Skip to main content
Avicenna Journal of Phytomedicine logoLink to Avicenna Journal of Phytomedicine
. 2017 Jul-Aug;7(4):334–344.

Hydro-alcoholic extract of Matricaria recutita exhibited dual anti-spasmodic effect via modulation of Ca2+ channels, NO and PKA2-kinase pathway in rabbit jejunum

Hamidreza yazdi 1,#, Akhtar Seifi 1,#, Shima Changizi 1, Vahid Khori 1,*, Fatemeh Hossini 1, Ali Davarian 1, Yahya Jand 1, Ayesheh Enayati 1, Masumeh Mazandarani 2, Fateme Nanvabashi 1
PMCID: PMC5580871  PMID: 28884083

Abstract

Objective:

Several studies have shown the antispasmodic activity of Matricaria recutita without detailing the underlying mechanism(s). The present study was designed to determine whether the antispasmodic mechanisms of M. recutita extract mediated via histaminergic/cholinergic receptors, Ca2+channels, activation of PKA2 and NO release in isolated rabbit jejunum.

Materials and Methods:

The concentration- dependent (3 × 10-3–1.3 × 10-2 mg/ml) antispasmodic effect of the hydro-alcoholic extract of M. recutita flowers was studied in isolated rabbit jejunum. The isolated jejunum preparations were divided into seven groups, including the pharmacological probes that modulate cholinergic, histaminergic, and nitrergic receptors, as well as PKA2.

Results:

M. recutita inhibited spontaneous smooth muscle contractility of the jejunum in a concentration-dependent manner (3 × 10-3–1.3 × 10-2 mg/ml) and reduced both K+- and Ca2+-induced contractions, which is similar to the effect of verapamil. The antispasmodic effect of M. recutita was inhibited by H89 (a PKA2 inhibitor). The myorelaxant effect of M. recutita increased in the presence of ACh/His and H89.

Conclusion:

M. recutita evoked antispasmodic and spasmolytic effects mediated through different signaling pathways. Our results have shown this dual inhibitory effect is mediated by blocking Ca2+ channels, activating His and ACh receptors, releasing NO, and activating PKA2.

Key Words: Matricaria, Cholinergic Agents, Histamine Agents, Jejunum, Rabbit, Nitric Oxide

Introduction

Matricaria recutita (German chamomile, Asteraceae Matricaria chamomilla L. var. recutita) is one of the most abundant annual medicinal herbs in the world. It has been used as an anti-inflammatory drug, an analgesic for gastrointestinal pain, as well as an antiseptic, an anti-microbial, and a sedative remedy in traditional medicine in Europe and Asia (McKay, et al., 2006). Clinical and animal studies have shown that M. recutita extract is effective as a smooth muscle relaxant (Ammon, et al., 2006; mazandarani, et al., 2011). It contains potentially active chemical constituents such as terpenoids, flavonoids, coumarins, and spiroethers. However, chamomile’s main active constituents are chamazulene, apigenin and bisabolol. It seems that flavonoids are the most potent antispasmodic products of M. recutita, while its other important components such as bisabolol, chamazulene, and α–bisabolol have anti-inflammatory effects (Abad, et al., 2011).

Flavonoids are not produced by the human body and, generally, daily diet is the main available source for them. Different disciplines such as chemistry, biochemistry and pharmacy have provided evidence of the vital role of flavonoids (Peng, et al., 2003).

Forster and Achterrath-Tuckermann studied the antispasmodic effects of M. recutita and its flavonoids in guinea-pig ileum. They found that the antispasmodic effect of M. recutita is 91% greater than that of papaverine, a myorelaxant alkaloid from the poppy plant (Achterrath-Tuckermann, et al., 1980; Forster, et al., 1980). Apigenin is a major flavonoid found in M. recutita and it is recognized for its antiphlogistic, antispasmodic and antibacterial effects (Guenwaldr, et al., 1998; Murti, et al., 2012). In terms of gastrointestinal effects, flavonoids decrease smooth muscle contractions, possibly by interfering with calcium movement through cell membranes (Di Carlo, et al., 1999).

Among numerous studies on the antispasmodic mechanism of M. recutita, few have considered downstream signaling cascade of this plant, especially NO and PKA2 pathways. In the present study, we hypothesized that M. recutita induces its anti-spasmodic effect via several signaling cascades including Ca2+ channel blockade, NO release, ACh/His receptors, and PKA2 activation. Here, we investigated 1) the concentration-dependent anti-spasmodic effect of M. recutita in the presence of contractions induced by K+ and Ca2+, 2) anti-spasmodic mechanism of M. recutita in the presence of the ACh/His blocker, and NOS and PKA2 inhibitors.

Materials and Methods

Drugs

Acetylcholine chloride (ACh), atropine sulfate, histamine dihydrochloride (His), cetirizine, L-NAME, l-arginine, potassium chloride, calcium chloride, verapamil hydrochloride, and H-89 were obtained from Sigma Chemicals Company, St. Louis, USA. All drugs were dissolved in distilled water and diluted in fresh Tyrode’s solution.

The hydro-alcoholic extract of M. recutita was obtained from Niak Pharmaceutical Company, Gorgan, Iran.

Plant material and phytochemical analysis of extract

All phytochemical procedures were performed as previously described (mazandarani et al., 2011). Fresh M. recutita flowers were harvested from a farm by Niak Pharmaceutical Company (Golestan province, Iran) in May 2014. The plant was identified in the herbarium of Azad University, Gorgan, Iran (RCMP-0100 code). M. recutita flowers were immediately separated, dried in an oven at 40 ºC, milled to a coarse powder (Mesh. 100) and extracted by the maceration method over 6 days using ethanol 70%. This hydro-alcoholic extract was then filtered and dried in vacuum evaporator (at 40–50 ºC). The percentage dried weight of the extract was 30.80% (Mohammedi, et al., 2011).

To analyze the phenolic and flavonoid contents of the extract, a 2.5 g sample of the dried extract was used. For each treatment, analyses were performed in triplicate.

Determination of total phenolic and flavonoid content

The total phenolic content of the extract was determined by the Folin–Ciocalteu method. Briefly, 0.2 ml of the extract was mixed with 0.5 ml of Folin–Ciocalteu reagent, 1.5 ml of 20% Na2CO3 (Merck, Germany) solution, and 7.8 ml of distilled water. The sample was left for 2 hr at room temperature and its absorbance at 765 nm was determined using a UV-visible spectrophotometer (Shimadzu, Japan). The result was expressed in terms of gallic acid (Merck, Germany) equivalent mg/g dry weight of the extract. The amount of flavonoids was determined with the colorimetric method by using aluminum chloride hexahydrate (AlCl3.6H2O; Merck, Germany). Absorbance at 395 nm was read using a UV-vis spectrophotometer (Shimadzu 1601). The result was expressed in terms of apigenin (Merck, Germany) equivalent mg/g dry weight of the extract. The total phenolic and flavonoid contents of the extract were 17.2 ± 1.3% and 5.6 ± 0.4%, respectively.

Animals

In this study, we used male New Zealand white rabbits weighing 1.8–2.5 kg (Pasteur Institute of Iran, Iran). These rabbits were housed in the animal house of Golestan University of Medical Sciences, Iran, under standard laboratory conditions and provided with animal food pellets and free access to water. However, food supply was ceased 12 hr prior to experimentation. The study was approved by the Institutional Animal Ethics Committee and carried out according to Golestan University of Medical Sciences’ guidelines for the use and care of laboratory animals.

Isolated tissue preparations

The anti-spasmodic activities of M. recutita were studied using isolated tissue from rabbit jejunum. The experiments were performed using jejunum sections taken from five rabbits. The rabbits were anaesthetized with sodium pentobarbital (40 mg/kg), and heparin (200 IU/kg) was used as an anticoagulant. The abdomen was exposed and opened. Several pieces of 2cm-long of jejunum were taken, cleaned, and transferred into a Petri dish containing Tyrode’s solution, where it was detached from the mesenteric attachments. Each jejunum segment was mounted vertically (with open lumen) under a resting tension of 0–20 g in 25 mL tissue baths containing Tyrode’s solution, connected to an isometric force transducer (N.TRI 202P, AD Instruments Pty Ltd., Sydney, Australia), and allowed to equilibrate for 20 min. The tissue-bathing medium was maintained at 37 °C (pH 7.4), and gassed with 95% oxygen and 5% carbon dioxide. The composition (mM) of the Tyrode’s solution used herein was as follows: NaCl (136.9 mM), KCl (2.7 mM), CaCl2 (1.8 mM), MgCl2 (1 mM), NaHCO3 (11.9 mM), NaH2PO4 (0.42 mM), and glucose (5.5 mM). For determination of Ca2+ channel blocking (CCB) activity, K+ (50 mM) was used to induce stable contraction of the preparations, as previously described (Farre, et al., 1991). When stable and sustained contraction was achieved, verapamil was added cumulatively to obtain the concentration-dependent inhibitory response (Van Rossum, 1963). To confirm the Ca2+ antagonist activity of the extract, the tissue was allowed to stabilize in normal Tyrode’s solution, which was then washed out with Ca2+-free Tyrode’s solution containing EDTA (0.1 mM) for 20 min. This solution was then replaced with K+-rich and Ca2+-free Tyrode’s solution of the following composition: KCl 50 mM, NaCl 91.04 mM, MgCl2 1.05 mM, NaHCO3 11.90 mM, NaH2PO4 0.42 mM, glucose 5.55 mM, and EDTA 0.1 mM. Following incubation for 20 min, concentration-response curves (CRCs) control of Ca2+ was obtained. When the CRCs of Ca2+ were found to be superimposable (usually after two cycles), the tissue was pretreated with the extract for 30 min to test the CCB effect. The CRCs of Ca2+ were reconstructed at different concentrations of M. recutita and other drugs.

Statistical analysis

All results are presented as mean ± SEM. All data were tested for normality before applying the statistical tests. Two groups of experimental data were compared using Student’s t-test. Concentration-dependent changes were examined using repeated-measures analysis of variance (ANOVA) and Scheffe contrasts. Changes in force were presented as a percentage of the maximal response induced by agonists (Acetylcholine, Histamine, KCL, and Ca2+). The EC50 values were obtained from cumulative concentration-effect curves via non-linear regression analysis of data points from individual concentration-effect curves plotted using GraphPad Prism 5 (GraphPad Software, Inc., CA, USA). A probability of 5% or lower was considered statistically significant.

Results

Phytochemical analysis of the flavonoid extract of M. recutita indicated the presence of quercetin, apigenin, luteolin, and patuletin. In the concentration-dependent model, the hydro-alcoholic extracts of M. recutita (3 × 10-3–13 × 10-3 mg/ml) inhibited spontaneous jejunum contractions, with an EC50 of 6.3 × 10–3 mg/ml (n = 5) (Figure 1). K+ (50 mM)-induced contractions of jejunum smooth muscle were inhibited by the M. recutita extract (EC50 = 6.5 × 10-3 mg/ml; n = 5) (Figure 2). M. recutita had nearly the same EC50 in the presence or absence of smooth muscle contraction (EC50 = 6.3 × 10-3 mg/ml).

Figure 1.

Figure 1

Concentration-dependent effects of M. recutita on the spontaneous contractions in isolated rabbit jejunum preparation (are presented as mean ±SEM, n=5). Mat: Matricaria recutita

Figure 2.

Figure 2

Inhibitory effect of M. recutita on spontaneous and high K+-induced contractions in isolated rabbit jejunum preparations. (Data are presented as mean±SEM, n=5

To determine whether the mechanism of antispasmodic activity of M. recutita was mediated through calcium channel blockade, jejunum was stabilized in a Ca2+-free Tyrode’s solution to inhibit smooth muscle contractions. Then, Ca2+ was added cumulatively (3 × 10-3–30 mM) in the presence and absence of M. recutita (8 × 10-3–1 × 10-2 mg/ml), wherein the extract caused a rightward shift in the Ca2+ concentration–response curves with EC50 = 3 × 10-3 and 7 × 10-3 (mM), respectively (Figure 3a). The inhibitory actions of the total extract and the flavonoid fraction were similar to that of verapamil (1 - 10 µM) with EC50 = 2.62 × 10-5 and 5.54 × 10-5 µM, respectively (n = 5), (Figures 3b, c and d).

Figure 3.

Figure 3

(a) and (b) show the concentration–response control curves of Ca2+ in the absence and presence of increasing concentrations of M. recutita and verapamil, respectively, constructed in Ca2+-free medium. (c) and (d) show the anti-spasmodic activity of M. recutita and flavonoids, respectively, in the presence of verapamil (Data are presented as mean ±SEM, n=5

Acetylcholine (ACh) caused a rapid increase in contractions at concentrations of 1–50 µM (EC50 = 5.9 × 10-6 µM). Pretreatment of the intestinal tissue with M. recutita extract (0.01 mg/ml) reversed the tissue’s response to ACh with EC50 = 1.8 × 10-6 mg/ml, where ACh inhibited basal jejunum contractions in a concentration-dependent manner (Figure 4a). Atropine (3µM) prevented the inhibitory effects of M. recutita in the presence of ACh (Figure 4b). The inhibitory effect of M. recutita in the presence of ACh was not observed in the presence of L-NAME, nadolol (EC50 = 4.42 × 10-6 mg/ml) (n = 5) (Figures 4c and d).

Figure 4.

Figure 4

(a) Comparison of dose–response curves of M. recutita in the presence of acetylcholine-induced contraction of isolated rabbit jejunum, (b) shows depressing effect of L-NAME on M.recutita in the presence of acetylcholine, (c) and (d) show effect of M.recutita extract in the presence of atropine and nadolol respectively ( Data are presented as mean ±SEM, n = 5

Histamine (0.2–200 µM) increased the basal contractions of smooth muscle in a concentration-dependent manner. M. recutita (0.01 mg/ml) caused a downward shift of the dose-response curve of histamine (Figure 5a). Similarly, the inhibitory effects of M. recutita on smooth muscle contraction were prevented by cetirizine (3 µM) (Figure 5b). Pretreatment of the extract with L-NAME abolished the inhibitory response of M. recutita (0.01 mg/ml) in the presence of histamine (EC50 = 1.19 × 10-6 mg/ml) (Figures 5c and d) (n = 5).

Figure 5.

Figure 5

a) and (b) show the effect of M. recutita on histamine-induced contraction of isolated rabbit jejunum in the absence and presence of L-NAME, respectively. (c) and (d) show the effect of M.recutita extract with cetirizine in the absence and presence of L-NAME, respectively (Data are presented as mean ±SEM, n = 5

L-arginine (100 mM) significantly reduced the antispasmodic effect of M. recutita. The inhibitory effect of M. recutita (3 × 10-3–13 × 10-3 mg/ml) on the intestinal smooth muscle was abrogated in the presence of L-name and L-arginine (EC50 = 1.99 × 10-8 mg/ml and 388.9 mg/ml), respectively (n = 5). (Figures 4c and d and Figures 5c and d)

In particular, to determine the possible involvement of PKA2 and NO in the anti-contractility effects of M. recutita, we used H89, a specific inhibitor of PKA2. Applying H89 (1 µM) in the presence of M. recutita increased tissue relaxation by about 53%. Furthermore, addition of histamine increased the inhibitory activity of the extract by about 38%, whereas ACh and L-arginine reduced its antispasmodic activity by 16% and 10%, respectively (Figures 4c and d and Figures 5c and d).

Discussion

In the present study, we demonstrated the anti-spasmodic and spasmolytic (relaxant) effects of the flavonoid components of M. recutita (3 × 10-3–1.3×10-2 mg/ml) in isolated rabbit jejunum. M. recutita evoked a spasmolytic effect on K+-induced spontaneous contractions, suggesting that calcium channels are involved in its spasmolytic actions. This effect was abrogated by L-NAME and amplified by H89. Consistent with this, the spasmolytic effect of M. recutita was mediated through the NO/PKA2 pathway. Our study showed that the spasmolytic effect of M. recutita was mediated through NO-activated cholinergic and histaminergic receptors.

In the present study, M. recutita prevented contractions induced by high potassium concentration, suggesting the possible role of voltage-dependent Ca2+ channels in its mechanism of action. It is well-documented that KCl induces smooth muscle contractions by activating calcium channels (Cortes, et al., 2006). Therefore, this data indicated the indirect Ca2+ channel blocking activity of M. recutita and its flavonoid fraction. Consistent with this, several studies have shown the potential Ca2+ channel blocking activity of M. recutita extract (Achterrath-Tuckermann et al., 1980; Forster et al., 1980; Rotondo, et al., 2009). In contrast, verapamil did not prevent the antispasmodic activity of M. recutita, suggesting that the M. recutita extract has a wide range of biological activity. M. recutita at low doses could augment the effects of verapamil and this is irrespective to indirect reaction with Ca2+channels.

Moreover, atropine and cetirizine blocked the spasmolytic effect of M. recutita, which probably shows a direct receptor-mediated effect of M. recutita. This possibly indicates direct mediatory roles of ACh/His receptors in the mechanism of action of M. recutita. In addition, several studies have proved the direct role of ACh and His in the muscle relaxant activity of M. recutita (Hagelauer, et al., 2006; Heinle, et al., 2006). Taken together, M. recutita-induced muscle relaxation, was partly mediated through ACh and His receptors.

Our data have shown that an NOS inhibitor (L-NAME) abrogated the relaxing effect of M. recutita. Along the lines of our results, Achterrath-Tuckermann et al. reported that M. recutita induced a greater anti-contractility effect in the presence of NO donors (Achterrath-Tuckermann et al., 1980). We hypothesized that the muscle relaxant effect of M. recutita is mediated by the release NO and activation of ACh/His receptors. Indeed, a study has demonstrated that ACh/His-induced smooth muscle contraction is mediated by a NO-dependent pathway (Grifoni, et al., 2000). Taken together, in the present study, M. recutita-induced smooth muscle relaxant activity was mediated via NO release and ACh/His receptors. Precise muscle relaxant signaling mechanism of M. recutita is yet to be determined.

Furthermore, our findings showed that the inhibitory effect of M. recutita on jejunum contraction is abrogated by L-arginine, due to an unknown mechanism. Applying of high concentrations of L-arginine can induce acidosis and this indirectly prevents the M. recutita action. In a separate group of the experiment, we adjusted the pH of medium and interestingly L-arginine showed the previous profile of action. Consistent with this data, few studies have shown that the inhibitory effect of M. recutita on the contractile tone of smooth muscle was restored by L-arginine, indicating a controversial dual role of NO as an inhibitor or a stimulator (Michel, et al., 2011; Westfall, et al., 2011). Consistent with this, it can be concluded that the controversial dual role of NO in the signaling cascades of M. recutita on intestinal motility depends on the basal contractility state of smooth muscle, and it is observable as a spasmolytic effect on contracting smooth muscle and a tonicising effect on relaxed muscle. Ammon et al indicated the dual activity of STW5 in both hypotonic and spastic states of isolated guinea pig ileum(Ammon et al., 2006). Taken together, NO and ACh/His receptors involved in the signaling cascades of M. recutita intestinal smooth muscle contractility

A previous study demonstrated that NO-evoked smooth muscle relaxation was induced by increasing cGMP and PKA2 (Hobbs, et al., 1996). To determine the possible role of PKA2 modulation in NO-induced smooth muscle relaxation, we analyzed the dual anti-spasmodic effects of M. recutita in the presence of a selective PKA2 inhibitor.

Our results showed that the effects of M. recutita were mediated directly or indirectly via PKA2. In fact, the inhibitory effects of M. recutita on jejunum contractions were augmented by H-89, indicating that the inhibitory effect of M. recutita was mediated through PKA2 activation. In agreement with our data, a study has shown that the spasmolytic effect of M. recutita is mediated by the inhibition of cAMP phosphodiesterase (Maschi, et al., 2008). In addition, the muscle relaxant function of GI smooth muscle tone was regulated by cAMP-PDE pathway and the flavonoid fraction of M. recutita inhibits cAMP activity (Murthy, 2006). Therefore, our data indicated that the PKA2-cAMP transduction pathway was involved in the inhibitory effect of M. recutita on smooth muscle contraction.

Consistent with our results, it has been proven that the gastrointestinal (GI) smooth muscles are synchronized by different mechanisms including the histaminergic, cholinergic, nitrergic pathways to stimulate PKA2 signaling cascade (Kochar, et al., 2011; Murthy, 2006). Moreover, PKA acts as a downstream and upstream signaling cascade to induce relaxation via several mechanisms, in smooth muscle cells.

As noted above, PKA and PKG act on upstream targets to regulate cAMP and cGMP levels by desensitizing receptors and stimulating PDE activities (Murthy, 2006). Moreover, a study demonstrated that the activation of ACh and His receptors is abolished by PKC inhibitors (Eto, et al., 2001).

In line with the aforementioned data, in our study, H89 potentiated smooth muscle relaxation induced by M. recutita, and this effect was decreased in the presence of ACh and L-arginine.

Therefore, we suggest that specific interplay among different signaling cascades including PKA2, ACh, His, and adrenergic receptors, as well as the nitrergic pathway, is required to mediate the antispasmodic effect of M. recutita in rabbit intestinal smooth muscle.

In this study, we were not able to determine the sequential pathway of NO and PKA2 activation in mediating the antispasmodic effects of M. recutita following the stimulation of ACh/His receptors. However, future studies should be conducted to provide sufficient evidence related to the downstream signaling pathways of M. recutita and focus on its possible link to ACh/His-receptors and the NO pathway.

In the present study, the concentration–response curves of M. recutita and its pure flavonoid fraction exhibited similar spasmolytic activity without significant changes in the EC50 values. Hence, the fundamental role of flavonoids in the anti-spasmodic effects of M. recutita was confirmed. Indeed, the total flavonoid fraction of M. recutita is used to prevent interfering actions of essential oils. Our previous study showed high amounts of apigenin and quercetin in the hydro-alcoholic extract of M. recutita (mazandarani et al., 2011). Thus, we suggest that the flavonoid fraction of M. recutita actively induces anti-spasmodic action. Moreover, a growing body of evidence suggests that apigenin and quercetin exhibit smooth muscle anti-spasmodic activities through a wide range of signaling cascades (Amira, et al., 2008; Gharzouli, et al., 2004). Consistent with our results, the myorelaxant activity of apigenin has been attributed to the inhibition of Ca2+ release or induction of eNOS and NO production in mouse gastric tissue (Chen, et al., 2010). In addition, Rotondo et al., showed significant NO production in response to an increase in intracellular calcium concentration by apigenin- and quercetin-induced gastric relaxation (Rotondo et al., 2009). Thus, we suggest that the total flavonoid fraction of M. recutita (apigenin, quercetin, etc.) mediates its inhibitory effect on smooth muscle contractions through blockade of calcium release and NO production.

The myorelaxant activity M. recutita is observed as dual anti-spasmodic and spasmolytic effects. Considering the fundamental role of Ca2+ channels as a possible end-effector step in the mechanism of action of M. recutita, we suggest that NO, His/ACh/ adrenergic receptors, and PKA2 are the main signaling components of M. recutita in the presence of spasmodic agents. Nevertheless, without analyzing the influence of M. recutita in the presence of several inhibitors or detecting direct kinase activity assay, we cannot determine whether the interactions among multiple mediators take place in parallel or in sequence. Meanwhile, further study is required to elucidate the mechanism of action of the various flavonoid components of M. recutita and the contributions of various signaling processes in the curative properties of this plant when uses in the treatment of gastrointestinal motility disorders.

Acknowledgment

The study was financially supported by Golestan University of Medical Sciences and authors thank for an unrestricted educational grant in support of this work, also authors thank Niak Pharmaceutical Company, Gorgan, Iran for providing of the extract.

Conflicts of interest

The authors declare no conflict of interest.

References

  1. Abad A, Nouri M, Gharjanie A, Tavakoli F. Effect of Matricaria chamomilla Hydroalcoholic Extract on Cisplatin-induced Neuropathy in Mice. Chin J Nat Meds. 2011;9:126–131. [Google Scholar]
  2. Achterrath-Tuckermann U, Kunde R, Flaskamp E, Isaac O, Thiemer K. Pharmacological investigations with compounds of chamomile V Investigations on the spasmolytic effect of compounds of chamomile and Kamillosan on the isolated guinea pig ileum. Planta Med. 1980;39:38–50. doi: 10.1055/s-2008-1074901. [DOI] [PubMed] [Google Scholar]
  3. Amira S, Rotondo A, Mule F. Relaxant effects of flavonoids on the mouse isolated stomach: structure-activity relationships. Eur J Pharmacol. 2008;599:126–130. doi: 10.1016/j.ejphar.2008.09.021. [DOI] [PubMed] [Google Scholar]
  4. Ammon HP, Kelber O, Okpanyi SN. Spasmolytic and tonic effect of Iberogast (STW 5) in intestinal smooth muscle. Phytomedicine. 2006;13 (Suppl 5):67–74. doi: 10.1016/j.phymed.2006.08.004. [DOI] [PubMed] [Google Scholar]
  5. Chen CC, Ke WH, Ceng LH, Hsieh CW, Wung BS. Calcium- and phosphatidylinositol 3-kinase/Akt-dependent activation of endothelial nitric oxide synthase by apigenin. Life Sci. 2010;87:743–749. doi: 10.1016/j.lfs.2010.10.014. [DOI] [PubMed] [Google Scholar]
  6. Cortes AR, Delgadillo AJ, Hurtado M, Dominguez-Ramirez AM, Medina JR, Aoki K. The antispasmodic activity of Buddleja scordioides and Buddleja perfoliata on isolated intestinal preparations. Biol Pharm Bull. 2006;29:1186–1190. doi: 10.1248/bpb.29.1186. [DOI] [PubMed] [Google Scholar]
  7. Di Carlo G, Mascolo N, Izzo AA, Capasso F. Flavonoids: old and new aspects of a class of natural therapeutic drugs. Life Sci. 1999;65:337–353. doi: 10.1016/s0024-3205(99)00120-4. [DOI] [PubMed] [Google Scholar]
  8. Eto M, Kitazawa T, Yazawa M, Mukai H, Ono Y, Brautigan DL. Histamine-induced vasoconstriction involves phosphorylation of a specific inhibitor protein for myosin phosphatase by protein kinase C alpha and delta isoforms. J Biol Chem. 2001;276:29072–29078. doi: 10.1074/jbc.M103206200. [DOI] [PubMed] [Google Scholar]
  9. Farre AJ, Colombo M, Fort M, Gutierrez B. Differential effects of various Ca2+ antagonists. Gen Pharmacol. 1991;22:177–181. doi: 10.1016/0306-3623(91)90331-y. [DOI] [PubMed] [Google Scholar]
  10. Forster HB, Niklas H, Lutz S. Antispasmodic effects of some medicinal plants. Planta Med. 1980;40:309–319. doi: 10.1055/s-2008-1074977. [DOI] [PubMed] [Google Scholar]
  11. Gharzouli K, Holzer P. Inhibition of guinea pig intestinal peristalsis by the flavonoids quercetin, naringenin, apigenin and genistein. Pharmacology. 2004;70:5–14. doi: 10.1159/000074237. [DOI] [PubMed] [Google Scholar]
  12. Grifoni SC, Bendhack LM. Relaxation induced by histamine is not endothelium dependent in tail arteries of L-NAME-treated rats. Gen Pharmacol. 2000;34:435–441. doi: 10.1016/s0306-3623(01)00081-7. [DOI] [PubMed] [Google Scholar]
  13. Guenwaldr J, Brendler T, Jaenicke C. Matricaria Chamomilla (Chamomile) In: M. N.J, editor. PDR for Herbal Medicines. Medical Economics Company; 1998. p. 961. [Google Scholar]
  14. Hagelauer D, Kelber O, Weiser D, Heinle H. Effects of STW 5 (Iberogast®) on prostaglandinF2α –induced contractions of ileum of mice in vitro. Planta Med. 2006;72:280. [Google Scholar]
  15. Heinle H, Hagelauer D, Pascht U, Kelber O, Weiser D. Intestinal spasmolytic effects of STW 5 (Iberogast) and its components. Phytomedicine. 2006;13 (Suppl 5):75–79. doi: 10.1016/j.phymed.2006.03.013. [DOI] [PubMed] [Google Scholar]
  16. Hobbs AJ, Ignarro LJ. Nitric oxide-cyclic GMP signal transduction system. Methods Enzymol. 1996;269:134–148. doi: 10.1016/s0076-6879(96)69016-8. [DOI] [PubMed] [Google Scholar]
  17. Kochar NI, Chandewal AV, Bakal RL, Kochar PN. Nitric oxide and the gastrointestinal tract. Int. J. Pharmacol. 2011;7:31–39. [Google Scholar]
  18. Maschi O, Cero ED, Galli GV, Caruso D, Bosisio E, Dell'Agli M. Inhibition of human cAMP-phosphodiesterase as a mechanism of the spasmolytic effect of Matricaria recutita L. J Agric Food Chem. 2008;56:5015–5020. doi: 10.1021/jf800051n. [DOI] [PubMed] [Google Scholar]
  19. Mazandarani M, hoseini F, Seifi A, Bayat H, Pourabouk M, Badaghabadi F, Rajaei M, Moheimani H, Khori V. Role of Histaminegic and calcium channels in the inhibitory effects of hydroalcoholic extract of Matricaria recutita L on isolated rabbit jejunum. Physiology and Pharmacology. 2011;15:361–370. [Google Scholar]
  20. McKay DL, Blumberg JB. A review of the bioactivity and potential health benefits of chamomile tea (Matricaria recutita L.) Phytother Res. 2006;20:519–530. doi: 10.1002/ptr.1900. [DOI] [PubMed] [Google Scholar]
  21. Michel T, Hoffman B. Treatment of myocardial ischemia and hypertension. In: Brunton, B, Chabner B, editors. Goodman and Gilman's The Pharmacological Basis of Therapeutics. McGraw-Hill Education; 2011. 763 pp. [Google Scholar]
  22. Mohammedi Z, Atik F. Impact of solvent extraction type on total polyphenols content and biological activity from Tamarix aphylla (L) karst. IJPBS. 2011:609–615. [Google Scholar]
  23. Murthy KS. Signaling for contraction and relaxation in smooth muscle of the gut. Annu Rev Physiol. 2006;68:345–374. doi: 10.1146/annurev.physiol.68.040504.094707. [DOI] [PubMed] [Google Scholar]
  24. Murti K, Panchal M, Gajera V, Solanki I. Pharmacological Properties of Matricaria recutita: A Review. Pharmacologia. 2012;3:348–351. [Google Scholar]
  25. Peng ZF, Strack D, Baumert A, Subramaniam R, Goh N K, Chia T F, Tan S N, Chia L S. Antioxidant flavonoids from leaves of Polygonum hydropiper L. Phytochemistry. 2003;62:219–228. doi: 10.1016/s0031-9422(02)00504-6. [DOI] [PubMed] [Google Scholar]
  26. Rotondo A, Serio R, Mule F. Gastric relaxation induced by apigenin and quercetin: analysis of the mechanism of action. Life Sci. 2009;85:85–90. doi: 10.1016/j.lfs.2009.04.022. [DOI] [PubMed] [Google Scholar]
  27. Van Rossum JM. Cumulative dose-response curves II Technique for the making of dose-response curves in isolated organs and the evaluation of drug parameters. Arch Int Pharmacodyn Ther. 1963;143:299–330. [PubMed] [Google Scholar]
  28. Westfall TC, Westfall DP. Neurotransmission: The Autonomic and Somatic Motor Nervous Systems. In: L. Brunton, B. chabner., editors. Goodman & Gilman's The Pharmacological Basis of Therapeutics. McGraw-Hill Education; 2011. p. 181. [Google Scholar]

Articles from Avicenna Journal of Phytomedicine are provided here courtesy of Mashhad University of Medical Sciences

RESOURCES