Skip to main content
Erschienen in: Critical Care 1/2020

Open Access 14.07.2020 | COVID-19 | Review

Role of angiotensin-converting enzyme 2 (ACE2) in COVID-19

verfasst von: Wentao Ni, Xiuwen Yang, Deqing Yang, Jing Bao, Ran Li, Yongjiu Xiao, Chang Hou, Haibin Wang, Jie Liu, Donghong Yang, Yu Xu, Zhaolong Cao, Zhancheng Gao

Erschienen in: Critical Care | Ausgabe 1/2020

Abstract

An outbreak of pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that started in Wuhan, China, at the end of 2019 has become a global pandemic. Both SARS-CoV-2 and SARS-CoV enter host cells via the angiotensin-converting enzyme 2 (ACE2) receptor, which is expressed in various human organs. We have reviewed previously published studies on SARS and recent studies on SARS-CoV-2 infection, named coronavirus disease 2019 (COVID-19) by the World Health Organization (WHO), confirming that many other organs besides the lungs are vulnerable to the virus. ACE2 catalyzes angiotensin II conversion to angiotensin-(1–7), and the ACE2/angiotensin-(1–7)/MAS axis counteracts the negative effects of the renin-angiotensin system (RAS), which plays important roles in maintaining the physiological and pathophysiological balance of the body. In addition to the direct viral effects and inflammatory and immune factors associated with COVID-19 pathogenesis, ACE2 downregulation and the imbalance between the RAS and ACE2/angiotensin-(1–7)/MAS after infection may also contribute to multiple organ injury in COVID-19. The SARS-CoV-2 spike glycoprotein, which binds to ACE2, is a potential target for developing specific drugs, antibodies, and vaccines. Restoring the balance between the RAS and ACE2/angiotensin-(1–7)/MAS may help attenuate organ injuries.

Graphical abstract

SARS-CoV-2 enters lung cells via the ACE2 receptor. The cell-free and macrophage-phagocytosed virus can spread to other organs and infect ACE2-expressing cells at local sites, causing multi-organ injury.
Hinweise
Wentao Ni, Xiuwen Yang and Deqing Yang contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
COVID-19
Coronavirus disease 2019
SARS
Severe acute respiratory syndrome
SARS-CoV-2
Severe acute respiratory syndrome coronavirus 2
ACE2
Angiotensin-converting enzyme 2
RAS
Renin-angiotensin system
MERS
Middle East Respiratory Syndrome
Ang-II
Angiotensin II
AT1R
Angiotensin type 1 receptor
AT2R
Angiotensin type 2 receptor
ARDS
Acute respiratory distress syndrome
DPP4
Dipeptidyl peptidase 4
PICs
Pro-inflammatory cytokines
MCP-1
Monocyte chemoattractant protein-1
IP-10
Interferon-gamma-inducible protein-10
ALI
Acute lung injury
ALT
Alanine aminotransferase
AST
Aspartate aminotransferase
GGT
Gamma-glutamyl transpeptidase
AKI
Acute kidney injury
CK
Creatine kinase
DIC
Disseminated intravascular coagulation
ACEI
ACE inhibitor
ARB
Angiotensin II receptor blocker

Background

At the end of 2019, an outbreak of a novel coronavirus (2019-nCoV) was reported in Wuhan, Hubei Province, China [1, 2]. The outbreak has become a global pandemic. This virus seems to be much more contagious than severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) and Middle East Respiratory Syndrome (MERS) coronavirus (MERS-CoV). By the end of June 8, 2020, there have been more than 7,000,000 confirmed cases of coronavirus disease 2019 (COVID-19), with nearly 400,000 fatalities worldwide.
Full-length genome sequencing revealed that 2019-nCoV shares 79.5% sequence identity with SARS-CoV, and pairwise protein sequence analysis found that it belonged to the class of SARS-related coronaviruses [3]. Both 2019-nCoV and SARS-CoV enter host cell via the same receptor, angiotensin-converting enzyme 2 (ACE2) [3]. Therefore, this virus was subsequently renamed SARS-CoV-2. Although the overall mortality rate of COVID-19 caused by SARS-CoV-2 is lower than that of SARS and MERS, organ dysfunction, such as acute respiratory distress syndrome (ARDS), acute cardiac injury, acute hepatic injury, and acute kidney injury are quite common in severe cases. ACE2, a homolog of angiotensin-converting enzyme (ACE), which is expressed in a variety of human organs and tissues, has extensive biological activities and can counteract the negative role of the renin-angiotensin system (RAS) in many diseases [46]. Considering that the spike protein of SARS-CoV-2 interacts with ACE2, as does that of SARS-CoV, COVID-19 may have a pathogenic mechanism similar to that of SARS. In this review, we will discuss the role of ACE2 in COVID-19, and its potential therapeutic targets, aiming to provide more information on the management of the epidemic.

RAS and ACE2

The RAS is a complex network that plays an important role in maintaining blood pressure as well as electrolyte and fluid homeostasis, affecting the function of many organs, such as the heart, blood vessels, and kidneys [6]. Angiotensin II (Ang-II), which is the most representative bioactive peptide in the RAS, widely participates in the progression of cardiovascular diseases, such as hypertension, myocardial infarction, and heart failure [7]. In the classic RAS, renin cleaves the substrate angiotensinogen to form the decapeptide angiotensin I (Ang-I), and then, ACE removes two amino acids at the carboxyl terminus of Ang-I to yield Ang-II (Fig. 1). To date, three Ang-II receptors have been identified, and the affinities of these receptors for Ang-II are similar, in the nanomolar range [7]. Among these receptors, angiotensin type 1 receptor (AT1R) binds to Ang-II, causing vasoconstriction, cell proliferation, inflammatory responses, blood coagulation, and extracellular matrix remodeling, whereas angiotensin type 2 receptor (AT2R) counteracts the aforementioned effects mediated by AT1R [8].
In 2000, two independent research groups discovered ACE2, a homolog of ACE, which can remove the carboxy-terminal phenylalanine in Ang-II to form the heptapeptide angiotensin-(1–7) [4, 9]. In addition, under the alternating effects of ACE2 and ACE, angiotensin-(1–7) can be formed without Ang-II (Fig. 1). In this metabolic pathway, Ang-I is first hydrolyzed by ACE2 to form angiotensin-(1–9), and angiotensin-(1–9) is then hydrolyzed by ACE to form angiotensin-(1–7). Ang-I can also be directly converted to angiotensin-(1–7) by endopeptidases and oligopeptidases [6]. Because of the higher affinity between ACE and Ang-I, the classical pathway of Ang-II to angiotensin-(1–7) is more common [6]. Angiotensin-(1–7), as a ligand, binds to the G-protein-coupled receptor MAS, which produces the opposite effect to that of Ang-II, and exerts a range of functions in multiple organs/systems [5, 6]. In addition to catalyzing the production of angiotensin-(1–7), ACE2 is involved in the uptake of amino acids in intestinal epithelial cells [10].

ACE2 mediates SARS-CoV-2 infection

Entry into host cells is the first step of viral infection. A spike glycoprotein on the viral envelope of the coronavirus can bind to specific receptors on the membrane of host cells. Previous studies have shown that ACE2 is a specific functional receptor for SARS-CoV [11]. Zhou et al. showed that SARS-CoV-2 can enter ACE2-expressing cells, but not cells without ACE2 or cells expressing other coronavirus receptors, such as aminopeptidase N and dipeptidyl peptidase 4 (DPP4), confirming that ACE2 is the cell receptor for SARS-CoV-2 [3]. Further studies showed that the binding affinity of the SARS-CoV-2 spike glycoprotein to ACE2 is 10- to 20-fold higher than that of SARS-CoV to ACE2 [12]. The probable mechanism of SARS-CoV-2 entry into host cells based on SARS-CoV studies is displayed in Fig. 2. Briefly, the receptor-binding domain of the spike glycoprotein binds to the tip of subdomain I of ACE2 [1114]. Membrane fusion of the virus and the host cell is activated after binding, and viral RNA is subsequently released into the cytoplasm, establishing infection. For SARS-CoV infection, intact ACE2 or its transmembrane domain is internalized together with the virus [15]. The catalytically active site of ACE2 is not occluded by the spike glycoprotein, and the binding process is independent of the peptidase activity of ACE2 [14]. Some transmembrane proteinases (such as a disintegrin and metallopeptidase domain 17 [ADAM17], transmembrane protease serine 2 [TMPRSS2], and TNF-converting enzyme) and proteins (such as vimentin and clathrin) may be involved in the binding and membrane fusion processes [1621]. For example, ADAM17 can cleave ACE2 to cause ectodomain shedding, and TMPRSS2 can cleave ACE2 to promote viral uptake [16, 17].
ACE2 is expressed in nearly all human organs in varying degrees. In the respiratory system, the traditional immunohistochemical method and recently introduced single-cell RNA-seq analysis revealed that ACE2 is mainly expressed on type II alveolar epithelial cells, but weakly expressed on the surface of epithelial cells in the oral and nasal mucosa and nasopharynx, indicating that the lungs are the primary target of SARS-CoV-2 [22, 23]. Moreover, ACE2 is highly expressed on myocardial cells, proximal tubule cells of the kidney, and bladder urothelial cells, and is abundantly expressed on the enterocytes of the small intestine, especially in the ileum [2224]. The cell-free and macrophage phagocytosis-associated virus may spread from the lungs to other organs with high ACE2 expression through blood circulation (Fig. 2). For example, up to 67% of patients who developed diarrhea during the course of SARS and quite a number of patients with COVID-19 showed enteric symptoms [2527]. Active viral replication in enterocytes of the small intestine has been reported, and SARS-CoV-2 has been successfully isolated from fecal specimens [28, 29].

ACE2 is associated with multi-organ injury in COVID-19

Autopsies of SARS patients showed that SARS-CoV infection can cause injury to multiple organs, such as the heart, kidney, liver, skeletal muscle, central nervous system, and adrenal and thyroid glands, besides the lungs [30, 31]. Most critically ill patients with COVID-19 also had multiple organ damage, including acute lung injury, acute kidney injury, cardiac injury, liver dysfunction, and pneumothorax [32]. As with SARS and COVID-19, organ injury is also frequently observed in MERS, especially the gastrointestinal tract and kidneys, while the incidence of acute cardiac injury is less common [3336]. Unlike SARS-CoV and SARS-CoV-2, MESR-CoV uses DPP4 as its entry receptor, which is mainly expressed on pneumocytes, multinucleated epithelial cells, and bronchial submucosal gland cells of the lungs; epithelial cells of the kidney and small intestine; and activated leukocytes [3739]. DPP4 is not abundantly expressed on myocardial cells [3739]. Therefore, this indicates that organ involvement and injury is strongly associated with receptor distribution in the body.
According to the results of a series of studies on SARS, the pathogenesis of COVID-19 should be complex. The virus-induced inflammatory responses, including the excessive expression of cytokines and chemokines, excessive recruitment of inflammatory cells, insufficient interferon response, and possible production of auto-antibodies are deemed to be vital factors in disease pathogenesis [30]. Pro-inflammatory cytokines (PICs) and chemokines in plasma, such as interleukin (IL)-1, IL-6, IL-12, IL-8, monocyte chemoattractant protein-1 (MCP-1), and interferon-gamma-inducible protein 10 (IP-10), are significantly elevated in the plasma of patients with SARS [40, 41]. Significantly increased plasma concentrations of these PICs were also found in severe patients with COVID-19 [1]. Autopsy studies of SARS patients further found that PICs and MCP-1 were highly expressed in SARS-CoV-infected ACE2+ cells, but not in tissues without infected ACE2+ cells, suggesting virus-induced local immune-mediated damage [42].
In addition to acting as the receptor for SARS-CoV and SARS-CoV-2, ACE2 hydrolyzes Ang-II to angiotensin-(1–7), and the ACE2/angiotensin-(1–7)/MAS counteracts the negative effects of the RAS and exerts anti-inflammatory effects [6, 43]. Several studies have shown that SARS-CoV infection can downregulate ACE2 expression on cells, thereby disrupting the physiological balance between ACE/ACE2 and Ang-II/angiotensin-(1–7) and subsequently causing severe organ injury [4447]. Given that SARS-CoV-2 is a species of SARS-related coronaviruses and uses ACE2 as its receptor, the downregulation of ACE2 expression may be involved in multiple organ injury in COVID-19.
Based on previous studies on SARS and recent studies on SARS-CoV-2, the multiple organ injury in COVID-19 (Fig. 3) and the possible role of ACE2 in organ injury are described below.

Acute lung injury

Although the mortality rate in COVID-19 is lower than that in SARS and MERS, numerous patients have acute lung injury (ALI) after infection [26, 32]. Similar to the pathological features of SARS and MERS, severe diffuse alveolar damage, such as extensive edema, hyaline membrane formation, inflammatory infiltrates, microthrombi formation, organization, and fibrosis, was also observed in COVID-19, but with more cellular fibromyxoid exudates in the alveoli and small airways [48, 49]. The role of the RAS and ACE2 in ARDS/ALI has drawn great attention since the outbreak of SARS in 2003. Clinical studies have found that ACE insertion/deletion polymorphism may be correlated with the severity of ARDS [50, 51]. High Ang-II levels in the lungs can increase vascular permeability and cause pulmonary edema [52, 53]. Several studies have revealed the protective effects of the ACE2/angiotensin-(1–7)/MAS axis in the lungs. It alleviates lung inflammation, fibrosis, and pulmonary arterial hypertension, as well as inhibiting cancer cell growth, tumor angiogenesis, and tumor metastasis [6, 5456]. In different animal models of ALI, ACE2-knockout mice exhibited enhanced vascular permeability, increased lung edema, neutrophil accumulation, and marked worsening of lung function compared with wild-type control mice [56]. Injection of recombinant human ACE2 protein or AT1R blockers into ACE2-knockout mice could decrease the degree of ALI [56].
SARS-CoV infection considerably reduces ACE2 expression in mouse lungs [46]. Further experiments showed that mere binding of recombinant SARS-CoV spike-Fc to human and mouse ACE2 could result in the downregulation of cell-surface ACE2 expression [46]. The spike-Fc protein worsened acid-induced ALI in wild-type mice, but did not affect the severity of lung failure in ACE2-knockout mice, indicating that the effect of spike protein on ALI is ACE2-specific [46]. Studies on influenza also found that ACE2 was significantly downregulated after H1N1 infection [57]. ACE2 deficiency significantly worsened the pathogenesis in infected mice, and inhibition of AT1 alleviated the severity of influenza H7N9 virus-induced lung injury [58, 59]. Moreover, Ang-II levels were elevated in H5N1- and H7N9-infected patients, which was associated with the severity of lung injury and predicted fatal outcomes in H7N9-infected patients [59, 60]. In patients with COVID-19, plasma Ang-II levels were markedly elevated and linearly associated with viral load and lung injury [61]. All these findings suggest that the RAS and ACE2 downregulation contribute to the pathogenesis of lung injury in COVID-19.

Acute cardiac injury

The heart abundantly expresses ACE2, indicating that it is vulnerable to SARS-Cov-2 infection. Autopsies of patients with SARS revealed that 35% of them (7 of 20) were positive for the SARS-CoV genome in cardiac tissue, and patients with SARS-CoV cardiac infections had a more aggressive illness and earlier mortality than those without [47]. Edema of the myocardial stroma, inflammatory cell infiltration, and atrophy of cardiac muscle fibers was observed in patients with SARS and myocardial damage [30, 31, 6264]. Cardiac injury is quite common among severely ill patients with COVID-19, and we found that early acute myocardial injury was associated with a higher risk of mortality [65].
The beneficial role of the ACE2/angiotensin-(1–7)/MAS axis in the heart has been well demonstrated [6]. It can induce vasorelaxation of coronary vessels, inhibit oxidative stress, attenuate pathological cardiac remodeling, and improve postischemic heart function [66]. ACE2 expression usually increases at the initial stage of heart injury, but decreases as the disease progresses [7]. ACE2 knockout in mice results in myocardial hypertrophy and interstitial fibrosis and accelerates heart failure [67, 68]. In addition, ACE2 knockout in mice aggravates cardiac dysfunction caused by diabetes [69]. In both SARS-CoV-infected mice and humans, ACE2 expression in myocardial cells is markedly downregulated in the heart [47]. According to recent studies [26, 70] and our data [65], a substantial number of patients with severe disease have hypertension as a comorbidity. Over-activation of the RAS may have already occurred in these individuals before infection. The significant downregulation of ACE2 and upregulation of Ang-II in COVID-19 results in RAS over-activation, and loss of the protective effects of angiotensin-(1–7) may aggravate and perpetuate cardiac injuries.

Digestive system injury

The gastrointestinal tract, especially the intestine, is vulnerable to SARS-CoV and SARS-CoV-2 infections. SARS-CoV particles have been detected in epithelial cells of the intestinal mucosa, but not in the esophagus and stomach [30, 42]. The main pathological finding in the intestines of patients with SARS was the depletion of mucosal lymphoid tissue [71]. Only mild focal inflammation was detected in the gastrointestinal tract [71]. These findings may explain why gastrointestinal manifestations in COVID-19 are not severe and are transient.
Many patients with COVID-19 show a slight to moderate increase in serum levels of alanine aminotransferase (ALT) and/or aspartate aminotransferase (AST) during the course of infection [26, 72]. Autopsies of SARS patients revealed fatty degeneration, hepatocyte necrosis, and cellular infiltration in the liver [30]. However, SARS-CoV was not detected in the hepatic tissue of most patients autopsied [30]. Both immunohistochemistry and single-cell RNA-seq analyses showed that hepatocytes, Kupffer cells, and the endothelial lining of the sinusoids were negative for ACE2; only cholangiocytes were positive for ACE2 [22, 23, 73]. Gamma-glutamyl transpeptidase (GGT), which reflects cholangiocyte damage, was elevated in some COVID-19 patients [74]. These findings indicate that most acute hepatic injury may not be due to virus infection, but is highly likely due to other causes, such as drug hepatotoxicity, hypoxia, and systemic inflammation. Whether SARS-CoV-2 causes damage to the bile ducts by binding with ACE2 on cholangiocytes requires further investigation.

Acute kidney injury

ACE2 is highly expressed in the kidney, especially in the apical membranes of proximal tubular epithelial cells, suggesting that the kidney is another target of SARS-CoV-2 [22, 23, 75]. Moreover, an imbalance between Ang-II and angiotensin-(1–7) caused by ACE2 deficiency may aggravate the vulnerability of the kidney to other factors causing acute kidney injury (AKI) [76]. SARS-CoV was detected in epithelial cells of the distal tubules, and viral sequences were identified in urinary samples from some patients [77, 78]. SARS-CoV-2 has also been isolated from urinary samples [79]. A retrospective analysis of 536 SARS patients showed that 6.7% of patients developed acute renal impairment during the course of the disease [80]. A large cohort study from New York showed that the incidence of AKI among patients with COVID-19 could reach 36.6% [81].

Other organ and tissue injuries

Pancreas

Pancreatic cells highly express ACE2, indicating that COVID-19 may affect the pancreas [82]. It has been reported that up to 16% of patients with severe COVID-19 have elevated serum amylase and lipase levels, with 7% displaying accompanying significant pancreatic changes on CT scans [83]. Clinical presentation of acute pancreatitis has been reported in patients with COVID-19 [84]. ACE2/angiotensin-(1–7) plays a protective role in diabetes by improving pancreatic β cell survival, stimulating insulin secretion, and reducing insulin resistance [6]. Studies have shown that, compared to patients with non-SARS pneumonia, many more SARS patients who had no previous diabetes and had not received steroid treatment developed insulin-dependent acute diabetes during hospitalization [85, 86]. Moreover, plasma glucose levels and diabetes are independent predictors of mortality in patients with SARS [86]. Autopsies of some SARS patients found atrophy and amyloid degeneration in most pancreatic islets, suggesting the virus causes damage to the islets [64]. Therefore, COVID-19 may also influence pancreatic function, similar to SARS, and glucose levels should be closely monitored, especially in patients with diabetes or glucocorticoid treatment.

Skeletal muscles

Muscle weakness and elevated serum creatine kinase (CK) levels were observed in more than 30% of patients with SARS [87]. Mildly to moderately elevated CK levels were also observed in patients with COVID-19 on admission [88]. Myofiber necrosis and atrophy were observed in skeletal muscle tissues, but no SARS-CoV particles were detected by electron microscopy [30, 89]. Recent studies revealed that the RAS plays an important role in the pathogenesis of various skeletal muscle disorders, and the ACE2/angiotensin-(1–7)/MAS axis exerts protective effects against muscle atrophy [6]. Nevertheless, whether SARS-CoV-2 attacks the muscles and whether the downregulation of ACE2 is associated with myopathy is unclear.

Central nervous system

ACE2 is widely present in the brain, predominantly in neurons, and participates in the neural regulation of broad physiological functions, such as cardiovascular and metabolic activities, stress response, and neurogenesis [6, 90, 91]. In a mouse model, SARS-CoV invaded the brain through the olfactory bulb and then spread transneuronally to other areas [92]. Olfactory and gustatory dysfunctions have been reported in many patients with COVID-19, suggesting the involvement of the olfactory bulb in SARS-CoV-2 infection [93, 94]. SARS-CoV was isolated from human brain tissue specimens [31, 95]. Autopsies showed edema and focal degeneration of neurons in the brains of patients with SARS [30, 31]. Many patients (78/214) had neurologic manifestations in COVID-19, and SARS-CoV-2 was detected in the cerebrospinal fluid of a patient with encephalitis [96, 97]. Considering that SARS-CoV-2 has a much higher affinity for its receptor (ACE2) than SARS-CoV, the former could be capable of infecting and damaging the central nervous system.

Blood vessels

ACE2 is also expressed in the endothelial cells of small and large blood vessels, and the vascular endothelium can produce angiotensin-(1–7) [6, 22]. The ACE2/angiotensin-(1–7)/MAS axis induces vasodilatory, antiproliferative, and antithrombotic effects in the vasculature [6]. SARS RNA can be detected in the endothelia of the small veins in many tissues [98]. Plasma D-dimer levels are significantly elevated in severely ill patients with COVID-19 [1, 32, 72], and the occurrence of disseminated intravascular coagulation (DIC) at the early stage of the disease is not rare. Viral infection and inflammatory responses damage the integrity of the vascular endothelium, causing increased permeability, coagulation activation, and microcirculation disturbances, which may contribute to organ injury in COVID-19.

Potential targets and drugs

As ACE2 is the receptor for both SARS-CoV and SARS-CoV-2, and some transmembrane proteinases such as ADAM17 and TMPRSS are involved in binding and membrane fusion processes, these sites may be potential targets in the development of antiviral drugs for COVID-19 treatment. For example, serum samples from patients with convalescent SARS can neutralize spike-driven entry of SARS-CoV-2 into host cells, suggesting that vaccines targeting the spike protein will be promising [18]. Studies have found that SARS-CoV-specific monoclonal antibodies and recombinant ACE2-Ig can potently neutralize SARS-CoV-2, and a hexapeptide of the receptor-binding domain of the spike protein binds to ACE2, thus blocking SARS-CoV entry [18, 99101].
The downregulation of ACE2 in organs after virus infection disturbs the local balance between the RAS and ACE2/angiotensin-(1–7)/MAS axis, which may be associated with organ injuries. Animal studies have found that ACE inhibitor (ACEI) therapy can increase plasma angiotensin-(1–7) levels, decrease plasma Ang-II levels, and increase cardiac ACE2 expression, whereas angiotensin II receptor blockers (ARBs) can increase the plasma levels of both Ang-II and angiotensin-(1–7) as well as the cardiac expression and activity of ACE2 [102]. Thus, the use of ACEIs/ARBs, renin inhibitors, and angiotensin-(1–7) analogs may attenuate organ injuries by blocking the renin-angiotensin pathway and/or increasing angiotensin-(1–7) levels [103]. Other animal studies showed that ALI mediated by SARS-CoV spike or the influenza virus in mice could be rescued by the use of ARBs [46, 60, 104]. In a population-based study, the application of ACEIs and ARBs significantly reduced the 30-day mortality rate of patients with pneumonia requiring hospitalization [105]. There are also concerns that treatment with ACEIs/ARBs may facilitate infection and increase the risk of developing severe and fatal COVID-19 by increasing ACE2 expression levels in target organs [106]. However, two large cohort studies showed that ACEIs/ARBs use was not associated with increased SARS-CoV-2 infection, but was associated with a lower risk of all-cause mortality in hospitalized patients [107, 108]. Further studies are needed to test the protective effects of ACEIs/ARBs in COVID-19.

Conclusions

The RAS and ACE2/angiotensin-(1–7)/MAS axis play important roles in various physiological and pathophysiological contexts. Both SARS-CoV-2 and SARS-CoV use ACE2 as the receptor for entry into host cells. Because ACE2 is highly expressed in various organs and tissues, SARS-CoV-2 not only invades the lungs but also attacks other organs with high ACE2 expression. The pathogenesis of COVID-19 is highly complex, with multiple factors involved. In addition to the direct viral effects and inflammatory and immune factors, the downregulation of ACE2 and imbalance between the RAS and ACE2/angiotensin-(1–7)/MAS axis may also contribute to the multiple organ injuries in COVID-19. The spike glycoprotein of SARS-CoV-2 is a potential target for the development of specific drugs, antibodies, and vaccines. Restoring the balance between the RAS and ACE2/angiotensin-(1–7)/MAS may help attenuate organ injuries in COVID-19.

Acknowledgements

We would like to thank Editage (www.​editage.​cn) for English language editing.
Not applicable.
Not applicable.

Competing interests

All authors declare 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
1.
Zurück zum Zitat Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497–506.PubMedPubMedCentral Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497–506.PubMedPubMedCentral
2.
Zurück zum Zitat Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, et al. A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med. 2020;382:727–33.PubMedPubMedCentral Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, et al. A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med. 2020;382:727–33.PubMedPubMedCentral
3.
Zurück zum Zitat Zhou P, Yang X, Wang X, Hu B, Zhang L, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–3.PubMedPubMedCentral Zhou P, Yang X, Wang X, Hu B, Zhang L, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–3.PubMedPubMedCentral
4.
Zurück zum Zitat Donoghue M, Hsieh F, Baronas E, Godbout K, Gosselin M, Stagliano N, et al. A novel angiotensin-converting enzyme-related carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1-9. Circ Res. 2000;87:E1–9.PubMed Donoghue M, Hsieh F, Baronas E, Godbout K, Gosselin M, Stagliano N, et al. A novel angiotensin-converting enzyme-related carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1-9. Circ Res. 2000;87:E1–9.PubMed
5.
Zurück zum Zitat Patel S, Rauf A, Khan H, Abu-Izneid T. Renin-angiotensin-aldosterone (RAAS): the ubiquitous system for homeostasis and pathologies. Biomed Pharmacother. 2017;94:317–25.PubMed Patel S, Rauf A, Khan H, Abu-Izneid T. Renin-angiotensin-aldosterone (RAAS): the ubiquitous system for homeostasis and pathologies. Biomed Pharmacother. 2017;94:317–25.PubMed
6.
Zurück zum Zitat Santos R, Sampaio W, Alzamora A, Motta-Santos D, Alenina N, Bader M, et al. The ACE2/angiotensin-(1-7)/MAS axis of the renin-angiotensin system: focus on angiotensin-(1-7). Physiol Rev. 2018;98:505–53.PubMed Santos R, Sampaio W, Alzamora A, Motta-Santos D, Alenina N, Bader M, et al. The ACE2/angiotensin-(1-7)/MAS axis of the renin-angiotensin system: focus on angiotensin-(1-7). Physiol Rev. 2018;98:505–53.PubMed
7.
Zurück zum Zitat Keidar S, Kaplan M, Gamliel-Lazarovich A. ACE2 of the heart: from angiotensin I to angiotensin (1-7). Cardiovasc Res. 2007;73:463–9.PubMed Keidar S, Kaplan M, Gamliel-Lazarovich A. ACE2 of the heart: from angiotensin I to angiotensin (1-7). Cardiovasc Res. 2007;73:463–9.PubMed
8.
Zurück zum Zitat Horiuchi M, Akishita M, Dzau V. Recent progress in angiotensin II type 2 receptor research in the cardiovascular system. Hypertension. 1999;33:613–21.PubMed Horiuchi M, Akishita M, Dzau V. Recent progress in angiotensin II type 2 receptor research in the cardiovascular system. Hypertension. 1999;33:613–21.PubMed
9.
Zurück zum Zitat Tipnis S, Hooper N, Hyde R, Karran E, Christie G, Turner A. A human homolog of angiotensin-converting enzyme. Cloning and functional expression as a captopril-insensitive carboxypeptidase. J Biol Chem. 2000;275:33238–43.PubMed Tipnis S, Hooper N, Hyde R, Karran E, Christie G, Turner A. A human homolog of angiotensin-converting enzyme. Cloning and functional expression as a captopril-insensitive carboxypeptidase. J Biol Chem. 2000;275:33238–43.PubMed
10.
Zurück zum Zitat Hashimoto T, Perlot T, Rehman A, Trichereau J, Ishiguro H, Paolino M, et al. ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature. 2012;487:477–81.PubMedPubMedCentral Hashimoto T, Perlot T, Rehman A, Trichereau J, Ishiguro H, Paolino M, et al. ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature. 2012;487:477–81.PubMedPubMedCentral
11.
Zurück zum Zitat Li W, Moore M, Vasilieva N, Sui J, Wong S, Berne M, et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 2003;426:450–4.PubMedPubMedCentral Li W, Moore M, Vasilieva N, Sui J, Wong S, Berne M, et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 2003;426:450–4.PubMedPubMedCentral
12.
Zurück zum Zitat Wrapp D, Wang N, Corbett K, Goldsmith J, Hsieh C, Abiona O, et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367:1260–3.PubMedPubMedCentral Wrapp D, Wang N, Corbett K, Goldsmith J, Hsieh C, Abiona O, et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367:1260–3.PubMedPubMedCentral
13.
Zurück zum Zitat Song W, Gui M, Wang X, Xiang Y. Cryo-EM structure of the SARS coronavirus spike glycoprotein in complex with its host cell receptor ACE2. PLoS Pathog. 2018;14:e1007236.PubMedPubMedCentral Song W, Gui M, Wang X, Xiang Y. Cryo-EM structure of the SARS coronavirus spike glycoprotein in complex with its host cell receptor ACE2. PLoS Pathog. 2018;14:e1007236.PubMedPubMedCentral
14.
Zurück zum Zitat Li F, Li W, Farzan M, Harrison S. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science. 2005;309:1864–8.PubMed Li F, Li W, Farzan M, Harrison S. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science. 2005;309:1864–8.PubMed
15.
Zurück zum Zitat Kuba K, Imai Y, Ohto-Nakanishi T, Penninger J. Trilogy of ACE2: a peptidase in the renin-angiotensin system, a SARS receptor, and a partner for amino acid transporters. Pharmacol Ther. 2010;128:119–28.PubMedPubMedCentral Kuba K, Imai Y, Ohto-Nakanishi T, Penninger J. Trilogy of ACE2: a peptidase in the renin-angiotensin system, a SARS receptor, and a partner for amino acid transporters. Pharmacol Ther. 2010;128:119–28.PubMedPubMedCentral
16.
Zurück zum Zitat Jia H, Look D, Tan P, Shi L, Hickey M, Gakhar L, et al. Ectodomain shedding of angiotensin converting enzyme 2 in human airway epithelia. Am J Physiol Lung Cell Mol Physiol. 2009;297:L84–96.PubMedPubMedCentral Jia H, Look D, Tan P, Shi L, Hickey M, Gakhar L, et al. Ectodomain shedding of angiotensin converting enzyme 2 in human airway epithelia. Am J Physiol Lung Cell Mol Physiol. 2009;297:L84–96.PubMedPubMedCentral
17.
Zurück zum Zitat Heurich A, Hofmann-Winkler H, Gierer S, Liepold T, Jahn O, Pöhlmann S. TMPRSS2 and ADAM17 cleave ACE2 differentially and only proteolysis by TMPRSS2 augments entry driven by the severe acute respiratory syndrome coronavirus spike protein. J Virol. 2014;88:1293–307.PubMedPubMedCentral Heurich A, Hofmann-Winkler H, Gierer S, Liepold T, Jahn O, Pöhlmann S. TMPRSS2 and ADAM17 cleave ACE2 differentially and only proteolysis by TMPRSS2 augments entry driven by the severe acute respiratory syndrome coronavirus spike protein. J Virol. 2014;88:1293–307.PubMedPubMedCentral
18.
Zurück zum Zitat Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen 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.PubMedPubMedCentral Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen 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.PubMedPubMedCentral
19.
20.
Zurück zum Zitat Yu Y, Chien S, Chen I, Lai C, Tsay Y, Chang S, Chang M. Surface vimentin is critical for the cell entry of SARS-CoV. J Biomed Sci. 2016;23:14.PubMedPubMedCentral Yu Y, Chien S, Chen I, Lai C, Tsay Y, Chang S, Chang M. Surface vimentin is critical for the cell entry of SARS-CoV. J Biomed Sci. 2016;23:14.PubMedPubMedCentral
21.
Zurück zum Zitat Inoue Y, Tanaka N, Tanaka Y, Inoue S, Morita K, Zhuang M, et al. Clathrin-dependent entry of severe acute respiratory syndrome coronavirus into target cells expressing ACE2 with the cytoplasmic tail deleted. J Virol. 2007;81:8722–9.PubMedPubMedCentral Inoue Y, Tanaka N, Tanaka Y, Inoue S, Morita K, Zhuang M, et al. Clathrin-dependent entry of severe acute respiratory syndrome coronavirus into target cells expressing ACE2 with the cytoplasmic tail deleted. J Virol. 2007;81:8722–9.PubMedPubMedCentral
22.
Zurück zum Zitat Hamming I, Timens W, Bulthuis M, Lely A, Navis G, van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 2004;203:631–7.PubMedPubMedCentral Hamming I, Timens W, Bulthuis M, Lely A, Navis G, van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 2004;203:631–7.PubMedPubMedCentral
23.
Zurück zum Zitat Zou X, Chen K, Zou J, Han P, Hao J, Han Z. The single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to Wuhan 2019-nCoV infection. Front Med. 2020;14:185–92.PubMed Zou X, Chen K, Zou J, Han P, Hao J, Han Z. The single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to Wuhan 2019-nCoV infection. Front Med. 2020;14:185–92.PubMed
24.
Zurück zum Zitat Zhang H, Li HB, Lyu JR, Lei XM, Li W, Wu G, et al. Specific ACE2 expression in small intestinal enterocytes may cause gastrointestinal symptoms and injury after 2019-nCoV infection. Int J Infect Dis. 2020;96:19–24.PubMedPubMedCentral Zhang H, Li HB, Lyu JR, Lei XM, Li W, Wu G, et al. Specific ACE2 expression in small intestinal enterocytes may cause gastrointestinal symptoms and injury after 2019-nCoV infection. Int J Infect Dis. 2020;96:19–24.PubMedPubMedCentral
25.
Zurück zum Zitat Leung W, To K, Chan P, Chan H, Wu A, Lee N, et al. Enteric involvement of severe acute respiratory syndrome-associated coronavirus infection. Gastroenterology. 2003;125:1011–7.PubMed Leung W, To K, Chan P, Chan H, Wu A, Lee N, et al. Enteric involvement of severe acute respiratory syndrome-associated coronavirus infection. Gastroenterology. 2003;125:1011–7.PubMed
27.
Zurück zum Zitat Liu K, Fang YY, Deng Y, Liu W, Wang MF, Ma JP, et al. Clinical characteristics of novel coronavirus cases in tertiary hospitals in Hubei Province. Chin Med J. 2020;133:1025–31.PubMedPubMedCentral Liu K, Fang YY, Deng Y, Liu W, Wang MF, Ma JP, et al. Clinical characteristics of novel coronavirus cases in tertiary hospitals in Hubei Province. Chin Med J. 2020;133:1025–31.PubMedPubMedCentral
30.
Zurück zum Zitat Gu J, Korteweg C. Pathology and pathogenesis of severe acute respiratory syndrome. Am J Pathol. 2007;170:1136–47.PubMedPubMedCentral Gu J, Korteweg C. Pathology and pathogenesis of severe acute respiratory syndrome. Am J Pathol. 2007;170:1136–47.PubMedPubMedCentral
31.
Zurück zum Zitat Gu J, Gong E, Zhang B, Zheng J, Gao Z, Zhong Y, et al. Multiple organ infection and the pathogenesis of SARS. J Exp Med. 2005;202:415–24.PubMedPubMedCentral Gu J, Gong E, Zhang B, Zheng J, Gao Z, Zhong Y, et al. Multiple organ infection and the pathogenesis of SARS. J Exp Med. 2005;202:415–24.PubMedPubMedCentral
32.
Zurück zum Zitat Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. 2020;8:475–81.PubMedPubMedCentral Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. 2020;8:475–81.PubMedPubMedCentral
33.
Zurück zum Zitat Hui D, Azhar E, Kim Y, Memish Z, Oh M, Zumla A. Middle East respiratory syndrome coronavirus: risk factors and determinants of primary, household, and nosocomial transmission. Lancet Infect Dis. 2018;18:e217–27.PubMedPubMedCentral Hui D, Azhar E, Kim Y, Memish Z, Oh M, Zumla A. Middle East respiratory syndrome coronavirus: risk factors and determinants of primary, household, and nosocomial transmission. Lancet Infect Dis. 2018;18:e217–27.PubMedPubMedCentral
34.
Zurück zum Zitat Assiri A, McGeer A, Perl TM, Price CS, Al Rabeeah AA, Cummings DA, et al. Hospital outbreak of Middle East Respiratory Syndrome coronavirus. N Engl J Med. 2013;369:407–16.PubMedPubMedCentral Assiri A, McGeer A, Perl TM, Price CS, Al Rabeeah AA, Cummings DA, et al. Hospital outbreak of Middle East Respiratory Syndrome coronavirus. N Engl J Med. 2013;369:407–16.PubMedPubMedCentral
35.
Zurück zum Zitat Hwang SM, Na BJ, Jung Y, Lim HS, Seo JE, Park SA, et al. Clinical and laboratory findings of Middle East Respiratory Syndrome coronavirus infection. Jpn J Infect Dis. 2019;72:160–7.PubMed Hwang SM, Na BJ, Jung Y, Lim HS, Seo JE, Park SA, et al. Clinical and laboratory findings of Middle East Respiratory Syndrome coronavirus infection. Jpn J Infect Dis. 2019;72:160–7.PubMed
36.
Zurück zum Zitat Alsaad KO, Hajeer AH, Al Balwi M, Al Moaiqel M, Al Oudah N, Al Ajlan A, et al. Histopathology of Middle East Respiratory Syndrome coronovirus (MERS-CoV) infection - clinicopathological and ultrastructural study. Histopathology. 2018;72:516–24.PubMed Alsaad KO, Hajeer AH, Al Balwi M, Al Moaiqel M, Al Oudah N, Al Ajlan A, et al. Histopathology of Middle East Respiratory Syndrome coronovirus (MERS-CoV) infection - clinicopathological and ultrastructural study. Histopathology. 2018;72:516–24.PubMed
37.
Zurück zum Zitat Raj VS, Mou H, Smits SL, Dekkers DH, Müller MA, Dijkman R, et al. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature. 2013;495:251–4.PubMedPubMedCentral Raj VS, Mou H, Smits SL, Dekkers DH, Müller MA, Dijkman R, et al. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature. 2013;495:251–4.PubMedPubMedCentral
38.
Zurück zum Zitat Lambeir Am, Durinx C, Scharpé S, De Meester I. Dipeptidyl-peptidase IV from bench to bedside: an update on structural properties, functions, and clinical aspects of the enzyme DPP IV. Crit Rev Clin Lab Sci 2003;40:209–294. Lambeir Am, Durinx C, Scharpé S, De Meester I. Dipeptidyl-peptidase IV from bench to bedside: an update on structural properties, functions, and clinical aspects of the enzyme DPP IV. Crit Rev Clin Lab Sci 2003;40:209–294.
39.
Zurück zum Zitat Boonacker E, Van Noorden CJ. The multifunctional or moonlighting protein CD26/DPPIV. Eur J Cell Biol. 2003;82:53–73.PubMed Boonacker E, Van Noorden CJ. The multifunctional or moonlighting protein CD26/DPPIV. Eur J Cell Biol. 2003;82:53–73.PubMed
40.
Zurück zum Zitat Wong CK, Lam CW, Wu AK, Ip WK, Lee NL, Chan IH, et al. Plasma inflammatory cytokines and chemokines in severe acute respiratory syndrome. Clin Exp Immunol. 2004;136:95–103.PubMedPubMedCentral Wong CK, Lam CW, Wu AK, Ip WK, Lee NL, Chan IH, et al. Plasma inflammatory cytokines and chemokines in severe acute respiratory syndrome. Clin Exp Immunol. 2004;136:95–103.PubMedPubMedCentral
41.
Zurück zum Zitat Zhang Y, Li J, Zhan Y, Wu L, Yu X, Zhang W, et al. Analysis of serum cytokines in patients with severe acute respiratory syndrome. Infect Immun. 2004;72:4410–5.PubMedPubMedCentral Zhang Y, Li J, Zhan Y, Wu L, Yu X, Zhang W, et al. Analysis of serum cytokines in patients with severe acute respiratory syndrome. Infect Immun. 2004;72:4410–5.PubMedPubMedCentral
42.
Zurück zum Zitat He L, Ding Y, Zhang Q, Che X, He Y, Shen H, et al. Expression of elevated levels of pro-inflammatory cytokines in SARS-CoV-infected ACE2+ cells in SARS patients: relation to the acute lung injury and pathogenesis of SARS. J Pathol. 2006;210:288–97.PubMedPubMedCentral He L, Ding Y, Zhang Q, Che X, He Y, Shen H, et al. Expression of elevated levels of pro-inflammatory cytokines in SARS-CoV-infected ACE2+ cells in SARS patients: relation to the acute lung injury and pathogenesis of SARS. J Pathol. 2006;210:288–97.PubMedPubMedCentral
43.
Zurück zum Zitat Rodrigues Prestes TR, Rocha NP, Miranda AS, Teixeira AL, Simoes-E-Silva AC. The anti-inflammatory potential of ACE2/angiotensin-(1-7)/Mas receptor axis: evidence from basic and clinical research. Curr Drug Targets. 2017;18:1301–13.PubMed Rodrigues Prestes TR, Rocha NP, Miranda AS, Teixeira AL, Simoes-E-Silva AC. The anti-inflammatory potential of ACE2/angiotensin-(1-7)/Mas receptor axis: evidence from basic and clinical research. Curr Drug Targets. 2017;18:1301–13.PubMed
44.
Zurück zum Zitat Haga S, Yamamoto N, Nakai-Murakami C, Osawa Y, Tokunaga K, Sata T, et al. Modulation of TNF-alpha-converting enzyme by the spike protein of SARS-CoV and ACE2 induces TNF-alpha production and facilitates viral entry. Proc Natl Acad Sci U S A. 2008;105:7809–14.PubMedPubMedCentral Haga S, Yamamoto N, Nakai-Murakami C, Osawa Y, Tokunaga K, Sata T, et al. Modulation of TNF-alpha-converting enzyme by the spike protein of SARS-CoV and ACE2 induces TNF-alpha production and facilitates viral entry. Proc Natl Acad Sci U S A. 2008;105:7809–14.PubMedPubMedCentral
45.
Zurück zum Zitat Glowacka I, Bertram S, Herzog P, Pfefferle S, Steffen I, Muench MO, et al. Differential downregulation of ACE2 by the spike proteins of severe acute respiratory syndrome coronavirus and human coronavirus NL63. J Virol. 2010;84:1198–205.PubMed Glowacka I, Bertram S, Herzog P, Pfefferle S, Steffen I, Muench MO, et al. Differential downregulation of ACE2 by the spike proteins of severe acute respiratory syndrome coronavirus and human coronavirus NL63. J Virol. 2010;84:1198–205.PubMed
46.
Zurück zum Zitat Kuba K, Imai Y, Rao S, Gao H, Guo F, Guan B, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat Med. 2005;11:875–9.PubMedPubMedCentral Kuba K, Imai Y, Rao S, Gao H, Guo F, Guan B, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat Med. 2005;11:875–9.PubMedPubMedCentral
47.
Zurück zum Zitat Oudit GY, Kassiri Z, Jiang C, Liu PP, Poutanen SM, Penninger JM, et al. SARS-coronavirus modulation of myocardial ACE2 expression and inflammation in patients with SARS. Eur J Clin Investig. 2009;39:618–25. Oudit GY, Kassiri Z, Jiang C, Liu PP, Poutanen SM, Penninger JM, et al. SARS-coronavirus modulation of myocardial ACE2 expression and inflammation in patients with SARS. Eur J Clin Investig. 2009;39:618–25.
49.
Zurück zum Zitat Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8(4):420–2.PubMedPubMedCentral Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8(4):420–2.PubMedPubMedCentral
50.
Zurück zum Zitat Marshall RP, Webb S, Bellingan GJ, Montgomery HE, Chaudhari B, McAnulty RJ, et al. Angiotensin converting enzyme insertion/deletion polymorphism is associated with susceptibility and outcome in acute respiratory distress syndrome. Am J Respir Crit Care Med. 2002;166:646–50.PubMed Marshall RP, Webb S, Bellingan GJ, Montgomery HE, Chaudhari B, McAnulty RJ, et al. Angiotensin converting enzyme insertion/deletion polymorphism is associated with susceptibility and outcome in acute respiratory distress syndrome. Am J Respir Crit Care Med. 2002;166:646–50.PubMed
51.
Zurück zum Zitat Cruces P, Díaz F, Puga A, Erranz B, Donoso A, Carvajal C, et al. Angiotensin-converting enzyme insertion/deletion polymorphism is associated with severe hypoxemia in pediatric ARDS. Intensive Care Med. 2012;38:113–9.PubMed Cruces P, Díaz F, Puga A, Erranz B, Donoso A, Carvajal C, et al. Angiotensin-converting enzyme insertion/deletion polymorphism is associated with severe hypoxemia in pediatric ARDS. Intensive Care Med. 2012;38:113–9.PubMed
52.
Zurück zum Zitat Marshall RP, Gohlke P, Chambers RC, Howell DC, Bottoms SE, Unger T, et al. Angiotensin II and the fibroproliferative response to acute lung injury. Am J Physiol Lung Cell Mol Physiol. 2004;286:L156–64.PubMed Marshall RP, Gohlke P, Chambers RC, Howell DC, Bottoms SE, Unger T, et al. Angiotensin II and the fibroproliferative response to acute lung injury. Am J Physiol Lung Cell Mol Physiol. 2004;286:L156–64.PubMed
53.
Zurück zum Zitat WSD T, Liao W, Zhou S, Mei D, Wong WF. Targeting the renin-angiotensin system as novel therapeutic strategy for pulmonary diseases. Curr Opin Pharmacol. 2018;40:9–17. WSD T, Liao W, Zhou S, Mei D, Wong WF. Targeting the renin-angiotensin system as novel therapeutic strategy for pulmonary diseases. Curr Opin Pharmacol. 2018;40:9–17.
54.
Zurück zum Zitat Jia H. Pulmonary angiotensin-converting enzyme 2 (ACE2) and inflammatory lung disease. Shock. 2016;46:239–48.PubMed Jia H. Pulmonary angiotensin-converting enzyme 2 (ACE2) and inflammatory lung disease. Shock. 2016;46:239–48.PubMed
55.
Zurück zum Zitat Feng Y, Wan H, Liu J, Zhang R, Ma Q, Han B, et al. The angiotensin-converting enzyme 2 in tumor growth and tumor-associated angiogenesis in non-small cell lung cancer. Oncol Rep. 2010;23:941–8.PubMed Feng Y, Wan H, Liu J, Zhang R, Ma Q, Han B, et al. The angiotensin-converting enzyme 2 in tumor growth and tumor-associated angiogenesis in non-small cell lung cancer. Oncol Rep. 2010;23:941–8.PubMed
56.
Zurück zum Zitat Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, et al. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 2005;436:112–6.PubMedPubMedCentral Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, et al. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 2005;436:112–6.PubMedPubMedCentral
57.
Zurück zum Zitat Liu X, Yang N, Tang J, Liu S, Luo D, Duan Q, et al. Downregulation of angiotensin-converting enzyme 2 by the neuraminidase protein of influenza A (H1N1) virus. Virus Res. 2014;185:64–71.PubMedPubMedCentral Liu X, Yang N, Tang J, Liu S, Luo D, Duan Q, et al. Downregulation of angiotensin-converting enzyme 2 by the neuraminidase protein of influenza A (H1N1) virus. Virus Res. 2014;185:64–71.PubMedPubMedCentral
58.
Zurück zum Zitat Yang P, Gu H, Zhao Z, Wang W, Cao B, Lai C, et al. Angiotensin-converting enzyme 2 (ACE2) mediates influenza H7N9 virus-induced acute lung injury. Sci Rep. 2014;4:7027.PubMedPubMedCentral Yang P, Gu H, Zhao Z, Wang W, Cao B, Lai C, et al. Angiotensin-converting enzyme 2 (ACE2) mediates influenza H7N9 virus-induced acute lung injury. Sci Rep. 2014;4:7027.PubMedPubMedCentral
59.
Zurück zum Zitat Zou Z, Yan Y, Shu Y, Gao R, Sun Y, Li X, et al. Angiotensin-converting enzyme 2 protects from lethal avian influenza A H5N1 infections. Nat Commun. 2014;5:3594.PubMed Zou Z, Yan Y, Shu Y, Gao R, Sun Y, Li X, et al. Angiotensin-converting enzyme 2 protects from lethal avian influenza A H5N1 infections. Nat Commun. 2014;5:3594.PubMed
60.
Zurück zum Zitat Huang F, Guo J, Zou Z, Liu J, Cao B, Zhang S, et al. Angiotensin II plasma levels are linked to disease severity and predict fatal outcomes in H7N9-infected patients. Nat Commun. 2014;5:3595.PubMed Huang F, Guo J, Zou Z, Liu J, Cao B, Zhang S, et al. Angiotensin II plasma levels are linked to disease severity and predict fatal outcomes in H7N9-infected patients. Nat Commun. 2014;5:3595.PubMed
61.
Zurück zum Zitat Liu Y, Yang Y, Zhang C, Huang F, Wang F, Yuan J, et al. Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury. Sci China Life Sci. 2020;63:364–74.PubMedPubMedCentral Liu Y, Yang Y, Zhang C, Huang F, Wang F, Yuan J, et al. Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury. Sci China Life Sci. 2020;63:364–74.PubMedPubMedCentral
62.
Zurück zum Zitat Chong PY, Chui P, Ling AE, Franks TJ, Tai DY, Leo YS, et al. Analysis of deaths during the severe acute respiratory syndrome (SARS) epidemic in Singapore: challenges in determining a SARS diagnosis. Arch Pathol Lab Med. 2004;128:195–204.PubMed Chong PY, Chui P, Ling AE, Franks TJ, Tai DY, Leo YS, et al. Analysis of deaths during the severe acute respiratory syndrome (SARS) epidemic in Singapore: challenges in determining a SARS diagnosis. Arch Pathol Lab Med. 2004;128:195–204.PubMed
63.
Zurück zum Zitat Ding Y, Wang H, Shen H, Li Z, Geng J, Han H, et al. The clinical pathology of severe acute respiratory syndrome (SARS): a report from China. J Pathol. 2003;200:282–9.PubMedPubMedCentral Ding Y, Wang H, Shen H, Li Z, Geng J, Han H, et al. The clinical pathology of severe acute respiratory syndrome (SARS): a report from China. J Pathol. 2003;200:282–9.PubMedPubMedCentral
64.
Zurück zum Zitat Lang ZW, Zhang LJ, Zhang SJ, Meng X, Li JQ, Song CZ, et al. A clinicopathological study of three cases of severe acute respiratory syndrome (SARS). Pathology. 2003;35:526–31.PubMed Lang ZW, Zhang LJ, Zhang SJ, Meng X, Li JQ, Song CZ, et al. A clinicopathological study of three cases of severe acute respiratory syndrome (SARS). Pathology. 2003;35:526–31.PubMed
66.
Zurück zum Zitat Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, et al. Angiotensin-converting enzyme 2 and angiotensin 1-7: novel therapeutic targets. Nat Rev Cardiol. 2014;11:413–26.PubMedPubMedCentral Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, et al. Angiotensin-converting enzyme 2 and angiotensin 1-7: novel therapeutic targets. Nat Rev Cardiol. 2014;11:413–26.PubMedPubMedCentral
67.
Zurück zum Zitat Zhong J, Basu R, Guo D, Chow FL, Byrns S, Schuster M, et al. Angiotensin-converting enzyme 2 suppresses pathological hypertrophy, myocardial fibrosis, and cardiac dysfunction. Circulation. 2010;122:717–28 18 p following 28.PubMed Zhong J, Basu R, Guo D, Chow FL, Byrns S, Schuster M, et al. Angiotensin-converting enzyme 2 suppresses pathological hypertrophy, myocardial fibrosis, and cardiac dysfunction. Circulation. 2010;122:717–28 18 p following 28.PubMed
68.
Zurück zum Zitat Oudit GY, Kassiri Z, Patel MP, Chappell M, Butany J, Backx PH, et al. Angiotensin II-mediated oxidative stress and inflammation mediate the age-dependent cardiomyopathy in ACE2 null mice. Cardiovasc Res. 2007;75:29–39.PubMed Oudit GY, Kassiri Z, Patel MP, Chappell M, Butany J, Backx PH, et al. Angiotensin II-mediated oxidative stress and inflammation mediate the age-dependent cardiomyopathy in ACE2 null mice. Cardiovasc Res. 2007;75:29–39.PubMed
69.
Zurück zum Zitat Patel VB, Bodiga S, Basu R, Das SK, Wang W, Wang Z, et al. Loss of angiotensin-converting enzyme-2 exacerbates diabetic cardiovascular complications and leads to systolic and vascular dysfunction: a critical role of the angiotensin II/AT1 receptor axis. Circ Res. 2012;110:1322–35.PubMedPubMedCentral Patel VB, Bodiga S, Basu R, Das SK, Wang W, Wang Z, et al. Loss of angiotensin-converting enzyme-2 exacerbates diabetic cardiovascular complications and leads to systolic and vascular dysfunction: a critical role of the angiotensin II/AT1 receptor axis. Circ Res. 2012;110:1322–35.PubMedPubMedCentral
70.
Zurück zum Zitat Novel Coronavirus Pneumonia Emergency Response Epidemiology Team. The epidemiological characteristics of an outbreak of 2019 novel coronavirus diseases (COVID-19) in China. Zhonghua Liu Xing Bing Xue Za Zhi. 2020;41:145–51. Novel Coronavirus Pneumonia Emergency Response Epidemiology Team. The epidemiological characteristics of an outbreak of 2019 novel coronavirus diseases (COVID-19) in China. Zhonghua Liu Xing Bing Xue Za Zhi. 2020;41:145–51.
71.
Zurück zum Zitat Shi X, Gong E, Gao D, Zhang B, Zheng J, Gao Z, et al. Severe acute respiratory syndrome associated coronavirus is detected in intestinal tissues of fatal cases. Am J Gastroenterol. 2005;100:169–76.PubMed Shi X, Gong E, Gao D, Zhang B, Zheng J, Gao Z, et al. Severe acute respiratory syndrome associated coronavirus is detected in intestinal tissues of fatal cases. Am J Gastroenterol. 2005;100:169–76.PubMed
72.
Zurück zum Zitat Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, 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.PubMedPubMedCentral Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, 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.PubMedPubMedCentral
73.
Zurück zum Zitat Chai X, Hu L, Zhang Y, Han W, Lu Z, Ke A, et al. Specific ACE2 expression in cholangiocytes may cause liver damage after 2019-nCoV infection. Preprint. bioRxiv 931766. Posted online February 04, 2020. Chai X, Hu L, Zhang Y, Han W, Lu Z, Ke A, et al. Specific ACE2 expression in cholangiocytes may cause liver damage after 2019-nCoV infection. Preprint. bioRxiv 931766. Posted online February 04, 2020.
74.
Zurück zum Zitat Fan Z, Chen L, Li J, Cheng X, Yang J, Tian C, et al. Clinical features of COVID-19-related liver functional abnormality. Clin Gastroenterol Hepatol. 2020;18:1561–6.PubMedPubMedCentral Fan Z, Chen L, Li J, Cheng X, Yang J, Tian C, et al. Clinical features of COVID-19-related liver functional abnormality. Clin Gastroenterol Hepatol. 2020;18:1561–6.PubMedPubMedCentral
75.
Zurück zum Zitat Ye M, Wysocki J, William J, Soler MJ, Cokic I, Batlle D. Glomerular localization and expression of angiotensin-converting enzyme 2 and angiotensin-converting enzyme: implications for albuminuria in diabetes. J Am Soc Nephrol. 2006;17:3067–75.PubMed Ye M, Wysocki J, William J, Soler MJ, Cokic I, Batlle D. Glomerular localization and expression of angiotensin-converting enzyme 2 and angiotensin-converting enzyme: implications for albuminuria in diabetes. J Am Soc Nephrol. 2006;17:3067–75.PubMed
76.
Zurück zum Zitat Ortiz-Melo DI, Gurley SB. Angiotensin converting enzyme 2 and the kidney. Curr Opin Nephrol Hypertens. 2016;25:59–66.PubMed Ortiz-Melo DI, Gurley SB. Angiotensin converting enzyme 2 and the kidney. Curr Opin Nephrol Hypertens. 2016;25:59–66.PubMed
77.
Zurück zum Zitat Farcas GA, Poutanen SM, Mazzulli T, Willey BM, Butany J, Asa SL, et al. Fatal severe acute respiratory syndrome is associated with multiorgan involvement by coronavirus. J Infect Dis. 2005;191:193–7.PubMed Farcas GA, Poutanen SM, Mazzulli T, Willey BM, Butany J, Asa SL, et al. Fatal severe acute respiratory syndrome is associated with multiorgan involvement by coronavirus. J Infect Dis. 2005;191:193–7.PubMed
78.
Zurück zum Zitat Chan KH, Poon LL, Cheng VC, Guan Y, Hung IF, Kong J, et al. Detection of SARS coronavirus in patients with suspected SARS. Emerg Infect Dis. 2004;10:294–9.PubMedPubMedCentral Chan KH, Poon LL, Cheng VC, Guan Y, Hung IF, Kong J, et al. Detection of SARS coronavirus in patients with suspected SARS. Emerg Infect Dis. 2004;10:294–9.PubMedPubMedCentral
79.
Zurück zum Zitat Wang W, Xu Y, Gao R, Lu R, Han K, Wu G, et al. Detection of SARS-CoV-2 in different types of clinical specimens. JAMA. 2020;323:1843–4.PubMedCentralPubMed Wang W, Xu Y, Gao R, Lu R, Han K, Wu G, et al. Detection of SARS-CoV-2 in different types of clinical specimens. JAMA. 2020;323:1843–4.PubMedCentralPubMed
80.
Zurück zum Zitat Chu KH, Tsang WK, Tang CS, Lam MF, Lai FM, To KF, et al. Acute renal impairment in coronavirus-associated severe acute respiratory syndrome. Kidney Int. 2005;67:698–705.PubMedPubMedCentral Chu KH, Tsang WK, Tang CS, Lam MF, Lai FM, To KF, et al. Acute renal impairment in coronavirus-associated severe acute respiratory syndrome. Kidney Int. 2005;67:698–705.PubMedPubMedCentral
83.
Zurück zum Zitat Liu FL, Zou X, Fang W, Wu M, Li W, Zhang W et al . Highly ACE2 expression in pancreas may cause pancreas damage after SARS-CoV-2 infection. Preprint. medRxiv 2020. Posted online March 03, 2020. Liu FL, Zou X, Fang W, Wu M, Li W, Zhang W et al . Highly ACE2 expression in pancreas may cause pancreas damage after SARS-CoV-2 infection. Preprint. medRxiv 2020. Posted online March 03, 2020.
84.
Zurück zum Zitat Hadi A, Werge M, Kristiansen KT, et al. Coronavirus disease-19 (COVID-19) associated with severe acute pancreatitis: case report on three family members. Pancreatology. 2020;20:665–7.PubMed Hadi A, Werge M, Kristiansen KT, et al. Coronavirus disease-19 (COVID-19) associated with severe acute pancreatitis: case report on three family members. Pancreatology. 2020;20:665–7.PubMed
85.
Zurück zum Zitat Yang JK, Lin SS, Ji XJ, Guo LM. Binding of SARS coronavirus to its receptor damages islets and causes acute diabetes. Acta Diabetol. 2010;47:193–9.PubMed Yang JK, Lin SS, Ji XJ, Guo LM. Binding of SARS coronavirus to its receptor damages islets and causes acute diabetes. Acta Diabetol. 2010;47:193–9.PubMed
86.
Zurück zum Zitat Yang JK, Feng Y, Yuan MY, Yuan SY, Fu HJ, Wu BY, et al. Plasma glucose levels and diabetes are independent predictors for mortality and morbidity in patients with SARS. Diabet Med. 2006;23:623–8.PubMed Yang JK, Feng Y, Yuan MY, Yuan SY, Fu HJ, Wu BY, et al. Plasma glucose levels and diabetes are independent predictors for mortality and morbidity in patients with SARS. Diabet Med. 2006;23:623–8.PubMed
87.
Zurück zum Zitat Lee N, Hui D, Wu A, Chan P, Cameron P, Joynt GM, et al. A major outbreak of severe acute respiratory syndrome in Hong Kong. N Engl J Med. 2003;348:1986–94.PubMed Lee N, Hui D, Wu A, Chan P, Cameron P, Joynt GM, et al. A major outbreak of severe acute respiratory syndrome in Hong Kong. N Engl J Med. 2003;348:1986–94.PubMed
89.
Zurück zum Zitat Leung TW, Wong KS, Hui AC, To KF, Lai ST, Ng WF, et al. Myopathic changes associated with severe acute respiratory syndrome: a postmortem case series. Arch Neurol. 2005;62:1113–7.PubMed Leung TW, Wong KS, Hui AC, To KF, Lai ST, Ng WF, et al. Myopathic changes associated with severe acute respiratory syndrome: a postmortem case series. Arch Neurol. 2005;62:1113–7.PubMed
90.
Zurück zum Zitat Katsi V, Maragkoudakis S, Marketou M, Tsioufis C, Parthenakis F, Tousoulis D. The role of angiotensin-(1-7)/Mas axis and angiotensin type 2 receptors in the central nervous system in cardiovascular disease and therapeutics: a riddle to be solved. Curr Vasc Pharmacol. 2019;17:319–25.PubMed Katsi V, Maragkoudakis S, Marketou M, Tsioufis C, Parthenakis F, Tousoulis D. The role of angiotensin-(1-7)/Mas axis and angiotensin type 2 receptors in the central nervous system in cardiovascular disease and therapeutics: a riddle to be solved. Curr Vasc Pharmacol. 2019;17:319–25.PubMed
91.
Zurück zum Zitat Alenina N, Bader M. ACE2 in brain physiology and pathophysiology: evidence from transgenic animal models. Neurochem Res. 2019;44:1323–9.PubMed Alenina N, Bader M. ACE2 in brain physiology and pathophysiology: evidence from transgenic animal models. Neurochem Res. 2019;44:1323–9.PubMed
92.
Zurück zum Zitat Netland J, Meyerholz DK, Moore S, Cassell M, Perlman S. Severe acute respiratory syndrome coronavirus infection causes neuronal death in the absence of encephalitis in mice transgenic for human ACE2. J Virol. 2008;82:7264–75.PubMedPubMedCentral Netland J, Meyerholz DK, Moore S, Cassell M, Perlman S. Severe acute respiratory syndrome coronavirus infection causes neuronal death in the absence of encephalitis in mice transgenic for human ACE2. J Virol. 2008;82:7264–75.PubMedPubMedCentral
94.
Zurück zum Zitat Lechien JR, Chiesa-Estomba CM, De Siati DR, Horoi M, Le Bon SD, Rodriguez A, et al. Olfactory and gustatory dysfunctions as a clinical presentation of mild-to-moderate forms of the coronavirus disease (COVID-19): a multicenter European study. Eur Arch Otorhinolaryngol. 2020. https://doi.org/10.1007/s00405-020-05965-1. Lechien JR, Chiesa-Estomba CM, De Siati DR, Horoi M, Le Bon SD, Rodriguez A, et al. Olfactory and gustatory dysfunctions as a clinical presentation of mild-to-moderate forms of the coronavirus disease (COVID-19): a multicenter European study. Eur Arch Otorhinolaryngol. 2020. https://​doi.​org/​10.​1007/​s00405-020-05965-1.
95.
Zurück zum Zitat Xu J, Zhong S, Liu J, Li L, Li Y, Wu X, et al. Detection of severe acute respiratory syndrome coronavirus in the brain: potential role of the chemokine Mig in pathogenesis. Clin Infect Dis. 2005;41:1089–96.PubMed Xu J, Zhong S, Liu J, Li L, Li Y, Wu X, et al. Detection of severe acute respiratory syndrome coronavirus in the brain: potential role of the chemokine Mig in pathogenesis. Clin Infect Dis. 2005;41:1089–96.PubMed
98.
Zurück zum Zitat Zhang QL, Ding YQ, Hou JL, He L, Huang ZX, Wang HJ, et al. Detection of severe acute respiratory syndrome (SARS)-associated coronavirus RNA in autopsy tissues with in situ hybridization. Di Yi Jun Yi Da Xue Xue Bao. 2003;23(11):1125–7.PubMed Zhang QL, Ding YQ, Hou JL, He L, Huang ZX, Wang HJ, et al. Detection of severe acute respiratory syndrome (SARS)-associated coronavirus RNA in autopsy tissues with in situ hybridization. Di Yi Jun Yi Da Xue Xue Bao. 2003;23(11):1125–7.PubMed
99.
Zurück zum Zitat Lei C, Qian K, Li T, Zhang S, Fu W, Ding M, et al. Neutralization of SARS-CoV-2 spike pseudotyped virus by recombinant ACE2-Ig. Nat Commun. 2020;11(1):2070.PubMedPubMedCentral Lei C, Qian K, Li T, Zhang S, Fu W, Ding M, et al. Neutralization of SARS-CoV-2 spike pseudotyped virus by recombinant ACE2-Ig. Nat Commun. 2020;11(1):2070.PubMedPubMedCentral
100.
Zurück zum Zitat Tian X, Li C, Huang A, Xia S, Lu S, Shi Z, et al. Potent binding of 2019 novel coronavirus spike protein by a SARS coronavirus-specific human monoclonal antibody. Emerg Microbes Infect. 2020;9:382–5.PubMedPubMedCentral Tian X, Li C, Huang A, Xia S, Lu S, Shi Z, et al. Potent binding of 2019 novel coronavirus spike protein by a SARS coronavirus-specific human monoclonal antibody. Emerg Microbes Infect. 2020;9:382–5.PubMedPubMedCentral
101.
Zurück zum Zitat Aw S, Axmann M, Pfefferle S, Drosten C, Meyer B. A hexapeptide of the receptor-binding domain of SARS corona virus spike protein blocks viral entry into host cells via the human receptor ACE2. Antiviral Res. 2012;94:288–96. Aw S, Axmann M, Pfefferle S, Drosten C, Meyer B. A hexapeptide of the receptor-binding domain of SARS corona virus spike protein blocks viral entry into host cells via the human receptor ACE2. Antiviral Res. 2012;94:288–96.
102.
Zurück zum Zitat Ferrario CM, Jessup J, Chappell MC, Averill DB, Brosnihan KB, Tallant EA, et al. Effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockers on cardiac angiotensin-converting enzyme 2. Circulation. 2005;111:2605–10.PubMed Ferrario CM, Jessup J, Chappell MC, Averill DB, Brosnihan KB, Tallant EA, et al. Effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockers on cardiac angiotensin-converting enzyme 2. Circulation. 2005;111:2605–10.PubMed
104.
Zurück zum Zitat Yan Y, Liu Q, Li N, Du J, Li X, Li C, et al. Angiotensin II receptor blocker as a novel therapy in acute lung injury induced by avian influenza A H5N1 virus infection in mouse. Sci China Life Sci. 2015;58:208–11.PubMedPubMedCentral Yan Y, Liu Q, Li N, Du J, Li X, Li C, et al. Angiotensin II receptor blocker as a novel therapy in acute lung injury induced by avian influenza A H5N1 virus infection in mouse. Sci China Life Sci. 2015;58:208–11.PubMedPubMedCentral
105.
Zurück zum Zitat Mortensen EM, Nakashima B, Cornell J, Copeland LA, Pugh MJ, Anzueto A, et al. Population-based study of statins, angiotensin II receptor blockers, and angiotensin-converting enzyme inhibitors on pneumonia-related outcomes. Clin Infect Dis. 2012;55:1466–73.PubMedPubMedCentral Mortensen EM, Nakashima B, Cornell J, Copeland LA, Pugh MJ, Anzueto A, et al. Population-based study of statins, angiotensin II receptor blockers, and angiotensin-converting enzyme inhibitors on pneumonia-related outcomes. Clin Infect Dis. 2012;55:1466–73.PubMedPubMedCentral
107.
108.
Metadaten
Titel
Role of angiotensin-converting enzyme 2 (ACE2) in COVID-19
verfasst von
Wentao Ni
Xiuwen Yang
Deqing Yang
Jing Bao
Ran Li
Yongjiu Xiao
Chang Hou
Haibin Wang
Jie Liu
Donghong Yang
Yu Xu
Zhaolong Cao
Zhancheng Gao
Publikationsdatum
14.07.2020
Verlag
BioMed Central
Schlagwörter
COVID-19
SARS-CoV-2
Erschienen in
Critical Care / Ausgabe 1/2020
Elektronische ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-020-03120-0

Weitere Artikel der Ausgabe 1/2020

Critical Care 1/2020 Zur Ausgabe

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Hinter dieser Appendizitis steckte ein Erreger

23.04.2024 Appendizitis Nachrichten

Schmerzen im Unterbauch, aber sonst nicht viel, was auf eine Appendizitis hindeutete: Ein junger Mann hatte Glück, dass trotzdem eine Laparoskopie mit Appendektomie durchgeführt und der Wurmfortsatz histologisch untersucht wurde.

Ärztliche Empathie hilft gegen Rückenschmerzen

23.04.2024 Leitsymptom Rückenschmerzen Nachrichten

Personen mit chronischen Rückenschmerzen, die von einfühlsamen Ärzten und Ärztinnen betreut werden, berichten über weniger Beschwerden und eine bessere Lebensqualität.

Mehr Schaden als Nutzen durch präoperatives Aussetzen von GLP-1-Agonisten?

23.04.2024 Operationsvorbereitung Nachrichten

Derzeit wird empfohlen, eine Therapie mit GLP-1-Rezeptoragonisten präoperativ zu unterbrechen. Eine neue Studie nährt jedoch Zweifel an der Notwendigkeit der Maßnahme.

Update AINS

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