Skip to main content
Erschienen in: Critical Care 1/2021

Open Access 01.12.2021 | COVID-19 | Review

SARS-CoV-2 pneumonia—receptor binding and lung immunopathology: a narrative review

verfasst von: Maria Clara Saad Menezes, Diego Vinicius Santinelli Pestana, Gustavo Rosa Gameiro, Luiz Fernando Ferraz da Silva, Ėlodie Baron, Jean-Jacques Rouby, José Otavio Costa Auler Jr

Erschienen in: Critical Care | Ausgabe 1/2021

Abstract

The current pandemic of COVID-19 caused thousands of deaths and healthcare professionals struggle to properly manage infected patients. This review summarizes information about SARS-CoV-2 receptor binding dynamics and intricacies, lung autopsy findings, immune response patterns, evidence-based explanations for the immune response, and COVID-19-associated hypercoagulability.
Hinweise
Search strategy and selection criteria: references for this review were identified through searches of PubMed for articles published any date before September, 2020, by use of the terms “SARS-CoV-2”, “pneumonia”, “ARDS”, “ACE2”, “immunology” and “pathology”. Articles resulting from these searches and relevant references cited in those articles were reviewed. Articles published in English were included.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ACE
Angiotensin converting enzyme
ACE-2
Angiotensin converting enzyme 2
Ang
Angiotensin
ARDS
Acute respiratory distress syndrome
BK
Bradykinin
CARDS
COVID-19 acute respiratory distress syndrome
COVID-19
Coronavirus disease 19
DAD
Diffuse alveolar damage
GCSF
Granulocyte colony stimulating factor
HCoV
Human coronavirus
HLH
Haemophagocytic lymphohistiocytosis
ICU
Intensive care unit
IFN
Interferon
IL
Interleukin
MERS-CoV
Middle East respiratory syndrome coronavirus
SARS-CoV
Severe acute respiratory syndrome coronavirus
SARS-CoV-2
Severe acute respiratory syndrome coronavirus 2
S
Spike
TNF
Tumor necrosis factor

Introduction

In December 2019, a new type of human coronavirus (HCoV) was identified [1] and named severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), due to its similarity to severe acute respiratory syndrome coronavirus (SARS-CoV) [2, 3].
SARS-CoV-2 causes a disease named coronavirus disease 19 (COVID-19), which main symptoms are cough, fatigue, anorexia, myalgias, anosmia, ageusia and diarrhea. Even though most COVID-19 patients have moderate symptoms and a quick recovery, some patients develop COVID 19 acute respiratory distress syndrome (CARDS). In contrast to acute respiratory distress syndrome (ARDS), CARDS is initially characterized by severe hypoxemia associated to relatively preserved lung compliance until the development of more aggressive phases. Patients may present quite clinically comfortable with a "silent hypoxemia" in early stages [4]. Moreover, dissociation between the laboratory values and imaging presentation is not uncommon [5]. CARDS may be presented into two subtypes: type H (high elastance similar to conventional ARDS) and type L (low elastance) and recognizing them based on CT scan characteristics, for instance, may be paramount to provide appropriate care [6]. Most often, type L precedes type H which rarely appears as the primary form of severe SARS-CoV-2 pneumonia. Ventilation-perfusion mismatch with predominantly dead space areas over shunt portions of the lung could be a hallmark of CARDS [7]. A study advocates that a better alveolar recruitment and greater oxygenation is achieved with high PEEP values even in the L subtype with a high risk of haemodynamic compromise and alveolar hyperinflation [8].
Emerging and reemerging viral threats, such as HCoVs, have continued to challenge public health systems and incur economic and social costs to both individuals and countries [9]. Coronaviruses are enveloped non-segmented positive sense RNA viruses [10] that have long been considered inconsequential pathogens. However, in the twenty-first century, two highly pathogenic HCoVs—SARS-CoV and Middle East respiratory syndrome coronavirus—presumably emerged from animal reservoirs to cause global epidemics [11]. Given the high prevalence and wide distribution of coronaviruses, and increasing human–animal interface activities, novel coronaviruses are likely to emerge periodically as a consequence of frequent cross-species infections and occasional spillover events [12, 13]. Virus-induced direct pulmonary cytopathic effects, viral evasion of host immune responses and exuberant inflammatory responses are believed to play major roles in disease severity [14]. Yet, recent studies with humans who had severe SARS-CoV-2 pneumonia suggest that dysregulation of the immune response results in a compromising inflammation leading to CARDS and lethal outcomes [15]. In this review we aim to discuss recent advances in the understanding of SARS-CoV-2 pneumonia pathogenesis.

SARS-CoV-2 receptor binding

Angiotensin-converting enzyme 2 (ACE2) is a membrane-bound monocarboxypeptidase found ubiquitously in humans and expressed primarily in pulmonary endothelial cells, alveolar epithelial type II cells, heart, intestine and kidney [16, 17]. ACE2 catalytically removes the last amino acid of angiotensin II (Ang-II), thereby generating the vasodilatory, antifibrotic, antiproliferative and antigrowth peptide Ang-(1–7), which counterbalance the vasoactive Ang-II effects. Investigations focusing on ACE2 have revealed a variety of roles not just catalytic but also as an amino acid transporter and a viral receptor [18]. As shown in Table 1, recent studies have demonstrated that ACE2, which is the main entry receptor of SARS-CoV, is also related to SARS-CoV-2 pathogenicity [1, 19].
Table 1
SARS-CoV-2 and receptor binding
Mechanisms of SARS-CoV-2 receptor binding
SARS-CoV-2: similar receptor-binding domain structure to SARS-CoV [20]
SARS-CoV-2 uses angiotensin-converting enzyme 2 to target cells [1]
SARS-CoV-2 and SARS-CoV S protein affinity to angiotensin-converting enzyme 2: similar [21] or ~ 10- to 20-fold higher [22]
SARS-CoV-2 S protein: requires transmembrane protease serine 2 for S protein priming [26]
SARS-CoV-2 S protein: furin-like protease recognition pattern [25]
Soluble recombinant human ACE2 can inhibit SARS-CoV-2 infections [27]
COVID 19 patients: Greater number of angiotensin-converting enzyme 2-positive lung endothelial cells compared with uninfected controls [29]
Although SARS-CoV-2 shares similarity with coronaviruses isolated from bats, its receptor binding domain structure is very similar to that of SARS-CoV [1, 2, 20]. ACE2-expressing human airway epithelial cells cultures inoculated with SARS-CoV-2 show cytopathic effects 96 h after inoculation, including lack of cilia beating. In cultures not expressing ACE-2, cytopathic effects were not observed suggesting that SARS-CoV-2 uses ACE2 as a viral entry receptor [2]. Tight binding between SARS-CoV-2 spike (S) protein and ACE2 partially explains the efficient transmission of SARS-CoV-2 in humans. There is evidence that SARS-CoV-2 S protein binds to ACE2 with an affinity that is 10- to 20-fold higher than the affinity between SARS-CoV S protein and ACE2 [21, 22].
The nature of the cell protease that cleaves the S glycoprotein varies according to the coronavirus. There is evidence that SARS-CoV uses the cellular transmembrane protease serine 2 for S protein priming [23]. Recently, evidence was found that SARS-CoV-2 S protein also uses transmembrane protease serine 2 [24]. Additionally, SARS-CoV-2 S-protein sequence has a specific furin-like protease recognition pattern present in the vicinity of one of the maturation sites of the S protein that is absent in SARS-CoV sequences. Furin protease is a proprotein convertase that is responsible for the activation of precursor proteins, such as growth factors, hormones, receptors and adhesion molecules, as well as cell surface glycoproteins of infectious viruses, thereby having the potential to cleave specifically viral envelope glycoproteins, and enhance viral fusion with host cell membrane [25, 26].
Finally, recombinant human soluble ACE2 molecule—but not mouse soluble ACE2—can significantly inhibit SARS-CoV-2 infections and reduce viral load by a factor of 1000–5000 by reducing viral S protein binding to membrane-bound ACE2. This finding might be used for studying potential therapeutic interventions for COVID-19 [27]. In addition, SARS-CoV polyclonal antibodies inhibit the entry of SARS-CoV-2 into target cells, providing a basis for the design of vaccines (Fig. 1) [21].

Lung pathology and biomarkers of SARS-CoV-2-induced epithelial and endothelial cells injury

A series of lung autopsies of laboratory confirmed COVID-19 patients have contributed to elucidate the immunopathology behind SARS-CoV-2 pneumonia and the development of CARDS. The main findings are summarized in Table 2.
Table 2
Lung autopsy findings from COVID-19 patients
Lung pathology characteristic of COVID-19
Lung oedema [28, 29, 34]
Diffuse alveolar damage [28, 29, 3438]
Multiple thrombi on the distal lumen of pulmonary vessels [28, 29, 35, 38]
T cells around pulmonary vessels, bronchioles and within interstitium (mainly CD4 and CD8) [28, 29, 31, 34]
Intussusceptive angiogenesis predominating over sprouting angiogenesis [29]
Pulmonary endothelial cells: twice as many angiotensin-converting enzyme 2 when compared to pneumocytes [29]
Pulmonary endothelial cells: disruption of intercellular junctions, swelling, shrinking of capillary lumen, loss of contact with the basal membrane [29, 34]
Gross examination of lungs from patients with SARS-CoV-2 pneumonia revealed haemorrhagic lung oedema, unfrequently associated with pleural effusions and focal haemorrhages [28, 29]. Multiple thrombi are often visible within the lumen of pulmonary vessels (Figs. 2 and 3c). On light microscopy, perivascular lymphocytic inflammation with preservation of distal airways lumen is the main histological characteristic of SARS-CoV-2 pneumonia at early and late stages (Fig. 3a–d). SARS-CoV-2 pneumonia differs markedly from bacterial ventilator-associated pneumonia where polymorphonuclear leucocytic inflammation centered on an infected bronchiole is the typical histological pattern [3033]. As shown in Fig. 3e–g, CD8 and CD4-positive T-cells are the predominant lymphocytes identified around pulmonary vessels, bronchioles and within interstitial spaces [28, 29, 31, 34]. Sparse infiltrates of CD3-positive T-lymphocytes can be identified within the alveolar septa with a few CD20-positive B-lymphocytes. Diffuse alveolar damage (DAD), a nonspecific pattern observed in non-COVID ARDS, is also a frequent histological finding [28, 29, 31, 3438]. During the exudative (acute) phase, DAD is characterized by interstitial oedema, acute and chronic pulmonary inflammation, type 2-pneumocytes hyperplasia, and hyaline membrane formation (Fig. 3d). Lung aeration is preserved at the early phase of severe SARS CoV-2 pneumonia, explaining preservation of respiratory compliance and the characteristic lung ultrasound pattern of diffuse coalescent B lines (Fig. 3a) [33]. During the organizing (healing) phase that is observed after several days in the ICU, the features are similar to those of an organizing pneumonia: granulation tissue (loose accumulations of collagen-embedding fibroblasts and myofibroblasts) and mild chronic inflammation (lymphocytes and plasma cells) (Fig. 3d) [34]; those findings are more rarely found in SARS-CoV-2 patients because lung autopsy is often performed before the patients have entered the healing phase.
Multiple thrombi are present in the lumen of distal pulmonary vessels including capillaries [28, 29, 34, 35, 38]. These vascular obstructions are frequently observed in non-COVID ARDS [39] and cannot be considered as COVID-19 specific. Associated with vascular thrombi, an early and intense angiogenesis is observed [29]. Compared to patients with severe influenza A (H1N1) pneumonia, angiogenesis in COVID-19 patients is early and massive, resulting in distorted and chaotic alveolar plexus (Fig. 4b), increases with duration of hospitalization (Fig. 4e,f) and occurs predominantly by intussusception (Fig. 4c,e,f). In non-COVID ARDS, tortuous neovascularization is also present at the early and late phases [39] and angiogenesis mechanisms are unknown. Additional studies are required to elucidate how intussusceptive angiogenesis impacts the clinical outcome of COVID-19.
SARS-CoV-2 can be directly visualized by electron microscopy [2, 28, 29, 35, 3843] or evidenced on histologic slices by immunostaining [28, 29, 31, 36, 38, 40, 42]. A note of caution should be added: using electron microscopy, viral particles can be confused with cross sections of rough endoplasmic reticulum [4042]. Viral particles are not isolated and free in the cytoplasm, but multiple, inside membrane-bound cisternae located within the Golgi area of the rough endoplasmic reticulum. SARS-CoV-2 particles are found in alveolar type II cells with apparent viral cytopathic effect consisting of cytomegaly, and enlarged nuclei with bright, eosinophilic nucleoli [28, 38, 43], in distal airway epithelial cells [2, 38], in pulmonary [29] and renal [35, 40, 44] endothelial cells [Fig. 5]. There are strong arguments to think that SARS-CoV-2 predominantly infect endothelial cells. In addition to the perivascular accumulation of lymphocytes, pulmonary endothelial cells express twice as many ACE2 receptors for viral entry than pneumocytes [29]. Pulmonary and renal endothelial cells are frequently and morphologically injured with disruption of intercellular junctions, cell swelling, shrinking of the capillary lumen, and a loss of contact with the basal membrane, all findings consistent with a central role of endothelial cells in the vascular phase of COVID-19 [29].
Immunostaining demonstrates a prominent expression of SARS-CoV-2 Rp3 NP protein on alveolar epithelial cells, another evidence of direct infection by SARS-CoV-2 [43]. Finally, SARS-CoV viral particles and viral genome have been detected in monocytes and lymphocytes [45]. However, this finding has not yet been confirmed in SARS-CoV-2 infection. Viral injury of epithelial and endothelial cells observed in CARDS is indirectly confirmed by the increase in specific biomarkers. Increase in surfactant protein D plasma level, a biomarker of alveolar type II-pneumocyte injury, is associated with the development of CARDS and macrophage activating syndrome in critically ill patients with severe SARS-CoV-2 pneumonia [46]. Surfactant protein D level is also negatively correlated with PaO2/FiO2 ratio in those patients, suggesting that surfactant deficiency resulting from injured type II-pneumocytes may contribute to the development of atelectasis and hypoxemia. Unfortunately, the soluble form of the receptor for advanced glycation end product (sRAGE), a well-established biomarker of alveolar type I-pneumocyte injury in non-COVID ARDS [47], has not yet been reported in CARDS patients. It would be interesting to compare plasma and bronchoalveolar lavage sRAGE levels in critically ill patients with CARDS and non-COVID ARDS and assess whether the sRAGE level would allow to separate focal from nonfocal CARDS phenotypes and allow personalized mechanical ventilation [48]. Increase in Angiopoietin-2, soluble E-selectin, and intercellular adhesion molecule 1 plasma levels, all considered as biomarkers of endothelial injury, was predictive of CARDS and admission to the ICU [49, 50]. Interestingly, levels of Angiopoietin-2 were negatively correlated with pulmonary compliance in patients on mechanical ventilation. Very likely, viral infection of endothelial cells triggers high permeability type pulmonary oedema and diffuse alveolar damage, resulting in impaired respiratory mechanics [49]. Similarly, high initial plasma levels of intercellular adhesion molecule 1 were associated with severe forms of SARS-CoV-2 pneumonia, and significantly decreased with recovery, suggesting an alleviation of endothelial cell injury [50].

Circulating lymphocytes and neutrophils during SARS-CoV-2 pneumonia

Lymphocytes and neutrophils are involved in SARS-CoV-2-induced lung injury. Lymphocytopenia was present in 83.2% of 1099 patients with COVID-19 on admission [51]. Moreover, lymphocytopenia prevalence is higher in ICU than in non-ICU patients (85 vs 54%) and is a risk factor for CARDS [12, 44, 52]. Decrease in lymphocyte count mainly concerns CD8 [53], CD4 and CD3 T-cells [15].
Neutrophilia is also a common finding in severe COVID-19 and is considered a risk factor for CARDS and death [15]. Neutrophils’ antimicrobial and inflammatory functions are mediated by an armamentarium of proteins stored in granules and by the formation of neutrophil extracellular traps [54]. The toxic nature of these traps may pose, however, a threat to highly vascularized tissues such as the lungs. A cell-intrinsic program modifying the circulating neutrophils’ proteome and reducing the neutrophil extracellular traps-forming capacity, protects the lungs against neutrophil-induced inflammatory injury [55]. Finally, there is also evidence of neutrophils immunometabolic reprogramming in COVID-19 patients with increased cytosolic pyruvate kinase muscle, HIF-1α and lactate [56].

Cytokine responses during SARS-CoV-2 pneumonia

Cytokines are a broad category of small proteins (< 40 kDa) that are produced and released for cell signaling and immunomodulation [57]. An effective and well-coordinated immune response is the first line of defense against viral infections, whereas supraphysiologic immune response can cause organ damage.
The term ''cytokine storm'' is used to express the exuberant inflammatory response observed in severe viral infections [58]. Cytokine storm syndrome is a hyperinflammatory state characterized by fulminant multi-organ failure and elevation of cytokine levels [59]. In COVID-19, the immune response is characterized by high plasma levels of interleukins (IL-6, IL-2), interferons (IFN-y,) chemokines (CXCL10, CCL2, CCL3), growth factors (granulocyte colony stimulating factor) and tumor necrosis factor (TNFα). A high level of IL-10 has also been reported; however, the level of IL-10 was lower in patients with severe COVID-19 when compared to patients with mild COVID-19 [52, 53, 60, 61]. The cytokine profile in the serum is summarized in Table 3.
Table 3
Cytokine profile during SARS-CoV-2 pneumonia
Cytokine response during SARS-CoV-2 pneumonia
Severe COVID-19: higher IL-2, IL-7, granulocyte colony-stimulating factor, IP-10, macrophage inflammatory protein1 and tumor necrosis factor-α [44, 52]
Severe COVID-19: lower IL-10 when compared to stable COVID-19 [56]
IL-2, IL-4, IFN-γ and TNF-α: maximum serum level in severe SARS-CoV-2 pneumonia 3–6 days after the disease onset [53]
IL-6: sustained increase and started decreasing around 13–16 days in severe SARS-CoV-2 pneumonia [53]
Cytokine profile in COVID-19 patients resembles haemophagocytic lymphohistiocytosis [69]
Cytokine profile in COVID-19 ARDS less exuberant than in hyper inflammatory phenotype of non-COVID ARDS [72]
IL-6 plays a pivotal role in promoting the inflammatory response observed in severe SARS-CoV-2 pneumonia [62]. The baseline IL-6 plasma level is correlated with pneumonia severity and extension of computed tomography (CT) opacities [63]. Significant decreases in IL-6 and CT opacities are associated with patient’s recovery, whereas time-dependent increase in IL-6 predicts mortality [6365]. Tocilizumab, a humanized monoclonal antibody, specifically designed to bind soluble receptors for IL-6, could be a therapeutic option for treating severe CARDS [66]. However, a recently published randomized controlled trial did not show any reduction in disease aggravation, admission to the ICU and mortality [67]. Of note, IL-6 is involved not only in the activation of the immune system but also in regenerative processes (anti-inflammatory properties) [68].
Finally, the cytokine profile in COVID-19 patients resembles haemophagocytic lymphohistiocytosis (HLH) syndrome [69]. The cytokine profile of HLH is characterized by high levels of IFN-γ, TNF-α, IL-6, IL-10, and IL-12 [70], a similar pattern to what is found in severe COVID-19 [52, 53, 60, 61]. Other cardinal features of HLH such as cytopenias and hyperferritinemia are also a common finding in severe COVID-19 [51, 61]. HLH is an aggressive and life-threatening syndrome of excessive immune activation. The hyperinflammatory/dysregulated immune state is thought to be caused by the absence of normal downregulation by activated macrophages and lymphocytes causing an excessive cytokine production by macrophages, natural killer cells, and cytotoxic lymphocytes [71].
A note of caution must be added. The relevance of the cytokine storm to COVID-19 pathogenesis has been criticized. There is evidence that the cytokine profile in CARDS is less exuberant when compared with previous cohorts of patients with non-COVID ARDS and the median IL-6 level is 10- to 200-fold lower in CARDS when compared to the hyperinflammatory phenotype of non-COVID ARDS [72]. As a consequence, it is highly likely that the “cytokine storm” is observed in high inflammatory phenotypes of CARDS and is not a characteristic of SARS-CoV-2 pneumonia.

Possible causes of the “cytokine storm” in severe COVID-19 patients

The immune response seen in critically ill COVID-19 patients is characterized by lymphopenia, neutrophilia and the ''cytokine storm'', which mechanisms are incompletely understood. The host and viral mechanisms associated with SARS-CoV-2-induced immune response are summarized in Table 4.
Table 4
Host and viral mechanisms of SARS-CoV-2-pneumonia-associated pathogenesis
Propositions of host and viral mechanisms of SARS-CoV-2-associated pathogenesis
Direct viral induced pathology
Innate immune responses
Adaptive immune responses
- ACE2 impaired function
- Cytokine overproduction
- Impaired anti-inflammatory properties
- Endothelial dysfunction
- Neutrophilia
- Excessive cytokine production
- Denudation of the airways
 
- Amplification of local and systemic inflammation
The previously mentioned interaction of SARS-CoV-2 with ACE2 per se may be a primary step in the development of an exuberant lung inflammatory response. ACE2 cleaves Angiotensin I into Ang-(1–9) (an inactive peptide) which is converted to Ang-(1–7) (a peptide with vasodilatory properties), counterbalancing the effects of Ang-II. When ACE2 is attached by SARS-CoV-2 S protein, its intracellular domain induces down-regulation of ACE2 activity, promoting a shift towards the downstream pathway of Ang-II (activation of AngII/angiotensin type 1 receptor axis). Consequently, Ang-II/angiotensin type 1 receptor axis activation leads to glycoprotein 130-mediated activation of signal transducer and activator of transcription 3. This cascade, associated with direct stimulation of pattern recognition receptors, promotes an intense activity of nuclear factor kappa B, which in turns generates increased transcription of IL-6, and triggers the cytokine storm [18, 73].
Ang-(1–7) interacts with mitochondrial assembly receptor (in bronchial smooth muscle and epithelium) initiating an intracellular cascade that yields inhibition of the p38 mitogen-activated protein kinase and nuclear factor-kappa B pathways, ultimately leading to decreased levels of proinflammatory cytokines (such as IL-6, TNF-a, and IL-8) and decreased expression of leukocyte extravasation factors (like intercellular adhesion molecule-1 and vascular cell adhesion molecule-1). Ang-(1–7) also modulates the activities of the extracellular-signal-regulated kinase 1/2 pathway, which modulates the production of IL-10, making the downregulation of ACE2 even worse. Moreover, another function of ACE2 is to cleave terminal residue of [des-Arg9]-bradykinin (BK), a known pulmonary inflammatory factor. [des-Arg9]-BK is a constituent of the kinin-kallikrein system, which acts via BKB1 receptor (BKB1R) and bradykinin B2 receptor (BKB2R). BKB1R expression is modulated by inflammatory cytokines (i.e. IL-1ß and TNF-α via nuclear factor-kappa B activity) and its downstream effect promotes neutrophil migration to pulmonary tissue (via chemokine C-X-C motif chemokine 5), fibroblast growth factor-2 expression, and increased IL-1β and monocyte chemotactic protein 1 levels. BKB2R is stimulated by bradykinin and doesn't appear to be involved in the major events of SARS-CoV-2 pneumonia. In contrast, BKB1R as an important pathway by which down-regulation of ACE2 leads to inflammation [74].
Lymphopenia, characterized by a reduction in peripheral CD4 and CD8 T-cells, is also a prominent feature of severe COVID-19 [64]. As mentioned above, SARS-CoV-like viral particles and SARS-CoV RNA were isolated from peripheral T-lymphocytes [45]. As the receptor-binding domain from SARS-CoV and SARS-CoV-2 shares a lot of similarities [20], it is reasonable to hypothesize that SARS-CoV-2 can directly infect T-cells. This finding associated with other mechanisms such as cell death induced by Fas and Fas ligand interaction and TNF-α-related apoptosis-inducing ligand axis [75], could contribute to the lymphopenia. Although there is a decrease in the absolute count of CD8 and CD4 T-lymphocytes, those cells are found in an overactivated state, harboring high concentrations of cytotoxic granules able to induce severe immune injury [36]. On the other hand, the loss of CD4 T-lymphocytes may cause inflammation as a consequence of impaired production of anti-inflammatory cytokines [73].
Neutrophils and macrophages may also play a role in cytokine overproduction. In influenza infection, there is evidence that lung epithelial cells, macrophages, and dendritic cells all express cytokines via activation of pattern recognition receptors including toll-like receptors (3, 7 and 8) retinoic acid-inducible gene I, and the nucleotide-binding oligomerization domain-like receptor family members [76]. Further studies are needed to assess whether such mechanisms play a role in COVID 19.
Another source of activation of the innate immune response is the endothelialitis evidenced in autopsies from COVID-19 patients [29, 40]. It is possible that as in influenza infection, once the fragile endothelial layer is broken, cytokine and viral antigen exposure can amplify inflammation, with endothelial cells as a major source of pro-inflammatory cytokines [77]. Accordingly, Li et al. hypothesized that upon this barrier breakage monocytes and neutrophils can migrate to the infection site to clear alveolar exudates with virus particles and infected cells, resulting in a loop of uncontrolled inflammation [78].
Finally, the complement system also stands as a pathway contributing to the cytokine storm [74]. Viral activation of complement normally occurs through each one of the three axes: classical, alternative, and lectin pathways. Some of the downstream products of these pathways directly enhance the production of cytokines. The aflatoxin C5a can induce the release of TNF-α; C5b-C9 complex stimulates the secretion of IL-6 from vascular smooth muscle cells, and C3a works as a stimulus to the production of IL-1, IL-6, and TNF-α. Recent data suggest that SARS-CoV-2 is able to cause an aberrant activation of the complement cascade via viral nucleocapsid protein binding to mannose-associated serine protease-2; this interaction promotes activation of mannose-binding lectin, which leads to downstream stimulation of the complement cascade.
Outlining the importance of the cytokine storm control, the randomized multicenter controlled RECOVERY study demonstrated that a daily dexamethasone dose of 6 mg for 10 days reduces day-28 mortality by 15% in patients mechanically ventilated for a severe SARS-CoV-2 pneumonia [79]. A similar benefit is also found in non-COVID ARDS [80].

COVID-19-associated hypercoagulability

Severe SARS-CoV-2 pneumonia is associated with an increased risk of thromboembolic events, compared to the regular population and to non-COVID-19 ARDS patients [81].
Most of the observed complications are pulmonary embolisms and circuit occlusions during continuous renal replacement therapy or extracorporeal membrane oxygenation. Arterial complications such as strokes, myocardial infarction, renal and mesenteric infarction have also been reported [81]. Elevated levels of anti-phospholipid antibodies have been associated with such events [81]. Laboratory analysis consistently exhibits elevated serum levels of D-dimer and fibrin/fibrinogen degradation products, with mild elevated or normal values of prothrombin time, activated partial thromboplastin time, and platelet count. Serum levels of D-dimer > 2 mg/L are predictive of mortality in critically ill patients with severe SARS-CoV-2 pneumonia [82]. Relatively few cases of disseminated intravascular coagulation have been reported [81], suggesting specific mechanisms for COVID-19-associated hypercoagulability differing from those involved in non-COVD ARDS coagulopathy.
At least four causative mechanisms are suspected: activation of coagulation cascade by the cytokine storm (with involvement of IL-1, IL-6, and tissue factor), impaired functioning of the fibrinolytic system (due to increased release of plasminogen activator inhibitor-1 with a decreased activity of urokinase-type plasminogen activator), inflammation-induced endothelial injury and extensive activation of platelets by pro-inflammatory cytokines and exposition to damaged endothelium [83].
As shown in Fig. 6, macrophages and neutrophils play important roles in the pathogenesis of lung capillary thrombosis. High levels of macrophage inflammatory protein, monocyte chemotactic protein 1 and interferon-inducible protein 10 in the alveolar space as well as the presence of intravascular, extravascular, and intraalveolar neutrophil extracellular traps are illustrative of the importance of these cells in the development of the lung pathologic findings. Moreover, it is possible that SARS-CoV-2 infection is characterized by a direct association between hypercoagulability and inflammation: thrombin binding to proteinase-activated receptor-1 promotes further inflammation by increasing levels of cytokines. Murine models demonstrated decreased inflammation after use of proteinase-activated receptor-1 antagonists [84].
In critically ill patients with SARS-CoV-2 pneumonia, the use of low molecular weight heparin might be associated to a survival benefit, possibly due to a mixed anti-inflammatory/anticoagulation effect [82, 85]. Therefore, some important guidelines recommend use of prophylactic dose low-molecular-weight heparin or unfractionated heparin in all COVID-19 patients requiring hospital admission [86, 87]. Other data encourage the use of therapeutic anticoagulation in patients who met the criteria for sepsis-induced coagulopathy [81]. However, more research is needed to establish indications, safety, and efficacy parameters of thromboprophylaxis.

Conclusions

The pandemic of COVID-19 forced the scientific community to provide a fast response and a considerable amount of new data have been released since January 2020. The present article identified important pathophysiological landmarks of severe SARS-CoV-2 pneumonia. The binding of SARS-CoV-2 S protein to ACE2 is the main mechanism by which the virus invade target cells. SARS-CoV-2 infects predominantly endothelial cells of pulmonary vessels and capillaries because they express a high density of ACE2 receptors, creating a genuine pulmonary endothelialitis with high permeability-type pulmonary oedema, multiple vascular thrombosis, and neovascularization resulting from predominant intussusceptive angiogenesis. COVID-19 is an interstitial pneumonia characterized by the lung accumulation of lymphocytes around pulmonary vessels associated with lymphocytopenia, predominating on CD4 and CD8 T-cells. In severe forms, there is a cytokine storm resulting from interaction of SARS-CoV-2 with ACE2, overactivation of CD8 T-lymphocytes, loss of CD4 T-lymphocytes, impaired production of anti-inflammatory cytokines, and viral activation of complement through its classical, alternative, and lectin pathways. Last but not least, severe SARS-CoV-2 pneumonia is associated to systemic hypercoagulability resulting from SARS-CoV-2-induced endothelial injury, activation of platelets by exposition to damaged endothelium, activation of coagulation cascade by the cytokine storm, and impaired functioning of the fibrinolytic system. The exact mechanisms by which SARS-CoV-2-induced hypercoagulability remain incompletely elucidated and more research is necessary. The recent demonstration that dexamethasone reduces mortality in mechanically ventilated patients with severe SARS-CoV-2 pneumonia outlines the importance of elucidating pathophysiological mechanisms.

Acknowledgements

Figures 3a, c and d and 5a were gently provided by Brazilian Image Autopsy Study Group, Department of Pathology, Hospital das Clínicas HCFMUSP, Universidade de Sao Paulo.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis 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 licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence 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 licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
3.
Zurück zum Zitat Yuntao W, Wenzhe H, Yaowei H, et al. SARS-CoV-2 is an appropriate name for the new coronavirus. Lancet. 2020 21–27 March; 395(10228): 949–950 Yuntao W, Wenzhe H, Yaowei H, et al. SARS-CoV-2 is an appropriate name for the new coronavirus. Lancet. 2020 21–27 March; 395(10228): 949–950
4.
Zurück zum Zitat Gattinoni L, Chiumello D, Cairon P, et al. COVID-19 pneumonia: different respiratory treatments for different phenotypes? Intensive Care Med. 2020;46:1099–102.PubMedPubMedCentralCrossRef Gattinoni L, Chiumello D, Cairon P, et al. COVID-19 pneumonia: different respiratory treatments for different phenotypes? Intensive Care Med. 2020;46:1099–102.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Xu L, Xiaochun M. Acute respiratory failure in COVID-19: is it “typical” ARDS? Crit Care. 2020;24:198.CrossRef Xu L, Xiaochun M. Acute respiratory failure in COVID-19: is it “typical” ARDS? Crit Care. 2020;24:198.CrossRef
8.
Zurück zum Zitat Salvatore G, Mirabella L, Murgolo F, et al. Effects of positive end-expiratory pressure in “High Compliance” Severe Acute Respiratory Syndrome Coronavirus 2 Acute Respiratory Distress Syndrome, Critical Care Medicine: September 15, 2020—Volume Online First - Issue - doi: https://doi.org/10.1097/CCM.0000000000004640 Salvatore G, Mirabella L, Murgolo F, et al. Effects of positive end-expiratory pressure in “High Compliance” Severe Acute Respiratory Syndrome Coronavirus 2 Acute Respiratory Distress Syndrome, Critical Care Medicine: September 15, 2020—Volume Online First - Issue - doi: https://​doi.​org/​10.​1097/​CCM.​0000000000004640​
10.
Zurück zum Zitat Richman D, Whitley R, Hayden F. Clinical virology. 4th ed. Washington: ASM Press; 2016.CrossRef Richman D, Whitley R, Hayden F. Clinical virology. 4th ed. Washington: ASM Press; 2016.CrossRef
11.
Zurück zum Zitat Paules CI, Marston HD, Fauci AS. Coronavirus infections—more than just the common cold. JAMA. 2020;323:707–8.PubMedCrossRef Paules CI, Marston HD, Fauci AS. Coronavirus infections—more than just the common cold. JAMA. 2020;323:707–8.PubMedCrossRef
12.
Zurück zum Zitat Cui J, Li F, Shi ZL. Origin and evolution of pathogenic coronaviruses. Nat Rev Microbiol. 2019;17:181–92.CrossRefPubMed Cui J, Li F, Shi ZL. Origin and evolution of pathogenic coronaviruses. Nat Rev Microbiol. 2019;17:181–92.CrossRefPubMed
13.
14.
Zurück zum Zitat Channappanavar R, Perlman S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. Semin Immunopathol. 2017;39:529–39.PubMedPubMedCentralCrossRef Channappanavar R, Perlman S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. Semin Immunopathol. 2017;39:529–39.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Wu C, Chen X, Cai Y, et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 Pneumonia in Wuhan, China. JAMA Intern Med. 180:934–3. Wu C, Chen X, Cai Y, et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 Pneumonia in Wuhan, China. JAMA Intern Med. 180:934–3.
16.
Zurück zum Zitat Rice GI, Thomas DA, Grant PJ, et al. Evaluation of angiotensin-converting enzyme (ACE), its homologue ACE2 and neprilysin in angiotensin peptide metabolism. Biochem J. 2004;383:45–51.PubMedPubMedCentralCrossRef Rice GI, Thomas DA, Grant PJ, et al. Evaluation of angiotensin-converting enzyme (ACE), its homologue ACE2 and neprilysin in angiotensin peptide metabolism. Biochem J. 2004;383:45–51.PubMedPubMedCentralCrossRef
17.
19.
20.
Zurück zum Zitat Lu R, Zhao X, Li J, et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020;395:565–74.CrossRefPubMedPubMedCentral Lu R, Zhao X, Li J, et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet. 2020;395:565–74.CrossRefPubMedPubMedCentral
22.
23.
Zurück zum Zitat Matsuyama S, Nagata N, Shirato K, et al. Efficient activation of the severe acute respiratory syndrome coronavirus spike protein by the transmembrane protease TMPRSS2. J Virol. 2010;84:12658–64.PubMedPubMedCentralCrossRef Matsuyama S, Nagata N, Shirato K, et al. Efficient activation of the severe acute respiratory syndrome coronavirus spike protein by the transmembrane protease TMPRSS2. J Virol. 2010;84:12658–64.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Shulla A, Heald-Sargent T, Subramanya G, et al. A transmembrane serine protease is linked to the severe acute respiratory syndrome coronavirus receptor and activates virus entry. J Virol. 2011;85:873–82.PubMedCrossRef Shulla A, Heald-Sargent T, Subramanya G, et al. A transmembrane serine protease is linked to the severe acute respiratory syndrome coronavirus receptor and activates virus entry. J Virol. 2011;85:873–82.PubMedCrossRef
25.
Zurück zum Zitat Coutard B, Valle C, de Lamballerie X, et al. The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antiviral Res. 2020;176:104742.PubMedPubMedCentralCrossRef Coutard B, Valle C, de Lamballerie X, et al. The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antiviral Res. 2020;176:104742.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat 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;181:271–80.PubMedPubMedCentralCrossRef 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;181:271–80.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Monteil V, Kwon H, Prado P, et al. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell. 2020;181(4):905–13.PubMedPubMedCentralCrossRef Monteil V, Kwon H, Prado P, et al. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell. 2020;181(4):905–13.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Fox S, Akmatbekov A, Harbert L, et al. Pulmonary and cardiac pathology in African American patients with COVID-19: an autopsy series from New Orleans, . Lancet Respir Med. 2020;8:682.CrossRef Fox S, Akmatbekov A, Harbert L, et al. Pulmonary and cardiac pathology in African American patients with COVID-19: an autopsy series from New Orleans, . Lancet Respir Med. 2020;8:682.CrossRef
29.
Zurück zum Zitat Ackermann M, Verleden S, Kuehnel M, et al. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in covid-19. NEJM. 2020;383:122–4.CrossRef Ackermann M, Verleden S, Kuehnel M, et al. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in covid-19. NEJM. 2020;383:122–4.CrossRef
30.
Zurück zum Zitat Rouby JJ, Martin De Lassale E, Poete P et al. Nosocomial bronchopneumonia in the critically ill. Histologic and bacteriologic aspects. Am Rev Respir Dis. 1992;146:1059–66 Rouby JJ, Martin De Lassale E, Poete P et al. Nosocomial bronchopneumonia in the critically ill. Histologic and bacteriologic aspects. Am Rev Respir Dis. 1992;146:1059–66
31.
Zurück zum Zitat Barton L, Duval E, Stroberg E, et al. COVID-19 Autopsies, Oklahoma, USA. Am J Clin Path. 2020;153:725–33.PubMedCrossRef Barton L, Duval E, Stroberg E, et al. COVID-19 Autopsies, Oklahoma, USA. Am J Clin Path. 2020;153:725–33.PubMedCrossRef
33.
Zurück zum Zitat Monteiro RPA, Oliveira EP, Saldiva PHN, Dolhnikoff M. Duarte‐Neto AN; Brazilian Image Autopsy Study Group. Histological‐ultrasonographical correlation of pulmonary involvement in severe COVID‐19. Intensive Care Med. 2020; 46:1766–8. doi: https://doi.org/10.1007/s00134-020-06125-z. Monteiro RPA, Oliveira EP, Saldiva PHN, Dolhnikoff M. Duarte‐Neto AN; Brazilian Image Autopsy Study Group. Histological‐ultrasonographical correlation of pulmonary involvement in severe COVID‐19. Intensive Care Med. 2020; 46:1766–8. doi: https://​doi.​org/​10.​1007/​s00134-020-06125-z.
36.
Zurück zum Zitat Xu Z, Shi L, Wang Y, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8:420–2.PubMedPubMedCentralCrossRef Xu Z, Shi L, Wang Y, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8:420–2.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat 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;15:700–4.PubMedPubMedCentralCrossRef 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;15:700–4.PubMedPubMedCentralCrossRef
38.
39.
Zurück zum Zitat Tomashefski JF Jr. Pulmonary pathology of acute respiratory distress syndrome. Clin Chest Med. 2000;21:455.CrossRef Tomashefski JF Jr. Pulmonary pathology of acute respiratory distress syndrome. Clin Chest Med. 2000;21:455.CrossRef
43.
Zurück zum Zitat Zhang H, Zhou P, Wei Y, et al. Histopathologic changes and SARS-CoV-2 immunostaining in the lung of a patient with COVID-19. Ann Inter Med. 2020;172:629–32.CrossRef Zhang H, Zhou P, Wei Y, et al. Histopathologic changes and SARS-CoV-2 immunostaining in the lung of a patient with COVID-19. Ann Inter Med. 2020;172:629–32.CrossRef
44.
Zurück zum Zitat Su H, Yang M, Wan C, Yi LX, et al. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int. 2020;98(1):219–27.PubMedPubMedCentralCrossRef Su H, Yang M, Wan C, Yi LX, et al. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int. 2020;98(1):219–27.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Kerget B, Kerget F, Koçak AO, Kızıltunç A, Araz Ö, Uçar EY, et al. Are serum interleukin 6 and surfactant protein D levels associated with the clinical course of COVID-19? Lung. 2020;777–84. Kerget B, Kerget F, Koçak AO, Kızıltunç A, Araz Ö, Uçar EY, et al. Are serum interleukin 6 and surfactant protein D levels associated with the clinical course of COVID-19? Lung. 2020;777–84.
47.
Zurück zum Zitat Mrozek S, Jabaudon M, Jaber S, Paugam-Burtz C, Lefrant J-Y, Rouby J-J, et al. Elevated plasma levels of sRAGE are associated with nonfocal CT-based lung imaging in patients with ARDS: a prospective multicenter study. Chest. 2016;150:998–1007.PubMedCrossRef Mrozek S, Jabaudon M, Jaber S, Paugam-Burtz C, Lefrant J-Y, Rouby J-J, et al. Elevated plasma levels of sRAGE are associated with nonfocal CT-based lung imaging in patients with ARDS: a prospective multicenter study. Chest. 2016;150:998–1007.PubMedCrossRef
48.
Zurück zum Zitat Constantin JM, Jabaudon M, Lefrant JY, Jaber S, Quenot JP, Langeron O, et al.; AZUREA Network. Personalised mechanical ventilation tailored to lung morphology versus low positive end-expiratory pressure for patients with acute respiratory distress syndrome in France (the LIVE study): a multicentre, single-blind, randomised controlled trial. Lancet Respir Med. 2019 Oct;7(10):870–80. Constantin JM, Jabaudon M, Lefrant JY, Jaber S, Quenot JP, Langeron O, et al.; AZUREA Network. Personalised mechanical ventilation tailored to lung morphology versus low positive end-expiratory pressure for patients with acute respiratory distress syndrome in France (the LIVE study): a multicentre, single-blind, randomised controlled trial. Lancet Respir Med. 2019 Oct;7(10):870–80.
49.
Zurück zum Zitat Smadja DM, Guerin CL, Chocron R, Yatim N, Boussier J, Gendron N, et al. Angiopoietin-2 as a marker of endothelial activation is a good predictor factor for intensive care unit admission of COVID-19 patients. Angiogenesis. 2020;611–20. Smadja DM, Guerin CL, Chocron R, Yatim N, Boussier J, Gendron N, et al. Angiopoietin-2 as a marker of endothelial activation is a good predictor factor for intensive care unit admission of COVID-19 patients. Angiogenesis. 2020;611–20.
50.
Zurück zum Zitat Tong M, Jiang Y, Xia D, Xiong Y, Zheng Q, Chen F, et al. Elevated expression of serum endothelial cell adhesion molecules in COVID-19 patients. J Infect Dis. 2020;894–8. Tong M, Jiang Y, Xia D, Xiong Y, Zheng Q, Chen F, et al. Elevated expression of serum endothelial cell adhesion molecules in COVID-19 patients. J Infect Dis. 2020;894–8.
51.
Zurück zum Zitat Guan W, Ni Z, Hu Y, et al. Clinical characteristics of coronavirus disease 2019 in china. NEJM. 2020;382:1708–20.PubMedCrossRef Guan W, Ni Z, Hu Y, et al. Clinical characteristics of coronavirus disease 2019 in china. NEJM. 2020;382:1708–20.PubMedCrossRef
52.
54.
Zurück zum Zitat Cowland J, Borregaard N. Granulopoiesis and granules of human neutrophils. Immunol Rev. 2016;273:11–28.PubMedCrossRef Cowland J, Borregaard N. Granulopoiesis and granules of human neutrophils. Immunol Rev. 2016;273:11–28.PubMedCrossRef
55.
Zurück zum Zitat Adrover J, Aroca-Crevillen A, Crainiciuc G, et al. Programmed ‘disarming’ of the neutrophil proteome reduces the magnitude of inflammation. Nat Immunol. 2020;21:135–44.PubMedPubMedCentralCrossRef Adrover J, Aroca-Crevillen A, Crainiciuc G, et al. Programmed ‘disarming’ of the neutrophil proteome reduces the magnitude of inflammation. Nat Immunol. 2020;21:135–44.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat McElvaney OJ, McEvoy NL, McElvaney OF, Carroll TP, Murphy MP, Dunlea DM, et al. Characterization of the inflammatory response to severe COVID-19 illness. Am J Respir Crit Care Med. 2020;202:812–21.PubMedPubMedCentralCrossRef McElvaney OJ, McEvoy NL, McElvaney OF, Carroll TP, Murphy MP, Dunlea DM, et al. Characterization of the inflammatory response to severe COVID-19 illness. Am J Respir Crit Care Med. 2020;202:812–21.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Liu Q, Zhou Y, Yang Z. The cytokine storm of severe influenza and development of immunomodulatory therapy. Cell Mol Immunol. 2016;3:3–10.CrossRef Liu Q, Zhou Y, Yang Z. The cytokine storm of severe influenza and development of immunomodulatory therapy. Cell Mol Immunol. 2016;3:3–10.CrossRef
59.
Zurück zum Zitat Chousterman B, Swirski F, Weber G. Cytokine storm and sepsis disease pathogenesis. Semin Immunopathol. 2017;39:517–28.CrossRefPubMed Chousterman B, Swirski F, Weber G. Cytokine storm and sepsis disease pathogenesis. Semin Immunopathol. 2017;39:517–28.CrossRefPubMed
60.
Zurück zum Zitat Chen N, Zhou M, Dong X, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395:507–13.PubMedPubMedCentralCrossRef Chen N, Zhou M, Dong X, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395:507–13.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Ruan Q, Yang K, Wang W, et al. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020;46:846–8.PubMedCrossRef Ruan Q, Yang K, Wang W, et al. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020;46:846–8.PubMedCrossRef
63.
64.
Zurück zum Zitat 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;395:1054–62.PubMedPubMedCentralCrossRef 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;395:1054–62.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Salvarani C, Dolci G, Massari M, et al. RCT-TCZ-COVID-19 Study Group. Effect of tocilizumab vs standard care on clinical worsening in patients hospitalized With COVID-19 pneumonia: a randomized clinical trial. JAMA Intern Med. 2020;20:e206615. Salvarani C, Dolci G, Massari M, et al. RCT-TCZ-COVID-19 Study Group. Effect of tocilizumab vs standard care on clinical worsening in patients hospitalized With COVID-19 pneumonia: a randomized clinical trial. JAMA Intern Med. 2020;20:e206615.
69.
Zurück zum Zitat Mehta P, McAuley DF, Brown M, et al. HLH Across Speciality Collaboration, UK COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet. 2020;395(10229):1033–4.PubMedPubMedCentralCrossRef Mehta P, McAuley DF, Brown M, et al. HLH Across Speciality Collaboration, UK COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet. 2020;395(10229):1033–4.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Arico M, Danesino C, Pende D, et al. Pathogenesis of haemophagocytic lymphohistiocytosis. Br J Haematol. 2001;114:761–9.PubMedCrossRef Arico M, Danesino C, Pende D, et al. Pathogenesis of haemophagocytic lymphohistiocytosis. Br J Haematol. 2001;114:761–9.PubMedCrossRef
73.
Zurück zum Zitat Wan S, Yi Q, Fan S, et al. Relationships among lymphocyte subsets, cytokines, and the pulmonary inflammation index in coronavirus (COVID-19) infected patients. Br J Haematol. 2020;189:428–37.PubMedPubMedCentralCrossRef Wan S, Yi Q, Fan S, et al. Relationships among lymphocyte subsets, cytokines, and the pulmonary inflammation index in coronavirus (COVID-19) infected patients. Br J Haematol. 2020;189:428–37.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Peteranderl C, Herold S. The impact of the interferon/TNF-related apoptosis-inducing ligand signaling axis on disease progression in respiratory viral infection and beyond. Front Immunol. 2017;8:313.PubMedPubMedCentral Peteranderl C, Herold S. The impact of the interferon/TNF-related apoptosis-inducing ligand signaling axis on disease progression in respiratory viral infection and beyond. Front Immunol. 2017;8:313.PubMedPubMedCentral
77.
Zurück zum Zitat Teijaro J, Walsh K, Cahalan S, et al. Endothelial cells are central orchestrators of cytokine amplification during influenza virus infection. Cell. 2011;146:980–9.PubMedPubMedCentralCrossRef Teijaro J, Walsh K, Cahalan S, et al. Endothelial cells are central orchestrators of cytokine amplification during influenza virus infection. Cell. 2011;146:980–9.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Hui L, Liang L, Dingyu Z, Li H, Liu L, Zhang D, et al. SARS-CoV-2 and viral sepsis: observations and hypotheses. Lancet. 2020;395:1517–20.CrossRef Hui L, Liang L, Dingyu Z, Li H, Liu L, Zhang D, et al. SARS-CoV-2 and viral sepsis: observations and hypotheses. Lancet. 2020;395:1517–20.CrossRef
80.
Zurück zum Zitat Villar J, Ferrando C, Martínez D, et al. Dexamethasone treatment for the acute respiratory distress syndrome: a multicentre, randomised controlled trial. Lancet Respir Med. 2020;8:267–76.CrossRefPubMed Villar J, Ferrando C, Martínez D, et al. Dexamethasone treatment for the acute respiratory distress syndrome: a multicentre, randomised controlled trial. Lancet Respir Med. 2020;8:267–76.CrossRefPubMed
81.
Zurück zum Zitat Helms J, Tacquard C, Severac F, et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med. 2020;46(6):1089–98.PubMedCrossRefPubMedCentral Helms J, Tacquard C, Severac F, et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med. 2020;46(6):1089–98.PubMedCrossRefPubMedCentral
85.
Zurück zum Zitat Paranjpe I, Fuster V, Lala A, et al. Association of treatment dose anticoagulation with in-hospital survival among hospitalized patients with COVID-19. J Am Coll Cardiol. 2020;S0735–1097(20):35218–9. Paranjpe I, Fuster V, Lala A, et al. Association of treatment dose anticoagulation with in-hospital survival among hospitalized patients with COVID-19. J Am Coll Cardiol. 2020;S0735–1097(20):35218–9.
86.
Zurück zum Zitat Thachil J, Tang N, Gando S, et al. ISTH interim guidance on recognition and management of coagulopathy in COVID-19. J Thromb Haemost. 2020;18:1023–6.PubMedCrossRef Thachil J, Tang N, Gando S, et al. ISTH interim guidance on recognition and management of coagulopathy in COVID-19. J Thromb Haemost. 2020;18:1023–6.PubMedCrossRef
87.
Zurück zum Zitat Bikdeli B, Madhavan MV, Jimenez D, et al. COVID-19 and thrombotic or thromboembolic disease: Implications for prevention, antithrombotic therapy, and follow-up. J Am Coll Cardiol. 2020; S0735-1097(20): 35008-7. Bikdeli B, Madhavan MV, Jimenez D, et al. COVID-19 and thrombotic or thromboembolic disease: Implications for prevention, antithrombotic therapy, and follow-up. J Am Coll Cardiol. 2020; S0735-1097(20): 35008-7.
Metadaten
Titel
SARS-CoV-2 pneumonia—receptor binding and lung immunopathology: a narrative review
verfasst von
Maria Clara Saad Menezes
Diego Vinicius Santinelli Pestana
Gustavo Rosa Gameiro
Luiz Fernando Ferraz da Silva
Ėlodie Baron
Jean-Jacques Rouby
José Otavio Costa Auler Jr
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Schlagwort
COVID-19
Erschienen in
Critical Care / Ausgabe 1/2021
Elektronische ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-020-03399-z

Weitere Artikel der Ausgabe 1/2021

Critical Care 1/2021 Zur Ausgabe

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.