Direct Activation of Endothelial Cells by SARS-CoV-2 Nucleocapsid Protein Is Blocked by Simvastatin

Yisong Qian, Tianhua Lei, Parth S Patel, Chi H Lee, Paula Monaghan-Nichols, Hong-Bo Xin, Jianming Qiu, Mingui Fu, Yisong Qian, Tianhua Lei, Parth S Patel, Chi H Lee, Paula Monaghan-Nichols, Hong-Bo Xin, Jianming Qiu, Mingui Fu

Abstract

Emerging evidence suggests that endothelial activation plays a central role in the pathogenesis of acute respiratory distress syndrome (ARDS) and multiorgan failure in patients with coronavirus disease 2019 (COVID-19). However, the molecular mechanisms underlying endothelial activation in COVID-19 patients remain unclear. In this study, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral proteins that potently activate human endothelial cells were screened to elucidate the molecular mechanisms involved in endothelial activation. It was found that nucleocapsid protein (NP) of SARS-CoV-2 significantly activated human endothelial cells through Toll-like receptor 2 (TLR2)/NF-κB and mitogen-activated protein kinase (MAPK) signaling pathways. Moreover, by screening a natural microbial compound library containing 154 natural compounds, simvastatin was identified as a potent inhibitor of NP-induced endothelial activation. Remarkably, though the protein sequences of N proteins from coronaviruses are highly conserved, only NP from SARS-CoV-2 induced endothelial activation. The NPs from other coronaviruses such as SARS-CoV, Middle East respiratory syndrome coronavirus (MERS-CoV), HUB1-CoV, and influenza virus H1N1 did not activate endothelial cells. These findings are consistent with the results from clinical investigations showing broad endotheliitis and organ injury in severe COVID-19 patients. In conclusion, the study provides insights on SARS-CoV-2-induced vasculopathy and coagulopathy and suggests that simvastatin, an FDA-approved lipid-lowering drug, may help prevent the pathogenesis and improve the outcome of COVID-19 patients. IMPORTANCE Coronavirus disease 2019 (COVID-19), caused by the betacoronavirus SARS-CoV-2, is a worldwide challenge for health care systems. The leading cause of mortality in patients with COVID-19 is hypoxic respiratory failure from acute respiratory distress syndrome (ARDS). To date, pulmonary endothelial cells (ECs) have been largely overlooked as a therapeutic target in COVID-19, yet emerging evidence suggests that these cells contribute to the initiation and propagation of ARDS by altering vessel barrier integrity, promoting a procoagulative state, inducing vascular inflammation and mediating inflammatory cell infiltration. Therefore, a better mechanistic understanding of the vasculature is of utmost importance. In this study, we screened the SARS-CoV-2 viral proteins that potently activate human endothelial cells and found that nucleocapsid protein (NP) significantly activated human endothelial cells through TLR2/NF-κB and MAPK signaling pathways. Moreover, by screening a natural microbial compound library containing 154 natural compounds, simvastatin was identified as a potent inhibitor of NP-induced endothelial activation. Our results provide insights on SARS-CoV-2-induced vasculopathy and coagulopathy, and suggests that simvastatin, an FDA-approved lipid-lowering drug, may benefit to prevent the pathogenesis and improve the outcome of COVID-19 patients.

Keywords: COVID-19; SARS-CoV-2; endothelial activation; endothelial cells; nucleocapsid protein; simvastatin.

Figures

FIG 1
FIG 1
SARS-CoV2 nucleocapsid protein (NP) is a potent inducer of human endothelial cell activation. (a) HLMECs were incubated with SARS-CoV2 structural proteins (S, N, and E proteins; 1 μg/ml) and five nonstructural proteins (NSP1, NSP3, NSP5, NSP7, and NSP8; 1 μg/ml) for 8 h. (b) HLMECs were treated with 1 μg/ml of NP or 10 ng/ml of TNF-α for different incubation periods as indicated. (c) HLMECs were incubated with indicated concentrations of NP for 8 h. TNF-α at 10 ng/ml served as a positive control. (d) Different cultured cells, including mouse lung vascular endothelial cells (MECs), A549 cells, 293T cells, HUVECs, HAECs, HCAECs, HDMECs, and HLMECs, were treated with NP (1 μg/ml) for 8 h. The expression of ICAM-1, VCAM-1, and VE-cadherin was detected by Western blotting. β-Actin served as a loading control. (e) HLMECs were treated with PBS, NP (1 μg/ml), TNF-α (10 ng/ml), or lipopolysaccharide (LPS) (1 μg/ml) for 8 h. The total RNA was isolated and qPCR was performed for measuring the mRNA levels of TNF-α, ICAM-1, VCAM-1, MCP-1, and IL-6. (f) HLMECs were treated with PBS, NP (1 μg/ml), or TNF-α (10 ng/ml) for 8 h and cocultured with Zombie Red-labeled THP-1 cells for 1 h. After being washed, the adherent cells were imaged and quantitatively analyzed.
FIG 2
FIG 2
N protein activated NF-κB and MAPK signaling pathways in human endothelial cells. HLMECs were incubated with NP (1 μg/ml), SP (1 μg/ml), TNF-α (10 ng/ml), and LPS (1 μg/ml) for the indicated times. The phosphorylation of IKKs, p65, IκBα, ERK, JNK, and p38, as well as IκBα degradation, was detected by Western blotting. GAPDH served as a loading control.
FIG 3
FIG 3
N protein induced endothelial cell activation via the TLR2-mediated signaling pathway. (a) HLMECs were pretreated with inhibitors of endocytosis (Pitstop2 [12.5 μM] and Dynasore hydrate [12.5 μM]) and antagonists of TLR4 (LPS-RS [10 μg/ml]), TLR2 (CU-CPT22 [20 μM]), IL-1R (IL-1R antagonist [20 μM]), inhibitors of IKK (IKK16 [20 μM]), ERK (U0126 [20 μM]), JNK (JNK inhibitor V [20 μM]), and p38 (SB203580 [20 μM]) for 1 h followed by treatment with NP (1 μg/ml) for 8 h. (b) The Flag control and Flag-NP expression plasmids were transfected into HLMECs by electroporation. The whole-cell lysate was harvested after 48 h transfection. The expression of ICAM-1, VCAM-1, NP, and Flag was detected by Western blotting. (c) HLMECs were incubated with NP (1 μg/ml) for different time as indicated. After being washed, the cells were harvested, and NP was detected by Western blotting. (d) HLMECs were treated with the indicated concentrations of CU-CPT22 for 1 h followed by treatment with NP (1 μg/ml) for 8 h. (e) Wild-type (left) and TLR2-overexpressing 293T cells were treated with or without NP (1 μg/ml) for 15 min. pJNK and pP38 were detected by Western blotting. GAPDH served as a loading control. All experiments were repeated at least once.
FIG 4
FIG 4
Screening of chemicals for inhibition of N protein-induced endothelial activation. A total of 155 chemicals from microbial natural product library were added to HLMECs at 30 μM 1 h before the induction of N protein (1 μg/ml). The effect of chemicals on endothelial activation was measured by Western blotting with anti-ICAM-1 antibody. β-Actin served as a loading control. Arrows indicate effective compounds; stars indicate toxic compounds causing cell death. The experiments were repeated once.
FIG 5
FIG 5
Simvastatin is an effective inhibitor of N protein-induced endothelial activation in vitro. (a) HLMECs were pretreated with simvastatin, lovastatin, atorvastatin, mevastatin, and rosuvastatin at 30 μM for 1 h followed by treatment with NP (1 μg/ml) for 8 h. (b) HLMECs were pretreated with indicated concentrations of lovastatin and simvastatin for 1 h followed by treatment with NP (1 μg/ml) for 8 h. The expression of ICAM-1 and VCAM-1 was detected by Western blotting. (c) HLMECs were pretreated with indicated concentrations of lovastatin and simvastatin for 1 h by treatment with NP (1 μg/ml) for 15 min. The activation of NF-κB and MAPK signal pathways was detected by Western blotting. (d) HLMECs were pretreated with or without lovastatin (Lova) or simvastatin (Simva) followed by treatment with NP (1 μg/ml) for 8 h and then cocultured with Zombie Red-labeled THP-1 cells for 1 h. After washing, the adherent cells were imaged.
FIG 6
FIG 6
Chemical structures of simvastatin, lovastatin, mevastatin, atorvastatin, and rosuvastatin. Structures were adapted from Wikipedia (simvastatin, https://commons.wikimedia.org/w/index.php?curid=4486633; lovastatin, https://commons.wikimedia.org/wiki/File:Lovastatin.svg; mevastatin, https://commons.wikimedia.org/wiki/File:Mevastatin.svg; atorvastatin, https://commons.wikimedia.org/wiki/File:Atorvastatin.svg; rosuvastatin, https://commons.wikimedia.org/wiki/File:Rosuvastatin_structure.svg; all structures are available under open access licenses or are in the public domain).
FIG 7
FIG 7
The N protein from SARS-CoV2 but not the other coronaviruses potently induced endothelial cell activation. HLMECs were treated with or without five different recombinant viral N proteins (1 μg/ml), including SARS-CoV2, SARS-CoV, MERS-CoV, H7N9, and HKU1-CoV, for 8 h. The expression of ICAM-1 and VCAM-1 was detected by Western blotting. TNF-α (10 ng/ml) served as a positive control. GAPDH served as the loading control. The experiments were repeated at least once.

References

    1. Ackermann M, Verleden SE, Kuehnel M, Haverich A, Welte T, Laenger F, Vanstapel A, Werlein C, Stark H, Tzankov A, Li WW, Li VW, Mentzer SJ, Jonigk D. 2020. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in Covid-19. N Engl J Med 383:120–128. 10.1056/NEJMoa2015432.
    1. Kirtipal N, Bharadwaj S, Kang SG. 2020. From SARS to SARS-CoV-2, insights on structure, pathogenicity and immunity aspects of pandemic human coronaviruses. Infect Genet Evol 85:104502. 10.1016/j.meegid.2020.104502.
    1. de Wit E, van Doremalen N, Falzarano D, Munster VJ. 2016. SARS and MERS: recent insights into emerging coronaviruses. Nat Rev Microbiol 14:523–534. 10.1038/nrmicro.2016.81.
    1. Liu DX, Fung TS, Chong KK, Shukla A, Hilgenfeld R. 2014. Accessory proteins of SARS-CoV and other coronaviruses. Antiviral Res 109:97–109. 10.1016/j.antiviral.2014.06.013.
    1. Guan W-J, Ni Z-Y, Hu Y, Liang W-H, Ou C-Q, He J-X, Liu L, Shan H, Lei C-L, Hui DSC, Du B, Li L-J, Zeng G, Yuen K-Y, Chen R-C, Tang C-L, Wang T, Chen P-Y, Xiang J, Li S-Y, Wang J-L, Liang Z-J, Peng Y-X, Wei L, Liu Y, Hu Y-H, Peng P, Wang J-M, Liu J-Y, Chen Z, Li G, Zheng Z-J, Qiu S-Q, Luo J, Ye C-J, Zhu S-Y, Zhong N-S, China Medical Treatment Expert Group for Covid-19. 2020. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med 382:1708–1720. 10.1056/NEJMoa2002032.
    1. Carnevale S, Beretta P, Morbini P. 2020. Direct endothelial damage and vasculitis due to SARS-CoV-2 in small bowel submucosa of COVID-19 patient with diarrhea. J Med Virol 10:61–63. 10.1002/jmv.26119.
    1. Vrints CJM, Krychtiuk KA, Van Craenenbroeck EM, Segers VF, Price S, Heidbuchel H. 2020. Endothelialitis plays a central role in the pathophysiology of severe COVID-19 and its cardiovascular complications. Acta Cardiol 2:1–16. 10.1080/00015385.2020.1846921.
    1. Fraser DD, Patterson EK, Slessarev M, Gill SE, Martin C, Daley M, Miller MR, Patel MA, dos Santos CC, Bosma KJ, O’Gorman DB, Cepinskas G. 2020. Endothelial injury and glycocalyx degradation in critically ill coronavirus disease 2019 patients: implications for microvascular platelet aggregation. Crit Care Explor 2:e0194. 10.1097/CCE.0000000000000194.
    1. Guervilly C, Burtey S, Sabatier F, Cauchois R, Lano G, Abdili E, Daviet F, Arnaud L, Brunet P, Hraiech S, Jourde-Chiche N, Koubi M, Lacroix R, Pietri L, Berda Y, Robert T, Degioanni C, Velier M, Papazian L, Kaplanski G, Dignat-George F. 2020. Circulating endothelial cells as a marker of endothelial injury in severe COVID-19. J Infect Dis 222:1789–1793. 10.1093/infdis/jiaa528.
    1. Huertas A, Montani D, Savale L, Pichon J, Tu L, Parent F, Guignabert C, Humbert M. 2020. Endothelial cell dysfunction: a major player in SARS-CoV-2 infection (COVID-19)? Eur Respir J 56:2001634. 10.1183/13993003.01634-2020.
    1. Lei Y, Zhang J, Schiavon CR, He M, Chen L, Shen H, Zhang Y, Yin Q, Cho Y, Andrade L, Shadel GS, Hepokoski M, Lei T, Wang H, Zhang J, Yuan JX, Malhotra A, Manor U, Wang S, Yuan ZY, Shyy JY. 2021. SARS-CoV-2 spike protein impairs endothelial function via downregulation of ACE 2. Circ Res 128:1323–1326. 10.1161/circresaha.121.318902.
    1. Wang P, Luo R, Zhang M, Wang Y, Song T, Tao T, Li Z, Jin L, Zheng H, Chen W, Zhao M, Zheng Y, Qin J. 2020. A cross-talk between epithelium and endothelium mediates human alveolar-capillary injury during SARS-CoV-2 infection. Cell Death Dis 11:1042. 10.1038/s41419-020-03252-9.
    1. Teuwen LA, Geldhof V, Pasut A, Carmeliet P. 2020. COVID-19: the vasculature unleashed. Nat Rev Immunol 20:389–391. 10.1038/s41577-020-0343-0.
    1. Pons S, Fodil S, Azoulay E, Zafrani L. 2020. The vascular endothelium: the cornerstone of organ dysfunction in severe SARS-CoV-2 infection. Crit Care 24:353. 10.1186/s13054-020-03062-7.
    1. Lan J, Ge J, Yu J, Shan S, Zhou H, Fan S, Zhang Q, Shi X, Wang Q, Zhang L, Wang X. 2020. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 581:215–220. 10.1038/s41586-020-2180-5.
    1. Albini A, Di Guardo G, Noonan DM, Lombardo M. 2020. The SARS-CoV-2 receptor, ACE-2, is expressed on many different cell types: implications for ACE-inhibitor- and angiotensin II receptor blocker-based cardiovascular therapies. Intern Emerg Med 15:759–766. 10.1007/s11739-020-02364-6.
    1. Bhatnagar J, Gary J, Reagan-Steiner S, Estetter LB, Tong S, Tao Y, Denison AM, Lee E, deLeon-Carnes M, Li Y, Uehara A, Paden CR, Leitgeb B, Uyeki T, Martines RB, Ritter JM, Paddock CD, Shieh W-J, Zaki SR. 2021. Evidence of SARS-CoV-2 replication and tropism in the lungs, airways and vascular endothelium of patients with fatal COVID-19: an autopsy case-series. J Infect Dis 27:jiab039. 10.1093/infdis/jiab039.
    1. Korakas E, Ikonomidis I, Kousathana F, Balampanis K, Kountouri A, Raptis A, Palaiodimou L, Kokkinos A, Lambadiari V. 2020. Obesity and COVID-19: immune and metabolic derangement as a possible link to adverse clinical outcomes. Am J Physiol Endocrinol Metab 319:E105–E109. 10.1152/ajpendo.00198.2020.
    1. Lu R, Zhao X, Li J, Niu P, Yang B, Wu H, Wang W, Song H, Huang B, Zhu N, Bi Y, Ma X, Zhan F, Wang L, Hu T, Zhou H, Hu Z, Zhou W, Zhao L, Chen J, Meng Y, Wang J, Lin Y, Yuan J, Xie Z, Ma J, Liu WJ, Wang D, Xu W, Holmes EC, Gao GF, Wu G, Chen W, Shi W, Tan W. 2020. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet 395:565–574. 10.1016/S0140-6736(20)30251-8.
    1. Forster P, Forster L, Renfrew C, Forster M. 2020. Phylogenetic network analysis of SARS-CoV-2 genomes. Proc Natl Acad Sci USA 117:9241–9243. 10.1073/pnas.2004999117.
    1. Wawryk SO, Novotny JR, Wicks IP, Wilkinson D, Maher D, Salvaris E, Welch K, Fecondo J, Boyd AW. 1989. The role of the LFA-1/ICAM-1 interaction in human leukocyte homing and adhesion. Immunol Rev 108:135–161. 10.1111/j.1600-065x.1989.tb00016.x.
    1. Yong K, Khwaja A. 1990. Leucocyte cellular adhesion molecules. Blood Rev 4:211–225. 10.1016/0268-960X(90)90001-9.
    1. Fusté B, Mazzara R, Escolar G, Merino A, Ordinas A, Díaz-Ricart M. 2004. Granulocyte colony-stimulating factor increases expression of adhesion receptors on endothelial cells through activation of p38 MAPK. Haematologica 89:578–585.
    1. Iademarco MF, McQuillan JJ, Rosen GD, Dean DC. 1992. Characterization of the promoter for vascular cell adhesion molecule-1 (VCAM-1). J Biol Chem 267:16323–16329. 10.1016/S0021-9258(18)42004-2.
    1. Rodríguez C, Luque N, Blanco I, Sebastian L, Barberà JA, Peinado VI, Tura-Ceide O. 2021. Pulmonary endothelial dysfunction and thrombotic complications in COVID-19 patients. Am J Respir Cell Mol Biol 64:407–415. 10.1165/rcmb.2020-0359PS.
    1. Nascimento CJ, Schutt WR, Gorbunova EE, Mackow ER. 2020. Recombinant ACE2 expression is required for SARS-CoV-2 to infect primary human endothelial cells and induce inflammatory and procoagulative responses. mBio 11:e03185-20. 10.1128/mBio.03185-20.
    1. Khan A, Tahir Khan M, Saleem S, Junaid M, Ali A, Shujait Ali S, Khan M, Wei D-Q. 2020. Structural insights into the mechanism of RNA recognition by the N-terminal RNA-binding domain of the SARS-CoV-2 nucleocapsid phosphoprotein. Comput Struct Biotechnol J 18:2174–2184. 10.1016/j.csbj.2020.08.006.
    1. Chang CK, Hou MH, Chang CF, Hsiao CD, Huang TH. 2014. The SARS coronavirus nucleocapsid protein—forms and functions. Antiviral Res 103:39–50. 10.1016/j.antiviral.2013.12.009.
    1. Aboagye JO, Yew CW, Ng OW, Monteil VM, Mirazimi A, Tan YJ. 2018. Overexpression of the nucleocapsid protein of Middle East respiratory syndrome coronavirus up-regulates CXCL10. Biosci Rep 38:BSR20181059. 10.1042/BSR20181059.
    1. Zhu YG, Qu JM. 2009. Differential characteristics of the early stage of lung inflammation induced by SARS-CoV Nucleocapsid protein related to age in the mouse. Inflamm Res 58:312–320. 10.1007/s00011-009-8062-9.
    1. Gorbunova EE, Simons MJ, Gavrilovskaya IN, Mackow ER. 2016. The Andes virus nucleocapsid protein directs basal endothelial cell permeability by activating RhoA. mBio 7:e01747-16. 10.1128/mBio.01747-16.
    1. Bouhaddou M, Memon D, Meyer B, White KM, Rezelj VV, Correa Marrero M, Polacco BJ, Melnyk JE, Ulferts S, Kaake RM, Batra J, Richards AL, Stevenson E, Gordon DE, Rojc A, Obernier K, Fabius JM, Soucheray M, Miorin L, Moreno E, Koh C, Tran QD, Hardy A, Robinot R, Vallet T, Nilsson-Payant BE, Hernandez-Armenta C, Dunham A, Weigang S, Knerr J, Modak M, Quintero D, Zhou Y, Dugourd A, Valdeolivas A, Patil T, Li Q, Hüttenhain R, Cakir M, Muralidharan M, Kim M, Jang G, Tutuncuoglu B, Hiatt J, Guo JZ, Xu J, Bouhaddou S, Mathy CJP, Gaulton A, Manners EJ, et al.. 2020. The global phosphorylation landscape of SARS-CoV-2 infection. Cell 182:685–712.E19. 10.1016/j.cell.2020.06.034.
    1. Tong M, Jiang Y, Xia D, Xiong Y, Zheng Q, Chen F, Zou L, Xiao W, Zhu Y. 2020. Elevated expression of serum endothelial cell adhesion molecules in COVID-19 patients. J Infect Dis 222:894–898. 10.1093/infdis/jiaa349.
    1. Li T, Wang L, Wang H, Li X, Zhang S, Xu Y, Wei W. 2020. Serum SARS-CoV-2 nucleocapsid protein: a sensitivity and specificity early diagnostic marker for SARS-CoV-2 infection. Front Cell Infect Microb 10:470. 10.3389/fcimb.2020.00470.
    1. Gordon D. 2020. Statins may be a key therapeutic for Covid-19. Med Hypotheses 144:110001. 10.1016/j.mehy.2020.110001.
    1. Lima Martínez MM, Contreras MA, Marín W, D'Marco L. 2020. Statins in COVID-19: is there any foundation? Clin Invest Arterioscler 32:278–281. 10.1016/j.artere.2020.11.004.
    1. Teoh N, Farrell G. 2020. Statins as early therapy to mitigate COVID-19 (SARS-CoV-2)-associated ARDS and cytokine storm syndrome—time is of the essence. J Clin Transl Res 5:227–229.
    1. Tan WYT, Young BE, Lye DC, Chew DEK, Dalan R. 2020. Statin use is associated with lower disease severity in COVID-19 infection. Sci Rep 10:17458. 10.1038/s41598-020-74492-0.
    1. Zhang X-J, Qin J-J, Cheng X, Shen L, Zhao Y-C, Yuan Y, Lei F, Chen M-M, Yang H, Bai L, Song X, Lin L, Xia M, Zhou F, Zhou J, She Z-G, Zhu L, Ma X, Xu Q, Ye P, Chen G, Liu L, Mao W, Yan Y, Xiao B, Lu Z, Peng G, Liu M, Yang J, Yang L, Zhang C, Lu H, Xia X, Wang D, Liao X, Wei X, Zhang B-H, Zhang X, Yang J, Zhao G-N, Zhang P, Liu PP, Loomba R, Ji Y-X, Xia J, Wang Y, Cai J, Guo J, Li H. 2020. In-hospital use of statins is associated with a reduced risk of mortality among individuals with COVID-19. Cell Metab 32:176–187.E4. 10.1016/j.cmet.2020.06.015.
    1. Bifulco M, Gazzerro P. 2020. Statin therapy in COVID-19 infection: much more than a single pathway. Eur Heart J Cardiovasc Pharmacother 6:410–411. 10.1093/ehjcvp/pvaa055.
    1. Crespo MJ, Quidgley J. 2015. Simvastatin, atorvastatin, and pravastatin equally improve the hemodynamic status of diabetic rats. World J Diabetes 6:1168–1178. 10.4239/wjd.v6.i10.1168.
    1. Wang J, Jiang M, Chen X, Montaner LJ. 2020. Cytokine storm and leukocyte changes in mild versus severe SARS-CoV-2 infection: review of 3939 COVID-19 patients in China and emerging pathogenesis and therapy concepts. J Leukoc Biol 108:17–41. 10.1002/JLB.3COVR0520-272R.
    1. Sohn KM, Lee SG, Kim HJ, Cheon S, Jeong H, Lee J, Kim IS, Silwal P, Kim YJ, Paik S, Chung C, Park C, Kim YS, Jo EK. 2020. COVID-19 patients upregulate Toll-like receptor 4-mediated inflammatory signaling that mimics bacterial sepsis. J Korean Med Sci 35:e343. 10.3346/jkms.2020.35.e343.
    1. Li Y, Huang X, Guo F, Lei T, Li S, Monaghan-Nichols P, Jiang Z, Xin H-B, Fu M. 2020. TRIM65 E3 ligase targets VCAM-1 degradation to limit LPS-induced lung inflammation. J Mol Cell Biol 12:190–201. 10.1093/jmcb/mjz077.

Source: PubMed

3
Abonnieren