Basic Study
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Jun 27, 2022; 14(6): 1131-1141
Published online Jun 27, 2022. doi: 10.4254/wjh.v14.i6.1131
COVID-19 liver and gastroenterology findings: An in silico analysis of SARS-CoV-2 interactions with liver molecules
Gabrielle Caroline Peiter, Cristiano de Bem Torquato de Souza, Lucca Miketen de Oliveira, Luis Gustavo Pagliarin, Valentina Nunes Fontoura dos Anjos, Filipe Antônio França da Silva, Fabrício Freire de Melo, Kádima Nayara Teixeira
Gabrielle Caroline Peiter, Cristiano de Bem Torquato de Souza, Lucca Miketen de Oliveira, Luis Gustavo Pagliarin, Valentina Nunes Fontoura dos Anjos, Kádima Nayara Teixeira, Universidade Federal do Paraná, Campus Toledo, Toledo 85919-899, Paraná, Brazil
Filipe Antônio França da Silva, Fabrício Freire de Melo, Universidade Federal da Bahia, Campus Anísio Teixeira, Vitória da Conquista 45029-094, Bahia, Brazil
Author contributions: Peiter GC, de Souza CBT, Oliveira LM, dos Anjos VNF and Pagliarin LG performed the experiments, analyzed the results and wrote the manuscript; da Silva FAF analyzed the results and reviewed the manuscript; de Melo FF performed a critical analysis of the results and corrected the manuscript; Teixeira KN interpreted the data, performed a critical analysis of the results, corrected the manuscript and coordinated the study; all authors approved the final version of the manuscript.
Institutional review board statement: The study did not need approval by the Research Ethics Committee as it was an in silico study.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Data sharing statement: No additional data are available.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Kádima Nayara Teixeira, PhD, Professor, Universidade Federal do Paraná, Campus Toledo, Road 182, Km 320/321, Toledo 85919-899, Paraná, Brazil. kadimateixeira@ufpr.br
Received: December 3, 2021
Peer-review started: December 3, 2021
First decision: February 8, 2022
Revised: February 22, 2022
Accepted: May 16, 2022
Article in press: May 16, 2022
Published online: June 27, 2022
Abstract
BACKGROUND

Coronavirus disease 19 (COVID-19) has not only been shown to affect the respiratory system, but has also demonstrated variable clinical presentations including gastrointestinal tract disorders. In addition, abnormalities in liver enzymes have been reported indicating hepatic injury. It is known that severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) might infect cells via the viral receptor angiotensin-converting enzyme 2 (ACE2) which is expressed in several organs including the liver. The viral Spike glycoprotein binds to ACE2 and must be cleaved by Furin and Type 2 Serine Protease to enter the cells. After that, the Akt/mTOR signaling pathway is activated and several COVID-19 changes are triggered.

AIM

To analyze liver and gastrointestinal symptoms and cell signaling pathways triggered by SARS-CoV-2 infection due to virus-liver interactions in silico.

METHODS

In this in silico study, the three-dimensional structures of the Akt, mTORC1 and Furin (receptors) were selected from the Protein Data Bank (PDB) and the structures of inhibitors (ligands) MK-2206, CC-223 and Naphthofluorescein were selected from PubChem and ZINC databases. Ligand files were downloaded as 2D structures and converted to optimized 3D structures using ViewerLite 4.2 software. Marvin Sketch® software was used to calculate prediction of the protonated form of inhibitors in a physiological environment (pH 7.4). AutoDock Tools (ADT) software was used to calculate and delimit the Grid box used in the molecular docking of each structure selected in the PDB. In addition, protonated ligands were prepared for molecular docking using ADT software. Molecular docking was performed using ADT software tools connected to Vina software. Analysis of the amino acid residues involved in ligand interactions, as well as ligand twists, the atoms involved in interactions, bond type and strength of interactions were performed using PyMol® and Discovery Studio® (BIOVIA) software.

RESULTS

Molecular docking analysis showed that the mTORC1/CC-223 complex had affinity energy between the receptor and ligand of -7.7 kcal/moL with interactions ranging from 2.7 to 4.99 Å. There were four significant chemical bonds which involved two of five polypeptide chains that formed the FKBP12–Rapamycin-Binding (FRB) domain. The strongest was a hydrogen bond, the only polar interaction, and Van der Waals interactions shown to be present in 12 residues of mTORC1’s FRB domain. With regard to the Akt/MK-2206 complex there were three Van der Waals interactions and 12 chemical bonds in which seven residues of Akt were involved with all five rings of the MK-2206 structure. In this way, both ASP 388 and GLN 391 bind to the same MK-2206 ring, the smaller one. However, LYS 386 had four chemical bonds with the inhibitor, one with each structure ring, while LYS 387 binds two distinct rings. One of the MK-2206 inhibitor's rings which binds to LYS 387 also binds simultaneously to ILE 367 and LEU 385 residues, and the fifth ring of the structure was involved in a bond with the ALA 382 residue. The hydrogen bonds were the shortest bonds in the complex (2.61 and 3.08 Å) and all interactions had an affinity energy of -8.8 kcal/moL. The affinity energy in the Furin/Naphhofluorescein complex was -9.8 kcal/moL and involved six interactions ranging from 2.57 to 4.98 Å. Among them, two were polar and the others were non-polar, in addition to twelve more Van der Waals interactions. Two distinct hydrogen bonds were formed between Furin and its inhibitor involving GLN 388 and ALA 532 residues. ALA 532 also binds to two distinct rings of Naphthofluorescein, while TRP 531 residue has two simultaneous bonds with the inhibitor.

CONCLUSION

Liver infection and signaling pathways altered by SARS-CoV-2 can be modulated by inhibitors that demonstrate significant interaction affinity with human proteins, which could prevent the development of infection and symptoms.

Keywords: Bioinformatics, Cell signaling pathway, COVID-19, Liver injury, SARS-CoV-2

Core Tip: The classification of coronavirus disease 19 (COVID-19) as a respiratory disease caused the focus of studies to be directed in this direction. Therefore, mild clinical symptoms, such as gastrointestinal symptoms, have been little studied. Following the knowledge that COVID-19 is a systemic disease, studies on liver damage have become important. This study analyzed liver molecules targeted by severe acute respiratory syndrome coronavirus-2 infection using bioinformatics. Although these molecules are present in various organs due to the liver's central role in systemic metabolism, trying to understand metabolic changes in this organ will help understand systemic changes induced by the virus.