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

Thomas Menter, Jasmin D Haslbauer, Ronny Nienhold, Spasenija Savic, Helmut Hopfer, Nikolaus Deigendesch, Stephan Frank, Daniel Turek, Niels Willi, Hans Pargger, Stefano Bassetti, Joerg D Leuppi, Gieri Cathomas, Markus Tolnay, Kirsten D Mertz, Alexandar Tzankov, Thomas Menter, Jasmin D Haslbauer, Ronny Nienhold, Spasenija Savic, Helmut Hopfer, Nikolaus Deigendesch, Stephan Frank, Daniel Turek, Niels Willi, Hans Pargger, Stefano Bassetti, Joerg D Leuppi, Gieri Cathomas, Markus Tolnay, Kirsten D Mertz, Alexandar Tzankov

Abstract

Aims: Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has rapidly evolved into a sweeping pandemic. Its major manifestation is in the respiratory tract, and the general extent of organ involvement and the microscopic changes in the lungs remain insufficiently characterised. Autopsies are essential to elucidate COVID-19-associated organ alterations.

Methods and results: This article reports the autopsy findings of 21 COVID-19 patients hospitalised at the University Hospital Basel and at the Cantonal Hospital Baselland, Switzerland. An in-corpore technique was performed to ensure optimal staff safety. The primary cause of death was respiratory failure with exudative diffuse alveolar damage and massive capillary congestion, often accompanied by microthrombi despite anticoagulation. Ten cases showed superimposed bronchopneumonia. Further findings included pulmonary embolism (n = 4), alveolar haemorrhage (n = 3), and vasculitis (n = 1). Pathologies in other organ systems were predominantly attributable to shock; three patients showed signs of generalised and five of pulmonary thrombotic microangiopathy. Six patients were diagnosed with senile cardiac amyloidosis upon autopsy. Most patients suffered from one or more comorbidities (hypertension, obesity, cardiovascular diseases, and diabetes mellitus). Additionally, there was an overall predominance of males and individuals with blood group A (81% and 65%, respectively). All relevant histological slides are linked as open-source scans in supplementary files.

Conclusions: This study provides an overview of postmortem findings in COVID-19 cases, implying that hypertensive, elderly, obese, male individuals with severe cardiovascular comorbidities as well as those with blood group A may have a lower threshold of tolerance for COVID-19. This provides a pathophysiological explanation for higher mortality rates among these patients.

Keywords: COVID-19; SARS-CoV-2; autopsy; cardiovascular; kidney; lung; senile amyloidosis.

Conflict of interest statement

All authors declare that they have no conflicting interests.

© 2020 The Authors. Histopathology published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Gross lung findings. A, Typical appearance of coronavirus disease 2019 (COVID‐19) lungs; note the perceptibly thickened alveolar septae and congestive interstitial aspects and a thrombembolus in the lower lobe. Insert: detailed view highlighting interstitial congestion. B, Extensive bronchopneumonic infiltrates in a COVID‐19 patient suffering from superimposed suppurative pneumonia.
Figure 2
Figure 2
Microscopic lung findings. A, Exudative diffuse alveolar damage (DAD) showing discrete hyaline membranes and prominent capillary congestion [haematoxylin and eosin (H&E)]. Insert: immunohistochemistry (IHC) for fibrin(ogen) showing the extent of hyaline membranes. B, Syncytial cells of pneumocyte II origin (H&E). Insert: IHC for thyroid transcription factor 1. C, Extensive capillary congestion without DAD (H&E). D, Microthrombi in alveolar capillaries (IHC for fibrin).
Figure 3
Figure 3
Findings in other organs. A, Kidney showing acute tubular damage without evidence of increased inflammatory infiltrates [periodic acid–Schiff (PAS) stain]. B, Kidney showing disseminated intravascular coagulation (PAS). C, Florid splenitis showing increases in neutrophil numbers in the perifollicular and marginal zones of the spleen (PAS). D, Lymph node showing an increase in the number of plasmablasts in the interfollicular zone as well as congestion (haematoxylin and eosin). Insert: immunohistochemistry for multiple myeloma 1.
Figure 4
Figure 4
Electron microscopy findings. A,B, Podocyte cytoplasm with its foot processes on top of a glomerular basement membrane containing mitochondria (upper left corner) and multiple vesicles, two of which contain several small possible virus‐like particles with sizes between 70 nm and 110 nm (arrow). At higher magnification, the vesicles contain double membranes and the virus‐like particles show a ring of electron‐dense granules and a ragged outer contour (electron microscopy). C, An activated glomerular endothelial cell, and a vesicle close to the luminal border with virus‐like particles (arrow and insert), adjacent to an erythrocyte (electron‐dense structure at the top left) (electron microscopy). D, Cytoplasm of a proximal tubular epithelial cell on top of a basement membrane and adjacent collagen fibres (left side). The cytoplasm contains mitochondria, rough endoplasmic reticulum, and multiple vesicles, one of which contains virus‐like particles (arrow and insert, electron microscopy).

References

    1. Monto AS. Medical reviews. Coronaviruses. Yale J. Biol. Med. 1974; 47; 234–251.
    1. Mortensen ML, Ray CG, Payne CM et al Coronaviruslike particles in human gastrointestinal disease. Epidemiologic, clinical, and laboratory observations. Am. J. Dis. Child. 1985; 139; 928–934.
    1. Drosten C, Gunther S, Preiser W et al Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N. Engl. J. Med. 2003; 348; 1967–1976.
    1. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus AD, Fouchier RA. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N. Engl. J. Med. 2012; 367; 1814–1820.
    1. Gong SR, Bao LL. The battle against SARS and MERS coronaviruses: reservoirs and animal models. Animal Model Exp. Med. 2018; 1; 125–133.
    1. COVID‐19, Situation update worldwide . European Centre for Disease Prevention and Control. Available at: (accessed 23 April 2020).
    1. Rodriguez‐Morales AJ, Cardona‐Ospina JA, Clinical G‐O et al laboratory and imaging features of COVID‐ 19: a systematic review and meta‐analysis. Travel Med. Infect. Dis. 2020; 34: 101623.
    1. Tian S, Hu W, Niu L et al Pulmonary pathology of early‐phase 2019 novel coronavirus (COVID‐19) pneumonia in two patients with lung cancer. J. Thorac. Oncol. 2020; 15; 700–704.
    1. Xu Z, Shi L, Wang Y et al Pathological findings of COVID‐19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020; 8; 420–422.
    1. Karami P, Naghavi M, Feyzi A et al Mortality of a pregnant patient diagnosed with COVID‐ 19: a case report with clinical, radiological, and histopathological findings. Travel Med. Infect. Dis. 2020; 101665.
    1. Barton LM, Duval EJ, Stroberg E, Ghosh S, Mukhopadhyay S. COVID‐19 autopsies, Oklahoma, USA. Am. J. Clin. Pathol. 2020; 153; 725–733.
    1. Tian S, Xiong Y, Liu H et al Pathological study of the 2019 novel coronavirus disease (COVID‐19) through postmortem core biopsies. Mod. Pathol. 2020; 14; 1–8.
    1. Magro C, Mulvey JJ, Berlin D et al Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID‐19 infection: a report of five cases. Transl. Res. 2020; S1931–5244(20); 30070.
    1. Ding Y, Wang H, Shen H et al The clinical pathology of severe acute respiratory syndrome (SARS): a report from China. J. Pathol. 2003; 200; 282–289.
    1. Gu J, Gong E, Zhang B et al Multiple organ infection and the pathogenesis of SARS. J. Exp. Med. 2005; 202; 415–424.
    1. Franks TJ, Chong PY, Chui P et al Lung pathology of severe acute respiratory syndrome (SARS): a study of 8 autopsy cases from Singapore. Hum. Pathol. 2003; 34; 743–748.
    1. Hwang DM, Chamberlain DW, Poutanen SM et al Pulmonary pathology of severe acute respiratory syndrome in Toronto. Mod. Pathol. 2005; 18; 1–10.
    1. Guo Y, Korteweg C, McNutt MA, Gu J. Pathogenetic mechanisms of severe acute respiratory syndrome. Virus Res. 2008; 133; 4–12.
    1. Xu J, Zhao S, Teng T et al Systematic comparison of two animal‐to‐human transmitted human coronaviruses: SARS‐CoV‐2 and SARS‐CoV. Viruses 2020; 12; pii: E244.
    1. Ng DL, Al Hosani F, Keating MK et al Clinicopathologic, immunohistochemical, and ultrastructural findings of a fatal case of Middle East respiratory syndrome coronavirus infection in the United Arab Emirates, April 2014. Am. J. Pathol. 2016; 186; 652–658.
    1. Alsaad KO, Hajeer AH, Al Balwi M et al Histopathology of Middle East respiratory syndrome coronovirus (MERS‐CoV) infection—clinicopathological and ultrastructural study. Histopathology 2018; 72; 516–524.
    1. Hanley B, Lucas SB, Youd E, Swift B, Osborn M. Autopsy in suspected COVID‐19 cases. J. Clin. Pathol. 2020; 73; 239–242.
    1. Menter T, Bachmann M, Grieshaber S, Tzankov A. A more accurate approach to amyloid detection and subtyping: combining in situ Congo Red staining and immunohistochemistry. Pathobiology 2017; 84; 49–55.
    1. Ehmann MA, Medinger M, Bodenmann B et al Histologic features of hematopoietic stem cell transplant‐associated thrombotic microangiopathy are best percepted in deep skin biopsies and renal biopsies, while showing a significant overlap with changes related to severe acute graft‐versus‐host disease in gastrointestinal biopsies. Bone Marrow Transplant. 2020; Mar 30. doi: 10.1038/s41409-020-0880-y.
    1. Sterlacci W, Fiegl M, Juskevicius D, Tzankov A. Cluster analysis according to immunohistochemistry is a robust tool for non‐small cell lung cancer and reveals a distinct, immune signature‐defined subgroup. Appl. Immunohistochem. Mol. Morphol. 2020; 28; 274–283.
    1. Blood group distribution in Switzerland . Swiss transfusion SRC. Available at: (accessed 23 April 2020).
    1. Emami A, Javanmardi F, Pirbonyeh N, Akbari A. Prevalence of underlying diseases in hospitalized patients with COVID‐19: a systematic review and meta‐analysis. Arch. Acad. Emerg. Med. 2020; 8; e35.
    1. Chen T, Wu D, Chen H et al Clinical characteristics of 113 deceased patients with coronavirus disease 2019: retrospective study. Br. Med. J. 2020; 368; m1091.
    1. Perlman S, Netland J. Coronaviruses post‐SARS: update on replication and pathogenesis. Nat. Rev. Microbiol. 2009; 7; 439–450.
    1. Goldsmith CS, Tatti KM, Ksiazek TG et al Ultrastructural characterization of SARS coronavirus. Emerg. Infect. Dis. 2004; 10; 320–326.
    1. Wolfel R, Corman VM, Guggemos W et al Virological assessment of hospitalized patients with COVID‐2019. Nature 2020; 581; 465–469.
    1. Varga Z, Flammer AJ, Steiger P et al Endothelial cell infection and endotheliitis in COVID‐19. Lancet 2020; 395; 1417–1418.
    1. 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; 98; 219–227.
    1. Kissling S, Rotman S, Gerber C et al Collapsing glomerulopathy in a COVID‐19 patient. Kidney Int. 2020; S0085–2538(20); 30369.
    1. Tang N, Li D, Wang X, Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J. Thromb. Haemost. 2020; 18; 844–847.
    1. Nakagawa M, Tojo K, Sekijima Y, Yamazaki KH, Ikeda S. Arterial thromboembolism in senile systemic amyloidosis: report of two cases. Amyloid 2012; 19; 118–121.
    1. Yamamoto K, Takeshita K, Kojima T, Takamatsu J, Saito H. Aging and plasminogen activator inhibitor‐1 (PAI‐1) regulation: implication in the pathogenesis of thrombotic disorders in the elderly. Cardiovasc. Res. 2005; 66; 276–285.
    1. Cheng Y, Luo R, Wang K et al Kidney disease is associated with in‐hospital death of patients with COVID‐19. Kidney Int. 2020; 97; 829–838.
    1. Zhao J, Yang Y, Huang H et al Relationship between the ABO blood group and the COVID‐19 susceptibility (pre‐print, not peer‐reviewed). medRxiv 2020; 2020031120031096.
    1. Jenkins PV, O'Donnell JS. ABO blood group determines plasma von Willebrand factor levels: a biologic function after all? Transfusion 2006; 46; 1836–1844.
    1. Morelli VM, de Visser MC, van Tilburg NH et al ABO blood group genotypes, plasma von Willebrand factor levels and loading of von Willebrand factor with A and B antigens. Thromb. Haemost. 2007; 97; 534–541.
    1. Tirado I, Mateo J, Soria JM et al The ABO blood group genotype and factor VIII levels as independent risk factors for venous thromboembolism. Thromb. Haemost. 2005; 93; 468–474.
    1. Franchini M, Mannucci PM. ABO blood group and thrombotic vascular disease. Thromb. Haemost. 2014; 112; 1103–1109.
    1. Guillon P, Clement M, Sebille V et al Inhibition of the interaction between the SARS‐CoV spike protein and its cellular receptor by anti‐histo‐blood group antibodies. Glycobiology 2008; 18; 1085–1093.
    1. 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; 271–280.
    1. Zhang H, Penninger JM, Li Y, Zhong N, Slutsky AS. Angiotensin‐converting enzyme 2 (ACE2) as a SARS‐CoV‐2 receptor: molecular mechanisms and potential therapeutic target. Intensive Care Med. 2020; 46; 586–590.
    1. 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; 41; 1801–1803.
    1. Zhang P, Zhu L, Cai J 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; 126; 1671–1681.
    1. Huang C, Wang Y, Li X et al Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020; 395; 497–506.
    1. Liu B, Li M, Zhou Z, Guan X, Xiang Y. Can we use interleukin‐6 (IL‐6) blockade for coronavirus disease 2019 (COVID‐19)‐induced cytokine release syndrome (CRS)? J. Autoimmun. 2020; 111; 102452.
    1. Gralinski LE, Sheahan TP, Morrison TE et al Complement activation contributes to severe acute respiratory syndrome coronavirus pathogenesis. MBio 2018; 9; pii: e01753–18.
    1. Campbell CM, Kahwash R. Will complement inhibition be the new target in treating COVID‐19 related systemic thrombosis? Circulation 2020; 141; 1739–1741.
    1. Eculizumab (Soliris) in Covid‐19 infected patients (SOLID‐C19). . Available at: (accessed 23 April 2020).
    1. Dane K, Chaturvedi S. Beyond plasma exchange: novel therapies for thrombotic thrombocytopenic purpura. Hematology Am. Soc. Hematol. Educ. Program 2018; 2018; 539–547.
    1. Wu Y, Xu X, Chen Z et al Nervous system involvement after infection with COVID‐19 and other coronaviruses. Brain Behav. Immun. 2020; S0889–1591(20); 30357–30363.
    1. Du RH, Liang LR, Yang CQ et al Predictors of mortality for patients with COVID‐19 pneumonia caused by SARS‐CoV‐2: a prospective cohort study. Eur. Respir. J. 2020; 55; 2000524.
    1. Li B, Yang J, Zhao F et al Prevalence and impact of cardiovascular metabolic diseases on COVID‐19 in China. Clin. Res. Cardiol. 2020; 109; 531–538.
    1. Singh AK, Gupta R, Misra A. Comorbidities in COVID‐19: outcomes in hypertensive cohort and controversies with renin angiotensin system blockers. Diabetes Metab. Syndr. 2020; 14; 283–287.

Source: PubMed

3
S'abonner