47
views
0
recommends
+1 Recommend
5 collections
    1
    shares

      Call for Papers: Sex and Gender in Neurodegenerative Diseases

      Submit here before September 30, 2024

      About Neurodegenerative Diseases: 3.0 Impact Factor I 4.3 CiteScore I 0.695 Scimago Journal & Country Rank (SJR)

      Call for Papers: Green Renal Replacement Therapy: Caring for the Environment

      Submit here before July 31, 2024

      About Blood Purification: 3.0 Impact Factor I 5.6 CiteScore I 0.83 Scimago Journal & Country Rank (SJR)

      • Record: found
      • Abstract: found
      • Article: found

      Mechanisms of Stroke in COVID-19

      article-commentary

      Read this article at

      ScienceOpenPublisherPubMed
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          In this Commentary, we address mechanisms of stroke in patients with coronavirus disease 2019 (COVID-19) due to infection with the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). It should be noted that given the recency of the pandemic, most studies are small case series, so this evaluation should be regarded as preliminary. A review by a panel of the World Stroke Organization reported that the risk of ischemic stroke during COVID-19 is around 5% (95% confidence interval [CI]: 2.8–8.7%) [1]. COVID-19-related hemorrhagic strokes are far less common than ischemic strokes, but a few cases have been reported [2, 3, 4, 5]. The median time from diagnosis to ischemic stroke in one small single-center study was 10 days (IQR: 1–19) [6]. Patients with COVID-19 who had strokes were more likely to be older and have hypertension and higher levels of D-dimer[1]. Similarly, among 50 patients with ischemic stroke admitted in Wuhan, China, there was more comorbidity, lower platelet counts and leukocyte counts, and the patients had higher levels of D-dimers, cardiac troponin I, NT probrain natriuretic peptide, and interleukin-6 [7]. The strokes are commonly labeled as cryptogenic. In a retrospective review of 32 patients with COVID-19, 65.6% had cryptogenic stroke compared with 30.4% of contemporary controls (p = 0.003) and 25% of historical controls (p < 0.001). The next most frequent stroke category was cardioembolism (22%) [6]. It should be noted that diagnostic investigations could not be completed in some patients with COVID-19 and this might have contributed to the high rate of cryptogenic strokes. Strokes in patients with COVID-19 may be due to usual causes such as atherosclerosis, hypertension, and atrial fibrillation. In this review, however, we focus on mechanisms of stroke that appear to be directly related to COVID-19. It seems likely that these COVID-19-related mechanisms would also increase the risk of stroke in infected persons who harbor the more conventional stroke risk factors. Three main mechanisms appear to be responsible for the occurrence of ischemic strokes in COVID-19 [8, 9] (Fig. 1). These include a hypercoagulable state, vasculitis, and cardiomyopathy. While the pathogenesis of hemorrhagic strokes in the setting of COVID-19 has not been fully elucidated, it is possible that the affinity of the SARS-CoV-2 for ACE2 receptors, which are expressed in endothelial and arterial smooth muscle cells in the brain, allows the virus to damage intracranial arteries, causing vessel wall rupture[10]. In addition, it is possible that the cytokine storm that accompanies this disorder could be the cause of hemorrhagic strokes, as reported in a CO­VID-19 patient who developed an acute necrotizing encephalopathy associated with late parenchymal brain hemorrhages [4]. This massive release of cytokines may also damage and result in breakdown of the blood-brain barrier and cause hemorrhagic posterior reversible encephalopathy syndrome (PRES) [11]. Secondary hemorrhagic transformation of ischemic strokes has also been reported in COVID-19 patients [3, 12]. Such transformation may occur in the setting of endothelial damage or a consumption coagulopathy accompanying COVID-19 [12]. Hypercoagulable State Lee et al. [13] reported that 20–55% of patients hospitalized with COVID-19 have laboratory evidence of coagulopathy, with increased levels of D-dimer to above twice normal, slight prolongation of prothrombin time (1–3 s above normal), mild thrombocytopenia, and in late disease, decreased fibrinogen levels. A D-dimer level above 4 times normal was associated with a 5-fold increase in the likelihood of critical illness. Thachil et al. [14] published guidance on recognition and management of coagulopathy in COVID-19 from the International Society for Thrombosis and Haemostasis. They recommended monitoring of prothrombin time, D-dimer, platelet count, and fibrinogen and prophylactic anticoagulation with low-molecular weight (LMW) heparin in all patients with COVID-19. Yaghi et al. [6] reported that high levels of D-dimer were more common among patients with stroke and COVID-19 and suggested that hypercoagulability may underly much of stroke in this disease. They indicated that a randomized trial of therapeutic anticoagulation versus prophylactic anticoagulation is underway. Antiphospholipid antibodies (anticardiolipin and anti-β-glycoprotein I antibodies) have been documented in COVID-19 patients with multiple hemispheric infarcts and with concomitant elevation of prothrombin time, activated partial thromboplastin time (aPTT), fibrinogen, D-dimer, and CRP [15]. The lupus anticoagulant was reported to be present in 45% of patients with COVID-19 versus only 10% with anticardiolipin antibody in a study in France (n = 50) [16]. In another study, the lupus anticoagulant was documented in 31 of 34 (91%) COVID-19 patients who had an elevated aPTT, but the frequency of venous thromboembolism was low in this group (2 patients; 6%) [17]. The significance of these findings is unclear as antiphospholipid antibodies were present in subjects with multiple other features of hypercoagulability, and the isolated finding of lupus anticoagulant was not associated with high risk of thromboembolism. In some patients, the combination of thrombocytopenia, prolonged prothrombin time, increased D-dimer and lactate dehydrogenase levels, and decreased fibrinogen concentrations is consistent with consumption coagulopathy that typically occurs in disseminated intravascular coagulation (DIC). Postmortem studies in patients dying of COVID-19 show microvascular platelet-fibrin-rich thrombotic depositions in the lungs as well as in other organs [18, 19, 20]. It has been suggested that viral invasion of the vascular endothelium triggers activation of the contact and complement systems which, in turn, initiates thrombotic and inflammatory cascades leading to internal organ injury [19, 21]. Weidman et al. [22] reviewed the role of inflammation in thrombosis (Fig. 2). While the relevance of microvascular thrombosis is becoming increasingly clear, a substantial proportion of patients with severe COVID-19 also develop large vessel occlusion. Although in situ thrombosis has been postulated to cause large artery occlusion, this seems unlikely. Red thrombus forms in the setting of stasis; it contains more entrapped red blood cells and less platelets. White thrombus, which forms in the setting of fast flow/high shear rates, contains less entrapped red blood cells and more platelet aggregates [23, 24, 25]. Red thrombus in large arteries probably forms only after a plaque rupture or arterial dissection and in fusiform aneurysms. Many of the patients with large artery occlusion have been younger patients with no vascular risk factors, so unlikely to have large plaques with plaque rupture. Bangalore et al. [26] report that 33% of COVID-19 patients with myocardial infarction did not have obstructive coronary artery disease. At Mt. Sinai Hospital in New York, after thrombectomy, most of the large arteries with occlusive thrombus appeared normal (M. Alberts, personal communication, May 28, 2020). Escalard et al. [27] reported that among 10 patients with stroke and COVID-19, 5 had large artery occlusions in multiple vascular territories. This suggests that large artery occlusion in COVID-19 may be mainly cardioembolic/paradoxical. In 2 studies, deep venous thrombosis occurred in 20–25% of patients admitted to critical care [28, 29], and in another study this occurred despite prophylaxis with LMW heparin. Klok et al. [30] reported that despite prophylaxis with LMW heparin, 31% of the 100 patients with COVID-19 pneumonia admitted to the ICU had an arterial (3.7%) or venous (96.3%) thrombotic manifestation; all arterial events were strokes, and 81% of all thrombotic events were pulmonary emboli. They suggested that prophylaxis should be implemented with higher than usual doses of LMW heparin. In a recent autopsy study of 12 cases from Germany, fresh thrombus in the deep venous system was detected in 7; in addition, there was a fresh thrombus in the prostatic venous plexus in 6 cases [31]. In a French study, 79% of 34 patients admitted to the ICU had DVT at 48 h after admission [32]. These findings suggest that paradoxical embolism could be a plausible mechanism of stroke in some patients with COVID-19 coagulopathy. Indeed, paradoxical embolism may account for some or many of the large artery ischemic strokes in young people, including extracranial occlusion of the common and internal carotid artery [33]. Vasculitis SARS-CoV-2 causes clinical COVID-19 by its affinity for the ACE2 receptors that are expressed in the lungs, heart, kidneys, and small bowel. These receptors are also abundant in the vascular endothelium [34], where infection elicits an inflammatory response (a lymphocytic “endotheliitis”) [18] that has been postulated as one of the substrates for the thrombotic complications of this infection. In a recent pathological study in patients with COVID-19 infection (2 autopsies and 1 surgical biopsy), Varga et al. [18] documented viral inclusions in endothelial cells in the kidneys, heart, lungs, and small bowel, with associated widespread endothelial dysfunction and apoptosis. The authors speculated that a virus-induced state of systemic impaired microcirculatory function in different vascular beds may form the basis for the multiorgan failure that characterizes the severe cases of COVID-19. Vessels may not only be inflamed by a direct local effect of SARS-CoV-2 on the ACE2 receptors in the vascular endothelium but also by a systemic immune response to the pathogen (“cytokine storm”). In the case of COVID-19, several cytokines, including IL1B, IFNγ, IP10, and MCP1 have been found to be markedly elevated, especially in patients with severe disease and high rates of mortality [35]. Ackermann et al. [36] reported an autopsy series of 7 cases of COVID-19 compared with 7 cases of influenza. They noted that distinctive pulmonary features in COVID-19 included diffuse alveolar damage with perivascular T-cell infiltration, severe endothelial injury associated with the presence of intracellular virus and disrupted cell membranes, widespread thrombosis with microangiopathy, and angiogenesis [36]. The role that these various vascular and immune-mediated factors play in the pathogenesis of stroke in COVID-19 patient remains unclear. More data are needed regarding angiographic and postmortem studies of the cervical-cerebral vasculature in order to evaluate the presence and magnitude of vasculitis and its potential role in in-situ clot formation as a potential mechanism of ischemic stroke. It seems unlikely that angiography will disclose an endotheliitis, although it may show large vessel occlusion. The large vessel occlusions are also in atypical locations, such as extensive thrombus emanating from the common carotid artery. Whether such large artery thrombi form at the site, or are embolic, remains to be determined. As discussed above and below, it seems likely that some or many of the large artery occlusions may be embolic. The clinical stroke observations have included instances in which predominantly large-vessel territorial infarcts have occurred in subjects with conventional risk factors (age, hypertension, diabetes, atrial fibrillation, and ischemic heart disease) who were infected by SARS-CoV-2 and had high levels of inflammatory markers (such as CRP and ferritin) and markers of coagulation and fibrinolysis (such as D-dimer, fibrinogen, and lupus anticoagulant) [9]. The vascular images provided were indicative of large vessel occlusion without features to suggest an alternative mechanism such as multifocal or widespread vascular wall inflammation (vasculitis). A recent report from Italy compared stroke patients with (n = 43) and without (n = 68) COVID-19. Baseline characteristics including age, sex, and vascular risk factors were similar between the 2 groups [37]. Hemorrhagic strokes were less common in COVID-19 patients (7.0 vs. 13.4%). However, initial stroke severity, measured by the NIHSS, was greater in COVID-19 than in non-COVID-19 patients (10 vs. 4), suggesting involvement of large vessels. Unfortunately, neither vessel images nor stroke subtype classification were reported. Other small series have confirmed cases of large vessel vascular occlusion, and some authors have stressed a high frequency of stroke in young COVID-19 subjects (ages 33–49) with a low prevalence of conventional stroke risk factors and elevated markers of inflammation (ferritin) and coagulation (D-dimer and fibrinogen) [8]. The high frequency of ischemic stroke in young subjects with COVID-19 and a paucity of vascular risk factors raises the possibility that mechanisms peculiar to COVID-19 may be responsible. These could include abnormalities in the vascular endothelium related to direct viral invasion and inflammation (“endotheliitis”), along with the results of a “cytokine storm” (commonly observed in this patient group). A virus-induced prothrombotic state may play an additional role, potentially synergistic, in the pathogenesis of ischemic strokes of all types in COVID-19 patients. An additional prothrombotic factor is impaired endothelial expression of heparan sulfate, which promotes thrombosis [37]. It is clear that more data in this area are required in order to establish more precisely the potential contribution of these various processes in the pathogenesis of stroke in COVID-19 patients. Cardiomyopathy There are a number of mechanisms for cardiac involvement in COVID-19 patients [38, 39, 40]. There may be direct invasion by the virus, causing a myocarditis, with resultant injury and even death of cardiomyocytes. This may due to the affinity of the virus for ACE2, a membrane-bound aminopeptidase, which acts as a portal of viral entry, and downregulation of ACE2, leading to myocardial dysfunction [36]. The heart may also be indirectly affected by the systemic inflammatory state during the severe phase of the infection related to the cytokine storm where many “bystander” end-organs are damaged [40]. There is also increased cardiac stress due to respiratory failure and hypoxemia from the infection, leading to stress cardiomyopathy. An additional possible mechanism for cardiac damage in COVID-19 is stimulation of the sympathetic nervous system, predisposing to stress cardiomyopathy and cardiac arrhythmias [41, 42]. These may lead to arrhythmias and heart failure with preserved ejection fraction. The consequent intracardiac thrombus formation, possibly compounded by the hypercoagulable states, raises the risk of subsequent cardioembolic stroke. Some of the published case series of stroke during COVID-19 have not evaluated cardioembolic mechanisms [8, 9]. An early report from China and the previous report from Singapore on SARS-CoV-1 infection developing stroke during that 2002–2003 outbreak demonstrated cardioembolic mechanisms in 36 and 60% of their cases, respectively [43]. Yaghi et al. [6] reported that stroke patients with COVID-19 were more likely to be young men with elevated troponin levels compared with historical controls. Implications for Therapy Anticoagulation All patients admitted to intensive care should receive prophylaxis against venous thrombosis, with at least LMW heparin [14]. In patients with stroke, a cardioembolic mechanism should be strongly suspected, and if there are findings to support that suspicion, such as infarction in multiple cerebral vascular territories (or systemic and cerebral emboli), cardiomyopathy with significant ventricular dyskinesia, atrial fibrillation, or right-to-left shunt, the patient should probably receive therapeutic doses of anticoagulation [44]. A theoretical consideration, based on the experience of Klok et al. [30], is that perhaps DOACs (which target only one clotting factor) might be less effective than heparin or warfarin, which target multiple clotting factors [45, 46]. Warfarin interferes with the synthesis of factors II, VII, IX, and X [47]. Atarashi et al. [46] suggested that the reason warfarin causes more intracerebral hemorrhage is that it targets factor VII. It seems likely that interference with multiple clotting factors may be why warfarin is more effective than DOACs in patients with mechanical heart valves [48, 49, 50]. Tang et al. [51] reported that anticoagulation reduced mortality in COVID-19 patients with coagulopathy. In reply, Asakura and Ogawa [52] noted that some features of the coagulopathy in COVID-19 suggest DIC and recommended a combination of heparin and nafamostat mesylate, a treatment used for DIC in Japan. Thachil et al. [53] responded with a discussion of possible benefits of unfractionated heparin versus LMW heparin. Besides anticoagulation, there may be reason to consider thrombolytic therapy. Wang et al. [54] suggested from a small case series that tissue plasminogen activator may be beneficial in acute respiratory distress syndrome in COVID-10. Anti-Inflammatory Therapies Based on their benefit as anti-inflammatory agents in cardiovascular clinical trials, therapies such as IL-6R monoclonal antibodies (tocilizumab), TNF-α inhibitors (etanercept and infliximab), and IL-1β antagonists (the monoclonal antibody canakinumab and the anti-cytokine anakinra) have been suggested as potentially beneficial for COVID-19 patients [55]. However, the main concern related to the use of immunosuppression, including corticosteroids, is that it may delay the elimination of the virus [56] and increase the risk of secondary infection, especially in those with an impaired immune system [57]. Though there is no evidence from randomized controlled trials, anti-inflammatory agents, including corticosteroids, are empirically used for severe complications in patients with COVID-19, such as ARDS, and acute cardiac and renal involvement [58]. Whether systemic corticosteroids or other anti-inflammatory agents can be of value in the selected cases of stroke in which vasculitis is suspected is a matter of debate. Neurologists should carefully balance the risk and benefit ratio before starting anti-inflammatory therapies. Antiviral Therapy Beigel et al. [59] reported that in a randomized controlled trial in 1,059 patients hospitalized for COVID-19, remdesivir was associated with a shorter median recovery time (11 days, 95% CI: 9–12), compared with placebo (15 days, 95% CI: 13–19), and a lower 14-day mortality of 7.1% with remdesivir versus 11.9% with placebo (hazard ratio for death, 0.70; 95% CI: 0.47–1.04). Stroke was not mentioned in the report. Future Research The list of clinical trials registered at ClinicalTrials.gov can be found at https://clinicaltrials.gov/ct2/results?cond = COVID-19. Important therapeutic targets in these trials include virus neutralization, prevention and treatment of thrombotic complications, and inhibition of the cytokine storm. Such trials test a wide range of therapies from convalescent and hyperimmune plasma for virus neutralization to tissue plasminogen activator for treatment of microvascular thrombosis and from hemodialysis with reconditioning of immune cells to complement inhibitors, selective cytokine inhibitors, and colchicine to prevent the cytokine storm, based on a study on coronary artery disease [60]. Conclusions A number of mechanisms are involved in stroke in COVID-19, including a hypercoagulable state, DIC, necrotizing encephalopathy, vasculitis, and cardiomyopathy. It seems likely that anticoagulation will play a substantial role in the management of stroke in COVID-19. Further evidence is needed from larger studies. Conflict of Interest Statement Dr. Ay is employed by Takeda Pharmaceutical Company Limited; none of the other authors has a disclosure that is relevant to this topic. Author Contributions J.D.S. wrote the first and final draft; each of the other authors contributed to revisions.

          Related collections

          Most cited references58

          • Record: found
          • Abstract: found
          • Article: not found

          Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China

          Summary Background A recent cluster of pneumonia cases in Wuhan, China, was caused by a novel betacoronavirus, the 2019 novel coronavirus (2019-nCoV). We report the epidemiological, clinical, laboratory, and radiological characteristics and treatment and clinical outcomes of these patients. Methods All patients with suspected 2019-nCoV were admitted to a designated hospital in Wuhan. We prospectively collected and analysed data on patients with laboratory-confirmed 2019-nCoV infection by real-time RT-PCR and next-generation sequencing. Data were obtained with standardised data collection forms shared by WHO and the International Severe Acute Respiratory and Emerging Infection Consortium from electronic medical records. Researchers also directly communicated with patients or their families to ascertain epidemiological and symptom data. Outcomes were also compared between patients who had been admitted to the intensive care unit (ICU) and those who had not. Findings By Jan 2, 2020, 41 admitted hospital patients had been identified as having laboratory-confirmed 2019-nCoV infection. Most of the infected patients were men (30 [73%] of 41); less than half had underlying diseases (13 [32%]), including diabetes (eight [20%]), hypertension (six [15%]), and cardiovascular disease (six [15%]). Median age was 49·0 years (IQR 41·0–58·0). 27 (66%) of 41 patients had been exposed to Huanan seafood market. One family cluster was found. Common symptoms at onset of illness were fever (40 [98%] of 41 patients), cough (31 [76%]), and myalgia or fatigue (18 [44%]); less common symptoms were sputum production (11 [28%] of 39), headache (three [8%] of 38), haemoptysis (two [5%] of 39), and diarrhoea (one [3%] of 38). Dyspnoea developed in 22 (55%) of 40 patients (median time from illness onset to dyspnoea 8·0 days [IQR 5·0–13·0]). 26 (63%) of 41 patients had lymphopenia. All 41 patients had pneumonia with abnormal findings on chest CT. Complications included acute respiratory distress syndrome (12 [29%]), RNAaemia (six [15%]), acute cardiac injury (five [12%]) and secondary infection (four [10%]). 13 (32%) patients were admitted to an ICU and six (15%) died. Compared with non-ICU patients, ICU patients had higher plasma levels of IL2, IL7, IL10, GSCF, IP10, MCP1, MIP1A, and TNFα. Interpretation The 2019-nCoV infection caused clusters of severe respiratory illness similar to severe acute respiratory syndrome coronavirus and was associated with ICU admission and high mortality. Major gaps in our knowledge of the origin, epidemiology, duration of human transmission, and clinical spectrum of disease need fulfilment by future studies. Funding Ministry of Science and Technology, Chinese Academy of Medical Sciences, National Natural Science Foundation of China, and Beijing Municipal Science and Technology Commission.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found

            Remdesivir for the Treatment of Covid-19 — Final Report

            Abstract Background Although several therapeutic agents have been evaluated for the treatment of coronavirus disease 2019 (Covid-19), none have yet been shown to be efficacious. Methods We conducted a double-blind, randomized, placebo-controlled trial of intravenous remdesivir in adults hospitalized with Covid-19 with evidence of lower respiratory tract involvement. Patients were randomly assigned to receive either remdesivir (200 mg loading dose on day 1, followed by 100 mg daily for up to 9 additional days) or placebo for up to 10 days. The primary outcome was the time to recovery, defined by either discharge from the hospital or hospitalization for infection-control purposes only. Results A total of 1063 patients underwent randomization. The data and safety monitoring board recommended early unblinding of the results on the basis of findings from an analysis that showed shortened time to recovery in the remdesivir group. Preliminary results from the 1059 patients (538 assigned to remdesivir and 521 to placebo) with data available after randomization indicated that those who received remdesivir had a median recovery time of 11 days (95% confidence interval [CI], 9 to 12), as compared with 15 days (95% CI, 13 to 19) in those who received placebo (rate ratio for recovery, 1.32; 95% CI, 1.12 to 1.55; P<0.001). The Kaplan-Meier estimates of mortality by 14 days were 7.1% with remdesivir and 11.9% with placebo (hazard ratio for death, 0.70; 95% CI, 0.47 to 1.04). Serious adverse events were reported for 114 of the 541 patients in the remdesivir group who underwent randomization (21.1%) and 141 of the 522 patients in the placebo group who underwent randomization (27.0%). Conclusions Remdesivir was superior to placebo in shortening the time to recovery in adults hospitalized with Covid-19 and evidence of lower respiratory tract infection. (Funded by the National Institute of Allergy and Infectious Diseases and others; ACTT-1 ClinicalTrials.gov number, NCT04280705.)
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Endothelial cell infection and endotheliitis in COVID-19

              Cardiovascular complications are rapidly emerging as a key threat in coronavirus disease 2019 (COVID-19) in addition to respiratory disease. The mechanisms underlying the disproportionate effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection on patients with cardiovascular comorbidities, however, remain incompletely understood.1, 2 SARS-CoV-2 infects the host using the angiotensin converting enzyme 2 (ACE2) receptor, which is expressed in several organs, including the lung, heart, kidney, and intestine. ACE2 receptors are also expressed by endothelial cells. 3 Whether vascular derangements in COVID-19 are due to endothelial cell involvement by the virus is currently unknown. Intriguingly, SARS-CoV-2 can directly infect engineered human blood vessel organoids in vitro. 4 Here we demonstrate endothelial cell involvement across vascular beds of different organs in a series of patients with COVID-19 (further case details are provided in the appendix). Patient 1 was a male renal transplant recipient, aged 71 years, with coronary artery disease and arterial hypertension. The patient's condition deteriorated following COVID-19 diagnosis, and he required mechanical ventilation. Multisystem organ failure occurred, and the patient died on day 8. Post-mortem analysis of the transplanted kidney by electron microscopy revealed viral inclusion structures in endothelial cells (figure A, B ). In histological analyses, we found an accumulation of inflammatory cells associated with endothelium, as well as apoptotic bodies, in the heart, the small bowel (figure C) and lung (figure D). An accumulation of mononuclear cells was found in the lung, and most small lung vessels appeared congested. Figure Pathology of endothelial cell dysfunction in COVID-19 (A, B) Electron microscopy of kidney tissue shows viral inclusion bodies in a peritubular space and viral particles in endothelial cells of the glomerular capillary loops. Aggregates of viral particles (arrow) appear with dense circular surface and lucid centre. The asterisk in panel B marks peritubular space consistent with capillary containing viral particles. The inset in panel B shows the glomerular basement membrane with endothelial cell and a viral particle (arrow; about 150 nm in diameter). (C) Small bowel resection specimen of patient 3, stained with haematoxylin and eosin. Arrows point to dominant mononuclear cell infiltrates within the intima along the lumen of many vessels. The inset of panel C shows an immunohistochemical staining of caspase 3 in small bowel specimens from serial section of tissue described in panel D. Staining patterns were consistent with apoptosis of endothelial cells and mononuclear cells observed in the haematoxylin-eosin-stained sections, indicating that apoptosis is induced in a substantial proportion of these cells. (D) Post-mortem lung specimen stained with haematoxylin and eosin showed thickened lung septa, including a large arterial vessel with mononuclear and neutrophilic infiltration (arrow in upper inset). The lower inset shows an immunohistochemical staining of caspase 3 on the same lung specimen; these staining patterns were consistent with apoptosis of endothelial cells and mononuclear cells observed in the haematoxylin-eosin-stained sections. COVID-19=coronavirus disease 2019. Patient 2 was a woman, aged 58 years, with diabetes, arterial hypertension, and obesity. She developed progressive respiratory failure due to COVID-19 and subsequently developed multi-organ failure and needed renal replacement therapy. On day 16, mesenteric ischaemia prompted removal of necrotic small intestine. Circulatory failure occurred in the setting of right heart failure consequent to an ST-segment elevation myocardial infarction, and cardiac arrest resulted in death. Post-mortem histology revealed lymphocytic endotheliitis in lung, heart, kidney, and liver as well as liver cell necrosis. We found histological evidence of myocardial infarction but no sign of lymphocytic myocarditis. Histology of the small intestine showed endotheliitis (endothelialitis) of the submucosal vessels. Patient 3 was a man, aged 69 years, with hypertension who developed respiratory failure as a result of COVID-19 and required mechanical ventilation. Echocardiography showed reduced left ventricular ejection fraction. Circulatory collapse ensued with mesenteric ischaemia, and small intestine resection was performed, but the patient survived. Histology of the small intestine resection revealed prominent endotheliitis of the submucosal vessels and apoptotic bodies (figure C). We found evidence of direct viral infection of the endothelial cell and diffuse endothelial inflammation. Although the virus uses ACE2 receptor expressed by pneumocytes in the epithelial alveolar lining to infect the host, thereby causing lung injury, the ACE2 receptor is also widely expressed on endothelial cells, which traverse multiple organs. 3 Recruitment of immune cells, either by direct viral infection of the endothelium or immune-mediated, can result in widespread endothelial dysfunction associated with apoptosis (figure D). The vascular endothelium is an active paracrine, endocrine, and autocrine organ that is indispensable for the regulation of vascular tone and the maintenance of vascular homoeostasis. 5 Endothelial dysfunction is a principal determinant of microvascular dysfunction by shifting the vascular equilibrium towards more vasoconstriction with subsequent organ ischaemia, inflammation with associated tissue oedema, and a pro-coagulant state. 6 Our findings show the presence of viral elements within endothelial cells and an accumulation of inflammatory cells, with evidence of endothelial and inflammatory cell death. These findings suggest that SARS-CoV-2 infection facilitates the induction of endotheliitis in several organs as a direct consequence of viral involvement (as noted with presence of viral bodies) and of the host inflammatory response. In addition, induction of apoptosis and pyroptosis might have an important role in endothelial cell injury in patients with COVID-19. COVID-19-endotheliitis could explain the systemic impaired microcirculatory function in different vascular beds and their clinical sequelae in patients with COVID-19. This hypothesis provides a rationale for therapies to stabilise the endothelium while tackling viral replication, particularly with anti-inflammatory anti-cytokine drugs, ACE inhibitors, and statins.7, 8, 9, 10, 11 This strategy could be particularly relevant for vulnerable patients with pre-existing endothelial dysfunction, which is associated with male sex, smoking, hypertension, diabetes, obesity, and established cardiovascular disease, all of which are associated with adverse outcomes in COVID-19.
                Bookmark

                Author and article information

                Journal
                CED
                Cerebrovasc Dis
                10.1159/issn.1015-9770
                Cerebrovascular Diseases
                S. Karger AG
                1015-9770
                1421-9786
                20 July 2020
                :
                :
                : 1-8
                Affiliations
                [_a] aStroke Prevention & Atherosclerosis Research Centre, Robarts Research Institute, Western University, London, Ontario, Canada
                [_b] bDepartment of Neurology, Universidade Federal Fluminense and Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
                [_c] cDepartment of Neurology, University of Kentucky School of Medicine, Lexington, Kentucky, USA
                [_d] dDepartment of Neurology and Radiology, Harvard School of Medicine and Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, USA
                [_e] eNeurovascular Imaging Research Core and UCLA Stroke Center, Department of Neurology, University of California, Los Angeles, California, USA
                [_f] fDepartment of Neurology, Emory University, Atlanta, Georgia, USA
                [_g] gSchool of Medicine, Universidad Espíritu Santo, Samborondón, Ecuador
                [_h] hMedical School, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Washington, Australia
                [_i] iRaffles Neuroscience Centre, Raffles Hospital, Singapore, Singapore
                Author notes
                *J. David Spence, Stroke Prevention and Atherosclerosis Research Centre, Robarts Research Institute, 1400 Western Road, London, ON N6G 2V4 (Canada), dspence@robarts.ca
                Author information
                https://orcid.org/0000-0002-5109-8736
                Article
                509581 Cerebrovasc Dis
                10.1159/000509581
                32690850
                fd8a97b1-9fdf-4282-873b-a7be7eddf743
                © 2020 S. Karger AG, Basel

                Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher. Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug. Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.

                History
                : 05 June 2020
                : 22 June 2020
                Page count
                Figures: 2, Pages: 8
                Categories
                Commentary

                Internal medicine,Respiratory medicine,Clinical Psychology & Psychiatry,Microbiology & Virology,Infectious disease & Microbiology

                Comments

                Comment on this article