Skip to main content

Prospect of vasoactive intestinal peptide therapy for COPD/PAH and asthma: a review

Abstract

There is mounting evidence that pulmonary arterial hypertension (PAH), asthma and chronic obstructive pulmonary disease (COPD) share important pathological features, including inflammation, smooth muscle contraction and remodeling. No existing drug provides the combined potential advantages of reducing vascular- and bronchial-constriction, and anti-inflammation. Vasoactive intestinal peptide (VIP) is widely expressed throughout the cardiopulmonary system and exerts a variety of biological actions, including potent vascular and airway dilatory actions, potent anti-inflammatory actions, improving blood circulation to the heart and lung, and modulation of airway secretions. VIP has emerged as a promising drug candidate for the treatment of cardiopulmonary disorders such as PAH, asthma, and COPD. Clinical application of VIP has been limited in the past for a number of reasons, including its short plasma half-life and difficulty in administration routes. The development of long-acting VIP analogues, in combination with appropriate drug delivery systems, may provide clinically useful agents for the treatment of PAH, asthma, and COPD. This article reviews the physiological significance of VIP in cardiopulmonary system and the therapeutic potential of VIP-based agents in the treatment of pulmonary diseases.

1. Introduction

Vasoactive intestinal peptide (VIP) is a 28-amino-acid peptide, which was first isolated from upper intestine, and has been characterized as a vasodilatory peptide [1]. VIP has a very widespread distribution in the central and peripheral nervous systems [2]. It is one of the most abundant neuropeptides found in the cardiovascular system and airways [2–5]. This neuropeptide exerts a wide range of biological actions, such as positive inotropic and chronotropic effects, pulmonary and coronary vasodilatation, bronchodilation, and anti-inflammatory effects, and thus it influences many aspects of cardiopulmonary function [6–8]. Studies using VIP deficient animals and using animal models of diseases have indicated that VIP has significant therapeutic potential in the treatment of cardiopulmonary diseases, including pulmonary arterial hypertension (PAH), chronic obstructive pulmonary disease (COPD) and asthma [9–11].

Clinical manifestation of PAH

PAH is a disabling chronic disorder of the pulmonary vasculature, which is characterized by abnormal pulmonary vascular proliferation and remodeling, vasoconstriction, perivascular inflammation, and thrombosis, leading to elevated pulmonary arterial pressure, increases in peripheral vascular resistance, and it ultimately results in right heart failure and death [12, 13]. The past two decades have seen significant advances with the development and clinical implementation of a number of medications for the treatment of PAH: prostanoids, endothelin-1 receptor antagonists, and phosphodiesterase type 5 inhibitors. However, the results remain unsatisfactory, with persistent high mortality, insufficient clinical improvement and no convincing report of any reversal of the disease process [12, 13]. In addition, the current PAH therapy requires a cocktail of drugs to manage PAH symptoms and often leads to drug intolerance [14]. Therefore, it is necessary to develop additional novel therapeutic approaches that target the various components of this multifactorial disease. VIP provides the combined potential advantages of lowering pulmonary arterial pressure, improving blood circulation to the heart and lung, reducing inflammation of the heart and lung tissues, and is readily accepted by the body because it is natural to it [1–8]. Based on its multiple biological actions, the development of controlled release airway drug-delivery system with VIP has emerged as a novel therapeutic strategy for the treatment of PAH.

Clinical manifestation of COPD and asthma

Chronic inflammatory airway diseases such as bronchial asthma or COPD are major contributors to the global burden of disease. COPD is characterized by a chronic, slowly progressive airway disorder resulting from a combination of pulmonary emphysema and irreversible reduction in the caliber of the small airways of the lung, resulting in airflow limitation [15]. Asthma is a complex, persistent, inflammatory disease characterized by airway hyperresponsiveness in association with airway inflammation. Although there are many allopathic treatments, including bronchodilators and corticosteroids, there is no single medication that is effective against both the inflammatory and bronchoconstrictive components of asthma [16]. VIP exerts functions not only as a vasodilator and bronchodilator but also as a potent immunomodulator [1, 7, 8], thus VIP has significant therapeutic potential in the treatment of pulmonary diseases, including: PAH, asthma and COPD. However, VIP-based drugs are not yet in clinical use, possibly because the poor metabolic stability and difficulty in administration routes. The development of long-acting VIP analogues, in combination with appropriate drug delivery systems, may provide clinically useful agents for the treatment of PAH/asthma/COPD. This article reviews the physiological significance of VIP in cardiopulmonary system and the therapeutic potential of VIP-based agents in the treatment of pulmonary diseases.

2. Expression and distribution of VIP in cardiovascular-pulmonary system

VIP is co-localized with acetylcholine in postganglionic parasympathetic neurons in the cardiovascular and respiratory systems [17]. In the mammalian heart, VIP was found in nerve fibers associated with atrial and ventricular myocardium, conduction system, and coronary vessels [18–21]. Immunofluorescent and radioimmunoassay studies have localized VIP to neuronal cell bodies of the intrinsic cardiac ganglia, axons and dendrites, and presynaptic nerve terminals from which VIP is released as a nonadrenergic-noncholinergic neurotransmitter [22]. In the peripheral nervous system, VIP is present in sympathetic ganglia, the vagus nerves, some motor nerves such as the sciatic nerve, autonomic nerves that supply exocrine glands, vascular and nonvascular smooth muscle, and ganglion-like clusters of neuronal cell bodies that provide 'intrinsic' organ innervation [18, 23].

VIP is abundantly present in normal human lungs [1, 2, 24]. VIP-immunoreactivity (IR)-containing cells are present in the tracheobronchial smooth muscle layer and glands of airways, and within the walls of pulmonary and bronchial vessels [25, 26]. VIP-IR nerve fibers are found as branching networks in the respiratory tract [4]. The frequency of these VIP-ergic fibers decreases as the airways become smaller, and only a few VIP-ergic fibers are present in bronchioles and alveolar space [26]. The pattern of VIP-ergic nerve fiber distribution largely follows that of cholinergic nerves, which is consistent with the colocalization of VIP with acetylcholine [27]. VIP is also co-localized with nitric oxide synthase (NOS) in human and guinea-pig airways [28–30]. In human airways, a co-localized immunoreactivity of VIP and NOS is found in airway intrinsic neuronal perikarya [28, 30]. Furthermore, VIP has also been identified in some sensory nerves, including sub-epithelial airway nerves [27, 31]; as well as in immune cells such as mast cells [32], eosinophils [33, 34], and in different mononuclear cells and polymorphonuclear leukocytes [35]. A deficiency of VIP in the respiratory system is considered to be a pathogenetic factor in pulmonary disease [36, 37].

3. VIP release and metabolism

Circulating VIP in men is found in low plasma levels. However, an increase in plasma concentration has been detected in conditions, such as gastrointestinal stimulation, during strenuous exercise, acute myocardial infarction and gastrointestinal tumors [38–41]. Circulating VIP is produced from VIP-containing nerve fibers. Many VIP-containing nerves have a perivascular distribution and it thus seems likely that VIP can exert important local effects without producing a detectable increase in systemic levels [42]. Myocardial blood vessels and also pulmonary blood vessels are innervated by VIP immunoreactive nerve fibers, which cause vascular smooth muscle dilation [18, 23]. Endogenous VIP is released by high frequency nerve stimulation and also is released by neostigmine, as well as by serotonin, dopaminergic agonists such as bromocriptine and apomorphine, prostaglandins (PGE, PGD) and nerve growth factor [43, 44].

Under physiological conditions, VIP is mainly cleaved by endopeptidase, whereas in states of airway inflammation, mast cell enzymes dominate the degradation of VIP [45–47]. VIP is readily degraded by enzymes, including neutral endopeptidase, mast cell-derived tryptase and chymase, thus preventing it from relaxing vascular or tracheal smooth muscle [45–47].

4. VIP receptors in cardiovascular-pulmonary system

The biological effects of VIP are mediated by two type II G-protein-coupled receptors: VPAC1 and VPAC2 [48]. Stimulation of VPAC receptors by VIP causes dose-dependent activation of adenylate cyclase, which increases cAMP concentrations, and activates cAMP- and cGMP-dependent protein kinases and leads to smooth muscle relaxation via decreasing intracellular calcium levels [49]. While VIP binds both VPAC1 and VPAC2 receptors with high affinity, VIP can also bind with low affinity to the pituitary adenylate cyclase activating peptide (PACAP) receptor. PACAP is another secretin family member peptide that exhibits extensive similarities to VIP and shares VIP receptors and functions [50].

High densities of VIP binding sites were found in the pulmonary vascular smooth muscle layer and in airway smooth muscle of large, but not smaller airways. VIP binding sites are also present in sub-mucosal glands, airway epithelium and in alveolar walls [24, 51]. In the human upper respiratory tract, VIP receptors were found on submucosal glands, epithelial cells, and arterial but not sinusoidal vessels [5]. VIP receptors are also expressed in innate immune cell types, including human mast cells, neutrophils, and peripheral blood monocytes, and murine macrophages and dendritic cells [52–56]. VIP is thought to play a role in regulating immunity and inflammation. Studies using VPAC2 receptor knockout mice and transgenic mice overexpressing the VPAC2 receptor have revealed that the receptor regulates the balance between T-helper type 1 and 2 lymphocytes (Th1 and Th2 cells) by stimulating production of more Th2-type cytokines, which mediate hypersensitivity reactions (e.g. allergy) [57, 58]. Thus, this receptor is believed to play an important functional role in the respiratory tract by regulation of immune effects of VIP in allergic diseases such as allergic bronchial asthma.

The wide spread presence of VIP receptors in a variety of tissues and organ systems has led to the potential limitation of its clinical application. Intravenous administration of VIP has been shown to ameliorate histamine-induced bronchoconstriction in asthmatic subjects; while it also caused cardiovascular side effects by decreasing systemic blood pressure, inducing tachycardia and cutaneous flushing [59]. Thus, the development of effective drug delivery systems with airway delivery capability for VIP-based respiratory therapy represents a possible therapeutic strategy.

5. Role of VIP in heart and blood vessels

VIP is a potent vasodilator in coronary and pulmonary blood vessels, as well as other systemic blood vessels. The presence of VIP nerve fibers and their receptors in the coronary and pulmonary arteries strongly suggests that this peptide is important in the regulation of cardiopulmonary blood flow. VIP induces endothelium-independent relaxation in most of the vascular beds, including cat cerebral artery, dog isolated carotid artery, pig coronary artery, and bovine pulmonary artery [3–6].

There is direct evidence that VIP acts on heart muscle in various experimental system. VIP exerts a primary positive inotropic effect on cardiac muscle. In dogs, VIP infusion increases cardiac contractility and improves ventricular-vascular coupling, thus VIP enhances delivery of mechanical energy from the LV to the circulatory bed [60]. In isolated atrial or ventricular muscle, VIP, increases developed isometric force and is greater than isoproterenol in enhancing ventricular muscle contractile force [61]. VIP also exerts a primary positive chronotropic effect in the heart. Injection of VIP directly into the dog sinoatrial artery increases heart rate by 37%, VIP also dose-dependently shortens the atrioventricular conduction time, decreases the atrial and ventricular refractory periods [61, 62]. Endogenously released VIP increases atrial and ventricular contractility, and heart rate. Stimulation of the parasympathetic (vagal) nerves, during muscarinic and β-adrenergic receptor blockade in dogs, increases the atrial contractile force by 32%, increases heart rate by 37%, and also increases right ventricular contraction and relaxation by 28 and 33%, respectively [63, 64]. In patients with acute myocardial infarction, the VIP concentration in the plasma may increase by 33-62% within 6 h of the onset of symptoms [41]. Upon acute coronary ischemia, VIP is released from neurons in the coronary vessels and myocardium, and may also be released from the splanchnic viscera, and can act as a vasodilator to reduce myocardial ischemia [18, 65].

6. Biological actions of VIP in airway

VIP is a potent vasodilator of airway smooth muscle in vitro and in vivo. In isolated tracheal or bronchial segments, VIP attenuates the constrictor effect of histamine, prostaglandine F2α, endothelin, leukotriene D4, kallikrein and neurokinin A [66, 67]. The bronchodilatory effect of VIP in human bronchi is almost 100 times more potent than adrenergic dilatation by isoproterenol, and VIP is the most potent endogenous bronchodilator described so far [68]. VIP is also involved in the regulation of airway mucus secretion. High density VIP-expressing nerve fibers and VPAC2 mRNA have been found in airway submucosal glands [25, 69]. The role of VIP in airway mucus secretion has been controversial. VIP has been shown to have both stimulation and inhibition effects on airway secretion. In the human trachea, VIP inhibited methacholine-stimulated release of glycoproteins and lysozyme [70]. In the upper airways, VIP was shown to stimulate lactoferrin secretion from human nasal mucosal cells, but had little effects on mucous glycoprotein release [71]. VIP inhibits cholinergic secretion in ferret trachea, whereas it stimulates cholinergic secretion in the cat trachea [72, 73]. Therefore, the importance of VIP in airway mucus secretion appears to differ from species to markers examined. Future studies using human tissue and cells need to be performed in order to further elucidate the role of VIP on mucus secretion that associated with hypersecretory diseases such as COPD or asthma.

7. VIP in inflammatory response

Progressive pulmonary inflammation is the hallmark of airway diseases, including asthma, COPD and PAH. VIP has been shown to exert immunomodulating and anti-inflammatory activities through VIP specific receptors [74]. VIP inhibits the release of mediators from pulmonary mast cells, interacts with T lymphocytes, prevents lung injury due to xanthine oxidase and may act as a free radical scavenger [75–78]. VIP also inhibits the production of IL-6, IL-12, TNF alpha, and nitric oxide, and stimulates IL-10 production, and these effects are mostly mediated through the constitutively expressed VPAC1 receptor at the transcriptional level via modulation of NFκB and cAMP responsive element (CRE)-binding or ets-2 complexes [79]. Dunzendorfer et al. have suggested that VIP has an anti-inflammatory effect on eosinophils, reporting that VIP inhibited eosinophil migration and production of IL-16 in vitro, which subsequently inhibited chemotaxis of lymphocytes [80, 81]. Delgado et al. also reported that VIP inhibited LPS-induced inflammatory pathways in monocytes and macrophages via cAMP-dependent or independent mechanisms [55]. In addition, it has been suggested that VIP functions as an important T helper-differentiating factor that promotes Th2-like and inhibits Th1-like immune response via several mechanisms, including preferential survival of Th2 effectors and generation of memory Th2 cells [82]. In vitro studies show that VIP treatment leads to the induction of IL-4 and IL-5 in macrophages, and leads to the inhibition of IFN-gamma and IL-2 in antigen-primed CD4 T cells [83]. Mice lacking VPAC2 showed increased Th1-type responses which were characterized by an enhanced delayed type hypersensitivity and a diminished immediate-type hypersensitivity [58] In contrast, T cell over-expression of VPAC2 led to a deviation from the normal CD4 T cell cytokine expression profile toward a Th2-like profile with elevated blood IgE and IgG1 levels and increased eosinophil numbers. These transgenic mice also showed increased cutaneous allergic reactions, and a decreased delayed-type hypersensitivity [58]. Future study should further examine the immune-regulatory role of VIP using animal models with T cell-related diseases such as allergic asthma.

8. Therapeutic potential of VIP in PAH

The main pathological features of PAH in the pulmonary vasculature are perivascular inflammation, thrombosis, abnormal growth of vascular smooth muscle cells and extracellular matrix accumulation, leading to remodeling of the pulmonary vessel wall, obstruct pulmonary blood flow and ultimately cause right heart failure. Current treatment of PAH, which includes the use of prostacyclins, endothelin receptor antagonists, and phosphodiesterase type 5 inhibitors, either alone or in combination, have only limited efficacy in the improvement of clinical symptoms, hemodynamics, and long-term survival [12–14]. VIP has a large spectrum of biological functions including potent dilatory actions in pulmonary blood vessels and airway smooth muscles, potent anti-inflammatory actions, inhibition of vascular smooth muscle cell proliferation, enhancing would healing, regulation of cell growth and survival, and modulation of airway secretions. Therefore, using VIP-based drugs to target the various components of this multifactorial disease could be a novel therapeutic approach for the treatment of PAH.

In monocrotaline-induced pulmonary hypertension in rabbits, VIP dose-dependently decreased pulmonary artery pressure and pulmonary vascular resistance [83]. Application of VIP to patients with primary pulmonary hypertension results in substantial improvement of hemodynamic and prognostic parameters of the disease without side effects [36]. It decreased the mean pulmonary artery pressure in these patients, increased cardiac output, and mixed- venous oxygen saturation [36]. Said indicated that VIP gene is a key modulator of pulmonary vascular remodeling and inflammation [84]. Mice lacking VIP gene developed moderately severe PAH, with right ventricular hypertrophy, and thickened pulmonary artery, as well as perivascular inflammatory cell infiltrates in the lung [85]. Treatment of the mice with VIP attenuated both the vascular remodeling and right ventricular remodeling [85]. Right heart failure is a hallmark of severe PAH, and ultimately leading to death. In animals and in humans, infusion of VIP increases the epicardial coronary artery cross-sectional area by 27%, decreases coronary vascular resistance by 46%, and increases coronary artery blood flow by 200% [20, 86]. Application of VIP to patients also increases the left ventricular fraction shortening by 38% and significantly increases left ventricular contractility [86, 87]. Therefore, addition to its actions on decreasing pulmonary artery pressure, VIP also protects the heart.

9. Therapeutic potential of VIP in COPD/asthma

Chronic inflammatory airway diseases such as COPD and bronchial asthma continue to be an important cause of morbidity, mortality, and health-care cost worldwide. The key clinical features of asthma are airflow obstruction and airway hyperresponsiveness that caused by airway inflammation [16]. Many of the inflammatory events in asthma are thought to be mediated by Th2 cells. It also involves mast cells, eosinophils, neutrophils and mesenchymal cells such as epithelial cells, fibroblasts, smooth muscle cells and endothelial cells. The inflammatory mediators, including cytokines, chemokines, adhesion molecules, proteinases and growth factors released by these cells participant in this process at various stages and interact to maintain and amplify the inflammatory response [11]. Two categories of drugs are currently used in asthma therapies: bronchodilators and anti-inflammatory drugs. Despite the availability of these medications, the asthma epidemic continues to increase. The key clinical feature of COPD is airflow limitation results from airway constriction and irreversible reduction in the caliber of the small airways of the lung. Cigarette smoking is an important risk factor of COPD. The airflow limitation or obstruction that happens in COPD is caused by a mixture of small airway disease, parenchymal destruction (emphysema) and in many cases, increased airway responsiveness (asthma) [15]. Studies have shown that there is a large overlap of up to 30% between people who have a clinical diagnosis of COPD and asthma [88]. There is also a high incidence of mild to moderate PAH prevalence, reaching to 50% in advanced chronic obstructive COPD [89]. As Said suggested that PAH/asthma/COPD share important pathological features, including inflammation, smooth muscle contraction and remodeling [90]. Inflammation has long been acknowledged as a key feature of the asthma and COPD [11, 15, 16, 88, 89]. Perivascular inflammation has also been increasingly recognized as a significant component of clinical and experimental PAH phenotypes [91]. In these diseases there is increased resistance in, and narrowing of, airways and pulmonary arteries, respectively, due to airway and pulmonary vasoconstriction, smooth muscle constriction, and thickening of the walls caused by smooth muscle and other cell proliferation known as remodeling [90]. Muscularisation and remodeling of smaller pulmonary arteries are essential pathological lesions in PAH [92]. Airway remodeling caused by airway inflammation includes an increase in airway wall thickness, fibrosis, smooth muscle mass and vascularity, as well as abnormalities in extracellular matrix composition [89, 93]. These shared pathological features suggest possible common underlying mechanism among PAH/asthma/COPD.

Mice with targeted deletion of VIP gene, simultaneously express airway hyperresponsiveness with airway inflammation, together with PAH, pulmonary vascular remodeling and perivascular inflammation. Treatment of the mice with VIP reversed both sets of phenotypic changes, confirming that they result from the absence of the VIP gene [10, 84]. Recently, attention has been drawn to the therapeutic potential of VIP for the clinical treatment of COPD/asthma on the basis that VIP acts as a neurotransmitter, the dominant mechanism of human airway and vascular relaxation, and its anti-inflammatory properties. Neutrophil accumulation in the airway is a characteristic feature of COPD and asthma. VIP and its analogues have been shown to inhibit antigen- or cytokine-induced neutrophil recruitment in the airway in vivo [94]. VIP has also been shown to attenuate the cigarette smoke extract-induced apoptotic death of rat alveolar L2 cells, and protect against human bronchial epithelial cell damage, enhance airway wound healing [95, 96]. Recent studies show that inhalable powder formulation of VIP derivative, IK312532 attenuates airway inflammation in ovalbumin challenge-induced asthma/COPD -like rats and in cigarette smoke-exposed rats [9, 97, 98].

10. VIP for clinical use

The key to the therapeutic use of VIP in human disease is in its delivery. Firstly, VIP is degraded quickly by enzymes, catalytic antibodies, and spontaneous hydrolysis in biological fluids. Secondly, systemic administration of VIP has been shown to cause cardiovascular side effects [59]. To overcome the limited clinical effectiveness of native VIP, VIP incorporated into phospholipids has been used successfully in animal models of pulmonary hypertension [99]. Furthermore, several peptidase-resistant VIP-analogues have been developed [100]. VIP analogue, Ro 25-1553 causes a concentration-dependent relaxation of airway and pulmonary artery preparations, with an EC50 of approximately 10 nM and a maximal relaxation of 70%-75% of the induced tone [101]. In patients with asthma, inhalation of a selective VPAC2 receptor agonist Ro 25-1553 causes a bronchodilatory effect. The corresponding maximum bronchodilatory effect during 24 hours was similar for Ro 25-1553 and the reference bronchodilator formoterol (beta-2 adrenoceptor agonist). However, the bronchodilatory effect of Ro 25-1553 was attenuated 5 hours after inhalation whereas formoterol still had a bronchodilatory effect 12 hours after inhalation [102]. Therefore, the development of effective drug delivery systems for VIP-based respiratory therapy remains a significant challenge. It is possible to envisage that development of controlled-release biodegradable VIP-based drug system, particularly with airway delivery capability would have very significant therapeutic benefits in the treatment of cardiopulmonary diseases, including PAH, COPD and asthma.

11. Conclusion

This article describes the physiological significance of VIP and its therapeutic potential for the treatment of cardiopulmonary diseases, including PAH, asthma, and COPD. VIP exerts a variety of actions, including potent dilatory actions in pulmonary blood vessels and airway smooth muscles, potent anti-inflammatory and anti-proliferative actions, regulation of cell growth and survival, and modulation of airway secretions. PAH, asthma and COPD share key mechanisms of pathogenesis, including inflammation, smooth muscle contraction and remodeling. No other existing or potential drug provides the combined potential advantages of lowering pulmonary arterial pressure, reducing bronchoconstriction, improving blood circulation to the heart and lung, reducing inflammation of the heart and lung tissues, and enhancing wound healing of bronchial epithelial cells. Therefore, development of drug delivery system for VIP-based respiratory therapy may be a promising strategy for the treatment of PAH, asthma and COPD.

Abbreviations

VIP:

vasoactive intestinal peptide

VIP-IR:

VIP-immunoreactivity

PAH:

pulmonary arterial hypertension

COPD:

chronic obstructive pulmonary disease

PACAP:

pituitary adenylate cyclase activating peptide

VPAC1:

VIP/PACAP receptor type1

VPAC2:

VIP/PACAP receptor type 2

NOS:

nitric oxide synthase

CRE:

cAMP responsive element.

References

  1. Said SI, Mutt V: Polypeptide with broad biological activity: isolation from small intestine. Science. 1970, 169: 1217-8. 10.1126/science.169.3951.1217.

    Article  CAS  PubMed  Google Scholar 

  2. Said SI: Vasoactive intestinal peptide. J Endocrinol Invest. 1986, 9: 191-200.

    Article  CAS  PubMed  Google Scholar 

  3. Said SI, Mutt V, Yoshida T, Hara N: Vasoactive polypeptides from normal lung. Chest. 1975, 67: 44S-10.1378/chest.67.2_Supplement.44S.

    Article  CAS  PubMed  Google Scholar 

  4. Ghatei MA, Sheppard MN, O'Shaughnessy DJ, Adrian TE, McGregor GP, Polak JM, Bloom SR: Regulatory peptides in the mammalian respiratory tract. Endocrinology. 1982, 111: 1248-54. 10.1210/endo-111-4-1248.

    Article  CAS  PubMed  Google Scholar 

  5. Baraniuk JN, Lundgren JD, Okayama M, Mullol J, Merida M, Shelhamer JH, Kaliner MA: Vasoactive intestinal peptide in human nasal mucosa. J Clin Invest. 1990, 86: 825-31. 10.1172/JCI114780.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  6. Dvoráková MC: Cardioprotective role of the VIP signaling system. Timely Top Med Cardiovasc Dis. 2005, 9: E33-

    PubMed  Google Scholar 

  7. Saga T, Said SI: Vasoactive intestinal peptide relaxes isolated strips of human bronchus, pulmonary artery, and lung parenchyma. Trans Assoc Am Physicians. 1984, 97: 304-10.

    CAS  PubMed  Google Scholar 

  8. Smalley SG, Barrow PA, Foster N: Immunomodulation of innate immune responses by vasoactive intestinal peptide (VIP): its therapeutic potential in inflammatory disease. Clin Exp Immunol. 2009, 157: 225-34. 10.1111/j.1365-2249.2009.03956.x.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Onoue S, Misaka S, Aoki Y, Karaki S, Kuwahara A, Ohide A, Mizumoto T, Yamada S: Inhalable powder formulation of vasoactive intestinal peptide derivative, [R15,20,21, L17]-VIP-GRR, attenuated neutrophilic airway inflammation in cigarette smoke-exposed rats. Eur J Pharm Sci. 2010, 41: 508-14. 10.1016/j.ejps.2010.08.007.

    Article  CAS  PubMed  Google Scholar 

  10. Szema AM, Hamidi SA, Lyubsky S, Dickman KG, Mathew S, Abdel-Razek T, Chen JJ, Waschek JA, Said SI: Mice lacking the VIP gene show airway hyperresponsiveness and airway inflammation, partially reversible by VIP. Am J Physiol Lung Cell Mol Physiol. 2006, 291: L880-6. 10.1152/ajplung.00499.2005.

    Article  CAS  PubMed  Google Scholar 

  11. Onoue S, Yamada S, Yajima T: Bioactive analogues and drug delivery systems of vasoactive intestinal peptide (VIP) for the treatment of asthma/COPD. Peptides. 2007, 28: 1640-50. 10.1016/j.peptides.2007.04.009.

    Article  CAS  PubMed  Google Scholar 

  12. Yuan JX, Rubin LJ: Pathogenesis of pulmonary arterial hypertension: the need for multiple hits. Circulation. 2005, 111: 534-8. 10.1161/01.CIR.0000156326.48823.55.

    Article  PubMed  Google Scholar 

  13. Gandhi R, Baker N, Shek A, Yeh J, Bishop D: Pulmonary arterial hypertension: bridging the gap between efficacy, quality of life, and cost-effectiveness. Formulary. 2010, 45: 190-199.

    CAS  Google Scholar 

  14. Sahara M, Takahashi T, Imai Y, Nakajima T, Yao A, Morita T, Hirata Y, Nagai R: New insights in the treatment strategy for pulmonary arterial hypertension. Cardiovasc Drugs Ther. 2006, 20: 377-86. 10.1007/s10557-006-0498-3.

    Article  PubMed  Google Scholar 

  15. Mannino DM, Buist AS: Global burden of COPD: risk factors, prevalence, and future trends. Lancet. 2007, 370: 765-73. 10.1016/S0140-6736(07)61380-4.

    Article  PubMed  Google Scholar 

  16. Berger WE, Cline DC: Asthma and COPD: definitions, epidemiology, and treatment guidelines. Postgrad Med. 2005, 118: 2-8. 10.3810/pgm.12.2005.suppl44.232.

    Article  PubMed  Google Scholar 

  17. Lundberg JM: Pharmacology of cotransmission in the autonomic nervous system: integrative aspects on amines, neuropeptides, adenosine triphosphate, amino acids and nitric oxide. Pharmacol Rev. 1996, 48: 113-78.

    CAS  PubMed  Google Scholar 

  18. Henning RJ, Sawmiller DR: Vasoactive intestinal peptide: cardiovascular effects. Cardiovasc Res. 2001, 49: 27-37. 10.1016/S0008-6363(00)00229-7.

    Article  CAS  PubMed  Google Scholar 

  19. Della NG, Papka RE, Furness JB, Costa M: Vasoactive intestinal peptide-like immunoreactivity in nerves associated with the cardiovascular system of guinea-pigs. Neuroscience. 1983, 9: 605-19. 10.1016/0306-4522(83)90177-X.

    Article  CAS  PubMed  Google Scholar 

  20. Brum JM, Bove AA, Sufan Q, Reilly W, Go VL: Action and localization of vasoactive intestinal peptide in the coronary circulation: evidence for nonadrenergic, noncholinergic coronary regulation. J Am Coll Cardiol. 1986, 7: 406-13. 10.1016/S0735-1097(86)80513-7.

    Article  CAS  PubMed  Google Scholar 

  21. Slavíková J: Distribution of peptide-containing neurons in the developing rat right atrium, studied using immunofluorescence and confocal laser scanning. Neurochem Res. 1997, 22: 1013-21.

    Article  PubMed  Google Scholar 

  22. Kuncová J, Slavíková J, Reischig J: Distribution of vasoactive intestinal polypeptide in the rat heart: effect of guanethidine and capsaicin. Ann Anat. 2003, 185: 153-61.

    Article  PubMed  Google Scholar 

  23. Fahrenkrug J: VIP and autonomic neurotransmission. Pharmacol Ther. 1989, 41: 515-34. 10.1016/0163-7258(89)90129-0.

    Article  CAS  PubMed  Google Scholar 

  24. Groneberg DA, Rabe KF, Fischer A: Novel concepts of neuropeptide-based drug therapy: vasoactive intestinal polypeptide and its receptors. Eur J Pharmacol. 2006, 533: 182-94. 10.1016/j.ejphar.2005.12.055.

    Article  CAS  PubMed  Google Scholar 

  25. Dey RD, Shannon WA, Said SI: Localization of VIP-immunoreactive nerves in airways and pulmonary vessels of dogs, cat, and human subjects. Cell Tissue Res. 1981, 220: 231-8.

    CAS  PubMed  Google Scholar 

  26. Lundberg JM, Fahrenkrug J, Hökfelt T, Martling CR, Larsson O, Tatemoto K, Anggård A: Co-existence of peptide HI (PHI) and VIP in nerves regulating blood flow and bronchial smooth muscle tone in various mammals including man. Peptides. 1984, 5: 593-606. 10.1016/0196-9781(84)90090-1.

    Article  CAS  PubMed  Google Scholar 

  27. Laitinen A, Partanen M, Hervonen A, Pelto-Huikko M, Laitinen LA: VIP like immunoreactive nerves in human respiratory tract. Light and electron microscopic study. Histochemistry. 1985, 82: 313-9. 10.1007/BF00494059.

    Article  CAS  PubMed  Google Scholar 

  28. Fischer A, Hoffmann B: Nitric oxide synthase in neurons and nerve fibers of lower airways and in vagal sensory ganglia of man. Correlation with neuropeptides. Am J Respir Crit Care Med. 1996, 154: 209-16.

    Article  CAS  PubMed  Google Scholar 

  29. Fischer A, Canning BJ, Undem BJ, Kummer W: Evidence for an esophageal origin of VIP-IR and NO synthase-IR nerves innervating the guinea pig trachealis: a retrograde neuronal tracing and immunohistochemical analysis. J Comp Neurol. 1998, 394: 326-34. 10.1002/(SICI)1096-9861(19980511)394:3<326::AID-CNE4>3.0.CO;2-3.

    Article  CAS  PubMed  Google Scholar 

  30. Fischer A, Canning BJ, Kummer W: Correlation of vasoactive intestinal peptide and nitric oxide synthase with choline acetyltransferase in the airway innervation. Ann N Y Acad Sci. 1996, 805: 717-22. 10.1111/j.1749-6632.1996.tb17547.x.

    Article  CAS  PubMed  Google Scholar 

  31. Luts A, Sundler F: Peptide-containing nerve fibers in the respiratory tract of the ferret. Cell Tissue Res. 1989, 258: 259-67. 10.1007/BF00239446.

    Article  CAS  PubMed  Google Scholar 

  32. Cutz E, Chan W, Track NS, Goth A, Said SI: Release of vasoactive intestinal polypeptide in mast cells by histamine liberators. Nature. 1978, 275: 661-2. 10.1038/275661a0.

    Article  CAS  PubMed  Google Scholar 

  33. Aliakbari J, Sreedharan SP, Turck CW, Goetzl EJ: Selective localization of vasoactive intestinal peptide and substance P in human eosinophils. Biochem Biophys Res Commun. 1987, 148: 1440-5. 10.1016/S0006-291X(87)80293-0.

    Article  CAS  PubMed  Google Scholar 

  34. Metwali A, Blum AM, Ferraris L, Klein JS, Fiocchi C, Weinstock JV: Eosinophils within the healthy or inflamed human intestine produce substance P and vasoactive intestinal peptide. J Neuroimmunol. 1994, 52: 69-78. 10.1016/0165-5728(94)90164-3.

    Article  CAS  PubMed  Google Scholar 

  35. O'Dorisio MS, O'Dorisio TM, Cataland S, Balcerzak SP: Vasoactive intestinal polypeptide as a biochemical marker for polymorphonuclear leukocytes. J Lab Clin Med. 1980, 96: 666-72.

    PubMed  Google Scholar 

  36. Petkov V, Mosgoeller W, Ziesche R, Raderer M, Stiebellehner L, Vonbank K, Funk GC, Hamilton G, Novotny C, Burian B, Block LH: Vasoactive intestinal peptide as a new drug for treatment of primary pulmonary hypertension. J Clin Invest. 2003, 111: 1339-46.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  37. Haberl I, Frei K, Ramsebner R, Doberer D, Petkov V, Albinni S, Lang I, Lucas T, Mosgoeller W: Vasoactive intestinal peptide gene alterations in patients with idiopathic pulmonary arterial hypertension. Eur J Hum Genet. 2007, 15: 18-22. 10.1038/sj.ejhg.5201711.

    Article  CAS  PubMed  Google Scholar 

  38. Opstad PK: The plasma vasoactive intestinal peptide (VIP) response to exercise is increased after prolonged strain, sleep and energy deficiency and extinguished by glucose infusion. Peptides. 1987, 8: 175-8. 10.1016/0196-9781(87)90183-5.

    Article  CAS  PubMed  Google Scholar 

  39. Burhol PG, Lygren I, Waldum HL: Radioimmunoassay of vasoactive intestinal polypeptide in plasma. Scand J Gastroenterol. 1978, 13: 807-13. 10.3109/00365527809182195.

    Article  CAS  PubMed  Google Scholar 

  40. Bloom SR, Polak JM, Pearse AG: Vasoactive intestinal peptide and watery-diarrhoea syndrome. Lancet. 1973, 2: 14-6. 10.1016/S0140-6736(73)91947-8.

    Article  CAS  PubMed  Google Scholar 

  41. Gyöngyösi M, Németh J, Várkonyi T: Elevated levels of plasma vasoactive intestinal peptide in human acute myocardial infarction. Int J Cardiol. 1996, 56: 159-61.

    Article  PubMed  Google Scholar 

  42. Domschke S, Domschke W, Bloom SR, Mitznegg P, Mitchell SJ, Lux G, Strunz U: Vasoactive intestinal peptide in man: pharmacokinetics, metabolic and circulatory effects. Gut. 1978, 19: 1049-53. 10.1136/gut.19.11.1049.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  43. Lundberg JM, Fahrenkrug J, Brimijoin S: Characteristics of the axonal transport of vasoactive intestinal polypeptide (VIP) in nerves of the cat. Acta Physiol Scand. 1981, 112: 427-36. 10.1111/j.1748-1716.1981.tb06840.x.

    Article  CAS  PubMed  Google Scholar 

  44. Said SI: Vasoactive intestinal polypeptide (VIP): current status. Peptides. 1984, 5: 143-50. 10.1016/0196-9781(84)90197-9.

    Article  CAS  PubMed  Google Scholar 

  45. Goetzl EJ, Sreedharan SP, Turck CW, Bridenbaugh R, Malfroy B: Preferential cleavage of amino- and carboxyl-terminal oligopeptides from vasoactive intestinal polypeptide by human recombinant enkephalinase (neutral endopeptidase, EC 3.4.24.11). Biochem Biophys Res Commun. 1989, 158: 850-4. 10.1016/0006-291X(89)92800-3.

    Article  CAS  PubMed  Google Scholar 

  46. Caughey GH, Leidig F, Viro NF, Nadel JA: Substance P and vasoactive intestinal peptide degradation by mast cell tryptase and chymase. J Pharmacol Exp Ther. 1988, 244: 133-7.

    CAS  PubMed  Google Scholar 

  47. Lilly CM, Kobzik L, Hall AE, Drazen JM: Effects of chronic airway inflammation on the activity and enzymatic inactivation of neuropeptides in guinea pig lungs. J Clin Invest. 1994, 93: 2667-74. 10.1172/JCI117280.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  48. Laburthe M, Couvineau A: Molecular pharmacology and structure of VPAC Receptors for VIP and PACAP. Regul Pept. 2002, 108: 165-173. 10.1016/S0167-0115(02)00099-X.

    Article  CAS  PubMed  Google Scholar 

  49. Francis SH, Noblett BD, Todd BW, Wells JN, Corbin JD: Relaxation of vascular and tracheal smooth muscle by cyclic nucleotide analogs that preferentially activate purified cGMP-dependent protein kinase. Mol Pharmacol. 1988, 34: 506-17.

    CAS  PubMed  Google Scholar 

  50. Arimura A: Perspectives on pituitary adenylate cyclase activating polypeptide (PACAP) in the neuroendocrine, endocrine, and nervous systems. Jpn J Physiol. 1998, 48: 301-31. 10.2170/jjphysiol.48.301.

    Article  CAS  PubMed  Google Scholar 

  51. Carstairs JR, Barnes PJ: Visualization of vasoactive intestinal peptide receptors in human and guinea pig lung. J Pharmacol Exp Ther. 1986, 239: 249-255.

    CAS  PubMed  Google Scholar 

  52. Kulka M, Sheen CH, Tancowny BP, Grammer LC, Schleimer RP: Neuropeptides activate human mast cell degranulation and chemokine production. Immunology. 2008, 123: 398-410. 10.1111/j.1365-2567.2007.02705.x.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  53. Harfi I, D'Hondt S, Corazza F, Sariban E: Regulation of human polymorphonuclear leukocytes functions by the neuropeptide pituitary adenylate cyclase-activating polypeptide after activation of MAPKs. J Immunol. 2004, 173: 4154-63.

    Article  CAS  PubMed  Google Scholar 

  54. Lara-Marquez ML, O'Dorisio MS, Karacay B: Vasoactive intestinal peptide (VIP) receptor type 2 (VPAC2) is the predominant receptor expressed in human thymocytes. Ann NY Acad Sci. 2000, 921: 45-54. 10.1111/j.1749-6632.2000.tb06950.x.

    Article  CAS  PubMed  Google Scholar 

  55. Delgado M, Munoz-Elias EJ, Gomariz RP, Ganea D: Vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide prevent inducible nitric oxide synthase transcription in macrophages by inhibiting NF-kappa B and IFN regulatory factor 1 activation. J Immunol. 1999, 162: 4685-96.

    CAS  PubMed  Google Scholar 

  56. Delneste Y, Herbault N, Galea B, Magistrelli G, Bazin I, Bonnefoy JY, Jeannin P: Vasoactive intestinal peptide synergizes with TNF-alpha in inducing human dendritic cell maturation. J Immunol. 1999, 163: 3071-5.

    CAS  PubMed  Google Scholar 

  57. Goetzl EJ, Voice JK, Shen S, Dorsam G, Kong Y, West KM, Morrison CF, Harmar AJ: Enhanced delayed-type hypersensitivity and diminished immediate-type hypersensitivity in mice lacking the inducible VPAC2 receptor for vasoactive intestinal peptide. Proc Natl Acad Sci USA. 2001, 98: 13854-13859. 10.1073/pnas.241503798.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  58. Voice JK, Grinninger C, Kong Y, Bangale Y, Paul S, Goetzl EJ: Roles of vasoactive intestinal peptide (VIP) in the expression of different immune phenotypes by wild-type mice and T cell-targeted type II VIP receptor transgenic mice. J Immunol. 2003, 170: 308-314.

    Article  CAS  PubMed  Google Scholar 

  59. Morice A, Unwin RJ, Sever PS: Vasoactive intestinal peptide causes bronchodilatation and protects against histamine-induced bronchoconstriction in asthmatic subjects. Lancet. 1983, 2: 1225-7. 10.1016/S0140-6736(83)91272-2.

    Article  CAS  PubMed  Google Scholar 

  60. Colston JT, Freeman GL: Beneficial influence of vasoactive intestinal peptide on ventriculovascular coupling in closed-chest dogs. Am J Physiol. 1992, 263: H1300-5.

    CAS  PubMed  Google Scholar 

  61. Rigel DF, Grupp IL, Balasubramaniam A, Grupp G: Contractile effects of cardiac neuropeptides in isolated canine atrial and ventricular muscles. Am J Physiol. 1989, 257: H1082-7.

    CAS  PubMed  Google Scholar 

  62. Rigel DF, Lathrop DA: Vasoactive intestinal polypeptide facilitates atrioventricular nodal conduction and shortens atrial and ventricular refractory periods in conscious and anesthetized dogs. Circ Res. 1990, 67: 1323-33.

    Article  CAS  PubMed  Google Scholar 

  63. Henning RJ: Vagal stimulation during muscarinic and beta-adrenergic blockade increases atrial contractility and heart rate. J Auton Nerv Syst. 1992, 40: 121-9. 10.1016/0165-1838(92)90023-A.

    Article  CAS  PubMed  Google Scholar 

  64. Henning RJ, Feliciano L, Coers CM: Vagal nerve stimulation increases right ventricular contraction and relaxation and heart rate. Cardiovasc Res. 1996, 32: 846-53.

    Article  CAS  PubMed  Google Scholar 

  65. Lucia P, Caiola S, Coppola A, Maroccia E, Belli M, Buongiorno A, De Martinis C: Early increase of vasoactive intestinal peptide in acute myocardial infarction. Am Heart J. 1996, 132: 187-9. 10.1016/S0002-8703(96)90408-1.

    Article  CAS  PubMed  Google Scholar 

  66. Groneberg DA, Springer J, Fischer A: Vasoactive intestinal polypeptide as mediator of asthma. Pulm Pharmacol Ther. 2001, 14: 391-401. 10.1006/pupt.2001.0306.

    Article  CAS  PubMed  Google Scholar 

  67. Hamasaki Y, Saga T, Mojarad M, Said SI: Vasoactive intestinal peptide counteracts leukotriene D4-induced contractions of guinea pig trachea, lung, and pulmonary artery. Trans Assoc Am Physicians. 1983, 96: 406-11.

    CAS  PubMed  Google Scholar 

  68. Palmer JB, Cuss FM, Barnes PJ: VIP and PHM and their role in nonadrenergic inhibitory responses in isolated human airways. J Appl Physiol. 1986, 61: 1322-8.

    CAS  PubMed  Google Scholar 

  69. Groneberg DA, Hartmann P, Dinh QT, Fischer A: Expression and distribution of vasoactive intestinal polypeptide receptor VPAC(2) mRNA in human airways. Lab Invest. 2001, 81: 749-55. 10.1038/labinvest.3780283.

    Article  CAS  PubMed  Google Scholar 

  70. Coles SJ, Said SI, Reid LM: Inhibition by vasoactive intestinal peptide of glycoconjugate and lysozyme secretion by human airways in vitro. Am Rev Respir Dis. 1981, 124: 531-6.

    CAS  PubMed  Google Scholar 

  71. Baraniuk JN, Lundgren JD, Okayama M, Mullol J, Merida M, Shelhamer JH, Kaliner MA: Vasoactive intestinal peptide in human nasal mucosa. J Clin Invest. 1990, 86: 825-31. 10.1172/JCI114780.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  72. Webber SE, Widdicombe JG: The effect of vasoactive intestinal peptide on smooth muscle tone and mucus secretion from the ferret trachea. Br J Pharmacol. 1987, 91: 139-48.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  73. Shimura S, Sasaki T, Ikeda K, Sasaki H, Takishima T: VIP augments cholinergic-induced glycoconjugate secretion in tracheal submucosal glands. J Appl Physiol. 1988, 65: 2537-44.

    CAS  PubMed  Google Scholar 

  74. Delgado M, Abad C, Martinez C, Juarranz MG, Arranz A, Gomariz RP, Leceta J: Vasoactive intestinal peptide in the immune system: potential therapeutic role in inflammatory and autoimmune diseases. J Mol Med. 2002, 80: 16-24. 10.1007/s00109-001-0291-5.

    Article  CAS  PubMed  Google Scholar 

  75. Undem BJ, Dick EC, Buckner CK: Inhibition by vasoactive intestinal peptide of antigen-induced histamine release from guinea-pig minced lung. Eur J Pharmacol. 1983, 88: 247-50. 10.1016/0014-2999(83)90012-2.

    Article  CAS  PubMed  Google Scholar 

  76. O'Dorisio MS, Shannon BT, Fleshman DJ, Campolito LB: Identification of high affinity receptors for vasoactive intestinal peptide on human lymphocytes of B cell lineage. J Immunol. 1989, 142: 3533-6.

    PubMed  Google Scholar 

  77. Berisha H, Foda H, Sakakibara H, Trotz M, Pakbaz H, Said SI: Vasoactive intestinal peptide prevents lung injury due to xanthine/xanthine oxidase. Am J Physiol. 1990, 259: L151-5.

    CAS  PubMed  Google Scholar 

  78. Misra BR, Misra HP: Vasoactive intestinal peptide, a singlet oxygen quencher. J Biol Chem. 1990, 265: 15371-4.

    CAS  PubMed  Google Scholar 

  79. Leceta J, Gomariz RP, Martinez C, Abad C, Ganea D, Delgado M: Receptors and transcriptional factors involved in the anti-inflammatory activity of VIP and PACAP. Ann N Y Acad Sci. 2000, 921: 92-102. 10.1111/j.1749-6632.2000.tb06954.x.

    Article  CAS  PubMed  Google Scholar 

  80. Dunzendorfer S, Meierhofer C, Wiedermann CJ: Signalling in neuropeptide-induced migration of human eosinophils. J Leukoc Biol. 1998, 64: 828-34.

    CAS  PubMed  Google Scholar 

  81. Dunzendorfer S, Feistritzer C, Enrich B, Wiedermann CJ: Neuropeptide-induced inhibition of IL-16 release from eosinophils. Neuroimmunomodulation. 2002, 10: 217-23. 10.1159/000068324. 2003

    Article  CAS  PubMed  Google Scholar 

  82. Delgado M: VIP: a very important peptide in T helper differentiation. Trends Immunol. 2003, 24 (5): 221-4. 10.1016/S1471-4906(03)00069-3.

    Article  CAS  PubMed  Google Scholar 

  83. Gunaydin S, Imai Y, Takanashi Y, Seo K, Hagino I, Chang D, Shinoka T: The effects of vasoactive intestinal peptide on monocrotaline induced pulmonary hypertensive rabbits following cardiopulmonary bypass: a comparative study with isoproteronol and nitroglycerine. Cardiovasc Surg. 2002, 10: 138-45. 10.1016/S0967-2109(01)00126-0.

    Article  PubMed  Google Scholar 

  84. Said SI: The vasoactive intestinal peptide gene is a key modulator of pulmonary vascular remodeling and inflammation. Ann N Y Acad Sci. 2008, 1144: 148-53. 10.1196/annals.1418.014.

    Article  CAS  PubMed  Google Scholar 

  85. Said SI, Hamidi SA, Dickman KG, Szema AM, Lyubsky S, Lin RZ, Jiang YP, Chen JJ, Waschek JA, Kort S: Moderate pulmonary arterial hypertension in male mice lacking the vasoactive intestinal peptide gene. Circulation. 2007, 115: 1260-8.

    CAS  PubMed  Google Scholar 

  86. Popma JJ, Smitherman TC, Bedotto JB, Eichhorn EJ, Said SI, Dehmer GJ: Direct coronary vasodilation induced by intracoronary vasoactive intestinal peptide. J Cardiovasc Pharmacol. 1990, 16: 1000-6. 10.1097/00005344-199012000-00021.

    Article  CAS  PubMed  Google Scholar 

  87. Frase LL, Gaffney FA, Lane LD, Buckey JC, Said SI, Blomqvist CG, Krejs GJ: Cardiovascular effects of vasoactive intestinal peptide in healthy subjects. Am J Cardiol. 1987, 60: 1356-61. 10.1016/0002-9149(87)90619-9.

    Article  CAS  PubMed  Google Scholar 

  88. Soriano JB, Davis KJ, Coleman B, Visick G, Mannino D, Pride NB: The proportional Venn diagram of obstructive lung disease: two approximations from the United States and the United Kingdom. Chest. 2003, 124: 474-81. 10.1378/chest.124.2.474.

    Article  PubMed  Google Scholar 

  89. Zakynthinos E, Daniil Z, Papanikolaou G, Makris D: Pulmonary Hypertension in COPD: Pathophysiology and Therapeutic Targets. Curr Drug Targets. 2011, 12: 501-13. 10.2174/138945011794751483.

    Article  CAS  PubMed  Google Scholar 

  90. Said SI, Hamidi SA, Gonzalez Bosc L: Asthma and pulmonary arterial hypertension: do they share a key mechanism of pathogenesis?. Eur Respir J. 2010, 35: 730-4. 10.1183/09031936.00097109.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  91. Dorfmüller P, Perros F, Balabanian K, Humbert M: Inflammation in pulmonary arterial hypertension. Eur Respir J. 2003, 22: 358-63.

    Article  PubMed  Google Scholar 

  92. Stenmark KR, Mecham RP: Cellular and molecular mechanisms of pulmonary vascular remodeling. Annu Rev Physiol. 1997, 59: 89-144. 10.1146/annurev.physiol.59.1.89.

    Article  CAS  PubMed  Google Scholar 

  93. Homer RJ, Elias JA: Airway remodeling in asthma: therapeutic implications of mechanisms. Physiology (Bethesda). 2005, 20: 28-35.

    Article  CAS  Google Scholar 

  94. Sergejeva S, Hoshino H, Yoshihara S, Kashimoto K, Lötvall J, Lindén A: A synthetic VIP peptide analogue inhibits neutrophil recruitment in rat airways in vivo. Regul Pept. 2004, 117: 149-54. 10.1016/j.regpep.2003.10.002.

    Article  CAS  PubMed  Google Scholar 

  95. Onoue S, Ohmori Y, Endo K, Yamada S, Kimura R, Yajima T: Vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide attenuate the cigarette smoke extract-induced apoptotic death of rat alveolar L2 cells. Eur JBiochem. 2004, 271: 1757-67. 10.1111/j.1432-1033.2004.04086.x.

    Article  CAS  Google Scholar 

  96. Guan CX, Zhang M, Qin XQ, Cui YR, Luo ZQ, Bai HB, Fang X: Vasoactive intestinal peptide enhances wound healing and proliferation of human bronchial epithelial cells. Peptides. 2006, 27: 3107-14. 10.1016/j.peptides.2006.08.004.

    Article  CAS  PubMed  Google Scholar 

  97. Misaka S, Aoki Y, Karaki S, Kuwahara A, Mizumoto T, Onoue S, Yamada S: Inhalable powder formulation of a stabilized vasoactive intestinal peptide (VIP) derivative: anti-inflammatory effect in experimental asthmatic rats. Peptides. 2010, 31: 72-8. 10.1016/j.peptides.2009.09.032.

    Article  CAS  PubMed  Google Scholar 

  98. Misaka S, Sato H, Aoki Y, Mizumoto T, Onoue S, Yamada S: Novel vasoactive intestinal peptide derivatives with improved stability protect rat alveolar L2 cells from cigarette smoke-induced cytotoxicity and apoptosis. Peptides. 2011, 32: 401-7. 10.1016/j.peptides.2010.10.021.

    Article  CAS  PubMed  Google Scholar 

  99. Rubinstein I: Human VIP-alpha: an emerging biologic response modifier to treat primary pulmonary hypertension. Expert Rev Cardiovasc Ther. 2005, 3: 565-9. 10.1586/14779072.3.4.565.

    Article  CAS  PubMed  Google Scholar 

  100. Bolin DR, Cottrell J, Garippa R, Rinaldi N, Senda R, Simko B, O'Donnell M: Comparison of cyclic and linear analogs of vasoactive intestinal peptide. Drug Des Discov. 1996, 13: 107-14.

    CAS  PubMed  Google Scholar 

  101. Schmidt DT, Rühlmann E, Waldeck B, Branscheid D, Luts A, Sundler F, Rabe KF: The effect of the vasoactive intestinal polypeptide agonist Ro 25-1553 on induced tone in isolated human airways and pulmonary artery. Naunyn Schmiedebergs Arch Pharmacol. 2001, 364: 314-20. 10.1007/s002100100458.

    Article  CAS  PubMed  Google Scholar 

  102. Lindén A, Hansson L, Andersson A, Palmqvist M, Arvidsson P, Löfdahl CG, Larsson P, Lötvall J: Bronchodilation by an inhaled VPAC(2) receptor agonist in patients with stable asthma. Thorax. 2003, 58: 217-21.

    Article  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported in part by the World Class University program (R31-20029) funded by the Ministry of Education, Science and Technology", Republic of Korea.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dongmei Wu.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

All authors participated in drafting the manuscript. All authors read and approved the manuscript.

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://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

Wu, D., Lee, D. & Sung, Y.K. Prospect of vasoactive intestinal peptide therapy for COPD/PAH and asthma: a review. Respir Res 12, 45 (2011). https://doi.org/10.1186/1465-9921-12-45

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1465-9921-12-45

Keywords