Could a Lower Toll-like Receptor (TLR) and NF-κB Activation Due to a Changed Charge Distribution in the Spike Protein Be the Reason for the Lower Pathogenicity of Omicron?
Abstract
:1. Introduction
2. SARS-CoV-2 and COVID-19—The Virus and the Disease
3. Molecular Changes in the Omicron’s Spike Protein and Their Impact on Transmissibility, Immune Escape, and Pathogenicity
3.1. Mutations in the Omicron Spike Protein RBD Region Strengthen the Spike-ACE2 Interaction
3.2. Enhanced Escape from Therapeutic Antibodies and Immune Sera by Mutations in RBD and NTD
3.3. Mutations in S2
3.4. Mutations near to the S1/S2 Furin-Like Cleavage Site
4. Lower Pathogenicity of Omicron Compared to Previous VoCs
4.1. NF-κB Pathway Activation by SARS-CoV-2
4.2. SARS-CoV-2 Spike Protein Induces NF-κB
4.3. NF-κB Is Essential for SARS-CoV-2 Replication
4.4. NF-κB, Cytokines, and Hypoxia Enhance Furin Expression
4.5. Acute Viral Infections Such as Highly Pathogenic IAV and SARS-CoV-2 Depend on Furin and Stimulate Furin Expression
5. SARS-CoV-2 Activates Innate PRRs
5.1. SARS-CoV-2 Envelop E and Spike Protein Activate TLR2 and NF-κB
5.2. SARS-CoV-2 Spike Protein Activates TLR4 and NF-κB
5.3. TLR Activation during Different Highly Pathogenic Viral Infections
5.4. Activation of TLR4 by LPS
6. Discussion of an Integrated Mechanistic Model
- (1)
- Different components of SARS-CoV-2, in particular of the spike protein, have been demonstrated to activate TLRs, in particular TLR4 and TLR2.
- (2)
- Dimerization represents the general principle underlying the activation of TLRs, with activating PAMPs serving as molecular linkers promoting dimerization.
- (3)
- For dimerization, there is a minimal number of hydrophobic chains necessary which have to fit into hydrophobic pockets in order to provide sufficient hydrophobic interactions, as demonstrated for TLR4-MD-2, TLR2-TLR1, and TLR2-TLR6 complexes, respectively.
- (4)
- There is a common feature of the TLR activating viral glycoproteins (also including the SARS-CoV-2 spike protein) with all of them being membrane-bound proteins which contain hydrophobic domains necessary for fusion with the host cell membrane and having the potential to interact with the hydrophobic pockets of TLR complexes.
- (5)
- Negatively charged groups have been shown to be essential for dimerization, as illustrated for TLR4-MD-2/LPS complexes.
- (6)
- Interaction and dimerization of respective TLR complexes triggers the inherent downstream signaling pathways, mainly the NF-κB pathway.
- –
- Some hydrophobic domains of the SARS-CoV-2 spike protein can interact with the hydrophobic pockets of TLR-complexes leading to dimerization and activation. In particular, the hydrophobic six-helix bundle fusion core structure (6HB) in the post- fusion state of the SARS-CoV-2 spike protein can be hypothesized to fit into the hydrophobic pockets of MD-2-TLR4. Other hydrophobic domains of the spike protein, such as the three hydrophobic stretches in the S2 subunit of the trimer in prefusion state, may be speculated to fit for binding to TLR2-TLR1/6 complexes.
- –
- Distribution of charged amino acids can greatly affect binding to and dimerization of TLR complexes. The changed charge distribution on the Omicron spike protein with high accumulations of positively charged amino acid residues in the RBD and in the S2 subunit, together with loss of several negatively charged amino acids by substitutions in the Omicron spike protein, may prevent high affinity binding to the TLR complexes and/or insufficient dimerization of TLR complexes, leading to lower downstream signaling and lower pro-inflammatory activation, lower NF-kB activation, and related lower furin expression. Indeed, a lower NF-κB activation by the Omicron variant vs. a whole panel of previous variants including the D614G, Delta, Lambda, and Mu variant has been shown recently [187].
How Will this Impact the Virus Replication, Cellular Tropism, and Pathogenicity?
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Acknowledgments
Conflicts of Interest
References
- Viana, R.; Moyo, S.; Amoako, D.G.; Tegally, H.; Scheepers, C.; Althaus, C.L.; Anyaneji, U.J.; Bester, P.A.; Boni, M.F.; Chand, M.; et al. Rapid epidemic expansion of the SARS-CoV-2 Omicron variant in southern Africa. Nature 2022, 603, 679–686. [Google Scholar] [CrossRef]
- Dejnirattisai, W.; Huo, J.; Zhou, D.; Zahradnik, J.; Supasa, P.; Liu, C.; Duyvesteyn, H.M.E.; Ginn, H.M.; Mentzer, A.J.; Tuekprakhon, A.; et al. SARS-CoV-2 Omicron-B.1.1.529 leads to widespread escape from neutralizing antibody responses. Cell 2022, 185, 467–484.e15. [Google Scholar] [CrossRef]
- Meng, B.; Abdullahi, A.; Ferreira, I.; Goonawardane, N.; Saito, A.; Kimura, I.; Yamasoba, D.; Gerber, P.P.; Fatihi, S.; Rathore, S.; et al. Altered TMPRSS2 usage by SARS-CoV-2 Omicron impacts infectivity and fusogenicity. Nature 2022, 603, 706–714. [Google Scholar] [CrossRef]
- Kannan, S.R.; Spratt, A.N.; Sharma, K.; Chand, H.S.; Byrareddy, S.N.; Singh, K. Omicron SARS-CoV-2 variant: Unique features and their impact on pre-existing antibodies. J. Autoimmun. 2022, 126, 102779. [Google Scholar] [CrossRef]
- Ye, G.; Liu, B.; Li, F. Cryo-EM structure of a SARS-CoV-2 omicron spike protein ectodomain. Nat. Commun. 2022, 13, 1214. [Google Scholar] [CrossRef]
- Shah, M.; Woo, H.G. Omicron: A Heavily Mutated SARS-CoV-2 Variant Exhibits Stronger Binding to ACE2 and Potently Escapes Approved COVID-19 Therapeutic Antibodies. Front. Immunol. 2021, 12, 830527. [Google Scholar] [CrossRef]
- Halfmann, P.J.; Iida, S.; Iwatsuki-Horimoto, K.; Maemura, T.; Kiso, M.; Scheaffer, S.M.; Darling, T.L.; Joshi, A.; Loeber, S.; Singh, G.; et al. SARS-CoV-2 Omicron virus causes attenuated disease in mice and hamsters. Nature 2022, 603, 687–692. [Google Scholar] [CrossRef]
- Wolter, N.; Jassat, W.; Walaza, S.; Welch, R.; Moultrie, H.; Groome, M.; Amoako, D.G.; Everatt, J.; Bhiman, J.N.; Scheepers, C.; et al. Early assessment of the clinical severity of the SARS-CoV-2 omicron variant in South Africa: A data linkage study. Lancet 2022, 399, 437–446. [Google Scholar] [CrossRef]
- Willett, B.J.; Grove, J.; MacLean, O.A.; Wilkie, C.; Logan, N.; Lorenzo, G.D.; Furnon, W.; Scott, S.; Manali, M.; Szemiel, A.; et al. The hyper-transmissible SARS-CoV-2 Omicron variant exhibits significant antigenic change, vaccine escape and a switch in cell entry mechanism. medRxiv 2022. [Google Scholar] [CrossRef]
- Abdelrahman, Z.; Li, M.; Wang, X. Comparative Review of SARS-CoV-2, SARS-CoV, MERS-CoV, and Influenza A Respiratory Viruses. Front. Immunol. 2020, 11, 552909. [Google Scholar] [CrossRef]
- Eslami, N.; Aghbash, P.S.; Shamekh, A.; Entezari-Maleki, T.; Nahand, J.S.; Sales, A.J.; Baghi, H.B. SARS-CoV-2: Receptor and Co-receptor Tropism Probability. Curr Microbiol. 2022, 79, 133. [Google Scholar] [CrossRef]
- 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]
- Matsuyama, S.; Ujike, M.; Morikawa, S.; Tashiro, M.; Taguchi, F. Protease-mediated enhancement of severe acute respiratory syndrome coronavirus infection. Proc. Natl. Acad. Sci. USA 2005, 102, 12543–12547. [Google Scholar] [CrossRef] [Green Version]
- Simmons, G.; Gosalia, D.N.; Rennekamp, A.J.; Reeves, J.D.; Diamond, S.L.; Bates, P. Inhibitors of cathepsin L prevent severe acute respiratory syndrome coronavirus entry. Proc. Natl. Acad. Sci. USA 2005, 102, 11876–11881. [Google Scholar] [CrossRef] [Green Version]
- Belouzard, S.; Millet, J.K.; Licitra, B.N.; Whittaker, G.R. Mechanisms of coronavirus cell entry mediated by the viral spike protein. Viruses 2012, 4, 1011–1033. [Google Scholar] [CrossRef] [Green Version]
- Shah, M.; Woo, H.G. Molecular Perspectives of SARS-CoV-2: Pathology, Immune Evasion, and Therapeutic Interventions. Mol. Cell 2021, 44, 408–421. [Google Scholar] [CrossRef]
- Watanabe, Y.; Allen, J.D.; Wrapp, D.; McLellan, J.S.; Crispin, M. Site-specific glycan analysis of the SARS-CoV-2 spike. Science 2020, 369, 330–333. [Google Scholar] [CrossRef]
- Shajahan, A.; Pepi, L.E.; Rouhani, D.S.; Heiss, C.; Azadi, P. Glycosylation of SARS-CoV-2: Structural and functional insights. Anal. Bioanal. Chem. 2021, 413, 7179–7193. [Google Scholar] [CrossRef]
- Xia, S.; Zhu, Y.; Liu, M.; Lan, Q.; Xu, W.; Wu, Y.; Ying, T.; Liu, S.; Shi, Z.; Jiang, S.; et al. Fusion mechanism of 2019-nCoV and fusion inhibitors targeting HR1 domain in spike protein. Cell. Mol. Immunol. 2020, 17, 765–767. [Google Scholar] [CrossRef]
- Sah, P.; Fitzpatrick, M.C.; Zimmer, C.F.; Abdollahi, E.; Juden-Kelly, L.; Moghadas, S.M.; Singer, B.H.; Galvani, A.P. Asymptomatic SARS-CoV-2 infection: A systematic review and meta-analysis. Proc. Natl. Acad. Sci. USA 2021, 118, e2109229118. [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.e8. [Google Scholar] [CrossRef]
- Varga, Z.; Flammer, A.J.; Steiger, P.; Haberecker, M.; Andermatt, R.; Zinkernagel, A.S.; Mehra, M.R.; Schuepbach, R.A.; Ruschitzka, F.; Moch, H. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020, 395, 1417–1418. [Google Scholar] [CrossRef]
- Ackermann, M.; Verleden, S.E.; Kuehnel, M.; Haverich, A.; Welte, T.; Laenger, F.; Vanstapel, A.; Werlein, C.; Stark, H.; Tzankov, A.; et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N. Engl. J. Med. 2020, 383, 120–128. [Google Scholar] [CrossRef]
- Sokolowska, M.; Lukasik, Z.M.; Agache, I.; Akdis, C.A.; Akdis, D.; Akdis, M.; Barcik, W.; Brough, H.A.; Eiwegger, T.; Eljaszewicz, A.; et al. Immunology of COVID-19: Mechanisms, clinical outcome, diagnostics, and perspectives-A report of the European Academy of Allergy and Clinical Immunology (EAACI). Allergy 2020, 75, 2445–2476. [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]
- Xu, Z.; Shi, L.; Wang, Y.; Zhang, J.; Huang, L.; Zhang, C.; Liu, S.; Zhao, P.; Liu, H.; Zhu, L.; et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020, 8, 420–422. [Google Scholar] [CrossRef]
- Wang, D.; Hu, B.; Hu, C.; Zhu, F.; Liu, X.; Zhang, J.; Wang, B.; Xiang, H.; Cheng, Z.; Xiong, Y.; et al. Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. J. Am. Med. Assoc. 2020, 323, 1061–1069. [Google Scholar] [CrossRef]
- Zheng, Z.; Peng, F.; Xu, B.; Zhao, J.; Liu, H.; Peng, J.; Li, Q.; Jiang, C.; Zhou, Y.; Liu, S.; et al. Risk factors of critical & mortal COVID-19 cases: A systematic literature review and meta-analysis. J. Infect. 2020, 81, e16–e25. [Google Scholar]
- Biswas, M.; Rahaman, S.; Biswas, T.K.; Haque, Z.; Ibrahim, B. Association of Sex, Age, and Comorbidities with Mortality in COVID-19 Patients: A Systematic Review and Meta-Analysis. Intervirology 2021, 64, 36–47. [Google Scholar] [CrossRef]
- Kang, S.J.; Jung, S.I. Age-Related Morbidity and Mortality among Patients with COVID-19. J. Infect. Chemother. 2020, 52, 154–164. [Google Scholar] [CrossRef]
- O’Driscoll, M.; Ribeiro Dos Santos, G.; Wang, L.; Cummings, D.A.T.; Azman, A.S.; Paireau, J.; Fontanet, A.; Cauchemez, S.; Salje, H. Age-specific mortality and immunity patterns of SARS-CoV-2. Nature 2021, 590, 140–145. [Google Scholar] [CrossRef]
- Carsana, L.; Sonzogni, A.; Nasr, A.; Rossi, R.S.; Pellegrinelli, A.; Zerbi, P.; Rech, R.; Colombo, R.; Antinori, S.; Corbellino, M.; et al. Pulmonary post-mortem findings in a series of COVID-19 cases from northern Italy: A two-centre descriptive study. Lancet Infect. Dis. 2020, 20, 1135–1140. [Google Scholar] [CrossRef]
- Acharya, D.; Liu, G.; Gack, M.U. Dysregulation of type I interferon responses in COVID-19. Nat. Rev. Immunol. 2020, 20, 397–398. [Google Scholar] [CrossRef]
- Tay, M.Z.; Poh, C.M.; Renia, L.; MacAry, P.A.; Ng, L.F.P. The trinity of COVID-19: Immunity, inflammation and intervention. Nat. Rev. Immunol. 2020, 20, 363–374. [Google Scholar] [CrossRef]
- Schett, G.; Sticherling, M.; Neurath, M.F. COVID-19: Risk for cytokine targeting in chronic inflammatory diseases? Nat. Rev. Immunol. 2020, 20, 271–272. [Google Scholar] [CrossRef] [Green Version]
- Moore, J.B.; June, C.H. Cytokine release syndrome in severe COVID-19. Science 2020, 368, 473–474. [Google Scholar] [CrossRef] [Green Version]
- Huang, Y.; Tu, M.; Wang, S.; Chen, S.; Zhou, W.; Chen, D.; Zhou, L.; Wang, M.; Zhao, Y.; Zeng, W.; et al. Clinical characteristics of laboratory confirmed positive cases of SARS-CoV-2 infection in Wuhan, China: A retrospective single center analysis. Travel Med. Infect. Dis. 2020, 36, 101606. [Google Scholar] [CrossRef]
- Chua, R.L.; Lukassen, S.; Trump, S.; Hennig, B.P.; Wendisch, D.; Pott, F.; Debnath, O.; Thurmann, L.; Kurth, F.; Volker, M.T.; et al. COVID-19 severity correlates with airway epithelium-immune cell interactions identified by single-cell analysis. Nat. Biotechnol. 2020, 38, 970–979. [Google Scholar] [CrossRef]
- Huang, J.; Hume, A.J.; Abo, K.M.; Werder, R.B.; Villacorta-Martin, C.; Alysandratos, K.D.; Beermann, M.L.; Simone-Roach, C.; Lindstrom-Vautrin, J.; Olejnik, J.; et al. SARS-CoV-2 Infection of Pluripotent Stem Cell-Derived Human Lung Alveolar Type 2 Cells Elicits a Rapid Epithelial-Intrinsic Inflammatory Response. Cell Stem Cell 2020, 27, 962–973.e7. [Google Scholar] [CrossRef]
- Hojyo, S.; Uchida, M.; Tanaka, K.; Hasebe, R.; Tanaka, Y.; Murakami, M.; Hirano, T. How COVID-19 induces cytokine storm with high mortality. Inflamm. Regen. 2020, 40, 37. [Google Scholar] [CrossRef]
- Hirano, T.; Murakami, M. COVID-19: A New Virus, but a Familiar Receptor and Cytokine Release Syndrome. Immunity 2020, 52, 731–733. [Google Scholar] [CrossRef]
- Hong, K.S.; Ahn, J.H.; Jang, J.G.; Lee, J.H.; Kim, H.N.; Kim, D.; Lee, W. GSK-LSD1, an LSD1 inhibitor, quashes SARS-CoV-2-triggered cytokine release syndrome in-vitro. Signal Transduct. Target. Ther. 2020, 5, 267. [Google Scholar] [CrossRef]
- McAleavy, M.; Zhang, Q.; Ehmann, P.J.; Xu, J.; Wipperman, M.F.; Ajithdoss, D.; Pan, L.; Wakai, M.; Simonson, R.; Gadi, A.; et al. The Activin/FLRG Pathway Associates with Poor COVID-19 Outcomes in Hospitalized Patients. Mol. Cell. Biol. 2022, 42, e0046721. [Google Scholar] [CrossRef]
- Hariharan, A.; Hakeem, A.R.; Radhakrishnan, S.; Reddy, M.S.; Rela, M. The Role and Therapeutic Potential of NF-kappa-B Pathway in Severe COVID-19 Patients. Inflammopharmacology 2021, 29, 91–100. [Google Scholar] [CrossRef]
- Kircheis, R.; Haasbach, E.; Lueftenegger, D.; Heyken, W.T.; Ocker, M.; Planz, O. NF-kappaB Pathway as a Potential Target for Treatment of Critical Stage COVID-19 Patients. Front. Immunol. 2020, 11, 598444. [Google Scholar] [CrossRef]
- Neufeldt, C.J.; Cerikan, B.; Cortese, M.; Frankish, J.; Lee, J.Y.; Plociennikowska, A.; Heigwer, F.; Prasad, V.; Joecks, S.; Burkart, S.S.; et al. SARS-CoV-2 infection induces a pro-inflammatory cytokine response through cGAS-STING and NF-kappaB. Commun. Biol. 2022, 5, 45. [Google Scholar] [CrossRef]
- Lo, M.W.; Kemper, C.; Woodruff, T.M. COVID-19: Complement, Coagulation, and Collateral Damage. J. Immunol. 2020, 205, 1488–1495. [Google Scholar] [CrossRef]
- Holter, J.C.; Pischke, S.E.; de Boer, E.; Lind, A.; Jenum, S.; Holten, A.R.; Tonby, K.; Barratt-Due, A.; Sokolova, M.; Schjalm, C.; et al. Systemic complement activation is associated with respiratory failure in COVID-19 hospitalized patients. Proc. Natl. Acad. Sci. USA 2020, 117, 25018–25025. [Google Scholar] [CrossRef]
- Polycarpou, A.; Howard, M.; Farrar, C.A.; Greenlaw, R.; Fanelli, G.; Wallis, R.; Klavinskis, L.S.; Sacks, S. Rationale for targeting complement in COVID-19. EMBO Mol. Med. 2020, 12, e12642. [Google Scholar] [CrossRef]
- Yu, J.; Yuan, X.; Chen, H.; Chaturvedi, S.; Braunstein, E.M.; Brodsky, R.A. Direct activation of the alternative complement pathway by SARS-CoV-2 spike proteins is blocked by factor D inhibition. Blood 2020, 136, 2080–2089. [Google Scholar] [CrossRef]
- Fletcher-Sandersjoo, A.; Bellander, B.M. Is COVID-19 associated thrombosis caused by overactivation of the complement cascade? A literature review. Thromb. Res. 2020, 194, 36–41. [Google Scholar] [CrossRef]
- Mussbacher, M.; Salzmann, M.; Brostjan, C.; Hoesel, B.; Schoergenhofer, C.; Datler, H.; Hohensinner, P.; Basilio, J.; Petzelbauer, P.; Assinger, A.; et al. Cell Type-Specific Roles of NF-kappaB Linking Inflammation and Thrombosis. Front. Immunol. 2019, 10, 85. [Google Scholar] [CrossRef] [Green Version]
- McCarthy, C.G.; Wilczynski, S.; Wenceslau, C.F.; Webb, R.C. A new storm on the horizon in COVID-19: Bradykinin-induced vascular complications. Vascul. Pharmacol. 2021, 137, 106826. [Google Scholar] [CrossRef]
- Leppkes, M.; Knopf, J.; Naschberger, E.; Lindemann, A.; Singh, J.; Herrmann, I.; Sturzl, M.; Staats, L.; Mahajan, A.; Schauer, C.; et al. Vascular occlusion by neutrophil extracellular traps in COVID-19. Ebiomedicine 2020, 58, 102925. [Google Scholar] [CrossRef]
- De Buhr, N.; von Kockritz-Blickwede, M. The Balance of Neutrophil Extracellular Trap Formation and Nuclease Degradation: An Unknown Role of Bacterial Coinfections in COVID-19 Patients? mBio 2021, 12, e03304-20. [Google Scholar] [CrossRef]
- Kvietys, P.R.; Fakhoury, H.M.A.; Kadan, S.; Yaqinuddin, A.; Al-Mutairy, E.; Al-Kattan, K. COVID-19: Lung-Centric Immunothrombosis. Front. Cell. Infect. Microbiol. 2021, 11, 679878. [Google Scholar] [CrossRef]
- Gando, S.; Wada, T. Pathomechanisms Underlying Hypoxemia in Two COVID-19-Associated Acute Respiratory Distress Syndrome Phenotypes: Insights From Thrombosis and Hemostasis. Shock 2022, 57, 1–6. [Google Scholar] [CrossRef]
- Thachil, J.; Tang, N.; Gando, S.; Falanga, A.; Cattaneo, M.; Levi, M.; Clark, C.; Iba, T. ISTH interim guidance on recognition and management of coagulopathy in COVID-19. J. Thromb. Haemost. 2020, 18, 1023–1026. [Google Scholar] [CrossRef]
- Gu, S.X.; Tyagi, T.; Jain, K.; Gu, V.W.; Lee, S.H.; Hwa, J.M.; Kwan, J.M.; Krause, D.S.; Lee, A.I.; Halene, S.; et al. Thrombocytopathy and endotheliopathy: Crucial contributors to COVID-19 thromboinflammation. Nat. Rev. Cardiol. 2021, 18, 194–209. [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, eabd3876. [Google Scholar] [CrossRef]
- Klok, F.A.; Kruip, M.; van der Meer, N.J.M.; Arbous, M.S.; Gommers, D.; Kant, K.M.; Kaptein, F.H.J.; van Paassen, J.; Stals, M.A.M.; Huisman, M.V.; et al. Confirmation of the high cumulative incidence of thrombotic complications in critically ill ICU patients with COVID-19: An updated analysis. Thromb. Res. 2020, 191, 148–150. [Google Scholar] [CrossRef]
- Skendros, P.; Mitsios, A.; Chrysanthopoulou, A.; Mastellos, D.C.; Metallidis, S.; Rafailidis, P.; Ntinopoulou, M.; Sertaridou, E.; Tsironidou, V.; Tsigalou, C.; et al. Complement and tissue factor-enriched neutrophil extracellular traps are key drivers in COVID-19 immunothrombosis. J. Clin. Investig. 2020, 130, 6151–6157. [Google Scholar] [CrossRef]
- George, J.N.; Nester, C.M. Syndromes of thrombotic microangiopathy. N. Engl. J. Med. 2014, 371, 1847–1848. [Google Scholar] [CrossRef] [Green Version]
- Brocklebank, V.; Wood, K.M.; Kavanagh, D. Thrombotic Microangiopathy and the Kidney. Clin. J. Am. Soc. Nephrol. 2018, 13, 300–317. [Google Scholar] [CrossRef] [Green Version]
- Menter, T.; Haslbauer, J.D.; Nienhold, R.; Savic, S.; Hopfer, H.; Deigendesch, N.; Frank, S.; Turek, D.; Willi, N.; Pargger, H.; et al. Postmortem examination of COVID-19 patients reveals diffuse alveolar damage with severe capillary congestion and variegated findings in lungs and other organs suggesting vascular dysfunction. Histopathology 2020, 77, 198–209. [Google Scholar] [CrossRef]
- Gobeil, S.M.; Henderson, R.; Stalls, V.; Janowska, K.; Huang, X.; May, A.; Speakman, M.; Beaudoin, E.; Manne, K.; Li, D.; et al. Structural diversity of the SARS-CoV-2 Omicron spike. bioRxiv, 2022; in press. [Google Scholar]
- Mannar, D.; Saville, J.W.; Zhu, X.; Srivastava, S.S.; Berezuk, A.M.; Tuttle, K.S.; Marquez, A.C.; Sekirov, I.; Subramaniam, S. SARS-CoV-2 Omicron variant: Antibody evasion and cryo-EM structure of spike protein-ACE2 complex. Science 2022, 375, 760–764. [Google Scholar] [CrossRef]
- Yin, W.; Xu, Y.; Xu, P.; Cao, X.; Wu, C.; Gu, C.; He, X.; Wang, X.; Huang, S.; Yuan, Q.; et al. Structures of the Omicron spike trimer with ACE2 and an anti-Omicron antibody. Science 2022, 375, 1048–1053. [Google Scholar] [CrossRef]
- Barton, M.I.; MacGowan, S.A.; Kutuzov, M.A.; Dushek, O.; Barton, G.J.; van der Merwe, P.A. Effects of common mutations in the SARS-CoV-2 Spike RBD and its ligand, the human ACE2 receptor on binding affinity and kinetics. eLife 2021, 10, e70658. [Google Scholar] [CrossRef]
- Luan, B.; Wang, H.; Huynh, T. Enhanced binding of the N501Y-mutated SARS-CoV-2 spike protein to the human ACE2 receptor: Insights from molecular dynamics simulations. FEBS Lett. 2021, 595, 1454–1461. [Google Scholar] [CrossRef]
- Mansbach, R.A.; Chakraborty, S.; Nguyen, K.; Montefiori, D.C.; Korber, B.; Gnanakaran, S. The SARS-CoV-2 Spike variant D614G favors an open conformational state. Sci. Adv. 2021, 7, eabf3671. [Google Scholar] [CrossRef]
- Zhang, L.; Jackson, C.B.; Mou, H.; Ojha, A.; Peng, H.; Quinlan, B.D.; Rangarajan, E.S.; Pan, A.; Vanderheiden, A.; Suthar, M.S.; et al. SARS-CoV-2 spike-protein D614G mutation increases virion spike density and infectivity. Nat. Commun. 2020, 11, 6013. [Google Scholar] [CrossRef]
- Tarke, A.; Sidney, J.; Kidd, C.K.; Dan, J.M.; Ramirez, S.I.; Yu, E.D.; Mateus, J.; da Silva Antunes, R.; Moore, E.; Rubiro, P.; et al. Comprehensive analysis of T cell immunodominance and immunoprevalence of SARS-CoV-2 epitopes in COVID-19 cases. Cell Rep. Med. 2021, 2, 100204. [Google Scholar] [CrossRef]
- McCallum, M.; Czudnochowski, N.; Rosen, L.E.; Zepeda, S.K.; Bowen, J.E.; Walls, A.C.; Hauser, K.; Joshi, A.; Stewart, C.; Dillen, J.R.; et al. Structural basis of SARS-CoV-2 Omicron immune evasion and receptor engagement. Science 2022, 375, 864–868. [Google Scholar] [CrossRef]
- Wang, P.; Nair, M.S.; Liu, L.; Iketani, S.; Luo, Y.; Guo, Y.; Wang, M.; Yu, J.; Zhang, B.; Kwong, P.D.; et al. Antibody resistance of SARS-CoV-2 variants B.1.351 and B.1.1.7. Nature 2021, 593, 130–135. [Google Scholar] [CrossRef]
- McCarthy, K.R.; Rennick, L.J.; Nambulli, S.; Robinson-McCarthy, L.R.; Bain, W.G.; Haidar, G.; Duprex, W.P. Recurrent deletions in the SARS-CoV-2 spike glycoprotein drive antibody escape. Science 2021, 371, 1139–1142. [Google Scholar] [CrossRef]
- Rani, P.R.; Imran, M.; Lakshmi, J.V.; Jolly, B.; Jain, A.; Surekha, A.; Senthivel, V.; Chandrasekhar, P.; Divakar, M.K.; Srinivasulu, D.; et al. Symptomatic reinfection of SARS-CoV-2 with spike protein variant N440K associated with immune escape. J. Med. Virol. 2021, 93, 4163–4165. [Google Scholar] [CrossRef]
- Weisblum, Y.; Schmidt, F.; Zhang, F.; DaSilva, J.; Poston, D.; Lorenzi, J.C.; Muecksch, F.; Rutkowska, M.; Hoffmann, H.H.; Michailidis, E.; et al. Escape from neutralizing antibodies by SARS-CoV-2 spike protein variants. eLife 2020, 9, e61312. [Google Scholar] [CrossRef]
- Greaney, A.J.; Starr, T.N.; Gilchuk, P.; Zost, S.J.; Binshtein, E.; Loes, A.N.; Hilton, S.K.; Huddleston, J.; Eguia, R.; Crawford, K.H.D.; et al. Complete Mapping of Mutations to the SARS-CoV-2 Spike Receptor-Binding Domain that Escape Antibody Recognition. Cell Host Microbe 2021, 29, 44–57.e9. [Google Scholar] [CrossRef]
- Liu, Z.; VanBlargan, L.A.; Bloyet, L.M.; Rothlauf, P.W.; Chen, R.E.; Stumpf, S.; Zhao, H.; Errico, J.M.; Theel, E.S.; Liebeskind, M.J.; et al. Identification of SARS-CoV-2 spike mutations that attenuate monoclonal and serum antibody neutralization. Cell Host Microbe 2021, 29, 477–488.e4. [Google Scholar] [CrossRef]
- Arora, P.; Rocha, C.; Kempf, A.; Nehlmeier, I.; Graichen, L.; Winkler, M.S.; Lier, M.; Schulz, S.; Jack, H.M.; Cossmann, A.; et al. The spike protein of SARS-CoV-2 variant A.30 is heavily mutated and evades vaccine-induced antibodies with high efficiency. Cell. Mol. Immunol. 2021, 18, 2673–2675. [Google Scholar] [CrossRef]
- Starr, T.N.; Greaney, A.J.; Dingens, A.S.; Bloom, J.D. Complete map of SARS-CoV-2 RBD mutations that escape the monoclonal antibody LY-CoV555 and its cocktail with LY-CoV016. Cell Rep. Med. 2021, 2, 100255. [Google Scholar] [CrossRef]
- Baum, A.; Fulton, B.O.; Wloga, E.; Copin, R.; Pascal, K.E.; Russo, V.; Giordano, S.; Lanza, K.; Negron, N.; Ni, M.; et al. Antibody cocktail to SARS-CoV-2 spike protein prevents rapid mutational escape seen with individual antibodies. Science 2020, 369, 1014–1018. [Google Scholar] [CrossRef]
- Shekhar, N.; Sarma, P.; Prajapat, M.; Avti, P.; Kaur, H.; Raja, A.; Singh, H.; Bhattacharya, A.; Sharma, S.; Kumar, S.; et al. In Silico Structure-Based Repositioning of Approved Drugs for Spike Glycoprotein S2 Domain Fusion Peptide of SARS-CoV-2: Rationale from Molecular Dynamics and Binding Free Energy Calculations. mSystems 2020, 5, e00382-20. [Google Scholar] [CrossRef]
- Schutz, D.; Ruiz-Blanco, Y.B.; Munch, J.; Kirchhoff, F.; Sanchez-Garcia, E.; Muller, J.A. Peptide and peptide-based inhibitors of SARS-CoV-2 entry. Adv. Drug Deliv. Rev. 2020, 167, 47–65. [Google Scholar] [CrossRef]
- 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]
- Kleine-Weber, H.; Elzayat, M.T.; Hoffmann, M.; Pohlmann, S. Functional analysis of potential cleavage sites in the MERS-coronavirus spike protein. Sci. Rep. 2018, 8, 16597. [Google Scholar] [CrossRef]
- Ujike, M.; Nishikawa, H.; Otaka, A.; Yamamoto, N.; Yamamoto, N.; Matsuoka, M.; Kodama, E.; Fujii, N.; Taguchi, F. Heptad repeat-derived peptides block protease-mediated direct entry from the cell surface of severe acute respiratory syndrome coronavirus but not entry via the endosomal pathway. J. Virol. 2008, 82, 588–592. [Google Scholar] [CrossRef] [Green Version]
- Ni, D.; Lau, K.; Turelli, P.; Raclot, C.; Beckert, B.; Nazarov, S.; Pojer, F.; Myasnikov, A.; Stahlberg, H.; Trono, D. Structural analysis of the Spike of the Omicron SARS-COV-2 variant by cryo-EM and implications for immune evasion. bioRxiv 2021. [Google Scholar] [CrossRef]
- Whittaker, G.R. SARS-CoV-2 spike and its adaptable furin cleavage site. Lancet Microbe 2021, 2, e488–e489. [Google Scholar] [CrossRef]
- 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.e5. [Google Scholar] [CrossRef]
- Sasaki, M.; Uemura, K.; Sato, A.; Toba, S.; Sanaki, T.; Maenaka, K.; Hall, W.W.; Orba, Y.; Sawa, H. SARS-CoV-2 variants with mutations at the S1/S2 cleavage site are generated in vitro during propagation in TMPRSS2-deficient cells. PLoS Pathog. 2021, 17, e1009233. [Google Scholar] [CrossRef]
- Bestle, D.; Heindl, M.R.; Limburg, H.; Van Lam van, T.; Pilgram, O.; Moulton, H.; Stein, D.A.; Hardes, K.; Eickmann, M.; Dolnik, O.; et al. TMPRSS2 and furin are both essential for proteolytic activation of SARS-CoV-2 in human airway cells. Life Sci. Alliance 2020, 3, e202000786. [Google Scholar] [CrossRef]
- Peacock, T.P.; Goldhill, D.H.; Zhou, J.; Baillon, L.; Frise, R.; Swann, O.C.; Kugathasan, R.; Penn, R.; Brown, J.C.; Sanchez-David, R.Y.; et al. The furin cleavage site in the SARS-CoV-2 spike protein is required for transmission in ferrets. Nat. Microbiol. 2021, 6, 899–909. [Google Scholar] [CrossRef]
- Sasaki, M.; Toba, S.; Itakura, Y.; Chambaro, H.M.; Kishimoto, M.; Tabata, K.; Intaruck, K.; Uemura, K.; Sanaki, T.; Sato, A.; et al. SARS-CoV-2 Bearing a Mutation at the S1/S2 Cleavage Site Exhibits Attenuated Virulence and Confers Protective Immunity. mBio 2021, 12, e0141521. [Google Scholar] [CrossRef]
- Johnson, B.A.; Xie, X.; Bailey, A.L.; Kalveram, B.; Lokugamage, K.G.; Muruato, A.; Zou, J.; Zhang, X.; Juelich, T.; Smith, J.K.; et al. Loss of furin cleavage site attenuates SARS-CoV-2 pathogenesis. Nature 2021, 591, 293–299. [Google Scholar] [CrossRef]
- Saito, A.; Irie, T.; Suzuki, R.; Maemura, T.; Nasser, H.; Uriu, K.; Kosugi, Y.; Shirakawa, K.; Sadamasu, K.; Kimura, I.; et al. Enhanced fusogenicity and pathogenicity of SARS-CoV-2 Delta P681R mutation. Nature 2022, 602, 300–306. [Google Scholar] [CrossRef]
- Lubinski, B.; Fernandes, M.H.V.; Frazier, L.; Tang, T.; Daniel, S.; Diel, D.G.; Jaimes, J.A.; Whittaker, G.R. Functional evaluation of the P681H mutation on the proteolytic activation of the SARS-CoV-2 variant B.1.1.7 (Alpha) spike. iScience 2022, 25, 103589. [Google Scholar] [CrossRef]
- Yang, X.-J. SARS-COV-2 γ variant acquires spike P681H or P681R for improved viral fitness. bioRxiv 2021. [Google Scholar] [CrossRef]
- Kircheis, R.; Schuster, M.; Planz, O. COVID-19: Mechanistic Model of the African Paradox Supports the Central Role of the NF-kappaB Pathway. Viruses 2021, 13, 1887. [Google Scholar] [CrossRef]
- Iacobucci, G. Covid-19: Runny nose, headache, and fatigue are commonest symptoms of omicron, early data show. Br. Med. J. 2021, 375, n3103. [Google Scholar] [CrossRef]
- Wang, L.; Berger, N.A.; Kaelber, D.C.; Davis, P.B.; Volkow, N.D.; Xu, R. Comparison of outcomes from COVID infection in pediatric and adult patients before and after the emergence of Omicron. medRxiv 2022. [Google Scholar] [CrossRef]
- Liao, M.; Liu, Y.; Yuan, J.; Wen, Y.; Xu, G.; Zhao, J.; Cheng, L.; Li, J.; Wang, X.; Wang, F.; et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat. Med. 2020, 26, 842–844. [Google Scholar] [CrossRef]
- Khan, S.; Shafiei, M.S.; Longoria, C.; Schoggins, J.W.; Savani, R.C.; Zaki, H. SARS-CoV-2 spike protein induces inflammation via TLR2-dependent activation of the NF-kappaB pathway. eLife 2021, 10, e68563. [Google Scholar] [CrossRef]
- Sohn, K.M.; Lee, S.G.; Kim, H.J.; Cheon, S.; Jeong, H.; Lee, J.; Kim, I.S.; Silwal, P.; Kim, Y.J.; Paik, S.; et al. COVID-19 Patients Upregulate Toll-like Receptor 4-mediated Inflammatory Signaling That Mimics Bacterial Sepsis. J. Korean Med. Sci. 2020, 35, e343. [Google Scholar] [CrossRef]
- Patra, T.; Meyer, K.; Geerling, L.; Isbell, T.S.; Hoft, D.F.; Brien, J.; Pinto, A.K.; Ray, R.B.; Ray, R. SARS-CoV-2 spike protein promotes IL-6 trans-signaling by activation of angiotensin II receptor signaling in epithelial cells. PLoS Pathog. 2020, 16, e1009128. [Google Scholar] [CrossRef]
- Hsu, A.C.Y.; Wang, G.; Reid, A.T.; Veerati, P.C.; Pathinayake, P.S.; Daly, K.; Mayall, J.R.; Hansbro, P.M.; Horvat, J.C.; Wang, F.; et al. SARS-CoV-2 Spike protein promotes hyper-inflammatory response that can be ameliorated by Spike-antagonistic peptide and FDA-approved ER stress and MAP kinase inhibitors in vitro. bioRxiv 2020. [Google Scholar] [CrossRef]
- Olajide, O.A.; Iwuanyanwu, V.U.; Lepiarz-Raba, I.; Al-Hindawi, A.A. Induction of Exaggerated Cytokine Production in Human Peripheral Blood Mononuclear Cells by a Recombinant SARS-CoV-2 Spike Glycoprotein S1 and Its Inhibition by Dexamethasone. Inflammation 2021, 44, 1865–1877. [Google Scholar] [CrossRef]
- Petruk, G.; Puthia, M.; Petrlova, J.; Samsudin, F.; Stromdahl, A.C.; Cerps, S.; Uller, L.; Kjellstrom, S.; Bond, P.J.; Schmidtchen, A.A. SARS-CoV-2 spike protein binds to bacterial lipopolysaccharide and boosts proinflammatory activity. J. Mol. Cell Biol. 2020, 12, 916–932. [Google Scholar] [CrossRef]
- Colunga Biancatelli, R.M.L.; Solopov, P.A.; Sharlow, E.R.; Lazo, J.S.; Marik, P.E.; Catravas, J.D. The SARS-CoV-2 spike protein subunit S1 induces COVID-19-like acute lung injury in Kappa18-hACE2 transgenic mice and barrier dysfunction in human endothelial cells. Am. J. Physiol. Lung. Cell Mol. Physiol. 2021, 321, L477–L484. [Google Scholar] [CrossRef]
- Wang, W.; Ye, L.; Ye, L.; Li, B.; Gao, B.; Zeng, Y.; Kong, L.; Fang, X.; Zheng, H.; Wu, Z.; et al. Up-regulation of IL-6 and TNF-alpha induced by SARS-coronavirus spike protein in murine macrophages via NF-kappaB pathway. Virus Res. 2007, 128, 1–8. [Google Scholar] [CrossRef]
- Nilsson-Payant, B.E.; Uhl, S.; Grimont, A.; Doane, A.S.; Cohen, P.; Patel, R.S.; Higgins, C.A.; Acklin, J.A.; Bram, Y.; Chandar, V.; et al. The NF-kappaB Transcriptional Footprint Is Essential for SARS-CoV-2 Replication. J. Virol. 2021, 95, e0125721. [Google Scholar] [CrossRef]
- Ehrhardt, C.; Ruckle, A.; Hrincius, E.R.; Haasbach, E.; Anhlan, D.; Ahmann, K.; Banning, C.; Reiling, S.J.; Kuhn, J.; Strobl, S.; et al. The NF-kappaB inhibitor SC75741 efficiently blocks influenza virus propagation and confers a high barrier for development of viral resistance. Cell. Microbiol. 2013, 15, 1198–1211. [Google Scholar] [CrossRef]
- Haasbach, E.; Reiling, S.J.; Ehrhardt, C.; Droebner, K.; Ruckle, A.; Hrincius, E.R.; Leban, J.; Strobl, S.; Vitt, D.; Ludwig, S.; et al. The NF-kappaB inhibitor SC75741 protects mice against highly pathogenic avian influenza A virus. Antivir. Res. 2013, 99, 336–344. [Google Scholar] [CrossRef]
- Thomas, G. Furin at the cutting edge: From protein traffic to embryogenesis and disease. Nat. Rev. Mol. Cell Biol. 2002, 3, 753–766. [Google Scholar] [CrossRef] [Green Version]
- Shapiro, J.; Sciaky, N.; Lee, J.; Bosshart, H.; Angeletti, R.H.; Bonifacino, J.S. Localization of endogenous furin in cultured cell lines. J. Histochem. Cytochem. 1997, 45, 3–12. [Google Scholar] [CrossRef] [Green Version]
- Scudellari, M. How the coronavirus infects cells—And why Delta is so dangerous. Nature 2021, 595, 640–644. [Google Scholar] [CrossRef]
- Bourne, G.L.; Grainger, D.J. Development and characterisation of an assay for furin activity. J. Immunol. Methods 2011, 364, 101–108. [Google Scholar] [CrossRef]
- Uhlen, M.; Fagerberg, L.; Hallstrom, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, A.; Kampf, C.; Sjostedt, E.; Asplund, A.; et al. Proteomics. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef]
- Pesu, M.; Muul, L.; Kanno, Y.; O’Shea, J.J. Proprotein convertase furin is preferentially expressed in T helper 1 cells and regulates interferon gamma. Blood 2006, 108, 983–985. [Google Scholar] [CrossRef] [Green Version]
- Oksanen, A.; Aittomaki, S.; Jankovic, D.; Ortutay, Z.; Pulkkinen, K.; Hamalainen, S.; Rokka, A.; Corthals, G.L.; Watford, W.T.; Junttila, I.; et al. Proprotein convertase FURIN constrains Th2 differentiation and is critical for host resistance against Toxoplasma gondii. J. Immunol. 2014, 193, 5470–5479. [Google Scholar] [CrossRef] [Green Version]
- Murphy, T.L.; Cleveland, M.G.; Kulesza, P.; Magram, J.; Murphy, K.M. Regulation of interleukin 12 p40 expression through an NF-kappa B half-site. Mol. Cell. Biol. 1995, 15, 5258–5267. [Google Scholar] [CrossRef] [Green Version]
- Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-kappaB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar]
- Zhu, L.; Liu, H.W.; Yang, Y.; Hu, X.X.; Li, K.; Xu, S.; Li, J.B.; Ke, G.; Zhang, X.B. Near-Infrared Fluorescent Furin Probe for Revealing the Role of Furin in Cellular Carcinogenesis and Specific Cancer Imaging. Anal. Chem. 2019, 91, 9682–9689. [Google Scholar] [CrossRef]
- Kumar, V.; Behera, R.; Lohite, K.; Karnik, S.; Kundu, G.C. p38 kinase is crucial for osteopontin-induced furin expression that supports cervical cancer progression. Cancer Res. 2010, 70, 10381–10391. [Google Scholar] [CrossRef] [Green Version]
- Adu-Agyeiwaah, Y.; Grant, M.B.; Obukhov, A.G. The Potential Role of Osteopontin and Furin in Worsening Disease Outcomes in COVID-19 Patients with Pre-Existing Diabetes. Cells 2020, 9, 2528. [Google Scholar] [CrossRef]
- McMahon, S.; Grondin, F.; McDonald, P.P.; Richard, D.E.; Dubois, C.M. Hypoxia-enhanced expression of the proprotein convertase furin is mediated by hypoxia-inducible factor-1: Impact on the bioactivation of proproteins. J. Biol. Chem. 2005, 280, 6561–6569. [Google Scholar] [CrossRef] [Green Version]
- Taylor, C.T.; Cummins, E.P. The role of NF-kappaB in hypoxia-induced gene expression. Ann. N. Y. Acad Sci. 2009, 1177, 178–184. [Google Scholar] [CrossRef]
- Han, S.; Xu, W.; Wang, Z.; Qi, X.; Wang, Y.; Ni, Y.; Shen, H.; Hu, Q.; Han, W. Crosstalk between the HIF-1 and Toll-like receptor/nuclear factor-kappaB pathways in the oral squamous cell carcinoma microenvironment. Oncotarget 2016, 7, 37773–37789. [Google Scholar] [CrossRef]
- Riddell, J.R.; Maier, P.; Sass, S.N.; Moser, M.T.; Foster, B.A.; Gollnick, S.O. Peroxiredoxin 1 stimulates endothelial cell expression of VEGF via TLR4 dependent activation of HIF-1alpha. PLoS ONE 2012, 7, e50394. [Google Scholar] [CrossRef] [Green Version]
- Spirig, R.; Djafarzadeh, S.; Regueira, T.; Shaw, S.G.; von Garnier, C.; Takala, J.; Jakob, S.M.; Rieben, R.; Lepper, P.M. Effects of TLR agonists on the hypoxia-regulated transcription factor HIF-1alpha and dendritic cell maturation under normoxic conditions. PLoS ONE 2010, 5, e0010983. [Google Scholar] [CrossRef] [Green Version]
- Peyssonnaux, C.; Cejudo-Martin, P.; Doedens, A.; Zinkernagel, A.S.; Johnson, R.S.; Nizet, V. Cutting edge: Essential role of hypoxia inducible factor-1alpha in development of lipopolysaccharide-induced sepsis. J. Immunol. 2007, 178, 7516–7519. [Google Scholar] [CrossRef]
- Jiang, H.; Hu, R.; Sun, L.; Chai, D.; Cao, Z.; Li, Q. Critical role of Toll-like receptor 4 in hypoxia-inducible factor 1alpha activation during trauma/hemorrhagic shocky induced acute lung injury after lymph infusion in mice. Shock 2014, 42, 271–278. [Google Scholar] [CrossRef]
- Yang, X.; Chen, G.T.; Wang, Y.Q.; Xian, S.; Zhang, L.; Zhu, S.M.; Pan, F.; Cheng, Y.X. TLR4 promotes the expression of HIF-1alpha by triggering reactive oxygen species in cervical cancer cells in vitro-implications for therapeutic intervention. Mol. Med. Rep. 2018, 17, 2229–2238. [Google Scholar]
- Braun, E.; Sauter, D. Furin-mediated protein processing in infectious diseases and cancer. Clin. Transl. Immunol. 2019, 8, e1073. [Google Scholar] [CrossRef] [Green Version]
- Tse, L.V.; Hamilton, A.M.; Friling, T.; Whittaker, G.R. A novel activation mechanism of avian influenza virus H9N2 by furin. J. Virol. 2014, 88, 1673–1683. [Google Scholar] [CrossRef] [Green Version]
- De Greef, J.C.; Slutter, B.; Anderson, M.E.; Hamlyn, R.; O’Campo Landa, R.; McNutt, E.J.; Hara, Y.; Pewe, L.L.; Venzke, D.; Matsumura, K.; et al. Protective role for the N-terminal domain of alpha-dystroglycan in Influenza A virus proliferation. Proc. Natl. Acad. Sci. USA 2019, 116, 11396–11401. [Google Scholar] [CrossRef] [Green Version]
- Li, D.; Liu, X.; Zhang, L.; He, J.; Chen, X.; Liu, S.; Fu, J.; Fu, S.; Chen, H.; Fu, J.; et al. COVID-19 disease and malignant cancers: The impact for the furin gene expression in susceptibility to SARS-CoV-2. Int. J. Biol. Sci. 2021, 17, 3954–3967. [Google Scholar] [CrossRef]
- Kocyigit, A.; Sogut, O.; Durmus, E.; Kanimdan, E.; Guler, E.M.; Kaplan, O.; Yenigun, V.B.; Eren, C.; Ozman, Z.; Yasar, O. Circulating furin, IL-6, and presepsin levels and disease severity in SARS-CoV-2-infected patients. Sci. Prog. 2021, 104, 368504211026119. [Google Scholar] [CrossRef]
- Kermani, N.Z.; Song, W.J.; Badi, Y.; Versi, A.; Guo, Y.; Sun, K.; Bhavsar, P.; Howarth, P.; Dahlen, S.E.; Sterk, P.J.; et al. Sputum ACE2, TMPRSS2 and FURIN gene expression in severe neutrophilic asthma. Respir. Res. 2021, 22, 10. [Google Scholar] [CrossRef]
- Brake, S.J.; Eapen, M.S.; McAlinden, K.D.; Markos, J.; Haug, G.; Larby, J.; Chia, C.; Hardikar, A.; Singhera, G.K.; Hackett, T.L.; et al. SARS-CoV-2 (COVID-19) Adhesion Site Protein Upregulation in Small Airways, Type 2 Pneumocytes, and Alveolar Macrophages of Smokers and COPD—Possible Implications for Interstitial Fibrosis. Int. J. Chron. Obstruct. Pulmon. Dis. 2022, 17, 101–115. [Google Scholar] [CrossRef]
- Zalinger, Z.B.; Elliott, R.; Rose, K.M.; Weiss, S.R. MDA5 Is Critical to Host Defense during Infection with Murine Coronavirus. J. Virol. 2015, 89, 12330–12340. [Google Scholar] [CrossRef] [Green Version]
- Cervantes-Barragan, L.; Zust, R.; Weber, F.; Spiegel, M.; Lang, K.S.; Akira, S.; Thiel, V.; Ludewig, B. Control of coronavirus infection through plasmacytoid dendritic-cell-derived type I interferon. Blood 2007, 109, 1131–1137. [Google Scholar] [CrossRef] [Green Version]
- Channappanavar, R.; Fehr, A.R.; Zheng, J.; Wohlford-Lenane, C.; Abrahante, J.E.; Mack, M.; Sompallae, R.; McCray, P.B., Jr.; Meyerholz, D.K.; Perlman, S. IFN-I response timing relative to virus replication determines MERS coronavirus infection outcomes. J. Clin. Investig. 2019, 129, 3625–3639. [Google Scholar] [CrossRef]
- Zheng, M.; Williams, E.P.; Malireddi, R.K.S.; Karki, R.; Banoth, B.; Burton, A.; Webby, R.; Channappanavar, R.; Jonsson, C.B.; Kanneganti, T.D. Impaired NLRP3 inflammasome activation/pyroptosis leads to robust inflammatory cell death via caspase-8/RIPK3 during coronavirus infection. J. Biol. Chem. 2020, 295, 14040–14052. [Google Scholar] [CrossRef]
- Sheahan, T.; Morrison, T.E.; Funkhouser, W.; Uematsu, S.; Akira, S.; Baric, R.S.; Heise, M.T. MyD88 is required for protection from lethal infection with a mouse-adapted SARS-CoV. PLoS Pathog. 2008, 4, e1000240. [Google Scholar] [CrossRef] [Green Version]
- Hadjadj, J.; Yatim, N.; Barnabei, L.; Corneau, A.; Boussier, J.; Smith, N.; Pere, H.; Charbit, B.; Bondet, V.; Chenevier-Gobeaux, C.; et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 2020, 369, 718–724. [Google Scholar] [CrossRef]
- El-Zayat, S.R.; Sibaii, H.; Mannaa, F.A. Toll-like receptors activation, signaling, and targeting: An overview. Bull. Natl. Res. Cent. 2019, 43, 187. [Google Scholar] [CrossRef] [Green Version]
- Zheng, M.; Karki, R.; Williams, E.P.; Yang, D.; Fitzpatrick, E.; Vogel, P.; Jonsson, C.B.; Kanneganti, T.D. TLR2 senses the SARS-CoV-2 envelope protein to produce inflammatory cytokines. Nat. Immunol. 2021, 22, 829–838. [Google Scholar] [CrossRef]
- Planès, R.; Bert, J.-B.; Tairi, S.; Benmohamed, L.; Bahraoui, E. SARS-CoV-2 Envelope protein (E) binds and activates TLR2: A novel target for COVID-19 interventions. bioRxiv 2021. [Google Scholar] [CrossRef]
- Aboudounya, M.M.; Heads, R.J. COVID-19 and Toll-Like Receptor 4 (TLR4): SARS-CoV-2 May Bind and Activate TLR4 to Increase ACE2 Expression, Facilitating Entry and Causing Hyperinflammation. Mediat. Inflamm. 2021, 2021, 8874339. [Google Scholar] [CrossRef]
- Mogensen, T.H.; Paludan, S.R. Reading the viral signature by Toll-like receptors and other pattern recognition receptors. J. Mol. Med. 2005, 83, 180–192. [Google Scholar] [CrossRef] [PubMed]
- Turner, N.A. Inflammatory and fibrotic responses of cardiac fibroblasts to myocardial damage associated molecular patterns (DAMPs). J. Mol. Cell. Cardiol. 2016, 94, 189–200. [Google Scholar] [CrossRef] [PubMed]
- Bhattacharyya, S.; Wang, W.; Qin, W.; Cheng, K.; Coulup, S.; Chavez, S.; Jiang, S.; Raparia, K.; De Almeida, L.M.V.; Stehlik, C.; et al. TLR4-dependent fibroblast activation drives persistent organ fibrosis in skin and lung. JCI Insight 2018, 3, e98850. [Google Scholar] [CrossRef] [Green Version]
- Chow, J.C.; Young, D.W.; Golenbock, D.T.; Christ, W.J.; Gusovsky, F. Toll-like receptor-4 mediates lipopolysaccharide-induced signal transduction. J. Biol. Chem. 1999, 274, 10689–10692. [Google Scholar] [CrossRef] [Green Version]
- Kuzmich, N.N.; Sivak, K.V.; Chubarev, V.N.; Porozov, Y.B.; Savateeva-Lyubimova, T.N.; Peri, F. TLR4 Signaling Pathway Modulators as Potential Therapeutics in Inflammation and Sepsis. Vaccines 2017, 5, 34. [Google Scholar] [CrossRef] [Green Version]
- Molteni, M.; Gemma, S.; Rossetti, C. The Role of Toll-Like Receptor 4 in Infectious and Noninfectious Inflammation. Mediat. Inflamm. 2016, 2016, 6978936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roshan, M.H.; Tambo, A.; Pace, N.P. The Role of TLR2, TLR4, and TLR9 in the Pathogenesis of Atherosclerosis. Int. J. Inflam. 2016, 2016, 1532832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patra, M.C.; Shah, M.; Choi, S. Toll-like receptor-induced cytokines as immunotherapeutic targets in cancers and autoimmune diseases. Semin. Cancer Biol. 2020, 64, 61–82. [Google Scholar] [CrossRef]
- Garcia Bueno, B.; Caso, J.R.; Madrigal, J.L.; Leza, J.C. Innate immune receptor Toll-like receptor 4 signalling in neuropsychiatric diseases. Neurosci. Biobehav. Rev. 2016, 64, 134–147. [Google Scholar] [CrossRef]
- Mukherjee, S.; Karmakar, S.; Babu, S.P. TLR2 and TLR4 mediated host immune responses in major infectious diseases: A review. Braz. J. Infect. Dis. 2016, 20, 193–204. [Google Scholar] [CrossRef] [Green Version]
- Choudhury, A.; Mukherjee, S. In silico studies on the comparative characterization of the interactions of SARS-CoV-2 spike glycoprotein with ACE-2 receptor homologs and human TLRs. J. Med. Virol. 2020, 92, 2105–2113. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Kuang, M.; Li, J.; Zhu, L.; Jia, Z.; Guo, X.; Hu, Y.; Kong, J.; Yin, H.; Wang, X.; et al. SARS-CoV-2 spike protein interacts with and activates TLR41. Cell Res. 2021, 31, 818–820. [Google Scholar] [CrossRef]
- Shirato, K.; Kizaki, T. SARS-CoV-2 spike protein S1 subunit induces pro-inflammatory responses via toll-like receptor 4 signaling in murine and human macrophages. Heliyon 2021, 7, e06187. [Google Scholar] [CrossRef] [PubMed]
- Aboudounya, M.M.; Holt, M.R.; Heads, R.J. SARS-CoV-2 Spike S1 glycoprotein is a TLR4 agonist, upregulates ACE2 expression and induces pro-inflammatory M1 macrophage polarisation. bioRxiv 2021. [Google Scholar] [CrossRef]
- Russell, C.D.; Unger, S.A.; Walton, M.; Schwarze, J. The Human Immune Response to Respiratory Syncytial Virus Infection. Clin. Microbiol. Rev. 2017, 30, 481–502. [Google Scholar] [CrossRef] [Green Version]
- Guo, X.J.; Thomas, P.G. New fronts emerge in the influenza cytokine storm. Semin. Immunopathol. 2017, 39, 541–550. [Google Scholar] [CrossRef]
- Bixler, S.L.; Goff, A.J. The Role of Cytokines and Chemokines in Filovirus Infection. Viruses 2015, 7, 5489–5507. [Google Scholar] [CrossRef]
- Srikiatkhachorn, A.; Mathew, A.; Rothman, A.L. Immune-mediated cytokine storm and its role in severe dengue. Semin. Immunopathol. 2017, 39, 563–574. [Google Scholar] [CrossRef]
- Channappanavar, R.; Perlman, S. Pathogenic human coronavirus infections: Causes and consequences of cytokine storm and immunopathology. Semin. Immunopathol. 2017, 39, 529–539. [Google Scholar] [CrossRef]
- Rallabhandi, P.; Phillips, R.L.; Boukhvalova, M.S.; Pletneva, L.M.; Shirey, K.A.; Gioannini, T.L.; Weiss, J.P.; Chow, J.C.; Hawkins, L.D.; Vogel, S.N.; et al. Respiratory syncytial virus fusion protein-induced toll-like receptor 4 (TLR4) signaling is inhibited by the TLR4 antagonists Rhodobacter sphaeroides lipopolysaccharide and eritoran (E5564) and requires direct interaction with MD-2. mBio 2012, 3, e00218-12. [Google Scholar] [CrossRef] [Green Version]
- Lai, C.Y.; Strange, D.P.; Wong, T.A.S.; Lehrer, A.T.; Verma, S. Ebola Virus Glycoprotein Induces an Innate Immune Response In vivo via TLR4. Front. Microbiol. 2017, 8, 1571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Okumura, A.; Pitha, P.M.; Yoshimura, A.; Harty, R.N. Interaction between Ebola virus glycoprotein and host toll-like receptor 4 leads to induction of proinflammatory cytokines and SOCS1. J. Virol. 2010, 84, 27–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harrison, S.C. Viral membrane fusion. Virology 2015, 479–480, 498–507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Modhiran, N.; Watterson, D.; Muller, D.A.; Panetta, A.K.; Sester, D.P.; Liu, L.; Hume, D.A.; Stacey, K.J.; Young, P.R. Dengue virus NS1 protein activates cells via Toll-like receptor 4 and disrupts endothelial cell monolayer integrity. Sci. Transl. Med. 2015, 7, 304ra142. [Google Scholar] [CrossRef] [Green Version]
- Akey, D.L.; Brown, W.C.; Dutta, S.; Konwerski, J.; Jose, J.; Jurkiw, T.J.; DelProposto, J.; Ogata, C.M.; Skiniotis, G.; Kuhn, R.J.; et al. Flavivirus NS1 structures reveal surfaces for associations with membranes and the immune system. Science 2014, 343, 881–885. [Google Scholar] [CrossRef] [Green Version]
- Iampietro, M.; Younan, P.; Nishida, A.; Dutta, M.; Lubaki, N.M.; Santos, R.I.; Koup, R.A.; Katze, M.G.; Bukreyev, A. Ebola virus glycoprotein directly triggers T lymphocyte death despite of the lack of infection. PLoS Pathog. 2017, 13, e1006397. [Google Scholar] [CrossRef]
- Escudero-Perez, B.; Volchkova, V.A.; Dolnik, O.; Lawrence, P.; Volchkov, V.E. Shed GP of Ebola virus triggers immune activation and increased vascular permeability. PLoS Pathog 2014, 10, e1004509. [Google Scholar] [CrossRef]
- Olejnik, J.; Forero, A.; Deflube, L.R.; Hume, A.J.; Manhart, W.A.; Nishida, A.; Marzi, A.; Katze, M.G.; Ebihara, H.; Rasmussen, A.L.; et al. Ebolaviruses Associated with Differential Pathogenicity Induce Distinct Host Responses in Human Macrophages. J. Virol. 2017, 91, e00179-17. [Google Scholar] [CrossRef] [Green Version]
- Younan, P.; Ramanathan, P.; Graber, J.; Gusovsky, F.; Bukreyev, A. The Toll-Like Receptor 4 Antagonist Eritoran Protects Mice from Lethal Filovirus Challenge. mBio 2017, 8, e00226-17. [Google Scholar] [CrossRef] [Green Version]
- Modhiran, N.; Watterson, D.; Blumenthal, A.; Baxter, A.G.; Young, P.R.; Stacey, K.J. Dengue virus NS1 protein activates immune cells via TLR4 but not TLR2 or TLR6. Immunol. Cell Biol. 2017, 95, 491–495. [Google Scholar] [CrossRef] [Green Version]
- Olejnik, J.; Hume, A.J.; Muhlberger, E. Toll-like receptor 4 in acute viral infection: Too much of a good thing. PLoS Pathog. 2018, 14, e1007390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, B.S.; Lee, J.O. Recognition of lipopolysaccharide pattern by TLR4 complexes. Exp. Mol. Med. 2013, 45, e66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ohto, U.; Fukase, K.; Miyake, K.; Shimizu, T. Structural basis of species-specific endotoxin sensing by innate immune receptor TLR4/MD-2. Proc. Natl. Acad. Sci. USA 2012, 109, 7421–7426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schromm, A.B.; Brandenburg, K.; Loppnow, H.; Zahringer, U.; Rietschel, E.T.; Carroll, S.F.; Koch, M.H.; Kusumoto, S.; Seydel, U. The charge of endotoxin molecules influences their conformation and IL-6-inducing capacity. J. Immunol. 1998, 161, 5464–5471. [Google Scholar] [PubMed]
- Schromm, A.B.; Brandenburg, K.; Loppnow, H.; Moran, A.P.; Koch, M.H.; Rietschel, E.T.; Seydel, U. Biological activities of lipopolysaccharides are determined by the shape of their lipid A portion. Eur. J. Biochem. 2000, 267, 2008–2013. [Google Scholar] [CrossRef]
- Du, X.; Tang, H.; Gao, L.; Wu, Z.; Meng, F.; Yan, R.; Qiao, S.; An, J.; Wang, C.; Qin, F.X. Omicron adopts a different strategy from Delta and other variants to adapt to host. Signal Transduct. Target. Ther. 2022, 7, 45. [Google Scholar] [CrossRef]
- Zhao, M.M.; Yang, W.L.; Yang, F.Y.; Zhang, L.; Huang, W.J.; Hou, W.; Fan, C.F.; Jin, R.H.; Feng, Y.M.; Wang, Y.C.; et al. Cathepsin L plays a key role in SARS-CoV-2 infection in humans and humanized mice and is a promising target for new drug development. Signal Transduct. Target. Ther. 2021, 6, 134. [Google Scholar] [CrossRef]
- Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.C.; Uhl, S.; Hoagland, D.; Moller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 2020, 181, 1036–1045.e9. [Google Scholar] [CrossRef]
- Konno, Y.; Kimura, I.; Uriu, K.; Fukushi, M.; Irie, T.; Koyanagi, Y.; Sauter, D.; Gifford, R.J.; Consortium, U.-C.; Nakagawa, S.; et al. SARS-CoV-2 ORF3b Is a Potent Interferon Antagonist Whose Activity Is Increased by a Naturally Occurring Elongation Variant. Cell Rep. 2020, 32, 108185. [Google Scholar] [CrossRef]
- Lei, X.; Dong, X.; Ma, R.; Wang, W.; Xiao, X.; Tian, Z.; Wang, C.; Wang, Y.; Li, L.; Ren, L.; et al. Activation and evasion of type I interferon responses by SARS-CoV-2. Nat. Commun. 2020, 11, 3810. [Google Scholar] [CrossRef]
- Park, M.D. Immune evasion via SARS-CoV-2 ORF8 protein? Nat. Rev. Immunol. 2020, 20, 408. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.; Zhang, X.; Wang, F.; Wang, P.; Kuang, E.; Li, X. Suppression of MDA5-mediated antiviral immune responses by NSP8 of SARS-CoV-2. bioRxiv 2020. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Kircheis, R.; Planz, O. Could a Lower Toll-like Receptor (TLR) and NF-κB Activation Due to a Changed Charge Distribution in the Spike Protein Be the Reason for the Lower Pathogenicity of Omicron? Int. J. Mol. Sci. 2022, 23, 5966. https://doi.org/10.3390/ijms23115966
Kircheis R, Planz O. Could a Lower Toll-like Receptor (TLR) and NF-κB Activation Due to a Changed Charge Distribution in the Spike Protein Be the Reason for the Lower Pathogenicity of Omicron? International Journal of Molecular Sciences. 2022; 23(11):5966. https://doi.org/10.3390/ijms23115966
Chicago/Turabian StyleKircheis, Ralf, and Oliver Planz. 2022. "Could a Lower Toll-like Receptor (TLR) and NF-κB Activation Due to a Changed Charge Distribution in the Spike Protein Be the Reason for the Lower Pathogenicity of Omicron?" International Journal of Molecular Sciences 23, no. 11: 5966. https://doi.org/10.3390/ijms23115966
APA StyleKircheis, R., & Planz, O. (2022). Could a Lower Toll-like Receptor (TLR) and NF-κB Activation Due to a Changed Charge Distribution in the Spike Protein Be the Reason for the Lower Pathogenicity of Omicron? International Journal of Molecular Sciences, 23(11), 5966. https://doi.org/10.3390/ijms23115966