Next Article in Journal
A Review of Lignocellulosic-Derived Nanoparticles for Drug Delivery Applications: Lignin Nanoparticles, Xylan Nanoparticles, and Cellulose Nanocrystals
Next Article in Special Issue
Stability of Metronidazole and Its Complexes with Silver(I) Salts under Various Stress Conditions
Previous Article in Journal
N-Acetylaspartyl-Glutamate Metabolism in the Cingulated Cortices as a Biomarker of the Etiology in ASD: A 1H-MRS Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Chloroquine and Hydroxychloroquine Interact Differently with ACE2 Domains Reported to Bind with the Coronavirus Spike Protein: Mediation by ACE2 Polymorphism

1
Laboratory of General Biology, Department of Biology, University of Ha’il, Ha’il 81451, Saudi Arabia
2
Section of Histology—Cytology, Medicine College of Tunis, University of Tunis El Manar, Djebel Lakhdhar Road, La Rabta-Tunis 1007, Tunisia
3
Laboratory of Histo-Embryology and Cytogenetic, Medicine College of Sfax, University of Sfax, Majida Boulila Road, Sfax 3029, Tunisia
4
Laboratory of Genetics, Department of Biology, Aydin Adnan Menderes University, Aydin 09010, Turkey
5
Molecular Diagnostic and Personalized Therapeutics Unit, University of Ha’il, Ha’il 81451, Saudi Arabia
*
Author to whom correspondence should be addressed.
Molecules 2021, 26(3), 673; https://doi.org/10.3390/molecules26030673
Submission received: 22 December 2020 / Revised: 18 January 2021 / Accepted: 21 January 2021 / Published: 28 January 2021
(This article belongs to the Special Issue Pharmaceutical Analysis and Drug Stability Evaluation)

Abstract

:
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection inducing coronavirus disease 2019 (COVID-19) is still an ongoing challenge. To date, more than 95.4 million have been infected and more than two million deaths have been officially reported by the WHO. Angiotensin-converting enzyme (ACE) plays a key role in the disease pathogenesis. In this computational study, seventeen coding variants were found to be important for ACE2 binding with the coronavirus spike protein. The frequencies of these allele variants range from 3.88 × 10−3 to 5.47 × 10−6 for rs4646116 (K26R) and rs1238146879 (P426A), respectively. Chloroquine (CQ) and its metabolite hydroxychloroquine (HCQ) are mainly used to prevent and treat malaria and rheumatic diseases. They are also used in several countries to treat SARS-CoV-2 infection inducing COVID-19. Both CQ and HCQ were found to interact differently with the various ACE2 domains reported to bind with coronavirus spike protein. A molecular docking approach revealed that intermolecular interactions of both CQ and HCQ exhibited mediation by ACE2 polymorphism. Further explorations of the relationship and the interactions between ACE2 polymorphism and CQ/HCQ would certainly help to better understand the COVID-19 management strategies, particularly their use in the absence of specific vaccines or drugs.

1. Introduction

Chloroquine (CQ) and its metabolite hydroxychloroquine (HCQ) (Figure 1) are mainly used to prevent and treat malaria and rheumatic diseases (including rheumatoid and idiopathic arthritis and systemic lupus erythematous), respectively [1]. Recently, Xu et al. (2018) [2] reported efficient effects of CQ and HCQ in the treatment of cancer via autophagy inhibition. The half-life of HCQ is about one month and it takes about six months for a full elimination from the body [3]. CQ and HCQ act as chemotherapeutic agents against erythrocytic case plasmodium parasites following malarial infection. Both increase the pH of the parasite’s vacuole leading to disruption of its development and asexual reproduction [4].
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) started in Wuhan China. It has caused the worldwide COVID-19 (coronavirus disease 2019) pandemic. Currently, there are no specific drugs or vaccines available and people are still dying mainly with acute respiratory distress syndrome (ARDS) which is one of the main severe complications of COVID-19 [5]. Throughout the ongoing COVID-19 pandemic, the use of CQ and HCQ has been allowed in several countries to treat the SARS-CoV-2 infected people. It has been reported that both CQ and HCQ interfere with various cellular levels and might have a wide range of antiviral potencies even on cancer cells [2,6,7]. In fact, both inhibit the SARS-CoV-2 viral replication [8], decrease antigen processing and its presentation [9,10], and decrease the cellular activity via low secretion of inflammatory cytokines and type 1 interferon [5]. CQ and HCQ might also interfere with angiotensin-converting enzyme 2 (ACE2) receptor which is involved in COVID-19 and its symptoms [11]. Strong interactions have been reported between the SARS-CoV-2 RBD domain of the S protein and ACE2 [12]. In fact, SARS-CoV-2 binds, and then invades the target cells through ACE2 [13]. So far, cells highly expressing ACE2 such as lung, kidney, and vascular endothelial cells may be targeted by SARS-CoV-2 [14,15].
While ACE1 and ACE2 showed only 42% amino acid similarity, both cleave amino acids from the C-terminal chain of peptides [16]. It has been reported that ACE polymorphism might play a crucial role in the pathogenesis of COVID-19 using a multiple regression model [17,18]. Several studies have reported that some drugs, including CQ, HCQ, remdesivir, and favipiravir are useful in COVID-19 treatment strategies but, currently, two vaccine types, including an mRNA-based new technology, are authorized and recommended for use and several vaccines are undergoing large-scale (phase 3) clinical trials [19,20,21]. There has been no evidence of the benefits of using CQ and HCQ in the therapy of COVID-19. However, it is worthwhile to assess the interaction modes with various targets of interest, which may be used for the future design of new drugs or new diseases. In fact, recently there was a clear interest in these drugs during the ongoing COVID-19. A recent study reported the interaction of CQ and HCQ with ACE2. Similarly, some phytochemical compounds displayed potential therapeutic targets of COVID-19 via inhibition of ACE2 [22,23]. As far as we know, to date, no study has reported the different interactions of these drugs, particularly both CQ and HCQ, with the various ACE2 domains reported to bind with coronavirus spike protein and the mediation by ACE2 polymorphism. Hence, the present study aimed at identifying the different ACE2 variants which may interact with the SARS-CoV-2 virus and investigate their potential interactions with both CQ and HCQ using computational approaches. An overview is also given about the pharmacokinetics of CQ and HCQ using an ADME (absorption, distribution, metabolism and excretion) approach.

2. Materials and Methods

2.1. Structure and Genetic ACE2 Polymorphism

The genetic variants of human ACE2 and the allele frequencies were collected from Ensembl Genome Browser [24,25] and gnomAD [26]. Appropriate filters were used to select only the coding region of the different ACE2 variants. Accordingly, the coding region of seventeen variants of ACE2 gene that have been previously reported to bind with both SARS-CoV and SARS-CoV-2 [27,28] were selected for this study (Table 1). The corresponding protein sequences of human ACE2 (Q9BYF1) were retrieved from UniProt. Structures of the proteins were identified by PDB-BLAST and obtained from the RCSB protein data bank [29]. The amino acid changes and the allele frequencies were assessed.

2.2. In Silico Approach of ADME, Pharmacokinetics, and Docking Study

Each of the seventeen ACE2 variant receptors was used separately to assess its interactions with both chloroquine (CQ) and hydroxychloroquine (HCQ). The three-dimensional (3D) structures of CQ and HCQ were retrieved from the PubChem website (CID 2719 and CID 3652, respectively). SMILES notations were used for assessment of the pharmacokinetic and ADME parameters. The physicochemical and pharmacokinetics properties were assessed and compared. ADME (absorption, distribution, metabolism and excretion) characteristics were checked using SwissADME. AutoDock Vina was used for the generation of the different binding poses based on the CHARMM force field [30]. The different variants were prepared; water molecules and heteroatoms were removed. Then, the processed proteins with polar hydrogens and Coleman charges were used to generate different poses. Regarding chirality, (S)-enantiomers particularly S-13a, of both CQ and HCQ were used. Redocking was performed to check the efficiency of the docking assay. The predicted binding affinity and the intermolecular bonds were monitored and analyzed. The intermolecular bonds including conventional hydrogen bonds, carbon-hydrogen bonds, alkyl, Pi-alkyl, halogen, and van der Waals were explored using DS visualizer 2016.

3. Results and Discussion

The global COVID-19 pandemic is still an ongoing challenge because SARS-CoV-2 infection constitutes a serious threat both to human life and socioeconomic development [5]. Two vaccine types are authorized and recommended for use and some other COVID-19 vaccines are undergoing large-scale (phase 3) clinical trials [20]. In this study, the genetic variants of human ACE2 and the allele frequencies were collected from Ensembl Genome Browser [24,31] and gnomAD [26]. Seventeen coding variants of ACE2 were found to bind with the coronavirus spike protein. The interactions of CQ and HCQ with these ACE2 domain variants is well mediated by ACE2 polymorphism. Recognition of these interactions might be useful for better prognostic or shortening the recovery time in COVID-19 hospitalized patients. In fact, some COVID-19 useful drugs have been reported to shorten the time of recovery in United States hospitalized patients infected with SARS-CoV-2 [21,32].

3.1. ACE2 Coding Variants

Human ACE2 protein contains 805 amino acids and has two functional domains, i.e., N-terminal peptidase M2 domain and C-terminal collectrin domain, which have been reported to contain the residues involved in the spike protein binding [27,33]. This binding site is considered to be an entry door for the virus and several vaccine approaches are based on shutting this entry door in the host cells to combat this unprecedented pandemic [34]. Ensembl Genome Browser and gnomAD exhibited 345 and 242 natural ACE2 coding variants, respectively. Nevertheless, only seventeen coding variants were found to be important for ACE2 binding with the coronavirus spike protein (Table 1). The frequencies of these allele variants range from 3.88 × 10−3 to 5.47 × 10−6 for rs4646116 (K26R) and rs1238146879 (P426A), respectively. These results parallel recent published findings [28,35], in which the authors reported some rare and common ACE2 variants susceptible to SARS-CoV-2 infection. The variant rs4646116 (K26R) has been reported to be the most frequent in the Ashkenzai Jewish population [36]. These frequencies may explain the infection rate for this highly contagious virus but also the possible non-strong relationship between ACE2 variants and COVID-19 severity in different populations [36,37].

3.2. Molecular Binding and Interaction Results

In this study, a comparison of the different binding scores of CQ and HCQ with the different allelic variant of ACE2 is reported. Table 2 shows the predicted binding affinities of the stable ACE2 variant–CQ or –HCQ complexes, number of conventional H-bonds, and the number of the closest interacting residues. Both CQ and HCQ were found to exhibit negative binding energy, ranging from −6 to −3 kcal·mol−1, with the different ACE2 allelic variants. Accordingly, all complexes of ACE2 variants and CQ or HCQ displayed negative docking scores. Thus, the disruption of coronavirus entry via ACE2 is thermodynamically possible by using CQ or HCQ. Further analyses using molecular dynamic approaches would confirm our results. Both CQ and HCQ interact differently with the seventeen different targeted ACE2 domains, which had been reported to bind with coronavirus spike protein. It could be deduced that CQ and HCQ efficiency might be mediated by the ACE2 polymorphism, as their interactions depend on the latter. In this study, (S)-enantiomers particularly S-13a of both CQ and HCQ were used for the molecular docking assay. In fact, it has been previously reported that (S)-enantiomers are consistently showing better activity than corresponding (R)-enantiomers, especially the antimalarial effects of CQ and its analogues [38]. The best affinity was predicted for the variant 8 (rs961360700, D355N) by −6 and −5.9 kcal·mol−1 for HCQ and CQ, respectively. The radar distribution of CQ and HCQ binding affinities towards the allelic variants of ACE2 showed superposition only in four alleles which are rs762890235 (P389H), rs755691167 (K68E), rs1299103394 (K26E), and rs778500138 (E35D) (Figure 2). Recently, it has been reported that CQ and HCQ also interact differently with fifteen protein targets of SARS-CoV-2 using molecular docking and dynamics [39]. This can interfere with the inhibitory activity of ACE2, which has been previously reported [22]. In this study, we highlight ACE2 polymorphism as possible interference with CQ and HCQ.
However, none of these superposed points was associated with a similar number of conventional hydrogen bonds or ACE2 interacting residues. It could be deduced that the terminal hydroxyl group, which makes the difference between CQ and HCQ, is conditioning and playing a marked influence in the binding affinities, number of conventional hydrogen bonds, and the interacting residues. This could confirm previous data of Fantini et al. (2020) [40] who reported differential interactions of CQ and HCQ with sialic acids, which is also used by the S protein of SARS-CoV-2 as an entry receptor. Recently, it was reported that some ACE2 variants decreased and some others increased the electrostatic attraction towards SARS-CoV-2, such as ACE2-K26R and ACE2-R219C [36]. Likewise, this study outlined that ACE2 variants interact differently with CQ and HCQ.
Nevertheless, the number of conventional H-bonds and the number of the closest interacting residues were slightly better with HCQ. In fact, regarding conventional hydrogen bonds (H-bonds), which constitute the best bond and the most used category in drug design [41,42], the highest number (n = 5) was found in the ACE2 (variant 16: rs1396769231, M383T)–HCQ complex. That was followed by ACE2 (variant 12: rs1299103394, K26E)–CQ, ACE2 (variant 6: rs766996587, M82I)–CQ, and ACE2 (variant 8: rs961360700, D355N)–HCQ complexes with four H-bonds each (Table 3).
ACE facilitate the invasion of the virus and its depletion from the cell membrane enhance the damaging effects, which result in tissue deterioration, particularly, in the respiratory tract. Similarly, the genetic variation within ACE2 polymorphism might result in various effects of the virus on the targeted tissues. Likewise, CQ and HCQ might interact differently with ACE2 variants.
This could be correlated with the geographical distribution of ACE2 genotype which has been previously reported [43]. For its entry within the cell, SARS-CoV-2 uses both ACE2 and the ganglioside-attached sialic acids [5,40]. Further studies on the interactions of CQ and HCQ with ganglioside-attached sialic acids could give general ideas about the possible actions of these drugs on the virus entry.

3.3. Pharmacokinetics and ADME Findings of CQ and HCQ

Pharmacokinetics and the most pertinent absorption, distribution, metabolism, and excretion (ADME) parameters of both CQ and HCQ were also assessed based on their absorption, distribution, metabolism, and excretion data. Table 4 exhibits the properties, lipophylicity, druglikeness, and pharmacokinetics of CQ and HCQ.
Both drugs obey the Lipinski’s rule and show comparable and almost similar properties associated with good oral absorption and bioavailability score (0.55), low TPSA values, and acceptable consensus Log Po/w, which allow them to fall in the pink colored zone of the polygon suggesting good oral bioavailability (bottom of Table 4). Regarding pharmacokinetics results, both CQ and HCQ were predicted to have high gastrointestinal (GI) absorption and were blood–brain–barrier (BBB) permeant. Neither of the two drugs is a substrate of the p-glycoprotein. Cytochrome P450 (CYP) isoforms, such as CYP1A2, CYP2C19, CYP2C9, CYP2D6, and CYP3A4 are commonly used in biotransformation of drugs and xenobiotics [44]. In this study, the screened CYP enzyme isoforms data indicate that both CQ and HCQ were found to be inhibitors of only three isoenzymes, i.e., CYP1A2, CYP3A4, and CYP2D6 isoforms. Both CQ and HCQ displayed negative skin permeability, thus, both are not suitable compounds for transdermal delivery.
Taken together, the pharmacokinetic and ADME properties of CQ and HCQ complications such as heart failure and several non-reversible disorders have been previously, reported [45]. Our ADME and pharmacokinetic results confirmed previous data about pharmacokinetics and pharmacogenomics of CQ and HCQ [46]. The importance of the pharmacokinetic, ADME, and QSAR (for quantitative structure–activity relationship) assays was previously largely reported in the design and the assessment of several drugs [41,47,48].
The reported CQ and HCQ interactions with the different ACE2 variants known to bind with SARS-CoV-2 might certainly explain the variety of responses and new mechanistic effects of these drugs. In fact, new mechanisms of actions still need to be further explored [41]. The allelic variation of ACE2 may affect the recognition and infection by SARS-CoV-2, and therefore the susceptibility to its causing disease (COVID-19), and also to the treatment [49]. The current study proved that ACE polymorphism might mediate both the infection and the treatment of COVID-19. The promising effects of both CQ and HCQ that have been reported by treating the virus infection via blocking the binding of the virus with ACE2 confirms our interactions’ results [5,11]. Nevertheless, this effect might be mediated by ACE2 polymorphism. While CQ and HCQ clinically showed no potential beneficial effect on COVID-19, their interaction with ACE2 variants may certainly be used for the future design of new drugs or new diseases.

4. Conclusions

In conclusion, both CQ and HCQ interact differently with the different targeted ACE2 domains, which have been reported to bind with the coronavirus spike protein. It could be deduced that CQ and HCQ efficiency might be mediated by the ACE2 polymorphism. By extrapolation, the selection of CQ or HCQ for SARS-CoV-2 infected patients may be based on the ACE2 allelic variants to guarantee a more efficient drug. Further explorations of the relationship and the interactions between ACE2 polymorphism and CQ/HCQ would certainly help to better understand the COVID-19 management strategies and shorten the recovery period, particularly, in the absence of specific vaccines or drugs. This would certainly contribute to avoiding CQ and HCQ complications such as acute toxicity, heart failure, and several non-reversible disorders which have been previously reported [45,50]. The results of the current study provide new and strong evidence regarding COVID-19 susceptibility and treatment as a result of the ACE2 polymorphism.

Author Contributions

Conceptualization, R.B.; methodology, R.B.; software, R.B.; validation, R.B., M.A., M.M.A., and F.B.; investigation, R.B.; writing—review and editing, R.B.; project administration, R.B., F.B. and M.M.A.; funding acquisition, M.M.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Deanship of Scientific Research, the University of Ha’il, grant number COVID1942.

Institutional Review Board Statement

This research project has been approved by the local ethical committee, letter number 9472/5/42, dated on 5 October 2020.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data available in a publicly accessible repository that does not issue DOIs. Publicly available datasets were analyzed in this study.

Acknowledgments

The authors would like to acknowledge Jahoor Alam (assistant professor in bioinformatics, University of Ha’il) for his support given in the collection of PDB format of the 17 concerned proteins.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are not available from the authors.

References

  1. Jorge, A.; Ung, C.; Young, L.H.; Melles, R.B.; Choi, H.K. Hydroxychloroquine retinopathy—Implications of research advances for rheumatology care. Nat. Rev. Rheumatol. 2018, 14, 693–703. [Google Scholar] [CrossRef] [PubMed]
  2. Xu, R.; Ji, Z.; Xu, C.; Zhu, J. The clinical value of using chloroquine or hydroxychloroquine as autophagy inhibitors in the treatment of cancers: A systematic review and meta-analysis. Medicine 2018, 97, e12912. [Google Scholar] [CrossRef] [PubMed]
  3. Lammers, A.; Brohet, R.; Theunissen, R.; Koster, C.; Rood, R.; Verhagen, D.; Brinkman, K.; Hassing, R.; Dofferhoff, A.; El Moussaoui, R.; et al. Early hydroxychloroquine but not chloroquine use reduces ICU admission in COVID-19 patients. Int. J. Infect. Dis. 2020, 101, 283–289. [Google Scholar] [CrossRef] [PubMed]
  4. Kaur, K.; Jain, M.; Reddy, R.P.; Jain, R. Quinolines and structurally related heterocycles as antimalarials. Eur. J. Med. Chem. 2010, 45, 3245–3264. [Google Scholar] [CrossRef] [PubMed]
  5. Badraoui, R.; Alrashedi, M.M.; El-May, M.V.; Bardakci, F. Acute respiratory distress syndrome: A life threatening associated complication of SARS-CoV-2 infection inducing COVID-19. J. Biomol. Struct. Dyn. 2020, 1–11. [Google Scholar] [CrossRef] [PubMed]
  6. Shaw, J.J.; Boyer, T.L.; Venner, E.; Beck, P.J.; Slamowitz, T.; Caste, T.; Hickman, A.; Raymond, M.H.; Costa-Pinheiro, P.; Jameson, M.J.; et al. Inhibition of Lysosomal Function Mitigates Protective Mitophagy and Augments Ceramide Nanoliposome–Induced Cell Death in Head and Neck Squamous Cell Carcinoma. Mol. Cancer Ther. 2020, 19, 2621–2633. [Google Scholar] [CrossRef]
  7. Liang, S.; Li, X.; Gao, C.; Zhang, L. microRNA-based autophagy inhibition as targeted therapy in pancreatic cancer. Biomed. Pharmacother. 2020, 132, 110799. [Google Scholar] [CrossRef]
  8. Pimentel, J.; Andersson, N. Chloroquine and its derivatives in the management of COVID-19: A scoping review. Biomedica 2020, 40, 80–95. [Google Scholar] [CrossRef]
  9. Ziegler, H.K.; Unanue, E.R. Decrease in macrophage antigen catabolism caused by ammonia and chloroquine is associated with inhibition of antigen presentation to T cells. Proc. Natl. Acad. Sci. USA 1982, 79, 175–178. [Google Scholar] [CrossRef] [Green Version]
  10. Gies, V.; Bekaddour, N.; Dieudonné, Y.; Guffroy, A.; Frenger, Q.; Gros, F.; Rodero, M.P.; Herbeuval, J.-P.; Korganow, A.-S. Beyond anti-viral effects of chloroquine/hydroxychloroquine. Front. Immunol. 2020, 11, 1409. [Google Scholar] [CrossRef]
  11. Wang, N.; Han, S.; Liu, R.; Meng, L.; He, H.; Zhang, Y.; Wang, C.; Lv, Y.; Wang, J.; Li, X.; et al. Chloroquine and hydroxychloroquine as ACE2 blockers to inhibit viropexis of 2019-nCoV Spike pseudotyped virus. Phytomedicine 2020, 79, 153333. [Google Scholar] [CrossRef] [PubMed]
  12. Xu, X.; Chen, P.; Wang, J.; Feng, J.; Zhou, H.; Li, X.; Zhong, W.; Hao, P. Evolution of the novel coronavirus from the ongoing Wuhan outbreak and modeling of its spike protein for risk of human transmission. Sci. China Life Sci. 2020, 63, 457–460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Yan, R.; Zhang, Y.; Li, Y.; Xia, L.; Guo, Y.; Zhou, Q. Structural basis for the recognition of the SARS-CoV-2 by full-length human ACE2. Science 2020, 367, 1444–1448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Zhao, Y.; Zhao, Z.; Wang, Y.; Zhou, Y.; Ma, Y.; Zuo, W. Single-cell RNA expression profiling of ACE2, the putative receptor of Wuhan 2019-nCov. bioRxiv 2020. [Google Scholar] [CrossRef]
  15. Zou, X.; Chen, K.; Zou, J.; Han, P.; Hao, J.; Han, Z. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front. Med. 2020, 14, 185–192. [Google Scholar] [CrossRef] [Green Version]
  16. Donoghue, M.; Hsieh, F.; Baronas, E.; Godbout, K.; Gosselin, M.; Stagliano, S.; Donovan, M.; Woolf, B.; Robison, K.; Jeyaseelan, R.; et al. A novel angiotensin-converting enzyme-related carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1–9. Circ. Res. 2000, 87, E1–E9. [Google Scholar] [CrossRef]
  17. Delanghe, J.R.; Speeckaert, M.M.; De Buyzere, M.L. COVID-19 infections are also affected by human ACE1 D/I polymorphism. Clin. Chem. Lab. Med. 2020, 58, 1125–1126. [Google Scholar] [CrossRef] [Green Version]
  18. Delanghe, J.R.; Speeckaert, M.M.; De Buyzere, M.L. The host’s angiotensin-converting enzyme polymorphism may explain epidemiological findings in COVID-19 infections. Clin. Chim. Acta 2020, 505, 192–193. [Google Scholar] [CrossRef]
  19. Knoll, M.D.; Wonodi, C. Oxford-AstraZeneca COVID-19 vaccine efficacy. Lancet 2021, 397, 72–74. [Google Scholar] [CrossRef]
  20. Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J.L.; Pérez Marc, G.; Moreira, E.D.; Zerbini, C.; et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N. Engl. J. Med. 2020, 383, 2603–2615. [Google Scholar] [CrossRef]
  21. Zhu, H.; Lu, H. The development of a quarantine strategy is an important path to a normalized response to COVID-19. Biosci. Trends 2020, 14, 396–398. [Google Scholar] [CrossRef] [PubMed]
  22. Abdelli, I.; Hassani, F.; Brikci, S.B.; Ghalem, S. In silico study the inhibition of angiotensin converting enzyme 2 receptor of COVID-19 by Ammoides verticillata components harvested from Western Algeria. J. Biomol. Struct. Dyn. 2020, 1–14. [Google Scholar] [CrossRef] [PubMed]
  23. Siddiqui, A.J.; Danciu, C.; Ashraf, S.A.; Moin, A.; Singh, R.; Alreshidi, M.; Patel, M.; Jahan, S.; Kumar, S.; Alkhinjar, M.I.M.; et al. Plants-derived biomolecules as potent antiviral phytomedicines: New insights on ethnobotanical evidences against coronaviruses. Plants 2020, 9, 1244. [Google Scholar] [CrossRef] [PubMed]
  24. Hunt, S.E.; McLaren, W.; Gil, L.; Thormann, A.; Schuilenburg, H.; Sheppard, D.; Parton, A.; Armean, I.M.; Trevanion, S.J.; Flicek, P.; et al. Ensembl variation resources. Database 2018, 2018, 1–12. [Google Scholar] [CrossRef] [PubMed]
  25. Newman, V.; Moore, B.; Sparrow, H.; Perry, E. The Ensembl Genome Browser: Strategies for Accessing Eukaryotic Genome Data. Methods Mol. Biol. 2018, 1757, 115–139. [Google Scholar] [CrossRef] [Green Version]
  26. Lek, M.; Karczewski, K.J.; Exome Aggregation Consortium; Minikel, E.V.; Samocha, K.E.; Banks, E.; Fennell, T.; O’Donnell-Luria, A.H.; Ware, J.S.; Hill, A.J.; et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 2016, 536, 285–291. [Google Scholar] [CrossRef] [Green Version]
  27. Li, F.; Li, W.; Farzan, M.; Harrison, S.C. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science 2005, 309, 1864–1868. [Google Scholar] [CrossRef]
  28. Hussain, M.; Jabeen, N.; Raza, F.; Shabbir, S.; Baig, A.A.; Amanullah, A.; Aziz, B. Structural variations in human ACE2 may influence its binding with SARS-CoV-2 spike protein. J. Med. Virol. 2020, 92, 1580–1586. [Google Scholar] [CrossRef] [Green Version]
  29. Goodsell, D.S.; Zardecki, C.; Di Costanzo, L.; Duarte, J.M.; Hudson, B.P.; Persikova, I.; Segura, J.; Shao, C.; Voigt, M.; Westbrook, J.D.; et al. RCSB Protein Data Bank: Enabling biomedical research and drug discovery. Protein Sci. 2020, 29, 52–65. [Google Scholar] [CrossRef] [Green Version]
  30. Trott, O.; Olson, A.J. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar] [CrossRef] [Green Version]
  31. Yates, A.D.; Achuthan, P.; Akanni, W.; Allen, J.; Allen, J.; Alvarez-Jarreta, J.; Amode, M.R.; Armean, I.M.; Azov, A.G.; Bennett, R.; et al. Ensembl 2020. Nucleic Acids Res. 2020, 48, D682–D688. [Google Scholar] [CrossRef] [PubMed]
  32. Paladugu, S.; Donato, A.A. Remdesivir improved time to recovery in adults hospitalized with COVID-19 and lower respiratory tract involvement. Ann. Intern. Med. 2020, 173, JC4. [Google Scholar] [CrossRef] [PubMed]
  33. Li, W.; Zhang, C.; Sui, J.; Kuhn, J.H.; Moore, M.J.; Luo, S.; Wong, S.-K.; Huang, I.-C.; Xu, K.; Vasilieva, N.; et al. Receptor and viral determinants of SARScoronavirus adaptation to human ACE2. EMBO J. 2005, 24, 1634–1643. [Google Scholar] [CrossRef] [Green Version]
  34. Mudgal, R.; Nehul, S.; Tomar, S. Prospects for mucosal vaccine: Shutting the door on SARS-CoV-2. Hum. Vaccin. Immunother. 2020, 16, 2921–2931. [Google Scholar] [CrossRef] [PubMed]
  35. Shikov, A.E.; Barbitoff, Y.A.; Glotov, A.S.; Danilova, M.M.; Tonyan, Z.N.; Nasykhova, Y.A.; Mikhailova, A.A.; Bespalova, O.N.; Kalinin, R.S.; Mirzorustamova, A.M.; et al. Analysis of the Spectrum of ACE2 Variation Suggests a Possible Influence of Rare and Common Variants on Susceptibility to COVID-19 and Severity of Outcome. Front. Genet. 2020, 29, 551220. [Google Scholar] [CrossRef]
  36. Ali, F.; Elserafy, M.; Alkordi, M.H.; Amin, M. ACE2 coding variants in different populations and their potential impact on SARS-CoV-2 binding affinity. Biochem. Biophys. Rep. 2020, 24, 100798. [Google Scholar] [CrossRef]
  37. Novelli, A.; Biancolella, M.; Borgiani, P.; Cocciadiferro, D.; Colona, V.L.; D’Apice, M.R.; Rogliani, P.; Zaffina, S.; Leonardis, F.; Campana, A.; et al. Analysis of ACE2 genetic variants in 131 Italian SARS-CoV-2-positive patients. Hum. Genom. 2020, 14, 29. [Google Scholar] [CrossRef]
  38. Sinha, M.; Dola, V.R.; Soni, A.; Agarwal, P.; Srivastava, K.; Haq, W.; Puri, S.K.; Katti, S.B. Synthesis of chiral chloroquine and its analogues as antimalarial agents. Bioorganic Med. Chem. 2014, 22, 5950–5960. [Google Scholar] [CrossRef]
  39. Skariyachan, S.; Gopal, D.; Chakrabarti, S.; Kempanna, P.; Uttarkar, A.; Muddebihalkar, A.G.; Niranjan, V. Structural and molecular basis of the interaction mechanism of selected drugs towards multiple targets of SARS-CoV-2 by molecular docking and dynamic simulation studies- deciphering the scope of repurposed drugs. Comput. Biol. Med. 2020, 126, 104054. [Google Scholar] [CrossRef]
  40. Fantini, J.; Di Scala, C.; Chahinian, H.; Yahi, N. Structural and molecular modelling studies reveal a new mechanism of action of chloroquine and hydroxychloroquine against SARS-CoV-2 infection. Int. J. Antimicrob. Agent 2020, 55, 105960. [Google Scholar] [CrossRef]
  41. Akacha, A.; Badraoui, R.; Rebai, T.; Zourgui, L. Effect of Opuntia ficus indica extract on methotrexate-induced testicular injury: A biochemical, docking and histological study. J. Biomol. Struct. Dyn. 2020, 1856187. [Google Scholar] [CrossRef]
  42. Hwang, D.; Ramsey, J.D.; Makita, N.; Sachse, C.; Jordan, R.; Sokolsky-Papkov, M.; Kabanov, A.V. Novel poly(2-oxazoline) block copolymer with aromatic heterocyclic side chains as a drug delivery platform. J. Control. Release 2019, 307, 261–271. [Google Scholar] [CrossRef] [PubMed]
  43. Saab, Y.B.; Gard, P.; Overall, A. The geographic distribution of the ACE II genotype: A novel finding. Genet. Res. 2007, 89, 259–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Badraoui, R.; Rebai, T.; Elkahoui, S.; Alreshidi, M.; Veettil, V.N.; Noumi, E.; Al-Motair, K.A.; Aouadi, K.; Kadri, A.; De Feo, V.; et al. Allium subhirsutum L. as a Potential Source of Antioxidant and Anticancer Bioactive Molecules: HR-LCMS Phytochemical Profiling, In Vitro and In Vivo Pharmacological Study. Antioxidants 2020, 9, 1003. [Google Scholar] [CrossRef]
  45. Chatre, C.; Roubille, F.; Vernhet, H.; Jorgensen, C.; Pers, Y.M. Cardiac complications attributed to chloroquine and hydroxychloroquine: A systematic review of the literature. Drug Saf. 2018, 41, 919–931. [Google Scholar] [CrossRef]
  46. Babayeva, M.; Loewy, Z. Repurposing Drugs for COVID-19: Pharmacokinetics and Pharmacogenomics of Chloroquine and Hydroxychloroquine. Pharmgenomics Pers. Med. 2020, 13, 531–542. [Google Scholar] [CrossRef]
  47. Curreli, F.; Victor, S.M.B.; Ahmed, S.; Drelich, A.; Tong, X.; Tseng, C.-T.K.; Hillyer, C.D.; Debnath, A.K. Stapled Peptides Based on Human Angiotensin-Converting Enzyme 2 (ACE2) Potently Inhibit SARS-CoV-2 Infection In Vitro. mBio 2020, 11. [Google Scholar] [CrossRef]
  48. Flores-Holguín, N.; Frau, J.; Glossman-Mitnik, D. Conceptual DFT-Based Computational Peptidology of Marine Natural Compounds: Discodermins A–H. Molecules 2020, 25, 4158. [Google Scholar] [CrossRef]
  49. Guo, X.; Chen, Z.; Xia, Y.; Lin, W.; Li, H. Investigation of the genetic variation in ACE2 on the structural recognition by the novel coronavirus (SARS-CoV-2). J. Transl. Med. 2020, 18, 321. [Google Scholar] [CrossRef]
  50. Della Porta, A.; Bornstein, K.; Coye, A.; Montrief, T.; Long, B.; Parris, M.A. Acute chloroquine and hydroxychloroquine toxicity: A review for emergency clinicians. Am. J. Emerg. Med. 2020, 38, 2209–2217. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of chloroquine (CQ, R=H) and hydroxychloroquine (HCQ, R=OH).
Figure 1. Chemical structure of chloroquine (CQ, R=H) and hydroxychloroquine (HCQ, R=OH).
Molecules 26 00673 g001
Figure 2. Radar distribution of chloroquine (CQ, blue color) and hydroxychloroquine (HQC, red color) binding energy towards the different variants of human ACE2. Note the superposition only in 4 allelic variants, i.e., rs143936283 (E329G), rs755691167 (K68E), rs1299103394 (K26E), and rs778500138 (E35D).
Figure 2. Radar distribution of chloroquine (CQ, blue color) and hydroxychloroquine (HQC, red color) binding energy towards the different variants of human ACE2. Note the superposition only in 4 allelic variants, i.e., rs143936283 (E329G), rs755691167 (K68E), rs1299103394 (K26E), and rs778500138 (E35D).
Molecules 26 00673 g002
Table 1. Genetic variants change amino acids and allele frequencies of human angiotensin-converting enzyme 2 (ACE2) reported to bind with coronavirus.
Table 1. Genetic variants change amino acids and allele frequencies of human angiotensin-converting enzyme 2 (ACE2) reported to bind with coronavirus.
Variant No.Genetic VariantAmino Acid
Change
Allele
Frequency
1rs4646116K26R3.88 × 10−3
2rs73635825S19P3.13 × 10−4
3rs146676783E37K3.9 × 10−5
4rs762890235P389H3.83 × 10−5
5rs143936283E329G3.44 × 10−5
6rs766996587M82I2.44 × 10−5
7rs1348114695E35K1.64 × 10−5
8rs961360700D355N1.17 × 10−5
9rs755691167K68E1.09 × 10−5
10rs1316056737D427Y1.09 × 10−5
11rs781255386T27A1.09 × 10−5
12rs1299103394K26E5.45 × 10−6
13rs759134032P84T5.47 × 10−6
14rs1238146879P426A5.47 × 10−6
15rs778500138E35DN/A
16rs1396769231M383TN/A
17rs1016777825R559SN/A
Table 2. Ligand receptor interactions between chloroquine or hydroxychloroquine and the different variants of human ACE2.
Table 2. Ligand receptor interactions between chloroquine or hydroxychloroquine and the different variants of human ACE2.
No.Genetic VariantChloroquine (CQ)Hydroxychloroquine (HCQ)
Affinity (Kcal/Mol)Conventional H-BondsNumber of Closest
Interacting Residues
Affinity (Kcal/mol)Conventional H-BondsNumber of Closest Interacting
Residues
1rs4646116−3.824−4.124
2rs73635825−3.533−3.633
3rs146676783−4.524−4.324
4rs762890235−4.217−4.333
5rs143936283−4.016−4.025
6rs766996587−3.843−3.733
7rs1348114695−4.015−4.125
8rs961360700−5.926−6.047
9rs755691167−4.724−4.726
10rs1316056737−4.023−3.737
11rs781255386−3.014−3.333
12rs1299103394−3.842−3.844
13rs759134032−4.326−4.225
14rs1238146879−3.825−4.136
15rs778500138−4.424−4.432
16rs1396769231−3.336−4.056
17rs1016777825−3.923−4.024
Table 3. The best interacting complexes of ACE2 variants’ active site residues and CQ or HCQ.
Table 3. The best interacting complexes of ACE2 variants’ active site residues and CQ or HCQ.
Receptor-Ligand Interactions, Distance in Angstroms
and 2D Interactions Diagrams
Receptor-Ligand 3D interaction Microphotographs
Chloroquine (CQ)rs1299103394 (K26E)—CQ
(CYS16)---(CQ) Conventional hydrogen bond: 3.702 Å;
(ILE27)---(CQ) Conventional hydrogen bond: 2.269 Å;
(SER10)---(CQ) Conventional hydrogen bond: 2.849 Å;
(PHE15)---(CQ) Conventional hydrogen bond: 3.572 Å;
(ALA18)---(CQ) Alkyl interaction: 4.047 Å;
(CYS16)---(CQ) Alkyl interaction: 4.802 Å;
(TRP37)---(CQ) Pi-Alkyl interaction: 5.195 Å.
Molecules 26 00673 i001
Molecules 26 00673 i002
rs766996587 (M82I)—CQ
(TYR33)---(CQ) Conventional hydrogen bond: 2.227 Å;
(GLN51)---(CQ) Conventional hydrogen bond: 1.983 Å;
(TYR33)---(CQ) Conventional hydrogen bond: 3.385 Å;
(TYR33)---(CQ) Conventional hydrogen bond: 3.361 Å;
(PRO34)---(CQ) Pi-Sigma interaction: 3.437 Å;
(TYR33,PRO34)---(CQ) Amide-Pi Stacked interaction: 4.757 Å;
(PRO34)---(CQ) Pi-Alkyl interaction: 5.295 Å.
Molecules 26 00673 i003
Molecules 26 00673 i004
Hydroxychloroquine (HCQ)rs1396769231 (M383T)—HCQ
(GLY28)---(HCQ) Conventional hydrogen bond: 2.900 Å;
(SER48)---(HCQ) Conventional hydrogen bond: 2.271 Å;
(SER48)---(HCQ) Conventional hydrogen bond: 3.177 Å;
(GLY28)---(HCQ) Conventional hydrogen bond: 3.296 Å;
(SER46)---(HCQ) Conventional hydrogen bond: 3.399 Å;
(PRO53)---(HCQ) Alkyl interaction: 5.246 Å;
(LYS27)---(HCQ) Alkyl interaction: 4.494 Å;
(ARG49)---(HCQ) Pi-Alkyl interaction: 5.201 Å;
(PRO53)---(HCQ) Pi-Alkyl interaction: 5.246 Å;
(PRO53)---(HCQ) Pi-Alkyl interaction: 3.918 Å.
Molecules 26 00673 i005
Molecules 26 00673 i006
rs961360700 (D355N)—HCQ
(THR26)---(HCQ) Conventional hydrogen bond: 2.344 Å;
(THR26)---(HCQ) Conventional hydrogen bond: 2.537 Å;
(HIS24)---(HCQ) Conventional hydrogen bond: 3.377 Å;
(THR26)---(HCQ) Conventional hydrogen bond: 3.270 Å;
(MET11)---(HCQ) Carbon hydrogen bond: 3.494 Å;
(VAL22)---(HCQ) Carbon hydrogen bond: 3.455 Å;
(TRP28)---(HCQ) Pi-Hydrogen bond: 3.018 Å;
(ALA21)---(HCQ) Alkyl interaction: 3.885 Å;
(MET11)---(HCQ) Alkyl interaction: 4.566 Å;
(LEU30)---(HCQ) Pi-Alkyl interaction: 5.174 Å.
Molecules 26 00673 i007
Molecules 26 00673 i008
Table 4. Physicochemical properties, lipophilicity, drug-likeness, and pharmacokinetics of chloroquine (CQ) and hydroxychloroquine (HCQ) based on their absorption, distribution, metabolism, and excretion (ADME) characteristics.
Table 4. Physicochemical properties, lipophilicity, drug-likeness, and pharmacokinetics of chloroquine (CQ) and hydroxychloroquine (HCQ) based on their absorption, distribution, metabolism, and excretion (ADME) characteristics.
EntryChloroquine (CQ)Hydroxychloroquine (HCQ)
Physicochemical Properties, Lipophilicity and Drug-Likeness
Molecular weight (g/mol)319.87335.87
No. heavy atoms2223
No. arom. heavy atoms1010
Fraction Csp30.500.50
No. rotatable bonds89
No. H-bond acceptors23
No. H-bond donors12
Molar Refractivity97.4198.57
TPSA (Å2)28.1648.39
Consensus Log Po/w4.153.29
Lipinski’s RuleYesYes
Bioavailability Score0.550.55
PAINS0 alert0 alert
Pharmacokinetics
Gastrointestinal absorptionHighHigh
BBB permeantYesYes
P-gp substrateNoNo
CYP1A2 inhibitorYesYes
CYP2C19 inhibitorNoNo
CYP2C9 inhibitorNoNo
CYP2D6 inhibitorYesYes
CYP3A4 inhibitorYesYes
Log Kp (cm/s)−4.96−5.81
Molecules 26 00673 i009 Molecules 26 00673 i010
ADME, absorption, distribution, metabolism and excretion; LIPO (lipophilicity), −0.7 < XLOGP3 < +5.0; SIZE, 150 g/mol < MV < 500 g/mol; POLAR (polarity), 20 Å2 <TPSA < 130 Å2; INSOLU (insolubility), 0 < Log S (ESOL) < 6; INSATU (insaturation), 0.25 < fraction Csp3 < 1; FLEX (flexibility), 0 < number of rotatable bonds < 9; TPSA, topological polar surface area; CYP, cytochrome P450. The colored zone is the suitable physicochemical space for oral bioavailability.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Badraoui, R.; Adnan, M.; Bardakci, F.; Alreshidi, M.M. Chloroquine and Hydroxychloroquine Interact Differently with ACE2 Domains Reported to Bind with the Coronavirus Spike Protein: Mediation by ACE2 Polymorphism. Molecules 2021, 26, 673. https://doi.org/10.3390/molecules26030673

AMA Style

Badraoui R, Adnan M, Bardakci F, Alreshidi MM. Chloroquine and Hydroxychloroquine Interact Differently with ACE2 Domains Reported to Bind with the Coronavirus Spike Protein: Mediation by ACE2 Polymorphism. Molecules. 2021; 26(3):673. https://doi.org/10.3390/molecules26030673

Chicago/Turabian Style

Badraoui, Riadh, Mohd Adnan, Fevzi Bardakci, and Mousa M. Alreshidi. 2021. "Chloroquine and Hydroxychloroquine Interact Differently with ACE2 Domains Reported to Bind with the Coronavirus Spike Protein: Mediation by ACE2 Polymorphism" Molecules 26, no. 3: 673. https://doi.org/10.3390/molecules26030673

Article Metrics

Back to TopTop