The activity of medicinal plants and secondary metabolites on eosinophilic inflammation

The activity of medicinal plants and secondary metabolites on eosinophilic inflammation

Pharmacological Research 62 (2010) 298–307 Contents lists available at ScienceDirect Pharmacological Research journal homepage: www.elsevier.com/loc...

327KB Sizes 0 Downloads 23 Views

Pharmacological Research 62 (2010) 298–307

Contents lists available at ScienceDirect

Pharmacological Research journal homepage: www.elsevier.com/locate/yphrs

Review

The activity of medicinal plants and secondary metabolites on eosinophilic inflammation Alexandre P. Rogerio a,∗ , Anderson Sá-Nunes b , Lúcia H. Faccioli c a

Universidade Federal do Triângulo Mineiro, Av. Getúlio Guaritá s/n, Uberaba, MG 38025-440, Brazil Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 1730, São Paulo, SP 05508-900, Brazil c Departamento de Análises Clínicas, Toxicológicas e Bromatológicas Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Café s/n, Ribeirão Preto, SP 14040-903, Brazil b

a r t i c l e

i n f o

Article history: Received 8 December 2009 Received in revised form 16 April 2010 Accepted 27 April 2010 Keywords: Medicinal plants Secondary metabolites Eosinophils Anti-eosinophilic activity

a b s t r a c t Eosinophils are leukocytes that are present in several body compartments and in the blood at relatively low numbers under normal conditions. However, an increase in the number of eosinophils, in the blood or in the tissues, is observed in allergic or parasitic disorders. Although some progress has been made in understanding the development of eosinophil-mediated inflammation in allergic and parasitic diseases, the discovery of new compounds to control eosinophilia has lagged behind other advances. Plant-derived secondary metabolites are the basis for many drugs currently used to treat pathologic conditions, including eosinophilic diseases. Several studies, including our own, have demonstrated that plant extracts and secondary metabolites can reduce eosinophilia and eosinophil recruitment in different experimental animal models. In this review, we summarize these studies and describe the anti-eosinophilic activity of various plant extracts, such as Ginkgo biloba, Allium cepa, and Lafoensia pacari, as well as those of secondary metabolites (compounds isolated from plant extracts), such as quercetin and ellagic acid. In addition, we highlight the medical potential of these plant-derived compounds for treating eosinophil-mediated inflammation, such as asthma and allergy. © 2010 Elsevier Ltd. All rights reserved.

Contents 1. 2. 3. 4. 5.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . TH 2 immune responses and eosinophil trafficking . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Treatment of eosinophil-mediated inflammation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Plants with anti-eosinophil activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

298 299 299 300 303 304 304

1. Introduction

Abbreviations: 5-LO, 5-lipoxygenase; BALF, bronchoalveolar lavage fluid; CD, cluster of differentiation; Cys-LTs, cysteinyl leukotrienes; ECP, eosinophil cationic protein; EDN, eosinophil-derived neurotoxin; EGID, eosinophilic gastrointestinal diseases; EPO, eosinophil peroxidase; GM-CSF, granulocyte-macrophage colony-stimulating factor; hRSV, human respiratory syncytial virus; IFN-␥, interferon-gamma; IgE, immunoglobulin E; IL, interleukin; LTs, leukotrienes; MBP, major basic protein; NF-␬B, nuclear factor-␬B; OVA, ovalbumin; PAF, plateletactivating factor; TGF-␣/␤, transforming growth factor-alpha/beta; TH , T helper; TNF-␣, tumor necrosis factor-alpha; VLMS, visceral larva migrans syndrome. ∗ Corresponding author. Tel.: +55 34 3318 5813; fax: +55 34 3312 1487. E-mail addresses: [email protected], [email protected] (A.P. Rogerio). 1043-6618/$ – see front matter © 2010 Elsevier Ltd. All rights reserved. doi:10.1016/j.phrs.2010.04.005

The eosinophil granulocyte was first described by Paul Ehrlich in 1879 [1], although some researchers had identified eosinophillike cells or its products a few years earlier. Eosinophils are derived from CD34+ hematopoietic progenitor cells within the bone marrow, which differentiate into mature eosinophils in the presence of the eosinophilopoietins [interleukin-(IL-)3, IL-5 and granulocytemacrophage colony-stimulating factor (GM-CSF)] [2–5] under the control of specific transcription factors [6–8]. Eosinophils are multifunctional cells and important sources of various inflammatory and regulatory cytokines [IL-2, IL-3, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, IL-13, IL-16, IL-18, transforming growth factor-alpha/beta (TGF-␣/␤), tumor necrosis factor-alpha (TNF-␣), interferon-gamma (IFN-␥)], chemokines (CCL3, CCL5, CCL11, and others), and lipid

A.P. Rogerio et al. / Pharmacological Research 62 (2010) 298–307

mediators [leukotrienes (LTs) and platelet-activating factor (PAF)] [9–11]. In addition, eosinophils produce and secrete four principal cationic proteins: major basic protein (MBP), eosinophil-derived neurotoxin (EDN), eosinophil cationic protein (ECP), and eosinophil peroxidase (EPO), all of which can cause damage to tissues [11–14]. In recent decades, considerable progress has been made in our understanding of eosinophil functions under physiologic and pathologic conditions by using molecular and biochemical assays, as well as knockout and transgenic animals. However, the true biologic role of eosinophils remains obscure [15,16]. Under physiologic conditions, the average frequency of eosinophils in peripheral blood is low (1–3%), although these cells are part of the resident cell population in tissues such as the gastrointestinal tract, thymus, and others [17–19]. Nevertheless, in the presence of allergic and parasitic diseases, the production and output of eosinophils by the bone marrow increase, leading to their accumulation in the blood (eosinophilia) as well as in different organs and tissues [2]. 2. TH 2 immune responses and eosinophil trafficking Initial exposure of the immune system to allergens or parasitic antigens leads to the activation of a subset of T cells, known as T helper 2 (TH 2) cells, which orchestrate the immune response to these exogenous antigens by secreting cytokines, including IL4, IL-5 and IL-13 [20–23]. IL-4 is the major differentiation factor for the TH 2-type immune response and also blocks differentiation towards a TH 1 axis by downregulating the transcription of IFN-␥ [24,25]. In addition, in conjunction with IL-13, IL-4 also induces immunoglobulin E (IgE) class switching in B cells [26,27]. Most of the IgE produced is bound to mast cells and basophils by their high-affinity Fc receptor (Fc␧RI) [28,29]. Subsequent exposure of immune cells to antigen induces degranulation of IgE-sensitized mast cells and release of both pre-formed and newly generated mediators, a process known as immediate or type I hypersensitivity [28]. These mediators, alone or in conjunction with TH 2 cytokines, increase the contractility of smooth muscle cells, the permeability of epithelial cells and the production of mucus. In addition, these mediators trigger the recruitment of macrophages, eosinophils, and basophils to the inflammatory site [30]. IL-5 is the major cytokine involved in the accumulation of eosinophils in the blood during allergic inflammation and parasitic infections. This cytokine is essential for eosinophil migration from the bone marrow to the blood [21,31] and specifically supports terminal differentiation and proliferation of eosinophil precursors, as well as the activation of mature eosinophils [32–35]. However, residual tissue eosinophils are still found in IL-5-deficient mice [36,37] and asthmatic patients treated with anti-IL-5 monoclonal antibody [38], suggesting the existence of a minor IL-5-independent eosinophil population. The trafficking of eosinophils into physiologic or inflammatory sites involves the interaction of cytokines, chemokines, lipid mediators and adhesion molecules. Under normal conditions, the migration of eosinophils is regulated by the constitutive expression of eotaxin-1 (CCL11; a selective chemokine of eosinophils) in the gastrointestinal tract, thymus and mammary glands [17,39,40]. Notably, trafficking of eosinophils into the uterus is also regulated by estrogen [39]. Under inflammatory conditions, the mediators implicated in leukocyte migration, IL-5 and eotaxins (CCL11, CCL24 and CCL26), have been shown to cooperate in selectively recruiting eosinophils [41–45], and recent studies have demonstrated an important role for the eotaxins receptor, CCR3, in eosinophil recruitment to tissues and body compartments [46]. In addition, IL-5 increases the pool of eotaxin-responsive cells and primes eosinophils to respond to CCR3 ligands [47]. Moreover, IL-4 and IL-13 are potent inducers of eotaxins and when eotaxins are exogenously administered, they collaborate with IL-5 to induce production of IL-13 in the lungs [47], suggesting the existence of a

299

positive feedback loop between specific cytokines and chemokines in eosinophil recruitment. In allergic or parasitic diseases, the local chemokine network appears to regulate eosinophil accumulation independently of IL-5, a mechanism that could also play an important role in eosinophil-mediated inflammatory processes [40]. In addition, lipid mediators seem to have important roles in allergic and parasitic diseases. These mediators include leukotriene B4 (LTB4 ) and the cysteinyl leukotrienes (Cys-LTs): LTC4 , LTD4 and LTE4 . LTB4 induces the activation and chemotaxis of neutrophils and eosinophils [48–50], while Cys-LTs induce constriction of airway smooth muscle, subepithelial fibrosis, plasma leakage and mucus hypersecretion [51]. These lipid mediators are produced by a variety of cells, including eosinophils, mast cells, and macrophages [52], from the metabolism of membrane-derived arachidonic acid via the activity of the enzyme 5-lipoxygenase (5-LO) and its helper protein 5-LO-activating protein [53–55]. Although eosinophil recruitment and eosinophilia are hallmarks of asthma and parasitic diseases, their presence is also associated with the pathogenesis of other conditions. For example, in inflammatory bowel disease, eosinophils usually represent only a small percentage of the infiltrating leukocytes [56,57]. However, their presence has been proposed as a negative prognostic indicator [56,58]. High accumulation of eosinophils without a known cause is also observed in eosinophilic gastrointestinal diseases (EGID) and is believed to be caused by TH 2-derived cytokines and chemokines [59]. Eosinophils have also been detected in the bronchoalveolar lavage fluid (BALF) of patients infected with respiratory syncytial virus (hRSV; a RNA virus of the family Paramyxoviridae) [60–62]; however, the role of eosinophils in the pathogenesis of this viral infection is still unclear. 3. Treatment of eosinophil-mediated inflammation Pharmacologic compounds capable of inhibiting eosinophil function or migration are desirable for the treatment of patients with atopy/allergy, parasitic disorders and other eosinophil-related diseases. The majority of clinically important medicines used for the treatment of inflammation of different origins, including those mediated by eosinophils, belong to the steroid (e.g., corticosteroids) or non-steroid (e.g., LT inhibitors or antagonists) anti-inflammatory drug families. Corticosteroids are the most established therapeutics for controlling virtually all types of inflammatory reactions, and they exert a strong effect on leukocyte recruitment when administered locally or systemically [63–65]. The effects of steroids on eosinophil distribution and function seem to result from a combination of direct and indirect mechanisms. The potential targets of glucocorticoid-mediated action include eosinophil generation, priming and recruitment of eosinophils and the production/release of eosinophil chemoattractants by distinct cellular sources, including mast cells, endothelial cells, epithelial cells, fibroblasts and other cell types [66–68]. The most striking effect of corticosteroids is their ability to inhibit the expression of multiple inflammatory mediators whose genes are regulated by transcription factors, such as nuclear factor-␬B (NF-␬B) [65]. Thus, inhibition of the NF-␬B pathway is associated with suppressed expression of genes encoding cytokines (e.g., IL-5), chemokines (e.g., eotaxins) and adhesion molecules (e.g., integrins), all of which play critical roles in controlling eosinophil recruitment [11,69,70]. Inhibitors of LT synthesis (such as zileuton, which directly inhibits 5-LO) or CysLT1 antagonists (such as montelukast, zafirlukast, and pranlukast) may also have complementary effects for the therapeutic treatment of asthma by reducing the requirement for corticosteroid use [51,55,71]. Although the drugs described above have potent antiinflammatory activity (individually or combined), they also have adverse side effects that severely limit their long-term use.

300

A.P. Rogerio et al. / Pharmacological Research 62 (2010) 298–307

Fig. 1. Potential target of medicinal plants or secondary metabolites in eosinophilic inflammation.

A promising approach for the treatment of allergy and other related diseases is the development of therapies to control the TH 2 immune response. For example, monoclonal antibodies against IgE [72], IL-4 [73], IL-5 [74] and IL-13 [75] have been tested in animal models, as well as in human preclinical trials. Although some of these antibodies have failed to demonstrate therapeutic efficacy, additional studies are still in progress [30]. Several other mediators, such as endothelins [76,77], PAF [78], tachykinins [79], bradykinin [80], adenosine [81] and others, are involved in the induction of eosinophilia and eosinophil recruitment. Consequently, antagonists to these mediators may also have therapeutic potential for the control of eosinophil-mediated inflammation. 4. Plants with anti-eosinophil activity A number of experimental models are able to reproduce one or more features of allergic responses and have been studied for a few decades, such as the classical ovalbumin-(OVA-)induced allergic asthma model [82,83] and the Toxocara canis helminth infection, also known as visceral larva migrans syndrome (VLMS) or toxocariasis [21,84–86]. Some other models induce recruitment of different cell types, predominantly eosinophils; these models include acute peritonitis induced by the polysaccharide-rich F1 fraction from Histoplasma capsulatum yeast [87,88] or the inoculation of a variety of molecules, such as cytokines [89], chemokines [90,91], lipid mediators [48,92,93] and several other substances. These experimental models differ from each other in the kinetics (acute or chronic) and localization (local or systemic) of the eosinophilic response. A more detailed description of the most commonly used models of eosinophilia, including the ones described above, and their characteristics can be found in Table 1. In the course of a continued search for bioactive natural products derived from plants, several groups, including our own,

have successfully employed experimental models to screen the pharmacologic activities of plant extracts, as well as isolated compounds (secondary metabolites). Thus, many plant extracts and secondary metabolites have been shown to reduce eosinophilia and/or eosinophil recruitment (Fig. 1). We have generated a catalog that describes many plant-derived products reported in the literature and includes information on administration routes, doses and species employed for the study of each type of preparation or isolated compound. Tables 2 and 3 display lists of published studies on different plant extracts and secondary metabolites, respectively. In this section, the major findings in the field will be discussed, with special attention to those agents already tested in preclinical and clinical human studies. Ginkgo biloba L. (Ginkgoaceae) is one of the most well-known plants in Chinese culture and has been used for therapeutic purposes for about 1,000 years. Its extracts are marketed worldwide to prevent or delay cognitive impairment associated with aging or neurodegenerative disorders. In addition, G. biloba leaves have been used for the treatment of airway diseases, such as asthma and bronchitis [94]. In fact, treatment of asthmatic patients with fluticasone propionate (by aerosol; 1,000 mg) plus a daily oral dose of G. biloba extract (240 mg) significantly reduced eosinophil and lymphocyte numbers in BALF compared to the asthmatic subgroups treated with fluticasone propionate alone [95], confirming the potential anti-asthmatic activity of the extract. Additionally, a concentrated liquor of G. biloba leaves also provided improvement of clinical symptoms and pulmonary functions of asthmatic patients when orally administrated [96]. Two major anti-eosinophilic mechanisms of G. biloba seem to explain the observed effects. First, the total flavonoid content in G. biloba leaves presents a pro-apoptotic effect on eosinophils [97]. Second, the ginkgolide BN52021, isolated from G. biloba, has been described as a specific PAF antagonist, inhibiting human eosinophil chemotaxis in vitro [98] and eosinophil infiltration into the lungs of OVA-sensitized guinea pigs

A.P. Rogerio et al. / Pharmacological Research 62 (2010) 298–307

301

Table 1 Experimental models of eosinophil-mediated inflammationa . Model

Eosinophil localization

Kinetics of eosinophil increase

References

Experimental allergic asthma model induced by OVA sensitization and challenge

Lungs (intranasal or aerosol challenge), pleural cavity (intrathoracic inoculation), skin (intradermal challenge)

[118,45–150]

Infection with T. canis eggs

Blood, peritoneal cavity, lungs

Infection with Strongyloides venezuelensis Inoculation of F1 fraction (␤-glucan) from yeast wall of H. capsulatum LTB4 administration

Blood, peritoneal cavity, lungs

From 1 to 24 days after challenge (model-dependent) Peak at 1–3 days after challenge (model-dependent) From 3 to 48 days Peak at 18–24 days Peak at 14 day

[21,20,31,151,117,152] [205]

Peritoneal cavity (intraperitoneal inoculation)

From 24 to 168 h Peak at 48 h

[87,88,153]

Measured at 2 and 24 hb

[48,92,152]

Eotaxin administration

Peritoneal cavity (intraperitoneal inoculation), bronchoalveolar lavage fluid (aerosol), skin (intradermal inoculation) Lungs (aerosol or intratracheal inoculation)

[91,155]

LPS administration

Pleural cavity (intrathoracic inoculation)

IL-5 administration

Bronchoalveolar lavage fluid (intratracheal inoculation), skin (intradermal inoculation), peritoneal cavity (intraperitoneal inoculation) Blood (intravenous inoculation) Skin (intradermal inoculation), bronchoalveolar lavage fluid (intratracheal inoculation)

From 6 h to 7 daysb Peak at 24 h From 24 to 96 h Peak at 48 h From 5 to 24 h Peak at 24 h From 30 min to 3 h Peak at 1 h From 2 to 4 hb (skin); 6–24 h (peritoneal cavity) Peak at 2 hb (skin); 24 h (peritoneal cavity) Measured at 48 h (bronchoalveolar lavage fluid) Measured at 4, 24 and 48 hb No apparent peak 48 h Peak at 48 hb From 4 to 48 h Peak at 24 h From 2 to 14 days (blood) From 2 to ∼80 days (lung) Peak at 7 days From 12 to 48 hb Peak at 24 h (mouse) and 48 h (ratb ) From 30 min to 6 hb Peak at 6 hb Measured at 2 and 4 h

IL-8 administration

Polybia paulista venom inoculation

Peritoneal cavity (intraperitoneal inoculation)

Large volume of saline

Peritoneal cavity (intraperitoneal inoculation)

Stem cell factor inoculation

Pleural cavity (intrathoracic inoculation)

Inoculation of sephadex particles

Blood and lungs (intravenous inoculation) Peritoneal cavity (intraperitoneal inoculation)

Zymosan-activated plasma inoculation C5a inoculation

Skin (intradermal inoculation)

Carrageenan

Pleural cavity (intrathoracic inoculation)

Bradykinin

Pleural cavity (intrathoracic inoculation)

PAF inoculation

Pleural cavity (intrathoracic inoculation), skin (intradermal inoculation) Pleural cavity (intrathoracic inoculation), skin (intradermal inoculation) Peritoneal cavity (intraperitoneal inoculation)

Compound 48/80 DMTI-II inoculation a b

Skin (intradermal inoculation)

From 6 to 48 hb Peak at 24 h From 24 to 48 hb Peak at 24 h From 6 to 120 h (pleural cavity) Peak at 24 h (pleural cavity) From 24 to 96 h (pleural cavity) Peak at 24 h (pleural cavity) From 4 to 24 h Peak at 16 h

[156] [89,157–159]

[90,157,159]

[160] [154,159] [149] [161–165]

[48] [48,90] [166] [166] [93,167] [48,93] [168]

Parasitic model, except T. canis model and infection with Strongyloides venezuelensis, not included. No further time points were analysed.

[99]. Unfortunately, despite its ability to suppress PAF- and antigeninduced bronchoconstriction in normal subjects, this compound was not able to inhibit the neutrophilia and eosinophilia in asthmatic patients under the same conditions [100]. Allium cepa L. (Alliaceae), commonly known as onion, is among the oldest of all cultivated plants, and its extracts have been used as therapeutic agents for thousands of years. Their effects include antimicrobial, antithrombotic and antitumor properties, among others [101]. In the therapy, a cream, which includes onion in its composition, has been used in human to treat burn scars, keloids and contractures [102]. Interestingly, in the murine model of OVA-induced allergic airway inflammation, A. cepa extract (50–200 mg/kg, p.o.) significantly decreased eosinophil numbers in BALF, as well as other hallmarks of asthma [103]. The flavonoid quercetin is found in high amounts in A. cepa. This secondary metabolite is also found in other vegetables and several fruits consumed in a regular diet, as well as in a wide variety of plants [104,105]. Usually quercetin is linked to sugars, such as glu-

cose (quercetin-3-glucoside; isoquercitrin) or rutinose (rutin), and exhibits many activities in humans, such as in overweight-obese carriers [106,107], and in animals, such as anti-inflammatory and anti-allergic properties [108], among other activities. By further exploring the medicinal properties of plant-derived molecules, we have demonstrated, for the first time, that treatment with quercetin (10 mg/kg) reduces eosinophils in blood, BALF and pulmonary parenchyma in the murine OVA-induced allergic model [109]. Similar results have been found by many others employing the same experimental model in mice and guinea pigs under different treatment routes with quercetin [110–112]. Thus, the potential effects of A. cepa extract and, perhaps, other plants in inhibiting eosinophilic inflammation [101,113] may be due to the quercetin content or to the association (additive or synergistic) of this compound with other secondary metabolites. Another plant with anti-eosinophilic potential is Lafoensia pacari, Jaumes St. Hilaire (Lythraceae). Its extract is traditionally used by the population of Mato Grosso State, Brazil to treat inflam-

302

A.P. Rogerio et al. / Pharmacological Research 62 (2010) 298–307

Table 2 Plant extracts with anti-eosinophil activitya . Plant species

Formulation

Model/disease

Species tested

Effective dose/route

References

Actinidia arguta (Siebold & Zucc.) Planch. ex Miq. (Actinidiaceae)

Dried fruits boiled in distilled water for 3 h. Extract filtered and concentrated, followed by a freeze-drying process. Powder dissolved in distilled water Ethanol extract from dried leaves and branches Aged garlic bulbs peeled, homogenized with distilled water, filtered and centrifuged. NH4 SO4 added to the supernatant (5%) and the solution centrifuged at 3600 × g for 30 min. Pellet resuspended in saline, dialyzed against buffered saline and sterilized Bulbs dried, finely ground, and percolated in 50% ethyl alcohol for 16 h at room temperature. This procedure was repeated four times, and the pooled extract was dried under reduced pressure (50 ± 1 ◦ C) in a rotary evaporator. The extract, was further partitioned into butanol Ethanol extract prepared (DA-9601) and dissolved in 3% hydroxypropyl methylcellulose Herbal preparation of the gum resin, called “S compound” (trade name). Sold by Rahul Pharma, Jammu Extract produced by Beaufour Ipsen, France (no details published) Plant materials were extracted three times with distilled water at 100 ◦ C for 2 h separately, filtered and evaporated on a rotator evaporator and finally dried using a freeze drier Stem bark ethanol extract dried to obtain a powder. Aqueous suspension of powder prepared for treatment Stem bark aqueous extract prepared by decoction and dried to obtain a powder. Aqueous suspension of powder prepared for treatment Aqueous extract prepared by infusion of dried leaves, filtered and lyophilized. Powder dissolved in saline for treatment Rice bran extracted with 70% ethanol for 24 h. Extract was filtered, concentrated and lyophilized. The dried extract (DA-9201) was suspended in a 1% hydroxypropyl methylcellulose solution for treatment Fresh leaves extracted with 1% (w/v) citric acid at 90 ◦ C for 30 min. Extract concentrated and mixed with n-butanol. Resulting n-butanol layer dried and the residue dissolved in distilled water for treatment Extract manufactured using liquid carbon dioxide with leaves of the cultivated Petasites variety PETZELL –

OVA-induced asthma

Mouse

300 mg/kg, p.o.

[169]

OVA-induced asthma

Mouse

200 and 400 mg/kg, p.o.

[170]

OVA-induced asthma

Mouse

200 mg/kg, i.p.

[113]

OVA-induced asthma

Mouse

50–200 mg/kg, p.o.

[103]

OVA-induced asthma

Mouse

30 and 100 mg/kg, p.o.

[171]

Bronchial asthma

Human

300 mg, p.o.

[172]

Asthma

Human

240 mg, p.o.

[95]

OVA-induced asthma

Mouse

200 and 400 mg/kg, p.o.

[173]

Toxocariasis F1-induced peritonitis OVA-induced asthma Toxocariasis

Mouse

200 mg/kg, p.o.

[31,119,117]

Mouse

50 mg/kg, p.o.

[174]

Allergic pleurisy, OVA-induced asthma

Mouse

2 mg/kg, i.p.

[175]

OVA-induced asthma

Mouse

100 and 300 mg/kg, p.o.

[176]

Asthma induced by house dust mite allergen

Mouse

1.5 mg/day, p.o.

[177]

Allergic rhinitis OVA-induced asthma

Human Mouse

8 mg/three times a day, p.o. 100 ␮g/mouse, i.n.

[178,179]

Allergic asthma

Rat



[180]

Allergic pleurisy

Mouse

100 mg/kg, p.o.

[181]

Asthma

Human

40 or 60 mg

[182]

OVA-induced asthma

Mouse

45–720 mg/kg, i.p.

[183]

Ailanthus altissima Swingle (Simaroubaceae) Allium sativum L. (Liliaceae)

Allium cepa L. (Liliaceae)

Artemisia asiatica Nakai (Asteraceae) Boswellia serrata Roxb. (Burseraceae) Ginkgo biloba L. (Ginkgoaceae) Citrus reticulata, Blanco (Rutaceae), Pinellia ternata (Thunb.) Breit (Araceae) and their combination Lafoensia pacari Jaumes St. Hilaire (Lythraceae) Mangifera indica L. (Anacardiaceae)

Nidularium procerum Lindiman (Bromeliaceae) Oryza sativa L. (Poaceae)

Perilla frutescens Britton (Laminaceae)

Petasites hybridus L. (Asteraceae) Salvia miltiorrhiza Bunge. (Labiatae)b Syzygium cumini (L.) Skeels (Myrtaceae)

Tripterygium wilfordii Hook.f. (Celastraceae)b Zingiber officinale Rosoce (Zingiberaceae)

a b

Aqueous extract from fresh leaves prepared by decoction, filtered and lyophilized. Powder dissolved in distilled water for treatment – ◦

Rhizoma dried at 70 C, ground to a fine powder and soaked in distilled water for 24 h. Extract was filtered, evaporated, concentrated and freeze dried. Aqueous solutions were prepared for treatment

Organized in alphabetic order. No access to the entire manuscript, no answer from the corresponding authors.

A.P. Rogerio et al. / Pharmacological Research 62 (2010) 298–307

303

Table 3 Secondary metabolites with anti-eosinophil activitya . Secondary metabolite

Chemical class

Model or disease

Dose and route

References

Andrographolide Apigenin Astragaloside IV Caffeic acid phenethyl ester Curcumin Ellagic acid

Diterpenoid Flavonoid Triterpene Polyphenol Polyphenol Polyphenol

0.5 and 1 mg/kg, i.p. 5 and 10 mg/kg, i.p. 50 mg/kg, p.o. 10 mg/kg, i.p. 20 mg/kg, i.p. 1 and 10 mg/kg, p.o.

[184] [185] [186] [187] [188] [119,118]

Epigallocatechin-3-gallate Genistein Gingerol Ginkgolide Glycyrrhizin ␣-Humulene Isoquecetrin Lupeol Luteolin Luteolin-7-O-glucoside Narirutin Kaempferol Nicotine

Flavonoid Flavonoid Polyphenol Terpenoid Triterpene Sesquiterpenes Flavonoid Triterpene Flavonoid Flavonoid Flavonoid Flavonoid Alkaloid

Allergic asthma in mice Allergic asthma in mice Allergic asthma in mice Allergic asthma in mice Allergic asthma in mice Peritonitis induced by H. capsulatum in mice Allergic asthma in mice Allergic asthma in guinea pigs Allergic asthma in guinea pigs Allergic asthma in mice Allergic asthma in guinea pigs Allergic asthma in mice Allergic asthma in mice Allergic asthma in mice Allergic asthma in mice Allergic asthma in mice Allergic asthma in mice Allergic asthma in mice Allergic asthma in mice Allergic asthma in rat

[129] [189] [183] [99] [190] [191] [109] [192] [193] [194] [195] [196] [197]

Nobiletin Piperineb 1H,8H-Pyrano [3,4-c]pyran-1,8-dioneb Quercetin

Flavonoid Polyphenol – Flavonoid

3-O-Methylquercetin Theophylline Thymoquinone Warifteine

Flavonoid Alkaloid Quinone Alkaloid

25 mg/kg, i.p. 15 mg/kg, i.p. 14 mg/kg, i.p. 25 mg/kg, p.o. 5–20 mg/kg, p.o. 50 mg/kg, p.o. 15 mg/kg, p.o. 60 mg/kg, p.o. 10 mg/kg, p.o. 50 and 100 mg/kg, p.o. 10 mg/kg, p.o. 30 and 90 mg/kg, s.c. 1 mg/kg, osmotic pump implanted s.c 1.5 and 5 mg/kg, i.p. – – 10 mg/kg, p.o. 10 mg/ml, inhalation 7.5 mg/kg, p.o. 8 and 16 mg/kg, i.p. 3–30 ␮mol/kg, i.p. 400 mg/day, p.o. 3 mg/kg, i.p. 50 ␮g/animal, p.o.

a b

Allergic asthma in rats Allergic asthma in mice Allergic asthma in mice Allergic asthma in mice Allergic asthma in guinea pigs Allergic asthma in guinea pigs Allergic asthma in mice Allergic asthma in mice Human asthma Allergic asthma in mice Allergic pleurisy and asthma models in mice

[198] [199] [200] [109,110–112,201]

[202] [132] [203] [204]

Organized in alphabetic order. No access to the entire manuscript, no answer from the corresponding authors.

mation and gastric ulcers [114,115]. In a clinical trial, however, L. pacari methanolic extract (500 mg) failed to eradicate Helicobacter pylori in dyspeptic urease-positive patients, although the extract was well-tolerated and many patients reported relief of symptoms [116]. Employing the asthma model induced by T. canis infection, our group demonstrated that oral treatment with an ethanolic extract of L. pacari (200 mg/kg) decreased the number of eosinophils recruited to several tissues and compartments such as blood, BALF and the peritoneal cavity [31,117]. Furthermore, in the OVA-induced asthma model, L. pacari extract was effective in decreasing eosinophil recruitment and the production of IL-4, IL5, and IL-13 in BALF [118]. In addition, L. pacari extract did not demonstrate cytotoxicity, either in vitro [31] or in vivo [117]. In an attempt to identify the molecule(s) responsible for the antieosinophil activity of L. pacari extract, we have used a model of peritonitis induced in mice by exposure to the F1 fraction of the H. capsulatum yeast wall [87]. This model of acute and localized eosinophilia was suitable for the bioassay-guided fractionation of L. pacari extract. In this manner, we were able to isolate and chemically characterize ellagic acid (a polyphenol) as the major active component in L. pacari extract [119]. To our knowledge, this was the first study to demonstrate the ability of ellagic acid to inhibit eosinophil recruitment. We also showed that ellagic acid (1 and 10 mg/kg, p.o.) induced a reduction in the numbers of eosinophils found in BALF and lung parenchyma in the asthma model. Furthermore, similar to the whole L. pacari extract, ellagic acid was also effective in decreasing the concentrations of IL-4, IL-5, and IL-13 in BALF [118]. In addition to this activity, ellagic acid also demonstrates several other biologic effects, including anti-oxidant, anti-cancer, and anti-allergic activity [120–122]. Concerning its toxicity, intake of pomegranate juice (Punica granatum L., Punicaceae) with an equivalent concentration of 121 mg/L of ellagic acid

produced no toxic effect in humans over 3 years and conferred the benefits of reduced common carotid intima-media thickness, blood pressure and low-density lipoprotein (LDL) oxidation [123]. Green tea, from Camellia sinensis L. (Theaceae), is widely consumed around the world and is prepared by drying and steaming fresh tea leaves. Flavonoids are the major secondary metabolites found in green tea, and epigallocatechin-3-gallate is the most abundant of these. In cancer research, phase I and/or II clinical trials have been conducted with the green tea extract [124] as well as its flavonoids (catechins) [125,126]. In addition, beneficial effects of catechins have also been reported with regard to inflammation [127], angiogenesis [128] and other processes. In the guinea pig OVA-induced asthma model, epigallocatechin-3-gallate (25 mg/kg, i.p.) reduced eosinophilic inflammation, as well as other inflammatory parameters [129]. Therefore, epigallocatechin-3-gallate might constitute an attractive molecule with potential interest for the treatment of asthma and related inflammatory and allergic diseases. In the management of obstructive airway diseases from diverse etiologies, the alkaloid theophylline is one of the oldest drugs in use [130,131], despite its weakness as a bronchodilator. Its use is often limited due to concerns regarding dose-related adverse effects, numerous drug interactions and a narrow therapeutic index. However, it has been demonstrated that a low oral dose of theophylline attenuates airway inflammatory responses to allergen inhalation in atopic asthma, extending the beneficial effect of this alkaloid to the therapy of airway diseases [132]. 5. Concluding remarks Natural products and plant derivatives used in folk medicine are of vast medical importance due to their potential as a source

304

A.P. Rogerio et al. / Pharmacological Research 62 (2010) 298–307

of molecules with pharmacologic properties. In fact, plant-derived drugs represent more than 30% of the current pharmaceutical market [133], including 60% of cancer drugs and 75% of infectious diseases drugs [134]. In addition, 25% of the drugs currently prescribed to patients are of plant origin, and, according to World Health Organization (WHO), 11% of the 252 drugs considered as basic and essential are derived from plants [135]. Although active plant-derived secondary metabolites can be randomly discovered, this process is laborious, with a success rate on the order of 1 new product per 10,000 plants screened [136]. Moreover, the isolation and identification of secondary metabolites requires a substantial amount of materials, financial resources and equipment [137–139]. In addition, bioassays need to be carried out in order to identify the fraction(s) that contains the active compound(s) and to quantify its purity [140]. Despite all of these obstacles, important scientific findings have been made in this area. For example, the term “medicinal plants” yielded around 50,000 entries in the U.S. National Library of Medicine (PubMed) from the National Institutes of Health as of April 2010. Several plant-derived extracts and secondary metabolites can directly influence the production of inflammatory mediators, as well as the production and activity of second messengers and the expression of transcription factors and key pro-inflammatory molecules [141–143]. In addition, some secondary metabolites provide relief of symptoms in a manner comparable to that obtained from allopathic medicines. Substances of natural origin with few or no side effects are desired as substitutes or complements to traditional chemical therapeutics [144]. Therefore, plant-derived extracts and secondary metabolites may constitute attractive therapeutic alternatives, and we expect that, in the near future, they will be used for the treatment of allergic diseases, such as asthma and related eosinophilic disorders. In this review, we presented an overall picture of the models for studying eosinophil-mediated pathologies and some of the advances in characterizing the anti-eosinophilic activity of medicinal plants and their secondary metabolites. Using these experimental models, we and others have demonstrated that plantderived products can markedly reduce eosinophil recruitment and/or eosinophilia, as well as other features involved in allergic and parasitic disorders, including the production of chemokines and TH 2 cytokines. We hope that this work will serve as a catalog of the extensive list of agents under investigation by researchers in the field of medicinal plants. Acknowledgement This study was supported by grants from Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq Grant no. 472601/2004-0), Fundac¸ão de Amparo à Pesquisa do Estado de São Paulo (FAPESP Grant no. 2009/07169-5) and Coordenac¸ão de Aperfeic¸oamento de Pessoal de Nível Superior (CAPES). The authors would like to thank Ana Cristina Hallal Prata for her contribution in the Fig. 1. References [1] Hirsch JG, Hirsch BI. Paul Ehrlich and the discovery of the eosinophil. New York: Grune & Stratton; 1980. [2] Rothenberg ME. Eosinophilia. N Engl J Med 1998;338:1592–600. [3] Lopez AF, Begley CG, Williamson DJ, Warren DJ, Vadas MA, Sanderson CJ. Murine eosinophil differentiation factor. An eosinophil-specific colony-stimulating factor with activity for human cells. J Exp Med 1986;163:1085–99. [4] Lopez AF, Sanderson CJ, Gamble JR, Campbell HD, Young IG, Vadas MA. Recombinant human interleukin 5 is a selective activator of human eosinophil function. J Exp Med 1988;167:219–24. [5] Takatsu K, Takaki S, Hitoshi Y. Interleukin-5 and its receptor system: implications in the immune system and inflammation. Adv Immunol 1994;57:145–90.

[6] McNagny K, Graf T. Making eosinophils through subtle shifts in transcription factor expression. J Exp Med 2002;195:F43–7. [7] Nerlov C, Graf T. Pu.1 induces myeloid lineage commitment in multipotent hematopoietic progenitors. Genes Dev 1998;12:2403–12. [8] Nerlov C, McNagny KM, Doderlein G, Kowenz-Leutz E, Graf T. Distinct c/ebp functions are required for eosinophil lineage commitment and maturation. Genes Dev 1998;12:2413–23. [9] Blanchard C, Rothenberg ME. Biology of the eosinophil. Adv Immunol 2009;101:81–121. [10] Lacy P, Moqbel R. Eosinophil cytokines. Chem Immunol 2000;76:134–55. [11] Rothenberg ME, Hogan SP. The eosinophil. Annu Rev Immunol 2006;24:147–74. [12] Gleich GJ, Loegering DA. Immunobiology of eosinophils. Annu Rev Immunol 1984;2:429–59. [13] Moqbel R, Lacy P. Exocytotic events in eosinophils and mast cells. Clin Exp Allergy 1999;29:1017–22. [14] Hamid Q, Tulic M. Immunobiology of asthma. Annu Rev Physiol 2009;71:489–507. [15] Simon D, Simon HU. Eosinophilic disorders. J Allergy Clin Immunol 2007;119:1291–300 [quiz 1301-1292]. [16] Venge P. Eosinophils and respiratory disease. Respir Med 2001;95:168–9. [17] Mishra A, Hogan SP, Lee JJ, Foster PS, Rothenberg ME. Fundamental signals that regulate eosinophil homing to the gastrointestinal tract. J Clin Invest 1999;103:1719–27. [18] Kendall MD. The morphology of perivascular spaces in the thymus. Thymus 1989;13:157–64. [19] Kato M, Kephart GM, Talley NJ, Wagner JM, Sarr MG, Bonno M, et al. Eosinophil infiltration and degranulation in normal human tissue. Anat Rec 1998;252:418–25. [20] Faccioli LH, Medeiros AI, Malheiro A, Pietro RC, Januario A, Vargaftig BB. Interleukin-5 modulates interleukin-8 secretion in eosinophilic inflammation. Mediators Inflamm 1998;7:41–7. [21] Faccioli LH, Mokwa VF, Silva CL, Rocha GM, Araujo JI, Nahori MA, et al. IL5 drives eosinophils from bone marrow to blood and tissues in a guinea-pig model of visceral larva migrans syndrome. Mediators Inflamm 1996;5:24–31. [22] Zheng W, Flavell RA. The transcription factor gata-3 is necessary and sufficient for th2 cytokine gene expression in CD4 T cells. Cell 1997;89:587–96. [23] Nakayama T, Yamashita M. Initiation and maintenance of Th2 cell identity. Curr Opin Immunol 2008;20:265–71. [24] Fiorentino DF, Bond MW, Mosmann TR. Two types of mouse T helper cell. IV. Th2 clones secrete a factor that inhibits cytokine production by Th1 clones. J Exp Med 1989;170:2081–95. [25] Nakamura T, Kamogawa Y, Bottomly K, Flavell RA. Polarization of IL-4- and IFN-gamma-producing CD4+ T cells following activation of naive CD4+ T cells. J Immunol 1997;158:1085–94. [26] de Vries JE, Punnonen J, Cocks BG, de Waal Malefyt R, Aversa G. Regulation of the human IgE response by IL4 and IL13. Res Immunol 1993;144:597–601. [27] Geha RS, Jabara HH, Brodeur SR. The regulation of immunoglobulin E classswitch recombination. Nat Rev Immunol 2003;3:721–32. [28] Gould HJ, Sutton BJ. IgE in allergy and asthma today. Nat Rev Immunol 2008;8:205–17. [29] Furuichi K, Rivera J, Isersky C. The receptor for immunoglobulin E on rat basophilic leukemia cells: effect of ligand binding on receptor expression. Proc Natl Acad Sci USA 1985;82:1522–5. [30] Holgate ST, Polosa R. Treatment strategies for allergy and asthma. Nat Rev Immunol 2008;8:218–30. [31] Rogerio AP, Sa-Nunes A, Albuquerque DA, Anibal FF, Medeiros AI, Machado ER, et al. Lafoensia pacari extract inhibits IL-5 production in toxocariasis. Parasite Immunol 2003;25:393–400. [32] Sanderson CJ, Warren DJ, Strath M. Identification of a lymphokine that stimulates eosinophil differentiation in vitro. Its relationship to interleukin 3, and functional properties of eosinophils produced in cultures. J Exp Med 1985;162:60–74. [33] Yamaguchi Y, Suda T, Suda J, Eguchi M, Miura Y, Harada N, et al. Purified interleukin 5 supports the terminal differentiation and proliferation of murine eosinophilic precursors. J Exp Med 1988;167:43–56. [34] Clutterbuck EJ, Sanderson CJ. Human eosinophil hematopoiesis studied in vitro by means of murine eosinophil differentiation factor (IL5): production of functionally active eosinophils from normal human bone marrow. Blood 1988;71:646–51. [35] Coeffier E, Joseph D, Vargaftig BB. Activation of guinea pig eosinophils by human recombinant IL-5. Selective priming to platelet-activating factoracether and interference of its antagonists. J Immunol 1991;147:2595– 602. [36] Foster PS, Hogan SP, Ramsay AJ, Matthaei KI, Young IG. Interleukin 5 deficiency abolishes eosinophilia, airways hyperreactivity, and lung damage in a mouse asthma model. J Exp Med 1996;183:195–201. [37] Hogan SP, Koskinen A, Foster PS. Interleukin-5 and eosinophils induce airway damage and bronchial hyperreactivity during allergic airway inflammation in BALB/c mice. Immunol Cell Biol 1997;75:284–8. [38] Phipps S, Flood-Page P, Menzies-Gow A, Ong YE, Kay AB. Intravenous antiIL-5 monoclonal antibody reduces eosinophils and tenascin deposition in allergen-challenged human atopic skin. J Invest Dermatol 2004;122:1406–12. [39] Gouon-Evans V, Pollard JW. Eotaxin is required for eosinophil homing into the stroma of the pubertal and cycling uterus. Endocrinology 2001;142: 4515–21.

A.P. Rogerio et al. / Pharmacological Research 62 (2010) 298–307 [40] Foster PS, Mould AW, Yang M, Mackenzie J, Mattes J, Hogan SP, et al. Elemental signals regulating eosinophil accumulation in the lung. Immunol Rev 2001;179:173–81. [41] Pope SM, Fulkerson PC, Blanchard C, Akei HS, Nikolaidis NM, Zimmermann N, et al. Identification of a cooperative mechanism involving interleukin13 and eotaxin-2 in experimental allergic lung inflammation. J Biol Chem 2005;280:13952–61. [42] Jose PJ, Griffiths-Johnson DA, Collins PD, Walsh DT, Moqbel R, Totty NF, et al. Eotaxin: a potent eosinophil chemoattractant cytokine detected in a guinea pig model of allergic airways inflammation. J Exp Med 1994;179:881–7. [43] Rothenberg ME, Luster AD, Lilly CM, Drazen JM, Leder P. Constitutive and allergen-induced expression of eotaxin mRNA in the guinea pig lung. J Exp Med 1995;181:1211–6. [44] Shinkai A, Yoshisue H, Koike M, Shoji E, Nakagawa S, Saito A, et al. A novel human cc chemokine, eotaxin-3, which is expressed in IL-4-stimulated vascular endothelial cells, exhibits potent activity toward eosinophils. J Immunol 1999;163:1602–10. [45] Zimmermann N, Hogan SP, Mishra A, Brandt EB, Bodette TR, Pope SM, et al. Murine eotaxin-2: a constitutive eosinophil chemokine induced by allergen challenge and IL-4 overexpression. J Immunol 2000;165:5839–46. [46] Humbles AA, Lu B, Friend DS, Okinaga S, Lora J, Al-Garawi A, et al. The murine CCR3 receptor regulates both the role of eosinophils and mast cells in allergeninduced airway inflammation and hyperresponsiveness. Proc Natl Acad Sci USA 2002;99:1479–84. [47] Zimmermann N, Hershey GK, Foster PS, Rothenberg ME. Chemokines in asthma: cooperative interaction between chemokines and IL-13. J Allergy Clin Immunol 2003;111:227–42 [quiz 243]. [48] Faccioli LH, Nourshargh S, Moqbel R, Williams FM, Sehmi R, Kay AB, et al. The accumulation of 111in-eosinophils induced by inflammatory mediators, in vivo. Immunology 1991;73:222–7. [49] Weller CL, Collington SJ, Brown JK, Miller HR, Al-Kashi A, Clark P, et al. Leukotriene B4, an activation product of mast cells, is a chemoattractant for their progenitors. J Exp Med 2005;201:1961–71. [50] Corhay JL, Henket M, Nguyen D, Duysinx B, Sele J, Louis R. Leukotriene B4 contributes to exhaled breath condensate and sputum neutrophil chemotaxis in COPD. Chest 2009;13:1047–54. [51] Salvi SS, Krishna MT, Sampson AP, Holgate ST. The anti-inflammatory effects of leukotriene-modifying drugs and their use in asthma. Chest 2001;119:1533–46. [52] Elias JA, Lee CG, Zheng T, Shim Y, Zhu Z. Interleukin-13 and leukotrienes: an intersection of pathogenetic schema. Am J Respir Cell Mol Biol 2003;28:401–4. [53] Miller DK, Gillard JW, Vickers PJ, Sadowski S, Leveille C, Mancini JA, et al. Identification and isolation of a membrane protein necessary for leukotriene production. Nature 1990;343:278–81. [54] Lewis RA, Austen KF, Soberman RJ. Leukotrienes and other products of the 5lipoxygenase pathway. Biochemistry and relation to pathobiology in human diseases. N Engl J Med 1990;323:645–55. [55] Peters-Golden M, Henderson Jr WR. Leukotrienes. N Engl J Med 2007;357:1841–54. [56] Desreumaux P, Nutten S, Colombel JF. Activated eosinophils in inflammatory bowel disease: do they matter? Am J Gastroenterol 1999;94:3396–8. [57] Walsh RE, Gaginella TS. The eosinophil in inflammatory bowel disease. Scand J Gastroenterol 1991;26:1217–24. [58] Nishitani H, Okabayashi M, Satomi M, Shimoyama T, Dohi Y. Infiltration of peroxidase-producing eosinophils into the lamina propria of patients with ulcerative colitis. J Gastroenterol 1998;33:189–95. [59] Blanchard C, Rothenberg ME. Chemotactic factors associated with eosinophilic gastrointestinal diseases. Immunol Allergy Clin North Am 2009;29:141–8, xi. [60] Dimova-Yaneva D, Russell D, Main M, Brooker RJ, Helms PJ. Eosinophil activation and cysteinyl leukotriene production in infants with respiratory syncytial virus bronchiolitis. Clin Exp Allergy 2004;34:555–8. [61] Garofalo R, Kimpen JL, Welliver RC, Ogra PL. Eosinophil degranulation in the respiratory tract during naturally acquired respiratory syncytial virus infection. J Pediatr 1992;120:28–32. [62] Harrison AM, Bonville CA, Rosenberg HF, Domachowske JB. Respiratory syncytial virus-induced chemokine expression in the lower airways: eosinophil recruitment and degranulation. Am J Respir Crit Care Med 1999;159:1918–24. [63] Flower RJ. Eleventh Gaddum memorial lecture. Lipocortin and the mechanism of action of the glucocorticoids. Br J Pharmacol 1988;94:987–1015. [64] Perretti M, Flower RJ. Modulation of IL-1-induced neutrophil migration by dexamethasone and lipocortin 1. J Immunol 1993;150:992–9. [65] Barnes PJ. Anti-inflammatory actions of glucocorticoids: molecular mechanisms. Clin Sci (Lond) 1998;94:557–72. [66] Barnes PJ. How corticosteroids control inflammation: quintiles prize lecture 2005. Br J Pharmacol 2006;148:245–54. [67] Barnes PJ, Adcock I. Anti-inflammatory actions of steroids: molecular mechanisms. Trends Pharmacol Sci 1993;14:436–41. [68] Schwiebert LM, Stellato C, Schleimer RP. The epithelium as a target of glucocorticoid action in the treatment of asthma. Am J Respir Crit Care Med 1996;154:S16–9 [discussion S19–20]. [69] Jia GQ, Gonzalo JA, Hidalgo A, Wagner D, Cybulsky M, Gutierrez-Ramos JC. Selective eosinophil transendothelial migration triggered by eotaxin via modulation of MAC-1/ICAM-1 and VLA-4/VCAM-1 interactions. Int Immunol 1999;11:1–10.

305

[70] Yamamoto H, Nagata M, Sakamoto Y. Cc chemokines and transmigration of eosinophils in the presence of vascular cell adhesion molecule 1. Ann Allergy Asthma Immunol 2005;94:292–300. [71] Montuschi P, Sala A, Dahlen SE, Folco G. Pharmacological modulation of the leukotriene pathway in allergic airway disease. Drug Discov Today 2007;12:404–12. [72] Holgate ST, Djukanovic R, Casale T, Bousquet J. Anti-immunoglobulin E treatment with omalizumab in allergic diseases: an update on anti-inflammatory activity and clinical efficacy. Clin Exp Allergy 2005;35:408–16. [73] Hart TK, Blackburn MN, Brigham-Burke M, Dede K, Al-Mahdi N, ZiaAmirhosseini P, et al. Preclinical efficacy and safety of pascolizumab (SB 240683): a humanized anti-interleukin-4 antibody with therapeutic potential in asthma. Clin Exp Immunol 2002;130:93–100. [74] Kips JC, O’Connor BJ, Langley SJ, Woodcock A, Kerstjens HA, Postma DS, et al. Effect of SCH55700, a humanized anti-human interleukin-5 antibody, in severe persistent asthma: a pilot study. Am J Respir Crit Care Med 2003;167:1655–9. [75] Bree A, Schlerman FJ, Wadanoli M, Tchistiakova L, Marquette K, Tan XY, et al. IL-13 blockade reduces lung inflammation after Ascaris suum challenge in cynomolgus monkeys. J Allergy Clin Immunol 2007;119:1251–7. [76] Landgraf RG, Jancar S. Endothelin A receptor antagonist modulates lymphocyte and eosinophil infiltration, hyperreactivity and mucus in murine asthma. Int Immunopharmacol 2008;8:1748–53. [77] Redington AE, Springall DR, Ghatei MA, Lau LC, Bloom SR, Holgate ST, et al. Endothelin in bronchoalveolar lavage fluid and its relation to airflow obstruction in asthma. Am J Respir Crit Care Med 1995;151:1034–9. [78] Barnes PJ, Chung KF, Page CP. Platelet-activating factor as a mediator of allergic disease. J Allergy Clin Immunol 1988;81:919–34. [79] Kraneveld AD, Nijkamp FP. Tachykinins and neuro-immune interactions in asthma. Int Immunopharmacol 2001;1:1629–50. [80] Bandeira-Melo C, Calheiros AS, Silva PM, Cordeiro RS, Teixeira MM, Martins MA. Suppressive effect of distinct bradykinin B2 receptor antagonist on allergen-evoked exudation and leukocyte infiltration in sensitized rats. Br J Pharmacol 1999;127:315–20. [81] Wilson CN. Adenosine receptors and asthma in humans. Br J Pharmacol 2008;155:475–86. [82] Kumar RK, Herbert C, Foster PS. The “classical” ovalbumin challenge model of asthma in mice. Curr Drug Targets 2008;9:485–94. [83] Muller E, Bergmann KC, Lachmann B, Vogel J. Experimental model of bronchial asthma (author’s transl). Z Erkr Atmungsorgane 1976;144:246–53. [84] Beaver PC, Snyder CH, Carrera GM, Dent JH, Lafferty JW. Chronic eosinophilia due to visceral larva migrans; report of three cases. Pediatrics 1952;9:7–19. [85] Pinelli E, Dormans J, Fonville M, van der Giessen J. A comparative study of toxocariasis and allergic asthma in murine models. J Helminthol 2001;75:137–40. [86] Kayes SG, Oaks JA. Development of the granulomatous response in murine toxocariasis. Initial events. Am J Pathol 1978;93:277–94. [87] Medeiros AI, Silva CL, Malheiro A, Maffei CM, Faccioli LH. Leukotrienes are involved in leukocyte recruitment induced by live Histoplasma capsulatum or by the beta-glucan present in their cell wall. Br J Pharmacol 1999;128:1529–37. [88] Medeiros AI, Malheiro A, Jose PJ, Conroy DM, Williams TJ, Faccioli LH. Differential release of MIP-1alpha and eotaxin during infection of mice by Histoplasma capsulatum or inoculation of beta-glucan. Inflamm Res 2004;53:351–4. [89] Iwama T, Nagai H, Suda H, Tsuruoka N, Koda A. Effect of murine recombinant interleukin-5 on the cell population in guinea-pig airways. Br J Pharmacol 1992;105:19–22. [90] Collins PD, Weg VB, Faccioli LH, Watson ML, Moqbel R, Williams TJ. Eosinophil accumulation induced by human interleukin-8 in the guinea-pig in vivo. Immunology 1993;79:312–8. [91] Griffiths-Johnson DA, Collins PD, Rossi AG, Jose PJ, Williams TJ. The chemokine, eotaxin, activates guinea-pig eosinophils in vitro and causes their accumulation into the lung in vivo. Biochem Biophys Res Commun 1993;197:1167– 72. [92] Richards IM, Sun FF, Taylor BM, Shields SK, Griffin RL, Morris J, et al. Contribution of leukotriene B4 to airway inflammation and the effect of antagonists. Ann N Y Acad Sci 1991;629:274–87. [93] Martins MA, Pasquale CP, e Silva PM, Pires AL, Ruffie C, Rihoux JP, et al. Interference of cetirizine with the late eosinophil accumulation induced by either PAF or compound 48/80. Br J Pharmacol 1992;105:176–80. [94] Jaracz S, Stromgaard K, Nakanishi K. Ginkgolides: selective acetylations, translactonization, and biological evaluation. J Org Chem 2002;67:4623–6. [95] Tang Y, Xu Y, Xiong S, Ni W, Chen S, Gao B, et al. The effect of Ginkgo biloba extract on the expression of PKCalpha in the inflammatory cells and the level of IL-5 in induced sputum of asthmatic patients. J Huazhong Univ Sci Technol Med Sci 2007;27:375–80. [96] Li MH, Zhang HL, Yang BY. Effects of ginkgo leave concentrated oral liquor in treating asthma. Zhongguo Zhong Xi Yi Jie He Za Zhi 1997;17:216–8. [97] Weng XJ, Chen LL, Zhang HQ. Effect of total flavonoid in leaves of Ginkgo biloba on the apoptosis of eosinophil in broncho alveloar lavage fluid. Yao Xue Xue Bao 2008;43:480–3. [98] Kurihara K, Wardlaw AJ, Moqbel R, Kay AB. Inhibition of platelet-activating factor (PAF)-induced chemotaxis and PAF binding to human eosinophils and neutrophils by the specific ginkgolide-derived PAF antagonist, BN 52021. J Allergy Clin Immunol 1989;83:83–90. [99] Lagente V, Boichot E, Carre C, Guinot P, Mencia-Huerta JM, Braquet P. Effects of the platelet activating factor antagonists BN 52021 and BN 50730 on antigen-

306

[100]

[101]

[102] [103]

[104] [105] [106]

[107]

[108] [109]

[110]

[111]

[112]

[113]

[114]

[115] [116]

[117]

[118]

[119]

[120] [121]

[122] [123]

[124]

[125]

[126]

[127]

[128]

A.P. Rogerio et al. / Pharmacological Research 62 (2010) 298–307 induced bronchial hyperresponsiveness and eosinophil infiltration in lung from sensitized guinea-pigs. Clin Exp Allergy 1993;23:1002–10. Hsieh KH. Effects of PAF antagonist, BN52021, on the PAF-, methacholine, and allergen-induced bronchoconstriction in asthmatic children. Chest 1991;99:877–82. Ali M, Thomson M, Afzal M. Garlic and onions: their effect on eicosanoid metabolism and its clinical relevance. Prostaglandins Leukot Essent Fatty Acids 2000;62:55–73. Stozkowska W, Raczynska K. Efficiency and application safety of Cepan cream. Observation of a new indication. Przegl Lek 2008;65:241–3. Kaiser P, Youssouf MS, Tasduq SA, Singh S, Sharma SC, Singh GD, et al. Anti-allergic effects of herbal product from Allium cepa (bulb). J Med Food 2009;12:374–82. Erlund I, Freese R, Marniemi J, Hakala P, Alfthan G. Bioavailability of quercetin from berries and the diet. Nutr Cancer 2006;54:13–7. Terao J, Kawai Y, Murota K. Vegetable flavonoids and cardiovascular disease. Asia Pac J Clin Nutr 2008;17(Suppl. 1):291–3. Egert S, Boesch-Saadatmandi C, Wolffram S, Rimbach G, Muller MJ. Serum lipid and blood pressure responses to quercetin vary in overweight patients by apolipoprotein E genotype. J Nutr 2010;140:278–84. Perez-Vizcaino F, Duarte J, Jimenez R, Santos-Buelga C, Osuna A. Antihypertensive effects of the flavonoid quercetin. Pharmacol Rep 2009;61:67– 75. Gabor M. Anti-inflammatory and anti-allergic properties of flavonoids. Prog Clin Biol Res 1986;213:471–80. Rogerio AP, Kanashiro A, Fontanari C, da Silva EV, Lucisano-Valim YM, Soares EG, et al. Anti-inflammatory activity of quercetin and isoquercitrin in experimental murine allergic asthma. Inflamm Res 2007;56:402–8. Moon H, Choi HH, Lee JY, Moon HJ, Sim SS, Kim CJ. Quercetin inhalation inhibits the asthmatic responses by exposure to aerosolized-ovalbumin in conscious guinea-pigs. Arch Pharm Res 2008;31:771–8. Jung CH, Lee JY, Cho CH, Kim CJ. Anti-asthmatic action of quercetin and rutin in conscious guinea-pigs challenged with aerosolized ovalbumin. Arch Pharm Res 2007;30:1599–607. Park HJ, Lee CM, Jung ID, Lee JS, Jeong YI, Chang JH, et al. Quercetin regulates Th1/Th2 balance in a murine model of asthma. Int Immunopharmacol 2009;9:261–7. Zare A, Farzaneh P, Pourpak Z, Zahedi F, Moin M, Shahabi S, et al. Purified aged garlic extract modulates allergic airway inflammation in BALB/c mice. Iran J Allergy Asthma Immunol 2008;7:133–41. Albuquerque DA, Juliani JM, Santos JA, Hosida PY, Borges S, Borralho CT. Efeito do extrato etanólico de Lafoensia pacari sobre a peritonite aguda em camundongos. 3a Reunião Especial da SBPC “Ecossistemas Costeiros - do Conhecimento a Gestão”. In: Programa e Anais. 1996. Solon S, Lopes L, Teixeira de Sousa Jr P, Schmeda-Hirschmann G. Free radical scavenging activity of Lafoensia pacari. J Ethnopharmacol 2000;72:173–8. da Mota Menezes V, Atallah AN, Lapa AJ, Catapani WR. Assessing the therapeutic use of Lafoensia pacari St. Hil. Extract (mangava-brava) in the eradication of Helicobacter pylori: double-blind randomized clinical trial. Helicobacter 2006;11:188–95. Rogerio AP, Sa-Nunes A, Albuquerque DA, Soares EG, Faccioli LH. Antieosinophilic effect of Lafoensia pacari in toxocariasis. Phytomedicine 2008;15:348–57. Rogerio AP, Fontanari C, Borducchi E, Keller AC, Russo M, Soares EG, et al. Antiinflammatory effects of Lafoensia pacari and ellagic acid in a murine model of asthma. Eur J Pharmacol 2008;580:262–70. Rogerio AP, Fontanari C, Melo MC, Ambrosio SR, de Souza GE, Pereira PS, et al. Anti-inflammatory, analgesic and anti-oedematous effects of Lafoensia pacari extract and ellagic acid. J Pharm Pharmacol 2006;58:1265–73. Choi YH, Yan GH. Ellagic acid attenuates immunoglobulin E-mediated allergic response in mast cells. Biol Pharm Bull 2009;32:1118–21. Gali HU, Perchellet EM, Klish DS, Johnson JM, Perchellet JP. Hydrolyzable tannins: potent inhibitors of hydroperoxide production and tumor promotion in mouse skin treated with 12-O-tetradecanoylphorbol-13-acetate in vivo. Int J Cancer 1992;51:425–32. Bell C, Hawthorne S. Ellagic acid, pomegranate and prostate cancer—a mini review. J Pharm Pharmacol 2008;60:139–44. Aviram M, Rosenblat M, Gaitini D, Nitecki S, Hoffman A, Dornfeld L, et al. Pomegranate juice consumption for 3 years by patients with carotid artery stenosis reduces common carotid intima-media thickness, blood pressure and LDL oxidation. Clin Nutr 2004;23:423–33. Laurie SA, Miller VA, Grant SC, Kris MG, Ng KK. Phase I study of green tea extract in patients with advanced lung cancer. Cancer Chemother Pharmacol 2005;55:33–8. Moyers SB, Kumar NB. Green tea polyphenols and cancer chemoprevention: multiple mechanisms and endpoints for phase II trials. Nutr Rev 2004;62:204–11. Luo H, Tang L, Tang M, Billam M, Huang T, Yu J, et al. Phase IIA chemoprevention trial of green tea polyphenols in high-risk individuals of liver cancer: modulation of urinary excretion of green tea polyphenols and 8hydroxydeoxyguanosine. Carcinogenesis 2006;27:262–8. Dona M, Dell’Aica I, Calabrese F, Benelli R, Morini M, Albini A, et al. Neutrophil restraint by green tea: inhibition of inflammation, associated angiogenesis, and pulmonary fibrosis. J Immunol 2003;170:4335–41. Oak MH, El Bedoui J, Schini-Kerth VB. Antiangiogenic properties of natural polyphenols from red wine and green tea. J Nutr Biochem 2005;16:1–8.

[129] Bani D, Giannini L, Ciampa A, Masini E, Suzuki Y, Menegazzi M, et al. Epigallocatechin-3-gallate reduces allergen-induced asthma-like reaction in sensitized guinea pigs. J Pharmacol Exp Ther 2006;317:1002–11. [130] Barnes PJ. Theophylline: new perspectives for an old drug. Am J Respir Crit Care Med 2003;167:813–8. [131] Guillot C, Fornaris M, Badier M, Orehek J. Spontaneous and provoked resistance to isoproterenol in isolated human bronchi. J Allergy Clin Immunol 1984;74:713–8. [132] Sullivan P, Bekir S, Jaffar Z, Page C, Jeffery P, Costello J. Antiinflammatory effects of low-dose oral theophylline in atopic asthma. Lancet 1994;343:1006–8. [133] Kirkpatrick P. Antibacterial drugs: stitching together naturally. Nat Rev Drug Discov 2002;1:748. [134] Cragg GM, Newman DJ, Snader KM. Natural products in drug discovery and development. J Nat Prod 1997;60:52–60. [135] Rates SM. Plants as source of drugs. Toxicon 2001;39:603–13. [136] Basso LA, da Silva LH, Fett-Neto AG, de Azevedo Jr WF, Moreira Ide S, Palma MS, et al. The use of biodiversity as source of new chemical entities against defined molecular targets for treatment of malaria, tuberculosis, and T-cell mediated diseases—a review. Mem Inst Oswaldo Cruz 2005;100:475–506. [137] Hamburger M, Hostettmann K. Analytical aspects of drugs of natural origin. J Pharm Biomed Anal 1989;7:1337–49. [138] Koehn FE, Carter GT. The evolving role of natural products in drug discovery. Nat Rev Drug Discov 2005;4:206–20. [139] Merfort I. Review of the analytical techniques for sesquiterpenes and sesquiterpene lactones. J Chromatogr A 2002;967:115–30. [140] Potterat O, Hamburger M. Drug discovery and development with plantderived compounds. Prog Drug Res 2008;65(45):47–118. [141] Calixto JB, Beirith A, Ferreira J, Santos AR, Filho VC, Yunes RA. Naturally occurring antinociceptive substances from plants. Phytother Res 2000;14: 401–18. [142] Calixto JB, Campos MM, Otuki MF, Santos AR. Anti-inflammatory compounds of plant origin. Part II. Modulation of pro-inflammatory cytokines, chemokines and adhesion molecules. Planta Med 2004;70:93–103. [143] Seelinger G, Merfort I, Schempp CM. Anti-oxidant, anti-inflammatory and anti-allergic activities of luteolin. Planta Med 2008;74:1667–77. [144] Verpoorte R. Exploration of nature’s chemodiversity: the role of secondary metabolites as leads in drug development. Drug Discov Today 1999;3:232–8. [145] Kung TT, Jones H, Adams 3rd GK, Umland SP, Kreutner W, Egan RW, et al. Characterization of a murine model of allergic pulmonary inflammation. Int Arch Allergy Immunol 1994;105:83–90. [146] Das AM, Flower RJ, Hellewell PG, Teixeira MM, Perretti M. A novel murine model of allergic inflammation to study the effect of dexamethasone on eosinophil recruitment. Br J Pharmacol 1997;121:97–104. [147] Facincone S, De Siqueira AL, Jancar S, Russo M, Barbuto JA, Mariano M. A novel murine model of late-phase reaction of immediate hypersensitivity. Mediators Inflamm 1997;6:127–33. [148] Kodama T, Matsuyama T, Miyata S, Nishimura H, Nishioka Y, Kitada O, et al. Kinetics of apoptosis in the lung of mice with allergic airway inflammation. Clin Exp Allergy 1998;28:1435–43. [149] Klein A, Talvani A, Cara DC, Gomes KL, Lukacs NW, Teixeira MM. Stem cell factor plays a major role in the recruitment of eosinophils in allergic pleurisy in mice via the production of leukotriene B4. J Immunol 2000;164:4271– 6. [150] Rogerio AP, Cardoso CR, Fontanari C, Souza MA, Afonso-Cardoso SR, Silva EV, et al. Anti-asthmatic potential of a d-galactose-binding lectin from Synadenium carinatum latex. Glycobiology 2007;17:795–804. [151] Anibal FF, Rogerio AP, Malheiro A, Machado ER, Martins-Filho OA, Andrade MC, et al. Impact of MK886 on eosinophil counts and phenotypic features in toxocariasis. Scand J Immunol 2007;65:344–52. [152] Malheiro A, Anibal FF, Martins-Filho OA, Teixeira-Carvalho A, Perini A, Martins MA, et al. Pcdna-IL-12 vaccination blocks eosinophilic inflammation but not airway hyperresponsiveness following murine Toxocara canis infection. Vaccine 2008;26:305–15. [153] Sa-Nunes A, Medeiros AI, Faccioli LH. Interleukin-5 mediates peritoneal eosinophilia induced by the F1 cell wall fraction of Histoplasma capsulatum. Braz J Med Biol Res 2004;37:343–6. [154] Oliveira SH, Faccioli LH, Cunha FQ, Ferreira SH. Role of resident peritoneal cells in eosinophil migration induced by saline. Int Arch Allergy Immunol 1994;104:323–31. [155] Fukuyama S, Inoue H, Aizawa H, Oike M, Kitaura M, Yoshie O, et al. Effect of eotaxin and platelet-activating factor on airway inflammation and hyperresponsiveness in guinea pigs in vivo. Am J Respir Crit Care Med 2000;161:1844–9. [156] Bozza PT, Castro-Faria-Neto HC, Pires AL, Silva PM, Martins MA, Cordeiro RS. Endotoxin induces eosinophil accumulation in the rat pleural cavity. Braz J Med Biol Res 1991;24:957–60. [157] Lagente V, Pruniaux MP, Junien JL, Moodley I. Modulation of cytokine-induced eosinophil infiltration by phosphodiesterase inhibitors. Am J Respir Crit Care Med 1995;151:1720–4. [158] Collins PD, Marleau S, Griffiths-Johnson DA, Jose PJ, Williams TJ. Cooperation between interleukin-5 and the chemokine eotaxin to induce eosinophil accumulation in vivo. J Exp Med 1995;182:1169–74. [159] Oliveira SH, Faccioli LH, Ferreira SH, Cunha FQ. Participation of interleukin-5, interleukin-8 and leukotriene B4 in eosinophil accumulation in two different experimental models. Mem Inst Oswaldo Cruz 1997;92(Suppl. 2):205–10.

A.P. Rogerio et al. / Pharmacological Research 62 (2010) 298–307 [160] de Paula L, Santos WF, Malheiro A, Carlos D, Faccioli LH. Differential modulation of cell recruitment and acute edema in a model of Polybia paulista venom-induced inflammation. Int Immunopharmacol 2006;6:182–9. [161] Laycock SM, Smith H, Spicer BA. Airway hyper-reactivity and blood, lung and airway eosinophilia in rats treated with sephadex particles. Int Arch Allergy Appl Immunol 1986;81:363–7. [162] Laycock SM, Smith H, Spicer BA. Airway hyper-reactivity and eosinophilia in rats treated with sephadex particles. Int Arch Allergy Appl Immunol 1987;82:347–8. [163] Cook RM, Musgrove NR, Smith H. Eosinophils and the granulomatous reaction in rats injected with sephadex particles. Pulm Pharmacol 1989;2:185–90. [164] Spicer BA, Baker RC, Hatt PA, Laycock SM, Smith H. The effects of drugs on sephadex-induced eosinophilia and lung hyper-responsiveness in the rat. Br J Pharmacol 1990;101:821–8. [165] Oliveira SH, Costa CH, Ferreira SH, Cunha FQ. Sephadex induces eosinophil migration to the rat and mouse peritoneal cavity: involvement of mast cells, LTB4, TNF-alpha, IL-8 and PAF. Inflamm Res 2002;51:144–53. [166] Ferreira HH, Medeiros MV, Lima CS, Flores CA, Sannomiya P, Autunes E, et al. Inhibition of eosinophil chemotaxis by chronic blockade of nitric oxide biosynthesis. Eur J Pharmacol 1996;310:201–7. [167] e Silva PM, Martins MA, Castro-Faria-Neto HC, Bozza PT, Pires AL, Cordeiro RS, et al. Nedocromil sodium prevents in vivo generation of the eosinophilotactic substance induced by PAF but fails to antagonize its effects. Br J Pharmacol 1992;105:436–40. [168] Mello GC, Desouza IA, Mariano NS, Ferreira T, Macedo ML, Antunes E. Mechanisms involved in the rat peritoneal leukocyte migration induced by a kunitz-type inhibitor isolated from Dimorphandra mollis seeds. Toxicon 2009;53:323–9. [169] Kim D, Kim SH, Park EJ, Kang CY, Cho SH, Kim S. Anti-allergic effects of PG102, a water-soluble extract prepared from Actinidia arguta, in a murine ovalbumininduced asthma model. Clin Exp Allergy 2009;39:280–9. [170] Jin MH, Yook J, Lee E, Lin CX, Quan Z, Son KH, et al. Anti-inflammatory activity of Ailanthus altissima in ovalbumin-induced lung inflammation. Biol Pharm Bull 2006;29:884–8. [171] Kim JY, Kim DY, Lee YS, Lee BK, Lee KH, Ro JY. DA-9601, Artemisia asiatica herbal extract, ameliorates airway inflammation of allergic asthma in mice. Mol Cells 2006;22:104–12. [172] Gupta I, Gupta V, Parihar A, Gupta S, Ludtke R, Safayhi H, et al. Effects of Boswellia serrata gum resin in patients with bronchial asthma: results of a double-blind, placebo-controlled, 6-week clinical study. Eur J Med Res 1998;3:511–4. [173] Ok IS, Kim SH, Kim BK, Lee JC, Lee YC. Pinellia ternata, Citrus reticulata, and their combinational prescription inhibit eosinophil infiltration and airway hyperresponsiveness by suppressing CCR3+ and Th2 cytokines production in the ovalbumin-induced asthma model. Mediators Inflamm 2009;2009:413270. [174] Sa-Nunes A, Rogerio AP, Medeiros AI, Fabris VE, Andreu GP, Rivera DG, et al. Modulation of eosinophil generation and migration by Mangifera indica L. extract (Vimang). Int Immunopharmacol 2006;6:1515–23. [175] Vieira-de-Abreu A, Amendoeira FC, Gomes GS, Zanon C, Chedier LM, Figueiredo MR, et al. Anti-allergic properties of the Bromeliaceae Nidularium procerum: inhibition of eosinophil activation and influx. Int Immunopharmacol 2005;5:1966–74. [176] Lee SH, Choi SM, Sohn YS, Kang KK, Yoo M. Effect of Oryza sativa extract on the progression of airway inflammation and remodeling in an experimental animal model of asthma. Planta Med 2006;72:405–10. [177] Sanbongi C, Takano H, Osakabe N, Sasa N, Natsume M, Yanagisawa R, et al. Rosmarinic acid in Perilla extract inhibits allergic inflammation induced by mite allergen, in a mouse model. Clin Exp Allergy 2004;34:971–7. [178] Thomet OA, Schapowal A, Heinisch IV, Wiesmann UN, Simon HU. Antiinflammatory activity of an extract of Petasites hybridus in allergic rhinitis. Int Immunopharmacol 2002;2:997–1006. [179] Brattstrom A, Schapowal A, Maillet I, Schnyder B, Ryffel B, Moser R. Petasites extract Ze 339 (PET) inhibits allergen-induced Th2 responses, airway inflammation and airway hyperreactivity in mice. Phytother Res 2010;24:680–5. [180] Li D, Xiong SD, Du DB. Inhibitory effects of Salvia miltiorrhiza injection coordinated with dexamethasone on interleukin-13 and eotaxin expression in lung of asthmatic rats. Zhongguo Zhong Xi Yi Jie He Za Zhi 2006;26:1007–10. [181] Brito FA, Lima LA, Ramos MF, Nakamura MJ, Cavalher-Machado SC, Siani AC, et al. Pharmacological study of anti-allergic activity of Syzygium cumini (L.) Skeels. Braz J Med Biol Res 2007;40:105–15. [182] Lin KX, Wang CZ, Qian GS. Effect of Tripterygium wilfordii on Th1, Th2 cytokines production in asthma patients. Zhongguo Zhong Xi Yi Jie He Za Zhi 2001;21:22–4.

307

[183] Ahui ML, Champy P, Ramadan A, Pham Van L, Araujo L, Brou Andre K, et al. Ginger prevents Th2-mediated immune responses in a mouse model of airway inflammation. Int Immunopharmacol 2008;8:1626–32. [184] Bao Z, Guan S, Cheng C, Wu S, Wong SH, Kemeny DM, et al. A novel antiinflammatory role for andrographolide in asthma via inhibition of the nuclear factor-kappab pathway. Am J Respir Crit Care Med 2009;179:657–65. [185] Choi JR, Lee CM, Jung ID, Lee JS, Jeong YI, Chang JH, et al. Apigenin protects ovalbumin-induced asthma through the regulation of GATA-3 gene. Int Immunopharmacol 2009;9:918–24. [186] Du Q, Chen Z, Zhou LF, Zhang Q, Huang M, Yin KS. Inhibitory effects of astragaloside IV on ovalbumin-induced chronic experimental asthma. Can J Physiol Pharmacol 2008;86:449–57. [187] Jung WK, Lee DY, Choi YH, Yea SS, Choi I, Park SG, et al. Caffeic acid phenethyl ester attenuates allergic airway inflammation and hyperresponsiveness in murine model of ovalbumin-induced asthma. Life Sci 2008;82:797–805. [188] Moon DO, Kim MO, Lee HJ, Choi YH, Park YM, Heo MS, et al. Curcumin attenuates ovalbumin-induced airway inflammation by regulating nitric oxide. Biochem Biophys Res Commun 2008;375:275–9. [189] Duan W, Kuo IC, Selvarajan S, Chua KY, Bay BH, Wong WS. Antiinflammatory effects of genistein, a tyrosine kinase inhibitor, on a guinea pig model of asthma. Am J Respir Crit Care Med 2003;167:185–92. [190] Ram A, Mabalirajan U, Das M, Bhattacharya I, Dinda AK, Gangal SV, et al. Glycyrrhizin alleviates experimental allergic asthma in mice. Int Immunopharmacol 2006;6:1468–77. [191] Rogerio AP, Andrade EL, Leite DF, Figueiredo CP, Calixto JB. Preventive and therapeutic anti-inflammatory properties of the sesquiterpene alphahumulene in experimental airways allergic inflammation. Br J Pharmacol 2009;158:1074–87. [192] Vasconcelos JF, Teixeira MM, Barbosa-Filho JM, Lucio AS, Almeida JR, de Queiroz LP, et al. The triterpenoid lupeol attenuates allergic airway inflammation in a murine model. Int Immunopharmacol 2008;8:1216–21. [193] Das M, Ram A, Ghosh B. Luteolin alleviates bronchoconstriction and airway hyperreactivity in ovalbumin sensitized mice. Inflamm Res 2003;52:101–6. [194] Jin M, Yang JH, Lee E, Lu Y, Kwon S, Son KH, et al. Antiasthmatic activity of luteolin-7-O-glucoside from Ailanthus altissima through the downregulation of T helper 2 cytokine expression and inhibition of prostaglandin E(2) production in an ovalbumin-induced asthma model. Biol Pharm Bull 2009;32:1500–3. [195] Funaguchi N, Ohno Y, La BL, Asai T, Yuhgetsu H, Sawada M, et al. Narirutin inhibits airway inflammation in an allergic mouse model. Clin Exp Pharmacol Physiol 2007;34:766–70. [196] Medeiros KC, Faustino L, Borduchi E, Nascimento RJ, Silva TM, Gomes E, et al. Preventive and curative glycoside kaempferol treatments attenuate the Th2-driven allergic airway disease. Int Immunopharmacol 2009;9:1540–8. [197] Mishra NC, Rir-Sima-Ah J, Langley RJ, Singh SP, Pena-Philippides JC, Koga T, et al. Nicotine primarily suppresses lung Th2 but not goblet cell and muscle cell responses to allergens. J Immunol 2008;180:7655–63. [198] Wu YQ, Zhou CH, Tao J, Li SN. Antagonistic effects of nobiletin, a polymethoxyflavonoid, on eosinophilic airway inflammation of asthmatic rats and relevant mechanisms. Life Sci 2006;78:2689–96. [199] Kim SH, Lee YC. Piperine inhibits eosinophil infiltration and airway hyperresponsiveness by suppressing T cell activity and th2 cytokine production in the ovalbumin-induced asthma model. J Pharm Pharmacol 2009;61:353–9. [200] Lee H, Han AR, Kim Y, Choi SH, Ko E, Lee NY, et al. A new compound, 1H,8H-pyrano[3,4-c]pyran-1,8-dione, suppresses airway epithelial cell inflammatory responses in a murine model of asthma. Int J Immunopathol Pharmacol 2009;22:591–603. [201] Rogerio AP, Dora CL, Andrade EL, Chaves JS, Silva LF, Lemos-Senna E, et al. Anti-inflammatory effect of quercetin-loaded microemulsion in the airways allergic inflammatory model in mice. Pharmacol Res 2009;61:288–97. [202] Ko WC, Shih CM, Chen MC, Lai YH, Chen JH, Chen CM, et al. Suppressive effects of 3-O-methylquercetin on ovalbumin-induced airway hyperresponsiveness. Planta Med 2004;70:1123–7. [203] El Gazzar M, El Mezayen R, Marecki JC, Nicolls MR, Canastar A, Dreskin SC. Anti-inflammatory effect of thymoquinone in a mouse model of allergic lung inflammation. Int Immunopharmacol 2006;6:1135–42. [204] Bezerra-Santos CR, Vieira-de-Abreu A, Barbosa-Filho JM, Bandeira-Melo C, Piuvezam MR, Bozza PT. Anti-allergic properties of Cissampelos sympodialis and its isolated alkaloid warifteine. Int Immunopharmacol 2006;6: 1152–60. [205] Machado ER, Ueta MT, Lourenc¸o EV, Anibal FF, Sorgi CA, Soares EG, RoqueBarreira MC, Medeiros AI, Faccioli LH. Leukotrienes play a role in the control of parasite burden in strongyloidiasis. J Immunol 2005;175:3892–9.