Next Article in Journal
NMR-Based Metabolomics of Blood Serum in Predicting Response to Induction Chemotherapy in Head and Neck Cancer—A Preliminary Approach
Next Article in Special Issue
Concomitance of Pericardial Tamponade and Pulmonary Embolism in an Invasive Mucinous Lung Adenocarcinoma with Atypical Presentation: Diagnostic and Therapeutic Pitfalls—Case Report and Literature Review
Previous Article in Journal
Impact of Maternal Pre-Pregnancy Underweight on Cord Blood Metabolome: An Analysis of the Population-Based Survey of Neonates in Pomerania (SNiP)
Previous Article in Special Issue
Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Aprotinin (I): Understanding the Role of Host Proteases in COVID-19 and the Importance of Pharmacologically Regulating Their Function

by
Juan Fernando Padín
1,
José Manuel Pérez-Ortiz
2,3,* and
Francisco Javier Redondo-Calvo
1,4,5,*
1
Department of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, 13971 Ciudad Real, Spain
2
Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Madrid, Spain
3
Instituto de Investigación Sanitaria HM Hospitales, 28015 Madrid, Spain
4
Department of Anaesthesiology and Critical Care Medicine, University General Hospital, 13005 Ciudad Real, Spain
5
Translational Research Unit, University General Hospital and Research Institute of Castilla-La Mancha (IDISCAM), 13005 Ciudad Real, Spain
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(14), 7553; https://doi.org/10.3390/ijms25147553
Submission received: 27 May 2024 / Revised: 6 July 2024 / Accepted: 8 July 2024 / Published: 10 July 2024

Abstract

:
Proteases are produced and released in the mucosal cells of the respiratory tract and have important physiological functions, for example, maintaining airway humidification to allow proper gas exchange. The infectious mechanism of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), takes advantage of host proteases in two ways: to change the spatial conformation of the spike (S) protein via endoproteolysis (e.g., transmembrane serine protease type 2 (TMPRSS2)) and as a target to anchor to epithelial cells (e.g., angiotensin-converting enzyme 2 (ACE2)). This infectious process leads to an imbalance in the mucosa between the release and action of proteases versus regulation by anti-proteases, which contributes to the exacerbation of the inflammatory and prothrombotic response in COVID-19. In this article, we describe the most important proteases that are affected in COVID-19, and how their overactivation affects the three main physiological systems in which they participate: the complement system and the kinin–kallikrein system (KKS), which both form part of the contact system of innate immunity, and the renin–angiotensin–aldosterone system (RAAS). We aim to elucidate the pathophysiological bases of COVID-19 in the context of the imbalance between the action of proteases and anti-proteases to understand the mechanism of aprotinin action (a panprotease inhibitor). In a second-part review, titled “Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions”, we explain in depth the pharmacodynamics, pharmacokinetics, toxicity, and use of aprotinin as an antiviral drug.

1. Introduction

Coronaviruses infect epithelial cells by recognising and binding to certain plasma membrane proteins. One of the most studied is the angiotensin-converting enzyme type 2 (ACE2). The anchoring mechanism of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), to this enzyme has consequences that are fundamental for the cell and is closely related to the disease it causes. To understand the pathophysiology COVID-19, it is necessary to explain the importance of ACE2, which goes beyond blood pressure control. In this first review, titled “Aprotinin (I): Understanding the Role of Host Proteases in COVID-19 and the Importance of Pharmacologically Regulating Their Function”, we explain the pathophysiological importance of ACE in the SARS-CoV-2 infection process, and how a cascade of events activates proteolytic pathways that constitute the most important causes of the disease. Understanding these mechanisms allows the development of new antiviral drugs, such as aprotinin, which is a broad-spectrum inhibitor of host proteases. In the second-part review, titled “Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions” [1], we describe the main pharmacodynamic, pharmacokinetic, and toxicological mechanisms of aprotinin for its use by inhalation in these conditions.

2. The SARS-CoV-2 Infectious Process

ACE2, the main target to which SARS-CoV-2 anchors in the viral infectious process, is expressed to a greater or lesser extent in the cells of the pulmonary, digestive, renal, and vascular endothelium [2,3]. Anchoring and fusion of the viral capsid to the host cell occurs through recognition of a virus envelope glycoprotein, known as the spike (S) protein, with this enzyme [4,5,6]. However, for the S protein to recognise it, it needs to undergo a post-translational activation process by endoproteolysis by proteases from the epithelial cells [7]. This two-step entry mechanism that involves activation by endogenous host proteases is common in viruses of the Paramyxoviridae, Orthomyxoviridae, Retroviridae, Herpesviridae, Flaviviridae, Filoviridae, Hepadnaviridae, Togaviridae, and Coronaviridae families [8,9]. In the specific case of SARS-CoV-2, the cleavage occurs in four redundant furin-like domains [10] located in the S1/S2 protein subunits of the virus [11]. Proteolysis is indispensable to separate and activate the S1 and S2 subunits, each of which performs distinct functions (Figure 1A) [12,13]. While the S1 subunit is responsible for binding to ACE2 with an affinity in the nanomolar range [14], the S2 subunit participates in the fusion between viral RNA and the cell membrane [13,15]. Unlike other coronaviruses, this proteolytic cleavage and presentation of the S1 subunit for anchoring to ACE2 is much more efficient for SARS-CoV-2 [16]. Therefore, it is essential to know the host proteases used by SARS-CoV-2 for its infectious process to prevent disease.
Among the known host proteases, trypsin, cysteine protease cathepsins, thermolysin, neutrophil elastase (NE), and activated clotting factors, such as plasminogen and factor Xa (FXa), have been described [13,17,18,19,20,21]. In addition, much attention has been paid to transmembrane serine protease type 2 (TMPRSS2), one of the proteases for which SARS-CoV-2 shows the strongest preference to infect epithelial cells [22,23]. Once anchorage to ACE2 occurs, and depending on which protease is used in this process, the virus can enter the cell via at least two mechanisms. If the S protein is cleaved via cathepsins, entry will occur through endosome formation. In contrast, if proteolysis is via TMPRSS2, entry occurs through the formation of a fusion pore between its membrane and those of the epithelial cell [24]. ACE2 is usually co-expressed with TMPRSS2, indicating the importance of this mechanism [25]. However, the endocytic pathway usually occurs when TMPRSS2 expression levels are insufficient, and endosome formation occurs via a clathrin-dependent pathway, internalising the virus bound to ACE2 [26]. The use of this endocytic pathway in respiratory cells that do not express TMPRSS2 explains why some SARS-CoV-2 variants, such as Omicron, have enhanced transmissibility [27,28].
Key points:
  • SARS-CoV-2 uses both soluble and membrane proteases of the host cell in its viral infection mechanism.
  • Depending on which protease you use will influence the way it enters the cell (e.g., endocytosis or through the formation of a fusion pore).

2.1. The Physiological Importance of ACE beyond Blood Pressure Control

Although ACE was initially described for its importance in the control of blood pressure through the renin–angiotensin–aldosterone system (RAAS) [29], it is now known to have relevance in other processes, such as renal embryonic development [30], reproduction [31,32], cell proliferation (e.g., haematopoiesis, myeloproliferation, and angiogenesis), inflammation, oxidative stress, and immunity [33,34,35]. In somatic tissues, ACE contains two catalytic domains, each with different affinities for protein substrates. While the carboxyl-terminal domain is specialised in cleaving angiotensin I to generate angiotensin II, the amino-terminal domain is also capable of cleaving other peptides, including enkephalin, the tetrapeptide N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP), neurotensins, substance P, and bradykinin, among others (Figure 2) [36]. It can exert its protease function while anchored to the plasma membrane, extracellularly when released from cells into the blood plasma or subcellularly in cell organelles. Thus, it is not only an endocrine, paracrine, and autocrine cellular communication system, it is also an intracellular communication system (between the cell organelles themselves) [37]. These locations are important for the development of its various cellular functions and are of great relevance for understanding COVID-19 as well as the mechanisms by which SARS-CoV-2 evades the immune system.

ACE in Immunity

ACE is expressed in organelles, such as the endoplasmic reticulum, where it has been shown to be catalytically active [38], performing certain cellular functions. Among these, it contributes to adaptive immunity by cleaving both endogenous and foreign peptides for presentation by antigen-presenting cells (macrophages and dendritic cells) via major histocompatibility complex type I (MHC-I) and II (MHC-II) to cluster of differentiation 8 (CD8)+ T cells (Figure 2) [38,39,40].
In addition, ACE can be overexpressed in myeloid cells, such as macrophages, neutrophils, and dendritic cells, thereby enhancing the innate immune response through the production of proinflammatory cytokines and nitric oxide [41]. ACE plays a role in modulating the inflammatory response and recruitment of inflammatory cells, such as mast cells and neutrophils, by regulating the concentration of mediators and/or the expression of proteins, such as cytokines, adhesion molecules, and the plasma contact system, also known as the kinin–kallikrein system (KKS). ACE regulates this system by clearing the proinflammatory peptide bradykinin [42]. Increased bradykinin contributes to the activation of the FXII coagulation and complement pathways [43]. ACE can be secreted into the extracellular environment (soluble ACE (sACE)) from these myeloid cells and act as a local or systemic regulator of the production of these peptides [41]. In neutrophils, ACE is essential for immune competence [44]. These effects are independent of the angiotensin-converting action and depend on other factors—for example, activation of nicotinamide adenine dinucleotide phosphate oxidase (NOX) by ACE increases reactive oxygen species (ROS, including the superoxide) production [41,44]. Neutrophils kill bacteria using phagocytic mechanisms and through the release of extracellular fibres, called neutrophil extracellular traps (NETs), which are in turn stimulated by ROS generation. These fibres are composed of DNA and proteins that bind to, trap, and consequently kill bacteria. Moreover, under certain conditions, monocytes overexpress ACE, triggering further differentiation of these cells into macrophages, which release cytokines and adhesion and transmigration molecules [45]. In addition, ACE is also involved in the production of nitric oxide, which is important for microbial defence by these cells (Figure 2) [46].
Finally, ACE is involved in the control of immunity through the clearance of peptides with immunosuppressive activity, such as AcSDKP [36,47]. Among many actions, this peptide can inhibit the G1–S cell cycle transition and thus maintains haematopoietic progenitor cells in a quiescent phase [48]. This prevents proliferation, migration, and cytokine release by myeloid cells [49,50]. In addition, it can prevent fibroblast collagen synthesis and deposition by inhibiting DNA synthesis and endothelin-1, and by blocking small mothers against decapentaplegic (Smad) signal transduction and the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway, thus preventing the action of cytokines, such as transforming growth factor-β (TGF-β) [51]. AcSDKP has antifibrotic and anti-inflammatory effects in the lung, heart, liver, and kidney [51,52]. Therefore, ACE may exert these effects by decreasing angiotensin II levels and increasing AcSDKP levels [50].
Key points:
  • ACE cleaves a multitude of peptides acting subcellularly, anchored to the plasma membrane, or extracellularly.
  • ACE is involved in cleaving peptides to be presented by antigen-presenting cells at MHCs. Its dysregulation may contribute to viral evasion mechanisms.
  • ACE is overexpressed in myeloid cells as a mechanism of immune response and inflammation.
  • ACE regulates the contact system and the KKS.
  • ACE regulates immunity through the clearance of peptides, such as Ac-SDKP.

3. The Protease and Anti-Protease System in the Lung and SARS-CoV-2 Infection

The respiratory epithelium is mucosal tissue that consists of more than 40 different cell types [53]. These cells coordinate their actions through the release of various mediators (mucins, cytokines, proteases, and anti-proteases) to maintain respiratory function and protect against foreign agents. Proteases that are released from the respiratory mucosa are involved in processes related to airway function, including mucus characteristics (i.e., its density or rheology) [54], mucociliary clearance [55,56], and the recruitment and function of immune cells [57,58]. This process is carried out by cleaving pre-proteins to their physiologically active forms, a process that is finely regulated by the action of anti-proteases to control excessive over-activation [59]. The human genome contains more than 600 proteases, which gives an idea of their importance [60].
When infection occurs, the respiratory epithelium starts to produce proteases, such as the type II transmembrane serine proteases (TTSPs). This family includes matrix metalloproteinases (MMPs), a disintegrin and metalloproteinases (ADAMs), a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs), cathepsins, proteinase-3, human neutrophil elastase, trypsin, chymotrypsin, prostasin, and TMPRSSs. In addition to the aforementioned processes, these proteases regulate tissue repair, blood coagulation (secondary haemostasis), fibrinolysis, and immune function [61]. Moreover, anti-proteases or serpins can be released from respiratory epithelia, including α-1-antitrypsin (serpin A1), plasminogen activator inhibitor 1 (serpin E1), and glia-derived nexin (serpin E2), which can reduce SARS-CoV-2 infection [62]. The underlying pulmonary pathological processes involve an imbalance between proteases and anti-proteases [63], and viruses are known to deregulate this balance [64,65,66,67]. Proteomics studies in cells infected in vitro with SARS-CoV-2 have shown dysregulation with increased expression of proteases and decreased expression of anti-proteases (e.g., SPINT1 (Kunitz-type protease inhibitor 1), SPINT2 (Kunitz-type protease inhibitor 2), tissue inhibitors of metalloproteinases 1/2 (TIMP1/2), amyloid beta precursor-like protein 2 (APLP2), cystatin C (CST3), and α-1-antitrypsin) [68,69]. Therefore, it is necessary to re-establish their balance to have a healthy mucosa.
Key points:
  • The respiratory mucosa controls its functions through a complex release system of proteases and anti-proteases.
  • Respiratory viruses in their infective process unbalance the action of proteases and anti-proteases.

3.1. Host Proteases in SARS-CoV-2 Infection

Coronaviruses, such as SARS-CoV-2, take advantage of the activity of multiple proteases to infect a host cell [21]. When respiratory epithelial cells become infected, hyperreactivity occurs, where proteases (e.g., trypsin, cathepsins, and/or elastases) are overexpressed and released from respiratory tract epithelial cells as well as myeloid cells. Excessive activity of these proteases significantly contributes to the inflammatory and/or infectious processes. We have already discussed the role of TTSPs, such as TMPRSS2, in the process of SARS-CoV-2 entry into a host epithelial cell [23]. Below, we briefly review other proteases that also play an important role in the pathological process in the respiratory tract.

3.1.1. ADAMs

ADAMs are a family of type I transmembrane proteins belonging to the adamalysin subfamily of metalloproteinases. The members of this family have a metalloprotease domain and an integrin-interacting domain (disintegrin domain), indicating that they have both protease and adhesion molecule activity. They participate in the cellular processes of migration, adhesion, and cell fusion. In addition, through their protease activity, they participate in cell signalling by cleaving certain protein domains (sheddase activity), leading to the release of cell membrane-associated proteins, such as cytokines, apoptotic ligands, growth factors, and receptors. One of the most studied examples is the production of TNF-α. ADAM17 is also known as the TNF-α convertase (TACE)—it cleaves transmembrane TNF (26 kDa) to release its active soluble form, TNF-α (17 kDa). This family of proteases also produce mucus at the bronchial level [70]. In COVID-19, ADAM17 is upregulated via internalisation of ACE2 after it binds to SARS-CoV-2, a phenomenon that has been observed with other coronaviruses [71]. In addition, ADAM17 proteolytically cleaves ACE2, releasing it in its soluble form (sACE2) and decreasing its expression in the cytoplasmic membrane [72]. This phenomenon contributes to the infectious process [73] as well as the release of TNF-α, interleukin 6 (IL-6), and other proinflammatory molecules, which aggravates the inflammatory process (Figure 1B) [73,74].

3.1.2. Elastases and Other Neutrophil Serine Proteases

Elastases are serine proteases that have important physiological functions through the cleavage of multiple protein substrates. In coronavirus infections, elastases, such as pancreatic elastase, are involved in cleavage of the S protein [75,76] via an elastase-specific domain in the S2 subunit [75]. For SARS-CoV-2, the involvement of elastases released from myeloid cells such as macrophages [77], but mostly neutrophils, has been investigated. Their importance lies in the viral entry process [78]. In addition, the release of human neutrophil elastase from neutrophils is associated with increased production of cathepsins and matrix metalloproteases [79], which promote infection. Along with human neutrophil elastase, neutrophils release other serine proteases, such as cathepsin G, proteinase-3, neutrophil serine protease-4 (NSP-4), azurocidin (AZU1), myeloperoxidase, myeloblastin (PRTN3), and transcobalamin-1 (TCN1), which are stored in azurophil granules [80]. The main functions are to degrade the extracellular matrix by digesting collagen, transmembrane proteins, pulmonary surfactant factors, and proteoglycans. Therefore, their release contributes to a process of permeabilisation of cellular barriers, inflammation, and alteration of lung functions. In addition, they participate in inflammation by processing and modifying cytokine functions, inhibiting anti-inflammatory factors, such as progranulin, activating surface receptors, such as Toll-like receptors (TLRs) through their protease action, and promoting cytokine release from monocytes and macrophages [80,81]. However, in COVID-19, the most prominent role of human neutrophil elastase is in NET formation and NETosis, a process in which neutrophils expel their citrullinated chromatin into the extracellular milieu. Combined with the action of released proteases, a network is produced with the capacity to trap platelets and red blood cells, linking the thromboembolic processes that occur during COVID-19 [82]. The exacerbation of these processes is partially caused by an imbalance between the release and activity of proteases secreted by neutrophils, as opposed to anti-proteases, such as α-1-antitrypsin and other inhibitors secreted from leucocytes [78].

3.1.3. Trypsin and Human Airway Trypsin-like Proteases (HATs)

Trypsin has been shown to increase the infectivity of several coronaviruses, including SARS-CoV-2, when the virus is already attached to its cellular target [13,76,83]. These mechanisms do not necessarily require binding to ACE2, but they favour increased membrane fusion. In certain SARS-CoV-2 variants (e.g., Delta) that have a greater capacity to produce syncytia [84], and therefore have greater membrane fusion activity, trypsin plays a key role in this infectious mechanism [85]. On the other hand, excess trypsin release contributes to cell barrier permeabilisation, inflammation, and impaired lung function [86].
HATs are preferentially anchored to the surface of bronchial and tracheal respiratory tract hair cells [87]. They can also be found in soluble forms in patients with respiratory diseases [88]. Coronaviruses that need to cleave proteins from their capsid during the infectious process can use these proteases [89]. In addition, they participate in the activation of epithelial sodium channels (ENaC) that hydrate the airway and facilitate mucociliary clearance [90]. ENaC and the coronavirus S protein share the same furin-like cleavage domain, and thus use the same proteases (e.g., TMPRSS2) for protein activation [91]. Hence, by modifying the activity of proteases after infection, coronaviruses may affect ENaC activity [91,92]. SARS-CoV-2 leads to the overexpression of proteases, such as TMPRSS2, at the host plasma membrane and simultaneously prevents these proteases from degrading ENaC, leaving them in an overactivated state [93]. This state accelerates viral entry into the host cell [94]. In addition, altered ENaC activity is consistent with symptoms following SARS-CoV-2 infection, such as a runny nose, ageusia, pulmonary oedema, and respiratory distress [91].
The increased release or expression of proteases, such as trypsin, HATs, and others secreted by neutrophils (e.g., elastase and proteinase-3), favours activation of protease-activated receptor type 2. This receptor promotes the release of inflammatory mediators [95,96,97] that aggravate the inflammatory process [98] and cell growth, contributing to airway remodelling through fibroblast proliferation [99]. In addition, they can cleave and activate the urokinase-type plasminogen-activated receptor [100], which also contributes to thromboembolic processes in patients with COVID-19 [98].

3.1.4. Cathepsins

Cysteine cathepsins are a family of papain-like proteases found intracellularly in organelles such as lysosomes, although they have also been observed in the cytosol, mitochondria, nucleus, plasma membranes, and the extracellular milieu [101,102]. Cathepsin secretion into the extracellular milieu is observed under physiological conditions; for example, cathepsin B, K, and L are secreted from thyroid epithelial cells to release thyroid hormones from thyroglobulins. However, excessive cathepsin release can also be triggered in pathological conditions that involve inflammation [102,103]. In infectious diseases, such as COVID-19, where inflammation is evident, their expression and secretion increase [80,104]. A relevant fact is the ability of cathepsins to modify the spatial conformation of proteins by cleaving certain domains to alter their functionality [102]. The high transmissibility of SARS-CoV-2 variants, such as Omicron, has been linked to a greater capacity to enter the cell via an endocytic pathway, thanks to cathepsin-mediated modification of the S protein to produce the correct spatial arrangement of the S2 subunit [24,27,28,105]. A large number of cathepsin subtypes have been proposed to be involved in SARS-CoV-2 infection [18,21]. Researchers have used in silico and in vitro approaches to determine the exact S protein cleavage sites [18,106]. Cathepsins are upregulated upon infection, which contributes to aggravating infection, especially in those tissues where other proteases, such as TMPRSS2, are not as present, thus allowing a new entry route to infect these cells [68,80]. Another relevant factor regarding the action of cathepsins in the extracellular milieu is that they are important initiators or suppressors of the activity of other proteases or cytogens involved in proteolytic cascades. For example, cathepsin G is one of the main proteases, along with human neutrophil elastase, involved in NET formation, inflammation, and thrombosis. The mechanisms involve inactivation of tissue factor inhibitory peptide and activation of protease-activated receptors [107]. In addition, cathepsins are potent activators of blood platelets [108], and cathepsin L activates the KKS [109].
Cathepsin F, L, S, and V are also present in endolysosomes, where they cleave proteins and contribute to maturation and processing functions. An example of these functions is the processing of ENaC subunits [110]. The presence of SARS-CoV-2 and its viral proteins in lysosomes interferes in the proteolytic cleavage of ENaC and in its maturation processes that alter its functionality. These changes contribute to COVID-19 symptoms, as ENaC is involved in proper homeostasis of the pulmonary fluid interface [91] and in the occurrence of electrolyte imbalance (i.e., hypokalaemia) due to altered renal function [93,111]. Another example is the processing of antigenic proteins in immune cells for presentation by MHC-I and MHC-II [112]. Moreover, lysosomal cathepsins influence the trafficking of SARS-CoV-2-specific proteins, such as the accessory protein open reading frame 3a (ORF3a), which is involved in virus infectivity and the formation of new virions [96,113]. Indeed, if cathepsins are inhibited, it can lead to degradation of new virions into cellular multivesicular bodies [114].
SARS-CoV-2 may use a lysosomal pathway that involves cathepsins to release newly formed virions from the host cell [115], although the mechanisms remain unclear. In addition, cathepsin L is involved in the upregulation and processing of heparanase, which is implicated in the release of viral progeny and their propagation [116,117,118]. Serum heparanase levels are increased in patients with COVID-19 and correlate with the severity of the disease [116,119]. Specifically, heparanase damages the glycocalyx of endothelial cells and promotes thromboembolism [120]. It causes the release of certain molecules from heparan sulphate proteoglycans of the glycocalyx that bind to it, including growth factors, cytokines, enzymes, and lipoproteins [121]. These molecules released by heparanase are involved in cell motility, angiogenesis, inflammation, coagulation, stimulation of autophagy, and exosome production [122,123].
Since cathepsins are involved in the mechanisms of entry, processing of viral proteins, and release of viral progeny to the cellular exterior, their inhibition is a very attractive and well-studied therapeutic strategy for the development of antiviral drugs against coronaviruses [124,125].
Key points:
  • SARS-CoV-2 infection upregulates ADAM-17, causing the adhesion of inflammatory cells and the release of chemoattractants, such as sACE2 and proinflammatory cytokines (IL-6 and TNF-α).
  • Elastases participate in viral entry into the host cell and the release of other proteases that amplify the infectious and inflammatory response, permeabilise cellular barriers, and alter lung functions. Neutrophil elastase participates in NETosis.
  • Trypsin promotes infection when the virus is attached to the cell membrane by increasing the membrane fusion process.
  • Coronaviruses prevent proteases from exercising their physiological functions of correct maturation of proteins, such as ENaC. This is involved in many of the symptoms caused by respiratory viruses.
  • Cathepsins allow the entry of SARS-CoV-2 via the endocytic route into those cells where other proteases, such as TMPRSS2, are not so present.
  • Cathepsins initiate and amplify the activation of pathways activated by proteases such as KKS, coagulation, or the formation of NETs.
  • Cathepsins participate in entry, maturation of viral proteins, and release of new viral progeny from the host cell.

3.2. Host Anti-Proteases in SARS-CoV-2 Infection

3.2.1. α-1-Antitrypsin (Serpin A1)

In addition to increasing protease activity, coronaviruses negatively modulate the action of anti-proteases, which contributes to an imbalance between proteases and anti-proteases that prevents proper respiratory function. In general, coronaviruses increase the degradation of α-1-antitrypsin (serpin A1), increasing its degradation products. This mechanism is associated with increased pathogenicity [126,127] because α-1-anthrypsin is one of the most important inhibitors of serine proteases in the lung and, therefore, has important anti-inflammatory actions [128]. The loss of α-1-anthrypsin activity is also relevant to the SARS-CoV-2 infectious process [126]. For example, α-1-antitrypsin inhibits TMPRSS2, which is required for S protein cleavage [129], thus aggravating the infection. Once infection has occurred, the inflammatory process starts, with IL-6 serving as one of the main mediators and a prognostic marker of the disease [130,131]. One of the main actions of IL-6 is to induce hepatic synthesis of α-1-antitrypsin. It does so through the formation of IL-6-soluble IL-6 receptor (sIL-6R) complexes and subsequent binding to glycoprotein 130 (gp130), which activates the Janus kinase (JAK)/signal transducer and activator of transcription (STAT), ERK, and phosphoinositide 3-kinase (PI3K) signalling pathways in liver cells, regulating gene transcription [132]. As we discuss later, this is due to an increase in the release of sIL-6R via the action of proteases, such as ADAM17 [73,133], as opposed to action on un-cleaved membrane receptors released by proteases. Hence, an increased IL-6/ α-1-antitrypsin ratio has been proposed as a biomarker for a poor prognosis in SARS-CoV-2 infection [130,131]. The fact that anti-proteases, such as α-1-antitrypsin—whose function is to inhibit proteases (e.g., human neutrophil elastase, cathepsins, and metalloproteases) that are activated when the infectious process occurs, as a regulatory control mechanism [128]—lose functionality results in infection and inflammation. In addition, it is involved in the regulation of coagulation by inhibiting thrombin [134]. Thus, patients with COVID-19 and attenuated α-1-antitrypsin production tend to have a poor prognosis [130,131]. Therefore, α-1-antitrypsin is being evaluated for its therapeutic utility in several trials for the treatment of COVID-19 (ClinicalTrials.gov IDs: NCT04385836, NCT04547140, NCT04495101, and NCT04817332 [134,135]).

3.2.2. CST3

Cystatins are a superfamily of proteins that are ubiquitously expressed in all nucleated cells and consist of at least one 100–120 amino acid inhibitory domain with protease activity. There are three types of cystatins: type I or Stefins, which are cytosolic proteins, type II, which are secreted from cells into the extracellular milieu, and type III or quininogens, which are multifunctional proteins found in the blood and other fluids [136]. CST3 is secreted in body fluids, such as saliva and urine [137]. It has antiviral activity against coronaviruses and, therefore, its recombinant forms have been proposed as antiviral drugs against them [138,139]. The antiviral mechanism is related to the ability to inhibit cysteine proteases, such as cathepsins, which are used by the virus to gain entry to a host cell via the endocytic pathway [136,139]. Cathepsin S and L, among others, have an important role in foreign protein digestion, processing, and loading onto MHC-I and MHC-II for presentation by antigen-presenting cells (including dendritic cells) to T lymphocytes. By inhibiting cathepsins, cystatins regulate the generation of peptidergic MHC antigenic complexes [140]. This process involves other related proteins that have the capacity to inhibit cathepsins. The p41 isoform of CD74 of MHC-II has recently been found to inhibit them, in addition to its antigen-presenting function. In turn, CD74 can be activated through the MHC-II master regulator MHC-II transactivator (CIITA) [141]. Interestingly, CIITA levels have been observed to decrease in both children and adults when the prognosis of COVID-19 is severe [142]. The importance of this chain of events—ranging from CIITA activation of p41 of CD74 to cathepsins as final effector proteins—is that in addition to participating in antigen presentation, they intervene in the antiviral response via interferon (IFN) [143]. Finally, inhibition of cathepsins causes virions to be redirected to a degradation pathway in multivesicular bodies or lysosomes. Hence, inactivation of these pathways plays a very important protective role via reduction of viral transcription, assembly, and release [144].
Paradoxically, many studies on COVID-19 have reported a positive correlation between elevated serum and urine CST3 levels and mortality [145,146,147]. CST3 is a highly sensitive biomarker in the assessment of cardiovascular and renal function because it is filtered only at the glomerular level. Of note, renal failure is one of the main causes of mortality in patients with COVID-19 [148,149]. On the other hand, in the saliva of patients with moderate or severe COVID-19, CST3 levels have been observed to be decrease with respect to healthy individuals [150,151,152]. In hospitalised patients, CST3 levels tend to increase in those who are symptomatic relative to those who are asymptomatic, although these values are always lower than in healthy individuals [150]. These differences in CST3 saliva and serum levels are most likely due to renal impairment and the fact that many of these patients are treated with glucocorticoids that increase CST3 plasma levels [153]. In addition, elevated levels of ILs, such as IL-6, downregulate cystatin levels, and vice versa [154]. Critically ill patients with COVID-19 have high IL-6 levels [131] and show an abnormal glomerular filtration rate (GFR). Specifically, the GFRcystatinC/eGFRcreatinine ratio is <0.6 due to the influence that cytokines, such as IL-6, have on the regulation of CST3, among other factors [155,156].

3.2.3. Other Anti-Proteases

Other anti-proteases that are affected after SARS-CoV-2 infection include secretory leucocyte protease inhibitor (SLPI) and elafin [157]. SLPI is one of the most important anti-proteases secreted by clear and goblet epithelial cells, submucosal glands, and leucocytes in the airways as a protective mechanism against damage [158]. In COVID-19, SLPI is released as an anti-inflammatory response after cytokine release [157]. Factors such as pulmonary surfactant A released from type II pneumocytes may contribute to its release, and infection of these cells by SARS-CoV-2 may impair these mechanisms [159,160]. In patients with COVID-19, although increased expression has been observed [157,161], it is not able to attenuate the action of proteases, such as human neutrophil elastase [157]. In COVID-19, there is an increased release of proteases, such as MMP-9 and/or elastase released from neutrophils, which may contribute to the clearance of SLPI. In severe cases of COVID-19, this intense protease activity leads to increased oxidative stress, which promotes neutrophil activation. This leads to the suppression of pathways, such as nuclear factor erythroid 2-related factor 2 (Nrf2), to poise this oxidative balance [162,163] and, consequently, an imbalance in anti-protease activity. The administration of SLPI in its recombinant form has been proposed as a therapeutic alternative to treat COVID-19 [164].
Key points:
  • Coronaviruses increase the degradation of anti-proteases, such as α-1-antitrypsin.
  • Cystatin is one of the main anti-proteases that regulate the action of cathepsins.

4. Consequences of SARS-CoV-2 Anchoring to ACE2

Cells of the respiratory epithelium are targets for more than 200 viruses, which infect them through recognition and anchoring to proteins on their plasma membrane [165]. Several binding proteins for SARS-CoV-2 have been proposed, including neurophilin-1 [166], metabotropic glutamate receptor subtype 2 (mGluR2) [167], kidney injury molecule-1 [168], heat shock protein A5 (HSPA5) or glucose-regulated protein-78 (GRP78) [169], basigin (CD147) [170], heparan sulphate [171], dipeptidyl peptidase 4/CD26 [172], GRP78, CD147 [173], and TLR4 [174]. However, ACE2 is considered the most important target [2,3] because it is the one that can best explain the pathological events that occur after infection. Its expression levels in different tissues predict the main target organs of SARS-CoV-2: digestive tract (ileum) > heart > kidney and urinary bladder > respiratory tract and lung >>> stomach and liver [175]. Specifically, ACE2 expression is much higher in alveolar tissue (type II pneumocytes) relative to the other parts of the respiratory tract, explaining the great damage the SARS-CoV-2 infection causes there [176].
With the entry of the virus through the airway, and the subsequent proteolytic cleavage of the S protein by host proteases, the S1 subunit interacts with ACE2 exposed on the surface of mucosal cells [4,15,16,23,25]. This anchoring facilitates the entry of the virus into epithelial cells. The mechanism involves internalisation of ACE2 and its degradation by a protein that regulates cholesterol: protein convertase subtilisin-kexin type 9 (PCSK9) [177]. Therefore, there is a reduction in ACE2 expression on the surface of the membranes of infected cells [178,179]. This reduction is not only due to internalisation and degradation. In addition, coronaviruses downregulate ACE2 mRNA levels in tissues, such as the lung and myocardium [180,181]. ACE2 expression levels in the lung have been associated with a protective factor against respiratory distress [182]. A partial explanation for ACE2 upregulation caused by the virus is linked to the overexpression of ADAM17 via a mechanism that is still unclear [71]. ADAM17 is involved in the removal of ACE2 ectodomains from the cytoplasmic membrane, which consequently leads to an increase in free sACE2 (Figure 1B) [73,183,184]. In addition, it also causes the release from the plasma membrane of proinflammatory factors, such as TNF-α and its receptors TNF receptor 1 and 2 [185,186]. The same is true for IL-6R: the release of sIL-6R and the formation of IL6–sIL-6R complexes, which in turn bind to gp130 that is expressed on many cell membranes, exacerbates IL-6 production through the JAK/STAT3 pathway [133,187]. Autopsy studies of patients who died from coronavirus infections revealed that ACE2+ cells, which are infected by the virus, produce the most proinflammatory cytokines [188]. Furthermore, increased accumulation of sACE2 throughout the infectious process correlates independently with mortality in SARS-CoV-2 infection [183,189,190,191]. Thus, the accumulation of sACE2 is a measure by ADAM17 activity and is related to the inflammatory process. On the other hand, ACE2 downregulation leads to an imbalance in the production of angiotensinogen-derived peptides and, consequently, an imbalance between the angiotensin II/angiotensin II receptor type 1 (AT1R) and its counterpart angiotensin 1–7/MAS receptor pathways [190,192]. In addition, ACE2 is responsible for the degradation of other peptides, such as apelin (an APJ receptor agonist) and des-Arg9-bradykinin (a bradykinin receptor B1 agonist), linking this protease to the KKS [193,194]. We will discuss the importance of bradykinin in COVID-19 later.
One of the main consequences of ACE2 downregulation is increased production of angiotensin II, which is closely correlated with the viral load [195]. Angiotensin II acts on the AT1R, which has been implicated in the control of blood pressure and electrolyte balance. However, after SARS-CoV-2 infection, AT1R exacerbates vasoconstriction, inflammation, cell proliferation, fibrosis, thrombosis, and oxidative stress via ROS production [196]. AT1R is expressed on myeloid cells (dendritic cells and macrophages), neutrophils, mononuclear cells, T and B lymphocytes, and non-immune tissue cells. The latter are the most reactive [133,197]. Their activation results in the release of inflammatory mediators, such as vascular endothelial growth factor (VEGF), prostaglandins, TNF-α, IL-1β, IL-6, IL-10, and ROS [133,198]. These actions are mediated by multiple signalling pathways, including NOX, NF-κB, ERK1/2, MAPK, and STAT1 [199,200]. These signalling pathways are the main molecular players in triggering a hyperinflammatory state (cytokine storm) and acute respiratory distress syndrome [196]. These mechanisms provide insight into why angiotensin II leads to apoptosis of the alveolar epithelium through AT1R [201].
Key points:
  • ACE2 is the most important entry target of SARS-CoV-2 since the dysfunction in its activity caused by the infection best explains the COVID-19 disease.
  • Viral entry causes an overexpression of ADAM17, a decrease in ACE2 in the plasma membrane, and an increase in its soluble forms, which causes inflammation due to the release of TNF-α and IL-6.
  • Dysregulation of ACE2 causes an increase in angiotensin II that contributes to the hyperinflammatory state, respiratory distress, and damage to the lung epithelium.

4.1. Dysregulation of ACE2 and Its Relation to Bradykinin

Kinins, including bradykinin, require the action of plasma or tissue kallikrein for their synthesis [202]. In the plasma, pre-kallikrein (Fletcher factor) is cleaved, transforming it to an enzyme with serine-protease activity. Kallikrein cleaves a pre-protein kininogen to produce high-molecular-weight kininogen (HMWK; also known as Fitzgerald factor) and bradykinin [203,204]. Tissue-derived kallikrein has different properties than plasma kallikrein [202]. It is released physiologically at lower concentrations, mainly from exocrine glands, such as the salivary glands, kidney, and pancreas [205]. In addition, it can be synthesised by the activity of polycarboxypeptidase, also known as angiotensinase C, which is constitutively expressed on the surface of endothelial cells [206]. When activated (e.g., by pathogens), this enzyme converts plasma pre-kallikrein to kallikrein. Kininogens can also be cleaved by serine proteases other than kallikreins, such as human neutrophil elastase, tryptase, cathepsins, and proteinase-3 [207,208]. These processes also increase the production of kinins, such as Lys-bradykinin from L- and H- kininogens, which are rapidly bio-transformed to bradykinin via plasma aminopeptidases [202,209,210].
One of the main functions of bradykinin is to participate in inflammation, pain, and innate immunity [211]. Kallikreins are involved in important physiological functions, such as activation of the intrinsic pathway of blood coagulation, its regulation, and fibrinolysis. In addition, bradykinin, HMWK, and FXII are involved in the contact system of innate immunity, which in turn participates in activation of the complement pathway (Figure 3) [211,212,213].
The KKS is considered an extension of the RAAS [214]. ACE has a greater affinity for bradykinin than for angiotensin II [215]. Thus, after kinin production, ACE2 preferentially degrades des-Arg9-bradykinin, whereas ACE preferentially metabolises bradykinin [193,194,216]. Considering the rapid activity of ACE, the half-life of bradykinin and Lys-bradykinin is only 27 s, so these proteins act locally. In addition, ACE bio-transforms 11% of bradykinin to des-Arg9-bradykinin, which increases its half-life ten-fold compared with bradykinin [216]. The different ACE isoforms, which have distinct activity, allow bradykinin to regulate angiotensin II activity and promote vasodilation, natriuresis, and hypotension through the bradykinin receptor B2 [217]. These ACE isoforms exert peptidylpeptidase and kinase II activities and can metabolise kinins and kallikreins (Figure 3) [218,219]. Furthermore, angiotensin II increases B2 receptor expression [220,221,222], and angiotensin II receptor type 2 stimulates the expression of angiotensinase C in endothelial cells and, thus, the production of bradykinin [223].
Regulation of the expression and activity of ACE and its isoforms is crucial. When ACE2 is downregulated, there will be a preferential increase in des-Arg9-bradykinin levels [224,225]. As mentioned previously, ACE2 is downregulated in patients with COVID-19 [178,179,183,190,191]. An increase in the ACE/ACE2 expression ratio has been associated with organ damage in these patients [224]. Moreover, Roche and Roche [226] suggested that plasma kinin concentrations should be monitored to help predict the severity of pulmonary problems. In addition, kinins have been implicated in pain and inflammation that is potentiated by plasmin [43]. Des-Arg9-bradykinin activates the B1 receptor with great affinity, leading to neutrophil recruitment, increased vascular permeability, and leucocyte extravasation into the lung [217]. Activation of this receptor leads to the release of chemokines, such as C-X-C motif chemokine 5 (CXCL5), from pulmonary epithelial cells, which in turn activates chemokine receptor type 2 (CXCR2) of neutrophils to facilitate their recruitment and infiltration into the lung and increasing inflammation [227]. In these inflammatory conditions, the B1 receptor is upregulated almost 3000-fold and the B2 receptor is upregulated 200-fold compared with the normal state, resulting in respiratory distress syndrome and multiorgan failure [216,224,225,228]. B1 and B2 receptor activity is highly sensitive to the action of cytokines and growth factors (e.g., IL-1β, IL-2, TNF-α, IFN-γ, and epidermal growth factor (EGF)) and toxins from microorganisms [216]. These effects, observed in patients with COVID-19, have also been detected in murine models with ACE2 downregulation. Furthermore, in these models, ACE2 downregulation exacerbates inflammation and pulmonary oedema via increased B1 receptor activation by elevated Des-Arg9-bradykinin levels [229]. The B2 receptor has also been linked to the production of fever, cough, bronchoconstriction, and increased airway resistance [216,230]. It is also important to note that while plasma kininogen and kallikrein concentrations are very low in healthy individuals, very high values are detected in patients with COVID-19 [216,225,228,231].
Bradykinin also increases the activity of chymase [232], an endopeptidase released by mast cells to increase angiotensin II release and thus enhance the inflammatory process [233]. Bradykinin increases renin synthesis and release through the B2 receptor and induction of prostaglandin E2 release [234]. In addition, tissue kallikreins can transform angiotensin I to angiotensin II [235]. In summary, bradykinin is substantially involved in the cytokine storm that occurs in patients with COVID-19 [225,230,236,237] and in the various symptoms that occur during the course of this disease.
Key points:
  • Bradykinin, HMWK, and FXII of coagulation form the backbone of the contact system of innate immunity and participate in complement activation.
  • The KKS is an extension of the RAAS. ACE has peptidylpeptidase and chymase II activity, so it not only metabolises angiotensin II but also kinases and kallikreins.
  • The imbalance of the ACE/ACE2 ratio is associated with an increase in des-Arg9-bradykinin, which is related to neutrophilia, inflammation, and increased lung tissue damage.
  • Des-Arg9-bradykinin is related to many of the symptomatic processes of COVID-19 (fever, cough, or bronchoconstriction).
  • By increasing chymase activity, bradykinin causes higher levels of angiotensin.

4.2. Involvement of the RAAS and the KKS in Thromboembolism

One of the most frequent causes of mortality after SARS-CoV-2 infection is the formation of micro-embolism and macro-embolism in the pulmonary and extrapulmonary vasculature [238]. The causes of these events are related to vascular endothelial and epithelial cellular dysfunction [239] that occurs after ACE2 internalisation. The phenomenon is related to three fundamental processes that occur after infection: (a) immune activation and production of proinflammatory cytokines by endothelial cells, (b) dysregulation of the RAAS, and (c) dysregulation of the KKS. In this section, we mainly discuss how RAAS and KKS dysregulation affect the thromboembolic processes.

4.2.1. Immune Activation in Thromboembolism

Patients with COVID-19 present an exacerbated inflammatory response, known as the cytokine storm [240]. Epithelial cells (especially pulmonary and vascular cells) play an important role in this phenomenon, leading to tissue damage and immuno-thrombosis. This syndrome, which has a high incidence in patients with COVID-19 (10–20%), is characterised by high morbidity (e.g., micro-embolism, macro-embolism, and/or multiorgan failure) and lethality [240]. This situation is caused by expression of the viral proteins open reading frame 3b (ORF3b), ORF6 and ORF8 in infected cells. Together with the nucleocapsid (N) protein, these proteins are involved in facilitating rapid viral replication in epithelial cells (e.g., the vascular endothelium) and delaying or suppressing the response to type I and II IFNs, a process that involves NF-κB [241]. Furthermore, ACE2 dysregulation occurs as a consequence of viral entry and the important roles that this enzyme has in immunity—including control of immune competence of myeloid cells and the clearance of peptides, such as AcSDKP, involved in activation of immunity [36,47], leading to dysregulated activity of CD8+ T lymphocytes, natural killer (NK) cells, and antigen-presenting cells. This causes a miscommunication between innate and adaptive immunity, inducing amplification of the cytokine-mediated inflammatory response, which is prolonged over time [242].
SARS-CoV-2 infection of endothelial cells [243] causes dysfunction either directly, via viral activation of signalling pathways (e.g., the S protein binds to ACE2 to cause calcium-dependent toxic effects in endothelial cells [244]), or indirectly by altering the endothelium-associated immune and inflammatory response [146,245]. This inappropriate response is also aggravated by infection of vascular basement membrane pericytes [245,246], which have high ACE2 expression [245,247]. This leads to an imbalance in the endothelium–pericyte relationship, which has consequences in the signalling and the release of proinflammatory and profibrotic factors, such as angiopoietin I or platelet-derived growth factor [161,248]. The combination of the correct production of these mediators and the degree of expression of their receptors, as well as pericyte death [249], triggers hypercoagulation, vascular permeability, oxidative stress, and the passage of toxins into adjacent tissues. This phenomenon has also been observed with SARS-CoV-2 [250], as demonstrated in autopsies of patients with COVID-19 [251].
SARS-CoV-2 also activates complement pathways [252,253,254], which are closely and reciprocally related to haemostasis. This activation may be due in part to endothelial damage and to the action of several SARS-CoV-2 structural proteins. The N protein enhances the activation of the complement lectin pathway [255], and the S protein activates the alternative complement pathway by binding to heparan sulphate on cell surfaces and to C4a [256,257]. Both complement pathways converge with activation of C3a and C5a [252,253], anaphylatoxins that increase immune cell recruitment and ROS production, perpetuate endothelial damage, and cause thrombosis [254,257,258]. C3a and C5a can stimulate the release of IL-6 and TNF from macrophages and other cells expressing the C3a/C5a receptors [259]. SARS-CoV-2 results in overexpression of these receptors, making these pathways more susceptible to activation [260]. In addition, C5a can lead to the release of tissue factor and plasminogen activator inhibitor peptide type I (PAI-1) from endothelial cells [257,260].
Key points:
  • SARS-CoV-2 causes an alteration in the endothelium–pericyte relationship, which leads to the release of inflammatory factors and cell death.
  • SARS-CoV-2, through structural proteins, activates the contact system and the complement pathway. This causes the endothelium to release factors that participate in thrombo-inflammation, such as tissue factor or PAI-1.

4.2.2. The RAAS in Thromboembolism

As mentioned previously, ACE downregulation is linked to the overexpression of ADAM17 that releases ACE2 anchored to the plasma membrane [71], increasing its soluble forms [73,189]. Since ACE2 regulates inflammation, its deregulation increases inflammation. One of the main culprits is ADAM17, which causes the release of proinflammatory factors such as TNF-α, TNF receptor 1 and 2, and IL-6R [185,186]. The release of soluble cytokine receptors and the formation of soluble complexes exacerbates the response through the JAK/STAT3 pathway [133,187]. This inflammatory state contributes to thromboembolic processes [239] and increases angiotensin II levels [195,198], which also contribute to the inflammatory response and coagulation. Angiotensin II is a prothrombotic substance, as it increases the production of PAI-1 in endothelial cells [261]. Thus, an increase in angiotensin II may contribute to the local microthrombus formation in alveolar capillaries that occurs in patients with COVID-19, as fibrin is not degraded by tissue plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) [262,263]. Finally, it is important to note once again that there is cooperation between the RAAS and the KKS. Due to RAAS deregulation, there is greater production of kinins, which also contribute to perturb haemostasis in patients with COVID-19.

4.2.3. The KKS in Thromboembolism

The main contact system components are FXII, HMWK, and pre-kallikrein, which circulate in the blood in the form of zymogens. They are activated after binding to antigenic molecules of pathogens with high structural variability, such as nucleic acids of microorganisms, NETs, and ferritin [211,264]. Numerous SARS-CoV-2 antigens (e.g., structural proteins, such as S1, N, M, and E) have a high capacity to bind to complement proteins and the contact system, leading to their activation [265,266]. Furthermore, remnants of these antigens after infection may continue to activate these innate immunity pathways, which might contribute to the symptoms in patients with long COVID [267]. The binding of these antigens to proteins of the contact system (HMWK and pre-kallikrein) triggers, through FXII, activation of the intrinsic pathway of blood coagulation [210] and the KKS [212], producing plasma kallikrein and bradykinin that provide feedback to FXII activation [268]. In addition, the contact system can be activated independently of FXII activity, including through angiotensinase C from endothelial cells [206,210] or increased release of proteases (e.g., trypsin) from glandular tissue in response to infection [61]. In COVID-19, the levels and activity of plasma kallikreins and kininogens are increased [216,225,228]. This increased kallikrein activity causes consumption of intrinsic coagulation pathway factors, an increase in the activated partial thromboplastin time [269], conversion of plasminogen to plasmin, and a state of hyperfibrinolysis [236]. Plasmin increases the production of bradykinin, contributing to an inflammatory state [270]. Bradykinin also contributes to this inflammatory and hyperfibrinolytic state through the release of tPA [271], implicating it in the coagulation imbalances that occur in patients with COVID-19 (hyperfibrinolysis or thromboembolism) [272]. In addition, plasmin and FXII can be reciprocally activated [43]. The pathological increase in FXII contributes not only to septic phenomena and blood dyscrasias, but also to fibroblast proliferation and pulmonary fibrosis [273], phenomena that occur in patients with COVID-19 [274].
Key points:
  • Angiotensin II is a factor of thrombo-inflammation in the pulmonary blood capillaries, by increasing the production of PAI-1.
  • Binding of SARS-CoV-2 antigens to the contact system activates the intrinsic coagulation pathway and KKS.
  • The increase in protease activity (e.g., kallikreins or PAR receptors) causes a state of hyperfibrinolysis through the release of tPA. This participates in septic phenomena, blood dyscrasias, fibroblast proliferation, and pulmonary fibrosis.

5. Conclusions

SARS-CoV-2 uses plasma membrane and soluble proteases in its infective mechanism. The way it enters the host cell may vary depending on which one is used (e.g., trypsin or cathepsins). In addition, proteases also participate in the replication and maturation of viral proteins, as well as the release of new virions.
Upon viral entry, proteases cleave the S protein to anchor to the host cell via ACE2. This event causes dysregulation of ACE2, increasing its soluble forms and decreasing the one anchored in the cell membrane, a process that is mediated by ADAM17. This has important consequences on inflammation.
The main consequence of ACE2 dysregulation is the increase in the ACE/ACE2 ratio. Because RAAS is an extension of KKS, the release and activity of proteases (e.g., kallikreins) will be unbalanced against the anti-proteases that regulate them. These are going to activate each other in a chain. This imbalance contributes to the infectious and proinflammatory mechanism, as well as to the imbalance of respiratory function. Furthermore, KKS is part of the contact system of innate immunity together with the complement system and activates the intrinsic coagulation pathway.
Angiotensin II and des-Arg9-bradykinin are going to be the two main final mediators of this entire chain of events. Among the consequences of this will be the affectation of the endothelium–pericyte relationship, fibroblast proliferation, the death of the vascular endothelium, hyperinflammation, and the release of procoagulant factors, which in the most severe cases cause hyperfibrinolysis, thrombosis, and sepsis. These protease-mediated mechanisms are critical to understanding COVID-19 disease. These mechanisms have not been explained in depth previously.
Aprotinin is a broad-spectrum inhibitor of the most important proteases involved in SARS-CoV-2 infection. We describe its pharmacodynamics, pharmacokinetics, toxicity, and potential for the treatment of various respiratory viruses in a second-part review, entitled “Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions” [1].

Author Contributions

J.F.P. and F.J.R.-C., study conception and design; J.F.P., F.J.R.-C. and J.M.P.-O., searching and selecting publications; J.F.P., F.J.R.-C. and J.M.P.-O., writing—original draft preparation; J.F.P., F.J.R.-C. and J.M.P.-O., writing—review and editing; J.F.P. and F.J.R.-C., supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We thank the ATAC team that participated in the study carried out in public hospitals in Castilla-La Mancha with inhaled aprotinin and those institutions from which we received support (Health Service of Castilla-La Mancha, SESCAM, and University of Castilla-La Mancha, UCLM).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Padín, J.-F.; Pérez-Ortiz, J.M.; Redondo-Calvo, F.J. Aprotinin (II): Inhalational Administration for the Treatment of COVID-19 and Other Viral Conditions. Int. J. Mol. Sci. 2024, 25, 7209. [Google Scholar] [CrossRef]
  2. Zou, X.; Chen, K.; Zou, J.; Han, P.; Hao, J.; Han, Z. Single-Cell RNA-Seq Data Analysis on the Receptor ACE2 Expression Reveals the Potential Risk of Different Human Organs Vulnerable to 2019-nCoV Infection. Front. Med. 2020, 14, 185–192. [Google Scholar] [CrossRef] [PubMed]
  3. Li, M.Y.; Li, L.; Zhang, Y.; Wang, X.S. Expression of the SARS-CoV-2 Cell Receptor Gene ACE2 in a Wide Variety of Human Tissues. Infect. Dis. Poverty 2020, 9, 45. [Google Scholar] [CrossRef] [PubMed]
  4. Shang, J.; Wan, Y.; Luo, C.; Ye, G.; Geng, Q.; Auerbach, A.; Li, F. Cell Entry Mechanisms of SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2020, 117, 11727–11734. [Google Scholar] [CrossRef] [PubMed]
  5. Wan, Y.; Shang, J.; Graham, R.; Baric, R.S.; Li, F. Receptor Recognition by the Novel Coronavirus from Wuhan: An Analysis Based on Decade-Long Structural Studies of SARS Coronavirus. J. Virol. 2020, 94, e00127-20. [Google Scholar] [CrossRef] [PubMed]
  6. Yan, R.; Zhang, Y.; Li, Y.; Xia, L.; Guo, Y.; Zhou, Q. Structural Basis for the Recognition of SARS-CoV-2 by Full-Length Human ACE2. Science 2020, 367, 1444–1448. [Google Scholar] [CrossRef] [PubMed]
  7. Wrobel, A.G.; Benton, D.J.; Xu, P.; Roustan, C.; Martin, S.R.; Rosenthal, P.B.; Skehel, J.J.; Gamblin, S.J. SARS-CoV-2 and Bat RaTG13 Spike Glycoprotein Structures Inform on Virus Evolution and Furin-Cleavage Effects. Nat. Struct. Mol. Biol. 2020, 27, 763–767. [Google Scholar] [CrossRef]
  8. Izaguirre, G. The Proteolytic Regulation of Virus Cell Entry by Furin and Other Proprotein Convertases. Viruses 2019, 11, 837. [Google Scholar] [CrossRef] [PubMed]
  9. Nagai, Y. Protease-Dependent Virus Tropism and Pathogenicity. Trends. Microbiol. 1993, 1, 81–87. [Google Scholar] [CrossRef]
  10. Hoffmann, M.; Kleine-Weber, H.; Pöhlmann, S. A Multibasic Cleavage Site in the Spike Protein of SARS-CoV-2 Is Essential for Infection of Human Lung Cells. Mol. Cell 2020, 78, 779–784.e5. [Google Scholar] [CrossRef]
  11. Coutard, B.; Valle, C.; de Lamballerie, X.; Canard, B.; Seidah, N.G.; Decroly, E. The Spike Glycoprotein of the New Coronavirus 2019-nCoV Contains a Furin-like Cleavage Site Absent in CoV of the Same Clade. Antivir. Res. 2020, 176, 104742. [Google Scholar] [CrossRef]
  12. Kam, Y.W.; Okumura, Y.; Kido, H.; Ng, L.F.; Bruzzone, R.; Altmeyer, R. Cleavage of the SARS Coronavirus Spike Glycoprotein by Airway Proteases Enhances Virus Entry into Human Bronchial Epithelial Cells in Vitro. PLoS ONE 2009, 4, e7870. [Google Scholar] [CrossRef] [PubMed]
  13. Kim, Y.; Jang, G.; Lee, D.; Kim, N.; Seon, J.W.; Kim, Y.H.; Lee, C. Trypsin Enhances SARS-CoV-2 Infection by Facilitating Viral Entry. Arch. Virol. 2022, 167, 441–458. [Google Scholar] [CrossRef] [PubMed]
  14. Fenouillet, E.; Barbouche, R.; Jones, I.M. Cell Entry by Enveloped Viruses: Redox Considerations for HIV and SARS-Coronavirus. Antioxid. Redox Signal. 2007, 9, 1009–1034. [Google Scholar] [CrossRef] [PubMed]
  15. Shulla, A.; Heald-Sargent, T.; Subramanya, G.; Zhao, J.; Perlman, S.; Gallagher, T. A Transmembrane Serine Protease Is Linked to the Severe Acute Respiratory Syndrome Coronavirus Receptor and Activates Virus Entry. J. Virol. 2011, 85, 873–882. [Google Scholar] [CrossRef]
  16. Wrapp, D.; Wang, N.; Corbett, K.S.; Goldsmith, J.A.; Hsieh, C.L.; Abiona, O.; Graham, B.S.; McLellan, J.S. Cryo-EM Structure of the 2019-nCoV Spike in the Prefusion Conformation. Science 2020, 367, 1260–1263. [Google Scholar] [CrossRef]
  17. Bhattacharyya, C.; Das, C.; Ghosh, A.; Singh, A.K.; Mukherjee, S.; Majumder, P.P.; Basu, A.; Biswas, N.K. SARS-CoV-2 Mutation 614G Creates an Elastase Cleavage Site Enhancing Its Spread in High AAT-Deficient Regions. Infect Genet. Evol. 2021, 90, 104760. [Google Scholar] [CrossRef]
  18. Bollavaram, K.; Leeman, T.H.; Lee, M.W.; Kulkarni, A.; Upshaw, S.G.; Yang, J.; Song, H.; Platt, M.O. Multiple Sites on SARS-CoV-2 Spike Protein Are Susceptible to Proteolysis by Cathepsins B, K, L, S, and V. Protein. Sci. 2021, 30, 1131–1143. [Google Scholar] [CrossRef]
  19. Ji, H.-L.; Zhao, R.; Matalon, S.; Matthay, M.A. Elevated Plasmin(Ogen) as a Common Risk Factor for COVID-19 Susceptibility. Physiol. Rev. 2020, 100, 1065–1075. [Google Scholar] [CrossRef]
  20. Oubahmane, M.; Hdoufane, I.; Bjij, I.; Lahcen, N.A.; Villemin, D.; Daoud, R.; Allali, A.E.; Cherqaoui, D. Host Cell Proteases Mediating SARS-CoV-2 Entry: An Overview. Curr. Top. Med. Chem. 2022, 22, 1776–1792. [Google Scholar] [CrossRef]
  21. Zabiegala, A.; Kim, Y.; Chang, K.-O. Roles of Host Proteases in the Entry of SARS-CoV-2. Anim. Dis. 2023, 3, 12. [Google Scholar] [CrossRef]
  22. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef]
  23. Takeda, M. Proteolytic Activation of SARS-CoV-2 Spike Protein. Microbiol. Immunol. 2022, 66, 15–23. [Google Scholar] [CrossRef]
  24. Gorący, A.; Rosik, J.; Szostak, B.; Ustianowski, Ł.; Ustianowska, K.; Gorący, J. Human Cell Organelles in SARS-CoV-2 Infection: An Up-to-Date Overview. Viruses 2022, 14, 1092. [Google Scholar] [CrossRef] [PubMed]
  25. Jackson, C.B.; Farzan, M.; Chen, B.; Choe, H. Mechanisms of SARS-CoV-2 Entry into Cells. Nat. Rev. Mol. Cell Biol. 2022, 23, 3–20. [Google Scholar] [CrossRef] [PubMed]
  26. Bayati, A.; Kumar, R.; Francis, V.; McPherson, P.S. SARS-CoV-2 Infects Cells after Viral Entry via Clathrin-Mediated Endocytosis. J. Biol. Chem. 2021, 296, 100306. [Google Scholar] [CrossRef] [PubMed]
  27. Willett, B.J.; Grove, J.; MacLean, O.A.; Wilkie, C.; De Lorenzo, G.; Furnon, W.; Cantoni, D.; Scott, S.; Logan, N.; Ashraf, S.; et al. SARS-CoV-2 Omicron Is an Immune Escape Variant with an Altered Cell Entry Pathway. Nat. Microbiol. 2022, 7, 1161–1179. [Google Scholar] [CrossRef]
  28. Pia, L.; Rowland-Jones, S. Omicron Entry Route. Nat. Rev. Immunol. 2022, 22, 144. [Google Scholar] [CrossRef]
  29. Skeggs, L.T. Discovery of the Two Angiotensin Peptides and the Angiotensin Converting Enzyme. Hypertension 1993, 21, 259–260. [Google Scholar] [CrossRef]
  30. Almeida, L.F.; Tofteng, S.S.; Madsen, K.; Jensen, B.L. Role of the Renin-Angiotensin System in Kidney Development and Programming of Adult Blood Pressure. Clin. Sci. 2020, 134, 641–656. [Google Scholar] [CrossRef]
  31. Speth, R.C.; Daubert, D.L.; Grove, K.L. Angiotensin II: A Reproductive Hormone Too? Regul. Pept. 1999, 79, 25–40. [Google Scholar] [CrossRef] [PubMed]
  32. Li, L.J.; Zhang, F.B.; Liu, S.Y.; Tian, Y.H.; Le, F.; Wang, L.Y.; Lou, H.Y.; Xu, X.R.; Huang, H.F.; Jin, F. Human Sperm Devoid of Germinal Angiotensin-Converting Enzyme Is Responsible for Total Fertilization Failure and Lower Fertilization Rates by Conventional in Vitro Fertilization. Biol. Reprod. 2014, 90, 125. [Google Scholar] [CrossRef] [PubMed]
  33. Aghsaeifard, Z.; Alizadeh, R. The Role of Angiotensin-Converting Enzyme in Immunity: Shedding Light on Experimental Findings. Endocr. Metab. Immune Disord. Drug Targets 2022, 22, 6–14. [Google Scholar] [CrossRef] [PubMed]
  34. Bernstein, K.E.; Khan, Z.; Giani, J.F.; Cao, D.Y.; Bernstein, E.A.; Shen, X.Z. Angiotensin-Converting Enzyme in Innate and Adaptive Immunity. Nat. Rev. Nephrol. 2018, 14, 325–336. [Google Scholar] [CrossRef] [PubMed]
  35. Passos-Silva, D.G.; Verano-Braga, T.; Santos, R.A. Angiotensin-(1-7): Beyond the Cardio-Renal Actions. Clin. Sci. 2013, 124, 443–456. [Google Scholar] [CrossRef] [PubMed]
  36. Bernstein, K.E.; Shen, X.Z.; Gonzalez-Villalobos, R.A.; Billet, S.; Okwan-Duodu, D.; Ong, F.S.; Fuchs, S. Different in Vivo Functions of the Two Catalytic Domains of Angiotensin-Converting Enzyme (ACE). Curr. Opin. Pharmacol. 2011, 11, 105–111. [Google Scholar] [CrossRef] [PubMed]
  37. Abadir, P.M.; Walston, J.D.; Carey, R.M. Subcellular Characteristics of Functional Intracellular Renin-Angiotensin Systems. Peptides 2012, 38, 437–445. [Google Scholar] [CrossRef] [PubMed]
  38. Shen, X.Z.; Billet, S.; Lin, C.; Okwan-Duodu, D.; Chen, X.; Lukacher, A.E.; Bernstein, K.E. The Carboxypeptidase ACE Shapes the MHC Class I Peptide Repertoire. Nat. Immunol. 2011, 12, 1078–1085. [Google Scholar] [CrossRef] [PubMed]
  39. Shen, X.Z.; Lukacher, A.E.; Billet, S.; Williams, I.R.; Bernstein, K.E. Expression of Angiotensin-Converting Enzyme Changes Major Histocompatibility Complex Class I Peptide Presentation by Modifying C Termini of Peptide Precursors. J. Biol. Chem. 2008, 283, 9957–9965. [Google Scholar] [CrossRef]
  40. Zhao, T.; Bernstein, K.E.; Fang, J.; Shen, X.Z. Angiotensin-Converting Enzyme Affects the Presentation of MHC Class II Antigens. Lab. Investig. 2017, 97, 764–771. [Google Scholar] [CrossRef]
  41. Cao, D.Y.; Saito, S.; Veiras, L.C.; Okwan-Duodu, D.; Bernstein, E.A.; Giani, J.F.; Bernstein, K.E.; Khan, Z. Role of Angiotensin-Converting Enzyme in Myeloid Cell Immune Responses. Cell Mol. Biol. Lett. 2020, 25, 31. [Google Scholar] [CrossRef] [PubMed]
  42. Jaspard, E.; Wei, L.; Alhenc-Gelas, F. Differences in the Properties and Enzymatic Specificities of the Two Active Sites of Angiotensin I-Converting Enzyme (Kininase II). Studies with Bradykinin and Other Natural Peptides. J. Biol. Chem. 1993, 268, 9496–9503. [Google Scholar] [CrossRef] [PubMed]
  43. Maas, C. Plasminflammation-An Emerging Pathway to Bradykinin Production. Front. Immunol. 2019, 10, 2046. [Google Scholar] [CrossRef] [PubMed]
  44. Khan, Z.; Shen, X.Z.; Bernstein, E.A.; Giani, J.F.; Eriguchi, M.; Zhao, T.V.; Gonzalez-Villalobos, R.A.; Fuchs, S.; Liu, G.Y.; Bernstein, K.E. Angiotensin-Converting Enzyme Enhances the Oxidative Response and Bactericidal Activity of Neutrophils. Blood 2017, 130, 328–339. [Google Scholar] [CrossRef] [PubMed]
  45. Trojanowicz, B.; Ulrich, C.; Seibert, E.; Fiedler, R.; Girndt, M. Uremic Conditions Drive Human Monocytes to Pro-Atherogenic Differentiation via an Angiotensin-Dependent Mechanism. PLoS ONE 2014, 9, e102137. [Google Scholar] [CrossRef] [PubMed]
  46. Okwan-Duodu, D.; Datta, V.; Shen, X.Z.; Goodridge, H.S.; Bernstein, E.A.; Fuchs, S.; Liu, G.Y.; Bernstein, K.E. Angiotensin-Converting Enzyme Overexpression in Mouse Myelomonocytic Cells Augments Resistance to Listeria and Methicillin-Resistant Staphylococcus Aureus. J. Biol. Chem. 2010, 285, 39051–39060. [Google Scholar] [CrossRef] [PubMed]
  47. Rieger, K.J.; Saez-Servent, N.; Papet, M.P.; Wdzieczak-Bakala, J.; Morgat, J.L.; Thierry, J.; Voelter, W.; Lenfant, M. Involvement of Human Plasma Angiotensin I-Converting Enzyme in the Degradation of the Haemoregulatory Peptide N-Acetyl-Seryl-Aspartyl-Lysyl-Proline. Biochem. J. 1993, 296 Pt 2, 373–378. [Google Scholar] [CrossRef] [PubMed]
  48. Lombard, M.N.; Sotty, D.; Wdzieczak-Bakala, J.; Lenfant, M. In Vivo Effect of the Tetrapeptide, N-Acetyl-Ser-Asp-Lys-Pro, on the G1-S Transition of Rat Hepatocytes. Cell Tissue Kinet. 1990, 23, 99–103. [Google Scholar] [CrossRef] [PubMed]
  49. Peng, H.; Carretero, O.A.; Liao, T.D.; Peterson, E.L.; Rhaleb, N.E. Role of N-Acetyl-Seryl-Aspartyl-Lysyl-Proline in the Antifibrotic and Anti-Inflammatory Effects of the Angiotensin-Converting Enzyme Inhibitor Captopril in Hypertension. Hypertension 2007, 49, 695–703. [Google Scholar] [CrossRef]
  50. Rasoul, S.; Carretero, O.A.; Peng, H.; Cavasin, M.A.; Zhuo, J.; Sanchez-Mendoza, A.; Brigstock, D.R.; Rhaleb, N.E. Antifibrotic Effect of Ac-SDKP and Angiotensin-Converting Enzyme Inhibition in Hypertension. J. Hypertens. 2004, 22, 593–603. [Google Scholar] [CrossRef]
  51. Douglas, R.G.; Ehlers, M.R.; Sturrock, E.D. Antifibrotic Peptide N-Acetyl-Ser-Asp-Lys-Pro (Ac-SDKP): Opportunities for Angiotensin-Converting Enzyme Inhibitor Design. Clin. Exp. Pharmacol. Physiol. 2013, 40, 535–541. [Google Scholar] [CrossRef]
  52. Masuyer, G.; Douglas, R.G.; Sturrock, E.D.; Acharya, K.R. Structural Basis of Ac-SDKP Hydrolysis by Angiotensin-I Converting Enzyme. Sci. Rep. 2015, 5, 13742. [Google Scholar] [CrossRef] [PubMed]
  53. Müller, L.; Jaspers, I. Epithelial Cells, the “Switchboard” of Respiratory Immune Defense Responses: Effects of Air Pollutants. Swiss. Med. Wkly. 2012, 142, w13653. [Google Scholar] [CrossRef]
  54. Fahy, J.V.; Dickey, B.F. Airway Mucus Function and Dysfunction. N. Engl. J. Med. 2010, 363, 2233–2247. [Google Scholar] [CrossRef]
  55. Knowles, M.R.; Boucher, R.C. Mucus Clearance as a Primary Innate Defense Mechanism for Mammalian Airways. J. Clin. Investig. 2002, 109, 571–577. [Google Scholar] [CrossRef]
  56. Bustamante-Marin, X.M.; Ostrowski, L.E. Cilia and Mucociliary Clearance. Cold Spring Harb. Perspect Biol. 2017, 9, a028241. [Google Scholar] [CrossRef]
  57. Aschner, Y.; Zemans, R.L.; Yamashita, C.M.; Downey, G.P. Matrix Metalloproteinases and Protein Tyrosine Kinases: Potential Novel Targets in Acute Lung Injury and ARDS. Chest 2014, 146, 1081–1091. [Google Scholar] [CrossRef] [PubMed]
  58. Kido, H.; Okumura, Y.; Takahashi, E.; Pan, H.Y.; Wang, S.; Yao, D.; Yao, M.; Chida, J.; Yano, M. Role of Host Cellular Proteases in the Pathogenesis of Influenza and Influenza-Induced Multiple Organ Failure. Biochim. Biophys. Acta 2012, 1824, 186–194. [Google Scholar] [CrossRef] [PubMed]
  59. Turk, B. Targeting Proteases: Successes, Failures and Future Prospects. Nat. Rev. Drug Discov. 2006, 5, 785–799. [Google Scholar] [CrossRef]
  60. Puente, X.S.; Sánchez, L.M.; Overall, C.M.; López-Otín, C. Human and Mouse Proteases: A Comparative Genomic Approach. Nat. Rev. Genet. 2003, 4, 544–558. [Google Scholar] [CrossRef]
  61. Szabo, R.; Wu, Q.; Dickson, R.B.; Netzel-Arnett, S.; Antalis, T.M.; Bugge, T.H. Type II Transmembrane Serine Proteases. Thromb. Haemost. 2003, 90, 185–193. [Google Scholar] [CrossRef]
  62. Rosendal, E.; Mihai, I.S.; Becker, M.; Das, D.; Frängsmyr, L.; Persson, B.D.; Rankin, G.D.; Gröning, R.; Trygg, J.; Forsell, M.; et al. Serine Protease Inhibitors Restrict Host Susceptibility to SARS-CoV-2 Infections. mBio 2022, 13, e0089222. [Google Scholar] [CrossRef] [PubMed]
  63. Greene, C.M.; McElvaney, N.G. Proteases and Antiproteases in Chronic Neutrophilic Lung Disease—Relevance to Drug Discovery. Br. J. Pharmacol. 2009, 158, 1048–1058. [Google Scholar] [CrossRef]
  64. Akaike, T.; Molla, A.; Ando, M.; Araki, S.; Maeda, H. Molecular Mechanism of Complex Infection by Bacteria and Virus Analyzed by a Model Using Serratial Protease and Influenza Virus in Mice. J. Virol. 1989, 63, 2252–2259. [Google Scholar] [CrossRef]
  65. Beppu, Y.; Imamura, Y.; Tashiro, M.; Towatari, T.; Ariga, H.; Kido, H. Human Mucus Protease Inhibitor in Airway Fluids Is a Potential Defensive Compound against Infection with Influenza A and Sendai Viruses1. J. Biochem. 1997, 121, 309–316. [Google Scholar] [CrossRef]
  66. Hennet, T.; Peterhans, E.; Stocker, R. Alterations in Antioxidant Defences in Lung and Liver of Mice Infected with Influenza A Virus. J. Gen. Virol. 1992, 73 Pt 1, 39–46. [Google Scholar] [CrossRef]
  67. Reichert, R.; Hochstrasser, K.; Werle, E. Der Proteaseninhibitorspiegel in menschlichem Nasensekret unter physiologischen und pathophysiologischen Bedingungen. Klin. Wochenschr. 1971, 49, 1234–1236. [Google Scholar] [CrossRef] [PubMed]
  68. Bojkova, D.; Klann, K.; Koch, B.; Widera, M.; Krause, D.; Ciesek, S.; Cinatl, J.; Münch, C. Proteomics of SARS-CoV-2-Infected Host Cells Reveals Therapy Targets. Nature 2020, 583, 469–472. [Google Scholar] [CrossRef]
  69. Zecha, J.; Lee, C.-Y.; Bayer, F.P.; Meng, C.; Grass, V.; Zerweck, J.; Schnatbaum, K.; Michler, T.; Pichlmair, A.; Ludwig, C.; et al. Data, Reagents, Assays and Merits of Proteomics for SARS-CoV-2 Research and Testing. Mol. Cell Proteom. 2020, 19, 1503–1522. [Google Scholar] [CrossRef] [PubMed]
  70. Garcia-Verdugo, I.; Descamps, D.; Chignard, M.; Touqui, L.; Sallenave, J.-M. Lung Protease/Anti-Protease Network and Modulation of Mucus Production and Surfactant Activity. Biochimie 2010, 92, 1608–1617. [Google Scholar] [CrossRef]
  71. Haga, S.; Yamamoto, N.; Nakai-Murakami, C.; Osawa, Y.; Tokunaga, K.; Sata, T.; Yamamoto, N.; Sasazuki, T.; Ishizaka, Y. 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. USA 2008, 105, 7809–7814. [Google Scholar] [CrossRef]
  72. Healy, E.F.; Lilic, M. A Model for COVID-19-Induced Dysregulation of ACE2 Shedding by ADAM17. Biochem. Biophys. Res. Commun. 2021, 573, 158–163. [Google Scholar] [CrossRef]
  73. Zipeto, D.; Palmeira, J.d.F.; Argañaraz, G.A.; Argañaraz, E.R. ACE2/ADAM17/TMPRSS2 Interplay May Be the Main Risk Factor for COVID-19. Front. Immunol. 2020, 11, 576745. [Google Scholar] [CrossRef]
  74. Gheblawi, M.; Wang, K.; Viveiros, A.; Nguyen, Q.; Zhong, J.-C.; Turner, A.J.; Raizada, M.K.; Grant, M.B.; Oudit, G.Y. Angiotensin-Converting Enzyme 2: SARS-CoV-2 Receptor and Regulator of the Renin-Angiotensin System: Celebrating the 20th Anniversary of the Discovery of ACE2. Circ. Res. 2020, 126, 1456–1474. [Google Scholar] [CrossRef] [PubMed]
  75. Belouzard, S.; Madu, I.; Whittaker, G.R. Elastase-Mediated Activation of the Severe Acute Respiratory Syndrome Coronavirus Spike Protein at Discrete Sites within the S2 Domain. J. Biol. Chem. 2010, 285, 22758–22763. [Google Scholar] [CrossRef] [PubMed]
  76. Matsuyama, S.; Ujike, M.; Morikawa, S.; Tashiro, M.; Taguchi, F. Protease-Mediated Enhancement of Severe Acute Respiratory Syndrome Coronavirus Infection. Proc. Natl. Acad. Sci. USA 2005, 102, 12543–12547. [Google Scholar] [CrossRef]
  77. Guizani, I.; Fourti, N.; Zidi, W.; Feki, M.; Allal-Elasmi, M. SARS-CoV-2 and Pathological Matrix Remodeling Mediators. Inflamm. Res. 2021, 70, 847–858. [Google Scholar] [CrossRef]
  78. Silva, V.; Radic, M. COVID-19 Pathology Sheds Further Light on Balance between Neutrophil Proteases and Their Inhibitors. Biomolecules 2022, 13, 82. [Google Scholar] [CrossRef] [PubMed]
  79. Geraghty, P.; Rogan, M.P.; Greene, C.M.; Boxio, R.M.; Poiriert, T.; O’Mahony, M.; Belaaouaj, A.; O’Neill, S.J.; Taggart, C.C.; McElvaney, N.G. Neutrophil Elastase Up-Regulates Cathepsin B and Matrix Metalloprotease-2 Expression. J. Immunol. 2007, 178, 5871–5878. [Google Scholar] [CrossRef]
  80. Akgun, E.; Tuzuner, M.B.; Sahin, B.; Kilercik, M.; Kulah, C.; Cakiroglu, H.N.; Serteser, M.; Unsal, I.; Baykal, A.T. Proteins Associated with Neutrophil Degranulation Are Upregulated in Nasopharyngeal Swabs from SARS-CoV-2 Patients. PLoS ONE 2020, 15, e0240012. [Google Scholar] [CrossRef]
  81. Pham, C.T. Neutrophil Serine Proteases Fine-Tune the Inflammatory Response. Int. J. Biochem. Cell Biol. 2008, 40, 1317–1333. [Google Scholar] [CrossRef] [PubMed]
  82. Szturmowicz, M.; Demkow, U. Neutrophil Extracellular Traps (NETs) in Severe SARS-CoV-2 Lung Disease. Int. J. Mol. Sci. 2021, 22, 8854. [Google Scholar] [CrossRef] [PubMed]
  83. Yang, Y.L.; Meng, F.; Qin, P.; Herrler, G.; Huang, Y.W.; Tang, Y.D. Trypsin Promotes Porcine Deltacoronavirus Mediating Cell-to-Cell Fusion in a Cell Type-Dependent Manner. Emerg. Microbes Infect. 2020, 9, 457–468. [Google Scholar] [CrossRef] [PubMed]
  84. Bussani, R.; Schneider, E.; Zentilin, L.; Collesi, C.; Ali, H.; Braga, L.; Volpe, M.C.; Colliva, A.; Zanconati, F.; Berlot, G.; et al. Persistence of Viral RNA, Pneumocyte Syncytia and Thrombosis Are Hallmarks of Advanced COVID-19 Pathology. EBioMedicine 2020, 61, 103104. [Google Scholar] [CrossRef] [PubMed]
  85. Yamazaki, E.; Yazawa, S.; Shimada, T.; Tamura, K.; Saga, Y.; Itamochi, M.; Inasaki, N.; Hasegawa, S.; Morinaga, Y.; Oishi, K.; et al. Activation of SARS-CoV-2 by Trypsin-like Proteases in the Clinical Specimens of Patients with COVID-19. Sci. Rep. 2023, 13, 11632. [Google Scholar] [CrossRef] [PubMed]
  86. Johnston, S.L.; Goldblatt, D.L.; Evans, S.E.; Tuvim, M.J.; Dickey, B.F. Airway Epithelial Innate Immunity. Front. Physiol. 2021, 12, 749077. [Google Scholar] [CrossRef] [PubMed]
  87. Takahashi, M.; Sano, T.; Yamaoka, K.; Kamimura, T.; Umemoto, N.; Nishitani, H.; Yasuoka, S. Localization of Human Airway Trypsin-like Protease in the Airway: An Immunohistochemical Study. Histochem. Cell Biol. 2001, 115, 181–187. [Google Scholar] [CrossRef]
  88. Yamaoka, K.; Masuda, K.; Ogawa, H.; Takagi, K.; Umemoto, N.; Yasuoka, S. Cloning and Characterization of the cDNA for Human Airway Trypsin-like Protease. J. Biol. Chem. 1998, 273, 11895–11901. [Google Scholar] [CrossRef] [PubMed]
  89. Laporte, M.; Naesens, L. Airway Proteases: An Emerging Drug Target for Influenza and Other Respiratory Virus Infections. Curr. Opin. Virol. 2017, 24, 16–24. [Google Scholar] [CrossRef]
  90. Anand, D.; Hummler, E.; Rickman, O.J. ENaC Activation by Proteases. Acta. Physiol. 2022, 235, e13811. [Google Scholar] [CrossRef]
  91. Brown, E.F.; Mitaera, T.; Fronius, M. COVID-19 and Liquid Homeostasis in the Lung-A Perspective through the Epithelial Sodium Channel (ENaC) Lens. Cells 2022, 11, 1801. [Google Scholar] [CrossRef] [PubMed]
  92. Gentzsch, M.; Rossier, B.C. A Pathophysiological Model for COVID-19: Critical Importance of Transepithelial Sodium Transport upon Airway Infection. Function 2020, 1, zqaa024. [Google Scholar] [CrossRef] [PubMed]
  93. Muhanna, D.; Arnipalli, S.R.; Kumar, S.B.; Ziouzenkova, O. Osmotic Adaptation by Na+-Dependent Transporters and ACE2: Correlation with Hemostatic Crisis in COVID-19. Biomedicines 2020, 8, 460. [Google Scholar] [CrossRef] [PubMed]
  94. Hou, Y.; Yu, T.; Wang, T.; Ding, Y.; Cui, Y.; Nie, H. Competitive Cleavage of SARS-CoV-2 Spike Protein and Epithelial Sodium Channel by Plasmin as a Potential Mechanism for COVID-19 Infection. Am. J. Physiol. Lung Cell Mol. Physiol. 2022, 323, L569–L577. [Google Scholar] [CrossRef] [PubMed]
  95. Carroll, E.L.; Bailo, M.; Reihill, J.A.; Crilly, A.; Lockhart, J.C.; Litherland, G.J.; Lundy, F.T.; McGarvey, L.P.; Hollywood, M.A.; Martin, S.L. Trypsin-Like Proteases and Their Role in Muco-Obstructive Lung Diseases. Int. J. Mol. Sci. 2021, 22, 5817. [Google Scholar] [CrossRef] [PubMed]
  96. Chen, S.; Zhu, Q.; Xiao, Y.; Wu, C.; Jiang, Z.; Liu, L.; Qu, J. Clinical and Etiological Analysis of Co-Infections and Secondary Infections in COVID-19 Patients: An Observational Study. Clin. Respir. J. 2021, 15, 815–825. [Google Scholar] [CrossRef] [PubMed]
  97. Chen, H.; Huang, S.; Chen, Q.; Liu, Q.; Lv, X. Trypsin May Induce Cytokine Storm in M1 Macrophages, Resulting in Critical Coronavirus Disease. Respir. Physiol. Neurobiol. 2022, 303, 103920. [Google Scholar] [CrossRef]
  98. Subramaniam, S.; Ruf, W.; Bosmann, M. Advocacy of Targeting Protease-Activated Receptors in Severe Coronavirus Disease 2019. Br. J. Pharmacol. 2022, 179, 2086–2099. [Google Scholar] [CrossRef] [PubMed]
  99. Matsushima, R.; Takahashi, A.; Nakaya, Y.; Maezawa, H.; Miki, M.; Nakamura, Y.; Ohgushi, F.; Yasuoka, S. Human Airway Trypsin-like Protease Stimulates Human Bronchial Fibroblast Proliferation in a Protease-Activated Receptor-2-Dependent Pathway. Am. J. Physiol. Lung Cell Mol. Physiol. 2006, 290, L385–L395. [Google Scholar] [CrossRef] [PubMed]
  100. Beaufort, N.; Leduc, D.; Eguchi, H.; Mengele, K.; Hellmann, D.; Masegi, T.; Kamimura, T.; Yasuoka, S.; Fend, F.; Chignard, M.; et al. The Human Airway Trypsin-like Protease Modulates the Urokinase Receptor (uPAR, CD87) Structure and Functions. Am. J. Physiol. Lung Cell Mol. Physiol. 2007, 292, L1263–L1272. [Google Scholar] [CrossRef]
  101. Vizovišek, M.; Fonović, M.; Turk, B. Cysteine Cathepsins in Extracellular Matrix Remodeling: Extracellular Matrix Degradation and Beyond. Matrix. Biol. 2019, 75–76, 141–159. [Google Scholar] [CrossRef]
  102. Vidak, E.; Javoršek, U.; Vizovišek, M.; Turk, B. Cysteine Cathepsins and Their Extracellular Roles: Shaping the Microenvironment. Cells 2019, 8, 264. [Google Scholar] [CrossRef]
  103. Berdowska, I. Cysteine Proteases as Disease Markers. Clin. Chim. Acta 2004, 342, 41–69. [Google Scholar] [CrossRef] [PubMed]
  104. Nie, X.; Qian, L.; Sun, R.; Huang, B.; Dong, X.; Xiao, Q.; Zhang, Q.; Lu, T.; Yue, L.; Chen, S.; et al. Multi-Organ Proteomic Landscape of COVID-19 Autopsies. Cell 2021, 184, 775–791.e14. [Google Scholar] [CrossRef]
  105. Pišlar, A.; Mitrović, A.; Sabotič, J.; Pečar Fonović, U.; Perišić Nanut, M.; Jakoš, T.; Senjor, E.; Kos, J. The Role of Cysteine Peptidases in Coronavirus Cell Entry and Replication: The Therapeutic Potential of Cathepsin Inhibitors. PLoS Pathog. 2020, 16, e1009013. [Google Scholar] [CrossRef] [PubMed]
  106. Zhao, M.M.; Zhu, Y.; Zhang, L.; Zhong, G.; Tai, L.; Liu, S.; Yin, G.; Lu, J.; He, Q.; Li, M.J.; et al. Novel Cleavage Sites Identified in SARS-CoV-2 Spike Protein Reveal Mechanism for Cathepsin L-Facilitated Viral Infection and Treatment Strategies. Cell Discov. 2022, 8, 53. [Google Scholar] [CrossRef] [PubMed]
  107. Iba, T.; Miki, T.; Hashiguchi, N.; Tabe, Y.; Nagaoka, I. Is the Neutrophil a “prima Donna” in the Procoagulant Process during Sepsis? Crit. Care 2014, 18, 230. [Google Scholar] [CrossRef]
  108. LaRosa, C.A.; Rohrer, M.J.; Benoit, S.E.; Rodino, L.J.; Barnard, M.R.; Michelson, A.D. Human Neutrophil Cathepsin G Is a Potent Platelet Activator. J. Vasc. Surg. 1994, 19, 306–320. [Google Scholar] [CrossRef]
  109. Lalmanach, G.; Naudin, C.; Lecaille, F.; Fritz, H. Kininogens: More than Cysteine Protease Inhibitors and Kinin Precursors. Biochimie 2010, 92, 1568–1579. [Google Scholar] [CrossRef]
  110. Rossier, B.C.; Stutts, M.J. Activation of the Epithelial Sodium Channel (ENaC) by Serine Proteases. Annu. Rev. Physiol. 2009, 71, 361–379. [Google Scholar] [CrossRef]
  111. Noori, M.; Nejadghaderi, S.A.; Sullman, M.J.M.; Carson-Chahhoud, K.; Ardalan, M.; Kolahi, A.-A.; Safiri, S. How SARS-CoV-2 Might Affect Potassium Balance via Impairing Epithelial Sodium Channels? Mol. Biol. Rep. 2021, 48, 6655–6661. [Google Scholar] [CrossRef] [PubMed]
  112. Rudensky, A.; Beers, C. Lysosomal Cysteine Proteases and Antigen Presentation. Ernst. Schering. Res. Found. Workshop. 2006, 56, 81–95. [Google Scholar] [CrossRef]
  113. Minakshi, R.; Padhan, K. The YXXΦ Motif within the Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) 3a Protein Is Crucial for Its Intracellular Transport. Virol. J. 2014, 11, 75. [Google Scholar] [CrossRef]
  114. Scarcella, M.; d’Angelo, D.; Ciampa, M.; Tafuri, S.; Avallone, L.; Pavone, L.M.; De Pasquale, V. The Key Role of Lysosomal Protease Cathepsins in Viral Infections. Int. J. Mol. Sci. 2022, 23, 9089. [Google Scholar] [CrossRef]
  115. Ghosh, S.; Dellibovi-Ragheb, T.A.; Kerviel, A.; Pak, E.; Qiu, Q.; Fisher, M.; Takvorian, P.M.; Bleck, C.; Hsu, V.W.; Fehr, A.R.; et al. β-Coronaviruses Use Lysosomes for Egress Instead of the Biosynthetic Secretory Pathway. Cell 2020, 183, 1520–1535.e14. [Google Scholar] [CrossRef] [PubMed]
  116. Buijsers, B.; Yanginlar, C.; de Nooijer, A.; Grondman, I.; Maciej-Hulme, M.L.; Jonkman, I.; Janssen, N.A.F.; Rother, N.; de Graaf, M.; Pickkers, P.; et al. Increased Plasma Heparanase Activity in COVID-19 Patients. Front. Immunol. 2020, 11, 575047. [Google Scholar] [CrossRef] [PubMed]
  117. Gomes, C.P.; Fernandes, D.E.; Casimiro, F.; da Mata, G.F.; Passos, M.T.; Varela, P.; Mastroianni-Kirsztajn, G.; Pesquero, J.B. Cathepsin L in COVID-19: From Pharmacological Evidences to Genetics. Front. Cell Infect. Microbiol. 2020, 10, 589505. [Google Scholar] [CrossRef] [PubMed]
  118. Kinaneh, S.; Khamaysi, I.; Karram, T.; Hamoud, S. Heparanase as a Potential Player in SARS-CoV-2 Infection and Induced Coagulopathy. Biosci. Rep. 2021, 41, BSR20210290. [Google Scholar] [CrossRef]
  119. Nadir, Y.; Brenner, B. Relevance of Heparan Sulfate and Heparanase to Severity of COVID-19 in the Elderly. Semin. Thromb. Hemost. 2021, 47, 348–350. [Google Scholar] [CrossRef]
  120. Drost, C.C.; Rovas, A.; Osiaevi, I.; Rauen, M.; van der Vlag, J.; Buijsers, B.; Salmenov, R.; Lukasz, A.; Pavenstädt, H.; Linke, W.A.; et al. Heparanase Is a Putative Mediator of Endothelial Glycocalyx Damage in COVID-19—A Proof-of-Concept Study. Front. Immunol. 2022, 13, 916512. [Google Scholar] [CrossRef]
  121. Masola, V.; Bellin, G.; Gambaro, G.; Onisto, M. Heparanase: A Multitasking Protein Involved in Extracellular Matrix (ECM) Remodeling and Intracellular Events. Cells 2018, 7, 236. [Google Scholar] [CrossRef] [PubMed]
  122. Sanderson, R.D.; Elkin, M.; Rapraeger, A.C.; Ilan, N.; Vlodavsky, I. Heparanase Regulation of Cancer, Autophagy and Inflammation: New Mechanisms and Targets for Therapy. FEBS J. 2017, 284, 42–55. [Google Scholar] [CrossRef] [PubMed]
  123. Jin, H.; Zhou, S. The Functions of Heparanase in Human Diseases. Mini. Rev. Med. Chem. 2017, 17, 541–548. [Google Scholar] [CrossRef] [PubMed]
  124. Simmons, G.; Gosalia, D.N.; Rennekamp, A.J.; Reeves, J.D.; Diamond, S.L.; Bates, P. Inhibitors of Cathepsin L Prevent Severe Acute Respiratory Syndrome Coronavirus Entry. Proc. Natl. Acad. Sci. USA 2005, 102, 11876–11881. [Google Scholar] [CrossRef] [PubMed]
  125. Liu, T.; Luo, S.; Libby, P.; Shi, G.P. Cathepsin L-Selective Inhibitors: A Potentially Promising Treatment for COVID-19 Patients. Pharmacol. Ther. 2020, 213, 107587. [Google Scholar] [CrossRef] [PubMed]
  126. McElvaney, O.F.; Asakura, T.; Meinig, S.L.; Torres-Castillo, J.L.; Hagan, R.S.; Gabillard-Lefort, C.; Murphy, M.P.; Thorne, L.B.; Borczuk, A.; Reeves, E.P.; et al. Protease-Anti-Protease Compartmentalization in SARS-CoV-2 ARDS: Therapeutic Implications. EBioMedicine 2022, 77, 103894. [Google Scholar] [CrossRef] [PubMed]
  127. Ren, Y.; He, Q.Y.; Fan, J.; Jones, B.; Zhou, Y.; Xie, Y.; Cheung, C.Y.; Wu, A.; Chiu, J.F.; Peiris, J.S.; et al. The Use of Proteomics in the Discovery of Serum Biomarkers from Patients with Severe Acute Respiratory Syndrome. Proteomics 2004, 4, 3477–3484. [Google Scholar] [CrossRef] [PubMed]
  128. Strnad, P.; McElvaney, N.G.; Lomas, D.A. Alpha. N. Engl. J. Med. 2020, 382, 1443–1455. [Google Scholar] [CrossRef]
  129. Azouz, N.P.; Klingler, A.M.; Callahan, V.; Akhrymuk, I.V.; Elez, K.; Raich, L.; Henry, B.M.; Benoit, J.L.; Benoit, S.W.; Noé, F.; et al. Alpha 1 Antitrypsin Is an Inhibitor of the SARS-CoV-2-Priming Protease TMPRSS2. Pathog. Immun. 2021, 6, 55–74. [Google Scholar] [CrossRef]
  130. Philippe, A.; Puel, M.; Narjoz, C.; Gendron, N.; Durey-Dragon, M.A.; Vedie, B.; Balduyck, M.; Chocron, R.; Hauw-Berlemont, C.; Sanchez, O.; et al. Imbalance between Alpha-1-Antitrypsin and Interleukin 6 Is Associated with in-Hospital Mortality and Thrombosis during COVID-19. Biochimie 2022, 202, 206–211. [Google Scholar] [CrossRef]
  131. McElvaney, O.J.; McEvoy, N.L.; McElvaney, O.F.; Carroll, T.P.; Murphy, M.P.; Dunlea, D.M.; Ní Choileáin, O.; Clarke, J.; O’Connor, E.; Hogan, G.; et al. Characterization of the Inflammatory Response to Severe COVID-19 Illness. Am. J. Respir. Crit. Care Med. 2020, 202, 812–821. [Google Scholar] [CrossRef] [PubMed]
  132. Schumertl, T.; Lokau, J.; Rose-John, S.; Garbers, C. Function and Proteolytic Generation of the Soluble Interleukin-6 Receptor in Health and Disease. Biochim. Biophys. Acta Mol. Cell Res. 2022, 1869, 119143. [Google Scholar] [CrossRef]
  133. Iwasaki, M.; Saito, J.; Zhao, H.; Sakamoto, A.; Hirota, K.; Ma, D. Inflammation Triggered by SARS-CoV-2 and ACE2 Augment Drives Multiple Organ Failure of Severe COVID-19: Molecular Mechanisms and Implications. Inflammation 2021, 44, 13–34. [Google Scholar] [CrossRef]
  134. Bai, X.; Hippensteel, J.; Leavitt, A.; Maloney, J.P.; Beckham, D.; Garcia, C.; Li, Q.; Freed, B.M.; Ordway, D.; Sandhaus, R.A.; et al. Hypothesis: Alpha-1-Antitrypsin Is a Promising Treatment Option for COVID-19. Med. Hypotheses 2021, 146, 110394. [Google Scholar] [CrossRef]
  135. Martini, F.; De Mattei, M.; Contini, C.; Tognon, M.G. Potential Use of Alpha-1 Anti-Trypsin in the Covid-19 Treatment. Front. Cell Dev. Biol. 2020, 8, 577528. [Google Scholar] [CrossRef]
  136. Turk, V.; Bode, W. The Cystatins: Protein Inhibitors of Cysteine Proteinases. FEBS Lett. 1991, 285, 213–219. [Google Scholar] [CrossRef]
  137. Tavéra, C.; Prévot, D.; Girolami, J.P.; Leung-Tack, J.; Collé, A. Tissue and Biological Fluid Distribution of Cysteine Proteinases Inhibitor: Rat Cystatin C. Biol. Chem. Hoppe Seyler 1990, 371, 187–192. [Google Scholar] [PubMed]
  138. Collins, A.R.; Grubb, A. Inhibitory Effects of Recombinant Human Cystatin C on Human Coronaviruses. Antimicrob. Agents Chemother 1991, 35, 2444–2446. [Google Scholar] [CrossRef]
  139. Collins, A.R.; Grubb, A. Cystatin D, a Natural Salivary Cysteine Protease Inhibitor, Inhibits Coronavirus Replication at Its Physiologic Concentration. Oral. Microbiol. Immunol. 1998, 13, 59–61. [Google Scholar] [CrossRef] [PubMed]
  140. Pierre, P.; Mellman, I. Developmental Regulation of Invariant Chain Proteolysis Controls MHC Class II Trafficking in Mouse Dendritic Cells. Cell 1998, 93, 1135–1145. [Google Scholar] [CrossRef]
  141. León Machado, J.A.; Steimle, V. The MHC Class II Transactivator CIITA: Not (Quite) the Odd-One-Out Anymore among NLR Proteins. Int. J. Mol. Sci. 2021, 22, 1074. [Google Scholar] [CrossRef] [PubMed]
  142. Girona-Alarcon, M.; Argüello, G.; Esteve-Sole, A.; Bobillo-Perez, S.; Burgos-Artizzu, X.P.; Bonet-Carne, E.; Mensa-Vilaró, A.; Codina, A.; Hernández-Garcia, M.; Jou, C.; et al. Low Levels of CIITA and High Levels of SOCS1 Predict COVID-19 Disease Severity in Children and Adults. iScience 2022, 25, 103595. [Google Scholar] [CrossRef] [PubMed]
  143. Bruchez, A.; Sha, K.; Johnson, J.; Chen, L.; Stefani, C.; McConnell, H.; Gaucherand, L.; Prins, R.; Matreyek, K.A.; Hume, A.J.; et al. MHC Class II Transactivator CIITA Induces Cell Resistance to Ebola Virus and SARS-like Coronaviruses. Science 2020, 370, 241–247. [Google Scholar] [CrossRef] [PubMed]
  144. Zi, M.; Xu, Y. Involvement of Cystatin C in Immunity and Apoptosis. Immunol. Lett. 2018, 196, 80–90. [Google Scholar] [CrossRef]
  145. Matuszewski, M.; Reznikov, Y.; Pruc, M.; Peacock, F.W.; Navolokina, A.; Júarez-Vela, R.; Jankowski, L.; Rafique, Z.; Szarpak, L. Prognostic Performance of Cystatin C in COVID-19: A Systematic Review and Meta-Analysis. Int. J. Environ. Res. Public Health 2022, 19, 14607. [Google Scholar] [CrossRef] [PubMed]
  146. Prasad, K.; Kulkarni, A.; Navikala, K.; Gowda, V.; Shaikh, M.A. Serum Cystatin C Levels as a Predictor of Severity and Mortality Among Patients With COVID-19 Infection. Cureus 2023, 15, e42003. [Google Scholar] [CrossRef]
  147. Zinellu, A.; Mangoni, A.A. Cystatin C, COVID-19 Severity and Mortality: A Systematic Review and Meta-Analysis. J. Nephrol. 2022, 35, 59–68. [Google Scholar] [CrossRef] [PubMed]
  148. Gupta, A.; Al-Tamimi, A.O.; Halwani, R.; Alsaidi, H.; Kannan, M.; Ahmad, F. Lipocalin-2, S100A8/A9, and Cystatin C: Potential Predictive Biomarkers of Cardiovascular Complications in COVID-19. Exp. Biol. Med. 2022, 247, 1205–1213. [Google Scholar] [CrossRef] [PubMed]
  149. Li, Y.; Yang, S.; Peng, D.; Zhu, H.M.; Li, B.Y.; Yang, X.; Sun, X.L.; Zhang, M. Predictive Value of Serum Cystatin C for Risk of Mortality in Severe and Critically Ill Patients with COVID-19. World J. Clin. Cases 2020, 8, 4726–4734. [Google Scholar] [CrossRef]
  150. Aita, A.; Battisti, I.; Contran, N.; Furlan, S.; Padoan, A.; Franchin, C.; Barbaro, F.; Cattelan, A.M.; Zambon, C.F.; Plebani, M.; et al. Salivary Proteomic Analysis in Asymptomatic and Symptomatic SARS-CoV-2 Infection: Innate Immunity, Taste Perception and FABP5 Proteins Make the Difference. Clin. Chim. Acta 2022, 537, 26–37. [Google Scholar] [CrossRef]
  151. Mohtasham, N.; Bargi, R.; Farshbaf, A.; Shahri, M.V.; Hesari, K.K.; Mohajertehran, F. Salivary Antiviral and Antibacterial Properties in the Encounter of SARS-CoV-2. Curr. Pharm. Des. 2023, 29, 2140–2148. [Google Scholar] [CrossRef] [PubMed]
  152. Muñoz-Prieto, A.; Rubić, I.; Gonzalez-Sanchez, J.C.; Kuleš, J.; Martínez-Subiela, S.; Cerón, J.J.; Bernal, E.; Torres-Cantero, A.; Vicente-Romero, M.R.; Mrljak, V.; et al. Saliva Changes in Composition Associated to COVID-19: A Preliminary Study. Sci. Rep. 2022, 12, 10879. [Google Scholar] [CrossRef] [PubMed]
  153. Larsson, A.O.; Hultström, M.; Frithiof, R.; Nyman, U.; Lipcsey, M.; Eriksson, M.B. Differential Bias for Creatinine- and Cystatin C- Derived Estimated Glomerular Filtration Rate in Critical COVID-19. Biomedicines 2022, 10, 2708. [Google Scholar] [CrossRef] [PubMed]
  154. Gren, S.T.; Janciauskiene, S.; Sandeep, S.; Jonigk, D.; Kvist, P.H.; Gerwien, J.G.; Håkansson, K.; Grip, O. The Protease Inhibitor Cystatin C Down-Regulates the Release of IL-β and TNF-α in Lipopolysaccharide Activated Monocytes. J. Leukoc. Biol. 2016, 100, 811–822. [Google Scholar] [CrossRef] [PubMed]
  155. Avotins, L.; Kroica, J.; Petersons, A.; Zentina, D.; Kravale, Z.; Saulite, A.; Racenis, K. eGFR cystatinC /eGFR Creatinine Ratio < 0.6 in Patients with SARS-CoV-2 Pneumonia: A Prospective Cohort Study. BMC Nephrol. 2023, 24, 269. [Google Scholar] [CrossRef]
  156. Liu, Y.; Xia, P.; Cao, W.; Liu, Z.; Ma, J.; Zheng, K.; Chen, L.; Li, X.; Qin, Y. Divergence between Serum Creatine and Cystatin C in Estimating Glomerular Filtration Rate of Critically Ill COVID-19 Patients. Ren. Fail. 2021, 43, 1104–1114. [Google Scholar] [CrossRef] [PubMed]
  157. Cambier, S.; Metzemaekers, M.; de Carvalho, A.C.; Nooyens, A.; Jacobs, C.; Vanderbeke, L.; Malengier-Devlies, B.; Gouwy, M.; Heylen, E.; Meersseman, P.; et al. Atypical Response to Bacterial Coinfection and Persistent Neutrophilic Bronchoalveolar Inflammation Distinguish Critical COVID-19 from Influenza. JCI Insight 2022, 7, e155055. [Google Scholar] [CrossRef]
  158. Hiemstra, P.S. Novel Roles of Protease Inhibitors in Infection and Inflammation. Biochem. Soc. Trans. 2002, 30, 116–120. [Google Scholar] [CrossRef]
  159. Melms, J.C.; Biermann, J.; Huang, H.; Wang, Y.; Nair, A.; Tagore, S.; Katsyv, I.; Rendeiro, A.F.; Amin, A.D.; Schapiro, D.; et al. A Molecular Single-Cell Lung Atlas of Lethal COVID-19. Nature 2021, 595, 114–119. [Google Scholar] [CrossRef]
  160. Ruaro, B.; Salton, F.; Braga, L.; Wade, B.; Confalonieri, P.; Volpe, M.C.; Baratella, E.; Maiocchi, S.; Confalonieri, M. The History and Mystery of Alveolar Epithelial Type II Cells: Focus on Their Physiologic and Pathologic Role in Lung. Int. J. Mol. Sci. 2021, 22, 2566. [Google Scholar] [CrossRef]
  161. Sibila, O.; Perea, L.; Albacar, N.; Moisés, J.; Cruz, T.; Mendoza, N.; Solarat, B.; Lledó, G.; Espinosa, G.; Barberà, J.A.; et al. Elevated Plasma Levels of Epithelial and Endothelial Cell Markers in COVID-19 Survivors with Reduced Lung Diffusing Capacity Six Months after Hospital Discharge. Respir. Res. 2022, 23, 37. [Google Scholar] [CrossRef]
  162. Olagnier, D.; Farahani, E.; Thyrsted, J.; Blay-Cadanet, J.; Herengt, A.; Idorn, M.; Hait, A.; Hernaez, B.; Knudsen, A.; Iversen, M.B.; et al. SARS-CoV2-Mediated Suppression of NRF2-Signaling Reveals Potent Antiviral and Anti-Inflammatory Activity of 4-Octyl-Itaconate and Dimethyl Fumarate. Nat. Commun. 2020, 11, 4938. [Google Scholar] [CrossRef]
  163. Laforge, M.; Elbim, C.; Frère, C.; Hémadi, M.; Massaad, C.; Nuss, P.; Benoliel, J.J.; Becker, C. Tissue Damage from Neutrophil-Induced Oxidative Stress in COVID-19. Nat. Rev. Immunol. 2020, 20, 515–516. [Google Scholar] [CrossRef]
  164. Mitra, P. Inhibiting Fusion with Cellular Membrane System: Therapeutic Options to Prevent Severe Acute Respiratory Syndrome Coronavirus-2 Infection. Am. J. Physiol. Cell Physiol. 2020, 319, C500–C509. [Google Scholar] [CrossRef] [PubMed]
  165. Mackie, P.L. The Classification of Viruses Infecting the Respiratory Tract. Paediatr. Respir. Rev. 2003, 4, 84–90. [Google Scholar] [CrossRef] [PubMed]
  166. Daly, J.L.; Simonetti, B.; Klein, K.; Chen, K.E.; Williamson, M.K.; Antón-Plágaro, C.; Shoemark, D.K.; Simón-Gracia, L.; Bauer, M.; Hollandi, R.; et al. Neuropilin-1 Is a Host Factor for SARS-CoV-2 Infection. Science 2020, 370, 861–865. [Google Scholar] [CrossRef] [PubMed]
  167. Wang, X.; Wen, Z.; Cao, H.; Luo, J.; Shuai, L.; Wang, C.; Ge, J.; Bu, Z.; Wang, J. Transferrin Receptor Protein 1 Cooperates with mGluR2 To Mediate the Internalization of Rabies Virus and SARS-CoV-2. J. Virol. 2023, 97, e0161122. [Google Scholar] [CrossRef]
  168. Yang, C.; Zhang, Y.; Zeng, X.; Chen, H.; Chen, Y.; Yang, D.; Shen, Z.; Wang, X.; Liu, X.; Xiong, M.; et al. Kidney Injury Molecule-1 Is a Potential Receptor for SARS-CoV-2. J. Mol. Cell Biol. 2021, 13, 185–196. [Google Scholar] [CrossRef]
  169. Elfiky, A.A. SARS-CoV-2 Spike-Heat Shock Protein A5 (GRP78) Recognition May Be Related to the Immersed Human Coronaviruses. Front. Pharmacol. 2020, 11, 577467. [Google Scholar] [CrossRef]
  170. Wang, K.; Chen, W.; Zhang, Z.; Deng, Y.; Lian, J.Q.; Du, P.; Wei, D.; Zhang, Y.; Sun, X.X.; Gong, L.; et al. CD147-Spike Protein Is a Novel Route for SARS-CoV-2 Infection to Host Cells. Signal Transduct. Target. Ther. 2020, 5, 283. [Google Scholar] [CrossRef]
  171. Shiliaev, N.; Lukash, T.; Palchevska, O.; Crossman, D.K.; Green, T.J.; Crowley, M.R.; Frolova, E.I.; Frolov, I. Natural and Recombinant SARS-CoV-2 Isolates Rapidly Evolve. J. Virol. 2021, 95, e0135721. [Google Scholar] [CrossRef] [PubMed]
  172. Vankadari, N.; Wilce, J.A. Emerging WuHan (COVID-19) Coronavirus: Glycan Shield and Structure Prediction of Spike Glycoprotein and Its Interaction with Human CD26. Emerg. Microbes Infect. 2020, 9, 601–604. [Google Scholar] [CrossRef] [PubMed]
  173. Maleksabet, H.; Rezaee, E.; Tabatabai, S.A. Host-Cell Surface Binding Targets in SARS-CoV-2 for Drug Design. Curr. Pharm. Des. 2022, 28, 3583–3591. [Google Scholar] [CrossRef] [PubMed]
  174. Aboudounya, M.M.; Heads, R.J. COVID-19 and Toll-Like Receptor 4 (TLR4): SARS-CoV-2 May Bind and Activate TLR4 to Increase ACE2 Expression, Facilitating Entry and Causing Hyperinflammation. Mediators. Inflamm. 2021, 2021, 8874339. [Google Scholar] [CrossRef]
  175. Beyerstedt, S.; Casaro, E.B.; Rangel, É. COVID-19: Angiotensin-Converting Enzyme 2 (ACE2) Expression and Tissue Susceptibility to SARS-CoV-2 Infection. Eur. J. Clin. Microbiol. Infect. Dis. 2021, 40, 905–919. [Google Scholar] [CrossRef] [PubMed]
  176. Ortiz, M.E.; Thurman, A.; Pezzulo, A.A.; Leidinger, M.R.; Klesney-Tait, J.A.; Karp, P.H.; Tan, P.; Wohlford-Lenane, C.; McCray, P.B.; Meyerholz, D.K. Heterogeneous Expression of the SARS-Coronavirus-2 Receptor ACE2 in the Human Respiratory Tract. EBioMedicine 2020, 60, 102976. [Google Scholar] [CrossRef] [PubMed]
  177. Essalmani, R.; Andréo, U.; Evagelidis, A.; Le Dévéhat, M.; Pereira Ramos, O.H.; Fruchart Gaillard, C.; Susan-Resiga, D.; Cohen, É.; Seidah, N.G. SKI-1/S1P Facilitates SARS-CoV-2 Spike Induced Cell-to-Cell Fusion via Activation of SREBP-2 and Metalloproteases, Whereas PCSK9 Enhances the Degradation of ACE2. Viruses 2023, 15, 360. [Google Scholar] [CrossRef]
  178. Verdecchia, P.; Cavallini, C.; Spanevello, A.; Angeli, F. The Pivotal Link between ACE2 Deficiency and SARS-CoV-2 Infection. Eur. J. Intern. Med. 2020, 76, 14–20. [Google Scholar] [CrossRef] [PubMed]
  179. Vieira, C.; Nery, L.; Martins, L.; Jabour, L.; Dias, R.; Simões E Silva, A.C. Downregulation of Membrane-Bound Angiotensin Converting Enzyme 2 (ACE2) Receptor Has a Pivotal Role in COVID-19 Immunopathology. Curr. Drug Targets 2021, 22, 254–281. [Google Scholar] [CrossRef]
  180. Kuba, K.; Imai, Y.; Rao, S.; Gao, H.; Guo, F.; Guan, B.; Huan, Y.; Yang, P.; Zhang, Y.; Deng, W.; et al. A Crucial Role of Angiotensin Converting Enzyme 2 (ACE2) in SARS Coronavirus-Induced Lung Injury. Nat. Med. 2005, 11, 875–879. [Google Scholar] [CrossRef]
  181. Oudit, G.Y.; Kassiri, Z.; Jiang, C.; Liu, P.P.; Poutanen, S.M.; Penninger, J.M.; Butany, J. SARS-Coronavirus Modulation of Myocardial ACE2 Expression and Inflammation in Patients with SARS. Eur. J. Clin. Investig. 2009, 39, 618–625. [Google Scholar] [CrossRef] [PubMed]
  182. Kuba, K.; Yamaguchi, T.; Penninger, J.M. Angiotensin-Converting Enzyme 2 (ACE2) in the Pathogenesis of ARDS in COVID-19. Front. Immunol. 2021, 12, 732690. [Google Scholar] [CrossRef] [PubMed]
  183. Benedetti, S.; Sisti, D.; Vandini, D.; Barocci, S.; Sudano, M.; Carlotti, E.; Teng, J.L.L.; Zamai, L. Circulating ACE2 Level and Zinc/Albumin Ratio as Potential Biomarkers for a Precision Medicine Approach to COVID-19. Adv. Biol. Regul. 2023, 89, 100973. [Google Scholar] [CrossRef] [PubMed]
  184. Oudit, G.Y.; Wang, K.; Viveiros, A.; Kellner, M.J.; Penninger, J.M. Angiotensin-Converting Enzyme 2-at the Heart of the COVID-19 Pandemic. Cell 2023, 186, 906–922. [Google Scholar] [CrossRef]
  185. Bell, J.H.; Herrera, A.H.; Li, Y.; Walcheck, B. Role of ADAM17 in the Ectodomain Shedding of TNF-Alpha and Its Receptors by Neutrophils and Macrophages. J. Leukoc. Biol. 2007, 82, 173–176. [Google Scholar] [CrossRef] [PubMed]
  186. Black, R.A.; Rauch, C.T.; Kozlosky, C.J.; Peschon, J.J.; Slack, J.L.; Wolfson, M.F.; Castner, B.J.; Stocking, K.L.; Reddy, P.; Srinivasan, S.; et al. A Metalloproteinase Disintegrin That Releases Tumour-Necrosis Factor-Alpha from Cells. Nature 1997, 385, 729–733. [Google Scholar] [CrossRef] [PubMed]
  187. Hojyo, S.; Uchida, M.; Tanaka, K.; Hasebe, R.; Tanaka, Y.; Murakami, M.; Hirano, T. How COVID-19 Induces Cytokine Storm with High Mortality. Inflamm. Regen. 2020, 40, 37. [Google Scholar] [CrossRef] [PubMed]
  188. He, L.; Ding, Y.; Zhang, Q.; Che, X.; He, Y.; Shen, H.; Wang, H.; Li, Z.; Zhao, L.; Geng, J.; 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, 288–297. [Google Scholar] [CrossRef] [PubMed]
  189. Oudit, G.Y.; Pfeffer, M.A. Plasma Angiotensin-Converting Enzyme 2: Novel Biomarker in Heart Failure with Implications for COVID-19. Eur. Heart J. 2020, 41, 1818–1820. [Google Scholar] [CrossRef]
  190. Wang, K.; Gheblawi, M.; Nikhanj, A.; Munan, M.; MacIntyre, E.; O’Neil, C.; Poglitsch, M.; Colombo, D.; Del Nonno, F.; Kassiri, Z.; et al. Dysregulation of ACE (Angiotensin-Converting Enzyme)-2 and Renin-Angiotensin Peptides in SARS-CoV-2 Mediated Mortality and End-Organ Injuries. Hypertension 2022, 79, 365–378. [Google Scholar] [CrossRef]
  191. Wang, J.; Zhao, H.; An, Y. ACE2 Shedding and the Role in COVID-19. Front. Cell Infect. Microbiol. 2021, 11, 789180. [Google Scholar] [CrossRef] [PubMed]
  192. Osman, I.O.; Melenotte, C.; Brouqui, P.; Million, M.; Lagier, J.-C.; Parola, P.; Stein, A.; La Scola, B.; Meddeb, L.; Mege, J.-L.; et al. Expression of ACE2, Soluble ACE2, Angiotensin I, Angiotensin II and Angiotensin-(1-7) Is Modulated in COVID-19 Patients. Front. Immunol. 2021, 12, 625732. [Google Scholar] [CrossRef] [PubMed]
  193. Vickers, C.; Hales, P.; Kaushik, V.; Dick, L.; Gavin, J.; Tang, J.; Godbout, K.; Parsons, T.; Baronas, E.; Hsieh, F.; et al. Hydrolysis of Biological Peptides by Human Angiotensin-Converting Enzyme-Related Carboxypeptidase. J. Biol. Chem. 2002, 277, 14838–14843. [Google Scholar] [CrossRef] [PubMed]
  194. Santos, R.A.S.; Sampaio, W.O.; Alzamora, A.C.; Motta-Santos, D.; Alenina, N.; Bader, M.; Campagnole-Santos, M.J. The ACE2/Angiotensin-(1-7)/MAS Axis of the Renin-Angiotensin System: Focus on Angiotensin-(1-7). Physiol. Rev. 2018, 98, 505–553. [Google Scholar] [CrossRef] [PubMed]
  195. Liu, M.Y.; Zheng, B.; Zhang, Y.; Li, J.P. Role and Mechanism of Angiotensin-Converting Enzyme 2 in Acute Lung Injury in Coronavirus Disease 2019. Chronic. Dis. Transl. Med. 2020, 6, 98–105. [Google Scholar] [CrossRef] [PubMed]
  196. El-Arif, G.; Khazaal, S.; Farhat, A.; Harb, J.; Annweiler, C.; Wu, Y.; Cao, Z.; Kovacic, H.; Abi Khattar, Z.; Fajloun, Z.; et al. Angiotensin II Type I Receptor (AT1R): The Gate towards COVID-19-Associated Diseases. Molecules 2022, 27, 2048. [Google Scholar] [CrossRef] [PubMed]
  197. Banu, N.; Panikar, S.S.; Leal, L.R.; Leal, A.R. Protective Role of ACE2 and Its Downregulation in SARS-CoV-2 Infection Leading to Macrophage Activation Syndrome: Therapeutic Implications. Life Sci. 2020, 256, 117905. [Google Scholar] [CrossRef] [PubMed]
  198. Mehrabadi, M.E.; Hemmati, R.; Tashakor, A.; Homaei, A.; Yousefzadeh, M.; Hemati, K.; Hosseinkhani, S. Induced Dysregulation of ACE2 by SARS-CoV-2 Plays a Key Role in COVID-19 Severity. Biomed. Pharmacother. 2021, 137, 111363. [Google Scholar] [CrossRef] [PubMed]
  199. Montecucco, F.; Pende, A.; Mach, F. The Renin-Angiotensin System Modulates Inflammatory Processes in Atherosclerosis: Evidence from Basic Research and Clinical Studies. Mediators. Inflamm. 2009, 2009, 752406. [Google Scholar] [CrossRef]
  200. Husain, K.; Hernandez, W.; Ansari, R.A.; Ferder, L. Inflammation, Oxidative Stress and Renin Angiotensin System in Atherosclerosis. World J. Biol. Chem. 2015, 6, 209–217. [Google Scholar] [CrossRef]
  201. Papp, M.; Li, X.; Zhuang, J.; Wang, R.; Uhal, B.D. Angiotensin Receptor Subtype AT(1) Mediates Alveolar Epithelial Cell Apoptosis in Response to ANG II. Am. J. Physiol. Lung Cell Mol. Physiol. 2002, 282, L713–L718. [Google Scholar] [CrossRef] [PubMed]
  202. Bhoola, K.D.; Figueroa, C.D.; Worthy, K. Bioregulation of Kinins: Kallikreins, Kininogens, and Kininases. Pharmacol. Rev. 1992, 44, 1–80. [Google Scholar] [PubMed]
  203. Kaplan, A.P.; Joseph, K.; Shibayama, Y.; Reddigari, S.; Ghebrehiwet, B. Activation of the Plasma Kinin Forming Cascade along Cell Surfaces. Int. Arch. Allergy Immunol. 2001, 124, 339–342. [Google Scholar] [CrossRef] [PubMed]
  204. Motta, G.; Shariat-Madar, Z.; Mahdi, F.; Sampaio, C.A.; Schmaier, A.H. Assembly of High Molecular Weight Kininogen and Activation of Prekallikrein on Cell Matrix. Thromb. Haemost. 2001, 86, 840–847. [Google Scholar] [PubMed]
  205. Rabito, S.F.; Orstavik, T.B.; Scicli, A.G.; Schork, A.; Carretero, O.A. Role of the Autonomic Nervous System in the Release of Rat Submandibular Gland Kallikrein into the Circulation. Circ. Res. 1983, 52, 635–641. [Google Scholar] [CrossRef] [PubMed]
  206. Yayama, K.; Kunimatsu, N.; Teranishi, Y.; Takano, M.; Okamoto, H. Tissue Kallikrein Is Synthesized and Secreted by Human Vascular Endothelial Cells. Biochim. Biophys. Acta 2003, 1593, 231–238. [Google Scholar] [CrossRef] [PubMed]
  207. Coffman, L.G.; Brown, J.C.; Johnson, D.A.; Parthasarathy, N.; D’Agostino, R.B.; Lively, M.O.; Hua, X.; Tilley, S.L.; Muller-Esterl, W.; Willingham, M.C.; et al. Cleavage of High-Molecular-Weight Kininogen by Elastase and Tryptase Is Inhibited by Ferritin. Am. J. Physiol. Lung Cell Mol. Physiol. 2008, 294, L505–L515. [Google Scholar] [CrossRef] [PubMed]
  208. Imamura, T.; Tanase, S.; Hayashi, I.; Potempa, J.; Kozik, A.; Travis, J. Release of a New Vascular Permeability Enhancing Peptide from Kininogens by Human Neutrophil Elastase. Biochem. Biophys. Res. Commun. 2002, 294, 423–428. [Google Scholar] [CrossRef] [PubMed]
  209. Marshall, P.; Heudi, O.; McKeown, S.; Amour, A.; Abou-Shakra, F. Study of Bradykinin Metabolism in Human and Rat Plasma by Liquid Chromatography with Inductively Coupled Plasma Mass Spectrometry and Orthogonal Acceleration Time-of-Flight Mass Spectrometry. Rapid. Commun. Mass. Spectrom. 2002, 16, 220–228. [Google Scholar] [CrossRef]
  210. Schmaier, A.H. The Contact Activation and Kallikrein/Kinin Systems: Pathophysiologic and Physiologic Activities. J. Thromb. Haemost. 2016, 14, 28–39. [Google Scholar] [CrossRef]
  211. Wu, Y. The Plasma Contact System as a Modulator of Innate Immunity. Curr. Opin. Hematol. 2018, 25, 389–394. [Google Scholar] [CrossRef] [PubMed]
  212. Kaplan, A.P.; Ghebrehiwet, B. The Plasma Bradykinin-Forming Pathways and Its Interrelationships with Complement. Mol. Immunol. 2010, 47, 2161–2169. [Google Scholar] [CrossRef] [PubMed]
  213. Rhaleb, N.-E.; Yang, X.-P.; Carretero, O.A. The Kallikrein-Kinin System as a Regulator of Cardiovascular and Renal Function. Compr. Physiol. 2011, 1, 971–993. [Google Scholar] [CrossRef] [PubMed]
  214. Schmaier, A.H. The Plasma Kallikrein-Kinin System Counterbalances the Renin-Angiotensin System. J. Clin. Investig. 2002, 109, 1007–1009. [Google Scholar] [CrossRef] [PubMed]
  215. Cyr, M.; Lepage, Y.; Blais, C.; Gervais, N.; Cugno, M.; Rouleau, J.L.; Adam, A. Bradykinin and Des-Arg(9)-Bradykinin Metabolic Pathways and Kinetics of Activation of Human Plasma. Am. J. Physiol. Heart Circ. Physiol. 2001, 281, H275–H283. [Google Scholar] [CrossRef]
  216. Tabassum, A.; Iqbal, M.S.; Sultan, S.; Alhuthali, R.A.; Alshubaili, D.I.; Sayyam, R.S.; Abyad, L.M.; Qasem, A.H.; Arbaeen, A.F. Dysregulated Bradykinin: Mystery in the Pathogenesis of COVID-19. Mediators. Inflamm. 2022, 2022, 7423537. [Google Scholar] [CrossRef] [PubMed]
  217. Marceau, F.; Bachelard, H.; Bouthillier, J.; Fortin, J.P.; Morissette, G.; Bawolak, M.T.; Charest-Morin, X.; Gera, L. Bradykinin Receptors: Agonists, Antagonists, Expression, Signaling, and Adaptation to Sustained Stimulation. Int. Immunopharmacol. 2020, 82, 106305. [Google Scholar] [CrossRef]
  218. Erdös, E.G.; Deddish, P.A. The Kinin System: Suggestions to Broaden Some Prevailing Concepts. Int. Immunopharmacol. 2002, 2, 1741–1746. [Google Scholar] [CrossRef] [PubMed]
  219. Lazartigues, E.; Feng, Y.; Lavoie, J.L. The Two fACEs of the Tissue Renin-Angiotensin Systems: Implication in Cardiovascular Diseases. Curr. Pharm. Des. 2007, 13, 1231–1245. [Google Scholar] [CrossRef] [PubMed]
  220. Chen, Z.; Tan, F.; Erdös, E.G.; Deddish, P.A. Hydrolysis of Angiotensin Peptides by Human Angiotensin I-Converting Enzyme and the Resensitization of B2 Kinin Receptors. Hypertension 2005, 46, 1368–1373. [Google Scholar] [CrossRef]
  221. Erdös, E.G.; Jackman, H.L.; Brovkovych, V.; Tan, F.; Deddish, P.A. Products of Angiotensin I Hydrolysis by Human Cardiac Enzymes Potentiate Bradykinin. J. Mol. Cell Cardiol. 2002, 34, 1569–1576. [Google Scholar] [CrossRef]
  222. Marcic, B.; Deddish, P.A.; Jackman, H.L.; Erdös, E.G. Enhancement of Bradykinin and Resensitization of Its B2 Receptor. Hypertension 1999, 33, 835–843. [Google Scholar] [CrossRef]
  223. Zhu, L.; Carretero, O.A.; Liao, T.D.; Harding, P.; Li, H.; Sumners, C.; Yang, X.P. Role of Prolylcarboxypeptidase in Angiotensin II Type 2 Receptor-Mediated Bradykinin Release in Mouse Coronary Artery Endothelial Cells. Hypertension 2010, 56, 384–390. [Google Scholar] [CrossRef] [PubMed]
  224. Tolouian, R.; Vahed, S.Z.; Ghiyasvand, S.; Tolouian, A.; Ardalan, M. COVID-19 Interactions with Angiotensin-Converting Enzyme 2 (ACE2) and the Kinin System; Looking at a Potential Treatment. J. Renal. Inj. Prev. 2020, 9, e19. [Google Scholar] [CrossRef]
  225. Garvin, M.R.; Alvarez, C.; Miller, J.I.; Prates, E.T.; Walker, A.M.; Amos, B.K.; Mast, A.E.; Justice, A.; Aronow, B.; Jacobson, D. A Mechanistic Model and Therapeutic Interventions for COVID-19 Involving a RAS-Mediated Bradykinin Storm. Elife 2020, 9, e59177. [Google Scholar] [CrossRef] [PubMed]
  226. Roche, J.A.; Roche, R. A Hypothesized Role for Dysregulated Bradykinin Signaling in COVID-19 Respiratory Complications. FASEB J. 2020, 34, 7265–7269. [Google Scholar] [CrossRef]
  227. Sodhi, C.P.; Wohlford-Lenane, C.; Yamaguchi, Y.; Prindle, T.; Fulton, W.B.; Wang, S.; McCray, P.B.; Chappell, M.; Hackam, D.J.; Jia, H. Attenuation of Pulmonary ACE2 Activity Impairs Inactivation of Des-Arg. Am. J. Physiol. Lung Cell Mol. Physiol. 2018, 314, L17–L31. [Google Scholar] [CrossRef]
  228. Kaplan, A.P.; Ghebrehiwet, B. Pathways for Bradykinin Formation and Interrelationship with Complement as a Cause of Edematous Lung in COVID-19 Patients. J. Allergy Clin. Immunol. 2021, 147, 507–509. [Google Scholar] [CrossRef] [PubMed]
  229. de Maat, S.; de Mast, Q.; Danser, A.H.J.; van de Veerdonk, F.L.; Maas, C. Impaired Breakdown of Bradykinin and Its Metabolites as a Possible Cause for Pulmonary Edema in COVID-19 Infection. Semin. Thromb. Hemost. 2020, 46, 835–837. [Google Scholar] [CrossRef]
  230. Ghahestani, S.-M.; Mahmoudi, J.; Hajebrahimi, S.; Sioofy-Khojine, A.-B.; Salehi-Pourmehr, H.; Sadeghi-Ghyassi, F.; Mostafaei, H. Bradykinin as a Probable Aspect in SARS-Cov-2 Scenarios: Is Bradykinin Sneaking out of Our Sight? Iran J. Allergy Asthma Immunol. 2020, 19, 13–17. [Google Scholar] [CrossRef]
  231. Martens, C.P.; Van Mol, P.; Wauters, J.; Wauters, E.; Gangnus, T.; Noppen, B.; Callewaert, H.; Feyen, J.H.M.; Liesenborghs, L.; Heylen, E.; et al. Dysregulation of the Kallikrein-Kinin System in Bronchoalveolar Lavage Fluid of Patients with Severe COVID-19. EBioMedicine 2022, 83, 104195. [Google Scholar] [CrossRef] [PubMed]
  232. Wei, C.C.; Hase, N.; Inoue, Y.; Bradley, E.W.; Yahiro, E.; Li, M.; Naqvi, N.; Powell, P.C.; Shi, K.; Takahashi, Y.; et al. Mast Cell Chymase Limits the Cardiac Efficacy of Ang I-Converting Enzyme Inhibitor Therapy in Rodents. J. Clin. Investig. 2010, 120, 1229–1239. [Google Scholar] [CrossRef] [PubMed]
  233. Dell’Italia, L.J.; Collawn, J.F.; Ferrario, C.M. Multifunctional Role of Chymase in Acute and Chronic Tissue Injury and Remodeling. Circ. Res. 2018, 122, 319–336. [Google Scholar] [CrossRef] [PubMed]
  234. Siragy, H.M.; Jaffa, A.A.; Margolius, H.S.; Carey, R.M. Renin-Angiotensin System Modulates Renal Bradykinin Production. Am. J. Physiol. 1996, 271, R1090–R1095. [Google Scholar] [CrossRef] [PubMed]
  235. Krivoy, N.; Schlüter, H.; Karas, M.; Zidek, W. Generation of Angiotensin II from Human Plasma by Tissue Kallikrein. Clin. Sci. 1992, 83, 477–482. [Google Scholar] [CrossRef] [PubMed]
  236. Colarusso, C.; Terlizzi, M.; Pinto, A.; Sorrentino, R. A Lesson from a Saboteur: High-MW Kininogen Impact in Coronavirus-induced Disease 2019. Br. J. Pharmacol. 2020, 177, 4866–4872. [Google Scholar] [CrossRef] [PubMed]
  237. van de Veerdonk, F.L.; Netea, M.G.; van Deuren, M.; van der Meer, J.W.; de Mast, Q.; Brüggemann, R.J.; van der Hoeven, H. Kallikrein-Kinin Blockade in Patients with COVID-19 to Prevent Acute Respiratory Distress Syndrome. Elife 2020, 9, e57555. [Google Scholar] [CrossRef] [PubMed]
  238. Poor, H.D. Pulmonary Thrombosis and Thromboembolism in COVID-19. Chest 2021, 160, 1471–1480. [Google Scholar] [CrossRef] [PubMed]
  239. Lim, M.S.; Mcrae, S. COVID-19 and Immunothrombosis: Pathophysiology and Therapeutic Implications. Crit. Rev. Oncol. Hematol. 2021, 168, 103529. [Google Scholar] [CrossRef]
  240. Dharra, R.; Kumar Sharma, A.; Datta, S. Emerging Aspects of Cytokine Storm in COVID-19: The Role of Proinflammatory Cytokines and Therapeutic Prospects. Cytokine 2023, 169, 156287. [Google Scholar] [CrossRef]
  241. Ramasamy, S.; Subbian, S. Critical Determinants of Cytokine Storm and Type I Interferon Response in COVID-19 Pathogenesis. Clin. Microbiol. Rev. 2021, 34, e00299-20. [Google Scholar] [CrossRef] [PubMed]
  242. Zhu, Q.; Xu, Y.; Wang, T.; Xie, F. Innate and Adaptive Immune Response in SARS-CoV-2 Infection-Current Perspectives. Front. Immunol. 2022, 13, 1053437. [Google Scholar] [CrossRef]
  243. Bernard, I.; Limonta, D.; Mahal, L.K.; Hobman, T.C. Endothelium Infection and Dysregulation by SARS-CoV-2: Evidence and Caveats in COVID-19. Viruses 2020, 13, 29. [Google Scholar] [CrossRef]
  244. Yang, K.; Liu, S.; Yan, H.; Lu, W.; Shan, X.; Chen, H.; Bao, C.; Feng, H.; Liao, J.; Liang, S.; et al. SARS-CoV-2 Spike Protein Receptor-Binding Domain Perturbates Intracellular Calcium Homeostasis and Impairs Pulmonary Vascular Endothelial Cells. Signal Transduct. Target. Ther. 2023, 8, 276. [Google Scholar] [CrossRef] [PubMed]
  245. Lui, K.O.; Ma, Z.; Dimmeler, S. SARS-CoV-2 Induced Vascular Endothelial Dysfunction: Direct or Indirect Effects? Cardiovasc. Res. 2024, 120, 34–43. [Google Scholar] [CrossRef]
  246. Rayner, S.G.; Hung, C.F.; Liles, W.C.; Altemeier, W.A. Lung Pericytes as Mediators of Inflammation. Am. J. Physiol. Lung Cell Mol. Physiol. 2023, 325, L1–L8. [Google Scholar] [CrossRef] [PubMed]
  247. Chen, L.; Li, X.; Chen, M.; Feng, Y.; Xiong, C. The ACE2 Expression in Human Heart Indicates New Potential Mechanism of Heart Injury among Patients Infected with SARS-CoV-2. Cardiovasc. Res. 2020, 116, 1097–1100. [Google Scholar] [CrossRef]
  248. Yuan, K.; Agarwal, S.; Chakraborty, A.; Condon, D.F.; Patel, H.; Zhang, S.; Huang, F.; Mello, S.A.; Kirk, O.I.; Vasquez, R.; et al. Lung Pericytes in Pulmonary Vascular Physiology and Pathophysiology. Compr. Physiol. 2021, 11, 2227–2247. [Google Scholar] [CrossRef]
  249. Cardot-Leccia, N.; Hubiche, T.; Dellamonica, J.; Burel-Vandenbos, F.; Passeron, T. Pericyte Alteration Sheds Light on Micro-Vasculopathy in COVID-19 Infection. Intensive Care Med. 2020, 46, 1777–1778. [Google Scholar] [CrossRef]
  250. Gavriilaki, E.; Anyfanti, P.; Gavriilaki, M.; Lazaridis, A.; Douma, S.; Gkaliagkousi, E. Endothelial Dysfunction in COVID-19: Lessons Learned from Coronaviruses. Curr. Hypertens. Rep. 2020, 22, 63. [Google Scholar] [CrossRef]
  251. Ackermann, M.; Verleden, S.E.; Kuehnel, M.; Haverich, A.; Welte, T.; Laenger, F.; Vanstapel, A.; Werlein, C.; Stark, H.; Tzankov, A.; et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N. Engl. J. Med. 2020, 383, 120–128. [Google Scholar] [CrossRef] [PubMed]
  252. Cugno, M.; Meroni, P.L.; Gualtierotti, R.; Griffini, S.; Grovetti, E.; Torri, A.; Lonati, P.; Grossi, C.; Borghi, M.O.; Novembrino, C.; et al. Complement Activation and Endothelial Perturbation Parallel COVID-19 Severity and Activity. J. Autoimmun. 2021, 116, 102560. [Google Scholar] [CrossRef] [PubMed]
  253. Eriksson, O.; Hultström, M.; Persson, B.; Lipcsey, M.; Ekdahl, K.N.; Nilsson, B.; Frithiof, R. Mannose-Binding Lectin Is Associated with Thrombosis and Coagulopathy in Critically Ill COVID-19 Patients. Thromb. Haemost. 2020, 120, 1720–1724. [Google Scholar] [CrossRef] [PubMed]
  254. Ramlall, V.; Thangaraj, P.M.; Meydan, C.; Foox, J.; Butler, D.; Kim, J.; May, B.; De Freitas, J.K.; Glicksberg, B.S.; Mason, C.E.; et al. Immune Complement and Coagulation Dysfunction in Adverse Outcomes of SARS-CoV-2 Infection. Nat. Med. 2020, 26, 1609–1615. [Google Scholar] [CrossRef] [PubMed]
  255. Gao, T.; Zhu, L.; Liu, H.; Zhang, X.; Wang, T.; Fu, Y.; Li, H.; Dong, Q.; Hu, Y.; Zhang, Z.; et al. Highly Pathogenic Coronavirus N Protein Aggravates Inflammation by MASP-2-Mediated Lectin Complement Pathway Overactivation. Signal Transduct. Target. Ther. 2022, 7, 318. [Google Scholar] [CrossRef]
  256. Niederreiter, J.; Eck, C.; Ries, T.; Hartmann, A.; Märkl, B.; Büttner-Herold, M.; Amann, K.; Daniel, C. Complement Activation via the Lectin and Alternative Pathway in Patients With Severe COVID-19. Front. Immunol. 2022, 13, 835156. [Google Scholar] [CrossRef]
  257. Sayyadi, M.; Hassani, S.; Shams, M.; Dorgalaleh, A. Status of Major Hemostatic Components in the Setting of COVID-19: The Effect on Endothelium, Platelets, Coagulation Factors, Fibrinolytic System, and Complement. Ann. Hematol. 2023, 102, 1307–1322. [Google Scholar] [CrossRef] [PubMed]
  258. Mastellos, D.C.; Pires da Silva, B.G.P.; Fonseca, B.A.L.; Fonseca, N.P.; Auxiliadora-Martins, M.; Mastaglio, S.; Ruggeri, A.; Sironi, M.; Radermacher, P.; Chrysanthopoulou, A.; et al. Complement C3 vs C5 Inhibition in Severe COVID-19: Early Clinical Findings Reveal Differential Biological Efficacy. Clin. Immunol. 2020, 220, 108598. [Google Scholar] [CrossRef] [PubMed]
  259. Polycarpou, A.; Howard, M.; Farrar, C.A.; Greenlaw, R.; Fanelli, G.; Wallis, R.; Klavinskis, L.S.; Sacks, S. Rationale for Targeting Complement in COVID-19. EMBO Mol. Med. 2020, 12, e12642. [Google Scholar] [CrossRef]
  260. Afzali, B.; Noris, M.; Lambrecht, B.N.; Kemper, C. The State of Complement in COVID-19. Nat. Rev. Immunol. 2022, 22, 77–84. [Google Scholar] [CrossRef]
  261. Vaughan, D.E.; Lazos, S.A.; Tong, K. Angiotensin II Regulates the Expression of Plasminogen Activator Inhibitor-1 in Cultured Endothelial Cells. A Potential Link between the Renin-Angiotensin System and Thrombosis. J. Clin. Investig. 1995, 95, 995–1001. [Google Scholar] [CrossRef] [PubMed]
  262. Nougier, C.; Benoit, R.; Simon, M.; Desmurs-Clavel, H.; Marcotte, G.; Argaud, L.; David, J.S.; Bonnet, A.; Negrier, C.; Dargaud, Y. Hypofibrinolytic State and High Thrombin Generation May Play a Major Role in SARS-COV2 Associated Thrombosis. J. Thromb. Haemost. 2020, 18, 2215–2219. [Google Scholar] [CrossRef]
  263. D’Alonzo, D.; De Fenza, M.; Pavone, V. COVID-19 and Pneumonia: A Role for the uPA/uPAR System. Drug Discov. Today 2020, 25, 1528–1534. [Google Scholar] [CrossRef] [PubMed]
  264. Maas, C.; Renné, T. Regulatory Mechanisms of the Plasma Contact System. Thromb. Res. 2012, 129 (Suppl. S2), S73–S76. [Google Scholar] [CrossRef] [PubMed]
  265. Stravalaci, M.; Pagani, I.; Paraboschi, E.M.; Pedotti, M.; Doni, A.; Scavello, F.; Mapelli, S.N.; Sironi, M.; Perucchini, C.; Varani, L.; et al. Recognition and Inhibition of SARS-CoV-2 by Humoral Innate Immunity Pattern Recognition Molecules. Nature Immunol. 2022, 23, 275–286. [Google Scholar] [CrossRef]
  266. Thomson, T.M.; Toscano-Guerra, E.; Casis, E.; Paciucci, R. C1 Esterase Inhibitor and the Contact System in COVID-19. Br. J. Haematol. 2020, 190, 520–524. [Google Scholar] [CrossRef]
  267. Savitt, A.G.; Manimala, S.; White, T.; Fandaros, M.; Yin, W.; Duan, H.; Xu, X.; Geisbrecht, B.V.; Rubenstein, D.A.; Kaplan, A.P.; et al. SARS-CoV-2 Exacerbates COVID-19 Pathology Through Activation of the Complement and Kinin Systems. Front. Immunol. 2021, 12, 767347. [Google Scholar] [CrossRef]
  268. Oehmcke-Hecht, S.; Köhler, J. Interaction of the Human Contact System with Pathogens-An Update. Front. Immunol. 2018, 9, 312. [Google Scholar] [CrossRef]
  269. Bailey, M.; Linden, D.; Guo-Parke, H.; Earley, O.; Peto, T.; McAuley, D.F.; Taggart, C.; Kidney, J. Vascular Risk Factors for COVID-19 ARDS: Endothelium, Contact-Kinin System. Front. Med. 2023, 10, 1208866. [Google Scholar] [CrossRef]
  270. Kleniewski, J.; Blankenship, D.T.; Cardin, A.D.; Donaldson, V. Mechanism of Enhanced Kinin Release from High Molecular Weight Kininogen by Plasma Kallikrein after Its Exposure to Plasmin. J. Lab. Clin. Med. 1992, 120, 129–139. [Google Scholar]
  271. Brown, N.J.; Gainer, J.V.; Stein, C.M.; Vaughan, D.E. Bradykinin Stimulates Tissue Plasminogen Activator Release in Human Vasculature. Hypertension 1999, 33, 1431–1435. [Google Scholar] [CrossRef] [PubMed]
  272. Oliveira, L.C.G.; Cruz, N.A.N.; Ricelli, B.; Tedesco-Silva, H.; Medina-Pestana, J.O.; Casarini, D.E. Interactions amongst Inflammation, Renin-Angiotensin-Aldosterone and Kallikrein-Kinin Systems: Suggestive Approaches for COVID-19 Therapy. J. Venom. Anim. Toxins Incl. Trop. Dis. 2021, 27, e20200181. [Google Scholar] [CrossRef] [PubMed]
  273. Wujak, L.; Didiasova, M.; Zakrzewicz, D.; Frey, H.; Schaefer, L.; Wygrecka, M. Heparan Sulfate Proteoglycans Mediate Factor XIIa Binding to the Cell Surface. J. Biol. Chem. 2015, 290, 7027–7039. [Google Scholar] [CrossRef]
  274. Meini, S.; Zanichelli, A.; Sbrojavacca, R.; Iuri, F.; Roberts, A.T.; Suffritti, C.; Tascini, C. Understanding the Pathophysiology of COVID-19: Could the Contact System Be the Key? Front. Immunol. 2020, 11, 2014. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The severe acute respiratory syndrome (SARS-CoV-2) infectious mechanism involves endoproteolysis of the spike (S) protein. (A) Representation of S protein cleavage (enlarged in the drawing) by transmembrane serine protease type 2 (TMPRSS2). Cleavage of the furin-like domain produces the active conformation of the S protein, exposing the S1 and S2 subunits. These subunits are responsible for anchoring to angiotensin-converting enzyme 2 (ACE2) located in the plasma membrane of the epithelial cell, and for membrane fusion and viral RNA release. Binding of the virus to ACE2 results in the activation, expression, and release of host proteases (B). Metalloproteases, such as a disintegrin and metalloproteinase 17 (ADAM17), cleave the membrane-bound receptors of tumour necrosis factor α (TNF-α) and interleukin 6 (IL-6), as well as ACE2, which are released into the extracellular medium in their respective soluble forms (sTNF receptor 1/2, sIL-6R, and sACE2, respectively). The cleavage and release exacerbate the inflammatory response and is one of the causes of the cytokine storm.
Figure 1. The severe acute respiratory syndrome (SARS-CoV-2) infectious mechanism involves endoproteolysis of the spike (S) protein. (A) Representation of S protein cleavage (enlarged in the drawing) by transmembrane serine protease type 2 (TMPRSS2). Cleavage of the furin-like domain produces the active conformation of the S protein, exposing the S1 and S2 subunits. These subunits are responsible for anchoring to angiotensin-converting enzyme 2 (ACE2) located in the plasma membrane of the epithelial cell, and for membrane fusion and viral RNA release. Binding of the virus to ACE2 results in the activation, expression, and release of host proteases (B). Metalloproteases, such as a disintegrin and metalloproteinase 17 (ADAM17), cleave the membrane-bound receptors of tumour necrosis factor α (TNF-α) and interleukin 6 (IL-6), as well as ACE2, which are released into the extracellular medium in their respective soluble forms (sTNF receptor 1/2, sIL-6R, and sACE2, respectively). The cleavage and release exacerbate the inflammatory response and is one of the causes of the cytokine storm.
Ijms 25 07553 g001
Figure 2. Somatic angiotensin-converting enzyme (ACE) with its two catalytic domains, and its main cellular functions. Somatic ACE has two catalytic domains with different affinities for protein substrates. (1) The carboxyl-terminal domain metabolises bradykinin to inactive peptides. It has greater affinity for bradykinin than it does for angiotensin I. It can regulate angiotensin activity via bradykinin metabolism. Increased angiotensin and bradykinin levels cause inflammation through the angiotensin II receptor type 1 (AT1R) and the bradykinin B1 receptor. In addition, the kallikrein–kinin system (KKS), which includes bradykinin and coagulation factor XII, together with the complement system constitute the contact system of innate immunity. (2) The amino-terminal domain of ACE degrades other peptide transmitters, such as substance P, enkephalins, and neurotensin. However, of great importance is the tetrapeptide N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP), which maintains myeloid immune cells in a quiescent state. In addition, this peptide blocks small mothers against decapentaplegic (Smad) signal transduction and the extracellular signal-regulated kinase (ERK) 1/2 pathway, which prevents cytokine formation. Thus, AcSDKP has anti-inflammatory and antifibrotic effects and is the counterpart to the effects of angiotensin and bradykinin. (3) ACE cleaves peptides that are 3–42 amino acids for antigen presentation to cluster of differentiation 8 (CD8)+ T cells by major histocompatibility complex type I and II (MHC-I and MHC-II) in antigen-presenting cells (APCs). (4) In COVID-19, ACE expression is dysregulated. ACE overexpression in myeloid cells is important for them to acquire immune competence. (5) In neutrophils, ACE actions are mediated by nicotinamide adenine dinucleotide phosphate oxidase (NOX). Overactivity of neutrophils increases oxidative stress and the release and activity of proteases, leading to the production of fibrosis and NETosis. (6) Finally, ACE dysregulation also increases the production of angiotensin II and bradykinin, which through the AT1R and the B1 receptor on myeloid cells act as one of the main chemotactic agents, contributing to cytokine storm and inflammation in COVID-19.
Figure 2. Somatic angiotensin-converting enzyme (ACE) with its two catalytic domains, and its main cellular functions. Somatic ACE has two catalytic domains with different affinities for protein substrates. (1) The carboxyl-terminal domain metabolises bradykinin to inactive peptides. It has greater affinity for bradykinin than it does for angiotensin I. It can regulate angiotensin activity via bradykinin metabolism. Increased angiotensin and bradykinin levels cause inflammation through the angiotensin II receptor type 1 (AT1R) and the bradykinin B1 receptor. In addition, the kallikrein–kinin system (KKS), which includes bradykinin and coagulation factor XII, together with the complement system constitute the contact system of innate immunity. (2) The amino-terminal domain of ACE degrades other peptide transmitters, such as substance P, enkephalins, and neurotensin. However, of great importance is the tetrapeptide N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP), which maintains myeloid immune cells in a quiescent state. In addition, this peptide blocks small mothers against decapentaplegic (Smad) signal transduction and the extracellular signal-regulated kinase (ERK) 1/2 pathway, which prevents cytokine formation. Thus, AcSDKP has anti-inflammatory and antifibrotic effects and is the counterpart to the effects of angiotensin and bradykinin. (3) ACE cleaves peptides that are 3–42 amino acids for antigen presentation to cluster of differentiation 8 (CD8)+ T cells by major histocompatibility complex type I and II (MHC-I and MHC-II) in antigen-presenting cells (APCs). (4) In COVID-19, ACE expression is dysregulated. ACE overexpression in myeloid cells is important for them to acquire immune competence. (5) In neutrophils, ACE actions are mediated by nicotinamide adenine dinucleotide phosphate oxidase (NOX). Overactivity of neutrophils increases oxidative stress and the release and activity of proteases, leading to the production of fibrosis and NETosis. (6) Finally, ACE dysregulation also increases the production of angiotensin II and bradykinin, which through the AT1R and the B1 receptor on myeloid cells act as one of the main chemotactic agents, contributing to cytokine storm and inflammation in COVID-19.
Ijms 25 07553 g002
Figure 3. The main actions of the kinin–kallikrein system (KKS). Proteases released from mucosal or myeloid cells provoke activation of factor XII (FXII → FXIIa). FXII can also be activated directly by binding to a viral antigen, as it is part of the contact system of innate immunity. Thus, it can activate the intrinsic coagulation pathway (shown in purple) or, via its serine protease action, plasma pre-kallikrein kininogen (PKK, Fletcher factor) to the kallikrein (KK) zymogen. These zymogens start a chain reaction of cleavage of other peptides to activate their proteolytic action (e.g., plasminogen or protease-activated receptors (PARs)). This process is involved in coagulation, inflammation, and activation of the complement pathway of the adhesion system of innate immunity. In this chain of events, plasma KK also produces high-molecular-weight kininogen (HMWK) and/or Fitzgerald factor, which lead to the production of bradykinin (BK) and des-Arg9-BK. PKK can also be released from tissues, which by cleavage of proteases, such as angiotensinase C, produces tissue KK that cleaves low-molecular-weight kininogen (LMWK) to produce Lys-des-Arg9-BK and kallidin (also called Lys-BK). In summary, this chain of activation from kininogens to zymogens constitutes the KKS (depicted in blue), and terminates in the production of BK, des-Arg9-BK, Lys-BK, and Lys-des-Arg9-BK (depicted in orange), each of which have greater or lesser affinity for the bradykinin B1 receptor (B1R) or bradykinin B2 receptor (B2R). Among many actions, B1R and B2R mediate inflammation, fibrosis, and oxidative stress. Furthermore, the importance of these mediators is that they are degraded to inactive peptides by kinases I and II, also known as angiotensin-converting enzyme 1 and 2 (ACE and ACE2, respectively), and by carboxypeptidases. The affinity of these enzymes for these peptides is greater than for angiotensin. Hence, the KKS is intimately related to the renin–angiotensin–aldosterone system.
Figure 3. The main actions of the kinin–kallikrein system (KKS). Proteases released from mucosal or myeloid cells provoke activation of factor XII (FXII → FXIIa). FXII can also be activated directly by binding to a viral antigen, as it is part of the contact system of innate immunity. Thus, it can activate the intrinsic coagulation pathway (shown in purple) or, via its serine protease action, plasma pre-kallikrein kininogen (PKK, Fletcher factor) to the kallikrein (KK) zymogen. These zymogens start a chain reaction of cleavage of other peptides to activate their proteolytic action (e.g., plasminogen or protease-activated receptors (PARs)). This process is involved in coagulation, inflammation, and activation of the complement pathway of the adhesion system of innate immunity. In this chain of events, plasma KK also produces high-molecular-weight kininogen (HMWK) and/or Fitzgerald factor, which lead to the production of bradykinin (BK) and des-Arg9-BK. PKK can also be released from tissues, which by cleavage of proteases, such as angiotensinase C, produces tissue KK that cleaves low-molecular-weight kininogen (LMWK) to produce Lys-des-Arg9-BK and kallidin (also called Lys-BK). In summary, this chain of activation from kininogens to zymogens constitutes the KKS (depicted in blue), and terminates in the production of BK, des-Arg9-BK, Lys-BK, and Lys-des-Arg9-BK (depicted in orange), each of which have greater or lesser affinity for the bradykinin B1 receptor (B1R) or bradykinin B2 receptor (B2R). Among many actions, B1R and B2R mediate inflammation, fibrosis, and oxidative stress. Furthermore, the importance of these mediators is that they are degraded to inactive peptides by kinases I and II, also known as angiotensin-converting enzyme 1 and 2 (ACE and ACE2, respectively), and by carboxypeptidases. The affinity of these enzymes for these peptides is greater than for angiotensin. Hence, the KKS is intimately related to the renin–angiotensin–aldosterone system.
Ijms 25 07553 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Padín, J.F.; Pérez-Ortiz, J.M.; Redondo-Calvo, F.J. Aprotinin (I): Understanding the Role of Host Proteases in COVID-19 and the Importance of Pharmacologically Regulating Their Function. Int. J. Mol. Sci. 2024, 25, 7553. https://doi.org/10.3390/ijms25147553

AMA Style

Padín JF, Pérez-Ortiz JM, Redondo-Calvo FJ. Aprotinin (I): Understanding the Role of Host Proteases in COVID-19 and the Importance of Pharmacologically Regulating Their Function. International Journal of Molecular Sciences. 2024; 25(14):7553. https://doi.org/10.3390/ijms25147553

Chicago/Turabian Style

Padín, Juan Fernando, José Manuel Pérez-Ortiz, and Francisco Javier Redondo-Calvo. 2024. "Aprotinin (I): Understanding the Role of Host Proteases in COVID-19 and the Importance of Pharmacologically Regulating Their Function" International Journal of Molecular Sciences 25, no. 14: 7553. https://doi.org/10.3390/ijms25147553

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop