Skip to main content
Springer Nature - PMC COVID-19 Collection logoLink to Springer Nature - PMC COVID-19 Collection
. 2020 May 5;69(7):641–643. doi: 10.1007/s00011-020-01353-x

SARS-CoV-2-mediated inflammatory response in lungs: should we look at RAGE?

Armando Rojas 1,, Ileana Gonzalez 1, Miguel A Morales 2
PMCID: PMC7200049  PMID: 32372149

In December 2019, a new type of coronavirus pneumonia (COVID-19) emerged in Wuhan, China, and spread rapidly all over the world, forcing the World Health Organization to officially declare on 30 January 2020, the COVID-19 as a global pandemic. Lung inflammation is the main cause of life-threatening respiratory disorders at the COVID-19 severe stage [1, 2].

The etiological agent of this new pandemic is a novel coronavirus, the SARS-CoV2, which uses the angiotensin converting enzyme 2 (ACE2) molecule as the receptor for viral cell entry [3]. ACE2 plays an important role in the renin–angiotensin system (RAS), and the imbalance between ACE/Ang II/AT1R pathway and ACE2/Ang (1–7)/Mas receptor pathway in the RAS system will lead to multi-system inflammation [4].

It is well known that increased ACE and Ang II are poor prognostic factors for severe pneumonia [5]. Conversely, different studies including systematic review and meta-analysis have shown that ACE inhibitors/ARBs have a protective role [6, 7]. Furthermore, inpatient use of ACEI/ARB in hypertensive hospitalized COVID-19 patients has been recently associated with lower risk of all-cause mortality compared with ACEI/ARB non-users [8].

Activation of the angiotensin II receptor type 1 (AT1R) by Ang II leads to the induction of NF-κB [9, 10], and subsequent inflammation through pathways distinct from those mediating classical Gq-induced signaling [11].

The receptor for advanced glycation end-products (RAGE), initially recognized for its ability to bind to Advanced Glycation End-products (AGEs), was subsequently found to be a pattern recognition receptor able to recognize several danger signals, including high mobility group box-1 (HMGB1)/amphoterin, S100/calgranulins, and amyloid-β peptide [12, 13].

At present, this multiligand pattern recognition receptor is considered as a key molecule in the onset and sustainment of the inflammatory response in many clinical entities [1417]. Furthermore, activation of RAGE causes not only an inflammatory gene expression profile but also a positive feed-forward loop, in which inflammatory stimuli activate NF-κB, which induces RAGE expression, followed by a sustained NF-κB activation [18].

The signaling cascades triggered by RAGE engagement are much more complex and diverse than initially thought, considering that RAGE-binding proteins located in either the cytoplasm and or on the plasma membrane can modulate RAGE-mediated signaling diversity, in addition to the conformational flexibility acquired after the engagement, ranging from homo-dimerization, homo-multimerization and even to hetero-dimerization [19, 20].

Noteworthy, a cognate ligand-independent mechanism for RAGE transactivation has been recently reported to occur following activation of the AT1R, in different cell types [21]. Activation of the AT1R by angiotensin II (Ang II) triggered the transactivation of the cytosolic tail of RAGE and NF-κB-driven proinflammatory gene expression, independent of the liberation of RAGE ligands or the ligand-binding ectodomain of RAGE. Furthermore, the adverse proinflammatory signaling events induced by AT1 receptor activation were attenuated when RAGE was deleted or transactivation of its cytosolic tail was inhibited.

At this point, it is important to highlight that RAGE is expressed at a low basal level in most healthy adult tissues, and its expression is up regulated during pathologic processes. However, pulmonary tissues express remarkably high basal levels of RAGE, where it seem to play a homeostatic physiological role in tissue morphology [22].

Although RAGE has been defined as a specific marker of AT1 cells, after cell injury [23], RAGE may also be expressed in type 2 alveolar epithelial (AT2) cells [24]. In addition to lung epithelium, RAGE expression has also been noted in many crucial cell types in lung physiology, such as vascular smooth muscle cells [25], airway smooth muscle cells [26], and endothelial cells [27].

Considering the abundance of both AT1R and RAGE expression in lungs, the RAGE transactivation produced by Ang II-mediated AT1R activation can run continuously; while, the virus-mediated imbalance of the ACE/Ang II/AT1R pathway is being produced by the binding of SARS-CoV-2 to ACE-2 molecules, and, thus, limiting its function as a RAS counter-regulator.

This new transactivation mechanism opens new questions, considering that RAGE is a highly polymorphic protein, on the possibility that some polymorphisms can alter these intermolecular protein–protein interactions. Furthermore, Ang II exerts several cytokine-like actions via the AT1R and by transactivation of several growth factor receptors, including EGF, platelet-derived growth factor, and IGF receptors [28, 29]. These conditions may then render a wide range of biological responses, as we are seeing in patients affected by COVID-19, where not all infected patients develop a severe respiratory illness.

Due to the compelling body of evidence supporting a crucial role of RAGE activation in many clinical entities, many efforts have been done to inhibit RAGE signaling, and although a very extensive variety of compounds of the most dissimilar nature has been reported as capable of inhibiting RAGE signaling, only a few have been evaluated in clinical trials [30]. Due to the magnitude of this pandemic and its associated costs, and considering that lung injury with severe respiratory failure is the leading cause of death in COVID-19, science cannot afford to rule out any approach to confront this daunting scenario. Although, many vaccine candidates are under development and different anti-RNA viral drugs clinical trials are in course, due to the current urgency to stop the pandemic, it is important to highlight that the more the knowledge generated about inflammatory bronchoalveolar pathophysiology of this disease, the greater the success of the rational design and/or the use of drugs for its treatment.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References.

  • 1.Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y. Clinical features of patients infected with 2019 novel coronavirus in Wuhan China. Lancet. 2020;395:497–506. doi: 10.1016/S0140-6736(20)30183-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lai CC, Shih TP, Ko WC, Tang HJ, Hsueh PR. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease-2019 (COVID-19): the epidemic and the challenges. Int J Antimicrob Agents. 2020;55:105924. doi: 10.1016/j.ijantimicag.2020.105924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, Müller MA, Drosten C, Pöhlmann S. SARS-CoV-2 cell entry depends on ace2 and tmprss2 and is blocked by a clinically proven protease inhibitor. Cell. 2020 doi: 10.1016/j.cell.2020.02.05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Mirabito-Colafella KM, Uijl E, Danser AH. Interference with the Renin-Angiotensin System (RAS): classical inhibitors and novel approaches. Encycl Endocr Dis. 2019 doi: 10.1016/b978-0-12-801238-3.65341-2. [DOI] [Google Scholar]
  • 5.van de Garde EM, Souverein PC, van den Bosch JM, Deneer VH, Leufkens HG. Angiotensin-converting enzyme inhibitor use and pneumonia risk in a general population. Eur Respir J. 2006;27:1217–1222. doi: 10.1183/09031936.06.00110005. [DOI] [PubMed] [Google Scholar]
  • 6.Caldeira D, Alarcão J, Vaz-Carneiro A, Costa J. Risk of pneumonia associated with use of angiotensin converting enzyme inhibitors and angiotensin receptor blockers: systematic review and meta-analysis. BMJ (Clin Res Ed) 2012;345:e4260. doi: 10.1136/bmj.e4260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Liu CL, Shau WY, Chang CH, Wu CS, Lai MS. Pneumonia risk and use of angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers. J Epidemiol. 2013;23:344–350. doi: 10.2188/jea.JE20120112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zhang P, Zhu L, Cai J, Lei F, Qin JJ, et al. Association of inpatient use of angiotensin converting enzyme inhibitors and angiotensin ii receptor blockers with mortality among patients with hypertension hospitalized with COVID-19. Circ Res. 2020 doi: 10.1161/CIRCRESAHA.120.317134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ruiz-Ortega M, Rupérez M, Esteban V, Rodriguez-Vita J, Sanchez-López E, Egido J. Modulation of angiotensin II effects, a potential novel approach to inflammatory and immune diseases. Curr Med Chem. 2003;2:379–394. [Google Scholar]
  • 10.Li XC, Zhuo JL. Nuclear factor-kappa B as a hormonal intracellular signaling molecule: focus on angiotensin II-induced cardiovascular and renal injury. Curr Opin Nephrol Hypertens. 2008;17:37–43. doi: 10.1097/MNH.0b013e3282f2903c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Seta K, Nanamori M, Modrall JG, Neubig RR, Sadoshima J. AT1 receptor mutant lacking heterotrimeric G protein coupling activates the Src-Ras-ERK pathway without nuclear translocation of ERKs. J Biol Chem. 2002;277:9268–9277. doi: 10.1074/jbc.M109221200. [DOI] [PubMed] [Google Scholar]
  • 12.Rojas A, Delgado-López F, González I, Pérez-Castro R, Romero J, Rojas I. The receptor for advanced glycation end-products: a complex signaling scenario for a promiscuous receptor. Cell Signal. 2013;25:609–614. doi: 10.1016/j.cellsig.2012.11.022. [DOI] [PubMed] [Google Scholar]
  • 13.González I, Romero J, Rodríguez BL, Pérez-Castro R, Rojas A. The immunobiology of the receptor of advanced glycation end-products: trends and challenges. Immunobiology. 2013;218:790–797. doi: 10.1016/j.imbio.2012.09.005. [DOI] [PubMed] [Google Scholar]
  • 14.Rojas A, Figueroa H, Morales E. Fueling inflammation at tumor microenvironment: the role of multiligand/RAGE axis. Carcinogenesis. 2010;31:334–341. doi: 10.1093/carcin/bgp322. [DOI] [PubMed] [Google Scholar]
  • 15.Rojas A, Mercadal E, Figueroa H, Morales MA. Advanced Glycation and ROS: a link between diabetes and heart failure. Curr Vasc Pharmacol. 2008;6:44–51. doi: 10.2174/157016108783331312. [DOI] [PubMed] [Google Scholar]
  • 16.D'Agati V, Schmidt AM. RAGE and the pathogenesis of chronic kidney disease. Nat Rev Nephrol. 2010;6:352–360. doi: 10.1038/nrneph.2010.54. [DOI] [PubMed] [Google Scholar]
  • 17.Yan SF, Ramasamy R, Schmidt AM. Mechanisms of disease: advanced glycation end-products and their receptor in inflammation and diabetes complications. Nat Clin Pract Endocrinol Metab. 2008;4:285–293. doi: 10.1038/ncpendmet0786. [DOI] [PubMed] [Google Scholar]
  • 18.Bierhaus A, Schiekofer S, Schwaninger M, Andrassy M, Humpert PM, Chen J, Hong M, Luther T, Henle T, Klöting I, Morcos M, Hofmann M, Tritschler H, Weigle B, Kasper M, Smith M, Perry G, Schmidt AM, Stern DM, Häring HU, Schleicher E, Nawroth PP. Diabetes-associated sustained activation of the transcription factor nuclear factor-kappa B. Diabetes. 2001;50:2792–2808. doi: 10.2337/diabetes.50.12.2792. [DOI] [PubMed] [Google Scholar]
  • 19.Sakaguchi M, Murata H, Aoyama Y, Hibino T, Putranto EW, Ruma IM, Inoue Y, Sakaguchi Y, Yamamoto K, Kinoshita R, et al. DNAX-activating protein 10 (DAP10) membrane adaptor associates with receptor for advanced glycation end products (RAGE) and modulates the RAGE-triggered signaling pathway in human keratinocytes. J Biol Chem. 2014;289:23389–23402. doi: 10.1074/jbc.M114.573071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sakaguchi M, Murata H, Yamamoto K, Ono T, Sakaguchi Y, Motoyama A, Hibino T, Kataoka K, Huh NH. TIRAP, an adaptor protein for TLR2/4, transduces a signal from RAGE phosphorylated upon ligand binding. PLoS ONE. 2011;6:e23132. doi: 10.1371/journal.pone.0023132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Pickering RJ, Tikellis C, Rosado CJ, Tsorotes D, Dimitropoulos A, Smith M, Huet O, Seeber RM, Abhayawardana R, Johnstone EK, Golledge J, Wang Y, Jandeleit-Dahm KA, Cooper ME, Pfleger KD, Thomas MC. Transactivation of RAGE mediates angiotensin-induced inflammation and atherogenesis. J Clin Invest. 2019;129:406–421. doi: 10.1172/JCI99987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Oczypok EA, Perkins TN, Oury TD. All the "RAGE" in lung disease: The receptor for advanced glycation endproducts (RAGE) is a major mediator of pulmonary inflammatory responses. Paediatr Respir Rev. 2017;23:40–49. doi: 10.1016/j.prrv.2017.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Uchida T, Shirasawa M, Ware LB, Kojima K, Hata Y, Makita K, Mednick G, Matthay ZA, Matthay MA. Receptor for advanced glycation end-products is a marker of type I cell injury in acute lung injury. Am J Respir Crit Care Med. 2006;173:1008–1015. doi: 10.1164/rccm.200509-1477OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Katsuoka F, Kawakami Y, Arai T, Imuta H, Fujiwara M, Kanma H, et al. Type II alveolar epithelial cells in lung express receptor for advanced glycation end products (RAGE) gene. Biochem Biophys Res Commun. 1997;238:512–516. doi: 10.1006/bbrc.1997.7263. [DOI] [PubMed] [Google Scholar]
  • 25.Prasad K. AGE-RAGE Stress in the Pathophysiology of Pulmonary Hypertension and its Treatment. Int J Angiol. 2019;28:71–79. doi: 10.1055/s-0039-1687818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Nakamura K, Sakaguchi M, Matsubara H, Akagi S, Sarashina T, Ejiri K, Akazawa K, Kondo M, Nakagawa K, Yoshida M, Miyoshi T, Ogo T, Oto T, Toyooka S, Higashimoto Y, Fukami K, Ito H. Crucial role of RAGE in inappropriate increase of smooth muscle cells from patients with pulmonary arterial hypertension. PLoS ONE. 2018;13(9):e0203046. doi: 10.1371/journal.pone.0203046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Polverino F, Celli BR, Owen CA. COPD as an endothelial disorder: endothelial injury linking lesions in the lungs and other organs? (2017 Grover Conference Series) Pulm Circ. 2018;8:2045894018758528. doi: 10.1177/2045894018758528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Heeneman S, Haendeler J, Saito Y, Ishida M, Berk BC. Angiotensin II induces transactivation of two different populations of the platelet-derived growth factor β receptor. Key role for the p66 adaptor protein Shc. J Biol Chem. 2000;275:15926–15932. doi: 10.1074/jbc.M909616199. [DOI] [PubMed] [Google Scholar]
  • 29.Du J, Sperling LS, Marrero MB, Phillips L, Delafontaine P. G-protein and tyrosine kinase receptor cross-talk in rat aortic smooth muscle cells: thrombin- and angiotensin II-induced tyrosine phosphorylation of insulin receptor substrate-1 and insulin-like growth factor 1 receptor. Biochem Biophys Res Commun. 1996;218:934–939. doi: 10.1006/bbrc.1996.0165. [DOI] [PubMed] [Google Scholar]
  • 30.Rojas A, Morales M, Gonzalez I, Araya P. Inhibition of RAGE axis signaling: a pharmacological challenge. Curr Drug Targets. 2019;20:340–346. doi: 10.2174/1389450119666180820105956. [DOI] [PubMed] [Google Scholar]

Articles from Inflammation Research are provided here courtesy of Nature Publishing Group

RESOURCES