15
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Endotheliitis and Endothelial Dysfunction in Patients with COVID-19: Its Role in Thrombosis and Adverse Outcomes †

      editorial

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          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

          Coronavirus disease 2019 (COVID-19) is a contagious disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV2), emerging in Wuhan, China and developing into a pandemic with rapidly emerging cardiovascular manifestations. In the United States, there are approximately 1.7 million reported cases with over 100,000 deaths as of 30 May 2020 [1]. Severe SARS-CoV-2 infection is more commonly observed in patients with specific comorbidities, including cardiovascular disease, diabetes and obesity, yet the mechanism of this relationship is unclear. SARS-CoV2 binds to the angiotensin-converting enzyme 2 (ACE2) receptor, which is abundantly expressed in human tissues including lung epithelium, myocardium, and vascular endothelium [2]. The virus has been shown to directly infect engineered human blood vessel organoids in vitro [3]. A rapidly accumulating body of evidence suggests that COVID-19 causes vascular derangements as a consequence of endothelial cell infection by the virus, contributing to observed cardiovascular and pulmonary complications. Advanced age, hypertension, diabetes, smoking and coronary artery disease are risk factors for severe COVID-19, conditions which are all associated with vascular endothelial dysfunction. Recent reports have also shown a robust and independent association between obesity and the severity of COVID-19 infection, even in the absence of other co-morbidities [4,5]. Obesity is a chronic inflammatory state associated with dysregulated endocrine and paracrine actions of adipocyte-derived factor, which in turn disrupt vascular homeostasis and cause endothelial dysfunction. While the mechanisms through which obesity exacerbates COVID-19 infection are not fully understood, endothelial dysfunction may be the common link [6,7]. A prothrombotic state is evident in COVID-19 patients, with elevated D-dimer levels, arterial and deep venous thrombosis, pulmonary embolism, strokes, and intracardiac and microvascular thrombi. It is postulated that endothelial dysfunction and endotheliitis (inflammation of the blood vessel wall) result in thrombus formation. A new pulmonary complication called microvascular COVID-19 lung vessels obstructive thromboinflammatory syndrome (MicroCLOTS) has been described in severe COVID-19 and likely represents an atypical form of acute respiratory distress syndrome (ARDS) [8]. MicroCLOTS is a progressive, diffuse endothelial thromboinflammatory syndrome which is characterized by the development of microvascular pulmonary thrombosis. On a background of endothelial dysfunction, endotheliitis (endothelialitis) likely plays a central role in the development of COVID-19 related thromboembolic phenomenon and pulmonary injury [8]. Since the vascular endothelium is a dynamic endocrine, paracrine, and autocrine organ with a vital role in regulating vascular tone and homoeostasis [9], its dysfunction leads to detrimental shifts in the vascular equilibrium towards vasoconstriction (manifesting clinically as organ ischemia, infarction and intrapulmonary shunting), inflammation, and a pro-coagulant state resulting in thrombosis [10]. A recent study has shown that angiopoietin-2 is significantly elevated in critical COVID-19 patients, suggesting its role as a strong prognostic biomarker [11]. The notion of angiopoitein-2 being a marker of endothelial damage relates to its presence and release from endothelial Weibel–Palade bodies has reinforced the hypothesis of a COVID-19-associated microvascular dysfunction [11]. The presence of viral inclusion structures has also been demonstrated in endothelial cells, consistent with direct viral infection of these cells; histologic assessment of these specimens revealed endotheliitis of the submucosal vessels [12]. The authors theorize that the presence of viral elements in the endothelium, by inclusion via the ACE2 receptor, recruits immune cells, thereby resulting in widespread endothelial dysfunction associated with apoptosis. Similarly, an autopsy case series [13] demonstrated severe pulmonary endothelial injury associated with the presence of intracellular virus and disrupted cell membranes [13]. Histologic analysis of pulmonary vessels also showed widespread thrombosis with microangiopathy; alveolar capillary microthrombi were nine times as prevalent in patients with Covid-19 compared to patients with influenza. Similarly another study, which examined skin and lung tissues from COVID-19 patients, showed that the pattern of COVID-19 pneumonitis was predominantly a pauci-inflammatory septal capillary injury [14]. This was accompanied by significant deposits of terminal complement components C5b-9 (membrane attack complex), C4d, and mannose binding lectin (MBL)-associated serine protease (MASP)2 in the microvasculature [14]. This is consistent with the hypothesis that COVID-19 infection causes a calamitous microvascular injury syndrome which includes activation of complement pathways, endotheliitis and an associated thrombotic state [15]. Future studies will likely focus on the role of endothelial dysfunction and endotheliitis in the pathogenesis of COVID and its related complications. While there are several invasive (e.g., coronary epicardial vasoreactivity (QCA) using provocation testing with intracoronary acetylcholine) and non-invasive (e.g., flow-mediated-dilation and peripheral arterial tonometry) tools to assess endothelial dysfunction, serum biomarkers will likely emerge as surrogate evidence for endotheliitis [9]. Similarly, addressing endothelial dysfunction and inhibiting the inflammatory response will likely be the underpinnings of a successful preventive and therapeutic strategy. The observations of EC viral infection and subsequent injury provide a rationale for therapies aimed at stabilizing the endothelium with anti-inflammatory and immune-modulating drugs. Clinical use of inhibitors of complement such as eculizumab (anti-C5 monoclonocal antibody) and RUKONEST (C1 inhibitor) have been reported [16,17]. In addition to more widespread use of IL-6 inhibitors such as tocilizumab, TNF inhibitors [18], ACE inhibitors [19], statins [20] and endothelin receptor antagonists [21] are being actively pursued. In addition to the aforementioned inflammation-modulating therapy, another important approach to reversing endothelial dysfunction is to enhance the vasoprotective effect of nitric oxide (NO), which is well known to have a wide range of biological properties involving vasodilation, angiogenesis, and anti-thrombosis. In the setting of endothelial dysfunction, there is impaired nitric oxide bioavailability either by diminished production by endothelial nitric oxide synthase or excess oxidative degradation. Ongoing research is examining the use of inhaled NO in COVID-19 with dual effect as a pulmonary vasodilator and direct antiviral activity by interfering with S-protein-ACE-2 interaction [22,23]. NO is amongst the most vital vasodilator molecules, and also inhibits other important events in the development of platelet adhesion and aggregation [9]. Finally, the role of anticoagulation is being pursued aggressively given the development of venous and arterial thrombosis in these patients. In conclusion, understanding the relationship between COVID-19, endotheliitis, pre-existing endothelial dysfunction, and observed endothelial injury will be imperative to developing new therapeutic targets to alter the trajectory of this pandemic. We also believe that the understanding of the relationship between endothelial health and susceptibility to severe COVID-19 and other infections may have implications for public health policy in the future.

          Related collections

          Most cited references18

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

            Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19

            Progressive respiratory failure is the primary cause of death in the coronavirus disease 2019 (Covid-19) pandemic. Despite widespread interest in the pathophysiology of the disease, relatively little is known about the associated morphologic and molecular changes in the peripheral lung of patients who die from Covid-19.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: A report of five cases

              Acute respiratory failure and a systemic coagulopathy are critical aspects of the morbidity and mortality characterizing infection with severe acute respiratory distress syndrome-associated coronavirus-2 (SARS-CoV-2), the etiologic agent of Coronavirus disease 2019 (COVID-19). We examined skin and lung tissues from 5 patients with severe COVID-19 characterized by respiratory failure (n=5) and purpuric skin rash (n=3). The pattern of COVID-19 pneumonitis was predominantly a pauci-inflammatory septal capillary injury with significant septal capillary mural and luminal fibrin deposition and permeation of the inter-alveolar septa by neutrophils. No viral cytopathic changes were observed and the diffuse alveolar damage (DAD) with hyaline membranes, inflammation, and type II pneumocyte hyperplasia, hallmarks of classic ARDS, were not prominent. These pulmonary findings were accompanied by significant deposits of terminal complement components C5b-9 (membrane attack complex), C4d, and mannose binding lectin (MBL)-associated serine protease (MASP)2, in the microvasculature, consistent with sustained, systemic activation of the alternative and lectin-based complement pathways. The purpuric skin lesions similarly showed a pauci-inflammatory thrombogenic vasculopathy, with deposition of C5b-9 and C4d in both grossly involved and normally-appearing skin. In addition, there was co-localization of COVID-19 spike glycoproteins with C4d and C5b-9 in the inter-alveolar septa and the cutaneous microvasculature of two cases examined. In conclusion, at least a subset of sustained, severe COVID-19 may define a type of catastrophic microvascular injury syndrome mediated by activation of complement pathways and an associated procoagulant state. It provides a foundation for further exploration of the pathophysiologic importance of complement in COVID-19, and could suggest targets for specific intervention.
                Bookmark

                Author and article information

                Journal
                J Clin Med
                J Clin Med
                jcm
                Journal of Clinical Medicine
                MDPI
                2077-0383
                15 June 2020
                June 2020
                : 9
                : 6
                : 1862
                Affiliations
                [1 ]Division of Cardiovascular Medicine, University of Connecticut, Farmington, CT 06105, USA; mosleh@ 123456uchc.edu (W.M.); kachen@ 123456uchc.edu (K.C.)
                [2 ]Division of Cardiovascular Medicine, VACT Healthcare System, West Haven, CT 06111, USA; steven.pfau@ 123456yale.edu
                [3 ]Division of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT 06510, USA
                [4 ]Division of Cardiovascular Medicine, Saint Francis Hospital, Hartford, CT 06205, USA
                Author notes
                [* ]Correspondence: vashist@ 123456uchc.edu
                [†]

                Brief Title: COVID-19, Endotheliitis and Endothelial Dysfunction.

                Article
                jcm-09-01862
                10.3390/jcm9061862
                7356402
                32549229
                c5eebf70-9617-4bc1-98d2-e6a280136716
                © 2020 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 04 June 2020
                : 11 June 2020
                Categories
                Editorial

                Comments

                Comment on this article