The Fight against COVID-19 on the Multi-Protease Front and Surroundings: Could an Early Therapeutic Approach with Repositioning Drugs Prevent the Disease Severity?
Abstract
:1. Introduction
Literature Search Strategy
2. The Binding of SARS-CoV-2 to ACE2 Receptor Is Just the Tip of the Iceberg: The Submerged Front of Multi-Proteases
2.1. Furin Priming
2.2. TMPRSS2
2.3. Cathepsin L-Dependent P-Pathway
2.4. ADAM17 and Soluble ACE2
3. Co-Receptors for ACE2: Sialic Acids, Neuropilin-1, Heparan Sulfate and Integrins
3.1. Sialic Acids
3.2. Neuropilin-1
3.3. Heparan Sulfate
3.4. Integrins
4. Other Receptors for SARS-CoV-2 Entry: CD147 and GRP78
4.1. CD147
4.2. GRP78
5. Effectiveness of Anti-inflammatory, Antibiotic and Anti-coagulant Treatments in the Early Stage of Infection
5.1. Usefulness of Anti-inflammatory Drugs Administered Upstream of Hyperinflammation
5.2. Usefulness of Antibiotics Administered Upstream of Hyperinflammation
5.3. Coagulation Factors and an Exacerbate Production of Antibodies Enhance Viral Entry
6. Conclusions and Prospective
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Acknowledgments
Conflicts of Interest
References
- Hussain, M.; Jabeen, N.; Amanullah, A.; Baig, A.A.; Aziz, B.; Shabbir, S.; Raza, F.; Uddin, N. Molecular docking between human TMPRSS2 and SARS-CoV-2 spike protein: Conformation and intermolecular interactions. AIMS Microbiol. 2020, 6, 350–360. [Google Scholar] [CrossRef] [PubMed]
- Gavriatopoulou, M.; Korompoki, E.; Fotiou, D.; Ntanasis-Stathopoulos, I.; Psaltopoulou, T.; Kastritis, E.; Terpos, E.; Dimopoulos, M.A. Organ-specific manifestations of COVID-19 infection. Clin. Exp. Med. 2020, 20, 493–506. [Google Scholar] [CrossRef] [PubMed]
- Xie, J.; Covassin, N.; Fan, Z.; Singh, P.; Gao, W.; Li, G.; Kara, T.; Somers, V.K. Association Between Hypoxemia and Mortality in Patients With COVID-19. Mayo Clin. Proc. 2020, 95, 1138–1147. [Google Scholar] [CrossRef] [PubMed]
- Devaux, C.A.; Rolain, J.M.; Raoult, D. ACE2 receptor polymorphism: Susceptibility to SARS-CoV-2, hypertension, multi-organ failure, and COVID-19 disease outcome. J. Microbiol. Immunol. Infect. 2020, 53, 425–435. [Google Scholar] [CrossRef]
- Borczuk, A.C.; Salvatore, S.P.; Seshan, S.V.; Patel, S.S.; Bussel, J.B.; Mostyka, M.; Elsoukkary, S.; He, B.; Del Vecchio, C.; Fortarezza, F.; et al. COVID-19 pulmonary pathology: A multi-institutional autopsy cohort from Italy and New York City. Mod. Pathol. 2020, 33, 2156–2168. [Google Scholar] [CrossRef]
- Senapati, S.; Banerjee, P.; Bhagavatula, S.; Kushwaha, P.P.; Kumar, S. Contributions of human ACE2 and TMPRSS2 in determining host-pathogen interaction of COVID-19. J. Genet. 2021, 100. [Google Scholar]
- Shang, J.; Wan, Y.; Luo, C.; Ye, G.; Geng, Q.; Auerbach, A.; Li, F. Cell entry mechanisms of SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2020, 117, 11727–11734. [Google Scholar] [CrossRef]
- Zamorano Cuervo, N.; Grandvaux, N. ACE2: Evidence of role as entry receptor for SARS-CoV-2 and implications in comorbidities. Elife 2020, 9, e61390. [Google Scholar] [CrossRef]
- Chiappelli, F. CoViD-19 Susceptibility. Bioinformation 2020, 16, 501–504. [Google Scholar] [CrossRef]
- Wang, Q.; Zhang, Y.; Wu, L.; Niu, S.; Song, C.; Zhang, Z.; Lu, G.; Qiao, C.; Hu, Y.; Yuen, K.Y.; et al. Structural and Functional Basis of SARS-CoV-2 Entry by Using Human ACE2. Cell 2020, 181, 894–904. [Google Scholar] [CrossRef]
- Perlot, T.; Penninger, J.M. ACE2—From the renin-angiotensin system to gut microbiota and malnutrition. Microbes Infect. 2013, 15, 866–873. [Google Scholar] [CrossRef]
- Heurich, A.; Hofmann-Winkler, H.; Gierer, S.; Liepold, T.; Jahn, O.; Pohlmann, S. TMPRSS2 and ADAM17 cleave ACE2 differentially and only proteolysis by TMPRSS2 augments entry driven by the severe acute respiratory syndrome coronavirus spike protein. J. Virol. 2014, 88, 1293–1307. [Google Scholar] [CrossRef] [Green Version]
- Vardhan, S.; Sahoo, S.K. Virtual screening by targeting proteolytic sites of furin and TMPRSS2 to propose potential compounds obstructing the entry of SARS-CoV-2 virus into human host cells. J. Tradit. Complement. Med. 2021, in press. [Google Scholar] [CrossRef]
- Benton, D.J.; Wrobel, A.G.; Xu, P.; Roustan, C.; Martin, S.R.; Rosenthal, P.B.; Skehel, J.J.; Gamblin, S.J. Receptor binding and priming of the spike protein of SARS-CoV-2 for membrane fusion. Nature 2020, 588, 327–330. [Google Scholar] [CrossRef]
- Belouzard, S.; Chu, V.C.; Whittaker, G.R. Activation of the SARS coronavirus spike protein via sequential proteolytic cleavage at two distinct sites. Proc. Natl. Acad. Sci. USA 2009, 106, 5871–5876. [Google Scholar] [CrossRef] [Green Version]
- Hoffmann, M.; Kleine-Weber, H.; Pohlmann, S. A Multibasic Cleavage Site in the Spike Protein of SARS-CoV-2 Is Essential for Infection of Human Lung Cells. Mol. Cell 2020, 78, 779–784. [Google Scholar] [CrossRef]
- Aguiar, J.A.; Tremblay, B.J.; Mansfield, M.J.; Woody, O.; Lobb, B.; Banerjee, A.; Chandiramohan, A.; Tiessen, N.; Cao, Q.; Dvorkin-Gheva, A.; et al. Gene expression and in situ protein profiling of candidate SARS-CoV-2 receptors in human airway epithelial cells and lung tissue. Eur. Respir. J. 2020, 56, 3. [Google Scholar] [CrossRef]
- Zhou, N.; Pan, T.; Zhang, J.; Li, Q.; Zhang, X.; Bai, C.; Huang, F.; Peng, T.; Zhang, J.; Liu, C.; et al. Glycopeptide Antibiotics Potently Inhibit Cathepsin L in the Late Endosome/Lysosome and Block the Entry of Ebola Virus, Middle East Respiratory Syndrome Coronavirus (MERS-CoV), and Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV). J. Biol. Chem. 2016, 291, 9218–9232. [Google Scholar] [CrossRef] [Green Version]
- Kastenhuber, E.R.; Jaimes, J.A.; Johnson, J.L.; Mercadante, M.; Muecksch, F.; Weisblum, Y.; Bram, Y.; Schwartz, R.E.; Whittaker, G.R.; Cantley, L.C. Coagulation factors directly cleave SARS-CoV-2 spike and enhance viral entry. bioRxiv 2021. [Google Scholar] [CrossRef]
- Frydman, G.H.; Streiff, M.B.; Connors, J.M.; Piazza, G. The Potential Role of Coagulation Factor Xa in the Pathophysiology of COVID-19: A Role for Anticoagulants as Multimodal Therapeutic Agents. TH Open 2020, 4, e288–e299. [Google Scholar] [CrossRef]
- Wielgat, P.; Rogowski, K.; Godlewska, K.; Car, H. Coronaviruses: Is Sialic Acid a Gate to the Eye of Cytokine Storm? From the Entry to the Effects. Cells 2020, 9, 1963. [Google Scholar] [CrossRef] [PubMed]
- Daly, J.L.; Simonetti, B.; Klein, K.; Chen, K.E.; Williamson, M.K.; Anton-Plagaro, C.; Shoemark, D.K.; Simon-Gracia, L.; Bauer, M.; Hollandi, R.; et al. Neuropilin-1 is a host factor for SARS-CoV-2 infection. Science 2020, 370, 861–865. [Google Scholar] [CrossRef] [PubMed]
- Dakal, T.C. SARS-CoV-2 attachment to host cells is possibly mediated via RGD-integrin interaction in a calcium-dependent manner and suggests pulmonary EDTA chelation therapy as a novel treatment for COVID 19. Immunobiology 2021, 226, 152021. [Google Scholar] [CrossRef] [PubMed]
- Sigrist, C.J.; Bridge, A.; Le Mercier, P. A potential role for integrins in host cell entry by SARS-CoV-2. Antivir. Res. 2020, 177, 104759. [Google Scholar] [CrossRef]
- Clausen, T.M.; Sandoval, D.R.; Spliid, C.B.; Pihl, J.; Perrett, H.R.; Painter, C.D.; Narayanan, A.; Majowicz, S.A.; Kwong, E.M.; McVicar, R.N.; et al. SARS-CoV-2 Infection Depends on Cellular Heparan Sulfate and ACE2. Cell 2020, 183, 1043–1057. [Google Scholar] [CrossRef]
- Yan, L.; Song, Y.; Xia, K.; He, P.; Zhang, F.; Chen, S.; Pouliot, R.; Weiss, D.J.; Tandon, R.; Bates, J.T.; et al. Heparan sulfates from bat and human lung and their binding to the spike protein of SARS-CoV-2 virus. Carbohydr. Polym. 2021, 260, 117797. [Google Scholar] [CrossRef]
- Wang, K.; Chen, W.; Zhang, Z.; Deng, Y.; Lian, J.Q.; Du, P.; Wei, D.; Zhang, Y.; Sun, X.X.; Gong, L.; et al. CD147-spike protein is a novel route for SARS-CoV-2 infection to host cells. Signal. Transduct. Target. Ther. 2020, 5, 283. [Google Scholar] [CrossRef]
- Ibrahim, I.M.; Abdelmalek, D.H.; Elshahat, M.E.; Elfiky, A.A. COVID-19 spike-host cell receptor GRP78 binding site prediction. J. Infect. 2020, 80, 554–562. [Google Scholar] [CrossRef]
- Iba, T.; Connors, J.M.; Levy, J.H. The coagulopathy, endotheliopathy, and vasculitis of COVID-19. Inflamm. Res. 2020, 69, 1181–1189. [Google Scholar] [CrossRef]
- Del Turco, S.; Vianello, A.; Ragusa, R.; Caselli, C.; Basta, G. COVID-19 and cardiovascular consequences: Is the endothelial dysfunction the hardest challenge? Thromb. Res. 2020, 196, 143–151. [Google Scholar] [CrossRef]
- Xu, H.; Zhong, L.; Deng, J.; Peng, J.; Dan, H.; Zeng, X.; Li, T.; Chen, Q. High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int. J. Oral. Sci. 2020, 12, 8. [Google Scholar] [CrossRef]
- Guzik, T.J.; Mohiddin, S.A.; Dimarco, A.; Patel, V.; Savvatis, K.; Marelli-Berg, F.M.; Madhur, M.S.; Tomaszewski, M.; Maffia, P.; D’Acquisto, F.; et al. COVID-19 and the cardiovascular system: Implications for risk assessment, diagnosis, and treatment options. Cardiovasc. Res. 2020, 116, 1666–1687. [Google Scholar] [CrossRef]
- Lan, J.; Ge, J.; Yu, J.; Shan, S.; Zhou, H.; Fan, S.; Zhang, Q.; Shi, X.; Wang, Q.; Zhang, L.; et al. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 2020, 581, 215–220. [Google Scholar] [CrossRef] [Green Version]
- Volz, E.; Hill, V.; McCrone, J.T.; Price, A.; Jorgensen, D.; O’Toole, A.; Southgate, J.; Johnson, R.; Jackson, B.; Nascimento, F.F.; et al. Evaluating the Effects of SARS-CoV-2 Spike Mutation D614G on Transmissibility and Pathogenicity. Cell 2021, 184, 64–75. [Google Scholar] [CrossRef]
- Khan, A.; Zia, T.; Suleman, M.; Khan, T.; Ali, S.S.; Abbasi, A.A.; Mohammad, A.; Wei, D.Q. Higher infectivity of the SARS-CoV-2 new variants is associated with K417N/T, E484K, and N501Y mutants: An insight from structural data. J. Cell. Physiol. 2021. [Google Scholar] [CrossRef]
- Du, L.; Kao, R.Y.; Zhou, Y.; He, Y.; Zhao, G.; Wong, C.; Jiang, S.; Yuen, K.Y.; Jin, D.Y.; Zheng, B.J. Cleavage of spike protein of SARS coronavirus by protease factor Xa is associated with viral infectivity. Biochem. Biophys. Res. Commun. 2007, 359, 174–179. [Google Scholar] [CrossRef]
- Tang, T.; Jaimes, J.A.; Bidon, M.K.; Straus, M.R.; Daniel, S.; Whittaker, G.R. Proteolytic Activation of SARS-CoV-2 Spike at the S1/S2 Boundary: Potential Role of Proteases beyond Furin. ACS Infect. Dis. 2021, 7, 264–272. [Google Scholar] [CrossRef]
- Walls, A.C.; Park, Y.J.; Tortorici, M.A.; Wall, A.; McGuire, A.T.; Veesler, D. Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 2020, 183, 1735. [Google Scholar] [CrossRef]
- Sakurai, Y.; Kolokoltsov, A.A.; Chen, C.C.; Tidwell, M.W.; Bauta, W.E.; Klugbauer, N.; Grimm, C.; Wahl-Schott, C.; Biel, M.; Davey, R.A. Ebola virus. Two-pore channels control Ebola virus host cell entry and are drug targets for disease treatment. Science 2015, 347, 995–998. [Google Scholar] [CrossRef] [Green Version]
- Gunaratne, G.S.; Yang, Y.; Li, F.; Walseth, T.F.; Marchant, J.S. NAADP-dependent Ca2+ signaling regulates Middle East respiratory syndrome-coronavirus pseudovirus translocation through the endolysosomal system. Cell Calcium. 2018, 75, 30–41. [Google Scholar] [CrossRef]
- Oz, M.; Lorke, D.E. Multifunctional angiotensin converting enzyme 2, the SARS-CoV-2 entry receptor, and critical appraisal of its role in acute lung injury. Biomed. Pharm. 2021, 136, 111193. [Google Scholar] [CrossRef] [PubMed]
- Andersen, K.G.; Rambaut, A.; Lipkin, W.I.; Holmes, E.C.; Garry, R.F. The proximal origin of SARS-CoV-2. Nat. Med. 2020, 26, 450–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seyran, M.; Pizzol, D.; Adadi, P.; El-Aziz, T.M.A.; Hassan, S.S.; Soares, A.; Kandimalla, R.; Lundstrom, K.; Tambuwala, M.; Aljabali, A.A.A.; et al. Questions concerning the proximal origin of SARS-CoV-2. J. Med. Virol. 2021, 93, 1204–1206. [Google Scholar] [CrossRef] [PubMed]
- Fitzgerald, K. Furin Protease: From SARS CoV-2 to Anthrax, Diabetes, and Hypertension. Perm. J. 2020, 24, 24. [Google Scholar] [CrossRef]
- Van Lam van, T.; Ivanova, T.; Hardes, K.; Heindl, M.R.; Morty, R.E.; Bottcher-Friebertshauser, E.; Lindberg, I.; Than, M.E.; Dahms, S.O.; Steinmetzer, T. Design, Synthesis, and Characterization of Macrocyclic Inhibitors of the Proprotein Convertase Furin. ChemMedChem 2019, 14, 673–685. [Google Scholar] [CrossRef]
- Palit, P.; Chattopadhyay, D.; Thomas, S.; Kundu, A.; Kim, H.S.; Rezaei, N. Phytopharmaceuticals mediated Furin and TMPRSS2 receptor blocking: Can it be a potential therapeutic option for Covid-19? Phytomedicine 2021, 85, 153396. [Google Scholar] [CrossRef]
- Morais, A.H.A.; de Medeiros, A.F.; Medeiros, I.; de Lima, V.C.O.; Luz, A.B.S.; Maciel, B.L.L.; Passos, T.S. Tamarind (Tamarindus indica L.) Seed a Candidate Protein Source with Potential for Combating SARS-CoV-2 Infection in Obesity. Drug Target. Insights 2021, 15, 5–12. [Google Scholar] [CrossRef]
- Yamaya, M.; Nishimura, H.; Deng, X.; Kikuchi, A.; Nagatomi, R. Protease Inhibitors: Candidate Drugs to Inhibit Severe Acute Respiratory Syndrome Coronavirus 2 Replication. Tohoku J. Exp. Med. 2020, 251, 27–30. [Google Scholar] [CrossRef]
- Yamamoto, M.; Kiso, M.; Sakai-Tagawa, Y.; Iwatsuki-Horimoto, K.; Imai, M.; Takeda, M.; Kinoshita, N.; Ohmagari, N.; Gohda, J.; Semba, K.; et al. The Anticoagulant Nafamostat Potently Inhibits SARS-CoV-2 S Protein-Mediated Fusion in a Cell Fusion Assay System and Viral Infection In Vitro in a Cell-Type-Dependent Manner. Viruses 2020, 12, 629. [Google Scholar] [CrossRef]
- de Loyola, M.B.; Dos Reis, T.T.A.; de Oliveira, G.; da Fonseca Palmeira, J.; Arganaraz, G.A.; Arganaraz, E.R. Alpha-1-antitrypsin: A possible host protective factor against Covid-19. Rev. Med. Virol. 2021, 31, e2157. [Google Scholar] [CrossRef]
- Braga, L.; Ali, H.; Secco, I.; Chiavacci, E.; Neves, G.; Goldhill, D.; Penn, R.; Jimenez-Guardeno, J.M.; Ortega-Prieto, A.M.; Bussani, R.; et al. Drugs that inhibit TMEM16 proteins block SARS-CoV-2 spike-induced syncytia. Nature 2021, 594, 88–93. [Google Scholar] [CrossRef]
- Zhang, X.; Zhang, G.; Zhai, W.; Zhao, Z.; Wang, S.; Yi, J. Inhibition of TMEM16A Ca(2+)-activated Cl(-) channels by avermectins is essential for their anticancer effects. Pharmacol. Res. 2020, 156, 104763. [Google Scholar] [CrossRef]
- Maisonnasse, P.; Guedj, J.; Contreras, V.; Behillil, S.; Solas, C.; Marlin, R.; Naninck, T.; Pizzorno, A.; Lemaitre, J.; Goncalves, A.; et al. Hydroxychloroquine use against SARS-CoV-2 infection in non-human primates. Nature 2020, 585, 584–587. [Google Scholar] [CrossRef]
- Rebeaud, M.E.; Zores, F. SARS-CoV-2 and the Use of Chloroquine as an Antiviral Treatment. Front. Med. 2020, 7, 184. [Google Scholar] [CrossRef]
- Borba, M.G.S.; Val, F.F.A.; Sampaio, V.S.; Alexandre, M.A.A.; Melo, G.C.; Brito, M.; Mourao, M.P.G.; Brito-Sousa, J.D.; Baia-da-Silva, D.; Guerra, M.V.F.; et al. Effect of High vs. Low Doses of Chloroquine Diphosphate as Adjunctive Therapy for Patients Hospitalized With Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection: A Randomized Clinical Trial. JAMA Netw. Open 2020, 3, e208857. [Google Scholar] [CrossRef] [Green Version]
- Hoffmann, M.; Mosbauer, K.; Hofmann-Winkler, H.; Kaul, A.; Kleine-Weber, H.; Kruger, N.; Gassen, N.C.; Muller, M.A.; Drosten, C.; Pohlmann, S. Chloroquine does not inhibit infection of human lung cells with SARS-CoV-2. Nature 2020, 585, 588–590. [Google Scholar] [CrossRef]
- Perez-Miller, S.; Patek, M.; Moutal, A.; Duran, P.; Cabel, C.R.; Thorne, C.A.; Campos, S.K.; Khanna, R. Novel Compounds Targeting Neuropilin Receptor 1 with Potential To Interfere with SARS-CoV-2 Virus Entry. ACS Chem. Neurosci. 2021, 12, 1299–1312. [Google Scholar] [CrossRef]
- Yu, M.; Zhang, T.; Zhang, W.; Sun, Q.; Li, H.; Li, J.P. Elucidating the Interactions Between Heparin/Heparan Sulfate and SARS-CoV-2-Related Proteins-An Important Strategy for Developing Novel Therapeutics for the COVID-19 Pandemic. Front. Mol. Biosci. 2020, 7, 628551. [Google Scholar] [CrossRef]
- Tandon, R.; Sharp, J.S.; Zhang, F.; Pomin, V.H.; Ashpole, N.M.; Mitra, D.; McCandless, M.G.; Jin, W.; Liu, H.; Sharma, P.; et al. Effective Inhibition of SARS-CoV-2 Entry by Heparin and Enoxaparin Derivatives. J. Virol. 2021, 95, e01987-20. [Google Scholar] [CrossRef]
- Beddingfield, B.J.; Iwanaga, N.; Chapagain, P.P.; Zheng, W.; Roy, C.J.; Hu, T.Y.; Kolls, J.K.; Bix, G.J. The Integrin Binding Peptide, ATN-161, as a Novel Therapy for SARS-CoV-2 Infection. JACC Basic Transl. Sci. 2021, 6, 1–8. [Google Scholar] [CrossRef]
- Ulrich, H.; Pillat, M.M. CD147 as a Target for COVID-19 Treatment: Suggested Effects of Azithromycin and Stem Cell Engagement. Stem. Cell Rev. Rep. 2020, 16, 434–440. [Google Scholar] [CrossRef]
- Liu, C.; von Brunn, A.; Zhu, D. Cyclophilin A and CD147: Novel therapeutic targets for the treatment of COVID-19. Med. Drug Discov. 2020, 7, 100056. [Google Scholar] [CrossRef]
- Malek, A.E.; Granwehr, B.P.; Kontoyiannis, D.P. Doxycycline as a potential partner of COVID-19 therapies. IDCases 2020, 21, e00864. [Google Scholar] [CrossRef]
- Yates, P.A.; Newman, S.A.; Oshry, L.J.; Glassman, R.H.; Leone, A.M.; Reichel, E. Doxycycline treatment of high-risk COVID-19-positive patients with comorbid pulmonary disease. Ther. Adv. Respir. Dis. 2020, 14, 1753466620951053. [Google Scholar] [CrossRef]
- Allam, L.; Ghrifi, F.; Mohammed, H.; Hafidi, N.E.; Jaoudi, R.E.; Harti, J.E.; Lmimouni, B.; Belyamani, L.; Ibrahimi, A. Targeting the GRP78-Dependant SARS-CoV-2 Cell Entry by Peptides and Small Molecules. Bioinform. Biol. Insights. 2020, 14, 1177932220965505. [Google Scholar] [CrossRef]
- Balmeh, N.; Mahmoudi, S.; Mohammadi, N.; Karabedianhajiabadi, A. Predicted therapeutic targets for COVID-19 disease by inhibiting SARS-CoV-2 and its related receptors. Inform. Med. Unlocked 2020, 20, 100407. [Google Scholar] [CrossRef]
- Suter, F.; Consolaro, E.; Pedroni, S.; Moroni, C.; Pastò, E.; Paganini, M.V.; Pravettoni, G.; Cantarelli, U.; Rubis, N.; Perico, N.; et al. A Simple, Home-Therapy Algorithm to Prevent Hospitalization for COVID-19 Patients: A Retrospective Observational Matched-Cohort Study. medRxiv 2021. [Google Scholar] [CrossRef]
- Fuentes-Prior, P. Priming of SARS-CoV-2 S protein by several membrane-bound serine proteinases could explain enhanced viral infectivity and systemic COVID-19 infection. J. Biol. Chem. 2021, 100135. [Google Scholar] [CrossRef] [PubMed]
- Gemmati, D.; Bramanti, B.; Serino, M.L.; Secchiero, P.; Zauli, G.; Tisato, V. COVID-19 and Individual Genetic Susceptibility/Receptivity: Role of ACE1/ACE2 Genes, Immunity, Inflammation and Coagulation. Might the Double X-chromosome in Females Be Protective against SARS-CoV-2 Compared to the Single X-Chromosome in Males? Int. J. Mol. Sci. 2020, 21, 3474. [Google Scholar] [CrossRef] [PubMed]
- Guney, C.; Akar, F. Epithelial and Endothelial Expressions of ACE2: SARS-CoV-2 Entry Routes. J. Pharm. Pharm. Sci. 2021, 24, 84–93. [Google Scholar] [CrossRef] [PubMed]
- Fathema, K.; Hassan, M.N.; Mazumder, M.W.; Benzamin, M.; Ahmed, M.; Islam, M.R.; Haque, N.; Sutradhar, P.K.; Rahman, A.R.; Rukunuzzaman, M. COVID 19 in Children: Gastrointestinal, Hepatobiliary and Pancreatic Manifestation. Mymensingh Med. J. 2021, 30, 570–579. [Google Scholar]
- Kaur, U.; Chakrabarti, S.S.; Ojha, B.; Pathak, B.K.; Singh, A.; Saso, L.; Chakrabarti, S. Targeting Host Cell Proteases to Prevent SARS-CoV-2 Invasion. Curr. Drug Targets 2021, 22, 192–201. [Google Scholar] [CrossRef]
- Dalpiaz, E.L.; Lamas, A.Z.; Caliman, I.F.; Ribeiro, R.F., Jr.; Abreu, G.R.; Moyses, M.R.; Andrade, T.U.; Gouvea, S.A.; Alves, M.F.; Carmona, A.K.; et al. Correction: Sex Hormones Promote Opposite Effects on ACE and ACE2 Activity, Hypertrophy and Cardiac Contractility in Spontaneously Hypertensive Rats. PLoS ONE 2015, 10, e0133225. [Google Scholar] [CrossRef] [Green Version]
- Bahmad, H.F.; Abou-Kheir, W. Crosstalk between COVID-19 and prostate cancer. Prostate Cancer Prostatic Dis. 2020, 23, 561–563. [Google Scholar] [CrossRef]
- Thunders, M.; Delahunt, B. Gene of the month: TMPRSS2 (transmembrane serine protease 2). J. Clin. Pathol. 2020, 73, 773–776. [Google Scholar] [CrossRef]
- Latini, A.; Agolini, E.; Novelli, A.; Borgiani, P.; Giannini, R.; Gravina, P.; Smarrazzo, A.; Dauri, M.; Andreoni, M.; Rogliani, P.; et al. COVID-19 and Genetic Variants of Protein Involved in the SARS-CoV-2 Entry into the Host Cells. Genes 2020, 11, 1010. [Google Scholar] [CrossRef]
- Buchrieser, J.; Dufloo, J.; Hubert, M.; Monel, B.; Planas, D.; Rajah, M.M.; Planchais, C.; Porrot, F.; Guivel-Benhassine, F.; Van der Werf, S.; et al. Syncytia formation by SARS-CoV-2-infected cells. EMBO J. 2020, 39, e106267. [Google Scholar] [CrossRef]
- Maggio, R.; Corsini, G.U. Repurposing the mucolytic cough suppressant and TMPRSS2 protease inhibitor bromhexine for the prevention and management of SARS-CoV-2 infection. Pharmacol. Res. 2020, 157, 104837. [Google Scholar] [CrossRef]
- Depfenhart, M.; de Villiers, D.; Lemperle, G.; Meyer, M.; Di Somma, S. Potential new treatment strategies for COVID-19: Is there a role for bromhexine as add-on therapy? Intern. Emerg. Med. 2020, 15, 801–812. [Google Scholar] [CrossRef]
- Olaleye, O.A.; Kaur, M.; Onyenaka, C.C. Ambroxol Hydrochloride Inhibits the Interaction between Severe Acute Respiratory Syndrome Coronavirus 2 Spike Protein’s Receptor Binding Domain and Recombinant Human ACE2. bioRxiv 2020. [Google Scholar] [CrossRef]
- Tian, Y.; Kongsuphol, P.; Hug, M.; Ousingsawat, J.; Witzgall, R.; Schreiber, R.; Kunzelmann, K. Calmodulin-dependent activation of the epithelial calcium-dependent chloride channel TMEM16A. FASEB J. 2011, 25, 1058–1068. [Google Scholar] [CrossRef] [Green Version]
- Maertens, C.; Wei, L.; Voets, T.; Droogmans, G.; Nilius, B. Block by fluoxetine of volume-regulated anion channels. Br. J. Pharmacol. 1999, 126, 508–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, Y.D.; Cho, H.; Koo, J.Y.; Tak, M.H.; Cho, Y.; Shim, W.S.; Park, S.P.; Lee, J.; Lee, B.; Kim, B.M.; et al. TMEM16A confers receptor-activated calcium-dependent chloride conductance. Nature 2008, 455, 1210–1215. [Google Scholar] [CrossRef]
- Gomes, C.P.; Fernandes, D.E.; Casimiro, F.; da Mata, G.F.; Passos, M.T.; Varela, P.; Mastroianni-Kirsztajn, G.; Pesquero, J.B. Cathepsin L in COVID-19: From Pharmacological Evidences to Genetics. Front. Cell. Infect. Microbiol. 2020, 10, 589505. [Google Scholar] [CrossRef] [PubMed]
- Wang, N.; Shang, J.; Jiang, S.; Du, L. Subunit Vaccines against Emerging Pathogenic Human Coronaviruses. Front. Microbiol. 2020, 11, 298. [Google Scholar] [CrossRef] [PubMed]
- Kumar, B.K.; Sekhar, K.; Kunjiappan, S.; Jamalis, J.; Balana-Fouce, R.; Tekwani, B.L.; Sankaranarayanan, M. Druggable targets of SARS-CoV-2 and treatment opportunities for COVID-19. Bioorg. Chem. 2020, 104, 104269. [Google Scholar] [CrossRef] [PubMed]
- Riva, L.; Yuan, S.; Yin, X.; Martin-Sancho, L.; Matsunaga, N.; Pache, L.; Burgstaller-Muehlbacher, S.; De Jesus, P.D.; Teriete, P.; Hull, M.V.; et al. Discovery of SARS-CoV-2 antiviral drugs through large-scale compound repurposing. Nature 2020, 586, 113–119. [Google Scholar] [CrossRef]
- Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Kruger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280. [Google Scholar] [CrossRef]
- Wang, M.; Cao, R.; Zhang, L.; Yang, X.; Liu, J.; Xu, M.; Shi, Z.; Hu, Z.; Zhong, W.; Xiao, G. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020, 30, 269–271. [Google Scholar] [CrossRef]
- Zhao, H.; To, K.K.W.; Lam, H.; Zhou, X.; Chan, J.F.; Peng, Z.; Lee, A.C.Y.; Cai, J.; Chan, W.M.; Ip, J.D.; et al. Cross-linking peptide and repurposed drugs inhibit both entry pathways of SARS-CoV-2. Nat. Commun. 2021, 12, 1517. [Google Scholar] [CrossRef]
- McKee, D.L.; Sternberg, A.; Stange, U.; Laufer, S.; Naujokat, C. Candidate drugs against SARS-CoV-2 and COVID-19. Pharmacol. Res. 2020, 157, 104859. [Google Scholar] [CrossRef]
- Zipeto, D.; Palmeira, J.D.F.; Arganaraz, G.A.; Arganaraz, E.R. ACE2/ADAM17/TMPRSS2 Interplay May Be the Main Risk Factor for COVID-19. Front. Immunol. 2020, 11, 576745. [Google Scholar] [CrossRef]
- Zoufaly, A.; Poglitsch, M.; Aberle, J.H.; Hoepler, W.; Seitz, T.; Traugott, M.; Grieb, A.; Pawelka, E.; Laferl, H.; Wenisch, C.; et al. Human recombinant soluble ACE2 in severe COVID-19. Lancet Respir. Med. 2020, 8, 1154–1158. [Google Scholar] [CrossRef]
- Monteil, V.; Dyczynski, M.; Lauschke, V.M.; Kwon, H.; Wirnsberger, G.; Youhanna, S.; Zhang, H.; Slutsky, A.S.; Hurtado Del Pozo, C.; Horn, M.; et al. Human soluble ACE2 improves the effect of remdesivir in SARS-CoV-2 infection. EMBO Mol. Med. 2021, 13, e13426. [Google Scholar] [CrossRef]
- Chu, H.; Hu, B.; Huang, X.; Chai, Y.; Zhou, D.; Wang, Y.; Shuai, H.; Yang, D.; Hou, Y.; Zhang, X.; et al. Host and viral determinants for efficient SARS-CoV-2 infection of the human lung. Nat. Commun. 2021, 12, 134. [Google Scholar] [CrossRef]
- Mayi, B.S.; Leibowitz, J.A.; Woods, A.T.; Ammon, K.A.; Liu, A.E.; Raja, A. The role of Neuropilin-1 in COVID-19. PLoS Pathog. 2021, 17, e1009153. [Google Scholar] [CrossRef]
- Makowski, L.; Olson-Sidford, W.; John, W.W. Biological and Clinical Consequences of Integrin Binding via a Rogue RGD Motif in the SARS CoV-2 Spike Protein. Viruses 2021, 13, 146. [Google Scholar] [CrossRef]
- Petrosillo, N.; Viceconte, G.; Ergonul, O.; Ippolito, G.; Petersen, E. COVID-19, SARS and MERS: Are they closely related? Clin. Microbiol. Infect. 2020, 26, 729–734. [Google Scholar] [CrossRef]
- Kim, C.H. SARS-CoV-2 Evolutionary Adaptation toward Host Entry and Recognition of Receptor O-Acetyl Sialylation in Virus-Host Interaction. Int. J. Mol. Sci. 2020, 21, 4549. [Google Scholar] [CrossRef]
- Yang, L.; Han, Y.; Nilsson-Payant, B.E.; Gupta, V.; Wang, P.; Duan, X.; Tang, X.; Zhu, J.; Zhao, Z.; Jaffre, F.; et al. A Human Pluripotent Stem Cell-based Platform to Study SARS-CoV-2 Tropism and Model Virus Infection in Human Cells and Organoids. Cell Stem. Cell 2020, 27, 125–136. [Google Scholar] [CrossRef]
- Seyran, M.; Takayama, K.; Uversky, V.N.; Lundstrom, K.; Palu, G.; Sherchan, S.P.; Attrish, D.; Rezaei, N.; Aljabali, A.A.A.; Ghosh, S.; et al. The structural basis of accelerated host cell entry by SARS-CoV-2dagger. FEBS J. 2020. [Google Scholar] [CrossRef] [PubMed]
- 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. Agents. 2020, 55, 105960. [Google Scholar] [CrossRef] [PubMed]
- Jobe, A.; Vijayan, R. Neuropilins: C-end rule peptides and their association with nociception and COVID-19. Comput. Struct. Biotechnol. J. 2021. [Google Scholar] [CrossRef] [PubMed]
- Cantuti-Castelvetri, L.; Ojha, R.; Pedro, L.D.; Djannatian, M.; Franz, J.; Kuivanen, S.; van der Meer, F.; Kallio, K.; Kaya, T.; Anastasina, M.; et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science 2020, 370, 856–860. [Google Scholar] [CrossRef]
- Kofler, N.; Simons, M. The expanding role of neuropilin: Regulation of transforming growth factor-beta and platelet-derived growth factor signaling in the vasculature. Curr. Opin. Hematol. 2016, 23, 260–267. [Google Scholar] [CrossRef] [Green Version]
- Amraie, R.; Napoleon, M.A.; Yin, W.; Berrigan, J.; Suder, E.; Zhao, G.; Olejnik, J.; Gummuluru, S.; Muhlberger, E.; Chitalia, V.; et al. CD209L/L-SIGN and CD209/DC-SIGN act as receptors for SARS-CoV-2 and are differentially expressed in lung and kidney epithelial and endothelial cells. bioRxiv 2020. [Google Scholar] [CrossRef]
- Kielian, M. Enhancing host cell infection by SARS-CoV-2. Science 2020, 370, 765–766. [Google Scholar] [CrossRef]
- Yu, Y.; Bruzdoski, K.; Kostousov, V.; Hensch, L.; Hui, S.K.; Siddiqui, F.; Farooqui, A.; Kouta, A.; Zhang, F.; Fareed, J.; et al. Structural characterization of a clinically described heparin-like substance in plasma causing bleeding. Carbohydr. Polym. 2020, 244, 116443. [Google Scholar] [CrossRef]
- Gue, Y.X.; Gorog, D.A. Reduction in ACE2 may mediate the prothrombotic phenotype in COVID-19. Eur. Heart J. 2020, 41, 3198–3199. [Google Scholar] [CrossRef]
- Salah, H.M.; Mehta, J.L. Heparan sulfate consumption as a potential mechanism of intra-cardiac thrombosis in SARS-CoV-2 infection. Heart Lung 2021, 50, 242–243. [Google Scholar] [CrossRef]
- Lin, L.; Lu, L.; Cao, W.; Li, T. Hypothesis for potential pathogenesis of SARS-CoV-2 infection-a review of immune changes in patients with viral pneumonia. Emerg. Microbes. Infect. 2020, 9, 727–732. [Google Scholar] [CrossRef] [Green Version]
- Russo, V.; Cardillo, G.; Viggiano, G.V.; Mangiacapra, S.; Cavalli, A.; Fontanella, A.; Agrusta, F.; Bellizzi, A.; Amitrano, M.; Iannuzzo, M.; et al. Fondaparinux Use in Patients With COVID-19: A Preliminary Multicenter Real-World Experience. J. Cardiovasc. Pharmacol. 2020, 76, 369–371. [Google Scholar] [CrossRef]
- Tang, N.; Bai, H.; Chen, X.; Gong, J.; Li, D.; Sun, Z. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J. Thromb. Haemost. 2020, 18, 1094–1099. [Google Scholar] [CrossRef]
- Seffer, M.T.; Cottam, D.; Forni, L.G.; Kielstein, J.T. Heparin 2.0: A New Approach to the Infection Crisis. Blood Purif. 2021, 50, 28–34. [Google Scholar] [CrossRef]
- Carvacho, I.; Piesche, M. RGD-binding integrins and TGF-beta in SARS-CoV-2 infections—Novel targets to treat COVID-19 patients? Clin. Transl. Immunol. 2021, 10, e1240. [Google Scholar] [CrossRef]
- Meszaros, B.; Samano-Sanchez, H.; Alvarado-Valverde, J.; Calyseva, J.; Martinez-Perez, E.; Alves, R.; Shields, D.C.; Kumar, M.; Rippmann, F.; Chemes, L.B.; et al. Short linear motif candidates in the cell entry system used by SARS-CoV-2 and their potential therapeutic implications. Sci. Signal. 2021, 14, 655. [Google Scholar] [CrossRef]
- Zhu, X.; Song, Z.; Zhang, S.; Nanda, A.; Li, G. CD147: A novel modulator of inflammatory and immune disorders. Curr. Med. Chem. 2014, 21, 2138–2145. [Google Scholar] [CrossRef]
- Gautret, P.; Lagier, J.C.; Parola, P.; Hoang, V.T.; Meddeb, L.; Mailhe, M.; Doudier, B.; Courjon, J.; Giordanengo, V.; Vieira, V.E.; et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: Results of an open-label non-randomized clinical trial. Int. J. Antimicrob. Agents 2020, 56, 105949. [Google Scholar] [CrossRef]
- Crosnier, C.; Bustamante, L.Y.; Bartholdson, S.J.; Bei, A.K.; Theron, M.; Uchikawa, M.; Mboup, S.; Ndir, O.; Kwiatkowski, D.P.; Duraisingh, M.T.; et al. Basigin is a receptor essential for erythrocyte invasion by Plasmodium falciparum. Nature 2011, 480, 534–537. [Google Scholar] [CrossRef] [Green Version]
- Gadanec, L.K.; McSweeney, K.R.; Qaradakhi, T.; Ali, B.; Zulli, A.; Apostolopoulos, V. Can SARS-CoV-2 Virus Use Multiple Receptors to Enter Host Cells? Int. J. Mol. Sci. 2021, 22, 922. [Google Scholar] [CrossRef]
- Palmeira, A.; Sousa, E.; Koseler, A.; Sabirli, R.; Goren, T.; Turkcuer, I.; Kurt, O.; Pinto, M.M.; Vasconcelos, M.H. Preliminary Virtual Screening Studies to Identify GRP78 Inhibitors Which May Interfere with SARS-CoV-2 Infection. Pharmaceuticals 2020, 13, 132. [Google Scholar] [CrossRef] [PubMed]
- Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef] [PubMed]
- Shi, Y.; Wang, Y.; Shao, C.; Huang, J.; Gan, J.; Huang, X.; Bucci, E.; Piacentini, M.; Ippolito, G.; Melino, G. COVID-19 infection: The perspectives on immune responses. Cell Death Differ. 2020, 27, 1451–1454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, Y.; Fu, B.; Zheng, X.; Wang, D.; Zhao, C.; Qi, Y.; Sun, R.; Tian, Z.; Xu, X.; Wei, H. Pathogenic T-cells and inflammatory monocytes incite inflammatory storms in severe COVID-19 patients. Natl. Sci. Rev. 2020, 7, 998–1002. [Google Scholar] [CrossRef] [Green Version]
- Costela-Ruiz, V.J.; Illescas-Montes, R.; Puerta-Puerta, J.M.; Ruiz, C.; Melguizo-Rodriguez, L. SARS-CoV-2 infection: The role of cytokines in COVID-19 disease. Cytokine Growth Factor Rev. 2020, 54, 62–75. [Google Scholar] [CrossRef]
- Alexopoulou, L.; Holt, A.C.; Medzhitov, R.; Flavell, R.A. Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature 2001, 413, 732–738. [Google Scholar] [CrossRef]
- Wu, J.; Chen, Z.J. Innate immune sensing and signaling of cytosolic nucleic acids. Annu. Rev. Immunol. 2014, 32, 461–488. [Google Scholar] [CrossRef]
- Giamarellos-Bourboulis, E.J.; Netea, M.G.; Rovina, N.; Akinosoglou, K.; Antoniadou, A.; Antonakos, N.; Damoraki, G.; Gkavogianni, T.; Adami, M.E.; Katsaounou, P.; et al. Complex Immune Dysregulation in COVID-19 Patients with Severe Respiratory Failure. Cell Host Microbe. 2020, 27, 992–1000. [Google Scholar] [CrossRef]
- Mehta, M.; Purpura, L.J.; McConville, T.H.; Neidell, M.J.; Anderson, M.R.; Bernstein, E.J.; Dietz, D.E.; Laracy, J.; Gunaratne, S.H.; Miller, E.H.; et al. What about tocilizumab? A retrospective study from a NYC Hospital during the COVID-19 outbreak. PLoS ONE 2021, 16, e0249349. [Google Scholar] [CrossRef]
- Jagannathan, P.; Andrews, J.R.; Bonilla, H.; Hedlin, H.; Jacobson, K.B.; Balasubramanian, V.; Purington, N.; Kamble, S.; de Vries, C.R.; Quintero, O.; et al. Peginterferon Lambda-1a for treatment of outpatients with uncomplicated COVID-19: A randomized placebo-controlled trial. Nat. Commun. 2021, 12, 1967. [Google Scholar] [CrossRef]
- Della-Torre, E.; Lanzillotta, M.; Campochiaro, C.; Cavalli, G.; De Luca, G.; Tomelleri, A.; Boffini, N.; De Lorenzo, R.; Ruggeri, A.; Rovere-Querini, P.; et al. Respiratory Impairment Predicts Response to IL-1 and IL-6 Blockade in COVID-19 Patients With Severe Pneumonia and Hyper-Inflammation. Front. Immunol. 2021, 12, 675678. [Google Scholar] [CrossRef]
- Pontali, E.; Volpi, S.; Signori, A.; Antonucci, G.; Castellaneta, M.; Buzzi, D.; Montale, A.; Bustaffa, M.; Angelelli, A.; Caorsi, R.; et al. Efficacy of early anti-inflammatory treatment with high doses of intravenous anakinra with or without glucocorticoids in patients with severe COVID-19 pneumonia. J. Allergy Clin. Immunol. 2021, 147, 1217–1225. [Google Scholar] [CrossRef]
- Group, R.C.; Horby, P.; Lim, W.S.; Emberson, J.R.; Mafham, M.; Bell, J.L.; Linsell, L.; Staplin, N.; Brightling, C.; Ustianowski, A.; et al. Dexamethasone in Hospitalized Patients with Covid-19. N. Engl. J. Med. 2021, 384, 693–704. [Google Scholar]
- Castelli, V.; Cimini, A.; Ferri, C. Cytokine Storm in COVID-19: “When You Come Out of the Storm, You Won’t Be the Same Person Who Walked in”. Front. Immunol. 2020, 11, 2132. [Google Scholar] [CrossRef]
- Hasan, S.S.; Capstick, T.; Zaidi, S.T.R.; Kow, C.S.; Merchant, H.A. Use of corticosteroids in asthma and COPD patients with or without COVID-19. Respir. Med. 2020, 170, 106045. [Google Scholar] [CrossRef]
- Ramakrishnan, S.; Nicolau, D.V., Jr.; Langford, B.; Mahdi, M.; Jeffers, H.; Mwasuku, C.; Krassowska, K.; Fox, R.; Binnian, I.; Glover, V.; et al. Inhaled budesonide in the treatment of early COVID-19 (STOIC): A phase 2, open-label, randomised controlled trial. Lancet Respir. Med. 2021. [Google Scholar] [CrossRef]
- Rothan, H.A.; Bahrani, H.; Mohamed, Z.; Teoh, T.C.; Shankar, E.M.; Rahman, N.A.; Yusof, R. A combination of doxycycline and ribavirin alleviated chikungunya infection. PLoS ONE 2015, 10, e0126360. [Google Scholar] [CrossRef] [Green Version]
- Mosquera-Sulbaran, J.A.; Hernandez-Fonseca, H. Tetracycline and viruses: A possible treatment for COVID-19? Arch. Virol. 2021, 166, 1–7. [Google Scholar] [CrossRef]
- Chakraborti, S.; Bheemireddy, S.; Srinivasan, N. Repurposing drugs against the main protease of SARS-CoV-2: Mechanism-based insights supported by available laboratory and clinical data. Mol. Omics. 2020, 16, 474–491. [Google Scholar] [CrossRef]
- Wu, C.; Liu, Y.; Yang, Y.; Zhang, P.; Zhong, W.; Wang, Y.; Wang, Q.; Xu, Y.; Li, M.; Li, X.; et al. Analysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methods. Acta Pharm. Sin. B 2020, 10, 766–788. [Google Scholar] [CrossRef]
- McGonagle, D.; O’Donnell, J.S.; Sharif, K.; Emery, P.; Bridgewood, C. Immune mechanisms of pulmonary intravascular coagulopathy in COVID-19 pneumonia. Lancet Rheumatol. 2020, 2, e437–e445. [Google Scholar] [CrossRef]
- Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
- Kipshidze, N.; Dangas, G.; White, C.J.; Kipshidze, N.; Siddiqui, F.; Lattimer, C.R.; Carter, C.A.; Fareed, J. Viral Coagulopathy in Patients With COVID-19: Treatment and Care. Clin. Appl. Thromb. Hemost. 2020, 26, 1076029620936776. [Google Scholar] [CrossRef]
- Zuo, Y.; Estes, S.K.; Ali, R.A.; Gandhi, A.A.; Yalavarthi, S.; Shi, H.; Sule, G.; Gockman, K.; Madison, J.A.; Zuo, M.; et al. Prothrombotic autoantibodies in serum from patients hospitalized with COVID-19. Sci. Transl. Med. 2020, 12, 570. [Google Scholar] [CrossRef]
- Butenas, S.; Mann, K.G. Blood coagulation. Biochemistry 2002, 67, 3–12. [Google Scholar]
- Moschonas, I.C.; Tselepis, A.D. SARS-CoV-2 infection and thrombotic complications: A narrative review. J. Thromb. Thrombolysis 2021. [Google Scholar] [CrossRef]
- Song, E.; Zhang, C.; Israelow, B.; Lu-Culligan, A.; Prado, A.V.; Skriabine, S.; Lu, P.; Weizman, O.E.; Liu, F.; Dai, Y.; et al. Neuroinvasion of SARS-CoV-2 in human and mouse brain. J. Exp. Med. 2021, 218, e20202135. [Google Scholar] [CrossRef]
- Xiao, F.; Tang, M.; Zheng, X.; Liu, Y.; Li, X.; Shan, H. Evidence for Gastrointestinal Infection of SARS-CoV-2. Gastroenterology 2020, 158, 1831–1833. [Google Scholar] [CrossRef]
- Wang, C.; Li, W.; Drabek, D.; Okba, N.M.A.; van Haperen, R.; Osterhaus, A.; van Kuppeveld, F.J.M.; Haagmans, B.L.; Grosveld, F.; Bosch, B.J. A human monoclonal antibody blocking SARS-CoV-2 infection. Nat. Commun. 2020, 11, 2251. [Google Scholar] [CrossRef]
- Silva, T.F.; Tomiotto-Pellissier, F.; Sanfelice, R.A.; Goncalves, M.D.; da Silva Bortoleti, B.T.; Detoni, M.B.; Rodrigues, A.C.J.; Carloto, A.C.M.; Concato, V.M.; Siqueira, E.D.S.; et al. A 21st Century Evil: Immunopathology and New Therapies of COVID-19. Front. Immunol. 2020, 11, 562264. [Google Scholar] [CrossRef]
- Llitjos, J.F.; Leclerc, M.; Chochois, C.; Monsallier, J.M.; Ramakers, M.; Auvray, M.; Merouani, K. High incidence of venous thromboembolic events in anticoagulated severe COVID-19 patients. J. Thromb. Haemost. 2020, 18, 1743–1746. [Google Scholar] [CrossRef] [PubMed]
- Merrill, J.T.; Erkan, D.; Winakur, J.; James, J.A. Emerging evidence of a COVID-19 thrombotic syndrome has treatment implications. Nat. Rev. Rheumatol. 2020, 16, 581–589. [Google Scholar] [CrossRef] [PubMed]
- Cuker, A.; Tseng, E.K.; Nieuwlaat, R.; Angchaisuksiri, P.; Blair, C.; Dane, K.; Davila, J.; DeSancho, M.T.; Diuguid, D.; Griffin, D.O.; et al. American Society of Hematology 2021 guidelines on the use of anticoagulation for thromboprophylaxis in patients with COVID-19. Blood Adv. 2021, 5, 872–888. [Google Scholar] [CrossRef] [PubMed]
- Group, The UPMC REMAP-COVID; REMAP-CAP Investigators. Implementation of the Randomized Embedded Multifactorial Adaptive Platform for COVID-19 (REMAP-COVID) trial in a US health system-lessons learned and recommendations. Trials 2021, 22, 100. [Google Scholar]
- Flumignan, R.L.; Tinoco, J.D.S.; Pascoal, P.I.; Areias, L.L.; Cossi, M.S.; Fernandes, M.I.; Costa, I.K.; Souza, L.; Matar, C.F.; Tendal, B.; et al. Prophylactic anticoagulants for people hospitalised with COVID-19. Cochrane Database Syst. Rev. 2020, 10, CD013739. [Google Scholar]
- Akizawa, T.; Koshikawa, S.; Ota, K.; Kazama, M.; Mimura, N.; Hirasawa, Y. Nafamostat mesilate: A regional anticoagulant for hemodialysis in patients at high risk for bleeding. Nephron 1993, 64, 376–381. [Google Scholar] [CrossRef]
- Park, T.K.; Yang, J.H.; Jeon, K.; Choi, S.H.; Choi, J.H.; Gwon, H.C.; Chung, C.R.; Park, C.M.; Cho, Y.H.; Sung, K.; et al. Extracorporeal membrane oxygenation for refractory septic shock in adults. Eur. J. Cardiothorac. Surg. 2015, 47, e68–e74. [Google Scholar] [CrossRef] [Green Version]
- Kobayashi, T.; Terao, T.; Maki, M.; Ikenoue, T. Diagnosis and management of acute obstetrical DIC. Semin. Thromb. Hemost. 2001, 27, 161–167. [Google Scholar] [CrossRef]
- Paccaly, A.; Frick, A.; Rohatagi, S.; Liu, J.; Shukla, U.; Rosenburg, R.; Hinder, M.; Jensen, B.K. Pharmacokinetics of otamixaban, a direct factor Xa inhibitor, in healthy male subjects: Pharmacokinetic model development for phase 2/3 simulation of exposure. J. Clin. Pharmacol. 2006, 46, 37–44. [Google Scholar] [CrossRef]
- Stangier, J.; Clemens, A. Pharmacology, pharmacokinetics, and pharmacodynamics of dabigatran etexilate, an oral direct thrombin inhibitor. Clin. Appl. Thromb. Hemost. 2009, 15 (Suppl. 1), 9S–16S. [Google Scholar] [CrossRef]
- Liu, C.L.; Guo, J.; Zhang, X.; Sukhova, G.K.; Libby, P.; Shi, G.P. Cysteine protease cathepsins in cardiovascular disease: From basic research to clinical trials. Nat. Rev. Cardiol. 2018, 15, 351–370. [Google Scholar] [CrossRef]
SARS-CoV-2 | SARS-CoV | MERS-CoV | |
---|---|---|---|
Receptor | ACE2 [10] | ACE2 [12] | DPP4 (CD26) [10] |
Priming protease | TMPRSS2 [13] | TMPRSS2 [12] | |
Furin [14] | Furin [15] | TMPRSS2 [16] | |
Cathepsin-L [16] | Cathepsin-L [17] | Furin [16] | |
ADAM17 [17] | ADAM 17 [12] | Cathepsin-L [18] | |
Factor Xa [19] | Factor Xa [20] | ||
Co-receptors | Sialic Acids [21] | ||
NRP-1 [22] | Integrins [23] | Sialic Acids [21] | |
Integrins [24] | Heparan Sulfate [25] | ||
Heparan Sulfate [26] | |||
Other receptors | CD147 [27] | CD147 [17] | GRP78 [28] |
GRP78 [28] | GRP78 [28] |
Targets | Potential Drugs | Mechanism of Action | Refs |
---|---|---|---|
Furin | ambroxol, bromhexine and luteolin tamarind, polyphenols, limonin and gedunin | Block the S-protein cleavage activation and membrane fusion | [13,46,47] |
TMPRSS2 | camostat, nafamostat, limonin, gedunin, otamixaban dabigatran and α-1-antitrypsin | Block S-protein cleavage mediated not only by TMPRSS2 but also by other proteases | [13,48,49,50] |
ADAM17 | α-1-antitrypsin | Blocks S-protein cleavage | [50] |
Syncytia formation | niclosamide, trifluoperazine, serotonin reuptake inhibitors, ivermectin | Suppress the activity of TMEM16F, involved in syncytia formation | [51,52] |
Cathepsin L | chloroquine, hydroxychloroquine and 8P9R | Interfere with the endosomial pathway, increasing pH | [53,54,55,56] |
NRP-1 | heparin, natural products and small molecules | Block C-end rule peptide on NRP-1 | [57] |
Heparan sulfate | heparin/HS mimetics | Preclude the interaction between HS and S protein | [58,59] |
Integrins | ATN-161 | Inhibits the interaction between S protein and integrins | [60] |
CD147 | azithromycin, cyclophilin A, doxycycline | Interfere with ligands/CD147 interaction | [61,62,63,64] |
GRP78 | epigallocatechin gallate, omoeriodictyol, isorhamnetin, and curcumin, berbamine, OSU-03012 | Interfere with ligands/GRP78 interaction | [65,66] |
Factor Xa | heparin | Blocks S protein cleavage by Factor Xa and inhibits the coagulation | [20] |
Cyclo-oxygenase-2 | non-steroidal anti-inflammatory drugs | Prevent inflammatory cytokine storm | [67] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Vianello, A.; Del Turco, S.; Babboni, S.; Silvestrini, B.; Ragusa, R.; Caselli, C.; Melani, L.; Fanucci, L.; Basta, G. The Fight against COVID-19 on the Multi-Protease Front and Surroundings: Could an Early Therapeutic Approach with Repositioning Drugs Prevent the Disease Severity? Biomedicines 2021, 9, 710. https://doi.org/10.3390/biomedicines9070710
Vianello A, Del Turco S, Babboni S, Silvestrini B, Ragusa R, Caselli C, Melani L, Fanucci L, Basta G. The Fight against COVID-19 on the Multi-Protease Front and Surroundings: Could an Early Therapeutic Approach with Repositioning Drugs Prevent the Disease Severity? Biomedicines. 2021; 9(7):710. https://doi.org/10.3390/biomedicines9070710
Chicago/Turabian StyleVianello, Annamaria, Serena Del Turco, Serena Babboni, Beatrice Silvestrini, Rosetta Ragusa, Chiara Caselli, Luca Melani, Luca Fanucci, and Giuseppina Basta. 2021. "The Fight against COVID-19 on the Multi-Protease Front and Surroundings: Could an Early Therapeutic Approach with Repositioning Drugs Prevent the Disease Severity?" Biomedicines 9, no. 7: 710. https://doi.org/10.3390/biomedicines9070710
APA StyleVianello, A., Del Turco, S., Babboni, S., Silvestrini, B., Ragusa, R., Caselli, C., Melani, L., Fanucci, L., & Basta, G. (2021). The Fight against COVID-19 on the Multi-Protease Front and Surroundings: Could an Early Therapeutic Approach with Repositioning Drugs Prevent the Disease Severity? Biomedicines, 9(7), 710. https://doi.org/10.3390/biomedicines9070710