Next Article in Journal
Neuroprotective Effects of Growth Hormone (GH) and Insulin-Like Growth Factor Type 1 (IGF-1) after Hypoxic-Ischemic Injury in Chicken Cerebellar Cell Cultures
Next Article in Special Issue
Identification of Anti-Severe Acute Respiratory Syndrome-Related Coronavirus 2 (SARS-CoV-2) Oxysterol Derivatives In Vitro
Previous Article in Journal
Insight into the bZIP Gene Family in Solanum tuberosum: Genome and Transcriptome Analysis to Understand the Roles of Gene Diversification in Spatiotemporal Gene Expression and Function
Previous Article in Special Issue
Molecular Modeling Study of c-KIT/PDGFRα Dual Inhibitors for the Treatment of Gastrointestinal Stromal Tumors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Discovery of a Bradykinin B2 Partial Agonist Profile of Raloxifene in a Drug Repurposing Campaign

by
Patricia Gomez-Gutierrez
and
Juan J. Perez
*
Department of Chemical Engineering, Universitat Politecnica de Catalunya, ETSEIB, Av. Diagonal, 647, 08028 Barcelona, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(1), 257; https://doi.org/10.3390/ijms22010257
Submission received: 16 November 2020 / Revised: 14 December 2020 / Accepted: 28 December 2020 / Published: 29 December 2020
(This article belongs to the Special Issue Exploiting the Multitarget and Repositioned Drugs Approach)

Abstract

:
Covid-19 urges a deeper understanding of the underlying molecular mechanisms involved in illness progression to provide a prompt therapeutical response with an adequate use of available drugs, including drug repurposing. Recently, it was suggested that a dysregulated bradykinin signaling can trigger the cytokine storm observed in patients with severe Covid-19. In the scope of a drug repurposing campaign undertaken to identify bradykinin antagonists, raloxifene was identified as prospective compound in a virtual screening process. The pharmacodynamics profile of raloxifene towards bradykinin receptors is reported in the present work, showing a weak selective partial agonist profile at the B2 receptor. In view of this new profile, its possible use as a therapeutical agent for the treatment of severe Covid-19 is discussed.

1. Introduction

About 20% of patients infected with SARS-Cov-2 develop respiratory complications, including pneumonia, that may evolve to severe hypoxemia, acute respiratory distress syndrome (ARDS), pulmonary edema, shock, metabolic acidosis, coagulation dysfunction and multiple organ functional failure [1,2]. Covid-19 patients with severe symptoms exhibit an immune response characterized by elevated serum levels of diverse proinflammatory molecules, including interleukin-6 (IL-6) [3,4,5,6]. Presently, these patients are treated with anti-inflammatory drugs like dexamethasone [7] or cytokine inhibitors, combined with antibiotics to treat secondary infections, sometimes combined with remdesivir, an antiviral originally developed to treat hepatitis C and tested against the Ebola virus disease to reduce the viral load [8].
Treatment of Covid-19 urges a deeper understanding of the underlying molecular mechanisms involved in disease progression to provide a prompt therapeutical response with an adequate use of available drugs, including drug repurposing. Indeed, there is a number of ongoing clinical trials to test the efficacy of diverse FDA-approved drugs against Covid-19. Among them, a few involve assessment of the therapeutic efficacy of IL-6 inhibition, unfortunately without reporting conclusive results [9,10,11]. Recently, it was proposed that dysregulated bradykinin (BK) signaling could be the trigger of the cytokine storm observed in patients with severe Covid-19 [12,13,14,15]. The rational for this hypothesis relies on the increased availability of BK and its metabolite des-Arg9-BK in Covid-19 patients, due to the downregulation of angiotensin converting enzyme 2 (ACE2), the entry protein of SARS-Cov-2 [16]. Indeed, ACE2 downregulation increases the amount of angiotensin I processed by the angiotensin-converting enzyme (ACE) with a concomitant decrease of its degradation activity on BK. Moreover, levels of des-Arg9-BK are also increased due to the diminished degradative activity of ACE2 caused by its downregulation [12]. The BK hypothesis is consistent with the observed elevated levels of IL-6 in Covid-19 patients [17]. This new perspective suggests that inhibition of BK signaling may be a suitable therapy to avoid the cytokine storm and its consequences. Indeed, as a proof of principle, preliminary results from a limited study suggests that inhibition of the BK signaling is associated with a decrease of the oxygen supplementation needed by Covid-19 patients [18].
Nowadays, icatibant is the only BK antagonist approved as therapeutic agent for the treatment of hereditary angioedema [19], in spite of the number of selective BK ligands described in the literature [20]. The discovery of the bradykinin hypothesis prompted the launch of an ongoing clinical trial to assess the benefits of icatibant for the treatment of covid-19 patients [21]. In other to swiftly test the efficacy of alternative BK antagonists with diverse pharmacological profiles for the treatment of Covid-19, drug repurposing represents a valuable strategy [22]. To this aim, we recently carried out a virtual screening study on the DrugBank database [23] aimed at identifying BK ligands among drugs already approved by the FDA [24]. The study was carried out using four points common to the pharmacophores derived for the B1 and B2 bradykinin receptors recently reported that were used as queries for the search [25,26,27]. Hits identified were subsequently subjected to a docking study using the Molecular Operating Environment (MOE) program [28].
The virtual screening process permitted the identification of a small set of drugs as prospective nonselective BK ligands, as described elsewhere [24]. Specifically, the DrugBank database, containing a total of 1703 molecules, was screened for compounds that fulfilled a common pharmacophore for the B1 and B2 receptors. The study identified eight drugs including raloxifene, sildenafil, cefepime, cefpirome, imatinib, ponatinib, abemaciclib and entrectinib. Subsequently, the eight compounds were purchased and tested for their capacity to displace a reference ligand in any of the two bradykinin receptors B1 and B2, respectively at 20 µM. Three of the compounds including raloxifene, sildenafil and cefepime displaced the reference ligand from B2, which represents a 40% success rate as found in similar studies [29]. However, none of the compounds exhibited affinity for the B1 receptor at this concentration. This might be due to the steric hindrance that was not properly included in the virtual screening search. Raloxifene, the most potent antagonist identified was further investigated in a functional study in vitro, exhibiting an IC50 of ~16 µM. Moreover, the compound showed a weak partial agonist profile with a maximal activity of ~20%. Ralixofene is a selective estrogen receptor modulator, exerting estrogen agonist action in some target tissues while acting as an estrogen antagonist in others [30]. The compound was approved a few years ago for the treatment of osteoporosis in postmenopausal women, as well as for cancer chemoprotection. The discovery of a new pharmacodynamic profile of relixofene as a BK partial agonist opens an opportunity for its repurposing as a therapeutic agent for the treatment of severe Covid-19.

2. Results and Discussion

In order to have a preliminary insight into the affinity of ralixofene towards the bradykinin receptors, the compound was first tested at 20 µM for its capacity to displace a reference ligand from B1 and B2, respectively. Specifically, desArg10-kallidin was used in the case of the B1 receptor and NPC 567 for the B2 receptor. The results of this study indicate that at this concentration, ralixofene does not displace desArg10-kallidin at the B1 receptor, whereas the reference ligand at the B2 receptor is displaced ~54%. We subsequently proceeded to carry out a study of the pharmacodynamic profile of the compound towards the BK B2 receptor in vitro, using a functional efficacy measurement [31]. Accordingly, we tested its capacity to antagonize BK on Chinese Hamster Ovary (CHO) cells stably transfected with the B2 receptor.
As can be seen in Figure 1a, raloxifene antagonizes BK with an IC50 ~16 µM with an apparent dissociation constant KB = 1.8 µM, computed using the modified Cheng Prusoff equation [32]:
K B = IC 50 1 + A EC 50
where, IC50 = 16 µM corresponds to raloxifene and EC50 = 2.4 pM corresponds to BK, and A = 0.03 nM is the concentration of BK that was used in the experiments. We also tested its activity as an agonist on CHO cells stably transfected with the B2 receptor. Present results also showed (Figure 1b) that the compound exhibited a partial agonistic profile, with a maximum effect of 20% of BK that was achieved at around 20 µM.
The antagonistic profile of the compound can be rationalized from docking studies. Figure 2 shows the prospective bound conformation of raloxifene (orange) to the B2 receptor, superimposed with the conformation matching the query (yellow) from the virtual screening process. Despite fulfilling the four pharmacophore points, the directionality of the hydrogen bond interactions (not included in the query) was not optimal. However, in the docking process, the molecule was relaxed to satisfy new interactions that may improve its docking score. As can be seen in Figure 2, from its initial position (yellow), the molecule tilts to get a few favorable interactions (orange). Specifically, the phenoxyl moiety sacrifices a hydrogen bond interaction with Arg169 that is compensated with a new one with Asn104. This in turn, permits the phenoxyl aromatic ring to get closer to Trp86 and the carbonyl group establishes a hydrogen bond interaction with Arg169. On the other hand, some interactions are weakened, for example, the interaction of the bicycle moiety with Trp86 and the interaction of the pyridine nitrogen with Asp266. Partial fulfilment of the pharmacophore may explain the low IC50 exhibited by raloxifene [26]. On the other hand, the fact that raloxifene does not bind to the B1 can only be explained by steric interactions, since most of the residues between the two receptors are conserved [27]. Inspection of the results of the docking study reveals that the pyridine nitrogen of raloxifene exhibits a hydrogen bond with Thr286 that cannot not be established in B1, since the position is occupied by Leu294.
Bradykinin signaling though the B2 receptor induces a proinflammatory action as well as a hypotensive effect [33]. The proinflammatory activity of BK involves the stimulation of cytokines, like IL-6 and IL-8, through the MAPK/AP-1 signaling axis [17,34], whereas the hypotensive effects are due to vasodilation and increased vascular permeability. Due to the partial agonist profile of raloxifene, it is expected that the compound antagonizes the proinflammatory effects of BK, modulating the levels of diverse cytokines, including IL-6. To support this hypothesis, in clinical trials aimed at understanding the effect of hormone replacement therapy for menopausal women on the risk of cardiovascular disease, in contrast to other compounds raloxifene did not exhibit a proinflammatory profile [35,36] that could, in part, be explained by its antagonistic effect on the BK B2 receptor.
The beneficial vascular effects of ralixofene could, in part, be explained by its partial agonist profile towards the BK B2 receptor [36]. Thus, in a study aimed at assessing the vasoprotective effects of the compound, rats treated with raloxifene showed an increased reduction of systolic blood pressure on administration of bradykinin, suggesting an enhanced bioavailability of NO in these animals [37]. Moreover, diverse experiments subsequently confirmed a role of raloxifene inducing endothelium-dependent relaxation as due to the upregulation of nitric-oxide synthase (eNOS) expression [38,39,40]. Although these results can be due to the partial agonist profile of raloxifene reported in this work [33], the effect can also be explained as due to the activation of the GPR30 receptor [41]. Accordingly, further experimental work is necessary to differentiate both effects.
Present results strongly support the need to perform a deeper investigation on the use of raloxifene as a therapeutic agent for the treatment of Covid-19. As shown in the present communication, its pharmacological profile as a partial BK B2 agonist is expected to antagonize the inflammatory action of BK, shown to be upregulated in Covid-19 patients [12,13,14,15]. This, in turn, will produce a reduction of cytokine levels, including IL-6, which is the primary proinflammatory cytokine associated with the severity of the illness [5,6]. On the other hand, its partial agonism profile is expected to maintain a base level of BK, preserving its beneficial cardiovascular effects [42]. Despite the novel beneficial effects identified, caution should be paid due to the necessary higher dose required for the compound to be used for a novel therapeutic use. The treatment of osteoporosis requires an oral dose between 30 to 150 mg/d. Taking into account its poor bioavailability of 2%, a single dose of 100 mg gives a maximal concentration in plasma of ~2 nM [43]. Concentrations necessary for the therapeutical benefit of raloxifene towards the BK B2 receptor are expected to be, at most, around 100 times higher, which is within the therapeutic window of the drug [44].
The results of this work complement recent comments suggesting the use of raloxifene for the treatment of Covid-19 [45], based on its capacity to inhibit the IL-6/STAT3 signaling pathway [46,47]. In addition, the presumed antiviral profile of ralixofene against the SARS-Cov2 virus through its capability to block the two-pore channel TCP2 [48], as found for the ebola virus [49] has been underlined. In conclusion, the pharmacodynamic profile exhibited by ralidoxene described in the present report represents a reasonable base to test the compound for the treatment of Covid-19. Very recently a clinical trial in Italy has been approved to test ralixofene in COVID-19 patients with mild symptoms [50].

3. Materials and Methods

3.1. Computational Methods

Vitual screening on the DrugBank [23] was carried out using the Molecular Operating Environment (MOE) program [28], using four common points of the BK B1 and B2 pharmacophores described previously [25,26] that were used as a query. Previously, a 3D DrugBank database containing the 3D structure, together with a set of conformations of each molecule, was constructed as explained elsewhere [24]. Virtual screening was carried out on the subset of molecules with molecular weight between 200 and 600 (a total of 1703 molecules). Hits identified in the screening process were docked in the 3D models of the B1 and B2 bradykinin receptors, using a set of unique conformations resulting from thorough conformational searches for the diverse ligands studied and rank ordered by means of the MOE program [28].

3.2. In Vitro Assays

3.2.1. Ligand Displacement Assays

Raloxifene was tested for its ability to displace reference ligands on human recombinant bradykinin B1 or B2 receptors, respectively, expressed in CHO cells at 20 µM. For this purpose, first saturation isotherms were obtained with reference ligands ([3H] des-Arg10-BK (0.35 nM) in the case of B1, and [3H]-bradykinin (0.2 nM) for B2) incubated for 60 min at room temperature. Nonspecific binding was evaluated by adding a reference compound (desArg9[Leu8]-BK at 10 µM in the case of B1 and BK at 1 µM for B2). Antagonism of raloxifene was measured as a percent inhibition of specific binding of [3H]-bradykinin as control, obtained in the presence of raloxifene at 20 µM and using as reference compounds desArg10-KD for B1 and NPC-567 for B2.

3.2.2. Functional Efficacy Assays

Measurementent of the efficacy of the compound on the B2 receptor was carried out following the protocol described elsewhere [31]. The method is based on measuring differences in intracellular Ca2+ concentrations produced in diverse conditions on CHO cells expressing recombinant human B2 receptor by fluorometry. Agonism was measured through the capacity of the compound to increase Ca2+ concentration compared to BK (EC50 ~ 2.4 pM). Antagonism was measured through the capacity to antagonize BK based on the reduction of Ca2+ concentration.

4. Conclusions

The aim of the present study was to characterize the pharmacological profile of raloxifene towards the bradykinin receptors. The compound was identified as a micromolar ligand of the B2 receptor by virtual screening in a drug repurposing campaign. Results showed that raloxifene is a weak partial agonist toward the B2 receptor, with a 19% efficacy compared to bradykinin, with an apparent dissociation constant KB ~ 21 µM. The discovery of the bradykinin pharmacodynamics profile of raloxidene explains in part its observed vascular beneficial effects, although they could also be due to activation of the GPR30 receptor signaling pathway. On the other hand, it acts as a bradykinin antagonist for B2 with an IC50 ~ 21 µM.
Based on the bradykinin hypothesis on the trigger of the cytokine storm in severe Covid-19 [12,13,14,15], these results suggest that the compound could be used for its treatment. Present results support previous claims suggesting the use of raloxifene based on its capacity to inhibit the IL-6/STAT3 signaling pathway [45], and also for being a prospective antiviral agent for SARS-Cov2 as inhibitor of the two-pore channel TCP2 [48]. Very recently a clinical trial in Italy has been approved to test its efficacy in COVID-19 patients with mild symptoms [50].

Author Contributions

Conceptualization, J.J.P.; methodology, P.G.-G.; calculations and formal analysis, P.G.-G.; writing, J.J.P. All authors have read and agreed to the published version of the manuscript.

Funding

The present study was funded by the Spanish Ministry of Science and Innovation through the Carlos III Institute of Health under contract COV20/00052.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available upon request from the corresponding author.

Acknowledgments

The antagonist potency of the hits discovered in the in silico study were carried out by Cerep under contracts FR095-0020745 and FR095-0021802 and is gratefully acknowledged.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wang, Y.; Wang, Y.; Chen, Y.; Qin, Q. Unique epidemiological and clinical features of the emerging 2019 novel coronavirus pneumonia (COVID-19) implicate special control measures. J. Med. Virol. 2020, 92, 568–576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Hirano, T.; Murakami, M. COVID-19: A New Virus, but a Familiar Receptor and Cytokine Release Syndrome. Immunity 2020, 52, 731–733. [Google Scholar] [CrossRef] [PubMed]
  3. Iwasaki, M.; Saito, J.; Zhao, H.; Sakamoto, A.; Hirota, K.; Ma, D. Inflammation Triggered by SARS-CoV-2 and ACE2 Augment Drives Multiple Organ Failure of Severe COVID-19: Molecular Mechanisms and Implications. Inflammation 2020, 1–22. [Google Scholar] [CrossRef] [PubMed]
  4. Wu, Z.; McGoogan, J.M. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: Summary of a report of 72314 cases from the Chinese Center for Disease Control and Prevention. J. Am. Med. Assoc. 2020, 323, 1239–1242. [Google Scholar] [CrossRef] [PubMed]
  5. Ruan, Q.; Yang, K.; Wang, W.; Jiang, L.; Song, J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020, 46, 846–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X.; et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: A retrospective cohort study. Lancet 2020, 395, 1054–1062. [Google Scholar] [CrossRef]
  7. Horby, P.; Lim, W.S.; Emberson, J.R.; Mafham, M.; Bell, J.L.; Linsell, L.; Staplin, N.; Brightling, C.; Ustianowski, A.; Elmahi, E.; et al. Dexamethasone in Hospitalized Patients with Covid-19—Preliminary Report. N. Engl. J. Med. 2020. [Google Scholar] [CrossRef]
  8. Beigel, J.H.; Tomashek, K.M.; Dodd, L.E.; Mehta, A.K.; Zingman, B.S.; Kalil, A.C.; Hohmann, E.; Chu, H.Y.; Luetkemeyer, A.; Kline, S.; et al. Remdesivir for the Treatment of Covid-19-Final Report. N. Engl. J. Med. 2020, 383, 1813–1826. [Google Scholar] [CrossRef]
  9. Xua, X.; Hanb, M.; Lia, T.; Sun, W.; Wang, D.; Fu, B.; Zhou, Y.; Zheng, X.; Yange, Y.; Li, X.; et al. Effective treatment of severe COVID-19 patients with tocilizumab. Proc. Natl. Acad. Sci. USA 2020, 117, 10970–10975. [Google Scholar] [CrossRef]
  10. Campochiaro, C.; Dagna, L. The conundrum of interleukin-6 blockade in COVID-19. Lancet Rheumatol. 2020, 2, e579–e580. [Google Scholar] [CrossRef]
  11. Parr, J.B. Time to Reassess Tocilizumab’s Role in COVID-19 Pneumonia. JAMA Intern. Med. 2020. [Google Scholar] [CrossRef] [PubMed]
  12. Sodhi, C.P.; Wohlford-Lenane, C.; Yamaguchi, Y.; Prindle, T.; Fulton, W.B.; Wang, S.; McCray, P.B., Jr.; Chappell, M.; Hackam, D.J.; Jia, H. Attenuation of pulmonary ACE2 activity impairs inactivation of des-Arg9 bradykinin/BKB1R axis and facilitates LPS-induced neutrophil infiltration. Am. J. Physiol. Lung Cell. Mol. Physiol. 2018, 314, L17–L31. [Google Scholar] [CrossRef] [PubMed]
  13. Van de Veerdonk, F.L.; Netea, M.G.; van Deuren, M.; van der Meer, J.W.M.; de Mast, Q.; Brüggemann, R.J.; van der Hoeven, H. Kallikrein-kinin blockade in patients with COVID-19 to prevent acute respiratory distress syndrome. eLife 2020, 9, e57555. [Google Scholar] [CrossRef] [PubMed]
  14. De Maat, S.; de Mast, Q.; Danser, A.H.J.; van de Veerdonk, F.L.; Maas, C. Impaired Breakdown of Bradykinin and Its Metabolites as a Possible Cause for Pulmonary Edema in COVID-19 Infection. Semin. Thromb. Hemost. 2020, 46, 835–837. [Google Scholar] [CrossRef] [PubMed]
  15. Dagnino, A.A.; Campos, M.M.; Silva, R.B.M. Kinins and Their Receptors in Infectious Diseases. Pharmaceuticals 2020, 13, 215. [Google Scholar] [CrossRef]
  16. Wang, Q.; Zhang, Y.; Wu, L.; Niu, S.; Song, C.; Zhang, Z.; Lu, G.; Qiao, C.; Hu, Y.; Yuen, K. Structural and functional basis of SARS-CoV-2 entry by using human ACE2. Cell 2020, 181, 894–904. [Google Scholar] [CrossRef]
  17. Huang, C.-D.; Tliba, O.; Panettieri, R.A., Jr.; Amrani, Y. Bradykinin Induces Interleukin-6 Production in Human Airway Smooth Muscle Cells. Am. J. Respir. Cell Mol. Biol. 2003, 28, 330–338. [Google Scholar] [CrossRef]
  18. Van de Veerdonk, F.L.; Kouijzer, I.J.E.; de Nooijer, A.H.; van der Hoeven, H.G.; Maas, C.; Netea, M.G.; Brüggemann, R.J.M. Outcomes Associated with Use of a Kinin B2 Receptor Antagonist Among Patients With COVID-19. JAMA Netw. Open 2020, 3, e2017708. [Google Scholar] [CrossRef]
  19. Bork, K.; Yasothan, U.; Kirkpatrick, P. Icatibant. Nat. Rev. Drug Discov. 2008, 7, 801–802. [Google Scholar] [CrossRef]
  20. Marceau, F.; Bachelard, H.; Bouthillier, J.; Fortin, J.-P.; Morissette, J.-P.G.; Bawolak, M.-T.; Charest-Morin, X.; Gera, L. Bradykinin receptors: Agonists, antagonists, expression, signaling, and adaptation to sustained stimulation. Int. Immunopharmacol. 2020, 82, 106305. [Google Scholar] [CrossRef] [Green Version]
  21. SPY COVID TRIAL: An Adaptive Platform Trial to Reduce Mortality and Ventilator Requirements for Critically Ill Patients. Available online: https://clinicaltrials.gov/ct2/show/NCT04488081?term=icatibant&draw=3&rank=19 (accessed on 29 December 2020).
  22. Gns, H.S.; Saraswathy, G.R.; Muraharia, M.; Krishnamurthy, M. An update on Drug Repurposing: Re-written saga of the drug’s fate. Biomed. Pharm. 2019, 110, 700–716. [Google Scholar] [CrossRef] [PubMed]
  23. Wishart, D.S.; Feunang, Y.D.; Guo, A.C.; Lo, E.J.; Marcu, A.; Grant, J.R.; Sajed, T.; Johnson, D.; Li, C.; Sayeeda, Z.; et al. DrugBank 5.0: A major update to the DrugBank database for 2018. Nucleic Acids Res. 2018, 46, D1074–D1082. [Google Scholar] [CrossRef] [PubMed]
  24. Rasaeifar, B.; Gomez-Gutierrez, P.; Perez, J.J. Molecular Features of Non-Selective Small Molecule Antagonists of the Bradykinin Receptors. Pharmaceuticals 2020, 13, 259. [Google Scholar] [CrossRef] [PubMed]
  25. Lupala, C.L.; Gomez-Gutierrez, P.; Perez, J.J. New insights into the stereochemical requirements of the bradykinin B1 receptor antagonist binding. J. Mol. Graph. Model. 2016, 68, 184–196. [Google Scholar] [CrossRef] [Green Version]
  26. Lupala, C.L.; Gomez-Gutierrez, P.; Perez, J.J. New insights into the stereochemical requirements of the bradykinin B2 receptor antagonist binding. J. Comp. Aided Mol. Des. 2016, 30, 85–101. [Google Scholar] [CrossRef] [Green Version]
  27. Rasaeifar, B.; Lupala, C.S.; Gomez-Gutierrez, P.; Perez, J.J. Molecular Features Characterizing Non-peptide B1 and B2 Bradykinin Receptor Selectivity. Bioorg. Med. Chem. Lett. 2019, 29, 11–14. [Google Scholar] [CrossRef]
  28. Chemical Computing Group UCL. Molecular Operating Environment (MOE); January 2019; Chemical Computing Group UCL: Montreal, QC, Canada, 2020. [Google Scholar]
  29. Krishna, S.; Singh, D.K.; Meena, S.; Datta, D.; Siddiqi, M.I.; Banerjee, D. Pharmacophore-Based Screening and Identification of Novel Human Ligase I Inhibitors with Potential Anticancer Activity. J. Chem. Inf. Model. 2014, 54, 781–792. [Google Scholar] [CrossRef]
  30. Goldfrank, D.; Haytoglu, T.; Frishman, W.H.; Mohammad, Z. Raloxifene, a New Selective Estrogen Receptor Modulator. J. Clin. Pharmacol. 1999, 39, 767–774. [Google Scholar] [CrossRef]
  31. Simpson, P.B.; Woollacott, A.J.; Hill, R.G.; Seabrook, G.R. Functional characterization of bradykinin analogues on recombinant human bradykinin B1 and B2 receptors. Eur. J. Pharmacol. 2000, 392, 1–9. [Google Scholar] [CrossRef]
  32. Cheng, Y.; Prusoff, W.H. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem. Pharmacol. 1973, 22, 3099–3108. [Google Scholar]
  33. Leeb-Lundberg, L.M.F.; Marceau, F.; Muller-Esterl, W.; Pettibone, D.J.; Zuraw, B.L. International union of pharmacology. XLV. Classification of the kinin receptor family: From molecular mechanisms to pathophysiological consequences. Pharmacol. Rev. 2005, 57, 27–77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Hayashi, R.; Yamashita, N.; Matsui, S.; Fujita, T.; Araya, J.; Sassa, K.; Arai, N.; Yoshida, Y.; Kashii, T.; Maruyama, M.; et al. Bradykinin stimulates IL-6 and IL-8 production by human lung fibroblasts through ERK- and p38 MAPK-dependent mechanisms. Eur. Respir. J. 2000, 16, 452–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Gol, M.; Akanb, P.; Dogana, E.; Karas, C.; Saygili, U.; Posaci, C. Effects of estrogen, raloxifene, and hormone replacement therapy on serum C-reactive protein and homocysteine levels. Maturitas 2006, 53, 252–259. [Google Scholar] [CrossRef] [PubMed]
  36. Ciccone, M.M.; Scicchitano, P.; Cortese, F.; Gesualdo, M.; Zito, A.; Tesorio, M.; Guida, P.; Papagni, A.; Federici, A.; Cicinelli, E. Modulation of vascular tone control under isometric muscular stress: Role of estrogen receptors. Vasc. Pharmacol. 2013, 58, 127–133. [Google Scholar] [CrossRef] [PubMed]
  37. Wassmann, S.; Laufs, U.; Stamenkovic, D.; Linz, W.; Stasch, J.-P.; Ahlbory, K.; Rösen, R.; Böhm, M.; Nickenig, G. Raloxifene Improves Endothelial Dysfunction in Hypertension by Reduced Oxidative Stress and Enhanced Nitric Oxide Production. Circulation 2002, 10, 2083–2091. [Google Scholar] [CrossRef] [Green Version]
  38. Leung, F.P.; Yung, L.M.; Leung, H.S.; Au, C.L.; Yao, X.; Vanhoutte, P.M.; Laher, I.; Huang, Y. Therapeutic concentrations of raloxifene augment nitric oxide-dependent coronary artery dilatation in vitro. Br. J. Pharmacol. 2007, 152, 223–229. [Google Scholar] [CrossRef] [Green Version]
  39. Chan, C.; Leung, F.-P.; Yao, X.; Lau, C.-W.; Vanhoutte, P.M.; Huang, Y. Raloxifene Modulates Pulmonary Vascular Reactivity in Spontaneously Hypertensive Rats. J. Cardiovasc. Pharmacol. 2007, 49, 355–361. [Google Scholar] [CrossRef]
  40. Wong, C.M.; Au, C.L.; Tsang, S.Y.; Lau, C.W.; Yao, X.; Cai, Z.; Chung, A.C. Role of inducible nitric oxide synthase in endothelium-independent relaxation to raloxifene in rat aorta. Br. J. Pharmacol. 2017, 174, 718–733. [Google Scholar] [CrossRef] [Green Version]
  41. VandeVrede, L.; Abdelhamid, R.; Qin, Z.; Choi, J.; Piyankarage, S.; Luo, J.; Larson, J.; Bennett, B.M.; Thatcher, G.R.J. An NO Donor Approach to Neuroprotective and Procognitive Estrogen Therapy Overcomes Loss of NO Synthase Function and Potentially Thrombotic Risk. PLoS ONE 2013, 8, e70740. [Google Scholar] [CrossRef] [Green Version]
  42. Manolis, A.J.; Marketou, M.E.; Gavras, I.; Gavras, H. Cardioprotective properties of bradykinin: Role of the B2 receptor. Hypertens. Res. 2010, 33, 772–777. [Google Scholar] [CrossRef] [Green Version]
  43. Scott, J.A.; Da Camara, C.C.; Early, J.E. Raloxifene: A selective estrogen receptor modulator. Am. Fam. Physician 1999, 60, 1131–1139. [Google Scholar] [PubMed]
  44. Goldstein, S.R.; Siddhanti, S.; Ciaccia, A.V.; Plouffe, L., Jr. A pharmacological review of selective oestrogenreceptor modulators. Hum. Reprod. Update 2000, 6, 212–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Smetana, K., Jr.; Rosel, D.; Brábek, J. Raloxifene and Bazedoxifene Could Be Promising Candidates for Preventing the COVID-19 Related Cytokine Storm, ARDS and Mortality. In Vivo 2020, 34, 3027–3028. [Google Scholar] [CrossRef] [PubMed]
  46. Li, H.; Xiao, H.; Lin, L.; Jou, D.; Kumari, V.; Lin, J.; Li, C. Drug design targeting protein-protein interactions (PPIs) using multiple ligand simultaneous docking (MLSD) and drug repositioning: Discovery of raloxifene and bazedoxifene as novel inhibitors of IL-6/GP130 interface. J. Med. Chem. 2014, 57, 632–641. [Google Scholar] [CrossRef] [PubMed]
  47. Luo, P.; Shi, W.; Wang, Y.; Ma, H.; Liu, T.; Yan, D.; Huo, S.; Guo, J.; Wang, M.; Li, C.; et al. Raloxifene inhibits IL-6/STAT3 signaling pathway and protects against high-fat-induced atherosclerosis in ApoE-/-mice. Life Sci. 2020, 261, 118304. [Google Scholar] [CrossRef]
  48. Gil, C.; Ginex, T.; Maestro, I.; Nozal, V.; Barrado-Gil, L.; Cuesta-Geijo, M.A.; Urquiza, J.; Ramírez, D.; Alonso, C.; Campillo, N.E.; et al. COVID-19: Drug Targets and Potential Treatments. J. Med. Chem. 2020, 63, 12359–12386. [Google Scholar] [CrossRef]
  49. Penny, C.J.; Vassileva, K.; Jha, A.; Yuan, Y.; Chee, X.; Yates, E.; Mazzon, M.; Kilpatrick, B.S.; Muallem, S.; Marsh, M.; et al. Mining of Ebola virus entry inhibitors identifies approved drugs as two-pore channel pore blockers. Biochim. Biophys. Acta Mol. Cell Res. 2019, 1866, 1151–1161. [Google Scholar] [CrossRef]
  50. Multicenter, Adaptive, Randomized, Placebo-Controlled, Double Blind, Parallel-Group Phase 2/3 Trial, to Study Efficacy and Safety of Two Doses of Raloxifene in Adult Paucisymptomatic COVID-19 Patients. Available online: https://www.clinicaltrialsregister.eu/ctr-search/search?query=eudract_number:2020-003936-25 (accessed on 29 December 2020).
Figure 1. Functional efficacy study of raloxifene on CHO cells stably transfected with the B2 receptor (N = 2). (a) Study of the agonistic effect of the compound compared to bradykinin; (b) study of the efficacy of the compound antagonizing bradykinin.
Figure 1. Functional efficacy study of raloxifene on CHO cells stably transfected with the B2 receptor (N = 2). (a) Study of the agonistic effect of the compound compared to bradykinin; (b) study of the efficacy of the compound antagonizing bradykinin.
Ijms 22 00257 g001
Figure 2. Superposition of the prospective bound conformation of raloxifene onto the B2 receptor (orange) and the matching structure of raloxifene (yellow) from the virtual screening process. The 3D structure of the B2 receptor was constructed by homology modeling and reported previously [27]. Color spheres represent the pharmacophore points characteristic of the ligands binding to B2 receptor: dark blue represents a positive charge moiety; magenta a hydrogen bond accepting center; light blue a hydrogen bond donor/acceptor center; green an aromatic/lipophilic center.
Figure 2. Superposition of the prospective bound conformation of raloxifene onto the B2 receptor (orange) and the matching structure of raloxifene (yellow) from the virtual screening process. The 3D structure of the B2 receptor was constructed by homology modeling and reported previously [27]. Color spheres represent the pharmacophore points characteristic of the ligands binding to B2 receptor: dark blue represents a positive charge moiety; magenta a hydrogen bond accepting center; light blue a hydrogen bond donor/acceptor center; green an aromatic/lipophilic center.
Ijms 22 00257 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gomez-Gutierrez, P.; Perez, J.J. Discovery of a Bradykinin B2 Partial Agonist Profile of Raloxifene in a Drug Repurposing Campaign. Int. J. Mol. Sci. 2021, 22, 257. https://doi.org/10.3390/ijms22010257

AMA Style

Gomez-Gutierrez P, Perez JJ. Discovery of a Bradykinin B2 Partial Agonist Profile of Raloxifene in a Drug Repurposing Campaign. International Journal of Molecular Sciences. 2021; 22(1):257. https://doi.org/10.3390/ijms22010257

Chicago/Turabian Style

Gomez-Gutierrez, Patricia, and Juan J. Perez. 2021. "Discovery of a Bradykinin B2 Partial Agonist Profile of Raloxifene in a Drug Repurposing Campaign" International Journal of Molecular Sciences 22, no. 1: 257. https://doi.org/10.3390/ijms22010257

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop