effects in mouse cortex. Indeed, the treatment determined a significant reduction of lactate dehydrogenase (LDH) level (Figure 7), a well-known marker of cytotoxicity [45], at all pharmacological conditions; thus suggesting not only the capability to prevent the tissue damage induced by K+ 60 mM, but also an intrinsic biocompatibility when cortex was exposed to physiological K+ 3 and 15 mM. This biocompatibility is consistent with the abovementioned ecotoxicological predictions, and also mirrored by the inhibition of nitrite level (Figure 8), a reliable index of nitric oxide production and nitrosative stress [46], at all considered depolarizing conditions. This indicated not only the capability to contrast the burden of nitrosative stress induced by K+ 60 mM, but also protective effects occurring at physiological conditions. Finally, the mixture was also effective in blunting the K+ 60 mM-induced turnover of 5-HT, measured as 5HIIA/5-HT ratio (Figure 9), which is a reliable marker of MAO-A activity [47]. In this case, the mixture was effective at all depolarizing conditions to which the cortex was exposed. These findings further support the use of food supplements based on T. parthenium and S. alba for contrasting and/or preventing migraine attacks. The blunting effects on nitrite level and serotonin degradation could be due to more than one reason. On the one hand, we have to consider the intrinsic antiradical properties of both T. parthenium and S. alba extracts [17], present in this mixture. On the other hand, we have to highlight how the antiradical properties of herbal extracts are strictly related to enzyme inhibition effects, especially in the case of phenolic compounds [48]. Additionally, catechin has been suggested to act as a MAO-A inhibitor [49], whereas there is still a gap in the scientific literature about the effect of caftaric acid on this enzyme. Therefore, a docking approach was conducted with the aim of exploring putative interactions of caftaric acid with MAO-A, which could be predictive of enzyme inhibition effects [50]. The results of the virtual screening indicated the capability of caftaric acid to interact with the pocket/binding site of the enzyme through non-covalent interactions, including hydrogen, electrostatic and hydrophobic bonds (Figure 10). Specifically, it predicted the ability of caftaric acid to form hydrogen bonds with Ala-111, Phe-112, Thr-204, Pro-114, His-488, and to interact via electrostatic bond with Glu-492. A hydrophobic interaction was predicted with Phe-112, as well. Furthermore, the present and literature data showed micromolar affinities of caftaric acid and epicatechin, respectively, towards the MAO-A enzyme; thus suggesting the occurrence of enzyme inhibition effects by the mixture and underlying the observed reduction of 5-HT turnover. Considering the 5-HT depletion occurring in migraine, the inhibition of 5-HT turnover induced by the mixture could be of relevance for the prevention of attacks, in migraineurs.

3. Materials and Methods

3.1. Extracts

The dried extracts were provided by Cristalfarma S.r.l. (Milan, Italy). T. parthenium water extract (0.5% parthenolide w/w) and S. alba extract (15% salicin w/w) were rehydrated in water via ultrasound-assisted extraction (Trans-sonic T460 ultrasonic bath; Elma, Singen, Germany), as previously reported [51]. In the mixture, T. parthenium water extract and S. alba extracts were in the ratio 1:2 (w/w).

3.2. Qualitative Mass Spectrometer (MS) and High Performance Liquid Chromatography (HPLC) Analysis of Phenolic Compounds

The extract mixture was analyzed qualitatively using an expression compact mass spectrometer (CMS) (Advion, Ithaca, NY, USA) in negative and positive ion mode (m/z scan mode: 100–1200) [52]. Specifically, MS signal identification was realized through comparison with standard solutions and MS spectra present in the recognized Mass Bank Europe database. The mixture was also analyzed for phenol quantitative determination using a reversed-phase HPLC-DAD-MS in gradient elution mode, in agreement with previous studies [18,53]. The details of the analysis are reported in Supplementary Materials (Tables S1 and S2).

3.3. Protonic Magnetic Resonance (1H-NMR) Analysis

The extract mixture was analyzed via protonic magnetic resonance (1H-NMR). The Variant 300 MHz spectrometer in standard proton pulse sequence (s2pul) with pre-saturation was used for the analysis. 60 µL of D2O were added as internal lock to 600 µL of the sample (300 mg/mL). Pre-saturation was carried out with relaxation delay 1 s, pulse 45.0 degrees, acquisition time 1.706 s, width 4803.1 Hz and 128 number of scans; partial water suppression signals occurred around 4.80 ppm.

3.4. Artemia Salina and Daphnia Magna Toxicity Tests

The biocompatibility and cardiotoxicity of the extract mixture were predicted through the Artemia salina and Daphnia magna toxicity assays. For the A. salina assay, the mixture was tested in the concentration range 0.1–20 mg/mL. Data were expressed as LC50 values. In Daphnia magna, the cardiotoxicity was induced by ethanol 10% solution. The details about these tests are fully reported in our previous studies [22,54].

3.5. Ex Vivo CSD Paradigm

Male adult C57BL6 mice (3-month-old, weight 20–25 g, N = 24) were sacrificed by CO2 inhalation. The experimentation was approved by Local Ethical Committee (University “G. d’Annunzio” of Chieti-Pescara) and Italian Health Ministry (Italian Health Ministry Authorization Number: F4738.N.5QP). After the sacrifice, cortex specimens were immediately collected and maintained in a thermostatic shaking bath at 37 °C for 1 h (incubation period), in Krebs–Ringer buffer at different K+ concentrations (3, 15, 60 mM).
Specifically, the different K+ concentrations corresponded to basal, physiological depolarizing and neurotoxicity stimulus, respectively [55]. During the experimental procedure, isolated cortex was treated with the extract mixture (200 μg/mL).

3.6. High Performance Liquid Chromatography (HPLC) Analysis of Serotonin and 5-Hydroxyindolacetic Acid

Cortex serotonin (5-HT) and 5-hydroxyindolacetic acid (5HIIA) were extracted in HClO4 50 mM and their levels in the tissue homogenate were analyzed through an HPLC-EC apparatus, as previously reported [56]. Details are reported in Supplementary Materials.

3.7. In Silico Study

Regarding the docking analysis, the crystal structure of the protein was downloaded from the Protein Data Bank. The PDB code was: 2ZX5 [monoamine oxydase-A: MAO-A]. Putative interactions with caftaric acid were analyzed as previously reported [57]. Details are reported in Supplementary Materials.

3.8. Statistical Analysis

Statistical analysis was performed by GraphPad Prism™ (Version 5.00) software (GraphPad Software, Inc., San Diego, CA, USA). The statistical significance (p < 0.05) was evaluated through analysis of variance (ANOVA) followed by Newman–Keuls comparison multiple test. Regarding the ex vivo studies, the statistical analysis to calculate the number of animals used in each experiment (N = 4 per condition) was performed with the software G*Power (v3.1.9.4). Study potency (1-β) and significance level (α) were 0.8 and 0.05, respectively.

4. Conclusions

The results of the present study pointed to the efficacy of the mixture T. parthenium/S. alba, in an experimental model of migraine. In this regard, the mixture showed a good tolerability in cortex tissue, as evidenced by the reduction of LDH and nitrites, biomarkers of cytotoxicity and nitrosative stress, respectively. These effects could be related, albeit partially, to the prominent phytocompounds, namely salicin, caftaric acid and epicatechin. The phytochemical profile is also consistent with the inhibition of serotonin degradation, following cortex treatment, and the efficacy was demonstrated both in basal and neurotoxicity conditions, thus suggesting its use as both protection and prevention agent, in migraine. Based on the phytochemical composition, the inhibition of serotonin degradation could be related to the putative MAO-A inhibition by epicatechin and caftaric acid. Since MAO-A is deeply involved in 5-HT catabolism, and considering the 5-HT depletion occurring in migraine attacks, the inhibition of MAO-A activity could be of relevance in the management/prevention of migraine attacks. Moreover, our results add to a previous perspective study suggesting the association of T. parthenium and S. alba as an innovative pharmacological tool to prevent the onset of migraine, with an acceptable toxicological profile [19]. However, further studies conducted with alternative experimental methods are needed to confirm efficacy, especially through in vivo models, and to improve our knowledge about the mechanism of action.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/pr10020280/s1, Table S1: MS analysis conditions; Table S2: Gradient Elution Conditions.

Author Contributions

Conceptualization, G.O., C.F. and L.M.; methodology, C.F. and M.T.; software, C.F. and L.M.; validation, C.F. and L.M.; formal analysis, C.F. and G.Z.; investigation, A.C., L.R., S.L., A.A., M.L.L., S.C.D.S., C.G., C.C., M.P. and G.Z.; resources, L.M. and C.F.; data curation, C.F. and L.M.; writing—original draft preparation, C.F.; writing—review and editing, S.C.D.S.; A.A.; C.F. and M.T.; visualization, L.B.; supervision, L.M. and G.O.; project administration, L.M., G.O. and C.F.; funding acquisition, L.M., G.O. and C.F. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from Cristalfarma S.r.l. (Milan, Italy) that funded the Industrial Ph.D. project of Simonetta Cristina Di Simone entitled: ”From the Botanic Garden “Giardino dei Semplici” to modern rational phytotherapy: enhancement of biodiversity and study of plant extracts in complex mixtures for innovative phytotherapic applications”. Academic tutors are Luigi Menghini and Claudio Ferrante.

Institutional Review Board Statement

Animal experimental procedures were approved by the local ethical committee (University “G. d’Annunzio” of Chieti-Pescara) and Italian Health Ministry (Italian Health Ministry authorization N. F4738.N.5QP).

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The present study is part of the third mission activities of the Botanic Garden “Giardino dei Semplici” planned for the 20th anniversary of its establishment.

Conflicts of Interest

The study was funded by Cristalfarma S.r.l.

Sample Availability

Samples of the compounds are not available from the authors.

References

  1. Rajapakse, T.; Davenport, W.J. Phytomedicines in the treatment of Migraine. CNS Drugs 2019, 33, 399–415. [Google Scholar] [CrossRef] [PubMed]
  2. Hering, R.; Glover, V.; Pattichis, K.; Catarci, T.; Steiner, T.J. 5HT in migraine patients with medication-induced headache. Cephalalgia 1993, 13, 410–412. [Google Scholar] [CrossRef] [PubMed]
  3. Lance, J.W. 5-Hydroxytryptamine and its role in migraine. Eur. Neurol. 1991, 31, 279–281. [Google Scholar] [CrossRef] [PubMed]
  4. Massiou, H.; Bousser, M.G. Prophylactic drug treatment of migraine. Rev. Neurol. 2005, 161, 681–684. [Google Scholar] [CrossRef]
  5. Noseda, R.; Burstein, R. Migraine pathophysiology: Anatomy of the trigeminovascular pathway and associated neurological symptoms, cortical spreading depression, sensitization, and modulation of pain. Pain 2013, 154 (Suppl. S1), S44–S53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Edvinsson, L.; Haanes, K.A.; Warfvinge, K.; Krause, D.N. CGRP as the target of new migraine therapies—Successful translation from bench to clinic. Nat. Rev. Neurol. 2018, 14, 338–350. [Google Scholar] [CrossRef] [PubMed]
  7. Supornsilpchai, W.; Sanguanrangsirikul, S.; Maneesri, S.; Srikiatkhachorn, A. Serotonin depletion, cortical spreading depression, and trigeminal nociception. Headache 2006, 46, 34–39. [Google Scholar] [CrossRef] [PubMed]
  8. Lupi, C.; Benemei, S.; Guerzoni, S.; Pellesi, L.; Negro, A. Pharmacokinetics and pharmacodynamics of new acute treatments for migraine. Expert Opin. Drug Metab. Toxicol. 2019, 15, 189–198. [Google Scholar] [CrossRef]
  9. Loder, E.; Rizzoli, P. Pharmacologic prevention of migraine: A narrative review of the state of the art in 2018. Headache 2018, 58 (Suppl. S3), 218–229. [Google Scholar] [CrossRef]
  10. Frediani, F.; D’Arrigo, G.; Galli, A.; Altavilla, R.; Di Fiore, P. Exploring new strategy in erenumab therapy for migraine patients. Neurol. Sci. 2020, 41 (Suppl. S2), 507–508. [Google Scholar] [CrossRef]
  11. Wider, B.; Pittler, M.H.; Ernst, E. Feverfew for preventing migraine. Cochrane Database Syst. Rev. 2015, 4, CD002286. [Google Scholar] [CrossRef]
  12. Sangermani, R.; Boncimino, A. The use of nutraceutics in children’s and adolescent’s headache. Neurol. Sci. 2017, 38 (Suppl. S1), 121–124. [Google Scholar] [CrossRef]
  13. Pareek, A.; Suthar, M.; Rathore, G.S.; Bansal, V. Feverfew (Tanacetum parthenium L.): A systematic review. Pharmacogn. Rev. 2011, 5, 103–110. [Google Scholar] [CrossRef] [Green Version]
  14. Tawfeek, N.; Mahmoud, M.; Hamdan, D.I.; Sobeh, M.; Farrag, N.; Wink, M.; El-Shazly, A.M. Phytochemistry, pharmacology and medicinal uses of plants of the genus salix: An updated review. Front. Pharmacol. 2021, 12, 593856. [Google Scholar] [CrossRef]
  15. Guilbot, A.; Bangratz, M.; Ait Abdellah, S.; Lucas, C. A combination of coenzyme Q10, feverfew and magnesium for migraine prophylaxis: A prospective observational study. BMC Complementary Altern. Med. 2017, 17, 433. [Google Scholar] [CrossRef] [Green Version]
  16. Moscano, F.; Guiducci, M.; Maltoni, L.; Striano, P.; Ledda, M.G.; Zoroddu, F.; Raucci, U.; Villa, M.P.; Parisi, P. An observational study of fixed-dose Tanacetum parthenium nutraceutical preparation for prophylaxis of pediatric headache. Ital. J. Pediatr. 2019, 45, 36. [Google Scholar] [CrossRef]
  17. Ulrich-Merzenich, G.; Kelber, O.; Koptina, A.; Freischmidt, A.; Heilmann, J.; Müller, J.; Zeitler, H.; Seidel, M.F.; Ludwig, M.; Heinrich, E.; et al. Novel neurological and immunological targets for salicylate-based phytopharmaceuticals and for the anti-depressant imipramine. Phytomedicine 2012, 19, 930–939. [Google Scholar] [CrossRef]
  18. Di Giacomo, V.; Ferrante, C.; Ronci, M.; Cataldi, A.; Di Valerio, V.; Rapino, M.; Recinella, L.; Chiavaroli, A.; Leone, S.; Vladimir-Knežević, S.; et al. Multiple pharmacological and toxicological investigations on Tanacetum parthenium and Salix alba extracts: Focus on potential application as anti-migraine agents. Food Chem. Toxicol. 2019, 133, 110783. [Google Scholar] [CrossRef]
  19. Recinella, L.; Chiavaroli, A.; di Giacomo, V.; Antolini, M.D.; Acquaviva, A.; Leone, S.; Brunetti, L.; Menghini, L.; Ak, G.; Zengin, G.; et al. Anti-Inflammatory and neuromodulatory effects induced by tanacetum parthenium water extract: Results from in silico, in vitro and ex vivo studies. Molecules 2021, 26, 22. [Google Scholar] [CrossRef]
  20. Shrivastava, R.; Pechadre, J.C.; John, G.W. Tanacetum parthenium and Salix alba (Mig-RL) combination in migraine prophylaxis: A prospective, open-label study. Clin. Drug Investig. 2006, 26, 287–296. [Google Scholar] [CrossRef]
  21. Orlando, G.; Zengin, G.; Ferrante, C.; Ronci, M.; Recinella, L.; Senkardes, I.; Gevrenova, R.; Zheleva-Dimitrova, D.; Chiavaroli, A.; Leone, S.; et al. Comprehensive chemical profiling and multidirectional biological investigation of two wild anthemis species (Anthemis Tinctoria var. pallida and A. Cretica subsp. Tenuiloba): Focus on neuroprotective effects. Molecules 2019, 24, 2582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Chiavaroli, A.; Balaha, M.; Acquaviva, A.; Ferrante, C.; Cataldi, A.; Menghini, L.; Rapino, M.; Orlando, G.; Brunetti, L.; Leone, S.; et al. Phenolic characterization and neuroprotective properties of grape pomace extracts. Molecules 2021, 26, 6216. [Google Scholar] [CrossRef] [PubMed]
  23. Zhao, A.Q.; Zhao, J.H.; Zhang, S.Q.; Pan, Y.Y.; Huo, X.L. Determination of parthenolide in rat plasma by UPLC-MS/MS and its application to a pharmacokinetic study. J. Pharm. Biomed. Anal. 2016, 119, 99–103. [Google Scholar] [CrossRef] [PubMed]
  24. Kammerer, B.; Kahlich, R.; Biegert, C.; Gleiter, C.H.; Heide, L. HPLC-MS/MS analysis of willow bark extracts contained in pharmaceutical preparations. Phytochem. Anal. 2005, 16, 470–478. [Google Scholar] [CrossRef] [PubMed]
  25. Maier, T.; Sanzenbacher, S.; Kammerer, D.R.; Berardini, N.; Conrad, J.; Beifuss, U.; Carle, R.; Schieber, A. Isolation of hydroxycinnamoyltartaric acids from grape pomace by high-speed counter-current chromatography. J. Chromatogr. A 2006, 1128, 61–67. [Google Scholar] [CrossRef] [PubMed]
  26. Mavrommatis, A.; Giamouri, E.; Myrtsi, E.D.; Evergetis, E.; Filippi, K.; Papapostolou, H.; Koulocheri, S.D.; Zoidis, E.; Pappas, A.C.; Koutinas, A.; et al. Antioxidant Status of Broiler Chickens Fed Diets Supplemented with Vinification By-Products: A Valorization Approach. Antioxidants 2021, 10, 1250. [Google Scholar] [CrossRef]
  27. Gómez-Mejía, E.; Mikkelsen, L.H.; Rosales-Conrado, N.; León-González, M.E.; Madrid, Y.A. Combined approach based on matrix solid-phase dispersion extraction assisted by titanium dioxide nanoparticles and liquid chromatography to determine polyphenols from grape residues. J. Cromatogr. A 2021, 1644, 462128. [Google Scholar] [CrossRef]
  28. Goufo, P.; Singh, R.K.; Cortez, I. A reference list of phenolic compounds (including stilbenes) in Grapevine (Vitis vinifera L.) roots, woods, canes, stems, and leaves. Antioxidants 2020, 9, 398. [Google Scholar] [CrossRef]
  29. Emre, İ. The biochemical content and antioxidant capacities of endemic Tanacetum densum (Lab.) Schultz Bip. subsp. laxum, and Tanacetum densum (Lab.) Schultz Bip. subsp. amani Heywood growing in Turkey. Braz. J. Biol. 2021, 81, 1106–1114. [Google Scholar] [CrossRef]
  30. Muresan, M.; Benedec, D.; Vlase, L.; Oprean, R.; Toiu, A.; Oniga, I. Screening of polyphenolic compounds, antioxidant and antimicrobial properties of Tanacetum vulgare from Transylvania. Studia UBB Chem. 2015, 1, 127–138. [Google Scholar]
  31. Wu, C.; Chen, F.; Wang, X.; Wu, Y.; Dong, M.; He, G.; Galyean, R.D.; He, L.; Huang, G. Identification of antioxidant phenolic compounds in feverfew (Tanacetum parthenium) by HPLC-ESI-MS/MS and NMR. Phytochem. Anal. 2007, 18, 401–410. [Google Scholar] [CrossRef]
  32. Budny, M.; Zalewski, K.; Stolarski, M.J.; Wiczkowski, W.; Okorski, A.; Stryiński, R. The phenolic compounds in the young shoots of selected willow cultivars as a determinant of the plants’ attractiveness to cervids (Cervidae, Mammalia). Biology 2021, 10, 612. [Google Scholar] [CrossRef]
  33. Politi, M.; Zloh, M.; Pintado, M.E.; Castro, P.M.; Heinrich, M.; Prieto, J.M. Direct metabolic fingerprinting of commercial herbal tinctures by nuclear magnetic resonance spectroscopy and mass spectrometry. Phytochem. Anal. 2009, 20, 328–334. [Google Scholar] [CrossRef]
  34. Anastasiadi, M.; Zira, A.; Magiatis, P.; Haroutounian, S.A.; Skaltsounis, A.L.; Mikros, E. 1H NMR-based metabonomics for the classification of Greek wines according to variety, region, and vintage. Comparison with HPLC data. J. Agric. Food Chem. 2009, 57, 11067–11074. [Google Scholar] [CrossRef]
  35. Boffo, E.F.; Tavares, L.A.; Tobias, A.C.; Ferreira, M.M.; Ferreira, A.G. Identification of components of Brazilian honey by 1H NMR and classification of its botanical origin by chemometric methods. LWT 2012, 49, 55–63. [Google Scholar] [CrossRef] [Green Version]
  36. Orlando, G.; Recinella, L.; Chiavaroli, A.; Brunetti, L.; Leone, S.; Carradori, S.; Di Simone, S.; Ciferri, M.C.; Zengin, G.; Ak, G.; et al. Water extract from inflorescences of industrial hemp futura 75 variety as a source of anti-inflammatory, anti-proliferative and antimycotic agents: Results from in silico, in vitro and ex vivo studies. Antioxidants 2020, 9, 437. [Google Scholar] [CrossRef]
  37. Wang, Z.; Hwang, S.H.; Lim, S.S. Comprehensive profiling of minor tyrosinase inhibitors from Gastrodia elata using an off-line hyphenation of ultrafiltration, high-speed countercurrent chromatography, and high-performance liquid chromatography. J. Chromatogr. A 2017, 1529, 63–71. [Google Scholar] [CrossRef]
  38. Hwang, S.H.; Kim, H.-Y.; Guillen Quispe, Y.N.; Wang, Z.; Zuo, G.; Lim, S.S. Aldose reductase, protein glycation inhibitory and antioxidant of peruvian medicinal plants: The case of Tanacetum parthenium L. and its constituents. Molecules 2019, 24, 2010. [Google Scholar] [CrossRef] [Green Version]
  39. Ostolski, M.; Adamczak, M.; Brzozowski, B.; Wiczkowski, W. Antioxidant activity and chemical characteristics of supercritical CO2 and water extracts from willow and poplar. Molecules 2021, 26, 545. [Google Scholar] [CrossRef]
  40. Ramos, P.A.B.; Moreirinha, C.; Silva, S.; Costa, E.M.; Veiga, M.; Coscueta, E.; Santos, S.A.O.; Almeida, A.; Pintado, M.M.; Freire, C.S.R.; et al. The health-promoting potential of Salix spp. Bark polar extracts: Key insights on phenolic composition and in vitro bioactivity and biocompatibility. Antioxidants 2019, 8, 609. [Google Scholar] [CrossRef] [Green Version]
  41. Faria, A.; Pestana, D.; Teixeira, D.; Couraud, P.O.; Romero, I.; Weksler, B.; de Freitas, V.; Mateus, N.; Calhau, C. Insights into the putative catechin and epicatechin transport across blood-brain barrier. Food Funct. 2011, 2, 39–44. [Google Scholar] [CrossRef]
  42. Radulović, N.S.; Genčić, M.S.; Stojanović, N.M.; Randjelović, P.J.; Stojanović-Radić, Z.Z.; Stojiljković, N.I. Toxic essential oils. Part V: Behaviour modulating and toxic properties of thujones and thujone-containing essential oils of Salvia officinalis L.; Artemisia absinthium L.; Thuja occidentalis L. and Tanacetum vulgare L. Food Chem. Toxicol. 2017, 105, 355–369. [Google Scholar] [CrossRef] [PubMed]
  43. Cobley, J.N.; Fiorello, M.L.; Bailey, D.M. 13 reasons why the brain is susceptible to oxidative stress. Redox. Biol. 2018, 15, 490–503. [Google Scholar] [CrossRef]
  44. Halliwell, B.; Clement, M.V.; Ramalingam, J.; Long, L.H. Hydrogen peroxide. Ubiquitous in cell culture and in vivo? IUBMB Life 2000, 50, 251–257. [Google Scholar] [CrossRef] [PubMed]
  45. Cannizzaro, C.; D’Amico, M.; Preziosi, P.; Martire, M. Presynaptic effects of anandamide and WIN55, 212-2 on glutamatergic nerve endings isolated from rat hippocampus. Neurochem. Int. 2006, 48, 159–165. [Google Scholar] [CrossRef] [PubMed]
  46. Larner, S.F.; Wang, J.; Goodman, J.; Altman, M.B.O.; Xin, M.; Wang, K.K.W. In vitro neurotoxicity resulting from exposure of cultured neural cells to several types of nanoparticles. J. Cell Death 2017, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Tsikas, D. Assessment of lipid peroxidation by measuring malondialdehyde (MDA) and relatives in biological samples: Analytical and biological challenges. Anal. Biochem. 2017, 524, 13–30. [Google Scholar] [CrossRef] [PubMed]
  48. Lee, J.J.; Chang, C.K.; Liu, I.M.; Chi, T.C.; Yu, H.J.; Cheng, J.T. Changes in endogenous monoamines in aged rats. Clin. Exp. Pharmacol. Physiol. 2001, 28, 285–289. [Google Scholar] [CrossRef]
  49. Chatatikun, M.; Supjaroen, P.; Promlat, P.; Chantarangkul, C.; Waranuntakul, S.; Nawarat, J.; Tangpong, J.; Chiabchalard, A. Antioxidant and tyrosinase inhibitor properties of an aqueous extract og Garcinia atrovirdis Griff. Ex. T. Anderson fruit pericarps. Pharmacogn. J. 2020, 12, 71–78. [Google Scholar] [CrossRef] [Green Version]
  50. Reinheimer, J.B.; Bressan, G.N.; de Freitas, C.M.; Ceretta, A.P.C.; Krum, B.N.; Nogara, P.A.; Rodrigues, T.; Schwerz, J.P.; da Rocha, J.B.T.; Fachinetto, R. Effects of CATECHIN on reserpine-induced vacuous chewing movements: Behavioral and biochemical analysis. Naunyn-Schmiedeberg. Arch. Pharmacol. 2020, 393, 2439–2452. [Google Scholar] [CrossRef]
  51. Wang, Z.; Hwang, S.H.; Lim, S.S. Characterization of DHDP, a novel aldose reductase inhibitor isolated from Lysimachia christinae. J. Funct. Foods 2017, 37, 241–248. [Google Scholar] [CrossRef]
  52. Menghini, L.; Leporini, L.; Vecchiotti, G.; Locatelli, M.; Carradori, S.; Ferrante, C.; Zengin, G.; Recinella, L.; Chiavaroli, A.; Leone, S.; et al. Crocus sativus L. stigmas and byproducts: Qualitative fingerprint, antioxidant potentials and enzyme inhibitory activities. Food Res. Int. 2018, 109, 91–98. [Google Scholar] [CrossRef]
  53. Ferrante, C.; Chiavaroli, A.; Angelini, P.; Venanzoni, R.; Angeles Flores, G.; Brunetti, L.; Petrucci, M.; Politi, M.; Menghini, L.; Leone, S.; et al. Phenolic content and antimicrobial and anti-inflammatory effects of solidago virga-aurea, phyllanthus niruri, epilobium angustifolium, peumus boldus, and ononis spinosa extracts. Antibiotics 2020, 9, 783. [Google Scholar] [CrossRef]
  54. Orlando, G.; Chiavaroli, A.; Adorisio, S.; Delfino, D.V.; Brunetti, L.; Recinella, L.; Leone, S.; Zengin, G.; Acquaviva, A.; Angelini, P.; et al. Unravelling the phytochemical composition and the pharmacological properties of an optimized extract from the fruit from Prunus mahaleb L.: From traditional liqueur market to the pharmacy shelf. Molecules 2021, 26, 4422. [Google Scholar] [CrossRef]
  55. Raiteri, L.; Stigliani, S.; Zedda, L.; Raiteri, M.; Bonanno, G. Multiple mechanisms of transmitter release evoked by “pathologically” elevated extracellular [K+]: Involvement of transporter reversal and mitochondrial calcium. J. Neurochem. 2002, 80, 706–714. [Google Scholar] [CrossRef]
  56. Orlando, G.; Leone, S.; Ferrante, C.; Chiavaroli, A.; Mollica, A.; Stefanucci, A.; Macedonio, G.; Dimmito, M.P.; Leporini, L.; Menghini, L.; et al. Effects of Kisspeptin-10 on Hypothalamic Neuropeptides and Neurotransmitters Involved in Appetite Control. Molecules 2018, 23, 3071. [Google Scholar] [CrossRef] [Green Version]
  57. Vasileva, L.V.; Savova, M.S.; Amirova, K.M.; Balcheva-Sivenova, Z.; Ferrante, C.; Orlando, G.; Wabitsch, M.; Georgiev, M.I. Caffeic and chlorogenic acids synergistically activate browning program in human adipocytes: Implications of AMPK- and PPAR-mediated pathways. Int. J. Mol. Sci. 2020, 21, 9740. [Google Scholar] [CrossRef]
Figure 1. Qualitative mass spectrometry (MS) analysis conducted on the extract mixture. The MS analysis was conducted in positive ion mode (m/z scan mode: 100–1200), and the m/z ratios of 249.3 and 271.1 were monitored to identify the parthenolide. The present m/z ratios were selected on MassBankEurope database.
Figure 1. Qualitative mass spectrometry (MS) analysis conducted on the extract mixture. The MS analysis was conducted in positive ion mode (m/z scan mode: 100–1200), and the m/z ratios of 249.3 and 271.1 were monitored to identify the parthenolide. The present m/z ratios were selected on MassBankEurope database.
Processes 10 00280 g001
Figure 2. Qualitative mass spectrometry (MS) analysis conducted on the extract mixture. The MS analysis was conducted in negative ion mode (m/z scan mode: 100–1200), and the m/z ratio of 285.1 was monitored to identify the salicin. The present m/z ratio was selected on MassBankEurope database.
Figure 2. Qualitative mass spectrometry (MS) analysis conducted on the extract mixture. The MS analysis was conducted in negative ion mode (m/z scan mode: 100–1200), and the m/z ratio of 285.1 was monitored to identify the salicin. The present m/z ratio was selected on MassBankEurope database.
Processes 10 00280 g002
Figure 3. Chromatographic analysis of phenolic compounds, in the formula (20 mg/mL). Caftaric acid (peak #2; Rt: 9.483 min; Conc.: 123.93 µg/mL) and epicatechin (peak #5; Rt: 11.820 min; Conc.: 277.89 µg/mL) were the prominent compounds.
Figure 3. Chromatographic analysis of phenolic compounds, in the formula (20 mg/mL). Caftaric acid (peak #2; Rt: 9.483 min; Conc.: 123.93 µg/mL) and epicatechin (peak #5; Rt: 11.820 min; Conc.: 277.89 µg/mL) were the prominent compounds.
Processes 10 00280 g003
Figure 4. Qualitative composition of the formula shown by 1H-NMR analysis with suppression of water signal. (A) In the chemical shift range 3–4, the presence of sugar fraction is shown. In the chemical shift range 7–8 (B), the signals of phenolic compounds are shown.
Figure 4. Qualitative composition of the formula shown by 1H-NMR analysis with suppression of water signal. (A) In the chemical shift range 3–4, the presence of sugar fraction is shown. In the chemical shift range 7–8 (B), the signals of phenolic compounds are shown.
Processes 10 00280 g004
Figure 5. Effects induced by the mixture (mg/mL) on the survival of Artemia salina (brine shrimp lethality assay). ANOVA, p < 0.0001; *** p < 0.001, ** p < 0.01 vs. CTR.
Figure 5. Effects induced by the mixture (mg/mL) on the survival of Artemia salina (brine shrimp lethality assay). ANOVA, p < 0.0001; *** p < 0.001, ** p < 0.01 vs. CTR.
Processes 10 00280 g005
Figure 6. Effects induced by the mixture (2 mg/mL) on heart rate in the Daphnia magna toxicity model. ANOVA, p < 0.001; ** p< 0.01 vs. EtOH 10%, this last representing the positive control toxicity group.
Figure 6. Effects induced by the mixture (2 mg/mL) on heart rate in the Daphnia magna toxicity model. ANOVA, p < 0.001; ** p< 0.01 vs. EtOH 10%, this last representing the positive control toxicity group.
Processes 10 00280 g006
Figure 7. Inhibitory effect induced by the mixture (200 µg/mL) on lactate dehydrogenase (LDH) level, in isolated mouse cortex specimens. ANOVA, p < 0.001; ** p < 0.01, * p < 0.05 vs. K+ 60 mM.
Figure 7. Inhibitory effect induced by the mixture (200 µg/mL) on lactate dehydrogenase (LDH) level, in isolated mouse cortex specimens. ANOVA, p < 0.001; ** p < 0.01, * p < 0.05 vs. K+ 60 mM.
Processes 10 00280 g007
Figure 8. Inhibitory effect induced by the mixture (200 µg/mL) on nitrite level, in isolated mouse cortex specimens. ANOVA, p < 0.001; ** p < 0.01, * p < 0.05 vs. K+ 60 mM.
Figure 8. Inhibitory effect induced by the mixture (200 µg/mL) on nitrite level, in isolated mouse cortex specimens. ANOVA, p < 0.001; ** p < 0.01, * p < 0.05 vs. K+ 60 mM.
Processes 10 00280 g008
Figure 9. Inhibitory effect induced by the mixture (200 µg/mL) on serotonin degradation (5HIIA/5-HT), in isolated mouse cortex specimens. ANOVA p < 0.0001, *** p < 0.001, ** p < 0.01, * p < 0.05 vs. K+ 60 mM.
Figure 9. Inhibitory effect induced by the mixture (200 µg/mL) on serotonin degradation (5HIIA/5-HT), in isolated mouse cortex specimens. ANOVA p < 0.0001, *** p < 0.001, ** p < 0.01, * p < 0.05 vs. K+ 60 mM.
Processes 10 00280 g009
Figure 10. Putative interactions between caftaric acid and monoamine oxydase-A (MAO-A; PDB: 2ZX5). Free energy of binding (ΔG) and affinity (Ki) are −7.5 kcal/mol and 3.2 µM, respectively.
Figure 10. Putative interactions between caftaric acid and monoamine oxydase-A (MAO-A; PDB: 2ZX5). Free energy of binding (ΔG) and affinity (Ki) are −7.5 kcal/mol and 3.2 µM, respectively.
Processes 10 00280 g010
Table 1. Wavelengths of quantification, mass to charge (m/z) ratios and retention times related to the investigated phenolic compounds.
Table 1. Wavelengths of quantification, mass to charge (m/z) ratios and retention times related to the investigated phenolic compounds.
Standardm/zWavelengths (nm)Retention Time (min)Quantity
(µg/mL)
1Gallic acid169.12547.7303.26382
2Caftaric acid311.22549.483123.930
3Catechin289.32549.77318.4077
4Chlorogenic acid353.3125410.34028.0985
5Epicatechin289.325411.820277.890
6Caffeic acid179.1625412.9870.897624
7Syringic acid197.1725414.1831.65113
8Syringaldheyde181.1725417.99325.5515
9Chicoric acid473.3725419.9274.56034
10Coumaric acid163.0425420.7432.44522
11Ferulic acid193.125421.4400.538303
12Benzoic acid121.1225424.23715.6476
13Quercetin301.2425429.3600.155763
14Cinnamic acid147.1625429.7704.46950
15Hesperitin609.1925431.4600.101043
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Di Simone, S.C.; Acquaviva, A.; Libero, M.L.; Chiavaroli, A.; Recinella, L.; Leone, S.; Brunetti, L.; Politi, M.; Giannone, C.; Campana, C.; et al. The Association of Tanacetum parthenium and Salix alba Extracts Reduces Cortex Serotonin Turnover, in an Ex Vivo Experimental Model of Migraine. Processes 2022, 10, 280. https://doi.org/10.3390/pr10020280

AMA Style

Di Simone SC, Acquaviva A, Libero ML, Chiavaroli A, Recinella L, Leone S, Brunetti L, Politi M, Giannone C, Campana C, et al. The Association of Tanacetum parthenium and Salix alba Extracts Reduces Cortex Serotonin Turnover, in an Ex Vivo Experimental Model of Migraine. Processes. 2022; 10(2):280. https://doi.org/10.3390/pr10020280

Chicago/Turabian Style

Di Simone, Simonetta Cristina, Alessandra Acquaviva, Maria Loreta Libero, Annalisa Chiavaroli, Lucia Recinella, Sheila Leone, Luigi Brunetti, Matteo Politi, Claudia Giannone, Claudia Campana, and et al. 2022. "The Association of Tanacetum parthenium and Salix alba Extracts Reduces Cortex Serotonin Turnover, in an Ex Vivo Experimental Model of Migraine" Processes 10, no. 2: 280. https://doi.org/10.3390/pr10020280

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop