Next Article in Journal
TCGA PanCanAtlas Data Analysis Suggests Multiple Possibilities for Personalized Cancer Therapy
Previous Article in Journal
Variability of Selected Traits in Meadow Fescue (Festuca pratensis Huds.) Plants with Different Susceptibility to Seed Shattering
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Proceeding Paper

The Study of Natural Compounds as Antidepressants by Bioinformatics Methods †

1
Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independentei, No 91-95, 050095 Bucharest, Romania
2
Earth, Environmental and Life Sciences Section, Research Institute of the University of Bucharest, University of Bucharest, 1 B. P. Hașdeu St., 50567 Bucharest, Romania, Romania
3
Laser Department, National Institute for Laser, Plasma and Radiation Physics, 077125 Magurele, Romania
4
Department of Automatic Control and Systems Engineering, Politehnica University of Bucharest, 313 Splaiul Independenţei, 060042 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Presented at the 1st International Electronic Conference on Biomedicine, 1–26 June 2021; Available online: https://ecb2021.sciforum.net/.
Biol. Life Sci. Forum 2021, 7(1), 10; https://doi.org/10.3390/ECB2021-10268
Published: 31 May 2021
(This article belongs to the Proceedings of The 1st International Electronic Conference on Biomedicine)

Abstract

:
According to the World Health Organization, neuropsychiatric disorders pose an increasingly greater burden on health, society, and economy of countries. Currently, there are strategies to prevent some of these disorders, and there are treatments or means to alleviate their symptoms. In the case of depression, the medication consists of antidepressant drugs. Such drugs can also be used in other conditions like anxiety, pain, or insomnia. A shortcoming of the currently used antidepressants is the occurrence of side effects that sometimes are unbearable for the patient. In this respect, a promising direction is the usage of medicinal plants. Plant parts are rich in phytochemicals that could be beneficial in mental disorders by acting on various targets. Here, we investigated the antidepressant effect of ten natural compounds from sage, mint, and citrus. The biological activity of these compounds was investigated by calculating pKi values and affinities for dopamine receptor D2, serotonin transporter (SERT), and 5-hydroxytryptamine receptor 1A (5-HT1A) using quantitative structure-activity relationship (QSAR) models. Our results showed that linalyl acetate, 1,8-cineole, and neryl acetate could be efficient antidepressants and neuroleptics.

1. Introduction

According to the World Health Organization (WHO), depression is a prevalent mental disorder that seriously affects the function of patients inside their family, at their workplace, or school [1]. Patients with mild to severe depression are offered treatment as antidepressant drugs and psychotherapy [2,3]. The drugs prescribed in depression fall into three categories: monoamine oxidase inhibitors, tricyclic antidepressants, and second-generation antidepressants, like norepinephrine, serotonin, or serotonin–norepinephrine reuptake inhibitors [4,5]. These present a series of adverse side effects like anxiety, tachycardia, tremor, sedation, blurred vision, etc. [4,6]. The intensity of these secondary effects is sometimes unbearable for patients that become intolerant to the treatment [7]. In this context, natural products might represent an alternative to conventional drugs for the treatment of depression.
Natural compounds may be a viable alternative in the treatment of viral and bacterial infections, as well as adjuvants and treatments for a variety of diseases [8,9,10]. Up to date, the beneficial effect of plant compounds in depression has been extensively reviewed. From these, we can mention Chinese herbal medicines [11], Ayurvedic single or holistic approaches [12], and many other phytochemicals or medicinal herbs [4,13]. The usage of natural compounds for therapy imposes some challenges concerning their pharmacokinetic and pharmacodynamic properties [14], meaning that a thorough characterization of these compounds is required. Cheminformatics methods can be of great assistance, allowing the description of the compounds and prediction of their biological effect [15,16].
Using the experience from our previous studies in predicting the anti-Alzheimer effect of natural compounds from Mentha spicata essential oil [14] or in predicting the antidepressant effect and the targets of candidate compounds [17,18], here, we used cheminformatics methods to investigate the antidepressant potential of ten natural compounds from diverse sources: 1,8-cineole (eucalyptus, sage), limonene (peppermint, spearmint), sabinene (lemon, mint), resveratrol (skin of grapes), chamazulene (german chamomile, roman chamomile), germacrene D (peppermint), linalyl acetate (sage), nerol (common grapes), neryl acetate (lemon balm, peppermint), and quercetin (grape). These compounds were mentioned in the literature as having antidepressant or neuroprotective effects [19]. We calculated the pharmacokinetic profiles of compounds. The prediction of their antidepressant effect was performed by considering three drug targets in depression, namely the serotonin transporter (SERT), dopamine receptor 2 (D2), and 5-hydroxytryptamine receptor 1A (5-HT1A). In the case of each target, we built a quantitative structure-activity relationship (QSAR) model that was further used to assess the effect of considered natural compounds [20].

2. Materials and Methods

2.1. Ligand Selection and Assessment of Their Drug and Lead-Likeness Features

We selected ten natural compounds from various vegetable sources, namely 1,8-cineole, limonene, sabinene, resveratrol, chamazulene, germacrene D, linalyl acetate, nerol, neryl acetate, and quercetin based on previous studies mentioning their possible beneficial effects in neuropsychiatric treatments. Calculations were performed using the Simplified Molecular Input Line Entry (SMILES) files of selected compounds that were fetched from the PubChem database [21]. The three-dimensional structures of compounds were modeled using Molecular Operating Environment (MOE) software (https://www.chemcomp.com/Products.htm, accessed on 30 May 2021) by applying the MMFF94x force field, Gasteiger-type charges, and a 0.005 gradient. The drug-likeness features of selected compounds were evaluated using Lipinski [22], Veber [23], Ghose [24], and Egan [25] filters implemented in the SwissADME web tool [26].

2.2. Computational Pharmacokinetics Profiles of Natural Compounds

The absorption, distribution, metabolism, excretion (ADME), and toxicity profiles of compounds were determined using the pkCSM database [27]. From all the properties calculated, we focused on the intestinal absorption (human, %), blood-brain barrier (BBB) permeability (log BBB), central nervous system permeability (CNS), fraction un-bound (human) and feature of renal organic cation transporter 2 (OCT2), AMES toxicity, hepatotoxicity, LD50 (median lethal dose) and maximum tolerated dose (human).

2.3. Predicted Biological Activities of Natural Compounds on SERT, 5-HT1A and D2 by 3D-ALMOND-QSAR

We used a non-aligned 3D-QSAR method and the 3D-QSAR-ALMOND method [17] to predict the biological activities of natural compounds at SERT, 5-HT1A, and D2 receptors. The activities were evaluated using Pentacle software (http://www.moldiscovery.com/software/pentacle/, accessed on 30 May 2021) and were expressed as pKi values (log 1/Ki, Ki represents the inhibition constant). We built three QSAR models as QSAR-SERT, QSAR-5-HT1A, QSAR-D2 with good statistical parameters r2 fitted correlation coefficient (greater than 0.8) and q2 cross-validated correlation coefficient (greater than 0.6). In building the models, we considered a synthetic antidepressant and neuroleptic molecules in the training and validation sets. PDSP Ki Database—Psychoactive Drug Screening Program [28] was used to retrieve the experimentally determined Ki values of molecules from training and validation sets. The test sets considered in the case of all three QSAR models included the selected natural compounds, namely 1,8-cineole, limonene, sabinene, resveratrol, chamazulene, germacrene D, linalyl acetate, nerol, neryl acetate, and quercetin.

3. Results

3.1. Drug-Likeness Features and Pharmacokinetics Profiles of Compounds

The application of Lipinski, Ghose, Veber, and Egan rules showed that all considered natural compounds present drug-like features. Predicted absorption, distribution, elimination, and toxicity features of compounds show that: (i) all compounds present a good intestinal absorption, (ii) all compounds except for quercetin present a good BBB permeability, (iii) the compounds except sabiene and chamazulene present a good CNS permeability, (iv) all compounds recorded a low human fraction unbound, (v) the compounds are not substrates of renal OCT2, and (vi) the compounds do not present toxicity features like hepatotoxicity, cardiotoxicity, or AMES toxicity.

3.2. Predicted Biological Activities of Natural Compounds on SERT, 5-HT1A and D2

Three 3D-QSAR-ALMOND models were built by considering the activity of molecules on each target (QSAR-SERT, QSAR-5HT-1A, QSAR-D2). Good statistical parameters were obtained when the QSAR models simultaneously considered the contribution of several descriptors like hydrophobicity, electrostatic, hydrogen bond donor/acceptor in various combinations. The QSAR models were used to predict the biological activities of selected natural compounds at SERT, 5-HT1A, and D2.
The biological activity of compounds at SERT was predicted using the QSAR-SERT model. We observed that seven natural compounds (limonene, sabiene, chamazulene, germacrene D, linalyl acetate, nerol, and neryl acetate) have a strong antidepressant character.
When predicting the biological activity of natural compounds at 5-HT1A using the QSAR-5-HT1A model, we observed that the natural compounds have a medium antidepressant activity.
In order to determine the neuroleptic activity of compounds, we applied the QSAR-D2 model. Results show that natural compounds like 1,8-cineole, linalyl acetate, neryl acetate, and quercetin have high biological activity on this target.

4. Discussions

4.1. The Most Active Natural Compounds on SERT, 5-HT1A and D2 Active Sites

The biological activities of the natural compounds considered here were computed as pKi, where a high pKi value indicates a high biological activity [29]. Prediction results were compared with the most active drug from the training series used to build the QSAR model. In the QSAR-SERT model, the most active natural compounds are linalyl acetate, chamazulene, neryl acetate, nerol, and germacrene D, as revealed by the comparison with the antidepressant drug paroxetine. The most active compounds in the QSAR-5-HT1A model are 1,8-cineole and linalyl acetate, as determined by comparison with ziprasidone, the most active compound in the QSAR-5-HT1A training set. Relative to spiperone, the most active compound in the QSAR-D2 training set, the most active natural compounds appear to be quercetin, neryl acetate, linalyl acetate, and 1,8-cineole. These results point toward linalyl acetate, 1,8-cineole, and neryl acetate as promising antidepressant and neuroleptic molecules. In Figure 1, we present their three-dimensional structures relative to the spatial structures of the drugs used as a reference, namely paroxetine, ziprasidone, and spiperone.

4.2. The Drug-Likeness Features and Pharmacokinetics Profiles of the Most Active Compounds

Drug-like filter validation is correlated with possible drug actions and good bioavailability. The most active natural compounds comply with all drug-likeness rules. Pharmacokinetic investigations reveal that the most active natural compounds are well tolerated by the human body. In Table 1, we present the evaluation of hepatotoxicity, cardiotoxicity, and AMES toxicity in the case of the most active natural compounds—neryl acetate, 1,8-cineole, and linalyl acetate. We also present the same parameters calculated in the case of the most active drugs from QSAR models training sets, namely paroxetine, spiperone, and ziprasidone.
According to our predictions, the natural compounds are not hepatotoxic, AMES toxic, or hERG I and II inhibitors. In contrast, regularly used medications—spiperone, paroxetine, and ziprasidone, all have hepatotoxicity and are hERG II inhibitors. Moreover, paroxetine is AMES hazardous (Table 1).

5. Conclusions

Here, we investigated the potential of ten natural compounds (1,8-cineole, limonene, sabinene, resveratrol, chamazulene, germacrene D, linalyl acetate, nerol, neryl acetate, and quercetin) to exhibit antidepressant effects by acting on SERT and 5-HT1A receptors or neuroleptic activity acting on D2 receptors. This was performed using three QSAR models developed for predicting the activity of compounds at each target.
Before building the models, the compounds were filtered by drug and lead-likeness rules showing that all compounds comply with the rules, presenting drug-likeness and good bioavailability features. Moreover, the compounds were filtered based on their predicted ADME and toxicity profiles and all compounds resulted in having good BBB and CNS permeability while being non-toxic on the liver or heart and not mutagenic.
By applying the QSAR models that we built, we noticed that seven out of ten compounds should have good biological activity on SERT, two compounds should modulate 5-HT1A receptors, and four compounds should modulate D2 receptors. From these, linalyl acetate appears the only compound modulating all three protein targets, 1,8-cineole should modulate 5-HT1A and D2 receptors, and neryl acetate should modulate SERT and D2 receptors.

Author Contributions

Conceptualization, S.A.; methodology, S.A.; software, S.A. and C.B.; investigation, S.A. and A.M.U.; formal analysis, C.B.; writing—original draft preparation, A.M.U. and S.A.; writing—review and editing, M.M. and M.S.S. All authors have read and agreed to the published version of the manuscript.

Funding

The study was supported by UEFISCDI through the projects PN-III-P2-2.1-PED2019-1471 “New biocompatible shagaol and curcuminoid-like products used as adjuvantes in cancer radiotherapy”, Nucleu Programme, ctr. No. 16N/08.02.2019; and PN-III-P1-1.2-PCCDI-2017-0728 “Integrated project for the development of technologies dedicated to advanced medical treatments”.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the des (ign of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Depression. Available online: https://www.who.int/news-room/fact-sheets/detail/depression (accessed on 16 April 2021).
  2. Avram, S.; Milac, A.-L.; Mihailescu, D. 3D-QSAR Study Indicates an Enhancing Effect of Membrane Ions on Psychiatric Drugs Targeting Serotonin Receptor 5-HT1A. Mol. Biosyst. 2012, 8, 1418. [Google Scholar] [CrossRef]
  3. Yuan, Z.; Chen, Z.; Xue, M.; Zhang, J.; Leng, L. Application of Antidepressants in Depression: A Systematic Review and Meta-Analysis. J. Clin. Neurosci. 2020, 80, 169–181. [Google Scholar] [CrossRef] [PubMed]
  4. Lee, G.; Bae, H. Therapeutic Effects of Phytochemicals and Medicinal Herbs on Depression. BioMed Res. Int. 2017, 2017, 6596241. [Google Scholar] [CrossRef]
  5. Udrea, A.-M. Computational approaches of new perspectives in the treatment of depression during pregnancy. Farmacia 2018, 66, 680–687. [Google Scholar] [CrossRef]
  6. Khawam, E.A.; Laurencic, G.; Malone, D.A. Side Effects of Antidepressants: An Overview. Clevel. Clin. J. Med. 2006, 73, 351–353, 356–361. [Google Scholar] [CrossRef] [PubMed]
  7. Trivedi, M.H.; Fava, M.; Wisniewski, S.R.; Thase, M.E.; Quitkin, F.; Warden, D.; Ritz, L.; Nierenberg, A.A.; Lebowitz, B.D.; Biggs, M.M.; et al. Medication Augmentation after the Failure of SSRIs for Depression. N. Engl. J. Med. 2006, 354, 1243–1252. [Google Scholar] [CrossRef] [PubMed]
  8. Avram, S.; Puia, A.; Udrea, A.M.; Mihailescu, D.; Mernea, M.; Dinischiotu, A.; Oancea, F.; Stiens, J. Natural Compounds Therapeutic Features in Brain Disorders by Experimental, Bioinformatics and Cheminformatics Methods. Curr. Med. Chem. 2020, 27, 78–98. [Google Scholar] [CrossRef] [PubMed]
  9. Udrea, A.-M.; Mernea, M.; Buiu, C.; Avram, S. Scutellaria Baicalensis Flavones as Potent Drugs against Acute Respiratory Injury during SARS-CoV-2 Infection: Structural Biology Approaches. Processes 2020, 8, 1468. [Google Scholar] [CrossRef]
  10. Udrea, A.M.; Gradisteanu Pircalabioru, G.; Boboc, A.A.; Mares, C.; Dinache, A.; Mernea, M.; Avram, S. Advanced Bioinformatics Tools in the Pharmacokinetic Profiles of Natural and Synthetic Compounds with Anti-Diabetic Activity. Biomolecules 2021, 11, 1692. [Google Scholar] [CrossRef]
  11. Wang, Y.; Li, M.; Liang, Y.; Yang, Y.; Liu, Z.; Yao, K.; Chen, Z.; Zhai, S. Chinese Herbal Medicine for the Treatment of Depression: Applications, Efficacies and Mechanisms. Curr. Pharm. Des. 2017, 23, 5180–5190. [Google Scholar] [CrossRef] [PubMed]
  12. Sharma, R.; Kabra, A.; Rao, M.M.; Prajapati, P.K. Herbal and Holistic Solutions for Neurodegenerative and Depressive Disorders: Leads from Ayurveda. Curr. Pharm. Des. 2018, 24, 2597–2608. [Google Scholar] [CrossRef] [PubMed]
  13. Herbal Medicine for Depression, Anxiety and Insomnia: A Review of Psychopharmacology and Clinical Evidence—ScienceDirect. Available online: https://www.sciencedirect.com/science/article/pii/S0924977X1100071X (accessed on 22 December 2021).
  14. Avram, S.; Mernea, M.; Bagci, E.; Hritcu, L.; Borcan, L.-C.; Mihailescu, D.F. Advanced Structure-Activity Relationships Applied to Mentha Spicata L. Subsp. Spicata Essential Oil Compounds as AChE and NMDA Ligands, in Comparison with Donepezil, Galantamine and Memantine—New Approach in Brain Disorders Pharmacology. CNS Neurol. Disord. Drug Targets 2017, 16, 800–811. [Google Scholar] [CrossRef]
  15. Avram, S.; Movileanu, L.; Mihailescu, D.; Flonta, M.-L. Comparative Study of Some Energetic and Steric Parameters of the Wild Type and Mutants HIV-1 Protease: A Way to Explain the Viral Resistance. J. Cell. Mol. Med. 2002, 6, 251–260. [Google Scholar] [CrossRef]
  16. Milac, A.-L.; Avram, S.; Petrescu, A.-J. Evaluation of a Neural Networks QSAR Method Based on Ligand Representation Using Substituent Descriptors: Application to HIV-1 Protease Inhibitors. J. Mol. Graph. Model. 2006, 25, 37–45. [Google Scholar] [CrossRef]
  17. Avram, S.; Duda-Seiman, D.; Borcan, F.; Wolschann, P. QSAR-CoMSIA Applied to Antipsychotic Drugs with Their Dopamine D2 and Serotonine 5HT2A Membrane Receptors. J. Serbian Chem. Soc. 2011, 76, 263–281. [Google Scholar] [CrossRef]
  18. Avram, S.; Buiu, C.; Duda-Seiman, D.M.; Duda-Seiman, C.; Mihailescu, D. 3D-QSAR Design of New Escitalopram Derivatives for the Treatment of Major Depressive Disorders. Sci. Pharm. 2010, 78, 233–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Zhang, Y.; Long, Y.; Yu, S.; Li, D.; Yang, M.; Guan, Y.; Zhang, D.; Wan, J.; Liu, S.; Shi, A.; et al. Natural Volatile Oils Derived from Herbal Medicines: A Promising Therapy Way for Treating Depressive Disorder. Pharmacol. Res. 2021, 164, 105376. [Google Scholar] [CrossRef] [PubMed]
  20. Avram, S.; Stan, M.S.; Udrea, A.M.; Buiu, C.; Boboc, A.A.; Mernea, M. 3D-ALMOND-QSAR Models to Predict the Antidepressant Effect of Some Natural Compounds. Pharmaceutics 2021, 13, 1449. [Google Scholar] [CrossRef] [PubMed]
  21. Kim, S.; Chen, J.; Cheng, T.; Gindulyte, A.; He, J.; He, S.; Li, Q.; Shoemaker, B.A.; Thiessen, P.A.; Yu, B.; et al. PubChem in 2021: New Data Content and Improved Web Interfaces. Nucleic Acids Res. 2021, 49, D1388–D1395. [Google Scholar] [CrossRef] [PubMed]
  22. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and Computational Approaches to Estimate Solubility and Permeability in Drug Discovery and Development Settings. Adv. Drug Deliv. Rev. 1997, 23, 3–25. [Google Scholar] [CrossRef]
  23. Veber, D.F.; Johnson, S.R.; Cheng, H.-Y.; Smith, B.R.; Ward, K.W.; Kopple, K.D. Molecular Properties That Influence the Oral Bioavailability of Drug Candidates. J. Med. Chem. 2002, 45, 2615–2623. [Google Scholar] [CrossRef] [PubMed]
  24. Ghose, A.K.; Viswanadhan, V.N.; Wendoloski, J.J. A Knowledge-Based Approach in Designing Combinatorial or Medicinal Chemistry Libraries for Drug Discovery. 1. A Qualitative and Quantitative Characterization of Known Drug Databases. J. Comb. Chem. 1999, 1, 55–68. [Google Scholar] [CrossRef] [PubMed]
  25. Egan, W.J.; Merz, K.M.; Baldwin, J.J. Prediction of Drug Absorption Using Multivariate Statistics. J. Med. Chem. 2000, 43, 3867–3877. [Google Scholar] [CrossRef] [PubMed]
  26. Daina, A.; Michielin, O.; Zoete, V. SwissADME: A Free Web Tool to Evaluate Pharmacokinetics, Drug-Likeness and Medicinal Chemistry Friendliness of Small Molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Pires, D.E.V.; Blundell, T.L.; Ascher, D.B. PkCSM: Predicting Small-Molecule Pharmacokinetic and Toxicity Properties Using Graph-Based Signatures. J. Med. Chem. 2015, 58, 4066–4072. [Google Scholar] [CrossRef] [PubMed]
  28. PDSP. Available online: https://pdsp.unc.edu/databases/kidb.php (accessed on 22 December 2021).
  29. Tozar, T.; Santos Costa, S.; Udrea, A.-M.; Nastasa, V.; Couto, I.; Viveiros, M.; Pascu, M.L.; Romanitan, M.O. Anti-Staphylococcal Activity and Mode of Action of Thioridazine Photoproducts. Sci. Rep. 2020, 10, 18043. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The spatial structures of the most active natural compounds identified in the present study—neryl acetate, 1,8-cineole, and linalyl acetate and of the most active drugs that we considered in QSAR training sets—paroxetine, spiperone, and ziprasidone.
Figure 1. The spatial structures of the most active natural compounds identified in the present study—neryl acetate, 1,8-cineole, and linalyl acetate and of the most active drugs that we considered in QSAR training sets—paroxetine, spiperone, and ziprasidone.
Blsf 07 00010 g001
Table 1. Computed AMES, hepatotoxicity, hERG I and hERG II inhibitor for selected natural compounds and drugs.
Table 1. Computed AMES, hepatotoxicity, hERG I and hERG II inhibitor for selected natural compounds and drugs.
CompoundHepatotoxicityAMES ToxicityhERG I InhibitorhERG II Inhibitor
linalyl acetateNoNoNoNo
1,8-cineoleNoNoNoNo
neryl acetateNoNoNoNo
paroxetineYesYesNoYes
ziprasidoneYesNoNoYes
spiperoneYesNoNoYes
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Avram, S.; Stan, M.S.; Udrea, A.M.; Buiu, C.; Mernea, M. The Study of Natural Compounds as Antidepressants by Bioinformatics Methods. Biol. Life Sci. Forum 2021, 7, 10. https://doi.org/10.3390/ECB2021-10268

AMA Style

Avram S, Stan MS, Udrea AM, Buiu C, Mernea M. The Study of Natural Compounds as Antidepressants by Bioinformatics Methods. Biology and Life Sciences Forum. 2021; 7(1):10. https://doi.org/10.3390/ECB2021-10268

Chicago/Turabian Style

Avram, Speranta, Miruna Silvia Stan, Ana Maria Udrea, Catalin Buiu, and Maria Mernea. 2021. "The Study of Natural Compounds as Antidepressants by Bioinformatics Methods" Biology and Life Sciences Forum 7, no. 1: 10. https://doi.org/10.3390/ECB2021-10268

APA Style

Avram, S., Stan, M. S., Udrea, A. M., Buiu, C., & Mernea, M. (2021). The Study of Natural Compounds as Antidepressants by Bioinformatics Methods. Biology and Life Sciences Forum, 7(1), 10. https://doi.org/10.3390/ECB2021-10268

Article Metrics

Back to TopTop