Skip to main content
American Journal of Physiology - Lung Cellular and Molecular Physiology logoLink to American Journal of Physiology - Lung Cellular and Molecular Physiology
. 2020 Aug 12;319(4):L596–L602. doi: 10.1152/ajplung.00286.2020

Role of the renin-angiotensin system in the development of severe COVID-19 in hypertensive patients

Rodrigo Pacheco Silva-Aguiar 1,*, Diogo Barros Peruchetti 1,*, Patricia Rieken Macedo Rocco 1,2,3, Alvin H Schmaier 4,5, Patrícia Machado Rodrigues e Silva 3,6, Marco Aurélio Martins 3,6, Vinícius Frias Carvalho 3,6, Ana Acacia Sá Pinheiro 1,3, Celso Caruso-Neves 1,2,3,
PMCID: PMC7516382  PMID: 32783619

Abstract

A new form of severe acute respiratory syndrome (SARS) caused by SARS-coronavirus 2 (CoV-2), called COVID-19, has become a global threat in 2020. The mortality rate from COVID-19 is high in hypertensive patients, making this association especially dangerous. There appears to be a consensus, despite the lack of experimental data, that angiotensin II (ANG II) is linked to the pathogenesis of COVID-19. This process may occur due to acquired deficiency of angiotensin-converting enzyme 2 (ACE2), resulting in reduced degradation of ANG II. Furthermore, ANG II has a critical role in the genesis and worsening of hypertension. In this context, the idea that there is a surge in the level of ANG II with COVID-19 infection, causing multiple organ injuries in hypertensive patients becomes attractive. However, the role of other components of the renin angiotensin system (RAS) in this scenario requires elucidation. The identification of other RAS components in COVID-19 hypertension may provide both diagnostic and therapeutic benefits. Here, we summarize the pathophysiologic contributions of different components of RAS in hypertension and their possible correlation with poor outcome observed in hypertensive patients with COVID-19.

Keywords: angiotensin II, COVID-19, hypertension, renin-angiotensin system, SARS-CoV-2

INTRODUCTION

In early December 2019, a new form of severe acute respiratory syndrome (SARS) caused by a new virus, SARS-coronavirus-2 (CoV-2), was characterized in Wuhan and called COVID-19 (82). In addition to previously identified SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV-2 is the third coronavirus identified to replicate in the human lower respiratory tract (44). SARS-CoV-2 has higher transmissibility rate and greater adaptability to different environments, making the spread of this virus more difficult to contain and a greater threat to humanity.

The role of the renin angiotensin system (RAS) in the pathogenesis of COVID-19 begins with the observation that angiotensin-converting enzyme 2 (ACE2) is the respiratory membrane receptor of SARS-CoV-2 (20). Further, ACE2 is considered to be the major enzyme degrading ANG II into angiotensin (1–7) (ANG-(1–7) (63, 64, 68). It is proposed that in patients infected with SARS-CoV-2, an acquired deficiency (i.e., inhibition) of ACE2 occurs, resulting in an increase in ANG II (5). However, these events probably do not occur in isolation and may be associated with additional modifications in renin-angiotensin system (RAS) components, contributing to the overall pathophysiology of COVID-19 infection. In the present review, we highlight the possible participation of different components of RAS as an integrating system connecting undesirable worsening of COVID-19 with hypertension.

HYPERTENSION IS A RISK FACTOR FOR THE DEVELOPMENT OF SEVERE COVID-19

Epidemiologic data have demonstrated that hypertension is a pivotal comorbidity related to increased susceptibility to the severe form of COVID-19. In a follow-up study of 168 patients in China, Xie et al. (74) showed that hypertension was the most common comorbidity associated with COVID-19 mortality. In a meta-analysis, Zhao et al. (81) compiled data on ~56,000 patients affected by COVID-19. Hypertension was present in ~19% of all patients hospitalized and was correlated with a higher risk of COVID-19-related mortality. A meta-analysis of several scientific databases report showed that hypertensive patients have up to 2.5-fold higher risk of developing severe or fatal COVID-19 (30). Since RAS is the central biochemical pathway involved in hypertension (41, 54), it remains unclear to what extent this system contributes to the undesirable worsening of COVID-19 in hypertensive patients.

ANGIOTENSIN II AND CORONAVIRUS INFECTION: IS THERE A CAUSAL RELATIONSHIP?

There are two different types of RAS, systemic and tissue, that are activated by distinct signals and have different roles under physiologic and pathophysiologic conditions (41, 54). The production of ANG II in pulmonary vessels accounts for most of the circulating peptide. On the other hand, the synthesis of ANG II in extra-pulmonary tissues works to amplify its systemic level (4). Liu et al. (33) showed the level of ANG II in plasma was increased in patients with COVID-19 and was associated with lung injury. Based on the current knowledge obtained from other respiratory viruses, it is possible to propose a causative relationship among RAS, hypertension, and COVID-19.

Role of Plasma Renin Activity

Plasma renin activity (PRA) is used to predict the prognosis of hypertensive patients and determining the therapeutic approach. Essential hypertension can be stratified into two categories based on PRA: those with low PRA and those with normal-to-high PRA. Genest et al. (15) analyzed PRA and its correlation with cardiovascular complications in different types of hypertension, including essential and secondary hypertension. They observed that patients with higher PRA are at higher risk for developing severe cardiovascular complications. In line with this idea, Verma et al. (67), in the Heart Outcomes Prevention Evaluation (HOPE) study, observed that high PRA is an independent predictor for vascular damage and mortality in patients with atherosclerosis and diabetes. Based on these observations, we believe that determination of PRA in both hypertensive and nonhypertensive patients with COVID-19 would be a useful marker to indicate those with a worse prognosis to guide treatment decisions for the best therapeutic approach.

Role of ACE and ACE2

An interrelationship between ANG II level and ACE2 activity in SARS-CoV infection was initially proposed by Kuba et al. (28) using a murine model. They showed that the level of ANG II in the lung was increased after intravenous infusion of S spike protein from SARS-CoV, which correlated with inhibition of ACE2 activity and induction of lung injury. Sriram and Insel (60) proposed that inhibition of ACE2 by SARS-CoV-2 binding leads to imbalance in the action of ACE- and ACE2-derived peptides, ANG II and ANG-(1–7), respectively, and could be involved in the pathogenesis of COVID-19. In addition, the possible specific positive modulation of ACE activity mainly in hypertensive patients stricken by COVID-19 should be taken as an attractive possibility.

Deletion of ACE2 worsened lung injury in a murine model of H7N9 infection (79). Treatment with recombinant human ACE2 (rhACE2) improved acute lung injury in mice induced by acid aspiration or sepsis (23). Furthermore, a human soluble ACE2 binds to SARS-CoV-2 and inhibits the infection rate to 1,000–5,000 times in engineered organoids of human blood vessels and human kidney (39). Plasma ANG II level is increased in patients infected with H7N9 and H5N1, followed by a decrease in ACE2 activity, with no change in ACE activity (21, 84). Further, ACE activity increased and ACE2 decreased in the bronchoalveolar lavage fluid of animals and patients with acute respiratory distress syndrome (ARDS) (52, 70, 71). Thus, it can be hypothesized that, with a decrease in ACE2 followed by SARS-CoV-2 infection, ANG II would increase.

This idea is reinforced by the observation that influenza A H5N1 decreases ACE2 lung expression and enhances plasma ANG II levels in mice (84). Although, it is worth mentioning that unlike the SARS-CoV-2 virus, H5N1 infection is not mediated by ACE2, but RAS is involved in the pathogenesis of the ARDS as well. In addition, plasma ANG II levels is associated with disease severity in H7N9-infected patients (21). These observations indicate that RAS dysregulation could be a general mechanism associated with ARDS caused by different etiologies, including COVID-19, even with important differences among pathogenesis of distinct viral infections.

The interrelationship of ACE and ACE2 in the pathogenesis of essential hypertension has been proposed previously (22, 49). ACE2 has a protective role against the development of hypertension because it breaks down ANG II (54, 63), producing ANG-(1–7), which has cardioprotective effects (26). Thus, not only an increase in ANG II but also the loss of ANG-(1–7) could be involved in the development of hypertension. Although ACE expression is unaltered or increased in hypertensive animals, hypertension progression is associated with decreased ACE2 expression in the kidney and heart in hypertensive patients and animal models (11, 27, 45, 62).

Pharmacologic approaches aimed to increase the ACE2/ANG-(1–7) axis reduce blood pressure in spontaneously hypertensive rats (SHR) (11, 34) and attenuate cardiac dysfunction in ANG II-induced hypertension (83). An autopsy study of 20 patients diagnosed with hypertensive nephropathy or cardiomyopathy revealed a decrease in ACE2 expression in kidney, whereas ACE expression was increased in the kidney and heart (27). Little is known regarding the expression of ACE and ACE2 in the lung of hypertensive patients or animals.

The expression of ACE2 in specific tissues is also modulated by its shedding mediated by TNF-α converting enzyme (TACE)/ADAM-17, which is increased by ANG II (25, 43). TACE/ADAM-17 is involved in cardiovascular hypertrophy and perivascular fibrosis in a model of hypertension induced by ANG II in C57BL/6 mice (61). These findings indicate positive feedback between ACE2 inhibition and increased cardiac ANG II levels. Further, plasma ACE2 activity is an independent predictor of major adverse events in cardiac obstructive coronary disease and correlates positively with systolic blood pressure in patients with diabetes (47, 59).

TACE/ADAM-17 knockdown decreased SARS-CoV infection in human embryonic kidney 293T (HEK293T) cells (19). Based on the similarities between SARS-CoV and SARS-CoV-2 infection, we may speculate that ANG II increases entry of the virus into the target cell despite the lower expression of ACE2 at the cell membrane. This assessment is supported by the clinical observation that the severity of COVID-19 is not necessarily correlated to a high viral load (32), suggesting a pivotal role of the host response to the initial viral replication and the environment. In agreement, Xie et al. (75) showed age-related loss of ACE2 in the lungs. Despite this, increased mortality and worsened phenotype has been observed in elderly patients with COVID-19 (30).

Together, the findings suggest that ACE2 expression in the lung might contribute to increased infection rate of SARS-CoV-2. This process is followed by a decrease in ACE2 expression, leading to an increase in ANG II levels. On the other hand, the decreased ACE2 expression in the kidney and heart in hypertensive patients, could contribute to a further increase in plasma and/or tissue ANG II, a response possibly associated with an undesirable outcome in patients stricken by COVID-19. In addition, activation of TACE/ADAM-17 could be involved in the exacerbation of inflammation noted in COVID-19, due to increased shedding of pro-TNF-α and consequently increased concentrations of soluble TNF-α in the plasma.

Beyond ACE and ACE2: Role of Prolylcarboxypeptidase and Prolyl Oligopeptidase

Other proteases such as prolylcarboxypeptidase (PRCP) and prolyl oligopeptidase (POP), also called prolyl endopeptidase, may contribute to the surge in the level of ANG II in COVID-19 because they also cleave after Pro-X COOH-terminal peptide bonds like those in ANG II (52, 57). These serine carboxypeptidases have broad expression among the primary target tissues affected by COVID-19 and hypertension (12, 40). It is well established that these peptidases are present in cell membranes and lysosomes (58). PRCP has been called lysosomal carboxypeptidase because it was first discovered in the lysosomal fraction of kidney. It has been shown to be widely distributed by cell membranes (1, 55). It is also prominent in kidney and the hypothalamus in the brain (1, 69). POP function is better understood in the brain, where it correlates with inflammation in neurodegenerative diseases, but its role in the development of hypertension is still poorly defined (3). This enzyme is also highly expressed in lung, kidney, liver, and spleen, all of which are strongly associated with the pathogenesis of COVID-19 (12, 40). Recently, Serfozo et al. (53) proposed that POP is the main enzyme responsible for the conversion of ANG II into Ang-(1–7) in the systemic circulation, whereas ACE2 is more prominent in the lung and kidney, suggesting the potential involvement of POP as a determinant of the ANG II level in tissues infected by SARS-CoV-2. Nevertheless, its role in hypertension and its correlation with the severe form of COVID-19 have not yet been determined.

PRCP polymorphism (E112D) is associated with hypertension, and its expression is reduced in renovascular hypertensive rats (80). Recently, a novel polymorphism (rs12290550) was found to be associated with essential hypertension in a discrete Chinese ethnic group (72). PRCP gene trap mice (PRCPgt/gt) constitutively have a small, but significant, increase in blood pressure. Their vessels express increased reactive oxygen species (ROS), and cardiac hypertrophy and renal glomerular tubulization are observed. These animals also have a constitutive higher risk of induced arterial thrombosis (1, 36). Furthermore, Marangoni et al. (37) proposed that reduction in PRCP expression is associated with the SHR phenotype, a response not reversed by treatment of SHRs with the angiotensin type 1 receptor blocker (ARB) losartan. This finding is an indication that modification in PRCP expression seems to be a cause rather than a consequence of an increase in the tissue level of ANG II.

In kidney, ACE2 is the dominant ANG II-cleaving peptidase. However, in ace2−/− kidney, ANG-(1–7) is still produced by PRCP (18). At low pH in the kidney, however, PRCP is the dominant ANG II-degrading enzyme (18, 53). Because PRCP is expressed in the luminal membrane of renal proximal tubule, and this segment has a great capability to secrete H+, it is plausible to postulate that acidic microdomains, such as the space between microvilli of the brush border, can strongly favor the activity of this enzyme. Furthermore, PRCP cleaves ANG II in the distal nephron segments, where the luminal pH is acidic (36). However, the significance of these observations in the development of hypertension and susceptibility to severe COVID-19 is still an open matter.

One crucial clue comes from the observation that PRCP has an important effect on thrombosis and endothelial function (1). A reduction in PRCP levels (prcpgt/gt mice) is associated with a prothrombotic state and increase in ROS, which, in turn, leads to endothelial dysfunction and loss of anticoagulant properties. Curiously, these phenomena are similar to that observed in patients with severe COVID-19 (16). It is possible that hypertensive patients may have a decrease in the expression of PRCP in endothelial and renal cells, which make them more susceptible to severe COVID-19. In addition to this pathway, PRCP could contribute to COVID-19 pneumonia because it is a plasma prekallikrein (PK) activator to form plasma kallikrein (PKa) (51, 55, 73). The PKa formed cleaves high molecular weight kininogen to liberate bradykinin, which alone or when degraded by other carboxypeptidases to form des-Arg-BK, binds to bradykinin B2 and B1 receptors to stimulate the local edema seen in COVID-19 pneumonia. This topic is discussed further in other studies (65, 66).

Finally, there is a third possible mechanism whereby PRCP can contribute to hypertension. PRCP in the hypothalamus degrades α-melanocyte-stimulating hormone (α-MSH) so that it cannot stimulate the melanocortin 4 receptor to induce satiety and increase blood pressure (17, 69). In PRCP deficiency states, there is less degraded α-MSH binding to the melanocortin 4 receptor, leading to thin, hypertensive individuals (1, 69).

AT1R Mediating ANG II Effects During COVID-19 Progression

ANG II exerts its main effects through the angiotensin II type 1 receptor (AT1R), which belongs to the G protein-coupled receptor family and is a target for specific blockers (angiotensin type 1 receptor blockers, ARBs) in several diseases (14). The AT1R-mediated effects of ANG II include vasoconstriction, inflammation, and fibrosis (42). Several studies indicate a potential role of the ANG II/AT1R pathway in tissue damage caused by infections such as H7N9 and H5N1, as well as SARS-CoV and SARS-CoV-2 (21, 78, 84). Consistent with this interpretation, Kuba et al. (28), also showed that lung injury caused by SARS-CoV S protein infection in a murine model is attenuated by the ARB losartan.

There is consensus that an enhanced circulating level of IL-6 is a major hallmark of the profound inflammatory state seen in patients with COVID-19 associated with undesirable outcome (31). In addition, the use of cytokine-modulatory therapies, especially anti-IL-6 agents, for critically ill patients with COVID-19 offer a perspective to its treatment (2). In agreement, a phase 3 clinical trial is currently being performed (35). Nevertheless, the cost of and access to anti-IL-6 is a concern, especially as the numbers of cases worldwide continue to climb. On the other hand, Meng et al. (38) demonstrated that in hypertensive patients with COVID-19 treated with an ACE inhibitor/ARB, the levels of IL-6 decreased in peripheral plasma and was associated with a better outcome. Therefore, these drugs have potential to be a successful strategy for the treatment of COVID-19 provided the cost is not prohibitive. Furthermore, the use of an ARB could be beneficial for COVID-19 not only for inhibiting the proinflammatory actions of the ACE/ANG II/AT1R axis but also for inducing the anti-inflammatory actions of the ACE2/ANG (1–7)/MASR axis, which increases IL-10 levels (24). In addition, the use of an ACE inhibitor could be useful to the increase the level of ANG-(1–7) because it is well known that ACE cleaves ANG-(1–7) into ANG-(1–5) (48). In agreement, two trials on patients with COVID-19 are ongoing, one evaluating the therapeutic effect of rhACE2 (NCT04335136) and the other verifying the effect of intravenous ANG-(1–7) (NCT04332666).

The ANG II/AT1R pathway appears to be involved in the proinflammatory response in hypertensive patients and animal models (9, 29, 50). In essential hypertensive patients, AT1R gene expression is increased in leukocytes, including T cells (7, 8). Increased AT1R mRNA expression was detected in isolated adipocytes obtained from obese hypertensive patients compared with obese patients who were not hypertensive (13). Finally, AT1R (A1166C) gene polymorphism correlates with essential hypertension (6). Based on these findings, it is possible that a change in the expression or sensitivity of AT1R to the ANG II response in hypertensive patients may correlate with worse outcome in patients with COVID-19. How does ANG II increase COVID-19 susceptibility to a severe outcome?

Preexposure of ANG II is known to sensitize different tissues to a hyperresponsiveness to further ANG II exposure. In mice, preexposure to a low pressor dose of ANG II leads to an exacerbated hypertensive response to additional exposition to ANG II (77). This effect was correlated to the increase in mRNA expression of ANG II receptors, renin, angiotensinogen, and the increase in ACE/ACE2 ratio in the brain. RAS hyperresponsiveness is similarly sensitized by a high-fat diet (76). The authors showed that a high-fat diet upregulated RAS components in the brain. Furthermore, it has been shown that ANG II, through AT1R, induces a proinflammatory, hyper-responsive phenotype, including in infectious diseases such as malaria (56, 57, 85).

These observations are in accordance with data showing that patients with COVID-19 display lung T-cell infiltrate, with positive markers of exhaustion, likely caused by a hyperinflammatory stimulus (10). In addition, heterodimers between AT1R with bradykinin receptors, AT2R, or Mas may also alter ANG II binding to AT1R and subsequent signaling, as has been reported in preeclampsia (46). In this way, tissue ANG II could be an additional link between hypertension and severe COVID-19, functioning as an initial sensitizer, shifting the system toward a harmful phenotype, and thus contributing to the worsening of the disease noted in patients with COVID-19.

SUMMARY AND PERSPECTIVES

In conclusion, we postulate that there are several possible interaction points between hypertension and COVID-19, forming a dangerous cascade of events that could be responsible for the poor outcome of hypertensive patients with COVID-19 (Fig. 1). In this review, we have described at least three possible points of interaction between hypertension and COVID-19: 1) degradation of ANG II by different carboxypeptidases; 2) synthesis of ANG II; and 3) hypersensitization and/or increase in the expression of AT1R. All of them are associated with the ACE2/ANG-(1–7)/MASR pathway. The interaction of these pathways may form an amplifying loop able to upregulate ANG II levels to explain why hypertensive patients are more susceptible to the severe form of COVID-19. In addition, when hypertensive patients are stricken by COVID-19, there could be stimulation of the cellular pathways triggered by a presensitized AT1R, leading to a more intense proinflammatory response and tissue injury.

Fig. 1.

Fig. 1.

Proposed model to explain why hypertensive patients with coronavirus disease 2019 (COVID-19) progress to an undesirable outcome. In hypertensive patients with COVID-19, we propose that there is a further increase in the level of ANG II as well as higher responsivity of angiotensin II type 1 receptor (AT1R) to ANG II. The increase in the level of ANG II could be due to 1) a decrease in ANG II degradation by angiotensin-converting enzyme 2 (ACE2), prolylcarboxypeptidase (PRCP), and prolyl oligopeptidase (POP). ANG II could also decrease tissue ACE2 expression by increasing TNF-α converting enzyme (TACE)/ADAM17-mediated shedding, creating a positive feedback; 2) an increase in ANG II synthesis caused by an increase in renin and ACE activities. In addition to the increase in the level of ANG II, we also propose that the increase in expression and/or hyperresponsiveness of AT1R in hypertensive patients could potentiate the effect of ANG II on different tissues affected by COVID-19. In this context, the inhibition of AT1R could be a primary or coadjutant therapy for the treatment of patients with COVID-19. In agreement, a clinical trial of treatment with losartan is under way (NCT04328012). Red outline denotes reduced hypertension; blue outline denotes increased hypertension; dashed red outline denotes possible reduction in hypertension; dashed blue square denotes possibly increase in hypertension. *Possible therapeutic target; #possible biomarker.

GRANTS

This work was supported by grants from the following Brazilian agencies: Conselho Nacional de Desenvolvimento Científico e Tecnológico (https://www.cnpq.br): 304682/2015-2 (to A.A.S.P.), 303793/2015-5 (to C.C.-N.), Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro–FAPERJ (https://www.faperj.br): E-26/202.950/2016 (to A.A.S.P.), E-26/202.833/2017 (to C.C.-N.), and Rio Network of Innovation in nanosystems for the health (Nanohealth/FAPERJ) - E-26/010.000983/2019 (to A.A.S.P. and C.C.-N.).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

R.P.S.-A., D.B.P., and C.C.-N. prepared figure; R.P.S.-A, D.B.P. and C.C.-N. drafted manuscript; R.P.S.-A, D.B.P., P.R.M.R., A.H.S., P.M.R.S., M.A.M., V.F.C., A.A.S.P., and C.C.-N. edited and revised manuscript; and R.P.S.-A., D.B.P., P.R.M.R., A.H.S., P.M.R.S., M.A.M., V.F.C., A.A.S.P., and C.C.-N. approved final version of manuscript.

ACKNOWLEDGMENTS

The authors thank Mario Luiz da Silva Bandeira and Douglas Esteves Teixeira (FAPERJ TCT fellowships) for excellent technical support.

REFERENCES

  • 1.Adams GN, LaRusch GA, Stavrou E, Zhou Y, Nieman MT, Jacobs GH, Cui Y, Lu Y, Jain MK, Mahdi F, Shariat-Madar Z, Okada Y, D’Alecy LG, Schmaier AH. Murine prolylcarboxypeptidase depletion induces vascular dysfunction with hypertension and faster arterial thrombosis. Blood 117: 3929–3937, 2011. doi: 10.1182/blood-2010-11-318527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Arnaldez FI, O’Day SJ, Drake CG, Fox BA, Fu B, Urba WJ, Montesarchio V, Weber JS, Wei H, Wigginton JM, Ascierto PA. The Society for Immunotherapy of Cancer perspective on regulation of interleukin-6 signaling in COVID-19-related systemic inflammatory response. J Immunother Cancer 8: e000930, 2020. doi: 10.1136/jitc-2020-000930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Babkova K, Korabecny J, Soukup O, Nepovimova E, Jun D, Kuca K. Prolyl oligopeptidase and its role in the organism: attention to the most promising and clinically relevant inhibitors. Future Med Chem 9: 1015–1038, 2017. doi: 10.4155/fmc-2017-0030. [DOI] [PubMed] [Google Scholar]
  • 4.Benigni A, Cassis P, Remuzzi G. Angiotensin II revisited: new roles in inflammation, immunology and aging. EMBO Mol Med 2: 247–257, 2010. doi: 10.1002/emmm.201000080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Busse LW, Chow JH, McCurdy MT, Khanna AK. COVID-19 and the RAAS-a potential role for angiotensin II? Crit Care 24: 136, 2020. doi: 10.1186/s13054-020-02862-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Chandra S, Narang R, Sreenivas V, Bhatia J, Saluja D, Srivastava K. Association of angiotensin II type 1 receptor (A1166C) gene polymorphism and its increased expression in essential hypertension: a case-control study. PLoS One 9: e101502, 2014. doi: 10.1371/journal.pone.0101502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chon H, Gaillard CA, van der Meijden BB, Dijstelbloem HM, Kraaijenhagen RJ, van Leenen D, Holstege FC, Joles JA, Bluyssen HA, Koomans HA, Braam B. Broadly altered gene expression in blood leukocytes in essential hypertension is absent during treatment. Hypertension 43: 947–951, 2004. doi: 10.1161/01.HYP.0000123071.35142.72. [DOI] [PubMed] [Google Scholar]
  • 8.Coppo M, Bandinelli M, Berni A, Galastri S, Abbate R, Poggesi L, Marra F, Gensini GF, Boddi M. Ang II upregulation of the T-lymphocyte renin-angiotensin system is amplified by low-grade inflammation in human hypertension. Am J Hypertens 24: 716–723, 2011. doi: 10.1038/ajh.2011.32. [DOI] [PubMed] [Google Scholar]
  • 9.Dandona P, Dhindsa S, Ghanim H, Chaudhuri A. Angiotensin II and inflammation: the effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockade. J Hum Hypertens 21: 20–27, 2007. doi: 10.1038/sj.jhh.1002101. [DOI] [PubMed] [Google Scholar]
  • 10.Diao B, Wang C, Tan Y, Chen X, Liu Y, Ning L, Chen L, Li M, Liu Y, Wang G, Yuan Z, Feng Z, Zhang Y, Wu Y, Chen Y. Reduction and functional exhaustion of T cells in patients with coronavirus disease 2019 (COVID-19). Front Immunol 11: 827, 2020. doi: 10.3389/fimmu.2020.00827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Díez-Freire C, Vázquez J, Correa de Adjounian MF, Ferrari MF, Yuan L, Silver X, Torres R, Raizada MK. ACE2 gene transfer attenuates hypertension-linked pathophysiological changes in the SHR. Physiol Genomics 27: 12–19, 2006. doi: 10.1152/physiolgenomics.00312.2005. [DOI] [PubMed] [Google Scholar]
  • 12.Fagerberg L, Hallström BM, Oksvold P, Kampf C, Djureinovic D, Odeberg J, Habuka M, Tahmasebpoor S, Danielsson A, Edlund K, Asplund A, Sjöstedt E, Lundberg E, Szigyarto CA, Skogs M, Takanen JO, Berling H, Tegel H, Mulder J, Nilsson P, Schwenk JM, Lindskog C, Danielsson F, Mardinoglu A, Sivertsson A, von Feilitzen K, Forsberg M, Zwahlen M, Olsson I, Navani S, Huss M, Nielsen J, Ponten F, Uhlén M. Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol Cell Proteomics 13: 397–406, 2014. doi: 10.1074/mcp.M113.035600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Faloia E, Gatti C, Camilloni MA, Mariniello B, Sardu C, Garrapa GG, Mantero F, Giacchetti G. Comparison of circulating and local adipose tissue renin-angiotensin system in normotensive and hypertensive obese subjects. J Endocrinol Invest 25: 309–314, 2002. doi: 10.1007/BF03344010. [DOI] [PubMed] [Google Scholar]
  • 14.Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II signal transduction: an update on mechanisms of physiology and pathophysiology. Physiol Rev 98: 1627–1738, 2018. doi: 10.1152/physrev.00038.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Genest J, Boucher R, Kuchel O, Nowaczynski W. Renin in hypertension: how important as a risk factor? Can Med Assoc J 109: 475–478, 1973. [PMC free article] [PubMed] [Google Scholar]
  • 16.Giannis D, Ziogas IA, Gianni P. Coagulation disorders in coronavirus infected patients: COVID-19, SARS-CoV-1, MERS-CoV and lessons from the past. J Clin Virol 127: 104362, 2020. doi: 10.1016/j.jcv.2020.104362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Greenfield JR, Miller JW, Keogh JM, Henning E, Satterwhite JH, Cameron GS, Astruc B, Mayer JP, Brage S, See TC, Lomas DJ, O’Rahilly S, Farooqi IS. Modulation of blood pressure by central melanocortinergic pathways. N Engl J Med 360: 44–52, 2009. doi: 10.1056/NEJMoa0803085. [DOI] [PubMed] [Google Scholar]
  • 18.Grobe N, Weir NM, Leiva O, Ong FS, Bernstein KE, Schmaier AH, Morris M, Elased KM. Identification of prolyl carboxypeptidase as an alternative enzyme for processing of renal angiotensin II using mass spectrometry. Am J Physiol Cell Physiol 304: C945–C953, 2013. doi: 10.1152/ajpcell.00346.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Haga S, Yamamoto N, Nakai-Murakami C, Osawa Y, Tokunaga K, Sata T, Yamamoto N, Sasazuki T, Ishizaka Y. Modulation of TNF-α-converting enzyme by the spike protein of SARS-CoV and ACE2 induces TNF-α production and facilitates viral entry. Proc Natl Acad Sci USA 105: 7809–7814, 2008. doi: 10.1073/pnas.0711241105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, Müller MA, Drosten C, Pöhlmann S. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 181: 271–280.e8, 2020. doi: 10.1016/j.cell.2020.02.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Huang F, Guo J, Zou Z, Liu J, Cao B, Zhang S, Li H, Wang W, Sheng M, Liu S, Pan J, Bao C, Zeng M, Xiao H, Qian G, Hu X, Chen Y, Chen Y, Zhao Y, Liu Q, Zhou H, Zhu J, Gao H, Yang S, Liu X, Zheng S, Yang J, Diao H, Cao H, Wu Y, Zhao M, Tan S, Guo D, Zhao X, Ye Y, Wu W, Xu Y, Penninger JM, Li D, Gao GF, Jiang C, Li L. Angiotensin II plasma levels are linked to disease severity and predict fatal outcomes in H7N9-infected patients. Nat Commun 5: 3595, 2014. doi: 10.1038/ncomms4595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ibrahim MM. RAS inhibition in hypertension. J Hum Hypertens 20: 101–108, 2006. doi: 10.1038/sj.jhh.1001960. [DOI] [PubMed] [Google Scholar]
  • 23.Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, Yang P, Sarao R, Wada T, Leong-Poi H, Crackower MA, Fukamizu A, Hui CC, Hein L, Uhlig S, Slutsky AS, Jiang C, Penninger JM. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature 436: 112–116, 2005. doi: 10.1038/nature03712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Jia H. Pulmonary angiotensin-converting enzyme 2 (ACE2) and inflammatory lung disease. Shock 46: 239–248, 2016. doi: 10.1097/SHK.0000000000000633. [DOI] [PubMed] [Google Scholar]
  • 25.Jia HP, Look DC, Tan P, Shi L, Hickey M, Gakhar L, Chappell MC, Wohlford-Lenane C, McCray PB Jr. Ectodomain shedding of angiotensin converting enzyme 2 in human airway epithelia. Am J Physiol Lung Cell Mol Physiol 297: L84–L96, 2009. doi: 10.1152/ajplung.00071.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, Liu FF, Zhang K, Zhang C. Angiotensin-converting enzyme 2 and angiotensin 1-7: novel therapeutic targets. Nat Rev Cardiol 11: 413–426, 2014. doi: 10.1038/nrcardio.2014.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Koka V, Huang XR, Chung AC, Wang W, Truong LD, Lan HY. Angiotensin II up-regulates angiotensin I-converting enzyme (ACE), but down-regulates ACE2 via the AT1-ERK/p38 MAP kinase pathway. Am J Pathol 172: 1174–1183, 2008. doi: 10.2353/ajpath.2008.070762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kuba K, Imai Y, Rao S, Gao H, Guo F, Guan B, Huan Y, Yang P, Zhang Y, Deng W, Bao L, Zhang B, Liu G, Wang Z, Chappell M, Liu Y, Zheng D, Leibbrandt A, Wada T, Slutsky AS, Liu D, Qin C, Jiang C, Penninger JM. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat Med 11: 875–879, 2005. doi: 10.1038/nm1267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Landgraf SS, Wengert M, Silva JS, Zapata-Sudo G, Sudo RT, Takiya CM, Pinheiro AA, Caruso-Neves C. Changes in angiotensin receptors expression play a pivotal role in the renal damage observed in spontaneously hypertensive rats. Am J Physiol Renal Physiol 300: F499–F510, 2011. doi: 10.1152/ajprenal.00384.2010. [DOI] [PubMed] [Google Scholar]
  • 30.Lippi G, Plebani M. Laboratory abnormalities in patients with COVID-2019 infection. Clin Chem Lab Med 58: 1131–1134, 2020. doi: 10.1515/cclm-2020-0198. [DOI] [PubMed] [Google Scholar]
  • 31.Liu Y, Huang F, Xu J, Yang P, Qin Y, Cao M, Wang Z, Li X, Zhang S, Ye L, Lv J, Wei J, Xie T, Gao H, Xu KF, Wang F, Liu L, Jiang C. Anti-hypertensive Angiotensin II receptor blockers associated to mitigation of disease severity in elderly COVID-19 patients (Preprint). medRxiv 2020. doi: 10.1101/2020.03.20.20039586. [DOI]
  • 32.Liu Y, Yan LM, Wan L, Xiang TX, Le A, Liu JM, Peiris M, Poon LLM, Zhang W. Viral dynamics in mild and severe cases of COVID-19. Lancet Infect Dis 20: 656–657, 2020. doi: 10.1016/S1473-3099(20)30232-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Liu Y, Yang Y, Zhang C, Huang F, Wang F, Yuan J, Wang Z, Li J, Li J, Feng C, Zhang Z, Wang L, Peng L, Chen L, Qin Y, Zhao D, Tan S, Yin L, Xu J, Zhou C, Jiang C, Liu L. Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury. Sci China Life Sci 63: 364–374, 2020. doi: 10.1007/s11427-020-1643-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Lo J, Patel VB, Wang Z, Levasseur J, Kaufman S, Penninger JM, Oudit GY. Angiotensin-converting enzyme 2 antagonizes angiotensin II-induced pressor response and NADPH oxidase activation in Wistar-Kyoto rats and spontaneously hypertensive rats. Exp Physiol 98: 109–122, 2013. doi: 10.1113/expphysiol.2012.067165. [DOI] [PubMed] [Google Scholar]
  • 35.Maes B, Bosteels C, De Leeuw E, Declercq J, Van Damme K, Delporte A, Demeyere B, Vermeersch S, Vuylsteke M, Willaert J, Bollé L, Vanbiervliet Y, Decuypere J, Libeer F, Vandecasteele S, Peene I, Lambrecht BN. Correction to: Treatment of severely ill COVID-19 patients with anti-interleukin drugs (COV-AID): A structured summary of a study protocol for a randomised controlled trial. Trials 21: 556, 2020. doi: 10.1186/s13063-020-04519-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Maier C, Schadock I, Haber PK, Wysocki J, Ye M, Kanwar Y, Flask CA, Yu X, Hoit BD, Adams GN, Schmaier AH, Bader M, Batlle D. Prolylcarboxypeptidase deficiency is associated with increased blood pressure, glomerular lesions, and cardiac dysfunction independent of altered circulating and cardiac angiotensin II. J Mol Med (Berl) 95: 473–486, 2017. doi: 10.1007/s00109-017-1513-9. [DOI] [PubMed] [Google Scholar]
  • 37.Marangoni RA, Santos RA, Piccolo C. Deficient prolylcarboxypeptidase gene and protein expression in left ventricles of spontaneously hypertensive rats (SHR). Peptides 61: 69–74, 2014. doi: 10.1016/j.peptides.2014.08.016. [DOI] [PubMed] [Google Scholar]
  • 38.Meng J, Xiao G, Zhang J, He X, Ou M, Bi J, Yang R, Di W, Wang Z, Li Z, Gao H, Liu L, Zhang G. Renin-angiotensin system inhibitors improve the clinical outcomes of COVID-19 patients with hypertension. Emerg Microbes Infect 9: 757–760, 2020. doi: 10.1080/22221751.2020.1746200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Monteil V, Kwon H, Prado P, Hagelkrüys A, Wimmer RA, Stahl M, Leopoldi A, Garreta E, Hurtado Del Pozo C, Prosper F, Romero JP, Wirnsberger G, Zhang H, Slutsky AS, Conder R, Montserrat N, Mirazimi A, Penninger JM. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell 181: 905–913.e7, 2020. doi: 10.1016/j.cell.2020.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Myöhänen TT, Pyykkö E, Männistö PT, Carpen O. Distribution of prolyl oligopeptidase in human peripheral tissues and in ovarian and colorectal tumors. J Histochem Cytochem 60: 706–715, 2012. doi: 10.1369/0022155412453051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Nehme A, Zouein FA, Zayeri ZD, Zibara K. An update on the tissue renin angiotensin system and its role in physiology and pathology. J Cardiovasc Dev Dis 6: 14, 2019. doi: 10.3390/jcdd6020014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Nishimura H, Tsuji H, Masuda H, Nakagawa K, Nakahara Y, Kitamura H, Kasahara T, Sugano T, Yoshizumi M, Sawada S, Nakagawa M. Angiotensin II increases plasminogen activator inhibitor-1 and tissue factor mRNA expression without changing that of tissue type plasminogen activator or tissue factor pathway inhibitor in cultured rat aortic endothelial cells. Thromb Haemost 77: 1189–1195, 1997. doi: 10.1055/s-0038-1656136. [DOI] [PubMed] [Google Scholar]
  • 43.Patel VB, Clarke N, Wang Z, Fan D, Parajuli N, Basu R, Putko B, Kassiri Z, Turner AJ, Oudit GY. Angiotensin II induced proteolytic cleavage of myocardial ACE2 is mediated by TACE/ADAM-17: a positive feedback mechanism in the RAS. J Mol Cell Cardiol 66: 167–176, 2014. doi: 10.1016/j.yjmcc.2013.11.017. [DOI] [PubMed] [Google Scholar]
  • 44.Peeri NC, Shrestha N, Rahman MS, Zaki R, Tan Z, Bibi S, Baghbanzadeh M, Aghamohammadi N, Zhang W, Haque U. The SARS, MERS and novel coronavirus (COVID-19) epidemics, the newest and biggest global health threats: what lessons have we learned? Int J Epidemiol 49: 717–726, 2020. doi: 10.1093/ije/dyaa033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Prieto MC, González-Villalobos RA, Botros FT, Martin VL, Pagán J, Satou R, Lara LS, Feng Y, Fernandes FB, Kobori H, Casarini DE, Navar LG. Reciprocal changes in renal ACE/ANG II and ACE2/ANG 1-7 are associated with enhanced collecting duct renin in Goldblatt hypertensive rats. Am J Physiol Renal Physiol 300: F749–F755, 2011. doi: 10.1152/ajprenal.00383.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Quitterer U, Fu X, Pohl A, Bayoumy KM, Langer A, AbdAlla S. β-arrestin1 prevents preeclampsia by downregulation of mechanosensitive AT1–B2 receptor heteromers. Cell 176: 318–333.e19, 2019. doi: 10.1016/j.cell.2018.10.050. [DOI] [PubMed] [Google Scholar]
  • 47.Ramchand J, Patel SK, Srivastava PM, Farouque O, Burrell LM. Elevated plasma angiotensin converting enzyme 2 activity is an independent predictor of major adverse cardiac events in patients with obstructive coronary artery disease. PLoS One 13: e0198144, 2018. doi: 10.1371/journal.pone.0198144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Rice GI, Thomas DA, Grant PJ, Turner AJ, Hooper NM. Evaluation of angiotensin-converting enzyme (ACE), its homologue ACE2 and neprilysin in angiotensin peptide metabolism. Biochem J 383: 45–51, 2004. doi: 10.1042/BJ20040634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Santos RA. Angiotensin-(1-7). Hypertension 63: 1138–1147, 2014. doi: 10.1161/HYPERTENSIONAHA.113.01274. [DOI] [PubMed] [Google Scholar]
  • 50.Satou R, Penrose H, Navar LG. Inflammation as a regulator of the renin-angiotensin system and blood pressure. Curr Hypertens Rep 20: 100, 2018. doi: 10.1007/s11906-018-0900-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Schmaier AH, McCrae KR. The plasma kallikrein-kinin system: its evolution from contact activation. J Thromb Haemost 5: 2323–2329, 2007. doi: 10.1111/j.1538-7836.2007.02770.x. [DOI] [PubMed] [Google Scholar]
  • 52.Schouten LRA, Bos LDJ, Serpa Neto A, van Vught LA, Wiewel MA, Hoogendijk AJ, Bonten MJM, Cremer OL, Horn J, van der Poll T, Schultz MJ, Wösten-van Asperen RM; MARS consortium . Increased mortality in elderly patients with acute respiratory distress syndrome is not explained by host response. Intensive Care Med Exp 7: 58, 2019. doi: 10.1186/s40635-019-0270-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Serfozo P, Wysocki J, Gulua G, Schulze A, Ye M, Liu P, Jin J, Bader M, Myöhänen T, García-Horsman JA, Batlle D. Ang II (angiotensin II) conversion to angiotensin-(1–7) in the circulation is POP (prolyloligopeptidase)-dependent and ACE2 (angiotensin-converting enzyme 2)-independent. Hypertension 75: 173–182, 2020. doi: 10.1161/HYPERTENSIONAHA.119.14071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Sevá Pessôa B, van der Lubbe N, Verdonk K, Roks AJ, Hoorn EJ, Danser AH. Key developments in renin-angiotensin-aldosterone system inhibition. Nat Rev Nephrol 9: 26–36, 2013. doi: 10.1038/nrneph.2012.249. [DOI] [PubMed] [Google Scholar]
  • 55.Shariat-Madar Z, Mahdi F, Schmaier AH. Identification and characterization of prolylcarboxypeptidase as an endothelial cell prekallikrein activator. J Biol Chem 277: 17962–17969, 2002. doi: 10.1074/jbc.M106101200. [DOI] [PubMed] [Google Scholar]
  • 56.Silva-Filho JL, Caruso-Neves C, Pinheiro AA. Angiotensin II type-1 receptor (AT1R) regulates expansion, differentiation, and functional capacity of antigen-specific CD8+ T cells. Sci Rep 6: 35997, 2016. doi: 10.1038/srep35997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Silva-Filho JL, Caruso-Neves C, Pinheiro AA. Targeting angiotensin II type-1 receptor (AT1R) inhibits the harmful phenotype of Plasmodium-specific CD8+ T cells during blood-stage malaria. Front Cell Infect Microbiol 7: 42, 2017. doi: 10.3389/fcimb.2017.00042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Skidgel RA, Erdös EG. Cellular carboxypeptidases. Immunol Rev 161: 129–141, 1998. doi: 10.1111/j.1600-065X.1998.tb01577.x. [DOI] [PubMed] [Google Scholar]
  • 59.Soro-Paavonen A, Gordin D, Forsblom C, Rosengard-Barlund M, Waden J, Thorn L, Sandholm N, Thomas MC, Groop PH; FinnDiane Study Group . Circulating ACE2 activity is increased in patients with type 1 diabetes and vascular complications. J Hypertens 30: 375–383, 2012. doi: 10.1097/HJH.0b013e32834f04b6. [DOI] [PubMed] [Google Scholar]
  • 60.Sriram K, Insel PA. A hypothesis for pathobiology and treatment of COVID-19: The centrality of ACE1/ACE2 imbalance. Br J Pharmacol. In press. doi: 10.1111/bph.15082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Takayanagi T, Kawai T, Forrester SJ, Obama T, Tsuji T, Fukuda Y, Elliott KJ, Tilley DG, Davisson RL, Park JY, Eguchi S. Role of epidermal growth factor receptor and endoplasmic reticulum stress in vascular remodeling induced by angiotensin II. Hypertension 65: 1349–1355, 2015. doi: 10.1161/HYPERTENSIONAHA.115.05344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Tikellis C, Cooper ME, Bialkowski K, Johnston CI, Burns WC, Lew RA, Smith AI, Thomas MC. Developmental expression of ACE2 in the SHR kidney: a role in hypertension? Kidney Int 70: 34–41, 2006. doi: 10.1038/sj.ki.5000428. [DOI] [PubMed] [Google Scholar]
  • 63.Tipnis SR, Hooper NM, Hyde R, Karran E, Christie G, Turner AJ. A human homolog of angiotensin-converting enzyme. Cloning and functional expression as a captopril-insensitive carboxypeptidase. J Biol Chem 275: 33238–33243, 2000. doi: 10.1074/jbc.M002615200. [DOI] [PubMed] [Google Scholar]
  • 64.Turner AJ, Hiscox JA, Hooper NM. ACE2: from vasopeptidase to SARS virus receptor. Trends Pharmacol Sci 25: 291–294, 2004. doi: 10.1016/j.tips.2004.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.van de Veerdonk F, Netea MG, van Deuren M, van der Meer JW, de Mast Q, Bruggemann RJ, van der Hoeven H. Kinins and cytokines in COVID-19: a comprehensive pathophysiological approach. Preprints 2020. doi: 10.20944/preprints202004.0023.v1. [DOI]
  • 66.van de Veerdonk FL, Netea MG, van Deuren M, van der Meer JW, de Mast Q, Brüggemann RJ, van der Hoeven H. Kallikrein-kinin blockade in patients with COVID-19 to prevent acute respiratory distress syndrome. eLife 9: e57555, 2020. doi: 10.7554/eLife.57555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Verma S, Gupta M, Holmes DT, Xu L, Teoh H, Gupta S, Yusuf S, Lonn EM. Plasma renin activity predicts cardiovascular mortality in the Heart Outcomes Prevention Evaluation (HOPE) study. Eur Heart J 32: 2135–2142, 2011. doi: 10.1093/eurheartj/ehr066. [DOI] [PubMed] [Google Scholar]
  • 68.Vickers C, Hales P, Kaushik V, Dick L, Gavin J, Tang J, Godbout K, Parsons T, Baronas E, Hsieh F, Acton S, Patane M, Nichols A, Tummino P. Hydrolysis of biological peptides by human angiotensin-converting enzyme-related carboxypeptidase. J Biol Chem 277: 14838–14843, 2002. doi: 10.1074/jbc.M200581200. [DOI] [PubMed] [Google Scholar]
  • 69.Wallingford N, Perroud B, Gao Q, Coppola A, Gyengesi E, Liu ZW, Gao XB, Diament A, Haus KA, Shariat-Madar Z, Mahdi F, Wardlaw SL, Schmaier AH, Warden CH, Diano S. Prolylcarboxypeptidase regulates food intake by inactivating alpha-MSH in rodents. J Clin Invest 119: 2291–2303, 2009. doi: 10.1172/JCI37209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wösten-van Asperen RM, Bos AP, Bem RA, Dierdorp BS, Dekker T, van Goor H, Kamilic J, van der Loos CM, van den Berg E, Bruijn M, van Woensel JB, Lutter R. Imbalance between pulmonary angiotensin-converting enzyme and angiotensin-converting enzyme 2 activity in acute respiratory distress syndrome. Pediatr Crit Care Med 14: e438–e441, 2013. doi: 10.1097/PCC.0b013e3182a55735. [DOI] [PubMed] [Google Scholar]
  • 71.Wösten-van Asperen RM, Lutter R, Specht PA, Moll GN, van Woensel JB, van der Loos CM, van Goor H, Kamilic J, Florquin S, Bos AP. Acute respiratory distress syndrome leads to reduced ratio of ACE/ACE2 activities and is prevented by angiotensin-(1-7) or an angiotensin II receptor antagonist. J Pathol 225: 618–627, 2011. doi: 10.1002/path.2987. [DOI] [PubMed] [Google Scholar]
  • 72.Wu Y, Yang H, Xiao C. Genetic association study of prolylcarboxypeptidase polymorphisms with susceptibility to essential hypertension in the Yi minority of China: A case-control study based on an isolated population. J Renin Angiotensin Aldosterone Syst 21: 1470320320919586, 2020. doi: 10.1177/1470320320919586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Wu Y. Contact pathway of coagulation and inflammation. Thromb J 13: 17, 2015. doi: 10.1186/s12959-015-0048-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Xie J, Tong Z, Guan X, Du B, Qiu H. Clinical characteristics of patients who died of coronavirus disease 2019 in China. JAMA Netw Open 3: e205619, 2020. doi: 10.1001/jamanetworkopen.2020.5619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Xie X, Chen J, Wang X, Zhang F, Liu Y. Age- and gender-related difference of ACE2 expression in rat lung Life Sci 78: 2166–2171, 2006. [Erratum in Life Sci 79: 2499, 2006.] doi: 10.1016/j.lfs.2005.09.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Xue B, Thunhorst RL, Yu Y, Guo F, Beltz TG, Felder RB, Johnson AK. Central renin-angiotensin system activation and inflammation induced by high-fat diet sensitize angiotensin II-elicited hypertension. Hypertension 67: 163–170, 2016. doi: 10.1161/HYPERTENSIONAHA.115.06263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Xue B, Zhang Z, Johnson RF, Johnson AK. Sensitization of slow pressor angiotensin II (Ang II)-initiated hypertension: induction of sensitization by prior Ang II treatment. Hypertension 59: 459–466, 2012. doi: 10.1161/HYPERTENSIONAHA.111.185116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Yan Y, Liu Q, Li N, Du J, Li X, Li C, Jin N, Jiang C. 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 58: 208–211, 2015. doi: 10.1007/s11427-015-4814-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Yang P, Gu H, Zhao Z, Wang W, Cao B, Lai C, Yang X, Zhang L, Duan Y, Zhang S, Chen W, Zhen W, Cai M, Penninger JM, Jiang C, Wang X. Angiotensin-converting enzyme 2 (ACE2) mediates influenza H7N9 virus-induced acute lung injury. Sci Rep 4: 7027, 2014. doi: 10.1038/srep07027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Zhang Y, Hong XM, Xing HX, Li JP, Huo Y, Xu XP. E112D polymorphism in the prolylcarboxypeptidase gene is associated with blood pressure response to benazepril in Chinese hypertensive patients. Chin Med J (Engl) 122: 2461–2465, 2009. [PubMed] [Google Scholar]
  • 81.Zhao X, Zhang B, Li P, Ma C, Gu J, Hou P, Guo Z, Wu Z, Bai Y. Incidence, clinical characteristics and prognostic factor of patients with COVID-19: a systematic review and meta-analysis (Preprint). medRxiv 2020. doi: 10.1101/2020.03.17.20037572. [DOI]
  • 82.Zheng J. SARS-CoV-2: an emerging coronavirus that causes a global threat. Int J Biol Sci 16: 1678–1685, 2020. doi: 10.7150/ijbs.45053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Zhong J, Basu R, Guo D, Chow FL, Byrns S, Schuster M, Loibner H, Wang XH, Penninger JM, Kassiri Z, Oudit GY. Angiotensin-converting enzyme 2 suppresses pathological hypertrophy, myocardial fibrosis, and cardiac dysfunction. Circulation 122: 717–728, 2010. doi: 10.1161/CIRCULATIONAHA.110.955369. [DOI] [PubMed] [Google Scholar]
  • 84.Zou Z, Yan Y, Shu Y, Gao R, Sun Y, Li X, Ju X, Liang Z, Liu Q, Zhao Y, Guo F, Bai T, Han Z, Zhu J, Zhou H, Huang F, Li C, Lu H, Li N, Li D, Jin N, Penninger JM, Jiang C. Angiotensin-converting enzyme 2 protects from lethal avian influenza A H5N1 infections. Nat Commun 5: 3594, 2014. doi: 10.1038/ncomms4594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Zubcevic J, Santisteban MM, Perez PD, Arocha R, Hiller H, Malphurs WL, Colon-Perez LM, Sharma RK, de Kloet A, Krause EG, Febo M, Raizada MK. A single angiotensin II hypertensive stimulus is associated with prolonged neuronal and immune system activation in Wistar-Kyoto rats. Front Physiol 8: 592, 2017. doi: 10.3389/fphys.2017.00592. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Lung Cellular and Molecular Physiology are provided here courtesy of American Physiological Society

RESOURCES