27
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Long Covid: where we stand and challenges ahead

      review-article

      Read this article at

      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

          Post-acute sequelae of SARS-CoV-2 (PASC), also known as Post-Covid Syndrome, and colloquially as Long Covid, has been defined as a constellation of signs and symptoms which persist for weeks or months after the initial SARS-CoV-2 infection. PASC affects a wide range of diverse organs and systems, with manifestations involving lungs, brain, the cardiovascular system and other organs such as kidney and the neuromuscular system. The pathogenesis of PASC is complex and multifactorial. Evidence suggests that seeding and persistence of SARS-CoV-2 in different organs, reactivation, and response to unrelated viruses such as EBV, autoimmunity, and uncontrolled inflammation are major drivers of PASC. The relative importance of pathogenetic pathways may differ in different tissue and organ contexts. Evidence suggests that vaccination, in addition to protecting against disease, reduces PASC after breakthrough infection although its actual impact remains to be defined. PASC represents a formidable challenge for health care systems and dissecting pathogenetic mechanisms may pave the way to targeted preventive and therapeutic approaches.

          Related collections

          Most cited references120

          • 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

              Post-acute COVID-19 syndrome

              Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogen responsible for the coronavirus disease 2019 (COVID-19) pandemic, which has resulted in global healthcare crises and strained health resources. As the population of patients recovering from COVID-19 grows, it is paramount to establish an understanding of the healthcare issues surrounding them. COVID-19 is now recognized as a multi-organ disease with a broad spectrum of manifestations. Similarly to post-acute viral syndromes described in survivors of other virulent coronavirus epidemics, there are increasing reports of persistent and prolonged effects after acute COVID-19. Patient advocacy groups, many members of which identify themselves as long haulers, have helped contribute to the recognition of post-acute COVID-19, a syndrome characterized by persistent symptoms and/or delayed or long-term complications beyond 4 weeks from the onset of symptoms. Here, we provide a comprehensive review of the current literature on post-acute COVID-19, its pathophysiology and its organ-specific sequelae. Finally, we discuss relevant considerations for the multidisciplinary care of COVID-19 survivors and propose a framework for the identification of those at high risk for post-acute COVID-19 and their coordinated management through dedicated COVID-19 clinics.
                Bookmark

                Author and article information

                Contributors
                alberto.mantovani@humanitasresearch.it
                gianni.bussolati@unito.it
                Journal
                Cell Death Differ
                Cell Death Differ
                Cell Death and Differentiation
                Nature Publishing Group UK (London )
                1350-9047
                1476-5403
                7 September 2022
                7 September 2022
                : 1-10
                Affiliations
                [1 ]GRID grid.417728.f, ISNI 0000 0004 1756 8807, IRCCS Humanitas Research Hospital, ; via Manzoni 56, 20089 Rozzano Milan, Italy
                [2 ]GRID grid.452490.e, Department of Biomedical Sciences, , Humanitas University, ; Via Rita Levi Montalcini 4, 20072 Pieve Emanuele Milan, Italy
                [3 ]GRID grid.4868.2, ISNI 0000 0001 2171 1133, William Harvey Research Institute, Queen Mary University, ; London, EC1M 6BQ UK
                [4 ]Scientific Institute Stella Maris (IRCSS), Pisa, Italy
                [5 ]GRID grid.5395.a, ISNI 0000 0004 1757 3729, Department of Translational Research on New Technologies in Medicine and Surgery, , University of Pisa, ; Pisa, Italy
                [6 ]GRID grid.8142.f, ISNI 0000 0001 0941 3192, Institute of Pharmacology, Catholic University School of Medicine, and Fondazione Policlinico Universitario “A. Gemelli” Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), ; Rome, Italy
                [7 ]GRID grid.6530.0, ISNI 0000 0001 2300 0941, Department of Biology, , University of Rome Tor Vergata, ; Rome, Italy
                [8 ]GRID grid.428504.f, ISNI 0000 0004 1781 0034, Institute of Translational Pharmacology, CNR, ; Rome, Italy
                [9 ]GRID grid.5608.b, ISNI 0000 0004 1757 3470, Venetian Institute of Molecular Medicine, University of Padua, ; Padua, Italy
                [10 ]GRID grid.4527.4, ISNI 0000000106678902, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, ; Milan, Italy
                [11 ]GRID grid.466495.c, ISNI 0000 0001 2195 4282, Accademia Nazionale dei Lincei, ; Rome, Italy
                [12 ]GRID grid.7605.4, ISNI 0000 0001 2336 6580, University of Turin, ; Turin, Italy
                [13 ]GRID grid.11450.31, ISNI 0000 0001 2097 9138, University of Sassari, ; Sassari, Italy
                [14 ]GRID grid.25879.31, ISNI 0000 0004 1936 8972, Institute for Translational Medicine and Therapeutics, University of Pennsylvania, ; Philadelphia, PA USA
                [15 ]GRID grid.8404.8, ISNI 0000 0004 1757 2304, University of Florence, ; Florence, Italy
                [16 ]GRID grid.6530.0, ISNI 0000 0001 2300 0941, School of Medicine, , University of Rome “Tor Vergata”, ; Rome, Italy
                [17 ]GRID grid.7841.a, Department of Physics, , University of Rome ‘La Sapienza’’, ; Rome, Italy
                [18 ]Head External R&D Division, Glaxo Smith Kline Vaccines, Siena, Italy
                [19 ]GRID grid.7445.2, ISNI 0000 0001 2113 8111, Vaccines Research, Imperial College of London, ; London, UK
                [20 ]GRID grid.416651.1, ISNI 0000 0000 9120 6856, Istituto Superiore di Sanità, ; Rome, Italy
                [21 ]GRID grid.7445.2, ISNI 0000 0001 2113 8111, School of Public Health, , Imperial College London, ; London, UK
                Author information
                http://orcid.org/0000-0001-5578-236X
                http://orcid.org/0000-0003-2175-5069
                Article
                1052
                10.1038/s41418-022-01052-6
                9449925
                36071155
                cfbafdf8-a0a9-4662-9f3a-384399ace57a
                © The Author(s) 2022

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 9 June 2022
                : 5 August 2022
                : 14 August 2022
                Categories
                Review Article

                Cell biology
                infectious diseases,immunological disorders
                Cell biology
                infectious diseases, immunological disorders

                Comments

                Comment on this article