Does Angiotensin II Peak in Response to SARS-CoV-2?

Léder Leal Xavier, Paula Fernanda Ribas Neves, Lisiê Valeria Paz, Laura Tartari Neves, Pamela Brambilla Bagatini, Luís Fernando Saraiva Macedo Timmers, Alberto Antônio Rasia-Filho, Régis Gemerasca Mestriner, Andrea Wieck, Léder Leal Xavier, Paula Fernanda Ribas Neves, Lisiê Valeria Paz, Laura Tartari Neves, Pamela Brambilla Bagatini, Luís Fernando Saraiva Macedo Timmers, Alberto Antônio Rasia-Filho, Régis Gemerasca Mestriner, Andrea Wieck

Abstract

Human infection by the SARS-CoV-2 is causing the current COVID-19 pandemic. With the growing numbers of cases and deaths, there is an urgent need to explore pathophysiological hypotheses in an attempt to better understand the factors determining the course of the disease. Here, we hypothesize that COVID-19 severity and its symptoms could be related to transmembrane and soluble Angiotensin-converting enzyme 2 (tACE2 and sACE2); Angiotensin II (ANG II); Angiotensin 1-7 (ANG 1-7) and angiotensin receptor 1 (AT1R) activation levels. Additionally, we hypothesize that an early peak in ANG II and ADAM-17 might represent a physiological attempt to reduce viral infection via tACE2. This viewpoint presents: (1) a brief introduction regarding the renin-angiotensin-aldosterone system (RAAS), detailing its receptors, molecular synthesis, and degradation routes; (2) a description of the proposed early changes in the RAAS in response to SARS-CoV-2 infection, including biological scenarios for the best and worst prognoses; and (3) the physiological pathways and reasoning for changes in the RAAS following SARS-CoV-2 infection.

Keywords: Angiotensin-converting enzyme 2; COVID-19; SARS-CoV-2; angiotensin-II; immune activation; immune response.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Xavier, Neves, Paz, Neves, Bagatini, Timmers, Rasia-Filho, Mestriner and Wieck.

Figures

Figure 1
Figure 1
The roles of the RAAS in health and COVID-19: A better and worse scenarios—(A) Basal angiotensin system. Figure representing the classic RAAS pathway. Angiotensin I (ANG I) is produced from angiotensinogen by renin. ANG I is converted to Angiotensin II (ANG II) by Angiotensin-Converting Enzyme (ACE). ANG II exerts its actions by binding to AT1R. Angiotensin-Converting Enzyme 2 (ACE2), is responsible for converting ANG I to Angiotensin 1-9 (ANG 1-9) and ANG II to Angiotensin 1-7 (ANG 1-7). ANG 1-7 has opposite effects to ANG II, counterbalancing ANG II physiological outcome. ADAM 17 and TPMRSS2 are sheddases responsible for cleaving tACE2 into its soluble form, sACE2. ADAM17 cleaves tACE2 in a constitutively manner, while TMPRSS2 is implicated in SARS-CoV-2 infection, supporting virus entry the cell host. (B) COVID-19 Best Scenario (for a better understanding, follow the order of numbers). Increased TMPRSS2 activity due to viral entry resulting in reduced tACE2 and consequent increase in sACE2, unable to convert ANG II in ANG 1-7. This will lead to increased ANG II levels, and increased AT1R activation, resulting in higher ADAM-17 activity. Higher ADAM-17 activity will increase biologically active sACE2 levels, able to convert ANG II in ANG 1-7, resulting in no alteration in ANG 1-7 levels. In this scenario, the intense tACE2 cleavage promoted by ANG II/AT1R/ADAM-17 axis will reduce virus entry into the cells. (C) COVID-19 Worst Scenario Note that this scenario is closely related to an incapacity in promote the ANG II peak or AT1R down-regulation, as suggested to the use of ACE inhibitors, genetic factors (i.e. A blood type), diabetes mellitus and cardiovascular diseases. (for a better understanding, follow the order of numbers). Increased TMPRSS2 activity due to viral entry resulting in a mild reduction tACE2 as well as a mild increase in sACE2, unable to convert ANG II into ANG 1-7. Consequently, ANG II levels will remain equal or even decrease, as well as AT1R activation, and resulting in ANG 1-7 decrease. In this scenario, the equal or low levels of ANG II, AT1R, and ADAM-17 activities will be associated to a mild decrease in tACE2 and a substantial increase in sACE2 produced by TPMRSS2, increasing the virus entry into the cells.
Figure 2
Figure 2
Summary of proposed changes in the RAAS, and downstream effects following SARS-CoV-2 infection. In the best-case scenario, a peak of ANG II occurs immediately after SARS-CoV-2 infects the cell. The virus reduces tACE2 levels due to increased TMPRSS2 and ADAM-17 activity. Reduced tACE2 levels decrease virus entry into the cell, and, consequently, the viral load. The ANG II peak will also increase AT1R activation, modulating not only the ANG II effects, but also the host immune response, which produces TNF-α and IFN-γ in an appropriate time and amount, favoring better clinical outcomes. In the worst-case scenario, the host is unable to elevate ANG II levels. Even with reduced tACE2 due to virus TMPRSS2 activation, ANG II levels remain unaltered or reduced. Consequently, ADAM-17 activation is lower or unaltered, thus, more tACE2 is available, allowing more virus to enter the cell and replicate. Moreover, AT1R activation is also low or unaltered, which leads to an inappropriate early immune response, favoring poor clinical outcomes.
Figure 3
Figure 3
Angiotensin II and T lymphocytes activation during COVID-19 best scenario. (A) 1: Reduction in tACE2 following SARS-CoV-2 infection. 2: ANG II will increase and 3: act as a co-stimulatory molecule in T lymphocytes activation acting via AT1R, which are expressed in CD8+ cells. 4: ANG II is also expressed by T lymphocytes, enhancing AT1R activation in an autocrine and intracrine manner. 5:AT1R activation stimulates CD8+ cells to produce both TNF-α and IFN-γ. 6: TNF-α will increase TH1 differentiation, and, together with IFN-γ, activate the inflammatory response. 7: TNF- α will also activate ADAM-17 activity leading to 8: increase the biologically active sACE2 and, 9: maintaining ANG 1-7 levels. 10: ANG 1-7, which has anti-inflammatory characteristics, will counterbalance TNF-α and IFN-γ direct effects via MasR pathway. Taken together, these alterations would lead to a proper immune response to SARS-CoV-2 infection. (B) Angiotensin II and T lymphocytes activation during COVID-19 worse scenario. 1: tACE2 levels are reduced following SARS-CoV-2 infection. 2: ANG II levels remain the same or are even low (see Figure 1C, COVID-19 Worse Scenario), 3: maintaining the AT1R activation in CD8+ cells at normal levels. 4: As a result, IFN-γ and TNF-α levels remain the same, 5: affecting the beginning of inflammatory response. 6: ADAM-17 activity is equal or reduced, 7: leading to reduced biologically active sACE2 levels. 8: consequently, ANG 1-7 levels are reduced. 9: This unbalanced immune response could not be adequate to combat SARS-CoV-2 infection.

References

    1. Sohrabi C, Alsafi Z, O’Neill N, Khan M, Kerwan A, Al-Jabir A, et al. World Health Organization declares global emergency: A review of the 2019 novel coronavirus (COVID-19). Int J Surg (2020) 76:71–6. 10.1016/j.ijsu.2020.02.034
    1. Drawz P, Ghazi L. Advances in understanding the renin-angiotensin-aldosterone system (RAAS) in blood pressure control and recent pivotal trials of RAAS blockade in heart failure and diabetic nephropathy. F1000Research (2017) 6:1–10. 10.12688/f1000research.9692.1
    1. Ferrão FM. Renin-angiotensin system in the kidney: What is new? World J Nephrol (2014) 3:64–76. 10.5527/wjn.v3.i3.64
    1. Kashuba E, Bailey J, Allsup D, Cawkwell L. The kinin-kallikrein system: Physiological roles, pathophysiology and its relationship to cancer biomarkers. Biomarkers (2013) 18:279–96. 10.3109/1354750X.2013.787544
    1. Kuoppala A, Lindstedt KA, Saarinen J, Kovanen PT, Kokkonen JO. Inactivation of bradykinin by angiotensin-converting enzyme and by carboxypeptidase N in human plasma. Am J Physiol Hear Circ Physiol (2000) 278:1069–74. 10.1152/ajpheart.2000.278.4.h1069
    1. Fuchs S, Xiao HD, Hubert C, Michaud A, Campbell DJ, Adams JW, et al. Angiotensin-converting enzyme C-terminal catalytic domain is the main site of angiotensin I cleavage in vivo. Hypertension (2008) 51:267–74. 10.1161/HYPERTENSIONAHA.107.097865
    1. Jaspard E, Wei L, Alhenc-Gelas F. Differences in the properties and enzymatic specificities of the two active sites of angiotensin I-converting enzyme (kininase II). Studies with bradykinin and other natural peptides. J Biol Chem (1993) 268:9496–503.
    1. Atlas SA. The Renin-Angiotensin Aldosterone System: Pathophysiological Role and Pharmacologic Inhibition. J Manag Care Pharm (2007) 13:9–20. 10.18553/jmcp.2007.13.s8-b.9
    1. Ihara M, Urata H, Kinoshita A, Suzumiya J, Sasaguri M, Kikuchi M, et al. Increased chymase-dependent angiotensin II formation in human atherosclerotic aorta. Hypertension (1999) 33(6):1399–405. 10.1161/01.HYP.33.6.1399
    1. Kramkowski K, Mogielnicki A, Buczko W. The physiological significance of the alternative pathways of angiotensin II production. J Physiol Pharmacol (2006) 57(4):529–39.
    1. Crowley SD, Gurley SB, Herrera MJ, Ruiz P, Griffiths R, Kumar AP, et al. Angiotensin II causes hypertension and cardiac hypertrophy through its receptors in the kidney. Proc Natl Acad Sci USA (2006) 103:17985–90. 10.1073/pnas.0605545103
    1. Zisman LS. ACE and ACE2: A tale of two enzymes. Eur Heart J (2005) 26:322–4. 10.1093/eurheartj/ehi043
    1. Verdecchia P, Cavallini C, Spanevello A, Angeli F. The pivotal link between ACE2 deficiency and SARS-CoV-2 infection. Eur J Intern Med (2020) 76:0–1. 10.1016/j.ejim.2020.04.037
    1. Kuba K, Imai Y, Rao S, Gao H, Guo F, Guan B, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat Med (2005) 11:875–9. 10.1038/nm1267
    1. Sungnak W, Huang N, Bécavin C, Berg M, Queen R, Litvinukova M. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat Med (2020) 26:681–7. 10.1038/s41591-020-0868-6
    1. Hong PJ, Look DC, Tan P, Shi L, Hickey M, Gakhar L, et al. Ectodomain shedding of angiotensin converting enzyme 2 in human airway epithelia. Am J Physiol Lung Cell Mol Physiol (2009) 297:84–96. 10.1152/ajplung.00071.2009
    1. Black RA, Rauch CT, Kozlosky CJ, Peschon JJ, Slack JL, Wolfson MF, et al. A metalloproteinase disintegrin that releases tumour-necrosis factor-a from cells. Lett Nat (1997) 385120:729–33. 10.1038/385729a0
    1. Lambert DW, Yarski M, Warner FJ, Thornhill P, Parkin ET, Smith AI, et al. Tumor necrosis factor-α convertase (ADAM17) mediates regulated ectodomain shedding of the severe-acute respiratory syndrome-coronavirus (SARS-CoV) receptor, angiotensin-converting enzyme-2 (ACE2). J Biol Chem (2005) 280:30113–9. 10.1074/jbc.M505111200
    1. Zipeto D, Palmeira J da F, Argañaraz GA, Argañaraz ER. ACE2/ADAM17/TMPRSS2 Interplay May Be the Main Risk Factor for COVID-19. Front Immunol (2020) 11. 10.3389/fimmu.2020.576745
    1. Zoufaly A, Poglitsch M, Aberle JH, Hoepler W, Seitz T, Traugott M, et al. Human recombinant soluble ACE2 in severe COVID-19. Lancet Respir Med (2020) 8:1154–8. 10.1016/S2213-2600(20)30418-5
    1. 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) 88:1293–307. 10.1128/jvi.02202-13
    1. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell (2020) 181:271–80.e8. 10.1016/j.cell.2020.02.052
    1. Turner AJ, Hiscox JA, Hooper NM. ACE2: From vasopeptidase to SARS virus receptor. Trends Pharmacol Sci (2004) 25:291–4. 10.1016/j.tips.2004.04.001
    1. Shang J, Wan Y, Luo C, Ye G, Geng Q, Auerbach A, et al. Cell entry mechanisms of SARS-CoV-2. Proc Natl Acad Sci USA (2020) 2020:1–8. 10.1073/pnas.2003138117
    1. Stopsack KH, Mucci LA, Antonarakis ES, Nelson PS, Kantoff PW. TMPRSS2 and COVID-19: Serendipity or opportunity for intervention? Cancer Discovery (2020) 10(6):779–82. 10.1158/-20-0451
    1. Böttcher E, Matrosovich T, Beyerle M, Klenk H-D, Garten W, Matrosovich M. Proteolytic Activation of Influenza Viruses by Serine Proteases TMPRSS2 and HAT from Human Airway Epithelium. J Virol (2006) 80:9896–8. 10.1128/jvi.01118-06
    1. Bestle D, Heindl MR, Limburg H, van TVL, Pilgram O, Moulton H, et al. TMPRSS2 and furin are both essential for proteolytic activation and spread of SARS-CoV-2 in human airway epithelial cells and provide promising drug targets. bioRxiv (2020) 3(9). 10.1101/2020.04.15.042085
    1. Liu M, Wang T, Zhou Y, Zhao Y, Zhang Y, Li J. Potential Role of ACE2 in Coronavirus Disease 2019 (COVID-19) Prevention and Management. J Transl Intern Med (2020) 8:9–19. 10.2478/jtim-2020-0003
    1. Zemlin AE, Wiese OJ. Coronavirus disease 2019 (COVID-19) and the renin-angiotensin system: A closer look at angiotensin-converting enzyme 2 (ACE2). Ann Clin Biochem (2020) 0:1–12. 10.1177/0004563220928361
    1. Mohamed Saleem TS, Bharani K, Gauthaman K. ACE inhibitors - Angiotensin II receptor antagonists: A useful combination therapy for ischemic heart disease. Open Access Emerg Med (2010) 2:51–9. 10.2147/oaem.s10507
    1. Wösten-van Asperen RM, Lutter R, Specht PA, Moll GN, van Woensel JB, van der Loos CM, et al. Acute respiratory distress syndrome leads to reduced ratio of ACE/ACE2 activities and is prevented by angiotensin-(1-7) or an angiotensin II receptor antagonist. J Pathol (2011) 225:618—627. 10.1002/path.2987
    1. Gallagher PE, Ferrario CM, Tallant EA. MAP kinase/phosphatase pathway mediates the regulation of ACE2 by angiotensin peptides. Am J Physiol Cell Physiol (2008) 295:1169–74. 10.1152/ajpcell.00145.2008
    1. Tanaka Y, Tamura K, Koide Y, Sakai M, Tsurumi Y, Noda Y, et al. The novel angiotensin II type 1 receptor (AT1R)-associated protein ATRAP downregulates AT1R and ameliorates cardiomyocyte hypertrophy. FEBS Lett (2005) 579:1579–86. 10.1016/j.febslet.2005.01.068
    1. Imayama I, Ichiki T, Inanaga K, Ohtsubo H, Fukuyama K, Ono H, et al. Telmisartan downregulates angiotensin II type 1 receptor through activation of peroxisome proliferator-activated receptor γ. Cardiovasc Res (2006) 72:184–90. 10.1016/j.cardiores.2006.07.014
    1. Ichiki T, Usui M, Kato M, Unakoshi Y, Ito K, Egashira K, et al. Downregulation of Angiotensin II type 1 recptor gene transcription by nitric oxide. Am Hear Assoclatlon (1997) 10:342–8. 10.1161/01.HYP.31.1.342
    1. Glowacka I, Bertram S, Herzog P, Pfefferle S, Steffen I, Muench MO, et al. Differential Downregulation of ACE2 by the Spike Proteins of Severe Acute Respiratory Syndrome Coronavirus and Human Coronavirus NL63. J Virol (2010) 84:1198–205. 10.1128/jvi.01248-09
    1. Kai H, Kai M. Interactions of coronaviruses with ACE2, angiotensin II, and RAS inhibitors—lessons from available evidence and insights into COVID-19. Hypertens Res (2020) 1–7:648–54. 10.1038/s41440-020-0455-8
    1. Xu H, Zhong L, Deng J, Peng J, Dan H, Zeng X, et al. High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int J Oral Sci (2020) 12:1–5. 10.1038/s41368-020-0074-x
    1. Ellinghaus D, Degenhardt F, Bujanda L, Buti M, Albillos A, Invernizzi P, et al. Genomewide Association Study of Severe Covid-19 with Respiratory Failure. N Engl J Med (2020) 383(16):1–13. 10.1056/NEJMoa2020283
    1. Gallagher PE, Chappell MC, Ferrario CM, Tallant EA. Distinct roles for ANG II and ANG-(1-7) in the regulation of angiotensin-converting enzyme 2 in rat astrocytes. Am J Physiol Cell Physiol (2006) 290:420–6. 10.1152/ajpcell.00409.2004
    1. Ghosal S, Mukherjee JJ, Sinha B, Gangopadhyay KK. The effect of angiotensin converting enzyme inhibitors and angiotensin receptor blockers on death and severity of disease in patients with coronavirus disease 2019 (COVID-19): A meta-analysis. merRxiv (2020). 10.1101/2020.04.23.20076661
    1. Kuster GM, Pfister O, Burkard T, Zhou Q, Twerenbold R, Haaf P, et al. SARS-CoV2: should inhibitors of the renin-angiotensin system be withdrawn in patients with COVID-19? Eur Heart J (2020) 41(19):1801–3. 10.1093/eurheartj/ehaa235
    1. Mackey K, King VJ, Gurley S, Kiefer M, Liederbauer E, Vela K, et al. Risks and Impact of Angiotensin-Converting Enzyme Inhibitors or Angiotensin-Receptor Blockers on SARS-CoV-2 Infection in Adults. Ann Intern Med (2020) 1–10. 10.7326/m20-1515
    1. Liu Y, Yang Y, Zhang C, Huang F, Wang F, Yuan J, et al. Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury. Sci China Life Sci (2020) 63:364–74. 10.1007/s11427-020-1643-8
    1. Guo J, Huang F, Liu J, Chen Y, Wang W, Cao B, et al. The serum profile of hypercytokinemia factors identified in H7N9-infected patients can predict fatal outcomes. Sci Rep (2015) 5:1–10. 10.1038/srep10942
    1. Huang C, Wang Y, Li X, Ren L, Zhao J, Zhang L, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet (2020) 395:497–506. 10.1016/S0140-6736(20)30183-5
    1. Liu X, Yang N, Tang J, Liu S, Luo D, Duan Q, et al. Downregulation of angiotensin-converting enzyme 2 by the neuraminidase protein of influenza A (H1N1) virus. Virus Res (2014) 185:64–71. 10.1016/j.virusres.2014.03.010
    1. Zou Z, Yan Y, Shu Y, Gao R, Sun Y, Li X, et al. Angiotensin-converting enzyme 2 protects from lethal avian influenza A H5N1 infections. Nat Commun (2014) 5:1–7. 10.1038/ncomms4594
    1. Li W, Wong S-K, Li F, Kuhn JH, Huang I-C, Choe H, et al. Animal Origins of the Severe Acute Respiratory Syndrome Coronavirus: Insight from ACE2-S-Protein Interactions. J Virol (2006) 80:4211–9. 10.1128/jvi.80.9.4211-4219.2006
    1. Sheahan T, Rockx B, Donaldson E, Sims A, Pickles R, Corti D, et al. Mechanisms of Zoonotic Severe Acute Respiratory Syndrome Coronavirus Host Range Expansion in Human Airway Epithelium. J Virol (2008) 82:2274–85. 10.1128/jvi.02041-07
    1. Li W, Moore MJ, Vasilieva N, Sui J. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Lett Nat (2003) 426:450–4. 10.1038/nature02145
    1. Demogines A, Farzan M, Sawyer SL. Evidence for ACE2-Utilizing Coronaviruses (CoVs) Related to Severe Acute Respiratory Syndrome CoV in Bats. J Virol (2012) 86:6350–3. 10.1128/jvi.00311-12
    1. Yang CW, Lu LC, Chang CC, Cho CC, Hsieh WY, Tsai CH, et al. Imbalanced plasma ace and ace2 level in the uremic patients with cardiovascular diseases and its change during a single hemodialysis session. Ren Fail (2017) 39:719–28. 10.1080/0886022X.2017.1398665
    1. Magalhaes GS, Barroso LC, Reis AC, Rodrigues-Machado MG, Gregório JF, Motta-Santos D, et al. Angiotensin-(1-7) promotes resolution of eosinophilic inflammation in an experimental model of asthma. Front Immunol (2018) 9:58. 10.3389/fimmu.2018.00058
    1. Hao LY, Giasson BI, Bonini NM. DJ-1 is critical for mitochondrial function and rescues PINK1 loss of function. Proc Natl Acad Sci USA (2010) 107:9747–52. 10.1073/pnas.0911175107
    1. Soro-Paavonen A, Gordin D, Forsblom C, Rosengard-Barlund M, Waden J, Thorn L, et al. Circulating ACE2 activity is increased in patients with type 1 diabetes and vascular complications. J Hypertens (2012) 30:375–83. 10.1097/HJH.0b013e32834f04b6
    1. Ramchand J, Patel SK, Srivastava PM, Farouque O, Burrell LM. Elevated plasma angiotensin converting enzyme 2 activity is an independent predictor of major adverse cardiac events in patients with obstructive coronary artery disease. PloS One (2018) 13:1–11. 10.1371/journal.pone.0198144
    1. Sattar N, McInnes IB, McMurray JJV. Obesity a Risk Factor for Severe COVID-19 Infection: Multiple Potential Mechanisms. Circulation (2020) 44:1–8. 10.1161/circulationaha.120.047659
    1. Fang L, Karakiulakis G, Roth M. Are patients with hypertension and diabetes mellitus at increased risk for COVID-19 infection? The. Lancet Respir Med (2020) 8:e21. 10.1016/S2213
    1. Patel BM, Mehta AA. Aldosterone and angiotensin: Role in diabetes and cardiovascular diseases. Eur J Pharmacol (2012) 697:1–12. 10.1016/j.ejphar.2012.09.034
    1. Meira E, Cruz M, Miyazawa M, Gozal D. Putative contributions of circadian clock and sleep in the context of SARS-CoV-2 infection. Eur Respir J (2020) 55:1–7. 10.1183/13993003.01023-2020
    1. Cugini P, Lucia P, Centanni M. Circadian rhythms of plasma renin, aldosterone and cortisol. Effect of beta-adrenergic blockade by propranolol in patients with recent, advanced and complicated forms of essential hypertension. Chronobiologia (1982) 9(2):229–35.
    1. Cugini P, Lucia P, Tomassini R, Letizia C, Murano G, Scavo D, et al. Temporal correlation of some endocrine circadian rhythms in elderly subjects. Maturitas (1985) 7:175–86. 10.1016/0378-5122(85)90024-6
    1. Mochel JP, Fink M, Peyrou M, Desevaux C, Deurinck M, Giraudel JM, et al. Chronobiology of the renin-angiotensin-aldosterone system in dogs: Relation to blood pressure and renal physiology. Chronobiol Int (2013) 30:1144–59. 10.3109/07420528.2013.807275
    1. Herichova I, Zsoldosova K, Vesela A, Zeman M. Effect of angiotensin II infusion on rhythmic clock gene expression and local renin-angiotensin system in the aorta of Wistar rats. Endocr Regul (2014) 48:144–51. 10.4149/endo_2014_03_144
    1. Mazzoccoli G, Vinciguerra M, Carbone A, Relógio A. The circadian clock, the immune system, and viral infections: The intricate relationship between biological time and host-virus interaction. Pathogens (2020) 9. 10.3390/pathogens9020083
    1. Cao X. COVID-19: immunopathology and its implications for therapy. Nat Rev Immunol (2019) 2019:2019–20. 10.1038/s41577-020-0308-3
    1. Ruiz-Ortega M, Lorenzo O, Suzuki Y, Rupérez M, Egido J. Proinflammatory actions of angiotensins. Curr Opin Nephrol Hypertens (2001) 10(3):321–9. 10.1097/00041552-200105000-00005
    1. Tayeh MA, Scicli AG. Angiotensin II and bradykinin regulate the expression of P-selectin on the surface of endothelial cells in culture. Proc Assoc Am Physicians (1998) 110(5):412–21.
    1. Pastore L, Tessitore A, Martinotti S, Toniato E, Alesse E, Bravi MC, et al. Angiotensin II stimulates intercellular adhesion molecule-1 (ICAM-1) expression by human vascular endothelial cells and increases soluble ICAM-1 release in vivo. Circulation (1999) 100(15):1646–52. 10.1161/01.CIR.100.15.1646
    1. Sodhi CP, Nguyen J, Yamaguchi Y, Werts AD, Lu P, Ladd MR, et al. A Dynamic Variation of Pulmonary ACE2 Is Required to Modulate Neutrophilic Inflammation in Response to Pseudomonas aeruginosa Lung Infection in Mice. J Immunol (2019) 203(11):3000–12. 10.4049/jimmunol.1900579
    1. Wang K, Gheblawi M, Oudit GY. Angiotensin Converting Enzyme 2: A Double-Edged Sword. Circulation (2020) 142(5). 10.1161/CIRCULATIONAHA.120.047049
    1. Leow MKS. Clarifying the controversial risk-benefit profile of soluble ACE2 in COVID-19. Crit Care (2020) 24(1). 10.1186/s13054-020-03097-w
    1. Coppo M, Boddi M, Bandinelli M, Degl’Innocenti D, Ramazzotti M, Marra F, et al. Angiotensin II upregulates renin-angiotensin system in human isolated T lymphocytes. Regul Pept (2008) 151:1–6. 10.1016/j.regpep.2008.07.010
    1. Hoch NE, Guzik TJ, Chen W, Deans T, Maalouf SA, Gratze P, et al. Regulation of T-cell function by endogenously produced angiotensin II. Am J Physiol Regul Integr Comp Physiol (2009) 296:208–16. 10.1152/ajpregu.90521.2008
    1. Silva-Filho JL, Caruso-Neves C, Pinheiro AAS. Angiotensin II type-1 receptor (AT1 R) regulates expansion, differentiation, and functional capacity of antigen-specific CD8+ T cells. Sci Rep (2016) 6:1–17. 10.1038/srep35997
    1. Platten M, Youssef S, Eun MH, Ho PP, Han MH, Lanz TV, et al. Blocking angiotensin-converting enzyme induces potent regulatory T cells and modulates TH1- and TH17-mediated autoimmunity. Proc Natl Acad Sci USA (2009) 106:14948–53. 10.1073/pnas.0903958106
    1. Soy M, Keser G, Atagündüz P, Tabak F, Atagündüz I, Kayhan S. Cytokine storm in COVID-19: pathogenesis and overview of anti-inflammatory agents used in treatment. Clin Rheumatol (2020) 39(7):2085–94. 10.1007/s10067-020-05190-5
    1. Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, et al. Angiotensin II signal transduction: An update on mechanisms of physiology and pathophysiology. Physiol Rev (2018) 98:1627–738. 10.1152/physrev.00038.2017
    1. Grivennikov SI, Tumanov AV, Liepinsh DJ, Kruglov AA, Marakusha BI, Shakhov AN, et al. Distinct and nonredundant in vivo functions of TNF produced by T cells and macrophages/neutrophils: Protective and deleterious effects. Immunity (2005) 22:93–104. 10.1016/j.immuni.2004.11.016
    1. Singh A, Suresh M. A role for TNF in limiting the duration of CTL effector phase and magnitude of CD8 T cell memory. J Leukoc Biol (2007) 82:1201–11. 10.1189/jlb.0407240
    1. Jurewicz M, McDermott DH, Sechler JM, Tinckam K, Takakura A, Carpenter CB, et al. Human T and natural killer cells possess a functional renin-angiotensin system: Further mechanisms of angiotensin II-induced inflammation. J Am Soc Nephrol (2007) 18:1093–102. 10.1681/ASN.2006070707
    1. Kindler E, Thiel V. SARS-CoV and IFN: Too Little, Too Late Eveline. Cell Host Microbe Prev (2020) 19:139–41. 10.1016/j.chom.2016.01.012
    1. Channappanavar R, Fehr AR, Zheng J, Wohlford-Lenane C, Abrahante JE, Mack M, et al. IFN-I response timing relative to virus replication determines MERS coronavirus infection outcomes. J Clin Invest (2019) 129:3625–39. 10.1172/JCI126363
    1. Wen W, Su W, Tang H, Le W, Zhang X, Zheng Y, et al. Immune cell profiling of COVID-19 patients in the recovery stage by single-cell sequencing. Cell Discovery (2020) 6(31). 10.1038/s41421-020-0168-9
    1. Catanzaro M, Fagiani F, Racchi M, Corsini E, Govoni S, Lanni C. Immune response in COVID-19: addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2. Signal Transd Target Ther (2020) 5:84. 10.1038/s41392-020-0191-1
    1. Urra JM, Cabrera CM, Porras L, Ródenas I. Since January 2020 Elsevier has created a COVID-19 resource centre with free information in English and Mandarin on the novel coronavirus COVID-19. The COVID-19 resource centre is hosted on Elsevier Connect, the company’s public news and information. Clin Immunol (2020) 217:108486. 10.1016/j.clim.2020.108486
    1. Xiong Y, Liu Y, Cao L, Wang D, Guo M, Jiang A, et al. Transcriptomic characteristics of bronchoalveolar lavage fluid and peripheral blood mononuclear cells in COVID-19 patients. Emerg Microbes Infect (2020) 9:761–70. 10.1080/22221751.2020.1747363
    1. Zheng M, Gao Y, Wang G, Song G, Liu S, Sun D, et al. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell Mol Immunol (2020) 17:533–5. 10.1038/s41423-020-0402-2
    1. Zheng HY, Zhang M, Yang CX, Zhang N, Wang XC, Yang XP, et al. Elevated exhaustion levels and reduced functional diversity of T cells in peripheral blood may predict severe progression in COVID-19 patients. Cell Mol Immunol (2020) 17:541–3. 10.1038/s41423-020-0401-3

Source: PubMed

3
Tilaa