Vol 27, No 2 (2020)
Experts' Viewpoint
Published online: 2020-05-07

open access

Page views 10242
Article views/downloads 4803
Get Citation

Connect on Social Media

Connect on Social Media

COVID-19 — Toward a comprehensive understanding of the disease

Maciej M. Kowalik1, Piotr Trzonkowski2, Magdalena Łasińska-Kowara1, Andrzej Mital3, Tomasz Smiatacz4, Miłosz Jaguszewski5
Pubmed: 32378729
Cardiol J 2020;27(2):99-114.

Abstract

The evidence on the pathophysiology of the novel coronavirus SARS-CoV-2 infection is rapidly growing. Understanding why some patients suffering from COVID-19 are getting so sick, while others are not, has become an informal imperative for researchers and clinicians around the globe. The answer to this question would allow rationalizing the fear surrounding this pandemic. Understanding of the pathophysiology of COVID-19 relies on an understanding of interplaying mechanisms, including SARS-CoV-2 virulence, human immune response, and complex inflammatory reactions with coagulation playing a major role. An interplay with bacterial co-infections, as well as the vascular system and microcirculation affected throughout the body should also be examined. More importantly, a compre­hensive understanding of pathological mechanisms of COVID-19 will increase the efficacy of therapy and decrease mortality. Herewith, presented is the current state of knowledge on COVID-19: beginning from the virus, its transmission, and mechanisms of entry into the human body, through the pathological effects on the cellular level, up to immunological reaction, systemic and organ presentation. Last but not least, currently available and possible future therapeutic and diagnostic options are briefly commented on.

Article available in PDF format

View PDF Download PDF file

References

  1. Smiatacz T. It didn’t have to happen this way – what COVID-19 tells us about translational medicine. Eur J Transl Clin Med. 2020.
  2. Wu F, Zhao Su, Yu B, et al. A new coronavirus associated with human respiratory disease in China. Nature. 2020; 579(7798): 265–269.
  3. Zhou P, Yang XL, Wang XG, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020; 579(7798): 270–273.
  4. Wang M, Li M, Ren R, et al. International expansion of a novel SARS-CoV-2 mutant. J Virol. 2020.
  5. Wang W, Xu Y, Gao R, et al. Detection of sars-cov-2 in different types of clinical specimens. JAMA. 2020 [Epub ahead of print].
  6. Lescure FX, Bouadma L, Nguven D, et al. Clinical and virological data of the first cases of COVID-19 in Europe: a case series. Lancet Infect Dis. 2020.
  7. Yu F, Yan L, Wang N, et al. Quantitative detection and viral load analysis of sars-cov-2 in infected patients. Clin Infect Dis. 2020 [Epub ahead of print].
  8. Peng L, Liu J, Xu W, et al. SARS-CoV-2 can be detected in urine, blood, anal swabs, and oropharyngeal swabs specimens. J Med Virol. 2020 [Epub ahead of print].
  9. Paoli D, Pallotti F, Colangelo S, et al. Study of SARS-CoV-2 in semen and urine samples of a volunteer with positive naso-pharyngeal swab. J Endocrinol Invest. 2020 [Epub ahead of print].
  10. Kujawski STC. et al. COVID-19 Investigation Team. Clinical and virologic characteristics of the first 12 patients with coronavirus disease 2019 (COVID-19) in the United States. Nat Med. 2020 [Epub ahead of print].
  11. Zheng S, Fan J, Yu F, et al. Viral load dynamics and disease severity in patients infected with SARS-CoV-2 in Zhejiang province, China, January-March 2020: retrospective cohort study. BMJ. 2020; 369: m1443.
  12. Wang L, Li X, Chen H, et al. Coronavirus disease 19 infection does not result in acute kidney injury: an analysis of 116 hospitalized patients from Wuhan, China. Am J Nephrol. 2020: 1–6.
  13. Fang Z, Zhang Yi, Hang C, et al. Comparisons of viral shedding time of SARS-CoV-2 of different samples in ICU and non-ICU patients. J Infect. 2020 [Epub ahead of print].
  14. Wong SH, Lui RNs, Sung JJy. Covid-19 and the digestive system. J Gastroenterol Hepatol. 2020; 35(5): 744–748.
  15. Niżankowski R, Myśliwiec M, Szymański P et al. Zalecenia w COVID-19, Agencja Oceny Technologii Medycznych i Taryfikacji. Warsaw, 2020.
  16. van Doremalen N, Bushmaker T, Morris DH, et al. Aerosol and Surface Stability of SARS-CoV-2 as Compared with SARS-CoV-1. N Engl J Med. 2020; 382(16): 1564–1567.
  17. Anfinrud P, Stadnytskyi V, Bax CE, et al. Visualizing speech-generated oral fluid droplets with laser light scattering. N Engl J Med. 2020 [Epub ahead of print].
  18. Sungnak W, Huang Ni, Bécavin C, et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat Med. 2020 [Epub ahead of print].
  19. He Xi, Lau EHY, Wu P, et al. Temporal dynamics in viral shedding and transmissibility of COVID-19. Nat Med. 2020 [Epub ahead of print].
  20. Wei WE, Li Z, Chiew CJ, et al. Presymptomatic transmission of SARS-CoV-2 - Singapore, January 23-March 16, 2020. MMWR Morb Mortal Wkly Rep. 2020; 69(14): 411–415.
  21. Li F. Structure, function, and evolution of coronavirus spike proteins. Annu Rev Virol. 2016; 3(1): 237–261.
  22. Hoffmann M, Kleine-Weber H, Schroeder 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(2): 271–280.e8.
  23. Pan XW, Xu Da, Zhang H, et al. Identification of a potential mechanism of acute kidney injury during the COVID-19 outbreak: a study based on single-cell transcriptome analysis. Intensive Care Med. 2020 [Epub ahead of print].
  24. Zou L, Ruan F, Huang M, et al. SARS-CoV-2 Viral Load in Upper Respiratory Specimens of Infected Patients. N Engl J Med. 2020; 382(12): 1177–1179.
  25. Fried J, Ramasubbu K, Bhatt R, et al. The variety of cardiovascular presentations of COVID-19. Circulation. 2020.
  26. Nagata N, Iwata N, Hasegawa H, et al. Participation of both host and virus factors in induction of severe acute respiratory syndrome (SARS) in F344 rats infected with SARS coronavirus. J Virol. 2007; 81(4): 1848–1857.
  27. Petrosillo N, Viceconte G, Ergonul O, et al. COVID-19, SARS and MERS: are they closely related? Clin Microbiol Infect. 2020 [Epub ahead of print].
  28. Kikkert M. Innate immune evasion by human respiratory RNA viruses. J Innate Immun. 2020; 12(1): 4–20.
  29. Gaete-Argel A, Márquez CL, Barriga GP, et al. Strategies for success. Viral infections and membraneless organelles. Front Cell Infect Microbiol. 2019; 9: 336.
  30. Rawson TM, Moore LSP, Zhu N, et al. Bacterial and fungal co-infection in individuals with coronavirus: A rapid review to support COVID-19 antimicrobial prescribing. Clin Infect Dis. 2020 [Epub ahead of print].
  31. Trzonkowski P, Debska-Slizień A, Jankowska M, et al. Immunosenescence increases the rate of acceptance of kidney allotransplants in elderly recipients through exhaustion of CD4+ T-cells. Mech Ageing Dev. 2010; 131(2): 96–104.
  32. Trzonkowski P. Effect of anti-influenza vaccination on immune system in the elderly. Handbook on Immunosenescence. Basic Understanding and Clinical Applications. 2009: 1455–1486.
  33. Strindhall J, Nilsson BO, Löfgren S, et al. No immune risk profile among individuals who reach 100 years of age: findings from the swedish NONA immune longitudinal study. Exp Gerontol. 2007; 42(8): 753–761.
  34. Weyand CM, Goronzy JJ. Aging of the Immune System. Mechanisms and Therapeutic Targets. Ann Am Thorac Soc. 2016; 13 Suppl 5: S422–S428.
  35. Bellon M, Nicot C. Telomere dynamics in immune senescence and exhaustion triggered by chronic viral infection. Viruses. 2017; 9(10).
  36. Trzonkowski P, Myśliwska J, Szmit E, et al. Association between cytomegalovirus infection, enhanced proinflammatory response and low level of anti-hemagglutinins during the anti-influenza vaccination--an impact of immunosenescence. Vaccine. 2003; 21(25-26): 3826–3836.
  37. Zieliński M, Tarasewicz A, Zielińska H, et al. CD28 positive, cytomegalovirus specific cytotoxic T lymphocytes as a novel biomarker associated with cytomegalovirus viremia in kidney allorecipients. Journal of Clinical Virology, 2016. J Clin Virol. 2016; 83: 17–25.
  38. Lippi G, Mattiuzzi C, Sanchis-Gomar F, et al. Clinical and demographic characteristics of patients dying from COVID-19 in Italy versus China. J Med Virol. 2020 [Epub ahead of print].
  39. Trzonkowski P, Szmit E, Myśliwska J, et al. CD4+CD25+ T regulatory cells inhibit cytotoxic activity of CTL and NK cells in humans-impact of immunosenescence. Clin Immunol. 2006; 119(3): 307–316.
  40. Wang F, Nie J, Wang H, et al. Characteristics of peripheral lymphocyte subset alteration in COVID-19 pneumonia. J Infect Dis. 2020 [Epub ahead of print].
  41. Guan WJ, et al. Clinical Characteristics of Covid-19 in China. N Engl J Med. 2020.
  42. Wujtewicz M, Dylczyk-Sommer A, Aszkiełowicz A, et al. COVID-19 - what should anaethesiologists and intensivists know about it? Anaesthesiol Intensive Ther. 2020; 52(1): 34–41.
  43. Shi CS, Nabar NR, Huang NN, et al. SARS-coronavirus open reading frame-8b triggers intracellular stress pathways and activates NLRP3 inflammasomes. Cell Death Discov. 2019; 5: 101.
  44. Chen IY, Moivama M, Chang MF. Severe acute respiratory syndrome coronavirus viroporin 3a activates the NLRP3 inflammasome. Front Microbiol. 2019; 50.
  45. Jaume M, Yip MS, Cheung CY, et al. Anti-severe acute respiratory syndrome coronavirus spike antibodies trigger infection of human immune cells via a pH- and cysteine protease-independent FcγR pathway. J Virol. 2011; 85(20): 10582–10597.
  46. Hohdatsu T, Yamada M, Tominaga R, et al. Antibody-dependent enhancement of feline infectious peritonitis virus infection in feline alveolar macrophages and human monocyte cell line U937 by serum of cats experimentally or naturally infected with feline coronavirus. J Vet Med Sci. 1998; 60(1): 49–55.
  47. Siddiqi HK, Mehra MR. COVID-19 illness in native and immunosuppressed states: A clinical-therapeutic staging proposal. J Heart Lung Transplant. 2020; 39(5): 405–407.
  48. Zhao J, Yuan Q, Wang H, et al. Antibody responses to SARS-CoV-2 in patients of novel coronavirus disease. Clin Infect Dis. 2019.
  49. Trzonkowski P, Myśliwska J, Pawelec G, et al. From bench to bedside and back: the SENIEUR Protocol and the efficacy of influenza vaccination in the elderly. Biogerontology. 2009; 10(1): 83–94.
  50. Wills M, Akbar A, Beswick M, et al. Report from the second cytomegalovirus and immunosenescence workshop. Imm Ageing. 2011; 8(1).
  51. Fantuzzi G. Adipose tissue, adipokines, and inflammation. J Allergy Clin Immunol. 2005; 115(5): 911–9; quiz 920.
  52. Isobe KI, Nishio N, Hasegawa T. Immunological aspects of age-related diseases. World J Biol Chem. 2017; 8(2): 129–137.
  53. Williams H, Cassorla G, Pertsoulis N, et al. Human classical monocytes display unbalanced M1/M2 phenotype with increased atherosclerotic risk and presence of disease. Int Angiol. 2017; 36(2): 145–155.
  54. Roma-Lavisse C, Tagzirt M, Zawadzki C, et al. M1 and M2 macrophage proteolytic and angiogenic profile analysis in atherosclerotic patients reveals a distinctive profile in type 2 diabetes. Diab Vasc Dis Res. 2015; 12(4): 279–289.
  55. Myśliwska J, Wieckiewicz J, Hak L, et al. Interleukin 6 polymorphism corresponds to the number of severely stenosed coronary arteries. Eur Cytokine Netw. 2006; 17(3): 181–188.
  56. Dukat-Mazurek A, Bieniaszewska M, Hellmann A, et al. Association of cytokine gene polymorphisms with the complications of allogeneic haematopoietic stem cell transplantation. Hum Immunol. 2017; 78(11-12): 672–683.
  57. Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020; 395(10234): 1417–1418.
  58. Sherwood ER, Toliver-Kinsky T. Mechanisms of the inflammatory response. Best Pract Res Clin Anaesthesiol. 2004; 18(3): 385–405.
  59. Ciceri F, Beretta L, Scandrioglo AM, et al. Microvascular COVID-19 lung vessels obstructive thromboinflammatory syndrome (MicroCLOTS): an atypical acute respiratory distress syndrome working hypothesis. Crit Care Resusc. 2020.
  60. Klok FA, Kruip MJ, van der Meer NJM, et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res. 2020 [Epub ahead of print].
  61. Huang Z, Jiang Y, Chen J, et al. Inhibitors of the renin-angiotensin system: The potential role in the pathogenesis of COVID-19. Cardiol J. 2020 [Epub ahead of print].
  62. Tang N, Li D, Wang X, et al. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost. 2020; 18(4): 844–847.
  63. Danzi GB, Loffi M, Galeazzi G, et al. Acute pulmonary embolism and COVID-19 pneumonia: a random association? Eur Heart J. 2020 [Epub ahead of print].
  64. Giannis D, Ziogas IA, Gianni P. Coagulation disorders in coronavirus infected patients: COVID-19, SARS-CoV-1, MERS-CoV and lessons from the past. J Clin Virol. 2020 [Epub ahead of print]; 127: 104362.
  65. Thachil J, Tang N, Gando S, et al. ISTH interim guidance on recognition and management of coagulopathy in COVID-19. J Thromb Haemost. 2020; 18(5): 1023–1026.
  66. Idell S. Coagulation, fibrinolysis, and fibrin deposition in acute lung injury. Crit Care Med. 2003; 31(4 Suppl): S213–S220.
  67. Tersalvi G, Vicenzi M, Calabretta D, et al. Elevated troponin in patients with coronavirus disease 2019: possible mechanisms. J Card Fail. 2020 [Epub ahead of print].
  68. Kowalik MM, Smiatacz T, Hlebowicz M, et al. Coagulation, coma, and outcome in bacterial meningitis--an observational study of 38 adult cases. J Infect. 2007; 55(2): 141–148.
  69. Kowalik MM, Smiatacz T, Hlebowicz M. Role of coagulation in predicting the outcome of bacterial meningitis. Ann Neurol. 2008; 64(4): 473–4; author reply 474.
  70. Page MJ, Pretorius E. A champion of host defense: a generic large-scale cause for platelet dysfunction and depletion in infection. Semin Thromb Hemost. 2020; 46(3): 302–319.
  71. Zhou F, Yu T, Du R, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020; 395(10229): 1054–1062.
  72. Streiff MB, Agnelli G, Connors JM, et al. Guidance for the treatment of deep vein thrombosis and pulmonary embolism. J Thromb Thrombolysis. 2016; 41(1): 32–67.
  73. van Hylckama Vlieg A, Flinterman LE, Bare LA, et al. Genetic variations associated with recurrent venous thrombosis. Circ Cardiovasc Genet. 2014; 7(6): 806–813.
  74. Huang J, Huffman JE, Yamakuchi M, et al. Genome-wide association study for circulating tissue plasminogen activator levels and functional follow-up implicates endothelial STXBP5 and STX2. Arterioscler Thromb Vasc Biol. 2014; 34(5): 1093–1101.
  75. Germain M, Chasman D, de Haan H, et al. Meta-analysis of 65,734 Individuals Identifies TSPAN15 and SLC44A2 as Two Susceptibility Loci for Venous Thromboembolism. Am J Hum Genet. 2015; 96(4): 532–542.
  76. Haan Hde, Bezemer I, Doggen C, et al. Multiple SNP testing improves risk prediction of first venous thrombosis. Blood. 2012; 120(3): 656–663.
  77. Bruzelius M, Bottai M, Sabater-Lleal M, et al. Predicting venous thrombosis in women using a combination of genetic markers and clinical risk factors. J Thromb Haemost. 2015; 13(2): 219–227.
  78. Patnaik MM, Moll S. Inherited antithrombin deficiency: a review. Haemophilia. 2008; 14(6): 1229–1239.
  79. Wypasek E, Undas A. Protein C and protein S deficiency - practical diagnostic issues. Adv Clin Exp Med. 2013; 22(4): 459–467.
  80. Marongiu F, Grandone E, Barcellona D. Pulmonary thrombosis in 2019-nCoV pneumonia? J Thromb Haemost. 2020 [Epub ahead of print].
  81. Beun R, Kusadasi N, Sikma M, et al. Thromboembolic events and apparent heparin resistance in patients infected with SARS-CoV-2. Int J Lab Hematol. 2020 [Epub ahead of print].
  82. Joob B, Wiwanitkit V. Hemorrhagic problem among the patients with COVID-19: clinical summary of 41 thai infected patients. Clin Appl Thromb Hemost. 2020; 26: 1076029620918308.
  83. Tang N, Bai H, Chen X, et al. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J Thromb Haemost. 2020; 18(5): 1094–1099.
  84. Coto-Hernández R, Fábregas Ruano MT. Reply to. J Thromb Haemost. 2020 [Epub ahead of print].
  85. Moore HB, Barrett CD, Moore EE, et al. Is there a role for tissue plasminogen activator (tPA) as a novel treatment for refractory COVID-19 associated acute respiratory distress syndrome (ARDS)? J Trauma Acute Care Surg. 2020 [Epub ahead of print].
  86. Zhang Y, Xiao M, Zhang S, et al. Coagulopathy and Antiphospholipid Antibodies in Patients with Covid-19. N Engl J Med. 2020; 382(17): e38.
  87. Kowalik MM. Influence of intravascular coagulation on brain injury and clinical course in purulent meningitis. Przeglad Epidemiologiczny. 2006; 60(2): 273–280.
  88. Helms J, Kremer S, Merdji H, et al. Neurologic features in severe SARS-CoV-2 infection. N Engl J Med. 2020 [Epub ahead of print].
  89. Wu Z, McGoogan JM. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in china: summary of a report of 72 314 cases from the chinese center for disease control and prevention. JAMA. 2020 [Epub ahead of print].
  90. Onder G, Rezza G, Brusaferro S. Case-Fatality rate and characteristics of patients dying in relation to COVID-19 in italy. JAMA. 2020 [Epub ahead of print].
  91. Hu H, Yao Ni, Qiu Y. Comparing rapid scoring systems in mortality prediction of critical ill patients with novel coronavirus disease. Acad Emerg Med. 2020 [Epub ahead of print].
  92. Dzieciatkowski T, Szarpak L, Filipiak KJ, et al. COVID-19 challenge for modern medicine. Cardiol J. 2020 [Epub ahead of print].
  93. Kellum JA. KDIGO Clinical Practice Guideline for Acute Kidney Injury. Kidney International Supplements. 2012; 2(2): 1–141.
  94. Fanelli V, Fiorentino M, Cantaluppi V, et al. Acute kidney injury in SARS-CoV-2 infected patients. Crit Care. 2020; 24(1): 155.
  95. Su H, Yang M, Wan C, et al. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int. 2020 [Epub ahead of print].
  96. Zheng YY, Ma YT, Zhang JY, et al. COVID-19 and the cardiovascular system. Nat Rev Cardiol. 2020; 17(5): 259–260.
  97. Xiong TY, Redwood S, Prendergast B, et al. Coronaviruses and the cardiovascular system: acute and long-term implications. Eur Heart J. 2020 [Epub ahead of print].
  98. Guo T, Fan Y, Chen M, et al. Cardiovascular implications of fatal outcomes of patients with coronavirus disease 2019 (COVID-19). JAMA Cardiol. 2020 [Epub ahead of print].
  99. Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020; 395(10223): 497–506.
  100. Shi S, Qin Mu, Shen Bo, et al. Association of cardiac injury with mortality in hospitalized patients with COVID-19 in Wuhan, China. JAMA Cardiol. 2020 [Epub ahead of print].
  101. Hamming I, Timens W, Bulthuis MLC, et al. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 2004; 203(2): 631–637.
  102. Zou X, Chen Ke, Zou J, et al. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front Med. 2020 [Epub ahead of print].
  103. Chen L, Li X, Chen M, et al. The ACE2 expression in human heart indicates new potential mechanism of heart injury among patients infected with SARS-CoV-2. Cardiovasc Res. 2020; 116(6): 1097–1100.
  104. Mehra MR, Desai SS, Kuy S, et al. Cardiovascular disease, drug therapy, and mortality in covid-19. N Engl J Med. 2020 [Epub ahead of print].
  105. Brojakowska A, Narula J, Shimony R, et al. Clinical Implications of SARS-Cov2 Interaction with Renin Angiotensin System. J Am Coll Cardiol. 2020 [Epub ahead of print].
  106. Kuster GM, Pfister O, Burkard T, et al. SARS-CoV2: should inhibitors of the renin-angiotensin system be withdrawn in patients with COVID-19? Eur Heart J. 2020 [Epub ahead of print].
  107. Vaduganathan M, Vardeny O, Michel T, et al. Renin-Angiotensin-Aldosterone system inhibitors in patients with COVID-19. N Engl J Med. 2020; 382(17): 1653–1659.
  108. Danser AH, Epstein M, Batlle D. Renin-Angiotensin System Blockers and the COVID-19 Pandemic: At Present There Is No Evidence to Abandon Renin-Angiotensin System Blockers. Hypertension. 2020 [Epub ahead of print]: HYPERTENSIONAHA12015082.
  109. Maffia P, Guzik TJ. When, where, and how to target vascular inflammation in the post-CANTOS era? Eur Heart J. 2019; 40(30): 2492–2494.
  110. Clerkin KJ, Fried JA, Raikhelkar J, et al. Coronavirus disease 2019 (COVID-19) and cardiovascular disease. Circulation. 2020; 2020.
  111. Patel AB, Verma A. COVID-19 and angiotensin-converting enzyme inhibitors and angiotensin receptor blockers: what is the evidence? JAMA. 2020 [Epub ahead of print].
  112. Xu Z, Shi L, Wang Y, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020; 8(4): 420–422.
  113. Wang D, Hu Bo, Hu C, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020 [Epub ahead of print].
  114. Maekawa Y, Ouzounian M, Opavsky MA, et al. Connecting the missing link between dilated cardiomyopathy and viral myocarditis: virus, cytoskeleton, and innate immunity. Circulation. 2007; 115(1): 5–8.
  115. Ferrario CM, Jessup J, Chappell MC, et al. Effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockers on cardiac angiotensin-converting enzyme 2. Circulation. 2005; 111(20): 2605–2610.
  116. Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet. 2020; 395(10234): 1417–1418.
  117. Bonetti P, Lerman L, Lerman A. Endothelial dysfunction. Arterioscler Thromb Vasc Biol. 2003; 23(2): 168–175.
  118. Yin S, Huang M, Li D, et al. Difference of coagulation features between severe pneumonia induced by SARS-CoV2 and non-SARS-CoV2. J Thromb Thrombolysis. 2020 [Epub ahead of print].
  119. Driggin E, Madhavan MV, Bikdeli B, et al. Cardiovascular considerations for patients, health care workers, and health systems during the coronavirus disease 2019 (COVID-19) pandemic. J Am Coll Cardiol. 2020 [Epub ahead of print].
  120. Drummond GR, Vinh A, Guzik TJ, et al. Immune mechanisms of hypertension. Nat Rev Immunol. 2019; 19(8): 517–532.
  121. Sanders JM, Monogue ML, Jodlowski TZ, et al. Pharmacologic treatments for coronavirus disease 2019 (COVID-19): a review. JAMA. 2020 [Epub ahead of print].
  122. Molina JM, Delaugerre C, Le Goff J, et al. No evidence of rapid antiviral clearance or clinical benefit with the combination of hydroxychloroquine and azithromycin in patients with severe COVID-19 infection. Med Mal Infect. 2020 [Epub ahead of print].
  123. Cao B, Wang Y, Wen D, et al. A trial of lopinavir-ritonavir in adults hospitalized with severe COVID-19. N Engl J Med. 2020 [Epub ahead of print].
  124. Cortegiani A, Ingoglia G, Ippolito M, et al. A systematic review on the efficacy and safety of chloroquine for the treatment of COVID-19. J Crit Care. 2020 [Epub ahead of print].
  125. Gilead, Gilead’s Investigational Antiviral Remdesivir Receives U.S. Food and Drug Administration Emergency Use Authorization for the Treatment of COVID-19. 2020, Gilead.
  126. Wu C, Liu Y, Yang Y, et al. Analysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methods. Acta Pharm Sin B. 2020.
  127. Chu CM, Poon LLM, Cheng VCC, et al. Initial viral load and the outcomes of SARS. CMAJ. 2004; 171(11): 1349–1352.
  128. Jacobs JP, Stammers AH, St Louis J, et al. Extracorporeal membrane oxygenation in the treatment of severe pulmonary and cardiac compromise in COVID-19: experience with 32 patients. ASAIO J. 2020 [Epub ahead of print].
  129. Nicola M, O'Neill N, Sohrabi C, et al. Evidence based management guideline for the COVID-19 pandemic - review article. Int J Surg. 2020.
  130. Duan K, Liu B, Li C, et al. Effectiveness of convalescent plasma therapy in severe COVID-19 patients. Proc Natl Acad Sci U S A. 2020; 117(17): 9490–9496.
  131. Shen C, Wang Z, Zhao F, et al. Treatment of 5 critically ill patients with COVID-19 with convalescent plasma. JAMA. 2020 [Epub ahead of print].
  132. Dalli J, Winkler JW, Colas RA, et al. Resolvin D3 and aspirin-triggered resolvin D3 are potent immunoresolvents. Chem Biol. 2013; 20(2): 188–201.
  133. Colby JK, Abdulnour REE, Sham HoP, et al. Resolvin D3 and aspirin-triggered resolvin D3 are protective for injured epithelia. Am J Pathol. 2016; 186(7): 1801–1813.
  134. Arnardottir H, Dalli J, Colas R, et al. Aging delays resolution of acute inflammation in mice: reprogramming the host response with novel nano-proresolving medicines. J Immunol. 2014; 193(8): 4235–4244.
  135. Duvall MG, Levy BD. DHA- and EPA-derived resolvins, protectins, and maresins in airway inflammation. Eur J Pharmacol. 2016; 785: 144–155.