Next Article in Journal
Extraction of Cathepsin D-Like Protease from Neon Flying Squid (Ommastrephes bartramii) Viscera and Application in Antioxidant Hydrolysate Production
Next Article in Special Issue
The Role of Protein Tyrosine Phosphatase (PTP)-1B in Cardiovascular Disease and Its Interplay with Insulin Resistance
Previous Article in Journal / Special Issue
Promising Directions in Atherosclerosis Treatment Based on Epigenetic Regulation Using MicroRNAs and Long Noncoding RNAs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Marjoram Relaxes Rat Thoracic Aorta Via a PI3-K/eNOS/cGMP Pathway

1
Department of Nutrition, University of Petra, Amman, P.O. Box 961343, Amman 11196, Jordan
2
Department of Biology, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
3
Department of Biomedical Sciences, Lebanese International University, Beirut, P.O. Box 146404 Mazraa, Lebanon
4
Faculty of Public Health IV, Lebanese University, Beirut, P.O. Box 6573/14 Badaro, Lebanon
5
Department of Biomedical Sciences, Qatar University, Doha P.O. Box 2713, Qatar
6
Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
7
Department of Biology, United Arab Emirates University, Al Ain P.O. Box 15551, UAE
8
Department of Pharmacology and Toxicology, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
*
Authors to whom correspondence should be addressed.
Submission received: 15 April 2019 / Revised: 28 May 2019 / Accepted: 29 May 2019 / Published: 11 June 2019

Abstract

:
Despite pharmacotherapeutic advances, cardiovascular disease (CVD) remains the primary cause of global mortality. Alternative approaches, such as herbal medicine, continue to be sought to reduce this burden. Origanum majorana is recognized for many medicinal values, yet its vasculoprotective effects remain poorly investigated. Here, we subjected rat thoracic aortae to increasing doses of an ethanolic extract of Origanum majorana (OME). OME induced relaxation in a dose-dependent manner in endothelium-intact rings. This relaxation was significantly blunted in denuded rings. N(ω)-nitro-l-arginine methyl ester (L-NAME) or 1H-[1,2,4]oxadiazolo[4,3,-a]quinoxalin-1-one (ODQ) significantly reduced the OME-induced vasorelaxation. Cyclic guanosine monophosphate (cGMP) levels were also increased by OME. Moreover, wortmannin or LY294002 significantly reduced OME-induced vasorelaxation. Blockers of ATP-sensitive or Ca2+-activated potassium channels such as glibenclamide or tetraethylamonium (TEA), respectively, did not significantly affect OME-induced relaxation. Similarly, verapamil, a Ca2+ channel blocker, indomethacin, a non-selective cyclooxygenase inhibitor, and pyrilamine, a H1 histamine receptor blocker, did not significantly modulate the observed relaxation. Taken together, our results show that OME induces vasorelaxation via an endothelium-dependent mechanism involving the phosphoinositide 3-kinase (PI3-K)/ endothelial nitric oxide (NO) synthase (eNOS)/cGMP pathway. Our findings further support the medicinal value of marjoram and provide a basis for its beneficial intake. Although consuming marjoram may have an antihypertensive effect, further studies are needed to better determine its effects in different vascular beds.

1. Introduction

Cardiovascular disease (CVD) continues to cause more mortalities than any other disease [1]. Despite major pharmacotherapeutic advances, chronically elevated blood pressure persists as a major contributor to CVD-associated mortality [2]. Although many antihypertensive drugs continue to be developed, these synthetic drugs are not without many deleterious adverse effects. In addition to these synthetic drugs, the use of herbal medicine and plant-based therapies is increasing in many countries [3]. In recent years, more interest has been given to these therapies, largely due to their ease of availability, presumed low toxicity, and documented benefits [4,5,6,7,8]. Indeed, many of these plants have been employed for the particular treatment or management of cardiovascular disease [5,6,7,8,9].
Origanum majorana L. is a perennial subshrub with a distinguished fragrant foliage. It is native to southern Europe and the Mediterranean. It has been traditionally used as a folk medicine for many ailments, such as indigestion, headache and asthma [10,11]. Interestingly, the genus Origanum possesses bountiful biological properties, such as antioxidant, anti-inflammatory and anticholinesterase effects, as well as protection against aging and neurodegenerative disease [12]. In addition, marjoram elicits an in vitro microbicidal activity [13,14]. We have also shown that marjoram inhibits the malignant phenotype of breast cancer cells [15,16]. Relevantly, it has been reported that marjoram is used in the management/treatment of hypertension, although the mechanism of action was not elucidated [17].
Bioactives of marjoram, such as the monoterpene carvacrol, exhibit a potent inhibitory effect against bacterial and fungal infections, as well as antispasmodic effects [13]. Moreover, carvacrol can quench free radicals and ameliorate hypercontraction of aortic vasculature [18]. More importantly, carvacrol is known to play an important role in endothelium-dependent vasorelaxation of rat aortae via a mechanism that apparently involves potassium and calcium channels [19,20]. Similarly, thymol, another monoterpe found in marjoram, appears to induce relaxation of isolated rat aortae [20]. Hesperetin, another bioactive of marjoram [21], reduces systolic blood pressure in spontaneously hypertensive rats via an endothelium-dependent mechanism [22].
Despite these beneficial values of O. majorana or its bioactives, and to the best of our knowledge, no studies have so far investigated the vascular activity caused by O. majorana. Accordingly, the present study was undertaken to investigate and evaluate the potential vasorelaxatory effects of O. majorana on isolated rat aortae.

2. Materials and Methods

2.1. Preparation of the Extract

Origanum majorana, locally grown and commonly available, was collected in August 2014. Dried leaves were ground with a mortar and pestle, and the powder extracted with 70% EtOH for 3 h in a reflux apparatus at 70 ± 5 °C. Following filtration, the extract was collected and evaporated at 60 °C, and then it was lyophilized in a freeze-dryer to obtain a powder of the crude extract. This powder was weighed and kept at −20 °C until further use.

2.2. Animals

The study protocol was approved by the scientific committee on animal care and use in the Faculty of Public Health at the Lebanese University (Permit number for Samaha Marjoram Project: UL/FSPIV/07/2011). Experiments were performed in conformity with the National institutes of health legislation on the use of laboratory animals. Every effort was taken to minimize animal suffering. Male Sprague-Dawley (SD) rats (230–290 g body weight; 6–8 weeks old) were housed in an animal laboratory at an ambient temperature of 23 ± 2 ° C, under a 12 h:12 h dark: light cycle. Animals had access to a standard animal chow and tap water ad libitum.

2.3. Drugs and Chemicals

Acetylcholine, norepinephrine, tetraethylammonium (TEA), 1H-[1,2,4]oxadiazolo[4,3-alpha]quinoxalin-1-one (ODQ), atropine, 3-isobutyl-1-methylxanthine (IBMX), indomethacin, pyrilamine, verapamil, and Nω-nitro-l-arginine methyl ester (L-NAME) were purchased from Sigma-Aldrich Co. (St. Louis, MO, USA). LY294002 and Wortmannin was obtained from Tocris (Abingdon, UK) and Alexis Biochemicals (Lausen, Switzerland), respectively.

2.4. Arterial Ring Preparation and Vascular Reactivity

Male SD rats were euthanized by an overdose of pentobarbital (50 mg/kg body weight). Thoracic aortae were quickly dissected into a cold modified Krebs-Henseleit (KHB) buffer, as in our recent papers [7,8]. Aortae were cleaned of fat and other adhering connective tissues and cut into ring segments, which were suspended for the measurement of isometric force in organ chambers filled with KHB (pH 7.4), maintained at 37 °C, and bubbled with a gas mixture of 95% O2—5% CO2. The KH solution contained (in 10−3 M): NaCl (130), KCl (4.7), CaCl2·2H2O (1.6), Mg2SO4 7H2O (1.17), KH2PO4 (1.18), NaHCO3 (14.9), and glucose (5.5).
During the equilibration period (at least 30 min), the rings underwent isometric resting tension in a stepwise manner, as we recently described [7,8]. Cumulative concentrations (0.01–1 mg/mL,) of O. majorana (OME) were added, where at each dose of OME, the response curve would reach a plateau before the subsequent dose was added. To determine the endothelium-dependent relaxation, aortic rings with denuded endothelium were pre-contracted with norepinephrine (NE; 3 µM) and then exposed to incrementally increased concentrations of OME. As we recently published [8], denudation was done by gently rubbing, in a to and from motion, the lumen of the ring with a roughened piece of stainless wire. Generation of less than 15% relaxation with acetylcholine is considered indicative of a successful denudation.
When assessing the potential role of nitric oxide (NO), endothelium-intact rings were pre-incubated with L-NAME (100 μM), a NO synthase inhibitor, for 30 min. Rings were then contracted with NE, after which OME was added.
When determining the role of prostaglandins, histamine H1-receptors, muscarinic receptors, guanylate cyclase, or potassium channels in OME-induced relaxation, the following inhibitors were used: indomethacin (10 µM), a non-selective cyclooxygenase inhibitor; pyrilamine (10 μM), a histamine H1-receptor antagonist or atropine (10 μM), a muscarinic receptor antagonist; oxadiazole quinoxaline (ODQ, 10 µM), a soluble guanylate cyclase inhibitor; glibenclamide (10 µM), an ATP-sensitive potassium channels blocker; or tetraethylammonium (100 µM), a non-selective inhibitor of Ca2+-activated potassium channels. When the role of calcium channels was to be assessed, and as previously described [7,8,23], rings were pre-incubated with verapamil hydrochloride (1µM), a blocker of L-type calcium channels before the addition of OME.
To assess the potential role of phosphoinositide 3-kinase (PI3K)/Akt signaling, rings were pre-incubated with the PI3K inhibitors, LY294002 (10 µM) or wortmannin (100 nM), for 30 min. A cumulative dose response curve with increasing concentrations of OME followed.
To ensure that OME did not impart a significant irreversible effect on vascular responsiveness, aortic rings that had been exposed to OME were washed and exposed to norepinephrine (NE, 3 μM) or acetylcholine (ACh, 10 μM) anew. Finally, KCl (80 mM) was used to confirm viability of the rings.

2.5. cGMP Assay

Rings that had been pre-equilibrated were incubated in IBMX (a phosphodiesterase inhibitor; 0.1 mM) for 30 min before subsequent addition of NE. Then, these rings were re-equilibrated for an additional 30 min before adding OME. Tissues were then frozen in liquid nitrogen to stop the reaction, homogenized in trichloroacetic acid, and centrifuged at 10,000× g for 10 min. The supernatant was then extracted with water-saturated diethylether. Quantitation of protein or cyclic guanosine monophosphate (cGMP) in the extract was done by Bradford’s method or a specific immunoassay (GE Healthcare Life Sciences), Pittsburgh, PA, USA), respectively. Results are plotted as picomoles of cGMP per milligram of protein.

2.6. Graphing and Statistical Analysis

Graphing and determination of 50% effective dose (pED50) and EMAX, along with the confidence intervals, were performed by GraphPad Prism 6 software (GraphPad Software, San Diego, CA, USA). For statistical analysis, the same software package was used. Data are presented as means ± standard error (SE). Student’s t-test was used. If comparison between more than two means was needed, Analysis of variance (ANOVA) was used—either a one-way ANOVA with Tukey’s post hoc test or a two-way ANOVA with Sidak’s multiple comparison post hoc test. A p-value of less than 0.05 was considered statistically significant.

3. Results

3.1. Effect of Origanum majorana Extract on Norepinephrine-Induced Contraction

We first determined the concentration-dependent vasorelaxatory effects of OME (0.001–1 mg/mL) on endothelium-intact aortic rings. OME caused a dose-dependent relaxation with the maximal relaxant effect being 90 ± 5%, and a pED50 of −3.98, with a 95% confidence interval of −4.0 to −3.9 (Figure 1). The vehicle (ethanol) did not impart any significant effect on the rings.

3.2. Effect of Origanum majorana Extract on Endothelium-Intact or Endothelium-Denuded Aortic Rings Pre-Contracted with Norepinephrine

We next wished to determine the potential role of endothelium in OME-induced relaxation. In both endothelium-intact and endothelium-denuded aortic rings, OME caused a dose-dependent relaxation (Figure 2). The maximal relaxant effect was 95 ± 3% or 46 ± 4.0% for endothelium-intact or endothelium-denuded aortic rings, respectively (Figure 2). A pED50 of −3.96 with a 95% confidence interval of −4.0 to −3.9 was obtained for the endothelium intact rings, while a pED50 of −3.79 with a 95% confidence interval of −3.9 to −3.6 was obtained for the endothelium-denuded ones. This significant difference between the two conditions clearly demonstrates that endothelium is, at least in part, involved in OME-induced relaxation.

3.3. Role of Nitric Oxide and cGMP in Origanum majorana Extract-Induced Aortic Relaxation

NO and cGMP are two important signalling molecules that play a vital role in vasorelaxation. We next investigated the role of these molecules in the vasorelaxant effect of OME. OME-induced relaxation of endothelium-intact aortic rings was significantly diminished by the presence of L-NAME or ODQ. In the presence or absence of L-NAME, the maximal relaxant effect was 54 ± 5% or 94 ± 3%, respectively (Figure 3A), and pED50 was −3.7 or −3.9 with confidence intervals of −3.9 to −3.5 or −4 to −3.9, respectively. Likewise, the maximal relaxant effect in the presence or absence of ODQ was 52 ± 7% or 96 ± 4%, respectively (Figure 3B), and pED50 was −3.5 or −3.9 with confidence intervals of −3.7 to −3.3 or −4 to −3.8, respectively. Taken together, these data support the notion that both NO and cGMP play a significant role in OME-induced vasorelaxation.

3.4. Effect of Origanum majorana Extract on the Production of cGMP in Aortic Rings

Because OME-induced vasorelaxation was significantly reduced by L-NAME and ODQ, we next wished to determine if OME modulates the level of cGMP. Treatment with OME caused a significant and dose-dependent increase in the levels of cGMP (Figure 4). In control rings (vehicle-treated), the cGMP level was 2.8 ± 0.8 (mean ± standard error of the mean (SEM)) picomole/mg protein. However, in rings treated with 0.3 mg/mL OME, the cGMP level rose to a dramatic level of 28 ± 5 (mean ± SEM) picomole/mg protein (p < 0.001). Importantly, L-NAME and ODQ significantly inhibited OME-induced production of cGMP (data not shown).

3.5. Involvement of the PI3K Signaling Pathway in the Origanum majorana Extract-Induced Relaxation of Aortic Rings

Overwhelming evidence documents the contribution of the PI3K-Akt signaling to endothelium-dependent aortic relaxation. To determine if this signaling pathway is involved in OME-induced relaxation, we employed two inhibitors, wortmannin and LY294002. Indeed, pre-incubation with either inhibitor instigated a significant reduction in OME-induced vasorelaxation. In rings incubated in the absence of either inhibitor, the maximal relaxant effect was 91 ± 3% and pED50 was −4.1 with confidence interval of −4.1 to −3.9. In wortmannin pre-incubated rings, the maximal relaxant effect was 53 ± 6% (Figure 5) and pED50 was −3.5 with confidence interval of 3.7–3.4. Similar results were obtained when rings were pre-incubated with LY294002 (10 µM), where the maximal effect was 58%, pED50 was −3.7 and a 95% confidence interval of −3.8 to −6 (Figure 5). Together, our data clearly allude to the role of the PI3K signaling in OME-induced aortic relaxation.

3.6. Effect of K+ Channel Blockers on Origanum majorana Extract-Induced Relaxation

Potassium channels are known to play an important role in vasodilation [24,25,26]. Thus, we wished to determine the of role of these channels in OME-induced relaxation. The vasorelaxant effects of OME were not altered by incubation with K+ channel blockers tetraethylammonium (TEA, 5 mM) or glibenclamide (Glib, 10 μM). Indeed, in the presence or absence of glibenclamide, the OME-induced maximal relaxant effect was 90 ± 4% or 92 ± 4% (Figure 6A), and pED50 was −3.7 or −4.1 with confidence intervals of −3.9 to −3.7 or −4 to −3.8, respectively. Similarly, incubation with TEA did not affect OME-induced relaxation of endothelium-intact aortic rings (Figure 6B). In the presence of TEA, the maximal effect was 88%, pED50 was −4, and the 95% confidence interval was −4.1 to −4. Together, these data argue for the absence of a role for potassium channels in OME-induced relaxation.

3.7. Effects of Ca2+ Channel Blockers on Origanum majorana Extract-Induced Relaxation

To determine if calcium channels contribute to OME-induced relaxation, we pre-incubated aortic rings with verapamil (1 µM) before adding OME. There was no significant difference between the control and treated vessels (p > 0.05) (Figure 7). Indeed, the maximal relaxant effect in vehicle-treated or verapamil-incubated rings was not affected (91 ± 4% vs. 92 ± 3%) (Figure 7). pED50 was −4.0 or −3.9 with confidence intervals of −4.1 to −3.9 or −3.9 to −3.8 in the absence or presence of verapamil, respectively.

3.8. Effect of Atropine and Pyrilamine on Origanum majorana Extract-Induced Relaxation

Endothelial muscarinic M3 receptors have been shown to promote vasorelaxation by virtue of their ability to increase NO release [27]. We thus wished to examine the effect of muscarinic receptors on OME-induced relaxation. Aortic rings with intact endothelium were pre-treated with atropine (1 μM) for 30 min before NE (3 μM) pre-contraction. In the absence of atropine, the maximal relaxant effect was 96 ± 7% and a pED50 of −3.98 with confidence intervals of −4.1 to −3.8 (Figure 8A). However, in the presence of atropine, the maximal relaxant effect was dramatically reduced to 43 ± 9% and a pED50 of −3.8 with confidence intervals of −4.3 to −3.4. This clearly shows that muscarinic receptors are involved in OME-induced relaxation.
Endothelial histaminergic receptors, particularly H1 G protein-coupled receptors, are known to initiate vasorelaxation [28]. We thus investigated the influence of pyrilamine (also called mepyramine), a blocker of histamine H1-receptors on OME-induced relaxation. Compared to the control, incubation with pyrilamine did not significantly modulate OME-induced relaxation of endothelium-intact rings. In the absence of pyrilamine, the maximal relaxant effect was 88.8 ± 6.9% and a pED50 of −3.88 with confidence intervals of −4.0 to −3.7 (Figure 8B). In the presence of pyrilamine, there was only a marginal and insignificant reduction in the maximal relaxation value, which was noted to be 75 ± 6%. pED50 of pyrilamine-treated rings was found to be −3.82 with confidence intervals of −3.9 to −3.6, which is very similar to that obtained in the vehicle-treated rings (Figure 8B). Thus, histaminergic receptors do not seem to have a significant effect on OME-induced relaxation (p > 0.05).

3.9. Effect of Cyclooxygenase Pathway on Origanum majorana Extract-Induced Relaxation

Vascular tone is greatly affected by prostanoids generated via the activity of cyclocoxygenases [29]. We thus sought to determine the potential role of the cyclooxygenases in OME-induced relaxation. In the absence of indomethacin, the maximal relaxant effect was 90.9 ± 2.8% and a pED50 of −3.95 with confidence intervals of −4.0 to −3.9 (Figure 9). In the presence of indomethacin, the maximal relaxant effect was 89.6 ± 1.9% and a pED50 of −3.9 with confidence intervals of −3.9 to −3.8 (Figure 9). Therefore, cyclooxygenases do not play an important role in OME-induced relaxation (p > 0.05) (Figure 9).

4. Discussion

In this report, we elucidated the underlying signaling mechanisms implicated in rat aortic relaxation induced by the leaf extract of O. majorana, commonly known as marjoram. This plant contains a bounty of bioactives, such as thymol, carvacrol, p-cymene, sabinene hydrate, rosmarinic acid, γ-terpinene, ursolic acid, and many others [30,31]. These constituents possess antibacterial, antifungal, and antispasmodic activities, in addition to their ability to scavenge free radicals, inhibit acetylcholinesterase, and depress cardiac activity [32,33]. Some of these bioactives, like carvacrol, thymol, ursolic acid, and hesperetin indeed exert vasorelaxing activities [19,20,22,34]. In addition to these effects, we have recently shown that this plant can significantly inhibit the malignant phenotype of cancer cells [15,16]. To the best of our knowledge, this is the first report that dissects the mechanism for the vasodilator effects of marjoram. It is important to note that using the whole plant, part of it, or a crude extract may provide benefits over the use of individual bioactives. This is partly due to the notion that these herbs are commonly grown and consumed by people in rural areas where access to modern medicine is limited. In addition, these extracts can easily be prepared at home and are thus significantly less costly, making them relatively more appealing to be used. Importantly, oftentimes, the plant as a whole, or as a crude extract, may prove more effective than individual bioactives, likely due to a potential synergy between these molecules.
There is an intricate coordination between vasoconstrictors and vasodilators in regulating vasotone. The maestro of this well-orchestrated balance is the endothelial cell layer. Indeed, this layer secretes many vasoconstrictors, the most potent of which is endothelin, as well as vasodilators, the most prominent of which is NO. In endothelial cells, NO is generated from l-arginine by endothelial NO synthase (eNOS) activation, which is stimulated by the calcium-calmodulin complex or activated via the PI3K/Akt pathway [35]. The role of NO in vasorelaxation is overwhelmingly established and documented. Many stimuli, including the vascular endothelial growth factor, β-agonists, and shear-stress signals regulate NO production via a pathway involving phosphoinositide 3-kinase (PI3-K)/Akt signaling. This pathway is known to play an important role in endothelium-mediated vasorelaxation [36]. Indeed, the PI3-K/Akt pathway activates eNOS via phosphorylating serine residue at position 1177 [37,38,39]. Here, when we incubated rings with PI3-K/Akt inhibitors, there was a significant reduction in the OME-induced relaxation. In an ischemia/reperfusion injury model, it was recently reported that ginsenosides of panax ginseng promote coronary arterial flow by activating the PI3K/Akt/eNOS cascade. This establishes the cardioprotective effects conferred by isolated extracts of ginseng [40]. Similarly, in another ischemia-reperfusion injury model, stimulation of PI3K/Akt/NO cascade by a ginsenoside metabolite bestowed cardioprotection [41].
Nitric oxide is known to activate soluble guanylate cyclase (sGC) in vascular smooth muscle cells (VSMCs) and to promote the synthesis of a secondary intracellular messenger, cGMP. This cGMP elicits many biological effects including relaxation of vascular smooth musculature [42,43,44] and consequent cGMP-mediated vasodilation [45,46,47]. In this context, our data reveals that pretreatment with L-NAME, a NOS inhibitor, or ODQ, a sGC inhibitor, affect the vasorelaxant effects of OME. These results are a clear indication that the vasorelaxant effect of OME are directly related to the NO-cGMP pathway. Our results are similar to other reports showing several plant species causing aortic relaxation through NO/cGMP. Some of these plant species include Schizophyllum commune [48], Salvia fruticosa L. [7], Euphorbia humifusa Willd [49], Alpiniae zerumbet (Pers.) B. L. Burtt & R. M. Sm. (Zingiberaceae) [50], Rhus coriaria L. [8], and Mansoa hirsuta D.C. [51]. Furthermore, here in our study, we report that OME increased production of cGMP, clearly supporting the involvement of sGC-cGMP in OME-induced relaxation.
In VSMCs, the membrane potential is dynamically regulated via changes in K+ channel activity, which greatly influences the function of voltage-dependent calcium channels [52,53]. Interestingly, NO is known to have a direct effect on activation of the K+ channels [54]. Here, we employed two K+ channel blockers, namely glibenclamide, a highly selective blocker of ATP-sensitive K+ channels, and TEA, a blocker of voltage-sensitive K+ channels [55]. Our results show neither glibenclamide nor TEA appreciably modulated OME-induced effects. Thus, K+ channels (K+ATP or KV channels) are not involved in OME-induced aortic relaxation. This was very surprising to us due to the greatly significant role of these channels in vasorelaxation. Indeed, these channels play a critical role in regulating plasma membrane potential, and hence determining vascular tone [56,57,58]. It is already established that NO directly activates calcium-dependent potassium channels in VSMCs [59], leading to vasorelaxation by a hyperpolarization mechanism [60]. Our results argue for the absence of a role for Ca2+-dependent or ATP-driven potassium channels in OME-induced aortic relaxation. Our findings differ from other studies that show that certain plant extracts produce an endothelium-dependent vasodilatory effect via activation of the potassium channels. However, our results are in accordance with previous studies [61], which report that stimulation of potassium channels is not necessarily implicated in vasorelaxation. It is not unreasonable that activation of the NO/cGMP pathway produces vasodilation through alternative mechanisms.
The role of Ca2+ in VSMC contraction/relaxation is well-known. The two main sources for this Ca2+ are the extracellular space and intracellular stores like the sarcoplasmic reticulum. Other mechanisms, such as those involving protein kinase C (PKC) or Ca2+ sensitization, are also implicated in VSMC contractility [62]. By blocking calcium channels, danshen has been shown to potentiate endothelium-independent relaxation of rat coronary arteries [63]. However, our results show that inhibiting calcium channels did not alter OME-induced relaxation, suggesting that mechanisms independent of these channels are involved. This is similar to recently reported findings showing that L-type calcium channels may not affect extract-induced relaxation [7,8]. It is worth mentioning here that one of the limitations of this study is that we did not assess the effect of depleting extracellular calcium on the vasorelaxation involved. However, due to the fact that L-type calcium channels were not involved, one may argue that the absence or presence of extracellular calcium is unlikely to be involved.
Vasorelaxant responses initiated by histamine are known to be mediated by histamine H1 receptors [28], which are G protein-coupled receptors. In this report, we did not find any contributory role for these receptors in OME-induced relaxation. This is similar to other studies that have used other plants species such as S. fruticosa, Salvia miltiorrhiza, or R. coriaria [7,8,64].
Cyclooxygenases (COX-1 and COX-2) convert arachidonic acid to active prostanoids. These prostanoids are well-known to play important roles in vascular physiology and pathology, including vasorelaxatory mechanisms [65]. Prostacyclin (PGI2) is a potent vasorelaxant prostaglandin that is released by the endothelium. It increases the intracellular accumulation of 3′-5′-cyclic adenosine monophosphate (cAMP), which is a powerful vasodilator as well as a modulator of many mechanisms in VSMCs [66,67,68,69,70,71,72,73]. In our study, pre-treatment with indomethacin, a non-selective cyclooxygenase inhibitor, did not affect the vasorelaxant effects of OME. This result suggested that the vasorelaxant effect of OME did not have a relationship with the vascular prostacyclin pathway. This is similar to other reports showing no role for prostaglandins in relaxation of rat femoral arteries or aortic rings in response to danshen or S. fruticosa extract, respectively [7,74]. Similarly, we recently reported no role for these prostanoids in R. coriaria-induced aortic relaxation [8]. Contextually, our results are not surprising since danshen, S. fruticosa, R. coriaria and marjoram, all contain rosmarinic acid, which is known to inhibit both COX-1 and COX-2 [75].
Finally, an endothelium-independent effect may be involved, though this needs further experimentation. It is possible that the residual vasorelaxation is due to increased activity of adenylate cyclase. This pathway has indeed been reported to underlie the vasodilatory mechanism of others herbs, such as R. coriaria [8]. Importantly, two bioactives found in marjoram, namely kaempferol and luteolin, have been reported to contribute to vasodilatation of rat aortae by inhibiting phosphodiesterases [76]. As is well known, inhibiting phosphodiesterases leads to increased accumulation of cAMP, which itself is a potent vasodilator. However, more experiments are warranted before such an assertion is validated.

5. Conclusions

In conclusion, our results show that the vasorelaxant effect of OME was due to activation of the PI-3K pathway, as well as an endothelium-dependent increase in the accumulation of cGMP. Together, our findings support the use of marjoram in the management/treatment of hypertension, [17]. More research, particularly in animal models of hypertension, is needed to better support the antihypertensive potential of this herb. It is important to note that studies directed at determining which bioactives reach the bloodstream after oral consumption of marjoram or its extract are warranted.

Author Contributions

Conceptualization, A.H.E.; Data curation, A.B., E.B. and A.S.; Formal analysis, G.P., J.M., R.I. and A.H.E.; Funding acquisition, A.B., E.B. and A.H.E.; Investigation, A.S. and A.H.E.; Methodology, R.I. and K.I.; Project administration, A.B.; Resources, A.B. and A.H.E.; Writing—Original draft, K.I. and A.H.E.; Writing—Review & editing, A.B., E.B., A.S., G.P., J.M., R.I. and A.H.E.

Funding

This research was funded by a grant (Fund #: 5/4/2019) from the University of Petra (Aman, Jordan) to A.B., E.B. and A.H.E.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Benjamin, E.J.; Blaha, M.J.; Chiuve, S.E.; Cushman, M.; Das, S.R.; Deo, R.; de Ferranti, S.D.; Floyd, J.; Fornage, M.; Gillespie, C.; et al. Heart Disease and Stroke Statistics-2017 Update: A Report from the American Heart Association. Circulation 2017, 135, e146–e603. [Google Scholar] [CrossRef] [PubMed]
  2. WHO. Hypertension. Available online: https://www.who.int/news-room/fact-sheets/detail/hypertension (accessed on 22 May 2019).
  3. WHO. WHO Traditional Medicine Strategy. Available online: https://apps.who.int/iris/bitstream/handle/10665/92455/9789241506090_eng.pdf?sequence=1 (accessed on 22 May 2019).
  4. Xia, M.; Qian, L.; Zhou, X.; Gao, Q.; Bruce, I.C.; Xia, Q. Endothelium-independent relaxation and contraction of rat aorta induced by ethyl acetate extract from leaves of Morus alba (L.). J. Ethnopharmacol. 2008, 120, 442–446. [Google Scholar] [CrossRef] [PubMed]
  5. Al Disi, S.S.; Anwar, M.A.; Eid, A.H. Anti-hypertensive Herbs and their Mechanisms of Action: Part I. Front. Pharmacol. 2015, 6, 323. [Google Scholar] [CrossRef]
  6. Anwar, M.A.; Al Disi, S.S.; Eid, A.H. Anti-Hypertensive Herbs and Their Mechanisms of Action: Part II. Front. Pharmacol. 2016, 7, 50. [Google Scholar] [CrossRef] [PubMed]
  7. Anwar, M.A.; Samaha, A.A.; Ballan, S.; Saleh, A.I.; Iratni, R.; Eid, A.H. Salvia fruticosa Induces Vasorelaxation In Rat Isolated Thoracic Aorta: Role of the PI3K/Akt/eNOS/NO/cGMP Signaling Pathway. Sci. Rep. 2017, 7, 686. [Google Scholar] [CrossRef]
  8. Anwar, M.A.; Samaha, A.A.; Baydoun, S.; Iratni, R.; Eid, A.H. Rhus coriariaL. (Sumac) Evokes Endothelium-Dependent Vasorelaxation of Rat Aorta: Involvement of the cAMP and cGMP Pathways. Front. Pharmacol. 2018, 9, 688. [Google Scholar] [CrossRef] [PubMed]
  9. Ibarra-Alvarado, C.; Rojas, A.; Mendoza, S.; Bah, M.; Gutierrez, D.M.; Hernandez-Sandoval, L.; Martinez, M. Vasoactive and antioxidant activities of plants used in Mexican traditional medicine for the treatment of cardiovascular diseases. Pharm. Biol. 2010, 48, 732–739. [Google Scholar] [CrossRef] [PubMed]
  10. Banchio, E.; Bogino, P.C.; Zygadlo, J.; Giordanoa, W. Plant growth promoting rhizobacteria improve growth and essential oil yield in Origanum majorana L. Biochem. Syst. Ecol. 2008, 36, 766–771. [Google Scholar] [CrossRef]
  11. Baranauskiene, R.; Venskutonis, P.R.; Dewettinck, K.; Verhéc, R. Properties of oregano (Origanumvulgare L.), citronella (Cymbopogonnardus G.) and marjoram (Majoranahortensis L.) flavors encapsulated into milk protein-based matrices. Food Res. Int. 2006, 39, 413–425. [Google Scholar] [CrossRef]
  12. Loizzo, M.; Menichini, F.; Conforti, F.; Tundis, R.; Bonesi, M.; Saab, A. Chemical analysis, antioxidant, antiinflammatory and anticholinesterase activities of Origanum ehrenbergii Boiss and Origanum syriacum L. essential oils. Food Chem. 2009, 117, 174–180. [Google Scholar] [CrossRef]
  13. Hayouni el, A.; Chraief, I.; Abedrabba, M.; Bouix, M.; Leveau, J.Y.; Mohammed, H.; Hamdi, M. Tunisian Salvia officinalis L. and Schinus molle L. essential oils: Their chemical compositions and their preservative effects against Salmonella inoculated in minced beef meat. Int. J. Food Microbiol. 2008, 125, 242–251. [Google Scholar] [CrossRef] [PubMed]
  14. Muñoz, M.; Guevara, L.; Palop, A.; Tabera, J.; Fernandez, P.S. Determination of the effect of plant essential oils obtained by supercritical fluid extraction on the growth and viability of Listeria monocytogenes in broth and food systems using flow cytometry. LWT-Food Sci. Technol. 2009, 42, 220–227. [Google Scholar] [CrossRef]
  15. Al Dhaheri, Y.; Attoub, S.; Arafat, K.; Abuqamar, S.; Viallet, J.; Saleh, A.; Al Agha, H.; Eid, A.; Iratni, R. Anti-metastatic and anti-tumor growth effects of Origanum majorana on highly metastatic human breast cancer cells: Inhibition of NFκB signaling and reduction of nitric oxide production. PLoS ONE 2013, 8, e68808. [Google Scholar] [CrossRef] [PubMed]
  16. Al Dhaheri, Y.; Eid, A.; AbuQamar, S.; Attoub, S.; Khasawneh, M.; Aiche, G.; Hisaindee, S.; Iratni, R. Mitotic arrest and apoptosis in breast cancer cells induced by Origanum majorana extract: Upregulation of TNF-α and downregulation of survivin and mutant p53. PLoS ONE 2013, 8, e56649. [Google Scholar] [CrossRef] [PubMed]
  17. Tahraoui, A.; El-Hilaly, J.; Israili, Z.H.; Lyoussi, B. Ethnopharmacological survey of plants used in the traditional treatment of hypertension and diabetes in south-eastern Morocco (Errachidia province). J. Ethnopharmacol. 2007, 110, 105–117. [Google Scholar] [CrossRef] [PubMed]
  18. Shabir, H.; Kundu, S.; Basir, S.F.; Khan, L.A. Modulation of Pb(II) caused aortal constriction by eugenol and carvacrol. Biol. Trace Elem. Res. 2014, 161, 116–122. [Google Scholar] [CrossRef] [PubMed]
  19. Testai, L.; Chericoni, S.; Martelli, A.; Flamini, G.; Breschi, M.C.; Calderone, V. Voltage-operated potassium (Kv) channels contribute to endothelium-dependent vasorelaxation of carvacrol on rat aorta. J. Pharm. Pharmacol. 2016, 68, 1177–1183. [Google Scholar] [CrossRef]
  20. Peixoto-Neves, D.; Silva-Alves, K.S.; Gomes, M.D.; Lima, F.C.; Lahlou, S.; Magalhaes, P.J.; Ceccatto, V.M.; Coelho-de-Souza, A.N.; Leal-Cardoso, J.H. Vasorelaxant effects of the monoterpenic phenol isomers, carvacrol and thymol, on rat isolated aorta. Fundam. Clin. Pharmacol. 2010, 24, 341–350. [Google Scholar] [CrossRef]
  21. Erenler, R.; Sen, O.; Aksit, H.; Demirtas, I.; Yaglioglu, A.S.; Elmastas, M.; Telci, I. Isolation and identification of chemical constituents from Origanum majorana and investigation of antiproliferative and antioxidant activities. J. Sci. Food Agric. 2016, 96, 822–836. [Google Scholar] [CrossRef]
  22. Yamamoto, M.; Suzuki, A.; Hase, T. Short-term effects of glucosyl hesperidin and hesperetin on blood pressure and vascular endothelial function in spontaneously hypertensive rats. J. Nutr. Sci. Vitaminol. 2008, 54, 95–98. [Google Scholar] [CrossRef]
  23. Li, X.; Kim, H.Y.; Cui, H.Z.; Cho, K.W.; Kang, D.G.; Lee, H.S. Water extract of Zanthoxylum piperitum induces vascular relaxation via endothelium-dependent NO-cGMP signaling. J. Ethnopharmacol. 2010, 129, 197–202. [Google Scholar] [CrossRef]
  24. McGarr, G.W.; Fujii, N.; Muia, C.M.; Nishiyasu, T.; Kenny, G.P. The separate and combined effects of KCa and KATP channel blockade with NOS inhibition on cutaneous vasodilation and sweating in older men during heat stress. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2019. [Google Scholar] [CrossRef]
  25. Alaaeddine, R.A.; Mroueh, A.; Gust, S.; Eid, A.H.; Plane, F.; El-Yazbi, A.F. Impaired cross-talk between NO and hyperpolarization in myoendothelial feedback: A novel therapeutic target in early endothelial dysfunction of metabolic disease. Curr. Opin. Pharmacol. 2019, 45, 33–41. [Google Scholar] [CrossRef]
  26. Maaliki, D.; Shaito, A.A.; Pintus, G.; El-Yazbi, A.; Eid, A.H. Flavonoids in hypertension: A brief review of the underlying mechanisms. Curr. Opin. Pharmacol. 2019, 45, 57–65. [Google Scholar] [CrossRef]
  27. Attina, T.M.; Oliver, J.J.; Malatino, L.S.; Webb, D.J. Contribution of the M3 muscarinic receptors to the vasodilator response to acetylcholine in the human forearm vascular bed. Br. J. Clin. Pharmacol. 2008, 66, 300–303. [Google Scholar] [CrossRef]
  28. Van de Voorde, J.; Leusen, I. Role of the endothelium in the vasodilator response of rat thoracic aorta to histamine. Eur. J. Pharmacol. 1983, 87, 113–120. [Google Scholar] [CrossRef]
  29. Chen, H.F.; Xie, L.D.; Xu, C.S. The signal transduction pathways of heat shock protein 27 phosphorylation in vascular smooth muscle cells. Mol. Cell. Biochem. 2010, 333, 49–56. [Google Scholar] [CrossRef]
  30. Zgheib, R.; Chaillou, S.; Ouaini, N.; Kassouf, A.; Rutledge, D.; El Azzi, D.; El Beyrouthy, M. Chemometric Tools to Highlight the Variability of the Chemical Composition and Yield of Lebanese Origanum syriacum L. Essential Oil. Chem. Biodivers. 2016, 13, 1326–1347. [Google Scholar] [CrossRef]
  31. Bina, F.; Rahimi, R. Sweet Marjoram: A Review of Ethnopharmacology, Phytochemistry, and Biological Activities. J. Evid.-Based Complement. Altern. Med. 2017, 22, 175–185. [Google Scholar] [CrossRef]
  32. Marinelli, O.; Nabissi, M.; Morelli, M.B.; Torquati, L.; Amantini, C.; Santoni, G. ICOS-L as a Potential Therapeutic Target for Cancer Immunotherapy. Curr. Protein Pept. Sci. 2018, 19, 1107–1113. [Google Scholar] [CrossRef]
  33. Marchese, A.; Orhan, I.E.; Daglia, M.; Barbieri, R.; Di Lorenzo, A.; Nabavi, S.F.; Gortzi, O.; Izadi, M.; Nabavi, S.M. Antibacterial and antifungal activities of thymol: A brief review of the literature. Food Chem. 2016, 210, 402–414. [Google Scholar] [CrossRef]
  34. Aguirre-Crespo, F.; Vergara-Galicia, J.; Villalobos-Molina, R.; Javier Lopez-Guerrero, J.; Navarrete-Vazquez, G.; Estrada-Soto, S. Ursolic acid mediates the vasorelaxant activity of Lepechinia caulescens via NO release in isolated rat thoracic aorta. Life Sci. 2006, 79, 1062–1068. [Google Scholar] [CrossRef]
  35. Lorenz, M.; Wessler, S.; Follmann, E.; Michaelis, W.; Dusterhoft, T.; Baumann, G.; Stangl, K.; Stangl, V. A constituent of green tea, epigallocatechin-3-gallate, activates endothelial nitric oxide synthase by a phosphatidylinositol-3-OH-kinase-, cAMP-dependent protein kinase-, and Akt-dependent pathway and leads to endothelial-dependent vasorelaxation. J. Biol. Chem. 2004, 279, 6190–6195. [Google Scholar] [CrossRef]
  36. Isenovi, E.; Walsh, M.F.; Muniyappa, R.; Bard, M.; Diglio, C.A.; Sowers, J.R. Phosphatidylinositol 3-kinase may mediate isoproterenol-induced vascular relaxation in part through nitric oxide production. Metabolism 2002, 51, 380–386. [Google Scholar] [CrossRef]
  37. Zeng, G.; Nystrom, F.H.; Ravichandran, L.V.; Cong, L.N.; Kirby, M.; Mostowski, H.; Quon, M.J. Roles for insulin receptor, PI3-kinase, and Akt in insulin-signaling pathways related to production of nitric oxide in human vascular endothelial cells. Circulation 2000, 101, 1539–1545. [Google Scholar] [CrossRef]
  38. Isenovic, E.; Muniyappa, R.; Milivojevic, N.; Rao, Y.; Sowers, J.R. Role of PI3-kinase in isoproterenol and IGF-1 induced ecNOS activity. Biochem. Biophys. Res. Commun. 2001, 285, 954–958. [Google Scholar] [CrossRef]
  39. Hood, J.D.; Meininger, C.J.; Ziche, M.; Granger, H.J. VEGF upregulates ecNOS message, protein, and NO production in human endothelial cells. Am. J. Physiol. 1998, 274, H1054–H1058. [Google Scholar] [CrossRef]
  40. Yi, X.Q.; Li, T.; Wang, J.R.; Wong, V.K.W.; Luo, P.; Wong, I.Y.F.; Jiang, Z.H.; Liu, L.A.; Zhou, H. Total ginsenosides increase coronary perfusion flow in isolated rat hearts through activation of PI3K/Akt-eNOS signaling. Phytomed. Int. J. Phytother. Phytopharm. 2010, 17, 1006–1015. [Google Scholar] [CrossRef]
  41. Tsutsumi, Y.M.; Tsutsumi, R.; Mawatari, K.; Nakaya, Y.; Kinoshita, M.; Tanaka, K.; Oshita, S. Compound K, a metabolite of ginsenosides, induces cardiac protection mediated nitric oxide via Akt/PI3K pathway. Life Sci. 2011, 88, 725–729. [Google Scholar] [CrossRef]
  42. Paolocci, N.; Ekelund, U.E.; Isoda, T.; Ozaki, M.; Vandegaer, K.; Georgakopoulos, D.; Harrison, R.W.; Kass, D.A.; Hare, J.M. cGMP-independent inotropic effects of nitric oxide and peroxynitrite donors: Potential role for nitrosylation. Am. J. Physiol. Heart Circ. Physiol. 2000, 279, H1982–H1988. [Google Scholar] [CrossRef]
  43. Fleming, I.; Busse, R. Molecular mechanisms involved in the regulation of the endothelial nitric oxide synthase. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2003, 284, R1–R12. [Google Scholar] [CrossRef] [Green Version]
  44. Adachi, T.; Weisbrod, R.M.; Pimentel, D.R.; Ying, J.; Sharov, V.S.; Schoneich, C.; Cohen, R.A. S-Glutathiolation by peroxynitrite activates SERCA during arterial relaxation by nitric oxide. Nat. Med. 2004, 10, 1200–1207. [Google Scholar] [CrossRef]
  45. Forstermann, U.; Munzel, T. Endothelial nitric oxide synthase in vascular disease: From marvel to menace. Circulation 2006, 113, 1708–1714. [Google Scholar] [CrossRef]
  46. Deanfield, J.E.; Halcox, J.P.; Rabelink, T.J. Endothelial function and dysfunction: Testing and clinical relevance. Circulation 2007, 115, 1285–1295. [Google Scholar] [CrossRef]
  47. Stankevicius, E.; Kevelaitis, E.; Vainorius, E.; Simonsen, U. Role of nitric oxide and other endothelium-derived factors. Medicina 2003, 39, 333–341. [Google Scholar]
  48. Chen, H.; Li, S.; Wang, P.; Yan, S.; Hu, L.; Pan, X.; Yang, C.; Leung, G.P. Endothelium-dependent and -independent relaxation of rat aorta induced by extract of Schizophyllum commune. Phytomed. Int. J. Phytother. Phytopharm. 2014, 21, 1230–1236. [Google Scholar] [CrossRef]
  49. Wang, T.T.; Zhou, G.H.; Kho, J.H.; Sun, Y.Y.; Wen, J.F.; Kang, D.G.; Lee, H.S.; Cho, K.W.; Jin, S.N. Vasorelaxant action of an ethylacetate fraction of Euphorbia humifusa involves NO-cGMP pathway and potassium channels. J. Ethnopharmacol. 2013, 148, 655–663. [Google Scholar] [CrossRef]
  50. Tao, L.; Hu, H.S.; Shen, X.C. Endothelium-dependent vasodilatation effects of the essential oil from Fructus alpiniae zerumbet (EOFAZ) on rat thoracic aortic rings in vitro. Phytomed. Int. J. Phytother. Phytopharm. 2013, 20, 387–393. [Google Scholar] [CrossRef]
  51. Campana, P.R.; Braga, F.C.; Cortes, S.F. Endothelium-dependent vasorelaxation in rat thoracic aorta by Mansoa hirsuta D.C. Phytomed. Int. J. Phytother. Phytopharm. 2009, 16, 456–461. [Google Scholar] [CrossRef]
  52. Brayden, J.E. Potassium channels in vascular smooth muscle. Clin. Exp. Pharmacol. Physiol. 1996, 23, 1069–1076. [Google Scholar] [CrossRef]
  53. Nelson, M.T.; Quayle, J.M. Physiological roles and properties of potassium channels in arterial smooth muscle. Am. J. Physiol. 1995, 268, C799–C822. [Google Scholar] [CrossRef]
  54. Villar, I.C.; Francis, S.; Webb, A.; Hobbs, A.J.; Ahluwalia, A. Novel aspects of endothelium-dependent regulation of vascular tone. Kidney Int. 2006, 70, 840–853. [Google Scholar] [CrossRef] [Green Version]
  55. Novakovic, A.; Bukarica, L.G.; Kanjuh, V.; Heinle, H. Potassium channels-mediated vasorelaxation of rat aorta induced by resveratrol. Basic Clin. Pharmacol. Toxicol. 2006, 99, 360–364. [Google Scholar] [CrossRef]
  56. Tykocki, N.R.; Boerman, E.M.; Jackson, W.F. Smooth Muscle Ion Channels and Regulation of Vascular Tone in Resistance Arteries and Arterioles. Compr. Physiol. 2017, 7, 485–581. [Google Scholar] [CrossRef] [Green Version]
  57. Jackson, W.F. Ion channels and vascular tone. Hypertension 2000, 35, 173–178. [Google Scholar] [CrossRef]
  58. Sobey, C.G. Potassium channel function in vascular disease. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 28–38. [Google Scholar] [CrossRef]
  59. Bolotina, V.M.; Najibi, S.; Palacino, J.J.; Pagano, P.J.; Cohen, R.A. Nitric oxide directly activates calcium-dependent potassium channels in vascular smooth muscle. Nature 1994, 368, 850–853. [Google Scholar] [CrossRef]
  60. Archer, S.L.; Huang, J.M.; Hampl, V.; Nelson, D.P.; Shultz, P.J.; Weir, E.K. Nitric oxide and cGMP cause vasorelaxation by activation of a charybdotoxin-sensitive K channel by cGMP-dependent protein kinase. Proc. Natl. Acad. Sci. USA 1994, 91, 7583–7587. [Google Scholar] [CrossRef]
  61. Luna-Vazquez, F.J.; Ibarra-Alvarado, C.; Camacho-Corona, M.D.; Rojas-Molina, A.; Rojas-Molina, J.I.; Garcia, A.; Bah, M. Vasodilator Activity of Compounds Isolated from Plants Used in Mexican Traditional Medicine. Molecules 2018, 23, 1474. [Google Scholar] [CrossRef]
  62. Webb, R.C. Smooth muscle contraction and relaxation. Adv. Physiol. Educ. 2003, 27, 201–206. [Google Scholar] [CrossRef]
  63. Lam, F.F.; Yeung, J.H.; Chan, K.M.; Or, P.M. Relaxant effects of Danshen aqueous extract and its constituent danshensu on rat coronary artery are mediated by inhibition of calcium channels. Vasc. Pharmacol. 2007, 46, 271–277. [Google Scholar] [CrossRef]
  64. Lam, F.F.; Yeung, J.H.; Cheung, J.H. Mechanisms of the dilator action of Danshen (Salvia miltiorrhiza) on rat isolated femoral artery. J. Cardiovasc. Pharmacol. 2005, 46, 361–368. [Google Scholar] [CrossRef]
  65. Gerber, R.T.; Anwar, M.A.; Poston, L. Enhanced acetylcholine induced relaxation in small mesenteric arteries from pregnant rats: An important role for endothelium-derived hyperpolarizing factor (EDHF). Br. J. Pharmacol. 1998, 125, 455–460. [Google Scholar] [CrossRef]
  66. Faraci, F.M.; Heistad, D.D. Regulation of the cerebral circulation: Role of endothelium and potassium channels. Physiol. Rev. 1998, 78, 53–97. [Google Scholar] [CrossRef]
  67. Motawea, H.K.; Jeyaraj, S.C.; Eid, A.H.; Mitra, S.; Unger, N.T.; Ahmed, A.A.; Flavahan, N.A.; Chotani, M.A. Cyclic AMP-Rap1A signaling mediates cell surface translocation of microvascular smooth muscle α2C-adrenoceptors through the actin-binding protein filamin-2. Am. J. Physiol. Cell Physiol. 2013, 305, C829–C845. [Google Scholar] [CrossRef]
  68. Jeyaraj, S.C.; Unger, N.T.; Eid, A.H.; Mitra, S.; Paul El-Dahdah, N.; Quilliam, L.A.; Flavahan, N.A.; Chotani, M.A. Cyclic AMP-Rap1A signaling activates RhoA to induce α(2c)-adrenoceptor translocation to the cell surface of microvascular smooth muscle cells. Am. J. Physiol. Cell Physiol. 2012, 303, C499–C511. [Google Scholar] [CrossRef]
  69. Eid, A.H.; Chotani, M.A.; Mitra, S.; Miller, T.J.; Flavahan, N.A. Cyclic AMP acts through Rap1 and JNK signaling to increase expression of cutaneous smooth muscle α2C-adrenoceptors. Am. J. Physiol. Heart Circ. Physiol. 2008, 295, H266–H272. [Google Scholar] [CrossRef]
  70. Eid, A.H. cAMP induces adhesion of microvascular smooth muscle cells to fibronectin via an Epac-mediated but PKA-independent mechanism. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2012, 30, 247–258. [Google Scholar] [CrossRef]
  71. Chotani, M.A.; Mitra, S.; Eid, A.H.; Han, S.A.; Flavahan, N.A. Distinct cAMP signaling pathways differentially regulate –2C-adrenoceptor expression: Role in serum induction in human arteriolar smooth muscle cells. Am. J. Physiol. Heart Circ. Physiol. 2005, 288, H69–H76. [Google Scholar] [CrossRef]
  72. Dehaini, H.; Fardoun, M.; Abou-Saleh, H.; El-Yazbi, A.; Eid, A.A.; Eid, A.H. Estrogen in vascular smooth muscle cells: A friend or a foe? Vasc. Pharmacol. 2018, 111, 15–21. [Google Scholar] [CrossRef]
  73. Eid, A.H.; Maiti, K.; Mitra, S.; Chotani, M.A.; Flavahan, S.; Bailey, S.R.; Thompson-Torgerson, C.S.; Flavahan, N.A. Estrogen increases smooth muscle expression of α2C-adrenoceptors and cold-induced constriction of cutaneous arteries. Am. J. Physiol. Heart Circ. Physiol. 2007, 293, H1955–H1961. [Google Scholar] [CrossRef]
  74. Cao, J.; Wei, Y.J.; Qi, L.W.; Li, P.; Qian, Z.M.; Luo, H.W.; Chen, J.; Zhao, J. Determination of fifteen bioactive components in Radix et Rhizoma Salviae Miltiorrhizae by high-performance liquid chromatography with ultraviolet and mass spectrometric detection. Biomed. Chromatogr. BMC 2008, 22, 164–172. [Google Scholar] [CrossRef]
  75. Kelm, M.A.; Nair, M.G.; Strasburg, G.M.; DeWitt, D.L. Antioxidant and cyclooxygenase inhibitory phenolic compounds from Ocimum sanctum Linn. Phytomed. Int. J. Phytother. Phytopharm. 2000, 7, 7–13. [Google Scholar] [CrossRef]
  76. Duarte, J.; Perez Vizcaino, F.; Utrilla, P.; Jimenez, J.; Tamargo, J.; Zarzuelo, A. Vasodilatory effects of flavonoids in rat aortic smooth muscle. Structure-activity relationships. Gen. Pharmacol. 1993, 24, 857–862. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Effect of Origanum majorana (OME) extract on the vasorelaxation of aortic rings. Cumulative dose response curve for OME-induced relaxation of rat aortic rings was determined. Data expressed are mean ± standard error of the mean (SEM, n = 7).
Figure 1. Effect of Origanum majorana (OME) extract on the vasorelaxation of aortic rings. Cumulative dose response curve for OME-induced relaxation of rat aortic rings was determined. Data expressed are mean ± standard error of the mean (SEM, n = 7).
Biomolecules 09 00227 g001
Figure 2. Role of endothelium in OME-induced relaxation. Cumulative dose-response curves for OME in isolated norepinephrine (NE)-pre-contracted rat aortic rings either with intact (+E; black) or denuded endothelium (-E; red). Data are expressed as mean ± SEM (n = 7; p < 0.01 for +E vs. -E).
Figure 2. Role of endothelium in OME-induced relaxation. Cumulative dose-response curves for OME in isolated norepinephrine (NE)-pre-contracted rat aortic rings either with intact (+E; black) or denuded endothelium (-E; red). Data are expressed as mean ± SEM (n = 7; p < 0.01 for +E vs. -E).
Biomolecules 09 00227 g002
Figure 3. Role of nitrous oxide (NO) or cGMP in OME-induced relaxation. (A) Endothelium-intact rings were treated with cumulative doses of OME in the presence (red) or the absence (black) of Nω-nitro-l-arginine methyl ester (L-NAME) (inhibitor of eNOS, 100 µM). Data represent mean ± SEM (p < 0.01 for OME alone vs. L-NAME plus OME; n = 7). (B) Endothelium-intact rings were treated with cumulative doses of OME in the presence (red) or absence (black) of ODQ (inhibitor of soluble guanylate cyclase, 1 µM). Data represent mean ± SEM (p < 0.01 for OME vs. ODQ plus OME; n = 6).
Figure 3. Role of nitrous oxide (NO) or cGMP in OME-induced relaxation. (A) Endothelium-intact rings were treated with cumulative doses of OME in the presence (red) or the absence (black) of Nω-nitro-l-arginine methyl ester (L-NAME) (inhibitor of eNOS, 100 µM). Data represent mean ± SEM (p < 0.01 for OME alone vs. L-NAME plus OME; n = 7). (B) Endothelium-intact rings were treated with cumulative doses of OME in the presence (red) or absence (black) of ODQ (inhibitor of soluble guanylate cyclase, 1 µM). Data represent mean ± SEM (p < 0.01 for OME vs. ODQ plus OME; n = 6).
Biomolecules 09 00227 g003
Figure 4. Effect of OME on cGMP levels. Rings were treated without (control) or with increasing concentrations of OME. cGMP levels were quantitated by an immunoassay. Data are shown as mean ± SEM, n = 5. (* p < 0.01; ** p < 0.001).
Figure 4. Effect of OME on cGMP levels. Rings were treated without (control) or with increasing concentrations of OME. cGMP levels were quantitated by an immunoassay. Data are shown as mean ± SEM, n = 5. (* p < 0.01; ** p < 0.001).
Biomolecules 09 00227 g004
Figure 5. Effect of the PI3K pathway on OME-induced vasorelaxation. Rings with intact endothelium were treated with OME in the absence (black) or presence of phosphoinositide 3-kinase (PI3-K) inhibitors: Wortmannin (0.1 µM; red) or LY29400 (10 µM; blue). Data presented as mean ± SEM (n = 5; p < 0.01 for OME vs. Wortmannin + OME or LY29400 + OME).
Figure 5. Effect of the PI3K pathway on OME-induced vasorelaxation. Rings with intact endothelium were treated with OME in the absence (black) or presence of phosphoinositide 3-kinase (PI3-K) inhibitors: Wortmannin (0.1 µM; red) or LY29400 (10 µM; blue). Data presented as mean ± SEM (n = 5; p < 0.01 for OME vs. Wortmannin + OME or LY29400 + OME).
Biomolecules 09 00227 g005
Figure 6. Effect of potassium channel inhibitors on OME-induced vasorelaxation. Rings with intact endothelium were treated with OME in the absence (OME; black) or presence of (A) 10 µM of glibenclamide (Glib + OME; red), or (B) 100 µM of tetraethylammonium (TEA + OME; red). Data represent mean ± SEM (n = 6). p > 0.05 for OME alone vs. either Glib + OME or OME + TEA.
Figure 6. Effect of potassium channel inhibitors on OME-induced vasorelaxation. Rings with intact endothelium were treated with OME in the absence (OME; black) or presence of (A) 10 µM of glibenclamide (Glib + OME; red), or (B) 100 µM of tetraethylammonium (TEA + OME; red). Data represent mean ± SEM (n = 6). p > 0.05 for OME alone vs. either Glib + OME or OME + TEA.
Biomolecules 09 00227 g006
Figure 7. Role of calcium channels in OME-induced relaxation of aortic rings. Endothelium-intact aortic rings were pre-incubated with OME in the absence (OME; black) or presence of verapamil (verap) (1 µM; verap + OME; red). Data are presented as mean ± SEM (n = 5; p > 0.05).
Figure 7. Role of calcium channels in OME-induced relaxation of aortic rings. Endothelium-intact aortic rings were pre-incubated with OME in the absence (OME; black) or presence of verapamil (verap) (1 µM; verap + OME; red). Data are presented as mean ± SEM (n = 5; p > 0.05).
Biomolecules 09 00227 g007
Figure 8. Involvement of histaminic or muscarinic receptors in OME-induced relaxation. Rings with intact endothelium were incubated with cumulative doses of OME in the absence (OME; black) or presence of (A) 10 µM of atropine (natural alkaloid with antagonistic properties at muscarinic acetylcholine receptors; atropine + OME; red) or (B) 10 µM of pyrilamine (blocker of H1 histamine receptors; pyrilamine + OME; red). Data showed represent mean ± SEM (n = 6 or 5 for atropine or pyrilamine-treated rings, respectively). p < 0.05 for OME alone vs. either atropine + OME; p > 0.05 for OME alone vs. either pyrilamine + OME.
Figure 8. Involvement of histaminic or muscarinic receptors in OME-induced relaxation. Rings with intact endothelium were incubated with cumulative doses of OME in the absence (OME; black) or presence of (A) 10 µM of atropine (natural alkaloid with antagonistic properties at muscarinic acetylcholine receptors; atropine + OME; red) or (B) 10 µM of pyrilamine (blocker of H1 histamine receptors; pyrilamine + OME; red). Data showed represent mean ± SEM (n = 6 or 5 for atropine or pyrilamine-treated rings, respectively). p < 0.05 for OME alone vs. either atropine + OME; p > 0.05 for OME alone vs. either pyrilamine + OME.
Biomolecules 09 00227 g008
Figure 9. Effect of cyclooxygenase inhibition by indomethacin on OME-induced aortic relaxation. Rings with intact endothelium were pre-treated without (OME; black) or with indomethacin (a cyclooxygenase (COX)1/2 inhibitor, 10 µM; INDO + OME; red) followed by the addition of cumulative doses of OME. Data showed represent mean ± SEM (n = 5; p > 0.05).
Figure 9. Effect of cyclooxygenase inhibition by indomethacin on OME-induced aortic relaxation. Rings with intact endothelium were pre-treated without (OME; black) or with indomethacin (a cyclooxygenase (COX)1/2 inhibitor, 10 µM; INDO + OME; red) followed by the addition of cumulative doses of OME. Data showed represent mean ± SEM (n = 5; p > 0.05).
Biomolecules 09 00227 g009

Share and Cite

MDPI and ACS Style

Badran, A.; Baydoun, E.; Samaha, A.; Pintus, G.; Mesmar, J.; Iratni, R.; Issa, K.; Eid, A.H. Marjoram Relaxes Rat Thoracic Aorta Via a PI3-K/eNOS/cGMP Pathway. Biomolecules 2019, 9, 227. https://0-doi-org.brum.beds.ac.uk/10.3390/biom9060227

AMA Style

Badran A, Baydoun E, Samaha A, Pintus G, Mesmar J, Iratni R, Issa K, Eid AH. Marjoram Relaxes Rat Thoracic Aorta Via a PI3-K/eNOS/cGMP Pathway. Biomolecules. 2019; 9(6):227. https://0-doi-org.brum.beds.ac.uk/10.3390/biom9060227

Chicago/Turabian Style

Badran, Adnan, Elias Baydoun, Ali Samaha, Gianfranco Pintus, Joelle Mesmar, Rabah Iratni, Khodr Issa, and Ali H. Eid. 2019. "Marjoram Relaxes Rat Thoracic Aorta Via a PI3-K/eNOS/cGMP Pathway" Biomolecules 9, no. 6: 227. https://0-doi-org.brum.beds.ac.uk/10.3390/biom9060227

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