Skip to main content

Anti-inflammatory and anti-infectious effects of Evodia rutaecarpa (Wuzhuyu) and its major bioactive components

Abstract

This article reviews the anti-inflammatory relative and anti-infectious effects of Evodia rutaecarpa and its major bioactive components and the involvement of the nitric oxide synthases, cyclooxygenase, NADPH oxidase, nuclear factor kappa B, hypoxia-inducible factor 1 alpha, reactive oxygen species, prostaglandins, tumor necrosis factor, LIGHT, amyloid protein and orexigenic neuropeptides. Their potential applications for the treatment of endotoxaemia, obesity, diabetes, Alzheimer's disease and their uses as cardiovascular and gastrointestinal protective agents, analgesics, anti-oxidant, anti-atherosclerosis agents, dermatological agents and anti-infectious agents are highlighted. Stimulation of calcitonin gene-related peptide release may partially explain the analgesic, cardiovascular and gastrointestinal protective, anti-obese activities of Evodia rutaecarpa and its major bioactive components.

Introduction

Inflammation is a protective physiological response of an organism to chemical, physical, infectious agents, environmental toxins, ischemia or an antigen-antibody interaction. However, prolonged or overactive inflammation may cause tissue damage. Inflammation is very common manifested as body temperature change, edema, itch and pain, occasionally as serious as septic shock, tissue cirrhosis, necrosis or cancer. In the United States, over 500,000 patients suffer from sepsis triggered by severe systemic inflammation per year [1].

Various factors are involved in inflammation, such as calcium homeostasis, histamine, bradykinin, serotonin (5-HT), eicosanoids (prostaglandins, PG; thromboxanes, TX; leukotrienes, LT), platelet-activating factor, hormones (corticosterones), cytokines, interleukins (IL), chemotaxics, cyclooxygenase (COX), adhesion molecules, reactive oxygen species (ROS) (H2O2, O2-), nitric oxide (NO) and substance P. Cells taking part in inflammation are erythrocytes, neutrophils, basophils, eosinophils, platelet, natural killer cells, lymphocytes, mast cells, antigen presenting cells and dendritic cells [2]. Diseases and syndromes, such as arthritis, atherosclerosis, atopic dermatitis, brain or heart stroke, cancer, cataract, diabetes, neurodegeneration, pain, rhinitis and septic shock, are all related to inflammation.

Natural products may still be the most abundant sources for new drug development. Aspirin and corticosterone are two well known examples for anti-inflammatory products derived from Nature. Favonoids are potential therapeutic agents for the treatment of inflammation, heart disease and cancer [3]. This article reviews the anti-inflammatory relative and anti-infectious effects of Evodia rutaecarpa (Wuzhuyu) and its major bioactive components such as dehydroevodiamine (DeHE), evodiamine (Evo) and rutaecarpine (Rut).

Mechanisms of anti-inflammatory relative effects of Evodia rutaecarpa and its bioactive components are summarized in Additional file 1.

Effects on nitric oxide (NO) system and nitric oxide synthase (NOS)

While NO is involved in the blood pressure regulation, smooth muscle relaxation, platelet aggregation, neurotransmission, long-term potentiation, penile erection, apoptosis and immune response, over-expression of inducible nitric oxide synthase (iNOS) plays an important role in systemic or local, acute or chronic inflammation such as septic shock and rheumatoid arthritis [4, 5].

A study on the cardiovascular effects of DeHE, Evo and Rut found that Rut produced a full NO-dependent vasodilatation whereas Evo and DeHE produced a partially endothelium-dependent effect at 50% and 10% respectively [6]. Apart from endothelium dependence, alpha 1-adrenoceptor blockade, K+ channel activation and Ca2+ channel blockade were also involved in the vasorelaxant effect of DeHE [7]. Coupled with influx of extracellular calcium, Rut produced the endothelium-dependent vasorelaxant effect by activation of endothelium NOS and release of NO without pertussis toxin-sensitive Gi protein and other G proteins or phospholipase C activation being involved [8]. Another study using the whole-cell patch-clamp method found that Rut inhibited the L-type voltage-dependent calcium channels of rat vascular smooth muscle cells and increased NO release through opening of non-voltage-dependent calcium channels in the endothelial cells [9, 10].

In other smooth muscles, Evo was shown to possess a potent corporal relaxing effect attributed to endothelium-independent properties and was tested as a potential agent for the treatment of erectile dysfunction in aged animals [11].

DeHE was found to inhibit NO production by interfering not only with the priming signal initiated by interferon-gamma but also with iNOS synthesis while Evo affected the former only [12]. Ethanol extract of Evodia rutaecarpa dose-dependently prevented the circulation failure, vascular hyporeactivity to phenylephrine, liver dysfunction and reduced the NO over-production in plasma in lipopolysaccharide (LPS)-induced endotoxaemic rats [13]. Evodia rutaecarpa ethanol extract exhibited potent antioxidative effects in neutrophils and that in microglial cells Evodia rutaecarpa ethanol extract, DeHE, Evo and Rut all inhibited the LPS-induced NO production and iNOS expression [14].

Effects on nuclear factor kappa B (NF-kappa B), cyclooxygenase (COX), 5-lipoxygenase(5-LO), prostaglandins (PG), serotonin (5-HT), interleukins (IL), tumor necrosis factor-alpha (TNF-α) and LIGHT

COX and LO are enzynes involved in the metabolism arachidonic acid, thus formation of PG, IL, and other metabolites which related to inflammation [2].

A study found that Evo and Rut strongly inhibited PGE2 synthesis in LPS-treated RAW 264.7 cells and that Evo but not Rut inhibited COX-2 induction and NF-kappa B activation. Goshuyuamide ||, another Evodia rutaecarpa active component, inhibited 5-LO, thereby reducing leukotriene (LT) synthesis; however, these three compounds did not inhibit iNOS mediated NO production from cells up to 50 μM [15]. Another study reported that DeHE inhibited LPS-induced iNOS and COX-2 and their mRNAs expression in RAW 264.7 cells, probably through the suppression of NF-kappa B activation in the transcriptional level [16]. Evo was found to inhibit hypoxia-induced inflammatory response by repressing not only COX-2, COX-2 mRNA and iNOS expression but also PGE2 release in a concentration-dependent manner in RAW264.7 cells under hypoxia condition, mediated via dephosphorylation of the serine/threonine kinases Akt and p70S6 kinase regulating the translation process of hypoxia-inducible factor-1 alpha by Evo [17]. A study demonstrated that Rut is a new class of COX-2 inhibitor partially contributing its in vivo anti-inflammatory activities on lamda-carrageenan induced paw edema in rats [18].

Wuzhuyu Tang (WT), a Chinese medicine formula for migraine treatment, is composed of Evodia fruit, Ginger, Ginseng, and Jujube. A study on WT reported regulatory effects of various components in WT on tryptophan hydroxylase 2 (TPH2, the rate limiting enzyme for 5-HT biosynthesis in brain) promoter, suggesting that the effects of WT on migraine could be due to its stimulating effects on TPH2 promoter and promotion of the 5-HT synthesis and release in the brain [19].

In human mononuclear cells, 10% to 30% of Evodia rutaecarpa extracts were found to stimulate the secretion of IL-1 beta, IL-6, TNF-α and granulocyte-macrophage colony-stimulating factor; however, more than 40% of Evodia rutaecarpa extract lost its stimulating effect. Evodia rutaecarpa extract showed better stimulating effect when reacted with mononuclear cell for 18 or 24 hours than one or three hours [20, 21].

Homologous to Lymphotoxin, exhibits inducible expression, competes with Herpes Simplex Virus Glycoprotein D for binding to Herpes Virus entry Mediator (HVEM), a receptor on T lymphocytes (LIGHT) showed inducible expression and acted as a new player in the atherogenesis [22]. Evo and Rut decreased LIGHT-induced production of ROS, IL-8, monocyte chemoattractant protein-1, TNF-α, IL-6, and the expression of chemokine receptor (CCR) 1, CCR2 and intracellular adhesion molecule 1 and the phosphorylation of extracellular-signal-regulated kinases (ERK) 1/2 and p38 mitogen-activated protein kinase (MAPK) via decreasing ROS production and NADPH oxidase activation. Evo and Rut were considered as potential anti-atherosclerosis agents [23].

Capsaicin-like effects

Used as an analgesic, capsaicin, the major bioactive component of Capsicum frutescens L., is a vanilloid receptor agonist [24]. Capsaicin-sensitive sensory neurons are nociceptive neurons that release calcitonin gene-related peptide (CGRP) on activation. Capsaicin-sensitive sensory neurons are rich in transient receptor potential channel vanilloid type 1 (TRPV1) which plays a fundamental role in pain and involves in the protective effects on cardiovascular and gastrointestinal systems. A study found that TRPV1 could be activated by endogenous cannabinoids (anandamide, N-archidonoyl dopamine, N-oleoyldopamine) or by exogenous agonists such as capsaicin, Evo and Rut which in turn stimulated the CGRP relaease [25].

An earlier study found that oral administration of ethanol extract of Evodia rutaecarpa to mice reduced the acetic acid induced abdominal stretch [26]. Another study confirmed that Evo and Rut were partially responsible for the analgesic effects [27]. Limonin from Evodia rutaecarpa was also found to be analgesic [28].

Evo possesses vanilloid receptor agonistic activities comparable to capsaicin in guinea-pig isolated bronchus [29] and atria [30], and suppresses acetic acid-induced writhing by desensitizing visceral sensory nerves [31]. A study found that Evo was an agonist for the vanilloid receptor TRPV1 in rat, about 3-19 fold less potent than capsaicin [32]. Moreover, Evo was found to protect bovine serum albumin induced guinea-pig cardiac anaphylaxis by stimulation of CGRP release [33] and exert protection against myocardial ischemia-reperfusion injury in rats by activation of vanilloid receptors to stimulate the CGRP release [34].

Rut did not demonstrate bronchoconstrictive effects in guinea-pig isolated bronchus [29]. Rut increased the CGRP and decreased TNF-α with significant improvement of cardiac function and inhibition of the sinus tachycardia in antigen induced cardiac anaphylactic injury of guinea-pig hearts [35]. Rut was also found to release CGRP to inhibit vasoconstriction induced by anaphylaxis in guinea-pigs [36]. Similarly, the cardio-protective effect of Rut on myocardial ischemia-reperfusion injury was caused by vanilloid receptor activation to evoke CGRP release in normal [37] or spontaneously hypertensive rats (SHR) [38]. Rut inhibited hypoxia/reoxygenation induced apoptosis in primary rat hippocampal neurons via TRPV1-(Ca2+)i-dependent and phosphoinositide 3-kinase (PI3K)/Akt signaling pathway [39]. Furthermore, the protective effects of Rut on acetylsalicylic acid and stress-induced gastric mucosa injury were related to stimulation of endogenous CGRP release via activation of vanilloid receptor [40]. Rut also protected the gastric mucosa against injury induced by ethanol via stimulating the release of CGRP to attenuate ethanol-induced elevation of asymmetric dimethylarginine levels [41].

A review article reported that CGRP played an important role in the initiation, progression and maintenance of hypertension and that in contrast the increase in CGRP levels or the enhancement of vascular sensitivity response to CGRP served as a beneficial compensatory depressor role in the development of hypertension [42]. Furthermore, there are therapeutic possibilities of CGRP in hypertension [43]. Effects of Rut on cardiovascular system were reported to act through the release of CGRP, including the depressor and vasodilator [44], the hypotensive effects in the phenol-induced hypertensive rats [45], the hypotensive effects and reduction of mesenteric artery hypertrophy in removascular hypertensive rats [46] and the hypotensive and anti-platelet effects (inhibits the relaease of platelet-derived tissue factor) in SHR [47]. Effects of Rut to lower systolic blood pressure and reverse mesenteric artery remodeling were found to be related to increased expression of prolylcarboxypeptidase in the circulation and small arteries in renovascular hypertensive rats [48]. However, Rut inhibited platelet aggregation in human platelet-rich plasma by inhibiting TXA2 formation, phosphoinositide breakdown and phospholipase C [49–51]. CGRP could work as an endogenous protective substance to counteract endothelial progenitor cells senescence in hypertension and the accelerated endothelial progenitor cells senescence in hypertension is related to the reduction of CGRP while Rut could reverse endothelial progenitor cell senescence along with an elevation in CGRP production in SHR and reverse angiotensin II-induced CGRP mRNA expression in endothelial progenitor cells [52].

Rut solid dispersion significantly increased the blood concentration, accompanied by significant hypotensive effects in SHR in a dose-dependent manner [53]. The 14-N atom of Rut might be the key site for the activity and simple substitute in indole-ring or quinazoline-ring would not enhance the vasodilator effects unless in a proper position and with a proper group [54].

Effects on Alzheimer's disease

Alzheimer's disease, impairment of memory and cognitive ability caused by the loss of hippocampal and cortical neurons, is related to accumulations of beta-amyloid [55] and disproportionate deficiency of acetylcholine [56]. Treatment for Alzheimer's disease includes transmitter replacement therapies, anti-oxidants, neuronal calcium channel blockers, anti-apoptotic agents, anti-inflammatory agents, estrogens, nerve growth factors and drugs that inhibit secretase activity and prevent or block beta-amyloid formation in the brain [57, 58].

DeHE HCl was found to increase the cerebral blood flow in anesthetized cats [59]. In a screening of 29 natural products, Evodia rutaecarpa demonstrated a strong inhibitory effect on acetylcholinesterase in vitro and an anti-amnesic effect in vivo. The active component of Evodia rutaecarpa was identified as DeHE HCl [60]. A study suggested that DeHE HCl might be an effective drug not only for the Alzheimer's disease type but also for the vascular type of dementia [61]. Our study reported that DeHE pretreatment attenuated intracerebroventricular administration of beta-amyloid peptide (25-35) and intraperitoneal administration of scopolamine induced amnesia in mice [62]. Furthermore, pre-administration of DeHE via vena caudalis for one week effectively improved the Wortmannin and GF-109 203X (WT/GFX) induced spatial memory retention impairment of rats, antagonized tau hyperphosphorylation at multiple Alzheimer's disease site and arrested the overactivation of glycogen synthase kinase-3 induced by WT/GFX [63]. DeHE did not cause any serious adverse effects at the dose levels in the experimental animals [64]. Some novel inhibitors of acetyl- and butyrylcholinesterase derived from DeHE and Rut were also reported [65].

DeHE HCl could provide long-lasting facilitation of synaptic transmission that depended on the activation of both the muscarinic and N-methyl-D-aspartate receptors in the Cornu Ammonis area 1 region of rat hippocampal slices on the electrical stimulation evoked field excitatory postsynaptic potentials [66]; however, chronic exposure to DeHE concentration-dependently inhibited glutamate uptake and release in the cultured cerebellar cells [67]. In rat brain slices, DeHE attenuated calyculin A, a protein phosphatase (PP)-2A and PP-1inhibitor, and induced Alzheimer's disease-like tau hyperphosphorylation [68]. Evodia officinalis extract demonstrated the most protective effects among 10 kinds of plant extracts against the carboxy-terminal 105 amino acid fragments of amyloid precursor protein induced neurotoxicity [69].

Themoregulative effects, anti-obese, anti-adipogenic and anti-diabetic effects

Among the Evodia fruit alkaloids(hydroxy-Evo, Evo, Rut and evocarpine), Evo prevented the chlorpromazine induced decrease of body temperature in rats [70]; however, intraperitoneal injection of DeHE or Evo caused a dose-related hypothermia in afebrile rats at 20°C. Moreover, both DeHE and Evo attenuated the febrile response induced by intrahypothalamic injection of exogenous pyrogen in rats [71].

Evo was found to mimic the capsaicin-like anti-obese activities [72]; however, in uncoupling protein-1 (UCP1)-knockout mice, Evo triggered a UCP1-independent mechanism to prevent diet-induced obesity [73]. Furthermore, the anti-adipogenic effects of Evo were not blocked by the specific TRPV1 antagonist capsazepine in 3T3-L1 preadipocytes whereas Evo stimulated the phosphorylation of epidermal growth factor receptor (EGFR), protein kinase C alpha and ERK, all of which were reduced by EGFR inhibitor [74]. Evo inhibited human white preadipocyte differentiation accompanied by up-regulation of both GATA binding protein 2 and 3 mRNA and protein expression [75]. Evo also inhibited the adipocyte differentiation of 3T3-21 and C3H1OT1/2 cells and inhibited the obesity in db/db mice. Evo improved the undesirable effects of rosiglitazone, including adipogenesis, body weight gain and hepatotoxicity, while preserving its blood-glucose-lowering effects [76].

Orexin [77] and melanin-concentrating hormone (MCH) [78] regulate food intake, arousal and motivated behavior in lateral hypothalamic area. In fed and in hyperinsulinemic obese mice, insulin signaling led to nuclear exclusion of forkhead transcription factor Foxa2 and reduces expression of MCH and orexin [79]. As constitutive and conditional activation of Foxa2 in the brain increased neuronal MCH and orexin expression, it was suggested that pharmacological inhibition of Foxa2 phosphorylation might improve levels of physical activity, overall health and longevity [80].

Administration of Evo to juvenile rats decreased rate of food intake and body weight increase, reduced orexigenic neuropeptide Y (NPY) and agouti-gene related protein mRNA levels and NPY peptide level but increased the circulating level of leptin [81]. In high-fat-diet-induced (C57BL/6) and leptin-deficient (ob/ob) obese mice, Rut ameliorated obesity by inhibiting food intake [82].

Aldose reductase inhibitors are potential drugs for treating diabetic complications [83]. Rhetsinine from Evodia rutaecarpa inhibited aldose reductase activity and was considered potentially useful in the treatment of diabetic complications [84].

GI effects

One of the most important clinic application of Evodia Fructus is treatment of discomfort or chill of stomach.

Water extract of Evodia rutaecarpa inhibited the intestinal transit (anti-transit effect) and castor oil-induced diarrhea in mice [85]; however, the water extract of Evodia rutaecarpa protected the ethanol-induced rat gastric lesions [86, 87]. As mentioned earlier, the protective effects of Rut on acetylsalicylic acid, stress and ethanol-induced gastric mucosa injury were related to stimulation of endogenous CGRP release via activation of vanilloid receptor [40, 41].

Evo inhibited both gastric emptying and gastrointestinal transit in male rats via a mechanism involving cholecystokinin (CCK) release and CCK1 receptor activation [88]. DeHE HCl also exhibited anti-transit effect [64].

Anti-emetic effects of the ethanol extracts of WT were demonstrated via 5-HT and histamine receptors [89].

Dermatological applications

Among 100 herbal extracts screened for anti-oxidant activity and free radical scavenging activity, Evodia officinalis was one of the 14 potential sources of anti-oxidants [90].

Evodia rutaecarpa, Evo and Rut inhibited immunoglobulin E (IgE)-antigen complex-induced passive cutaneous anaphylaxis reaction and compound 48/80-induced scratching behaviors in mice. Evo and Rut inhibited IgE-antigen complex-induced TNF-α and IL-4 protein expression in RB2-2H3 cells, suggesting that Evo and Rut could be used for the treatment of atopic dermatitis and rhinitis [91].

Extract of Evodia officinalis showed a potent inhibitory effect on ultraviolet B (UVB) induced matrix metalloproteinase (MMP)-1 production in human skin fibroblasts [92]. A defined mixture composed of Rut, DeHE and evodin was shown to inhibit UVB-induced PGE2 released by keratinocytes in vitro and methyl nicotinate-induced erythema in human skin [93]. Rut also inhibited ultraviolet A (UVA) induced ROS generation and suppressed UVA or H2O2-induced increase in the expression of MMP-2 and MMP-9 in HaCaT human keratinocytes [94].

Anti-anoxic effects

Extract of Evodia rutaecarpa exerted an antianoxic effect in the KCN-induced anoxia model in mice [95]. Cholinergic mechanism was found to be involved in the antianoxic action of Evo which is an active component of Evodia rutaecarpa [96].

Anti-infectious effects

Anti-infectious, or chemotherapeutic, agents for the treatment of protozoal, helminth, and microbial diseases are not anti-inflammatory agents and different from the pharmacodynamic agents which affected the physiological, biochemical, or immunological function of host. The need to develop new chemotherapeutic agent for the widespred antibiotic-resistant pathogens are very important but less success.

Among 300 herbal extracts screened for the anti-hepatitis B surface antigen capability, Evodia rutaecarpa was one of the ten effective herbs [97]. Atanine (3-dimethylallyl-4-methoxy-2-quinolone) was found as an active anthelmintic compound in Evodia rutaecarpa [98]. Six quinolone alkaloids (ie evocarpine, 1-methyl-2-[(4Z,7Z)-4,7-tridecadienyl]-4(1H)-quinolone, 1-methyl-2-[(6Z,9Z)-6.9-pentadecadienyl]-4(1H)-quinolone, 1-methyl-2-undecyl-4(1H)-quinolone, dihydroevocarpine and 1-methyl-2-pentadecyl-4(1H)-quinolone) isolated from Evodia rutaecarpa showed potent anti-Helicobacter pylori activity [99]. Two alkyl quinolone compounds, namely 1-methyl-2-[(Z)-8-tridecenyl]-4-(1H)-quinolone and 1-methyl-2-[(Z)-7-tridecenyl]-4-(1H)-quinolone, from Evodia rutaecarpa were anti-bacterial agents highly selective in vitro against H. pylori and almost non-active against other intestinal pathogens [100]. In vivo studies on H. pylori infected Mongolian gerbils demonstrated that alkyl methyl quinolone compounds from Evodia rutaecarpa decreased the number of H. pylori and inhibited the H. pylori respiration [101, 102].

Three synthesized 2-alkenyl-4(1H)-quinolone compounds, one of which is found in Evodia rutaecarpa demonstrated vasodilating and antibacterial effects [103]. Evodia rutaecarpa extract was reported to possess bactericidal activity against gram-positive cocci, P aeruginose and C albicans [104]. Similarly, extracts of Evodia elleryana leaves, stem wood, stem bark, root wood, root bark and petrol, dichloromethane, ethyl acetate partition fractions showed a broad spectrum of anti-bacterial activity [105]. Extract of Evodia elleryana bark also inhibited Mycobacterium tuberculosis [106]. Ethyl acetate extract of Evodia fatraina stem bark showed moderate in vitro anti-malarial activity against Plasmodium falciparum while the ethanol extract exhibited 65% suppression of Plasmodium berghei in mice [107].

Conclusion

Stimulation of CGRP release may partially explain the analgesic, cardiovascular and gastrointestinal protective, anti-obese activities of Evodia rutaecarpa and its major bioactive components. Other direct actions by the active components of Evodia rutaecarpa on different targets may account for various pharmacological effects of Evodia rutaecarpa.

Abbreviations

5-HT:

5-hydroxytryptamine, serotonin

5-LO:

5-lipoxygenase

CCK:

cholecystokinin

CCR:

chemokine receptor

CGRP:

calcitonin gene-related peptide

COX:

cycloxygenase

DeHE:

dehydroevodiamine

EGFR:

epidermal growth factor receptor

ERK:

extracellular-signal-regulated kinases

Evo:

evodiamine

HSV:

herpes simplex virus

IgE:

immunoglobulin E

IL:

interleukin

iNOS:

inducible nitric oxide synthase

LIGHT:

Homologous to Lymphotoxin, exhibits inducible expression, competes with Herpes Simplex Virus Glycoprotein D for binding to Herpes Virus entry Mediator (HVEM), a receptor on T lymphocytes

LPS:

lipopolysaccharide

LT:

leukotriene

MCH:

melanin-concentrating hormone

MMP:

matrix metalloproteinase

NF-kappa B:

nuclear factor kappa B

NO:

nitric oxide

NOS:

nitric oxide synthase

NPY:

neuropeptide Y

PG:

prostaglandins

PP:

protein phosphatase

ROS:

reactive oxgen species

Rut:

rutaecarpine

SHR:

spontaneously hypertensive rats

TNF-α:

tumor necrosis factor-alpha

TPH2:

tryptophan hydroxylase 2

TRPV1:

transient receptor potential channel vanilloid type 1

TX:

thromboxanes

UCP1:

uncoupling protein-1

UVA:

ultraviolet A radiation

UVB:

ultraviolet B radiation

WT:

Wuzhuyu Tang

WT/GFX:

Wortmannin and GF-109 203X

References

  1. Angus DC: Epidemiology of severe sepsis in the United States: Analysis of incidence, outcome, and associated costs of care. Crit Care Med. 2001, 29: 1303-1310. 10.1097/00003246-200107000-00002.

    Article  CAS  PubMed  Google Scholar 

  2. Smyth EM, Burke A, Fitzgerald GA: Lipid-derived autacoids: Eicosanoids and platelet-activating factor. Goodman and Gilman's the Pharmacological Basis of Therapeutics. Edited by: Brunton LL, Lazo JS, Parker KL. 2006, New York: McGraw-Hill Med Pub Div, 653-670. 11

    Google Scholar 

  3. Elliott MJ, Chitham K, Theoharis LT: The effects of plant flavonoids on mammalian cells. Implications for inflammation, heart disease, and cancer. Pharmacol Rev. 2000, 52: 673-751.

    Google Scholar 

  4. Moncada S, Palmer RMJ, Higgs EA: Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol Rev. 1991, 43: 109-142.

    CAS  PubMed  Google Scholar 

  5. Stefanovic Racic M, Stadler J, Evans CH: Nitric oxide and arthritis. Arthritis Rheum. 1993, 36 (8): 1036-1044. 10.1002/art.1780360803.

    Article  CAS  PubMed  Google Scholar 

  6. Chiou WF, Liao JF, Chen CF: Comparative study of the vasodilatory effects of three quinazoline alkaloids isolated from Evodia rutaecarpa. J Nat Prod. 1996, 59 (4): 374-378. 10.1021/np960161+.

    Article  CAS  PubMed  Google Scholar 

  7. Chiou WF, Liao JF, Shum AY, Chen CF: Mechanisms of vasorelaxant effect of dehydroevodiamine: A bioactive isoquinazolinocarboline alkaloid of plant origin. J Cardiovasc Pharmacol. 1996, 27 (6): 845-853. 10.1097/00005344-199606000-00012.

    Article  CAS  PubMed  Google Scholar 

  8. Chiou WF, Shum AY, Liao JF, Chen CF: Studies of the cellular mechanisms underlying the vasorelaxant effects of rutaecarpine, a bioactive component extracted from a herbal drug. J Cardiovasc Pharmacol. 1997, 29 (4): 490-498. 10.1097/00005344-199704000-00010.

    Article  CAS  PubMed  Google Scholar 

  9. Wang GJ, Shan J, Pang PKT, Yang MCM, Chou CJ, Chen CF: The vasorelaxing action of rutaecarpine: Direct paradoxical effects of intracellular calcium concentration of vascular smooth muscle and endothelial cells. J Pharmacol Exp Ther. 1996, 276: 1016-1021.

    CAS  PubMed  Google Scholar 

  10. Wang GJ, Wu XC, Chen CF, Lin LC, Huang YT, Shan J, Pang PKT: Vasorelaxing action of rutaecarpine: Effects of rutaecarpine on calcium channel activities in vascular endothelial and smooth muscle cells. J Pharmacol Exp Ther. 1999, 289: 1237-1244.

    CAS  PubMed  Google Scholar 

  11. Chiou WF, Chen CF: Pharmacological profile of evodiamine in isolated rabbit corpus cavernosum. Eur J Pharmacol. 2002, 446: 151-159. 10.1016/S0014-2999(02)01762-4.

    Article  CAS  PubMed  Google Scholar 

  12. Chiou WF, Sung YJ, Liao JF, Shum AY, Chen CF: Inhibitory effect of dehydroevodiamine and evodiamine on nitric oxide production in cultured murine macrophages. J Nat Prod. 1997, 60 (7): 708-711. 10.1021/np960495z.

    Article  CAS  PubMed  Google Scholar 

  13. Chiou WF, Ko HC, Chen CF, Chou CJ: Evodia rutaecarpa, protects against circulation failure and organ dysfunction in endotoxaemic rats through modulating nitric oxide release. J Pharm Pharmacol. 2002, 54: 1399-1405. 10.1211/002235702760345491.

    Article  CAS  PubMed  Google Scholar 

  14. Ko HC, Wang YH, Liou KT, Chen CM, Chen CH, Wang WY, Chang S, Hou YC, Chen KT, Chen CF, Shen YC: Anti-inflammatory effects and mechanisms of the ethanol extract of Evodia rutaecarpa and its bioactive components on neutrophils and microglial cells. Eur J Pharmacol. 2007, 555 (2-3): 211-217. 10.1016/j.ejphar.2006.10.002.

    Article  CAS  PubMed  Google Scholar 

  15. Choi YH, Shin EM, Kim YS, Cai XF, Lee JJ, Kim HP: Anti-inflammatory principles from the fruits of Evodia rutaecarpa and their cellular action mechanisms. Arch Pharm Res. 2006, 29 (4): 293-297. 10.1007/BF02968573.

    Article  CAS  PubMed  Google Scholar 

  16. Noh EJ, Ahn KS, Shin EM, Jung SH, Kim YS: Inhibition of lipopolysaccharide-induced iNOS and COX-2 expression by dehydroevodiamine through suppression of NF-Kappa B activation in RAW 264.7 macrophages. Life Sci. 2006, 79: 695-701. 10.1016/j.lfs.2006.02.020.

    Article  CAS  PubMed  Google Scholar 

  17. Liu YN, Pan SL, Liao CH, Huang DY, Guh JH, Peng CY, Chang YL, Teng CM: Evodiamine represses hypoxia-induced inflammatory proteins expression and hypoxia-inducible factor 1 alpha accumulation in RAW 264.7. Shock. 2009, 32 (3): 263-269. 10.1097/SHK.0b013e31819940cb.

    Article  PubMed  CAS  Google Scholar 

  18. Moon TC, Murakami M, Kudo I, Son KH, Kim HP, Kang SS, Chang HW: A new class of COX-2 inhibitor, rutaecarpine from Evodia rutaecarpa. Inflamm Res. 1999, 48 (12): 621-625. 10.1007/s000110050512.

    Article  CAS  PubMed  Google Scholar 

  19. Wang Y, Lei F, Wang S, Hu J, Zhan H, Xing D, Du L: Regulatory effects of Wuzhuyutang (Evodia prescription) and its consisting herbs on TPH2 promoter. Zhongguo Zhong Yao Za Zhi. 2009, 34 (17): 2261-2264.

    PubMed  Google Scholar 

  20. Chang CP, Chang JY, Wang FY, Tseng J, Chang JG: The effect of Evodia rutaecarpa extract on cytokine secretion by human mononuclear cells in vitro. Am J Chin Med. 1995, 23 (2): 173-180. 10.1142/S0192415X95000237.

    Article  CAS  PubMed  Google Scholar 

  21. Chang JY, Yang TY, Chang CP, Chang JG: The effect of 'chi-han (hot nature)' Chinese herbs on the secretion of IL-1 beta and TNF-alpha by mononuclear cells. Kaohsiung J Med Sci. 1996, 12 (1): 18-24.

    CAS  PubMed  Google Scholar 

  22. Bobik A, Kalinina N: Tumor necrosis factor receptor and ligand superfamily family members TNFRSF 14 and LIGHT: New players in human atherogenesis. Arterioscler Thromb Vasc Biol. 2001, 21: 1873-1875.

    CAS  PubMed  Google Scholar 

  23. Heo SK, Yun HJ, Yi HS, Noh EK, Park SD: Evodiamine and rutaecarpine inhibit migration by LIGHT via suppression of NADPH oxidase activation. J Cell Biochem. 2009, 107 (1): 123-133. 10.1002/jcb.22109.

    Article  CAS  PubMed  Google Scholar 

  24. Caterina M, Julius D: The vanilloid receptor: a molecular gateway to the pain pathway. Annu Rev Neurosci. 2001, 24: 487-517. 10.1146/annurev.neuro.24.1.487.

    Article  CAS  PubMed  Google Scholar 

  25. Peng L, Li YJ: The vanilloid receptor TRPV-1: Role in cardiovascular and gastrointestinal protection. Eur J Pharmacol. 2010, 627 (1-3): 1-7. 10.1016/j.ejphar.2009.10.053.

    Article  CAS  PubMed  Google Scholar 

  26. Chow SY, Chen SM, Yang CM: Pharmacological studies on Chinese herbs (3): Analgesic effects of 27 Chinese drugs in mice. J Formosan Med Asso. 1976, 75: 349-357.

    CAS  Google Scholar 

  27. Matsuda H, Wu TX, Tanaka T, Linuma M, Kubo M: Antinociceptive activities of 70% methanol extract of Evodiae Fructus (fruit of Evodia rutaecarpa var. bodinieri ) and its alkaloidal components. Biol Pharm Bull. 1997, 20 (3): 243-248.

    Article  CAS  PubMed  Google Scholar 

  28. Matsuda H, Yoshikawa M, Linuma M, Kobo M: Antinociceptive and anti-inflammatory activities of limonin isolated from the fruits of Evodia rutaecarpa var. bodinieri. Planta Med. 1998, 64 (4): 339-342. 10.1055/s-2006-957447.

    Article  CAS  PubMed  Google Scholar 

  29. Kobayashi Y, Nakano Y, Hoshikuma KM, Yokoo Y, Kamiya T: The bronchoconstrictive action of evodiamine, an indoloquinazoline alkaloid isolated from the fruits of Evodia rutaecarpa, on guinea-pig isolated bronchus: Possible involvement on vanilloid receptors. Planta Med. 2000, 66 (6): 526-530. 10.1055/s-2000-8615.

    Article  CAS  PubMed  Google Scholar 

  30. Kobayashi Y, Hoshikuma K, Nakano Y, Yokoo Y, Kamiya T: The positive inotropic and chronotropic effects of evodiamine and rutaecarpine, indoloquinazoline alkaloids isolated from the fruits of Evodia rutaecarpa, on the guinea-pig isolated right atria: Possible involvement of vanilloid receptors. Planta Med. 2001, 67 (3): 244-248. 10.1055/s-2001-12008.

    Article  CAS  PubMed  Google Scholar 

  31. Kobayashi Y: The nociceptive and anti-nociceptive effects of evodiamine from fruits of Evodia rutaecarpa in mice. Planta Med. 2003, 69 (5): 425-428. 10.1055/s-2003-39701.

    Article  CAS  PubMed  Google Scholar 

  32. Pearce LV, Petukhov PA, Szabo T, Kedei N, Bizik F, Kozikowski AP, Blumberg PM: Evodiamine functions as an agonist for the vanilloid receptor TRPVI. Org Biomol Chem. 2004, 2 (16): 2281-2286. 10.1039/b404506h.

    Article  CAS  PubMed  Google Scholar 

  33. Rang WQ, Du YH, Hu CP, Ye F, Tan GS, Deng HW, Li YJ: Protective effects of calcitonin gene-related peptide-mediated evodiamine on guinea-pig cardiac anaphylaxis. Naunyn Schmiedebergs Arch Pharmacol. 2003, 367 (3): 306-311. 10.1007/s00210-002-0677-0.

    Article  CAS  PubMed  Google Scholar 

  34. Rang WQ, Du YH, Hu CP, Ye F, Xu KP, Peng J, Deng HW, Li YJ: Protective effects of evodiamine on myocardial ischemia-reperfusion injury in rats. Planta Med. 2004, 70 (12): 1140-1143. 10.1055/s-2004-835841.

    Article  CAS  PubMed  Google Scholar 

  35. Yi HH, Rang WQ, Deng PY, Hu CP, Liu GZ, Tan GS, Xu KP, Li YJ: Protective effects of rutaecarpine in cardiac anaphylactic injury is mediated by CGRP. Plant Med. 2004, 70 (12): 1135-1139. 10.1055/s-2004-835840.

    Article  CAS  Google Scholar 

  36. Yu J, Tan GS, Deng PY, Xu KP, Hu CP, Li YJ: Involvement of CGRP in the inhibitory effect of rutaecarpine on vasoconstriction induced by anaphylaxis in guinea pig. Regul Pept. 2005, 125 (1-3): 93-97. 10.1016/j.regpep.2004.08.001.

    Article  CAS  PubMed  Google Scholar 

  37. Hu CP, Xiao L, Deng HW, Li YJ: The cardioprotection of rutaecarpine is mediated by endogenous calcitonin release-gene peptide through activation of vanilloid receptors in guinea-pig hearts. Planta Med. 2002, 68 (8): 705-709. 10.1055/s-2002-33794.

    Article  CAS  PubMed  Google Scholar 

  38. Li D, Zhang XL, Chen L, Yang Z, Deng HW, Peng L, Li YJ: Calcitonin gene-related peptide mediates the cardioprotective effects of rutaecarpine against ischaemia-perfusion injury in spontaneously hypertensive rats. Clin Exp Pharmacol Physiol. 2009, 36 (7): 662-667. 10.1111/j.1440-1681.2008.05136.x.

    Article  CAS  PubMed  Google Scholar 

  39. Dai Z, Xiao J, Lin SY, Cui L, Hu GY, Jiang DJ: Rutaecarpine inhibits hypoxia/reoxygenation-induced apoptosis in rat hippocampal neurons. Neuropharmacology. 2008, 55 (8): 1307-1312. 10.1016/j.neuropharm.2008.08.030.

    Article  CAS  PubMed  Google Scholar 

  40. Wang L, Hu CP, Deng PY, Shen SS, Zhu HQ, Ding JS, Tan GS, Li YJ: The protective effects of rutaecarpine on gastric mucosa injury in rats. Planta Med. 2005, 71 (5): 416-419. 10.1055/s-2005-864135.

    Article  CAS  PubMed  Google Scholar 

  41. Liu YZ, Zhou Y, Li D, Wang L, Hu GY, Peng J, Li YJ: Reduction of asymmetric dimethylarginine in the protective effects of rutaecarpine on gastric mucosal injury. Can J Physiol Pharmacol. 2008, 86 (10): 675-681. 10.1139/Y08-073.

    Article  CAS  PubMed  Google Scholar 

  42. Deng PY, Li YJ: Calcitonin gene-related peptide and hypertension. Peptides. 2005, 26 (9): 1675-1685. 10.1016/j.peptides.2005.02.002.

    Article  CAS  Google Scholar 

  43. Márquez-Rodas L, Longo F, Rothlin RP, Balfagón G: Phathophysiology and the therapeutic possibilities of calcitonin gene-related peptide in hypertension. J Physiol Biochem. 2006, 62 (1): 45-46.

    Article  PubMed  Google Scholar 

  44. Hu CP, Xiao L, Deng HW, Li YJ: The depressor and vasodilator effects of rutaecarpine are mediated by calcitonin gene-related peptide. Planta Med. 2003, 69 (2): 125-129. 10.1055/s-2003-37703.

    Article  CAS  PubMed  Google Scholar 

  45. Deng PY, Ye F, Cai WJ, Tan GS, Hu CP, Deng HW, Li YJ: Stimulation of calcitonin gene-related peptide synthesis and release: mechanisms for a novel antihypertensive drug, rutaecarpine. J Hypertens. 2004, 22 (9): 1819-1829. 10.1097/00004872-200409000-00028.

    Article  CAS  PubMed  Google Scholar 

  46. Qin XP, Zeng SY, Li D, Chen QQ, Luo D, Zhang Z, Hu GY, Deng HW, Li YJ: Calcitonin gene-related peptide-mediated depressor effect and inhibiting vascular hypertrophy of rutaecarpine in renovascular hypertensive rats. J Cardiovasc Pharmacol. 2007, 50 (6): 654-659. 10.1097/FJC.0b013e3181579e7e.

    Article  CAS  PubMed  Google Scholar 

  47. Li D, Peng J, Xin HY, Luo D, Zhang YS, Zhou Z, Jiang DJ, Deng HW, Li YJ: Calcitonin gene-related peptide-mediated antihypertensive and anti-platelet effects by rutaecarpine in spontaneously hypertensive rats. Peptides. 2008, 29 (10): 1781-1788. 10.1016/j.peptides.2008.06.010.

    Article  CAS  PubMed  Google Scholar 

  48. Qin XP, Zeng SY, Tian HH, Deng SX, Ren JF, Zheng YB, Li D, Li YJ, Liao DF, Chen SY: Involvement of prolylcarboxypeptidase in the effect of rutaecarpine on the regression of mesenteric artery hypertrophy in renovascular hypertensive rats. Clin Exp Pharmacol Physiol. 2009, 36 (3): 319-324. 10.1111/j.1440-1681.2008.05079.x.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  49. Sheu JR, Hung WC, Lee YM, Yen MH: Mechanism of inhibition of platelet aggregation by rutaecarpine, an alkaloid isolated from Evodia rutaecarpa. Eur J Pharmacol. 1996, 318 (2-3): 469-475. 10.1016/S0014-2999(96)00789-3.

    Article  CAS  PubMed  Google Scholar 

  50. Sheu JR, Kan TC, Hung WC, Su CH, Lin CH, Lee YM, Yen MH: The antiplatelet activity of rutaecarpine, an alkaloid isolated from Evodia rutaecarpa, is mediated through inhibition of phospholipase C. Thromb Res. 1998, 92 (2): 53-64. 10.1016/S0049-3848(98)00112-1.

    Article  CAS  PubMed  Google Scholar 

  51. Sheu JR, Hung WC, Wu CH, Lee YM, Yen MH: Antithrombotic effect of rutaecarpine, an alkaloid isolated from Evodia rutaecarpa, on platelet plug formation in vivo experiments. Br J Pharmacol. 2000, 110 (1): 110-115.

    CAS  Google Scholar 

  52. Zhou Z, Peng J, Wang CJ, Li D, Li TT, Hu CP, Chen XP, Li YL: Accelerated senescence of endothelial progenitor cells in hypertension is related to the reduction of calcitonin gene-related peptide. J Hypertens. 2010, 28 (5): 931-939. 10.1097/HJH.0b013e3283399326.

    Article  CAS  PubMed  Google Scholar 

  53. Ding JS, Gao R, Li D, Peng J, Ran LL, Li YL: Solid dispersion of rutaecarpine improved its antihypertensive effect in spontaneously hypertensive rats. Biopharm Drug Dispos. 2008, 29 (9): 495-500. 10.1002/bdd.634.

    Article  CAS  PubMed  Google Scholar 

  54. Chen Z, Hu G, Li D, Chen J, Li Y, Zhou H, Xie Y: Synthesis and vasodilator effects of rutaecarpine analogues which might be involved transient receptor potential vanilloid subfamily, member I (TRPVI). Bioorg Med Chem. 2009, 17 (6): 2351-2359. 10.1016/j.bmc.2009.02.015.

    Article  CAS  PubMed  Google Scholar 

  55. Selkoe DJ, Podlisny MB: Deciphering the genetic basis of Alzheimer's disease. Ann Rev Genomics Hum Genet. 2002, 3: 67-99. 10.1146/annurev.genom.3.022502.103022.

    Article  CAS  Google Scholar 

  56. Perry EK: The cholinergic hypothesis-ten years on. Br Med Bull. 1986, 42: 63-69.

    CAS  PubMed  Google Scholar 

  57. Cumming JL, Vinters HV, Cole GM, Khachaturian ZS: Alzeheimer's disease: Etiologies, pathophysiology, cognitive reserve, and treatment opportunities. Neurology. 1998, 51 (Suppl 1): S2-S17.

    Article  Google Scholar 

  58. Parihar MS, Hemnani T: Alzheimer's disease pathogenesis and therapeutic interventions. J Clin Neurosci. 2004, 11 (5): 456-467. 10.1016/j.jocn.2003.12.007.

    Article  CAS  PubMed  Google Scholar 

  59. Haji A, Momose Y, Takeda R, Nakanishi S, Hiriachi T, Arisawa M: Increased feline cerebral blood flow induced by dehydroevodiamine hydrochloride from Evodia rutaecarpa. J Nat Prod. 1994, 57 (3): 387-389. 10.1021/np50105a009.

    Article  CAS  PubMed  Google Scholar 

  60. Park CH, Kim SH, Choi W, Lee YJ, Kim JS, Kang SS, Suh YH: Novel anticholinesterase and antiamnesic activities of dehydroevodiamine, a constituent of Evodia rutaecarpa. Planta Med. 1996, 62 (5): 405-409. 10.1055/s-2006-957926.

    Article  CAS  PubMed  Google Scholar 

  61. Park CH, Lee YJ, Lee SH, Choi SH, Kim HS, Jeong SJ, Kim SS, Suh YH: Dehydroevodiamine.HCl prevents impairment of learning and memory and neuronal loss in rat models of cognitive disturbance. J Neurochem. 2000, 74 (1): 244-253. 10.1046/j.1471-4159.2000.0740244.x.

    Article  CAS  PubMed  Google Scholar 

  62. Wang HH, Chou CJ, Liao JF, Chen CF: Dehydroevodiamine attenuates beta-amyloid peptide-induced amnesia in mice. Eur J Pharmacol. 2001, 413 (2-3): 221-225. 10.1016/S0014-2999(00)00913-4.

    Article  CAS  PubMed  Google Scholar 

  63. Peng JH, Zhang CE, Wei W, Hong XP, Pang XP, Wang JZ: Dehydroevodiamine attenuates tau hyerphosphorylation and spatial memory deficit induced by activation of glycogen synthase kinase-3 in rats. Neuropharmacology. 2007, 52 (7): 1521-1527. 10.1016/j.neuropharm.2007.02.008.

    Article  CAS  PubMed  Google Scholar 

  64. Park CH, Choi SH, Kim CH, Seo JH, Koo JW, Rah JC, Kim HS, Jeong SJ, Joo WS, Lee YL, Kim YS, Kim MS, Suh YH: General pharmacology of indole [2', 3':3,4]pyridol[2,1-b]quinazolinium-5,7,8,13-tetrahydro-14-methyl-5-oxo-chloride, a new anti-dementia agent. Arzneimittelforschung. 2003, 53 (6): 393-401.

    CAS  PubMed  Google Scholar 

  65. Decker M: Novel inhibitors of acetyl- and butyrylcholinesterase derived from the alkaloids dehydroevodiamine and rutaecarpine. Eur J Med Chem. 2005, 40 (3): 305-313. 10.1016/j.ejmech.2004.12.003.

    Article  CAS  PubMed  Google Scholar 

  66. Park EJ, Suh YH, Kim JY, Choi S, Lee CJ: Long-lasting facilitation by dehydroevodiamine HCI of synaptic responses evoked in the CA1 region of rat hippocampal slices. Neuroreport. 2003, 14 (3): 399-403. 10.1097/00001756-200303030-00020.

    Article  CAS  PubMed  Google Scholar 

  67. Lim DK, Lee YB, Kim HS: Effects of dehydroevodiamine exposure on glutamate release and uptake in the cultured cerebellar cells. Neurochem Res. 2004, 29 (2): 407-411. 10.1023/B:NERE.0000013745.17014.a3.

    Article  CAS  PubMed  Google Scholar 

  68. Fang J, Liu R, Tian Q, Hong XP, Wang SH, Cao FY, Pan XP, Wang JZ: Dehydroevodiamine attenuates calyculin A-induced tau hyperphosphorylation in rat brain slices. Acta Pharmacol Sin. 2007, 28 (11): 1717-1723. 10.1111/j.1745-7254.2007.00655.x.

    Article  CAS  PubMed  Google Scholar 

  69. Kim ST, Kim JD, Lyu YS, Lee MY, Kang HW: Neuroprotective effect of some plant extracts in cultured CT105-induced PC12 cells. Biol Pharm Bull. 2006, 29 (10): 2021-2024. 10.1248/bpb.29.2021.

    Article  CAS  PubMed  Google Scholar 

  70. Kano Y, Zong QN, Komatsu K: Pharmacological properties of galenical preparation, XIV. Body temperature retaining effect of the Chinese traditional medicine, 'goshuyu-to' and component crude drugs. Chem Pharm Bull (Tokyo). 1991, 39 (3): 690-692.

    Article  CAS  Google Scholar 

  71. Tsai TH, Lee TF, Chen CF, Wang LC: Thermoregulatory effects of alkaloids isolated from Wu-Chu-Yu in afebrile and febrile rats. Pharmacol Biochem Behav. 1995, 50 (2): 293-298. 10.1016/0091-3057(94)00317-C.

    Article  CAS  PubMed  Google Scholar 

  72. Kobayashi Y, Nakano Y, Kizaki M, Hoshikuma K, Yokoo Y, Kamiya T: Capsaicin-like anti-obese activities of evodiamine from fruits of Evodia rutaecarpa, a vanilloid receptor agonist. Planta Med. 2001, 67 (7): 628-633. 10.1055/s-2001-17353.

    Article  CAS  PubMed  Google Scholar 

  73. Wang T, Wang Y, Kontani Y, Kobayashi Y, Sato Y, Mori N, Yamashita H: Evodiamine improves diet-induced obesity in a uncoupling proteun-1-independent manner: Involvement of antiadipogenic mechanism and extracellularly regulated kinase/mitogen-activated protein kinase signaling. Endocrinology. 2008, 149 (1): 358-366. 10.1210/en.2007-0467.

    Article  CAS  PubMed  Google Scholar 

  74. Wang T, Wang Y, Yamashita H: Evodiamine inhibits adipogenesis via the EGFR-PKC alpha-ERK signaling pathway. FEBS. 2009, 583 (22): 3655-3659. 10.1016/j.febslet.2009.10.046.

    Article  CAS  Google Scholar 

  75. Hu Y, Fahmy H, Zjawiony JK, Davies GE: Inhibitory effect and transcriptional impact of berberine and evodiamine on human white preadipocyte differentiation. Fitoterapia. 2010, 81 (4): 259-268. 10.1016/j.fitote.2009.09.012.

    Article  CAS  PubMed  Google Scholar 

  76. Bak EJ, Park HG, Kim JM, Kin JM, Yoo YJ, Cha JH: Inhibitory effect of evodiamine alone and in combination with rosiglitazone on in vitro adipocyte differentiation and in vivo obesity related to diabetes. Int J Obes (Lond). 2010, 34 (2): 250-260. 10.1038/ijo.2009.223.

    Article  CAS  Google Scholar 

  77. Willie JT, Chemelli RM, Sinton CM, Yanagisawa W: To eat or to sleep? Orexin in the regulation of feeding and wakefulness. Neuroscience. 2001, 24: 429-458.

    CAS  Google Scholar 

  78. Shimada M, Tritos NA, Lowell BB, Flier JS, Maratos-Flie E: Mice lacking melanin-concentrating hormone are hypophagic and lean. Nature. 1998, 396: 670-674. 10.1038/25341.

    Article  CAS  PubMed  Google Scholar 

  79. Wolfrum C, Besser D, Luca E, Stoffel M: Insulin regulates the activity of forkhead transcription factor Hnf-3beta/Foxa-2 by Akt-mediated phosphorylation and nuclear/cytosolic localization. Proc Natl Acad Sci USA. 2003, 100: 11624-11629. 10.1073/pnas.1931483100.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  80. Silva JP, Von Meyenn F, Howell J, Thorens B, Wolfrum C: Regulation of adaptive behaviour during fasting by hypothalamic Foxa2. Nature. 2009, 462: 646-651. 10.1038/nature08589.

    Article  CAS  PubMed  Google Scholar 

  81. Shi J, Yan J, Lei Q, Zhao J, Chen K, Yang D, Zhaox X, Zhang Y: Intragastric administration of evodiamine suppresses NPY and AgRP gene expression in the hypothalamus and decreases food intake in rats. Brain Res. 2009, 1247: 71-78. 10.1016/j.brainres.2008.09.091.

    Article  CAS  PubMed  Google Scholar 

  82. Kim SJ, Lee SJ, Lee S, Chae S, Han MD, Mar W, Nam KW: Rutaecarpine ameliorates body weight gain through the inhibition of orexigenic neuropeptides NPY and AgRP in mice. Biochem Biophys Res Commun. 2009, 389 (3): 437-442. 10.1016/j.bbrc.2009.08.161.

    Article  CAS  PubMed  Google Scholar 

  83. Suzen S, Buyukbingol E: Recent studies of aldose reductase enzyme inhibition for diabetic complications. Curr Med Chem. 2003, 10 (15): 1329-1352. 10.2174/0929867033457377.

    Article  CAS  PubMed  Google Scholar 

  84. Kato A, Yasuko H, Goto H, Hollinshead J, Nash RJ, Adachi I: Inhibitory effect of rhetsinine isolated from Evodia rutaecarpa on aldose reductase activity. Phytomedicine. 2009, 16 (2-3): 258-261. 10.1016/j.phymed.2007.04.008.

    Article  CAS  PubMed  Google Scholar 

  85. Yu LL, Liao JF, Chen CF: Anti-diarrheal effect of water extract of Evodia Fructus in mice. J Ethnopharmacol. 2000, 73 (1-2): 39-45. 10.1016/S0378-8741(00)00267-1.

    Article  CAS  PubMed  Google Scholar 

  86. Yu X, Wu DZ: Protective effects of Evodia rutaecarpa, water extract on ethanol-induced rat gastric lesions. Zhongguo Zhong Yao Za Zhi. 2006, 31 (21): 1801-1803.

    PubMed  Google Scholar 

  87. Yu X, Wu DZ, Yuam JY, Zhang RR, Hu ZB: Gastroprotective effect of fructus evodiae water extract on ethanol-induced gastric lesions in rats. Am J Chin Med. 2006, 34 (6): 1027-1035. 10.1142/S0192415X06004491.

    Article  PubMed  Google Scholar 

  88. Wu CL, Hung CR, Chang FY, Lin LC, Pau KY, Wang PS: Effects of evodiamine on gastrointestinal motility in male rats. Eur J Pharmacol. 2002, 457 (2-3): 169-176. 10.1016/S0014-2999(02)02687-0.

    Article  CAS  PubMed  Google Scholar 

  89. Zhang T, Wang MW, Chen SW: Anti-emetic effect of ethanol extract from 'Wuzhuyu broth'. Zhougguo Zhong Yao Za Zhi. 2002, 27 (11): 862-866.

    Google Scholar 

  90. Kim BJ, Kim JH, Kim HP, Heo MY: Biological screening of 100 plant extracts for cosmetic use (II): Anti-oxidative activity and free radical scavenging activity. Int J Cosmet Sci. 1997, 19 (6): 299-307. 10.1111/j.1467-2494.1997.tb00194.x.

    Article  CAS  PubMed  Google Scholar 

  91. Shin YW, Bae EA, Cai XF, Lee JJ, Kim DH: In vitro and in vivo antiallergic effects on the fruits of Evodia rutaecarpa and its constituents. Biol Pharm Bull. 2007, 30 (1): 197-199. 10.1248/bpb.30.197.

    Article  CAS  PubMed  Google Scholar 

  92. Ho JN, Lee YH, Lee YD, Jun WJ, Kim HK, Hong BS, Shin DH, Cho HY: Inhibitory effect of aucubin isolated from Eucommia ulmoides against UVB-induced matrix metalloproteinase-1 production in human skin fibroblasts. Biosci Biotechnol Biochem. 2005, 69 (11): 2227-2231. 10.1271/bbb.69.2227.

    Article  CAS  PubMed  Google Scholar 

  93. Yarosh DB, Galvin JW, Nay SL, Pena AV, Canning MT, Broen DA: Anti-inflammatory activity in skin by biomimetic of Evodia rutaecarpa extract from traditional Chinese medicine. J Dermatol Sci. 2006, 42 (1): 13-21. 10.1016/j.jdermsci.2005.12.009.

    Article  PubMed  Google Scholar 

  94. Beak SM, Paek SH, Jahng Y, Lee YS, Kim JA: Inhibition of UVA irradiation-modulated signaling pathways by rutaecarpine, a quinazolinocarboline alkaloid, in human keratinocytes. Eur J Pharmacol. 2004, 49 (1-3): 19-25. 10.1016/j.ejphar.2004.07.065.

    Article  CAS  Google Scholar 

  95. Yamahara J, Yamada T, Kitani T, Naitoh Y, Fujmura H: Antianoxic action and active constituents of Evodia Fructus. Chem Pharm Bull (Tokyo). 1989, 37 (7): 1820-1822.

    Article  CAS  Google Scholar 

  96. Yamahara J, Yamada T, Kitani T, Naitoh Y, Fujimura H: Antianoxic action of evodiamine, an alkaloid in Evodia rutaecarpa fruit. J Ethnopharmacol. 1989, 27 (1-2): 185-192. 10.1016/0378-8741(89)90090-1.

    Article  CAS  PubMed  Google Scholar 

  97. Zheng MS, Zhang YZ: Anti-HBsAg herbs employing ELISA technique. Zhongguo Zhong Xi Yi Jie He Za Zhi. 1990, 10 (9): 560-562.

    CAS  Google Scholar 

  98. Perrett S, Whitfield PJ: Atanine (3-dimethylallyl-4-methoxy-2-quinolone), an alkaloid with anthelmintic activity from the Chinese medicinal plant, Evodia rutaecarpa. Planta Med. 1995, 61 (3): 276-278. 10.1055/s-2006-958073.

    Article  CAS  PubMed  Google Scholar 

  99. Rho TC, Bae EA, Kim DH, Oh WK, Kim BY, Ahn JS, Lee HS: Anti-Helicobacter pylori activity of quinolone alkaloids from Evodiae Fructus. Biol Pharm Bull. 1999, 22 (10): 1141-1143.

    Article  CAS  PubMed  Google Scholar 

  100. Hamasaki N, Ishii E, Tominaga K, Tezuka Y, Nagaoka T, Kadota S, Kuroki T, Yano I: Highly selective antibacterial activity of novel alkyl quinolone alkaloids from a Chinese herbal medicine, Gosyuyu (Wu-Chu-Yu), against Helicobacter pylori in vitro. Microbiol Immunol. 2000, 44 (1): 9-15.

    Article  CAS  PubMed  Google Scholar 

  101. Tominaga K, Higuchi K, Hamasaki N, Hamaguchi M, Takashima T, Tanigawa T, Watanabe T, Fujiwara Y, Tezuke Y, Nagaoka T, Kadota S, Ishii E, Kobayashi K, Arakawa T: In vivo action of novel alkyl methyl quinolone alkaloids against Helicobacter pylori. J Antimicrob Chemother. 2002, 50 (4): 547-552. 10.1093/jac/dkf159.

    Article  CAS  PubMed  Google Scholar 

  102. Tominaga K, Higuchi K, Hamasaki N, Tanigawa T, Sasaki E, Watanabe T, Fujiwara Y, Oshitani N, Arakawa T, Ishii E, Tezuka Y, Nagaoka T, Kadota S: Antibacterial activity of a Chinese herb medicine, Gosyuyu (Wu-Chu-Yu), against Helicobacter pylori. Nippon Rinsho. 2005, 63 (Suppl 11): 592-599.

    PubMed  Google Scholar 

  103. Tang Y, Wu F, Feng X, Huang L: Studies on synthesis and bioactivity of 2-alkenyl-4(1H)-quinolone. Yaoxue Xuebao. 1998, 33 (4): 269-274.

    CAS  Google Scholar 

  104. Thuille N, Fille M, Nagl M: Bactericidal activity of herbal extracts. Int J Hyg Environ Health. 2003, 206 (3): 217-221. 10.1078/1438-4639-00217.

    Article  PubMed  Google Scholar 

  105. Khan MR, Kihara M, Omolos AD: Antimicrobial activity of Evodia elleryana. Fitoterapia. 2000, 71 (1): 72-74. 10.1016/S0367-326X(99)00110-0.

    Article  CAS  PubMed  Google Scholar 

  106. Barrows LR, Powan E, Pond CD, Matainaho T: Anti-TB activity of Evodia elleryana bark extract. Fitoterapia. 2007, 78 (3): 250-252. 10.1016/j.fitote.2006.12.001.

    Article  PubMed Central  PubMed  Google Scholar 

  107. Ratsimamanga-Urverg S, Rasoanivo P, Rakoto-Ratsimamanga A, Le Bras J, Ramiliarisoa O, Savel J, Coulaud JP: Antimalarial activity and cytotoxicity of Evodia fatraina stem bark extracts. J Ethnopharmacol. 1991, 33 (3): 231-236. 10.1016/0378-8741(91)90082-O.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Chieh-Fu Chen.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

CFC proposed the review and wrote the manuscript. JFL searched the literature, compiled and reviewed the information and revised the manuscript. WFC reviewed the information on NO, NOS and endotoxaemic rats. YCS reviewed the information on neutrophils and microglial cells. GJW reviewed the information on vascular smooth muscle cell, endothelial cell and electropharmacology. All authors read and approved the final version of the manuscript.

Electronic supplementary material

13020_2010_108_MOESM1_ESM.DOC

Additional file 1: Mechanisms of anti-inflammatory relative effects of Evodia rutaecarpa and its bioactive components with potential clinic applications. The known mechanisms for anti-inflammatory effects of Evodia rutaecarpa extracts and its bioactive components such as dehydroevodiamine (DeHE), evodiamine (Evo) and rutaecarpine (Rut) are summarized and their potential clinic applications are suggested in this file. Some reported pharmacological effects of Wuzhuyu Tang (composed of Evodia fruit, Ginger, Ginseng, and Jujube) are also listed. Please refer to the text for the detail and references. (DOC 55 KB)

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Liao, JF., Chiou, WF., Shen, YC. et al. Anti-inflammatory and anti-infectious effects of Evodia rutaecarpa (Wuzhuyu) and its major bioactive components. Chin Med 6, 6 (2011). https://doi.org/10.1186/1749-8546-6-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1749-8546-6-6

Keywords