Loading

Clinical Research in Pulmonology

Adenosine A2A Receptor as a Target of Treatment for Pulmonary Arterial Hypertension

Review Article | Open Access

  • 1. Division of Biomedical Sciences, Drug Development Program, Federal University of Rio de Janeiro, Brazil
+ Show More - Show Less
Corresponding Authors
Gisele Zapata-Sudo, Division of Biomedical Sciences, Drug Development Program, Universidade Federal do Rio de Janeiro, Centro de Ciencias da Saude, Instituto de Ciencias Biomedicas, Bloco J, Sala 14, Rio de Janeiro, RJ, Brazil, 21941-590 PH/FAX 55-21-39386505
Abstract

Pulmonary arterial hypertension (PAH) is a rare condition characterized by small pulmonary artery remodeling that leads to chronic precapillary pulmonary hypertension, elevated pulmonary vascular resistance, and right heart failure. Therapeutic options for PAH remain limited despite the introduction of prostacyclin analogues, endothelin receptor antagonists, and phosphodiesterase 5 inhibitors within the last 15 years. Predominantly, these interventions address the endothelial and vascular dysfunctions associated with PAH, and simply delay progression of the disease rather than offer a cure. To improve treatment efficacy, emerging approaches are focusing on the pro-proliferative phenotype as the target. This phenotype underpins pulmonary vascular remodeling in the lung and contributes to impaired circulation and right heart failure. Several new targets have been investigated and validated in experimental PAH models. Further, the adenosinergic system, specifically the adenosine A2A receptor, is potentially a novel and efficient approach for PAH treatment. Herein, we provide a review of the effects of adenosine on the cardiopulmonary system, focusing on the contribution of the A2A receptor as a pharmacological target that induces pulmonary vasodilatation and cardioprotection in experimental PAH models.

Citation

Alencar AKN, Barreiro EJ, Sudo RT, Zapata-Sudo G (2014) Adenosine A2A Receptor as a Target of Treatment for Pulmonary Arterial Hypertension. Clin Res Pulmonol 2(2): 1021.

Keywords

•    Pulmonary arterial hypertension
•    Ventricular dysfunction
•    Pulmonary vascular remodeling
•    A2A adenosine agonist
•    Adenosinergic system 

ABBREVIATIONS

PAH: Pulmonary Arterial Hypertension; RV: Right Ventricle; EC: Endothelial Cell; SMC: Smooth Muscle Cell; MCT: Monocrotaline; HF: Heart Failure; A2AR: Adenosine A2A Receptor; ATP: Adenosine Triphosphate; cAMP: Cyclic Adenosine Monophosphate; SERCA2: Sarco/Endoplasmic Reticulum Ca2+- Atpase; MCT: Monocrotaline

INTRODUCTION

Pulmonary arterial hypertension (PAH) is characterized by deregulated pulmonary vascular remodeling that leads subsequently to increased pulmonary vascular resistance, right ventricular (RV) hypertrophy, RV dysfunction, uncompensated right heart failure, and premature death [1]. Therefore, PAH is associated with a poor prognosis. In its variety of presentations, this disease is estimated to affect up to 100 million people worldwide [2]. It is defined as an increase in mean pulmonary arterial pressure ≥ 25 mmHg at rest measured on right heart catheterization [3]. There are different hemodynamic definitions of PAH according to the pulmonary capillary wedge pressure, pulmonary vascular resistance, and cardiac output [4]. However, the current classification of PAH was agreed upon at the 4th World Symposium on Pulmonary Hypertension in 2008, in which PAH is separated into five groups (Table 1)[5,6].

Group 1 includes a variety of diseases that share several pathophysiological, histological, and prognostic characteristics. Idiopathic PAH defines group 1.1, in which patients of lack family history or a clearly identified risk factor for PAH. In hereditary PAH (group 1.2), loss-of-function mutations of the transforming growth factor β/bone morphogenetic protein (TGF-β/BMP) receptor superfamily, and more rarely, in activin receptor-like kinase type 1 have been identified as underlying mechanisms [6]. Drug- and toxin-induced forms of PAH are included in groups 1.3, and 1.4, as well as PAH associated with identified diseases (e.g., HIV). Persistent PAH of the newborn is included in group 1.5[6], and patients with pulmonary veno-oclusive disease and/or pulmonary capillary hemangiomatosis are classified in this group as well [1,5,6]. Group 2 includes patients with PAH secondary to left heart disease. Group 3 consists of all forms of PAH associated with disorders of the respiratory system, group 4, with all PAH caused by thromboembolic diseases, and group 5, with PAH caused by unclear multifactorial mechanisms [6].

The pathological characteristics of PAH include pulmonary arterial endothelial cell (EC) dysfunction, pulmonary artery EC and smooth muscle cell (SMC) proliferation, vasoconstriction, and in situ thrombosis. Furthermore, because sub-groups of PAH have common clinical characteristics, they are managed similarly [7]. However, despite the development of many new therapies over the last two decades, PAH remains an incurable disease process. If it is not interrupted, it will eventually become life threatening [8].

Many factors have been identified or proposed as contributing to vasoconstriction and vascular remodeling. These fall into several key, but somewhat interrelated and overlapping categories as follows: vasoactive factors, calcium signaling molecules, inflammatory mediators, growth factors, bone morphogenetic protein receptor 2 mutations, and metabolic dysfunction [6]. Over the past two decades, three major pathways (prostacyclin, endothelin, and nitric oxide [NO] pathways) have been established as being key elements in the development and progression of PAH [9-11]. These pathways have been targeted by PAH-specific therapies that fall into tree drug classes: prostacyclin (PGI2 ) analogues, endothelin receptor antagonists, and phosphodiesterase-5 inhibitors (iPDE5) [12]. Additionally, some of these drugs were shown to improve symptoms, survival, functional class, and time to clinical worsening. Further, the use of these agents in clinical practice has been associated with outcome improvements among patients with PAH compared with historical data; however, there is still room for improvement [11,13-15].

Pulmonary arterial vessel relaxation is one of the main goals of PAH treatment. Thus, new therapeutic approaches, which induce NO release and relaxation through different pathways, such as iPDE5 and PGI2 analogues, are emerging nowadays to improve the life quality of patients with PAH [12]. Moreover, stronger evidence suggesting that the adenosine A2A receptor (A2AR) could be a new target to treat PAH has been reported recently [16-18]. Therefore, in this review we focus on molecular mechanisms of relaxation induced by the activation of A2AR, and we comment on the newest findings of the literature concerning the aforementioned affirmation.

Table 1: Current classification of pulmonary arterial hypertension[5,6].

Group 1 - Pulmonary arterial hypertension
• Group 1.1 - Idiopathic pulmonary arterial hypertension
• Group 1.2 - Heritable pulmonary arterial hypertension
• Group 1.3 - Drug-induced and toxin-induced forms of pulmonary arterial hypertension
• Group 1.4 - Pulmonary arterial hypertension associated with identified diseases (for example, HIV and schistosomiasis infections)
Group 1.5 - Persistent pulmonary hypertension of the newborn
Group 1' - Pulmonary veno-occlusive disease and/or pulmonary capillary hemangiomatosis
Group 2 - Pulmonary hypertension resulting from left heart disease; this group represents the large group of patients with pulmonary venous hypertension
Group 3 - All forms of pulmonary hypertension associated with alveolar hypoxia and/or disorders of the respiratory system
Group 4 - All (thrombo)embolic diseases leading to pulmonary hypertension
Group 5 - Pulmonary hypertension with unclear multifactorial mechanisms (a group of diseases directly affecting the pulmonary vessels)

 

ADENOSINE AND ITS ENDOGENOUS ACTIONS

Adenosine is a purine nucleoside comprised of adenine and ribose joined by a glycosidic bond. It is a both a precursor and a metabolite of adenine nucleotides. Because all cells can use free energy derived from the catabolism of adenosine triphosphate (ATP) to perform various functions (e.g., synthesis, secretion, contraction, and ion transport), all cells are possible sources of adenosine. Many of the cells that “produce” adenosine also express adenosine receptors on their surface [19]. Activation of these receptors often leads to an overall reduction of work performed and oxygen consumed by cells and organs. For example, adenosine reduces the pacemaking rate of the cardiac sinoatrial (SA) cells, thereby reducing the heart rate and cardiac load [20].

Adenosine is mainly present in the cytoplasm in its phosphorylated forms, adenosine monophosphate (AMP), adenosine diphosphate (ADP), and ATP. All these forms are generated through AMP hydrolysis by ecto-5-nucleotidase enzyme, an integral part of energy regulation at the cellular level. Under physiological conditions adenosine is maintained at a low intracellular concentration (estimates range between ten and a few hundred nanomolar) by both S-adenosylhomocysteine and S-adenosylhomocysteine hydrolases [21]. In response to cellular stress and damage, ATP is released into the extracellular space and is rapidly dephosphorylated by extracellular nucleotidases [22], with the subsequent substantial increase of adenosine levels. Extracellular adenosine can then interact with G protein-coupled receptors, which are coupled to various second messenger systems (Figure 1). Alternatively, extracellular adenosine can be transported into cells by passive diffusion through a specific bi-directional transport system [23]. Both extra and intracellular adenosine particles can be deamined to inosine by the actions of the adenosine deaminase. Intracellular adenosine can be secreted to the extracellular fluid or phosphorylated back to ATP, reaction that is catalyzed by adenosine kinase [23-25].

Adenosine is present in several body tissues and is produced by different cell types. It is an important modulator of the central and peripheral nervous systems, and it is involved in the homeostasis of the cardiovascular, renal, respiratory, and immune systems [26]. Additionally, it plays an important role in energy transfer (during the transition from ATP to ADP) and signal transduction, through cyclic AMP (cAMP), which regulates many physiological and pathological processes. Thus, its rapid release in response to abnormal cellular conditions has two roles: initially, extracellular adenosine represents a “danger” signal and it is released rapidly upon tissue injury; as a consequence, increased extracellular levels of this nucleoside lead to a range of tissue responses for homeostasis restoration that can be considered organ-specific [26]. Evidence indicates that the adenosinergic system is essential in the mediation of intrinsic protection and in determining myocardial resistance to insult; therefore, adenosine may be considered as a potential cardioprotective molecule and its receptors could represent potential therapeutic targets in the setting of heart failure [25].

Adenosine receptors and their differential pattern of expression modulate a series of pleiotropic activities that are known to contribute to the control of inflammation, remodeling, and tissue repair [24]. Consequently, pharmacological manipulation of the adenosine signaling pathway is of great interest. This signaling pathway is currently under study as a therapeutic target for a number of respiratory diseases in conjunction with several molecules, with both agonist and antagonist activities, against known adenosine receptors involved in different conditions of the respiratory system, including PAH [24].

ADENOSINE RECEPTORS AND THEIR EFFECT ON THE CARDIOVASCULAR SYSTEM

For many years, adenosine has been known for its potent vasodilation effect, particularly in the coronary circulation [27]. Adenosine and its analogues have been shown to produce vasodilatation in canine basilar arteries [28], porcine coronary arteries [29], rat aorta [30], and dog carotid arteries [31]. Adenosine is released by several types of cells and activates four different membrane-spanning G protein-coupled receptors: A1 R, A2AR, A2BR, and A3 R. It can be released from the parenchymal tissue (including the endothelium) and after it interacts with specific extracellular receptors located on SMCs and ECs of the blood vessels to produce relaxation [5] (Figure 1). The release of adenosine increases under physiological stressful conditions, when it activates a homeostatic response (e.g., relaxation) to promote balance between the normal cells and tissue functions [32].

Both A1 R and A3 R are coupled negatively to adenylate cyclase through the Gi/o protein subunits; their activation causes a decrease in intracellular cAMP, and A1 R is linked to various kinase pathways such as protein kinase C, phosphoinositide 3 (PI3) kinase, and mitogen-activated protein (MAP) kinases. Furthermore, A1 R can directly activate K+ channels and inhibit Q-, P-, and N-type Ca2+ channels [33]. The A3 R is coupled both to Gi and Gq proteins, inhibits adenylyl cyclase, and stimulates phospholipase C (PLC). This receptor can also utilize phospholipase D, RhoA, Wnt, MAP kinase, and PI3 kinase pathways to control cell function [33]. Both A2AR and A2BR are coupled positively to adenylyl cyclase through the Gs protein, and their activation causes an increase in intracellular cAMP [34]. Stimulation of A2BR can trigger adenylyl cyclase activation via Gs and PLC activation via the Gq protein [33] (Figure 1).

The most extensively studied and accepted function mediated by the postsynaptic adenosine receptors is vasodilatation. In fact, the involvement of A2A and A2B receptor-mediated vasodilation in several vessels has been reported, namely in muscular arteries (mesenteric [35], renal [36], and coronary arteries [37]) and in elastic arteries, including the aorta of several species: guinea pigs [38], rats[39], and hamsters[40]. Adenosine relaxes precontracted isolated pulmonary arterial rings and its effects probably occur via A2A and A2B receptor activation [41]. In contrast, A1 R and A3 R, modulate negatively the A2A-B R-induced vasodilation [43-45]. A1 R appears to be involved in lowering the heart rate. Additionally, it plays a negative role in regulating blood pressure, causes contraction of vascular smooth muscle, and decreases coronary blood flow [45]. Thus, adenosine can act as a vasoconstrictor besides being a vasodilator. However, this depends on the interaction with specific subtypes and tissue localization. The number of actions after adenosine receptor activation, the large base of current knowledge, and the rapid pace of adenosine research suggest that additional clinical applications of adenosine receptor research are quite promising [19].

SIGNALING PATHWAYS OF THE A2A RECEPTOR-INDUCING RELAXATION AND ITS CARDIOPROTECTIVE EFFECTS

In the coronary arteries, vasodilatation is primarily caused by the activation of the A2AR [46-50]. Additionally, some studies demonstrated that the A2AR had an important protective role in the kidneys, lungs, and heart during ischemia/reperfusion injury. Further, the activation of A2AR promoted beneficial effects against lung ischemia/reperfusion injury when the A2AR agonist CGS21680 was administered before ischemia and during reperfusion [51]. These protective effects during ischemia/ reperfusion injury were closely related to extracellular-signalregulated kinases (ERK) and cAMP. The endogenous A2AR expressed in PC12 cells activated the ERK phosphorylation cascade, probably triggered by a rise in cAMP [52,53].

Organ-specific vascular hyporesponsiveness (thoracic aorta and left femoral, superior mesenteric, right renal, pulmonary, and middle cerebral arteries) following shock is closely related to the differential expression of A2ARs in the corresponding vasculatures under normal and shock conditions [54]. The participation of A2AR in regulating and protecting vascular reactivity following shock and its activation has a beneficial effect on hemorrhagic shock by improving vascular reactivity and hemodynamic parameters [54].

In an A2AR knockout mice model, a decreased adenosinemediated aortic relaxation was shown[45], and this fact can provide additional support to confirm the importance of A2AR on maintenance of the vascular tone. In further support of this concept, our research group synthetized a new compound that exhibited vasodilator and antihypertensive actions that were mediated by activation of the A2AR [55]. Some investigators have suggested that vascular relaxation in response to A2AR activation may be independent of ECs [56]. Others have shown a significant involvement of the ECs in A2AR-mediated relaxation [57]. This controversy, however, may be resolved by the findings of recent studies, which showed that the A2AR is located not only in the vascular endothelium but also in the vascular smooth cells [58], and confirmed that its activation is involved in vasodilation [59,60]. Activation of endothelial A2AR, which is coupled to a Gs protein, induces NO release by activating the adenylate cyclaseprotein kinase A pathway [61,62]. Moreover, activation of A2AR in vascular smooth cells increases activation of cAMP and protein kinase A, which leads to phosphorylation and opening of K+ channels. This effect, in turn, causes hyperpolarization and results in vasodilatation [63](Figure 2).

Along with vascular cells, A2AR is expressed in other myocardial cells types. Additionally, A2AR gene is expressed in mast cells [64], neutrophils [65], and CD4+ T cells [66]. Evidence of A2AR expression on myocytes has also been demonstrated in adult rat ventricular myocytes through the detection of A2AR messenger RNA (mRNA) [67] and by immunoblotting of ventricular cells of mice [68]. Although the numerous in vivo studies on A2AR implicate anti-inflammatory effects, the results of additional studies suggest that this protection could be caused in part because of direct myocardial effects [68-72].

The constitutive overexpression of A2AR in young mice was associated with increased cardiac contractility, higher heart rates, and a small increase in left ventricular mass. Viewed in terms of excitation-contraction coupling, the major alterations induced by A2AR overexpression were increased Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2) expression and Ca2+ uptake by the sarcoplasmic reticulum [73]. Thus, these findings suggest the beneficial effects of A2AR-mediated signaling on cardiac function and cardiac hemodynamics [74]. Moreover, adenosine inhibits collagen synthesis and hypertrophy of vascular smooth cells and cardiac fibroblasts via the cAMP pathway (A2AR and A2BR activation) [75].

As previously mentioned, PAH is characterized by excessive pulmonary vasoconstriction and abnormal vascular remodeling processes that usually affect all vessel layers (intima, media, and adventitia) and result in a severe loss of cross-sectional area, and therefore, increased RV after load [6]. Large pulmonary artery compliance is also decreased, contributing to strain on the RV [6]. Although the pathogenesis of PAH is incompletely understood, evidence suggests that PAH is associated with activation of inflammatory processes, endothelial damage and dysfunction, and abnormal coagulation [76]. Thus, the modification of pulmonary vascular structures causes an increased resistance on the RV load that induces RV dysfunction [77].

In this review, added to the beneficial actions of A2AR activation (vasodilatation and cardioprotective effects) reported to date, we also will describe the significance of A2AR agonist use during the pathogenesis of PAH and the potential of such a drug as a possible treatment of the disease.

A2A RECEPTOR AS A NEW TARGET FOR PAH TREATMENT

The development of PAH involves a complex interplay of multiple genetic, environmental, and hormonal abnormalities that lead to abnormal pulmonary vascular remodeling involving ECs, SMCs, and fibroblasts. Endogenous adenosine levels vary in different tissues, with adenosine levels being higher in the pulmonary than in the systemic circulation [78], for example, SMCs synthetize a substantial amount of adenosine [79], and extracellular adenosine levels increase markedly in response to hypoxia [80]. However, patients with PAH have lower plasma adenosine levels in their pulmonary circulation compared to control subjects, which indicates a possible deficiency in adenosine signaling in PAH [78]. Immunohistochemical analyses of lung parenchyma demonstrated A2AR expression in bronchiolar and alveolar epithelial cells, SMCs localized in bronchiolar and vessels walls, and ECs in pulmonary arteries [81]. Further, the A2BR receptor was expressed mainly in mast cells and macrophages, and the A1 R was expressed only in a few alveolar macrophages [81].

Several lines of experimental evidence suggest that adenosine may be a potential endogenous regulator of PAH development by balancing SMC growth and collagen synthesis and by maintaining vascular homeostasis in the systemic circulation. Adenosine can act at A2AR, induce powerful vasodilatation, and affect systemic blood pressure. It can exert a similar effect on pulmonary arteries [81]. Thus, adenosine action on A2AR regulates the pulmonary vascular remodeling and reduces exacerbated vasoconstriction.

Using an A2AR knockout mice model, Xu et al. provided the first evidence of the critical contribution of A2AR to the development of PAH. At the postnatal age of 14–16 weeks, A2AR KO mice exhibited characteristics of hemodynamic, histological, and ultrastructural changes of PAH: increased RV systolic pressure (RVSP) and increased RV mass, increased wall area and thickness, increased cellular proliferation in pulmonary resistance vessels, activation and hypertrophy of the SMCs and ECs, as well as collagen deposition in the adventitia of pulmonary arterial walls [17].

These investigators explained that spontaneous PAH and altered remodeling of pulmonary arteries at hemodynamic, histological, and ultrastructural levels are supported by the anatomical localization of A2AR in the vasculature primarily (where vasodilatation is mediated), added to the demonstration of the functional activation of A2AR in ECs (where adenosineinduced vasodilatation is mediated)[17]. Therefore, adenosine has been shown to cause vascular smooth muscle relaxation in the pulmonary circulation, and thus, it has been proposed for the therapy of clinical and experimental pulmonary hypertension. These findings suggest that the adenosine effect is likely to be mediated by A2AR in pulmonary vessels [17].

Curiously, our research group also investigated the A2AR involvement on regulation of cardiopulmonary physiology in rats with PAH induced by monocrotaline (MCT). Our results showed that after 14–28 days of MCT injection the animals exhibited a reduction in exercise capacity. This result corresponds well with the time course of the development of PAH [18]. When we treated the MCT-rats with a specific A2AR agonist (LASSBio-1386), the exercise capacity significantly improved compared with the control animals [18].

In a second study, we showed that another A2AR agonist synthetized by our laboratory (LASSBio-1359) was also capable of inducing pulmonary vascular relaxation and promoting the recovery of endothelial dysfunction of the pulmonary artery rings in rats with PAH [16]. MCT injection also induced fibromuscular hypertrophy and hyperplasia in the arteriole walls of rats with PAH. This condition increased the RVSP and led to RV hypertrophy. However, daily oral treatment with the A2AR agonists, which were administered to the rats with PAH in both studies, abolished the increase in RVSP and reduced RV hypertrophy, after the disease was established [16,18].

Another observation of our experimental data was a reduction of A2AR expression both on pulmonary tissue and on RV tissue from rats with PAH [18]. We were able to demonstrate for the first time that A2AR levels were altered on PAH. These data supported previous reports cited in this review that showed a downregulation of this receptor in different experimental models of cardiovascular disease. Further, in our MCT model of PAH, we also observed a decrease in RV SERCA2 protein expression and in Ca2+-ATPase activity at 28 days after MCT injection, in combination with the increased protein expression of total phospholamban that inhibits SERCA2 activity. Thus, it can be postulated that the upregulation of phospholamban inhibits SERCA2 activity and may be associated with the reduced A2AR expression and cardiac dysfunction [18] (Figure 3).

CONCLUDING REMARKS

Advances in our understanding of the pathogenetic role of adenosine in PAH may be translated into effective treatment options soon [24-26]. Considering the complex interplay driven by the different patterns of receptor distribution and/or affinity of the four known adenosine receptor subtypes in specific cell types at different stages of the disease, it is likely that combination of selective antagonist/agonists for different adenosine receptors subtypes will be required to obtain reasonable clinical efficacy. Alternatively, controlling the factors involved in driving adenosine concentrations in tissue may be also of great significance.

Thus, the data discussed herein indicate a role for A2AR in mediating beneficial effects during established PAH in rats as pulmonary vascular relaxation, reduction of pulmonary vessel and RV hypertrophy, amelioration of RV dysfunction, and exercise capacity. We provide a brief review of the importance of the adenosinergic system, specifically the A2AR, as new target for the treatment of PAH. Further, we consider that of new therapeutic strategies and careful attention to patients with PAH is need currently.

ACKNOWLEDGEMENTS

This work was supported by Conselho Nacional de Desenvolvimento Cientifico e Tecnológico (CNPq), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Programa de Apoio a Núcleos de Excelencia (PRONEX), Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Instituto Nacional de Ciência e Tecnologia (INCT-INOFAR).

REFERENCES

1. McLaughlin VV. Looking to the future: a new decade of pulmonary arterial hypertension therapy. Eur Respir Rev. 2011; 20: 262-269.

2. Simonneau G, Robbins IM, Beghetti M, Channick RN, Delcroix M, Denton CP, Elliott CG. Updated clinical classification of pulmonary hypertension. J Am Coll Cardiol. 2009; 54: S43-54.

3. Task Force for Diagnosis and Treatment of Pulmonary Hypertension of European Society of Cardiology (ESC); European Respiratory Society (ERS); International Society of Heart and Lung Transplantation (ISHLT), Galiè N, Hoeper MM, Humbert M, Torbicki A, Vachiery JL, et al. Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Respir J. 2009; 34: 1219-1263.

4. Montani D, Chaumais MC, Guignabert C, Günther S, Girerd B, Jaïs X, et al. Targeted therapies in pulmonary arterial hypertension. Pharmacol Ther. 2014; 141: 172-191.

5. dos Santos Fernandes CJ, Jardim CV, Hovnanian A, Hoette S, Dias BA, Souza S, et al. Survival in schistosomiasis-associated pulmonary arterial hypertension. J Am Coll Cardiol. 2010; 56: 715-720.

6. Schermuly RT, Ghofrani HA, Wilkins MR, Grimminger F. Mechanisms of disease: pulmonary arterial hypertension. Nat Rev Cardiol. 2011; 8: 443-455.

7. Simonneau G, Robbins IM, Beghetti M, Channick RN, Delcroix M, Denton CP, Elliott CG. Updated clinical classification of pulmonary hypertension. J Am Coll Cardiol. 2009; 54: S43-54.

8. Chin KM, Rubin LJ. Pulmonary arterial hypertension. J Am Coll Cardiol. 2008; 51: 1527-1538.

9. Humbert M, Sitbon O, Simonneau G. Treatment of pulmonary arterial hypertension. N Engl J Med. 2004; 351: 1425-1436.

10. Sitbon O, Morrell N. Pathways in pulmonary arterial hypertension: the future is here. Eur Respir Rev. 2012; 21: 321-327.

11. Galiè N, Ghofrani AH. New horizons in pulmonary arterial hypertension therapies. Eur Respir Rev. 2013; 22: 503-514.

12. Baliga RS, MacAllister RJ, Hobbs AJ. New perspectives for the treatment of pulmonary hypertension. Br J Pharmacol. 2011; 163: 125-140.

13. Humbert M, Sitbon O, Chaouat A, Bertocchi M, Habib G, Gressin V, Yaïci A. Survival in patients with idiopathic, familial, and anorexigenassociated pulmonary arterial hypertension in the modern management era. Circulation. 2010; 122: 156-163.

14. Ling Y, Johnson MK, Kiely DG, Condliffe R, Elliot CA, Gibbs JS. Changing demographics, epidemiology, and survival of incident pulmonary arterial hypertension: results from the pulmonary hypertension registry of the United Kingdom and Ireland. Am J Respir Crit Care Med. 2012; 186: 790-796.

15. Benza RL, Miller DP, Barst RJ, Badesch DB, Frost AE, McGoon MD. An evaluation of long-term survival from time of diagnosis in pulmonary arterial hypertension from the REVEAL Registry. Chest. 2012; 142: 448-456.

16. Alencar AK1, Pereira SL, Montagnoli TL, Maia RC, Kümmerle AE, Landgraf SS, Caruso-Neves C. Beneficial effects of a novel agonist of the adenosine A2A receptor on monocrotaline-induced pulmonary hypertension in rats. Br J Pharmacol. 2013; 169: 953-962.

17. Xu MH1, Gong YS, Su MS, Dai ZY, Dai SS, Bao SZ, Li N. Absence of the adenosine A2A receptor confers pulmonary arterial hypertension and increased pulmonary vascular remodeling in mice. J Vasc Res. 2011; 48: 171-183.

18. Alencar AK, Pereira SL, da Silva FE, Mendes LV, Cunha VN, Lima LM. N-acylhydrazone derivative ameliorates monocrotaline-induced pulmonary hypertension through the modulation of adenosine A2AR activity. Int J Cardiol. 2014; 173: 154-162.

19. Shryock JC, Belardinelli L. Adenosine and adenosine receptors in the cardiovascular system: biochemistry, physiology, and pharmacology. Am J Cardiol. 1997; 79: 2-10.

20. Newby AC. Adenosine and the concept of ‘‘retaliatory metabolites. Trends Biochem Sci. 1984; 9: 42-44.

21. Antonioli L, Fornai M, Colucci R, Ghisu N, Tuccori M, Del Tacca M, et al. Pharmacological modulation of adenosine system: novel options for treatment of inflammatory bowel diseases. Inflamm Bowel Dis. 2008; 14: 566-574.

22. Zhou Y, Schneider DJ, Blackburn MR. Adenosine signaling and the regulation of chronic lung disease. Pharmacol Ther. 2009; 123: 105- 116.

23. Haskó G, Cronstein BN. Adenosine: an endogenous regulator of innate immunity. Trends Immunol. 2004; 25: 33-39.

24. Caruso M, Alamo A, Crisafulli E, Raciti C, Fisichella A, Polosa R. Adenosine signaling pathways as potential therapeutic targets in respiratory disease. Expert Opin Ther Targets. 2013; 17: 761-772.

25. Del Ry S, Cabiati M, Lionetti V, Aquaro GD, Martino A, Mattii L, et al. Pacing-induced regional differences in adenosine receptors mRNA expression in a swine model of dilated cardiomyopathy. PLoS One. 2012; 7: e47011.

26. Fredholm BB. Adenosine, an endogenous distress signal, modulates tissue damage and repair. Cell Death Differ. 2007; 14: 1315-1323.

27. Jones CE, Mayer LR, Smith EE, Hurst TW. Relaxation of the isolated coronary artery by inosine: noninvolvement of the adenosine receptor. J Cardiovasc Pharmacol. 1981; 3: 612-621.

28. Shirahase H, Usui H, Manabe K, Kurahashi K, Fujiwara M. Endotheliumdependent contraction and -independent relaxation induced by adenine nucleotides and nucleoside in the canine basilar artery. J Pharmacol Exp Ther. 1988; 247: 1152-1157.

29. King AD, Milavec-Krizman M, Müller-Schweinitzer E. Characterization of the adenosine receptor in porcine coronary arteries. Br J Pharmacol. 1990; 100: 483-486.

30. Yen MH, Wu CC, Chiou WF. Partially endothelium-dependent vasodilator effect of adenosine in rat aorta. Hypertension. 1988; 11: 514-518.

31. D’Orléans-Juste P, Dion S, Mizrahi J, Regoli D. Effects of peptides and non-peptides on isolated arterial smooth muscles: role of endothelium. Eur J Pharmacol. 1985; 114: 9-21.

32. Della Latta V, Cabiati M, Rocchiccioli S, Del Ry S, Morales MA. The role of the adenosinergic system in lung fibrosis. Pharmacol Res. 2013; 76: 182-189.

33. Haskó G, Linden J, Cronstein B, Pacher P. Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug Discov. 2008; 7: 759-770.

34. Fredholm BB, Arslan G, Halldner L, Kull B, Schulte G, Wasserman W. Structure and function of adenosine receptors and their genes. Naunyn Schmiedebergs Arch Pharmacol. 2000; 362: 364-374.

35. Hiley CR, Bottrill FE, Warnock J, Richardson PJ. Effects of pH on responses to adenosine, CGS 21680, carbachol and nitroprusside in the isolated perfused superior mesenteric arterial bed of the rat. Br J Pharmacol. 1995; 116: 2641-2646.

36. Rump LC, Jabbari-T J, von Kügelgen I, Oberhauser V. Adenosine mediates nitric-oxide-independent renal vasodilation by activation of A2A receptors. J Hypertens. 1999; 17: 1987-1993.

37. Flood A, Headrick JP. Functional characterization of coronary vascular adenosine receptors in the mouse. Br J Pharmacol. 2001; 133: 1063- 1072.

38. Stoggall SM, Shaw JS. The coexistence of adenosine A1 and A2 receptors in guinea-pig aorta. Eur J Pharmacol. 1990; 190: 329-335.

39. Prentice DJ, Hourani SM. Activation of multiple sites by adenosine analogues in the rat isolated aorta. Br J Pharmacol. 1996; 118: 1509- 1517.

40. Prentice DJ, Hourani SM. Characterisation of adenosine receptors mediating relaxation in hamster isolated aorta. Naunyn Schmiedebergs Arch Pharmacol. 2000; 362: 427-434.

41. El-Kashef H, Elmazar MM, Al-Shabanah OA, Al-Bekairi AM. Effect of adenosine on pulmonary circulation of rabbits. Gen Pharmacol. 1999; 32: 307-313.

42. Mustafa SJ, Morrison RR, Teng B, Pelleg A. Adenosine receptors and the heart: role in regulation of coronary blood flow and cardiac electrophysiology. Handb Exp Pharmacol. 2009; 161-188.

43. Talukder MA, Morrison RR, Jacobson MA, Jacobson KA, Ledent C, Mustafa SJ. Targeted deletion of adenosine A(3) receptors augments adenosine-induced coronary flow in isolated mouse heart. Am J Physiol Heart Circ Physiol. 2002; 282: H2183-2189.

44. Ponnoth DS, Sanjani MS, Ledent C, Roush K, Krahn T, Mustafa SJ. Absence of adenosine-mediated aortic relaxation in A(2A) adenosine receptor knockout mice. Am J Physiol Heart Circ Physiol. 2009; 297: H1655-1660.

45. Abebe W, Makujina SR, Mustafa SJ. Adenosine receptor-mediated relaxation of porcine coronary artery in presence and absence of endothelium. Am J Physiol. 1994; 266: H2018-2025.

46. Belardinelli L, Shryock JC, Snowdy S, Zhang Y, Monopoli A, Lozza G, et al. The A2A adenosine receptor mediates coronary vasodilation. J Pharmacol Exp Ther. 1998; 284: 1066-1073.

47. Hein TW, Belardinelli L, Kuo L. Adenosine A2A receptors mediate coronary microvascular dilation to adenosine: role of nitric oxide and ATP-sensitive potassium channels. J Pharmacol Exp Ther. 1999; 291: 655-664.

48. Morrison RR, Talukder MA, Ledent C, Mustafa SJ. Cardiac effects of adenosine in A(2A) receptor knockout hearts: uncovering A(2B) receptors. Am J Physiol Heart Circ Physiol. 2002; 282: H437-444.

49. Talukder MA, Morrison RR, Mustafa SJ. Comparison of the vascular effects of adenosine in isolated mouse heart and aorta. Am J Physiol Heart Circ Physiol. 2002; 282: H49-57.

50. Gazoni LM, Laubach VE, Mulloy DP, Bellizzi A, Unger EB, Linden J, et al. Additive protection against lung ischemia-reperfusion injury by adenosine A2A receptor activation before procurement and during reperfusion. J Thorac Cardiovasc Surg. 2008; 135: 156-165.

51. Che J, Chan ES, Cronstein BN. Adenosine A2A receptor occupancy stimulates collagen expression by hepatic stellate cells via pathways involving protein kinase A, Src, and extracellular signal-regulated kinases 1/2 signaling cascade or p38 mitogen-activated protein kinase signaling pathway. Mol Pharmacol. 2007; 72: 1626-1636.

52. Zezula J, Freissmuth M. The A(2A)-adenosine receptor: a GPCR with unique features? Br J Pharmacol. 2008; 153 Suppl 1: S184-190.

53. Zhu Y, Liu L, Peng X, Ding X, Yang G, Li T,. Role of adenosine A2A receptor in organ-specific vascular reactivity following hemorrhagic shock in rats. J Surg Res. 2013; 184: 951-958.

54. Leal CM, Pereira SL, Kümmerle AE, Leal DM, Tesch R, de Sant’Anna CM, Fraga CA. Antihypertensive profile of 2-thienyl-3,4- methylenedioxybenzoylhydrazone is mediated by activation of the A2A adenosine receptor. Eur J Med Chem. 2012; 55: 49-57.

55. Pearl RG. Adenosine produces pulmonary vasodilation in the perfused rabbit lung via an adenosine A2 receptor. Anesth Analg. 1994; 79: 46- 51.

56. Martin PL, Potts AA. The endothelium of the rat renal artery plays an obligatory role in A2 adenosine receptor-mediated relaxation induced by 5’-N-ethylcarboxamidoadenosine and N6-cyclopentyladenosine. J Pharmacol Exp Ther. 1994; 270: 893-899.

57. Leal S, Sá C, Gonçalves J, Fresco P, Diniz C. Immunohistochemical characterization of adenosine receptors in rat aorta and tail arteries. Microsc Res Tech. 2008; 71: 703-709.

58. Lewis CD, Hourani SM, Long CJ, Collis MG. Characterization of adenosine receptors in the rat isolated aorta. Gen Pharmacol. 1994; 25: 1381-1387.

59. Prentice DJ, Hourani SM. Activation of multiple sites by adenosine analogues in the rat isolated aorta. Br J Pharmacol. 1996; 118: 1509- 1517.

60. Ikeda U, Kurosaki K, Ohya K, Shimada K. Adenosine stimulates nitric oxide synthesis in vascular smooth muscle cells. Cardiovasc Res. 1997; 35: 168-174.

61. Ray CJ, Marshall JM. The cellular mechanisms by which adenosine evokes release of nitric oxide from rat aortic endothelium. J Physiol. 2006; 570: 85-96.

62. Ko EA, Han J, Jung ID, Park WS. Physiological roles of K+ channels in vascular smooth muscle cells. J Smooth Muscle Res. 2008; 44: 65-81.

63. Marquardt DL, Walker LL, Heinemann S. Cloning of two adenosine receptor subtypes from mouse bone marrow-derived mast cells. J Immunol. 1994; 152: 4508-4515.

64. Fredholm BB, Zhang Y, van der Ploeg I. Adenosine A2A receptors mediate the inhibitory effect of adenosine on formyl-Met-Leu-Phe-stimulated respiratory burst in neutrophil leucocytes. Naunyn Schmiedebergs Arch Pharmacol. 1996; 354: 262-267.

65. Koshiba M, Rosin DL, Hayashi N, Linden J, Sitkovsky MV. Patterns of A2A extracellular adenosine receptor expression in different functional subsets of human peripheral T cells. Flow cytometry studies with anti-A2A receptor monoclonal antibodies. Mol Pharmacol. 1999; 55: 614-624.

66. Xu H, Stein B, Liang B. Characterization of a stimulatory adenosine A2a receptor in adult rat ventricular myocyte. Am J Physiol. 1996; 270: H1655-1661.

67. McIntosh VJ, Lasley RD. Adenosine receptor-mediated cardioprotection: are all 4 subtypes required or redundant? J Cardiovasc Pharmacol Ther. 2012; 17: 21-33.

68. Marala RB, Mustafa SJ. Immunological characterization of adenosine A2A receptors in human and porcine cardiovascular tissues. J Pharmacol Exp Ther. 1998; 286: 1051-1057.

69. Chandrasekera PC, McIntosh VJ, Cao FX, Lasley RD. Differential effects of adenosine A2a and A2b receptors on cardiac contractility. Am J Physiol Heart Circ Physiol. 2010; 299: H2082-2089.

70. Woodiwiss AJ, Honeyman TW, Fenton RA, Dobson JG Jr. Adenosine A2a-receptor activation enhances cardiomyocyte shortening via Ca2+-independent and -dependent mechanisms. Am J Physiol. 1999; 276: H1434-1441.

71. Monahan TS, Sawmiller DR, Fenton RA, Dobson JG Jr. Adenosine A(2a)-receptor activation increases contractility in isolated perfused hearts. Am J Physiol Heart Circ Physiol. 2000; 279: H1472-1481.

72. Chan TO, Funakoshi H, Song J, Zhang XQ, Wang J, Chung PH, et al. Cardiac-restricted overexpression of the A(2A)-adenosine receptor in FVB mice transiently increases contractile performance and rescues the heart failure phenotype in mice overexpressing the A(1)- adenosine receptor. Clin Transl Sci. 2008; 1: 126-133.

73. Feldman AM, Cheksis-Feiner E, Hamad E, Chan T. Adenosine receptor subtypes and the heart failure phenotype: translating lessons from mice to man. Trans Am Clin Climatol Assoc. 2011; 122: 198-214.

74. Dubey RK, Gillespie DG, Shue H, Jackson EK. A(2B) receptors mediate antimitogenesis in vascular smooth muscle cells. Hypertension. 2000; 35: 267-272.

75. Bi LQ, Zhu R, Kong H, Wu SL, Li N, Zuo XR, et al. Ruscogenin attenuates monocrotaline-induced pulmonary hypertension in rats. Int Immunopharmacol. 2013; 16: 7-16.

76. Janssen W, Schermuly RT, Kojonazarov B. The role of cGMP in the physiological and molecular responses of the right ventricle to pressure overload. Exp Physiol. 2013; 98: 1274-1278.

77. Saadjian AY, Paganelli F, Gaubert ML, Levy S, Guieu RP. Adenosine plasma concentration in pulmonary hypertension. Cardiovasc Res. 1999; 43: 228-236.

78. Dubey RK, Gillespie DG, Mi Z, Suzuki F, Jackson EK. Smooth muscle cell-derived adenosine inhibits cell growth. Hypertension. 1996; 27: 766-773.

79. Moudgil R, Michelakis ED, Archer SL. Hypoxic pulmonary vasoconstriction. J Appl Physiol (1985). 2005; 98: 390-403.

80. Varani K, Caramori G, Vincenzi F, Adcock I, Casolari P, Leung E, Maclennan S. Alteration of adenosine receptors in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2006; 173: 398-406.

81. Schindler CW, Karcz-Kubicha M, Thorndike EB, Müller CE, Tella SR, Ferré S, Goldberg SR. Role of central and peripheral adenosine receptors in the cardiovascular responses to intraperitoneal injections of adenosine A1 and A2A subtype receptor agonists. Br J Pharmacol. 2005; 144: 642-650.

Received : 03 Jul 2014
Accepted : 10 Oct 2014
Published : 13 Oct 2014
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X