Next Article in Journal
Molecular Mechanisms of Inhibition of Protein Amyloid Fibril Formation: Evidence and Perspectives Based on Kinetic Models
Next Article in Special Issue
Lipoprotein(a): Cardiovascular Disease, Aortic Stenosis and New Therapeutic Option
Previous Article in Journal
Advances in Intraoperative Flow Cytometry
Previous Article in Special Issue
Deciphering the Basis of Molecular Biology of Selected Cardiovascular Diseases: A View on Network Medicine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Proprotein Convertase Subtilisin/Kexin 6 in Cardiovascular Biology and Disease

1
Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
2
Vascular Function Laboratory, Human Aging Research Institute and School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(21), 13429; https://doi.org/10.3390/ijms232113429
Submission received: 6 September 2022 / Revised: 1 November 2022 / Accepted: 2 November 2022 / Published: 3 November 2022
(This article belongs to the Special Issue Molecular Mechanisms and Pathophysiology of Cardiovascular Diseases)

Abstract

:
Proprotein convertase subtilisin/kexin 6 (PCSK6) is a secreted serine protease expressed in most major organs, where it cleaves a wide range of growth factors, signaling molecules, peptide hormones, proteolytic enzymes, and adhesion proteins. Studies in Pcsk6-deficient mice have demonstrated the importance of Pcsk6 in embryonic development, body axis specification, ovarian function, and extracellular matrix remodeling in articular cartilage. In the cardiovascular system, PCSK6 acts as a key modulator in heart formation, lipoprotein metabolism, body fluid homeostasis, cardiac repair, and vascular remodeling. To date, dysregulated PCSK6 expression or function has been implicated in major cardiovascular diseases, including atrial septal defects, hypertension, atherosclerosis, myocardial infarction, and cardiac aging. In this review, we describe biochemical characteristics and posttranslational modifications of PCSK6. Moreover, we discuss the role of PCSK6 and related molecular mechanisms in cardiovascular biology and disease.

1. Introduction

Proteolytic cleavage is a key mechanism in regulating protein structure and function. The proprotein convertase subtilisin/kexin (PCSK) family consists of nine proteolytic enzymes that process a variety of proteins, including peptide hormones, growth factors, cell receptors, proteases, and adhesion molecules [1,2]. The PCSK-mediated pathways are essential for tissue homeostasis and physiological function. PCSKs are also exploited by pathogens to boost their infectivity. In COVID-19, for example, PCSK-mediated cleavage of spike protein on the surface of severe acute respiratory syndrome (SARS) coronavirus-2 particles is a crucial step for viral entry in human airways [3,4].
PCSK6, also known as paired basic amino acid cleaving enzyme 4 (PACE4) or subtilisin-like proprotein convertase 4 (SPC4), is a member of the PCSK family. PCSK6-mediated protein cleavage has been implicated in diverse biological processes, ranging from embryonic development to tissue senescence. Human genetic and animal model studies in recent years indicate that PCSK6 is an important regulator in cardiovascular biology and disease. In this review, we briefly describe the current knowledge regarding PCSK6 expression, biochemical and cellular mechanisms in zymogen activation, and protein substrate specificity. We also examine findings in Pcsk6-deficient mice under various conditions. Finally, we focus our discussion on the role of PCSK6 in heart development and aging, lipoprotein metabolism, blood pressure, and cardiovascular remodeling.

2. PCSK6 Gene and Tissue Expression

PCSK6 was identified from human hepatoma cells based on sequence similarities to other subtilisin-like proteases [5]. The human PCSK6 gene is located on chromosome 15 at q26.3, with 23 exons in a locus spanning ~228 kb [6]. PCSK6 orthologues are conserved in all vertebrates from fish to primates, an indication of functional significance.
PCSK6 mRNA expression has been detected in virtually all major organs, including the brain, heart, lung, liver, spleen, pancreas, kidney, skeletal muscle, uterus, ovary, and placenta [7]. Within the heart, high levels of PCSK6 mRNA are expressed in atrial and ventricular cardiomyocytes [8]. Based on proteomic analysis of human tissues, PCSK6 is also expressed in cardiac fibroblasts, endothelial cells, and smooth muscle cells (SMCs) [9]. It remains unclear if PCSK6 cleaves similar sets of proteins in different cardiac cell types.
In addition to normal tissues, upregulated PCSK6 expression has been found in many cancers, including lung cancer [10,11], breast cancer [12,13], prostate cancer [14,15], skin cancer [16,17], ovarian cancer [18,19], and thyroid cancer [20]. To date, PCSK6-mediated cleavage of metalloproteinases [17,21], signaling molecules [22,23], and apoptotic proteins [23,24] has been reported as a potential mechanism in tumor invasion and migration. In animal cancer models, PCSK6 inhibitors have been shown to inhibit tumor progression, indicating that PCSK6 is a potential cancer target [16,25,26]. In this review, our discussions focus on the role of PCSK6 in cardiovascular biology. More information on PCSK6 and cancers can be found in several comprehensive reviews [27,28,29].

3. Protein Domains and Post-Translational Modifications

The full-length human PCSK6 (NCBI protein accession number: NP_002561.1) consists of 969 amino acids, with a calculated mass of ~100 kDa. There are PCSK6 isoforms with shorter sequences from alternative mRNA slicing [6,14]. In cancer tissues, PCSK6 isoforms may differ from those in normal tissues [20]. Unlike furin, which has a transmembrane domain [1], PCSK6 is a secreted protein, consisting of an N-terminal signal peptide, a pro-domain, a subtilisin-like catalytic domain, a P or Homo B domain, and a C-terminal cysteine-rich domain [30] (Figure 1). Within the catalytic domain, the conserved active residues Asp, His, and Ser are at positions 205, 246, and 420, respectively.
PCSK6 is synthesized in a one-chain zymogen form. After the signal peptide is removed by signal peptidase in the endoplasmic reticulum (ER), PCSK6 undergoes a two-step autoactivation process, which resembles those in furin and PC5A activation [31,32]. The first autocleavage at RVKR149↓ between the pro-domain and the catalytic domain occurs in the ER [33] (Figure 2). The cleaved pro-domain is attached to the remaining fragment, thereby blocking PCSK6 catalytic activity. Moreover, the cleaved pro-domain acts as an intramolecular chaperone in protein folding and subsequent ER exiting [34]. Upon reaching the cell surface, PCSK6 binds to membrane-associated heparan sulfate proteoglycans and undergoes another autocleavage at KR117↓ within the pro-domain (Figure 2) [32]. This second cleavage removes the pro-domain, converting PCSK6 to a fully active enzyme [32].
The secretion and activation of PCSK6 are regulated by its own structural elements and interactions with other intracellular proteins. Within the cell, for example, association with reticulocalbin-3, a protein in the secretory pathway, promotes PCSK6 secretion and autoactivation [35]. The C-terminal cysteine-rich domain is inhibitory for PCSK6 intracellular trafficking. Deletion of a C-terminal segment or the entire domain increases PCSK6 secretion and autoactivation [36,37]. In addition to heparan sulfate proteoglycans on the cell surface [32,38], PCSK6 also binds to heparan sulfate proteoglycans in the extracellular matrix where it may activate matrix metalloproteinases [39]. In the cell-derived conditioned medium, PCSK6 also binds to heparin, which enhances PCSK6 activity [39]. The heparin-binding site on PCSK6 is in a positively charged segment, between residues 734 and 760, within the C-terminal cysteine-rich domain [39]. There is no evidence to indicate that PCSK6 binds to chondroitin sulfate [32,39]. To date, the heparan sulfate structures required for PCSK6 binding have not been determined experimentally [39]. Further studies will be important to define the biochemical and cellular mechanisms underlying PCSK6 intracellular trafficking and secretion.
In human PCSK6, there are three predicted N-glycosylation sites at N259, N914, and N932, respectively (Figure 1). These sites are conserved in PCSK6 homologs among mammalian species, with dogs being an exception, which have only two predicted N-glycosylation sites (corresponding to N259 and N932 in humans, respectively). In trypsin-like serine proteases, N-glycosylation facilitates protein folding, intracellular trafficking, cell surface expression, or secretion [40,41,42,43]. N-glycan-mediated interactions with calnexin, an ER chaperon, are critical for protein folding and ER exiting [41,44]. To date, the significance of N-glycosylation in PCSK6 remains unclear. Additional studies are required to verify if those predicted sites are N-glycosylated and to understand the potential role of N-glycans in PCSK6 biosynthesis and function.

4. Protein Substrates

Proteases break peptide bonds at preferred amino acid sequences. PCSK6 cleaves at sequences with paired basic residues, e.g., R-X-K/R-R, where X can be any amino acid, and K/R-R or non-paired basic residues, e.g., R-X-X-R [45,46]. These recognition sequences, however, are not unique for PCSK6, as many PCSKs exhibit similar substrate specificities in biochemical and cellular experiments [1]. Under physiological conditions, the function of individual PCSKs also depends on their spatial and temporal expression and regulatory mechanisms in specific tissue environments.
To date, numerous proteins have been reported as potential PCSK6 substrates. A partial list of the candidate substrates includes growth factors [47,48], cell surface receptors [49,50], blood clotting factors [51,52], matrix metalloproteinases (MMPs) [17,21], serine proteases [53,54], lysyl oxidase-like 2 [55], group X secretory phospholipase A2 [56], growth differentiation factor 15 [57], Nodal [58], bone morphogenetic protein-4 (BMP-4) [58,59], neural and hepatic peptides [60,61], bacterial toxins [45,62], and viral proteins [63,64]. These findings suggest a potential role of PCSK6 in diverse pathophysiological processes, ranging from embryonic development to tissue homeostasis to bacterial and viral infection. It is noted that the majority of the reported PCSK6 substrates are based on in vitro experiments. Many of the substrates are also cleaved by other PCSKs in similar experimental settings. It remains to be determined if the observed overlapping activities reflect the functional redundancy among PCSKs in vivo.

5. Findings from Pcsk6 Knockout (KO) Mice

5.1. Embryonic Development

In mice, Pcsk6 deficiency leads to lethality in ~25% embryos, which exhibit severe defects in heart formation, craniofacial patterning, and left–right axis specification by gestational day 15.5 [65]. The phenotype is primarily due to impaired processing and signaling of transforming growth factor beta (TGFβ) superfamily members, including Nodal, Bmp4, and Lefty, that are crucial in body axis specification during embryonic development [65,66,67]. These findings are consistent with the observed PCSK6 activity in processing TGFβ-like growth factors in vitro [59,65].
The incomplete penetrance of the embryonic lethality in Pcsk6 KO mice suggests that the Pcsk6 function in embryogenesis may be partially compensated by other PCSKs, particularly furin that exhibits similar Nodal and BMP4 processing activity in vitro and in vivo [58,59]. Moreover, both PCSK6 and furin are shown to bind to Cripto, a glycosylphosphatidylinositol (GPI)-anchored proteoglycan that localizes Nodal processing on the surface of embryonic cells [68]. It remains unclear how PCSK6 and furin activities are regulated, temporally and spatially, in embryonic tissues to achieve the overlapping but not identical function.

5.2. Age-Dependent Decline in Ovarian Function

The PCSK6 function in processing TGFβ-like growth factors appears dispensable for postnatal survival. The remaining three quarters of Pcsk6 KO mice that are born grow normally [65,69]. Both male and female Pcsk6 KO mice are fertile [65]. No major defects in liver and kidney function, blood coagulation, immune response, and tooth formation have been reported in these mice.
Studies in aged Pcsk6 KO mice, however, indicate a key role of Pcsk6 in preserving ovarian structure and function [69]. Female Pcsk6 KO mice at or older than nine months exhibit decreased estrus cyclicity, prolonged whelping intervals, increased serum follicle-stimulating hormone levels, and ovarian atrophy with less or no follicles [69]. Given the findings of PCSK6 expression in human and mouse ovarian granulosa cells and oocytes [70,71,72], the reproductive senescence in female Pcsk6 KO mice is likely related to impaired processing of Pcsk6 substrate(s) in the ovary. To date, such substrate(s) have yet to be defined. Among TGFβ superfamily members, oocyte-derived growth differentiation factor 9 (Gdf9) and Bmp15 are essential for folliculogenesis in mice [73]. In human genetic studies, GDF9 and BMP15 variants are found in women with premature ovarian failure and amenorrhea [74,75,76], suggesting that GDF9 and BMP15 could be potential PCSK6 substrates in the ovary.

5.3. Aggrecan Degradation in Articular Cartilage

Analysis of Pcsk6 KO mice substantiates a role of PCSK6 in aggrecan degradation and osteoarthritis [77]. Aggrecan is a proteoglycan with abundant chondroitin and keratan sulfate chains [78,79]. It aggregates on hyaluronan filaments in the extracellular matrix of articular cartilage, protecting the joint from the impact of compressive forces [78,79]. Increased aggrecan degradation is a key factor in the pathogenesis of osteoarthritis [80]. Members of the ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family, mainly ADAMTS-4 and ADAMTS-5, are major aggrecan-degrading enzymes in osteoarthritis [78,80].
In human osteoarthritic cartilage, PCSK6 has been identified as a key activator of ADAMTS-4 and ADAMTS-5 [81]. In mice, Pcsk6 deficiency alleviates pain in osteoarthritis models [77]. A PCSK6 variant allele is also associated with similar pain protection in patients with osteoarthritic knees [77]. Moreover, increased PCSK6 expression is found in synovial tissues in patients with rheumatoid arthritis [82]. PCSK6 inhibition reduces inflammatory responses in rat and human synoviocytes associated with rheumatoid arthritis [82,83,84]. These results highlight a crucial role for PCSK6 in cartilage remodeling and destruction under pathological conditions.

6. PCSK6 and Cardiovascular Pathophysiology

6.1. Atrial Septal Defects

Atrial septal defects are a common form of congenital heart disease [85]. As discussed earlier, Pcsk6 KO mice exhibit a plethora of cardiac abnormalities, including common atrium, double-outlet right ventricle, ventricular septal defects, persistent truncus arteriosus, and dextrocardia [65]. Common atrium is a severe type of atrial septal defect, in which the entire atrial septum is missing. In mice, Nodal and Bmp4 are downstream effectors of Pcsk6 in heart formation [65,66]. However, the upstream molecular network that regulates PCSK6 expression in developing hearts is not well defined.
TBX5, NKX2-5, and GATA4 are major transcription factors in heart development. Deleterious TBX5, NKX2-5, and GATA4 mutations are found in individuals with congenital heart disease, including atrial septal defects [86,87]. Protein odd-skipped-related 1 (OSR1) is a zinc-finger transcription factor, essential for cardiac progenitor growth and atrial septum formation [88]. In mouse embryonic hearts, Tbx5 is an immediate Osr1 upstream gene in the second heart field for atrial septation [89]. Osr1 deletion results in common atrium and embryonic death between E11.5 and E12.5 days [90]. The similar phenotype of atrial septal defects in Tbx5-, Osr1-, and Pcsk6-deficient mice suggests a possible TBX5-OSR1-PCSK6 pathway in promoting TGFβ-like growth factor signaling in atrial septum formation.
In agreement with this hypothesis, gene profiling in Tbx5- and Osr1-deficient embryos identifies Pcsk6 as one of the major genes in atrial septation regulated by Tbx5 and Osr1 [91]. Human genome-wide linkage analysis also suggests a connection between PCSK6 and congenital heart disease [92]. Moreover, a PCSK6 variant is found in a Spanish family with atrial septal defects and interatrial septal aneurysm [91]. These findings indicate that TBX5 and OSR1 are possible regulators in PCSK6 expression during heart development, providing new insights into the genetic mechanisms underlying atrial septal defects.

6.2. Endothelial Lipase Inactivation

Endothelial lipase, a member of the triglyceride lipase family, is a secreted protein, consisting of a signal peptide, a 40 kDa N-terminal catalytic domain, and a 28 kDa C-terminal domain [93]. Upon secretion, endothelial lipase binds to heparan sulfate proteoglycans on the cell surface, where it hydrolyzes triglycerides and phospholipids in plasma lipoproteins [94]. In addition to endothelial cells, endothelial lipase is expressed in hepatocytes, macrophages, and SMCs. Endothelial lipase modifies high-density lipoprotein (HDL) structure and metabolism, as indicated by high levels of plasma HDL cholesterol, bigger HDL particle sizes, and slower HDL clearance in endothelial lipase-deficient mice [95,96]. Conversely, plasma HDL cholesterol levels are decreased in endothelial lipase overexpressing mice [96,97]. In humans, deleterious variants in the LIPG gene, encoding endothelial lipase, are associated with increased plasma HDL cholesterol levels [98,99,100].
In membrane-bound serine proteases, ectodomain cleavage is a common mechanism in limiting protease activity on the cell surface [101,102]. A comparable mechanism exists in endothelial lipase inactivation on the cell surface. In the conditioned medium from human endothelial cells, a 40 kDa endothelial lipase fragment was detected [46,97]. A similar fragment was also found from human liver HepG2 cell culture [103]. As revealed by biochemical analyses, the fragment is derived by cleavage at a specific site, RNKR↓, which reduces the endothelial lipase activity [46,103]. Furin, PC5A, and PCSK6 are likely responsible for the cleavage [46,103]. Interestingly, lipoprotein lipase, another member of the triglyceride lipase family, contains an analogous site, RAKR↓, which is also cleaved by furin, PC5A, and PCSK6 in similar experiments [46]. These data indicate that PCSK-mediated endothelial lipase inactivation is a cellular mechanism in regulating lipoprotein metabolism.
Consistently, hepatic overexpression of pro-furin, an inhibitor of furin, PCSK5, and PCSK6 reduce endothelial lipase inactivation and lower plasma HDL cholesterol in mice [104]. In Lipg KO mice, such an effect of pro-furin on plasma HDL cholesterol is not observed, supporting a PCSK-endothelial lipase-dependent mechanism in HDL metabolism. Studies in those mice also validate a role of PCSK-mediated activation of an endogenous endothelial lipase inhibitor, angiopoietin-like 3 [104,105]. Further analyses in hepatocyte-specific Furin and Pcsk5 conditional KO and Pcsk6 global KO mice show that hepatic furin is primarily responsible for cleavage of endothelial lipase and angiopoietin-like 3 in vivo [106]. However, plasma levels of HDL cholesterol are only slightly reduced in the hepatocyte Furin conditional KO mice or not changed in the hepatocyte Pcsk5 conditional and Pcsk6 global KO mice, compared to that in WT mice [106]. These findings suggest functional redundancy among PCSKs in endothelial lipase inactivation, at least in mice.

6.3. Corin Activation and Hypertension

Atrial and B-type natriuretic peptides (ANP and BNP, respectively) are hormones in the natriuretic peptide system that preserves body fluid balance and cardiovascular homeostasis [107]. Genetic studies in mice and humans establish ANP as a key factor in blood pressure regulation [108,109,110]. Upon binding to its receptor, natriuretic peptide receptor A (also called guanylate cyclase A), ANP enhances renal salt excretion and relaxes blood vessels, thereby lowering blood volume and pressure. Variants in the NPPA gene, encoding ANP, are associated with increased risks of cardiovascular disease, such as hypertension, stroke, and heart disease [111,112].
Corin is a membrane-bound protease, highly expressed in the heart [113], where it converts pro-ANP to ANP [114,115,116]. Like most proteases, corin is produced in a pro-form, which is activated at a specific site, RMNKR↓ [117]. The cleavage sequence with paired basic residues indicates that corin is likely activated by one of the PCSKs. Indeed, PCSK6 has been identified as the corin activator [53]. Both PCSK6 and corin are expressed in cardiomyocytes, where PCSK6 activates corin on the cell surface [53,118]. In cultured murine cardiomyocytes, blocking Pcsk6 expression prevents corin activation [53]. In Pcsk6-deficient mice, corin activation and pro-ANP processing in the heart are eliminated [53]. Like Corin KO mice, Pcsk6-deficient mice develop salt-sensitive hypertension [53,119], indicating that PCSK6 is the corin activator in vivo and that this function cannot be substituted by other PCSKs.
In line with these findings, genetic studies support a role of PCSK6 in corin activation and cardiovascular function in humans. For example, several CORIN variants identified in hypertensive patients are defective in PCSK6-mediated activation [53,120]. PCSK6 variants are associated with hypertension [53,120] and coronary artery stenosis [121]. Studies in humans and rat models also indicate an important PCSK6-corin-ANP pathway in regulating renal aquaporin 2 and β-epithelial sodium channel expression in response to a high-salt diet [122], consistent with salt-sensitive hypertension in Pcsk6, Corin, and Nppa KO mice [53,109,119,123]. Moreover, reduced cardiac and renal PCSK6 and corin expression correlates with worsening cardiac and renal function in a rat heart failure model [124]. These data highlight the importance of PCSK6 in corin activation and body fluid-electrolyte homeostasis.

6.4. Vascular Remodeling in Atherosclerosis

Atherosclerosis is a major vascular disease, characterized by the formation of atherosclerotic plaques in the intima of medium- to large-sized arteries [125]. Depending on disease stages, the plaque usually contains lipid-packed macrophages, also called foam cells, and SMCs that are surrounded by accumulated extracellular matrix proteins and proteoglycans [125]. As the disease progresses, the macrophages and SMCs undergo apoptosis, creating a highly thrombotic necrotic core that is prone to rupture, thereby causing thrombosis formation [125]. To date, several lines of evidence point to a potential role of PCSK6 in regulating SMC migration, vascular remodeling, and atherosclerotic plaque formation.
In patients with aortic dissections in a Korean population, for example, genomic alternations are found in a locus where the PCSK6 gene is located [126]. Genome-wide expression analysis indicates elevated PCSK6 expression in atherosclerotic plaques [127,128]. In cultured human monocytes and endothelial cells, PCSK6 expression and activity are increased by pro-atherogenic lipid oxidation products [129]. In Pcsk6 KO mice, compromised vascular remodeling, as indicated by enlarged systolic and diastolic circumferences and reduced contractile SMC markers, is observed in carotid arteries exposed to increased blood flow [130]. Increased PCSK6 expression is also detected in smooth muscle α-actin (SMA) (an SMC marker) -positive cells in unstable carotid plaques, where inflammation and extracellular matrix degradation are active [131]. Moreover, PCSK6 expression in cultured human carotid SMCs is increased by proinflammatory factors, such as tumor necrosis factor and interferon-γ [131]. These findings suggest a connection between PCSK6 and SMC-derived cells in the vessel wall where inflammation and pathological remodeling occur.
Consistently, a recent human study links a PCSK6 variant with SMA-positive cell numbers in carotid stenosis lesions and artery wall thickness [132]. In human and rodent carotid arteries, increased PCSK6 expression correlates with SMC activation, intimal hyperplasia, and MMP2/MMP14 activation [132]. Conversely, decreased intimal hyperplasia and MMP14 activation are found in Pcsk6 KO mice with carotid artery ligation [132]. Moreover, aortic SMCs from Pcsk6 KO mice exhibit poor proliferation and migration induced by platelet-derived growth factor BB (PGDFBB), whereas in human SMCs overexpressing PCSK6, PDGFBB-stimulated cell proliferation and migration are increased [132]. PCSK6 is known to activate MMPs in cancers [133]. The latest findings suggest that PCSK6-mediated MMP activation may be important in SMC phenotypic changes and pathological vascular remodeling in atherosclerosis.

6.5. Cardiac Repair after Myocardial Infarction (MI)

MI triggers a series of cellular events, including cell death, inflammatory cell infiltration, and gradual wound healing with myofibroblast proliferation and ultimate scar formation [134]. Tissue remodeling depends on the interplay among various cell types, including immune cells, cardiomyocytes, fibroblasts, and vascular cells. Both autocrine and paracrine mechanisms are involved in cell–cell interactions in infarcted hearts [134].
Many serine proteases have been implicated in cardiac structure and function [135]. In a recent study, PCSK6 was identified as one of the highly secreted proteins from hypoxic cardiomyocytes [136]. The finding is confirmed in mouse hearts undergoing coronary artery ligation [136]. The PCSK6 expression and secretion in hypoxic cardiomyocytes promote TGFβ secretion from the same cells and subsequent SMAD (small and mothers against decapentapletic) signaling in cardiac fibroblasts [136]. Moreover, high levels of collagen I production and fibrosis-related gene expression (e.g., Col1a1, Col3a1, and Mmp14) are observed in cardiac fibroblasts treated with the PCSK6-containing conditioned medium from hypoxic cardiomyocytes [136]. These findings suggest that upregulated PCSK6 in ischemic cardiomyocytes activates TGFβ, which, in turn, binds to its receptor on cardiac fibroblasts, thereby enhancing downstream SMAD signaling to promote collagen production and cardiac fibrosis [136].
Increased fibrosis is a hallmark of poor cardiac remodeling, which impairs cardiac function. Consistently, PCSK6 overexpression in cardiomyocytes increases cardiac hypertrophy and fibrosis and decreases cardiac function in a mouse MI model [136]. Moreover, increased serum PCSK6 levels are observed in patients with acute MI, which peaks on day 3 post incidence [136]. Previously, increased ventricular, but not atrial, Pcsk6 expression was noticed post MI in a rat model [137]. These data support a role of PCSK6 in a paracrine mechanism, underlying cardiac remodeling after MI. In another study [138], serum PCSK6 levels were associated with cardiovascular events in a subset of patients undergoing coronary angiography. Further studies will be important to evaluate if serum PCSK6 can be used as a biomarker to predict cardiac remodeling and function in patients with heart disease.

6.6. Cardiac Senescence

In aging hearts, altered protein expression and signaling often lead to deteriorating cardiac structure and function. In addition to apoptosis, senescence is a common feature in aging cardiomyocytes, as indicated by DNA damage, dysregulated gene expression, increased oxidative stress, mitochondrial dysfunction, and poor contractility [139,140]. Natriuretic peptide-mediated signaling is critical in cardiomyocyte homeostasis [111]. In humans, variants in the NPPA gene are associated with impaired cardiovascular responsiveness in the elderly [141]. In rodents, decreased ANP secretion is found in aging hearts and senescent cardiomyocytes in culture [142,143].
PCSK6 is necessary for corin activation and ANP generation in the heart [53]. A recent study indicates that PCSK6 deficiency may contribute to senescence in cardiomyocytes [144]. In aged mouse hearts and senescent cardiomyocytes, Pcsk6 expression is reduced. Moreover, Pcsk6 downregulation causes senescence in cultured cardiomyocytes, as indicated by increased advanced glycation end products, oxidative stress, and apoptosis [144]. Conversely, Pcsk6 overexpression prevents senescence and dysfunction in cultured cardiomyocytes under similar experimental conditions [144].
The function of PCSK6 in cardiomyocyte senescence appears mediated, at least in part, by pathways related to ER stress. In aging mouse hearts and Pcsk6 knockdown cardiomyocytes, high levels of DNA-damage inducible transcript 3 (Ddit3) are observed [144]. Ddit3, also called C/EBP homologous protein, is a pro-apoptotic transcription factor inducted by ER stress [145]. In cardiomyocytes subjected to ER stress, Ddit3 expression is suppressed by PCSK6 expression [144], suggesting that PCSK6 may regulate cardiomyocyte senescence by reducing ER stress via a DDIT3-related mechanism. Consistent with these findings, increased ER stress is reported in human prostate cancer cells, in which the PCSK6 gene is downregulated [24]. In a mouse model of heart failure, Ddit3 deletion prevents ER-stress-induced cell death and cardiac dysfunction [146]. As discussed earlier, premature ovarian senescence is observed in Pcsk6 KO mice [69]. It will be important to examine if similar premature aging exists in other major organs in Pcsk6 KO mice.

7. Conclusions

PCSK6 is a multifunctional protease that acts in diverse tissues to modulate many pathophysiological processes, ranging from embryonic development to organ aging. Despite the apparent overlapping substrate specificity among PCSKs in biochemical and cellular studies, PCSK6 exhibits distinct physiological functions that are not fully compensated by other PCSKs, as indicated by findings in Pcsk6-deficient mice. The unique function of PCSK6 is likely achieved by mechanisms that regulate PCSK6 activity in specific cellular environments during various stages of life. To date, such regulatory mechanisms have yet to be fully understood.
PCSKs are important in the cardiovascular system. PCSK9, for example, is a key regulator in LDL receptor expression and lipid metabolism [147]. Currently, PCSK9 inhibitors are used to treat patients with familial hypercholesterolemia [147]. By acting on various growth factors and proteases, PCSK6 too plays a key role in cardiovascular development and homeostasis (Figure 3). To date, PCSK6 has been implicated in major cardiovascular diseases, such as atrial septal defects, hypertension, atherosclerosis, MI, and cardiac aging. More investigations are anticipated to determine if modulating PCSK6 expression and/or activity is a valid therapeutic strategy for cardiovascular disease.

Author Contributions

Q.W.: conceptualization and original draft preparation; Q.W. and S.C.: discussion, manuscript revision, and approval of the published version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported, in part, by grants from the National Natural Science Foundation of China (81873566 and 82071592) and the Priority Academic Program Development of Jiangsu Higher Education Institutes.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

Q.W. and S.C. are inventors in a PCSK6-related patent that is owned by the Cleveland Clinic in Cleveland, OH, USA.

References

  1. Seidah, N.G.; Prat, A. The biology and therapeutic targeting of the proprotein convertases. Nat. Rev. Drug Discov. 2012, 11, 367–383. [Google Scholar] [CrossRef] [PubMed]
  2. Shakya, M.; Lindberg, I. Mouse Models of Human Proprotein Convertase Insufficiency. Endocr. Rev. 2021, 42, 259–294. [Google Scholar] [CrossRef] [PubMed]
  3. Seidah, N.G.; Pasquato, A.; Andréo, U. How Do Enveloped Viruses Exploit the Secretory Proprotein Convertases to Regulate Infectivity and Spread? Viruses 2021, 13, 1229. [Google Scholar] [CrossRef] [PubMed]
  4. 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]
  5. Kiefer, M.C.; Tucker, J.E.; Joh, R.; Landsberg, K.E.; Saltman, D.; Barr, P.J. Identification of a second human subtilisin-like protease gene in the fes/fps region of chromosome 15. DNA Cell Biol. 1991, 10, 757–769. [Google Scholar] [CrossRef]
  6. Tsuji, A.; Hine, C.; Tamai, Y.; Yonemoto, K.; Yoshida, S.; Bando, M.; Sakai, E.; Mori, K.; Akamatsu, T.; Matsuda, Y. Genomic organization and alternative splicing of human PACE4 (SPC4), kexin-like processing endoprotease. J. Biochem. 1997, 122, 438–452. [Google Scholar] [CrossRef]
  7. Karlsson, M.; Zhang, C.; Méar, L.; Zhong, W.; Digre, A.; Katona, B.; Sjöstedt, E.; Butler, L.; Odeberg, J.; Dusart, P.; et al. A single-cell type transcriptomics map of human tissues. Sci. Adv. 2021, 7, eabh2169. [Google Scholar] [CrossRef]
  8. Beaubien, G.; Schäfer, M.K.; Weihe, E.; Dong, W.; Chrétien, M.; Seidah, N.G.; Day, R. The distinct gene expression of the pro-hormone convertases in the rat heart suggests potential substrates. Cell Tissue Res. 1995, 279, 539–549. [Google Scholar] [CrossRef]
  9. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Proteomics. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef]
  10. Mbikay, M.; Sirois, F.; Yao, J.; Seidah, N.G.; Chrétien, M. Comparative analysis of expression of the proprotein convertases furin, PACE4, PC1 and PC2 in human lung tumours. Br. J. Cancer 1997, 75, 1509–1514. [Google Scholar] [CrossRef]
  11. Lin, Y.E.; Wu, Q.N.; Lin, X.D.; Li, G.Q.; Zhang, Y.J. Expression of paired basic amino acid-cleaving enzyme 4 (PACE4) correlated with prognosis in non-small cell lung cancer (NSCLC) patients. J. Thorac. Dis. 2015, 7, 850–860. [Google Scholar] [PubMed]
  12. Patel, A.; García-Closas, M.; Olshan, A.F.; Perou, C.M.; Troester, M.A.; Love, M.I.; Bhattacharya, A. Gene-Level Germline Contributions to Clinical Risk of Recurrence Scores in Black and White Patients with Breast Cancer. Cancer Res. 2022, 82, 25–35. [Google Scholar] [CrossRef] [PubMed]
  13. Panet, F.; Couture, F.; Kwiatkowska, A.; Desjardins, R.; Guérin, B.; Day, R. PACE4 is an important driver of ZR-75-1 estrogen receptor-positive breast cancer proliferation and tumor progression. Eur. J. Cell Biol. 2017, 96, 469–475. [Google Scholar] [CrossRef] [PubMed]
  14. Couture, F.; Wang, L.; Dufour, F.; Chabot-Maheux, K.; Ekindi Ndongo, N.; Sabbagh, R.; Day, R. PACE4-altCT isoform of proprotein convertase PACE4 as tissue and plasmatic biomarker for prostate cancer. Sci. Rep. 2022, 12, 6066. [Google Scholar] [CrossRef]
  15. Bakrania, A.; Aubé, M.; Desjardins, R.; Sabbagh, R.; Day, R. Upregulation of PACE4 in prostate cancer is not dependent on E2F transcription factors. Can. J. Physiol. Pharmacol. 2020, 98, 477–481. [Google Scholar] [CrossRef]
  16. Weishaupt, C.; Mastrofrancesco, A.; Metze, D.; Kemper, B.; Stegemann, A.; Picardo, M.; Klein-Szanto, A.J.; Böhm, M. Paired Basic Amino Acid-cleaving Enzyme 4 (PCSK6): An Emerging New Target Molecule in Human Melanoma. Acta Derm. Venereol. 2020, 100, adv00157. [Google Scholar] [CrossRef]
  17. Bassi, D.E.; Lopez De Cicco, R.; Cenna, J.; Litwin, S.; Cukierman, E.; Klein-Szanto, A.J. PACE4 expression in mouse basal keratinocytes results in basement membrane disruption and acceleration of tumor progression. Cancer Res. 2005, 65, 7310–7319. [Google Scholar] [CrossRef] [Green Version]
  18. Chen, J.; Li, Y.; Wu, J.; Liu, Y.; Kang, S. Whole-exome sequencing reveals potential germline and somatic mutations in 60 malignant ovarian germ cell tumors. Biol. Reprod. 2021, 105, 164–178. [Google Scholar] [CrossRef]
  19. Longuespée, R.; Couture, F.; Levesque, C.; Kwiatkowska, A.; Desjardins, R.; Gagnon, S.; Vergara, D.; Maffia, M.; Fournier, I.; Salzet, M.; et al. Implications of Proprotein Convertases in Ovarian Cancer Cell Proliferation and Tumor Progression: Insights for PACE4 as a Therapeutic Target. Transl. Oncol. 2014, 7, 410–419. [Google Scholar] [CrossRef] [Green Version]
  20. Fradet, L.; Temmar, R.; Couture, F.; Belzile, M.; Fortier, P.H.; Day, R. Evaluation of PACE4 isoforms as biomarkers in thyroid cancer. J. Otolaryngol. Head Neck Surg. 2018, 47, 63. [Google Scholar] [CrossRef]
  21. Lapierre, M.; Siegfried, G.; Scamuffa, N.; Bontemps, Y.; Calvo, F.; Seidah, N.G.; Khatib, A.M. Opposing function of the proprotein convertases furin and PACE4 on breast cancer cells’ malignant phenotypes: Role of tissue inhibitors of metalloproteinase-1. Cancer Res. 2007, 67, 9030–9034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wang, P.; Wang, F.; Wang, L.; Pan, J. Proprotein convertase subtilisin/kexin type 6 activates the extracellular signal-regulated kinase 1/2 and Wnt family member 3A pathways and promotes in vitro proliferation, migration and invasion of breast cancer MDA-MB-231 cells. Oncol. Lett. 2018, 16, 145–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Tian, X.F.; Huang, G.M.; Zang, H.L.; Cao, H. PACE4 regulates apoptosis in human pancreatic cancer Panc-1 cells via the mitochondrial signaling pathway. Mol. Med. Rep. 2016, 14, 5205–5210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Guo, H.; Yao, Z.; Sun, B.; Hong, Q.; Yan, J.; Mu, D.; Li, J.; Sheng, H. PACE4 regulates apoptosis in human prostate cancer cells via endoplasmic reticulum stress and mitochondrial signaling pathways. Drug Des. Dev. Ther. 2015, 9, 5911–5923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kwiatkowska, A.; Couture, F.; Ait-Mohand, S.; Desjardins, R.; Dory, Y.L.; Guérin, B.; Day, R. Enhanced anti-tumor activity of the Multi-Leu peptide PACE4 inhibitor transformed into an albumin-bound tumor-targeting prodrug. Sci. Rep. 2019, 9, 2118. [Google Scholar] [CrossRef] [Green Version]
  26. Levesque, C.; Couture, F.; Kwiatkowska, A.; Desjardins, R.; Guérin, B.; Neugebauer, W.A.; Day, R. PACE4 inhibitors and their peptidomimetic analogs block prostate cancer tumor progression through quiescence induction, increased apoptosis and impaired neovascularisation. Oncotarget 2015, 6, 3680–3693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Bassi, D.E.; Fu, J.; Lopez de Cicco, R.; Klein-Szanto, A.J. Proprotein convertases: "master switches" in the regulation of tumor growth and progression. Mol. Carcinog. 2005, 44, 151–161. [Google Scholar] [CrossRef] [PubMed]
  28. Klein-Szanto, A.J.; Bassi, D.E. Proprotein convertase inhibition: Paralyzing the cell’s master switches. Biochem. Pharmacol. 2017, 140, 8–15. [Google Scholar] [CrossRef]
  29. Rose, M.; Duhamel, M.; Rodet, F.; Salzet, M. The Role of Proprotein Convertases in the Regulation of the Function of Immune Cells in the Oncoimmune Response. Front. Immunol. 2021, 12, 667850. [Google Scholar] [CrossRef]
  30. Nakayama, K.; Kim, W.; Torii, S.; Hosaka, M.; Nakagawa, T.; Ikemizu, J.; Baba, T.; Murakami, K. Identification of the fourth member of the mammalian endoprotease family homologous to the yeast Kex2 protease. Its testis-specific expression. J. Biol. Chem. 1992, 267, 5897–5900. [Google Scholar] [CrossRef]
  31. Anderson, E.D.; Molloy, S.S.; Jean, F.; Fei, H.; Shimamura, S.; Thomas, G. The ordered and compartment-specfific autoproteolytic removal of the furin intramolecular chaperone is required for enzyme activation. J. Biol. Chem. 2002, 277, 12879–12890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Mayer, G.; Hamelin, J.; Asselin, M.-C.; Pasquato, A.; Marcinkiewicz, E.; Tang, M.; Tabibzadeh, S.; Seidah, N.G. The regulated cell surface zymogen activation of the proprotein convertase PC5A directs the processing of its secretory substrates. J. Biol. Chem. 2008, 283, 2373–2384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Nagahama, M.; Taniguchi, T.; Hashimoto, E.; Imamaki, A.; Mori, K.; Tsuji, A.; Matsuda, Y. Biosynthetic processing and quaternary interactions of proprotein convertase SPC4 (PACE4). FEBS Lett. 1998, 434, 155–159. [Google Scholar] [CrossRef] [Green Version]
  34. Shinde, U.; Inouye, M. Propeptide-mediated folding in subtilisin: The intramolecular chaperone concept. Adv. Exp. Med. Biol. 1996, 379, 147–154. [Google Scholar]
  35. Tsuji, A.; Kikuchi, Y.; Sato, Y.; Koide, S.; Yuasa, K.; Nagahama, M.; Matsuda, Y. A proteomic approach reveals transient association of reticulocalbin-3, a novel member of the CREC family, with the precursor of subtilisin-like proprotein convertase, PACE4. Biochem. J. 2006, 396, 51–59. [Google Scholar] [CrossRef]
  36. Mains, R.E.; Berard, C.A.; Denault, J.B.; Zhou, A.; Johnson, R.C.; Leduc, R. PACE4: A subtilisin-like endoprotease with unique properties. Biochem. J. 1997, 321 Pt 3, 587–593. [Google Scholar] [CrossRef] [Green Version]
  37. Taniguchi, T.; Kuroda, R.; Sakurai, K.; Nagahama, M.; Wada, I.; Tsuji, A.; Matsuda, Y. A critical role for the carboxy terminal region of the proprotein convertase, PACE4A, in the regulation of its autocatalytic activation coupled with secretion. Biochem. Biophys. Res. Commun. 2002, 290, 878–884. [Google Scholar] [CrossRef]
  38. Nour, N.; Mayer, G.; Mort, J.S.; Salvas, A.; Mbikay, M.; Morrison, C.J.; Overall, C.M.; Seidah, N.G. The cysteine-rich domain of the secreted proprotein convertases PC5A and PACE4 functions as a cell surface anchor and interacts with tissue inhibitors of metalloproteinases. Mol. Biol. Cell 2005, 16, 5215–5226. [Google Scholar] [CrossRef] [Green Version]
  39. Tsuji, A.; Sakurai, K.; Kiyokage, E.; Yamazaki, T.; Koide, S.; Toida, K.; Ishimura, K.; Matsuda, Y. Secretory proprotein convertases PACE4 and PC6A are heparin-binding proteins which are localized in the extracellular matrix. Potential role of PACE4 in the activation of proproteins in the extracellular matrix. Biochim. Biophys. Acta 2003, 1645, 95–104. [Google Scholar] [CrossRef]
  40. Wang, H.; Zhou, T.; Peng, J.; Xu, P.; Dong, N.; Chen, S.; Wu, Q. Distinct roles of N-glycosylation at different sites of corin in cell membrane targeting and ectodomain shedding. J. Biol. Chem. 2015, 290, 1654–1663. [Google Scholar] [CrossRef] [Green Version]
  41. Sun, S.; Wang, L.; Zhang, S.; Zhang, C.; Chen, Y.; Wu, Q.; Dong, N. N-glycan in the scavenger receptor cysteine-rich domain of hepsin promotes intracellular trafficking and cell surface expression. Int. J. Biol. Macromol. 2020, 161, 818–827. [Google Scholar] [CrossRef] [PubMed]
  42. Martin, C.E.; Murray, A.S.; Sala-Hamrick, K.E.; Mackinder, J.R.; Harrison, E.C.; Lundgren, J.G.; Varela, F.A.; List, K. Posttranslational modifications of serine protease TMPRSS13 regulate zymogen activation, proteolytic activity, and cell surface localization. J. Biol. Chem. 2021, 297, 101227. [Google Scholar] [CrossRef]
  43. Jiang, J.; Yang, J.; Feng, P.; Zuo, B.; Dong, N.; Wu, Q.; He, Y. N-glycosylation is required for matriptase-2 autoactivation and ectodomain shedding. J. Biol. Chem. 2014, 289, 19500–19507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Wang, H.; Li, S.; Wang, J.; Chen, S.; Sun, X.L.; Wu, Q. N-glycosylation in the protease domain of trypsin-like serine proteases mediates calnexin-assisted protein folding. Elife 2018, 7, e35672. [Google Scholar] [CrossRef]
  45. Sucic, J.F.; Moehring, J.M.; Inocencio, N.M.; Luchini, J.W.; Moehring, T.J. Endoprotease PACE4 is Ca2+-dependent and temperature-sensitive and can partly rescue the phenotype of a furin-deficient cell strain. Biochem. J. 1999, 339 Pt 3, 639–647. [Google Scholar] [CrossRef] [PubMed]
  46. Jin, W.; Fuki, I.V.; Seidah, N.G.; Benjannet, S.; Glick, J.M.; Rader, D.J. Proprotein convertases [corrected] are responsible for proteolysis and inactivation of endothelial lipase. J. Biol. Chem. 2005, 280, 36551–36559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Siegfried, G.; Basak, A.; Prichett-Pejic, W.; Scamuffa, N.; Ma, L.; Benjannet, S.; Veinot, J.P.; Calvo, F.; Seidah, N.; Khatib, A.-M. Regulation of the stepwise proteolytic cleavage and secretion of PDGF-B by the proprotein convertases. Oncogene 2005, 24, 6925–6935. [Google Scholar] [CrossRef] [Green Version]
  48. Yuasa, K.; Masuda, T.; Yoshikawa, C.; Nagahama, M.; Matsuda, Y.; Tsuji, A. Subtilisin-like proprotein convertase PACE4 is required for skeletal muscle differentiation. J. Biochem. 2009, 146, 407–415. [Google Scholar] [CrossRef]
  49. Kim, W.; Zekas, E.; Lodge, R.; Susan-Resiga, D.; Marcinkiewicz, E.; Essalmani, R.; Mihara, K.; Ramachandran, R.; Asahchop, E.; Gelman, B.; et al. Neuroinflammation-Induced Interactions between Protease-Activated Receptor 1 and Proprotein Convertases in HIV-Associated Neurocognitive Disorder. Mol. Cell. Biol. 2015, 35, 3684–3700. [Google Scholar] [CrossRef] [Green Version]
  50. Kara, I.; Poggi, M.; Bonardo, B.; Govers, R.; Landrier, J.F.; Tian, S.; Leibiger, I.; Day, R.; Creemers, J.W.; Peiretti, F. The paired basic amino acid-cleaving enzyme 4 (PACE4) is involved in the maturation of insulin receptor isoform B: An opportunity to reduce the specific insulin receptor-dependent effects of insulin-like growth factor 2 (IGF2). J. Biol. Chem. 2015, 290, 2812–2821. [Google Scholar] [CrossRef] [Green Version]
  51. Rehemtulla, A.; Barr, P.J.; Rhodes, C.J.; Kaufman, R.J. PACE4 is a member of the mammalian propeptidase family that has overlapping but not identical substrate specificity to PACE. Biochemistry 1993, 32, 11586–11590. [Google Scholar] [CrossRef] [PubMed]
  52. Essalmani, R.; Susan-Resiga, D.; Guillemot, J.; Kim, W.; Sachan, V.; Awan, Z.; Chamberland, A.; Asselin, M.-C.; Ly, K.; Desjardins, R.; et al. Thrombin activation of protein C requires prior processing by a liver proprotein convertase. J. Biol. Chem. 2017, 292, 10564–10573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Chen, S.; Cao, P.; Dong, N.; Peng, J.; Zhang, C.; Wang, H.; Zhou, T.; Yang, J.; Zhang, Y.; Martelli, E.E.; et al. PCSK6-mediated corin activation is essential for normal blood pressure. Nat. Med. 2015, 21, 1048–1053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Oroszlán, G.; Dani, R.; Végh, B.M.; Varga, D.; Ács, A.V.; Pál, G.; Závodszky, P.; Farkas, H.; Gál, P.; Dobó, J. Proprotein Convertase Is the Highest-Level Activator of the Alternative Complement Pathway in the Blood. J. Immunol. 2021, 206, 2198–2205. [Google Scholar] [CrossRef] [PubMed]
  55. Okada, K.; Moon, H.J.; Finney, J.; Meier, A.; Mure, M. Extracellular Processing of Lysyl Oxidase-like 2 and Its Effect on Amine Oxidase Activity. Biochemistry 2018, 57, 6973–6983. [Google Scholar] [CrossRef]
  56. Layne, J.D.; Shridas, P.; Webb, N.R. Ectopically expressed pro-group X secretory phospholipase A2 is proteolytically activated in mouse adrenal cells by furin-like proprotein convertases: Implications for the regulation of adrenal steroidogenesis. J. Biol. Chem. 2015, 290, 7851–7860. [Google Scholar] [CrossRef] [Green Version]
  57. Li, J.J.; Liu, J.; Lupino, K.; Liu, X.; Zhang, L.; Pei, L. Growth Differentiation Factor 15 Maturation Requires Proteolytic Cleavage by PCSK3, -5, and -6. Mol. Cell. Biol. 2018, 38, e00249-18. [Google Scholar] [CrossRef] [Green Version]
  58. Constam, D.B.; Robertson, E.J. Regulation of bone morphogenetic protein activity by pro domains and proprotein convertases. J. Cell Biol. 1999, 144, 139–149. [Google Scholar] [CrossRef] [Green Version]
  59. Cui, Y.; Jean, F.; Thomas, G.; Christian, J.L. BMP-4 is proteolytically activated by furin and/or PC6 during vertebrate embryonic development. Embo J. 1998, 17, 4735–4743. [Google Scholar] [CrossRef] [Green Version]
  60. Scamuffa, N.; Basak, A.; Lalou, C.; Wargnier, A.; Marcinkiewicz, J.; Siegfried, G.; Chretien, M.; Calvo, F.; Seidah, N.G.; Khatib, A.-M. Regulation of prohepcidin processing and activity by the subtilisin-like proprotein convertases Furin, PC5, PACE4 and PC7. Gut 2008, 57, 1573–1582. [Google Scholar] [CrossRef]
  61. Watanabe, H.; Fitting, S.; Hussain, M.Z.; Kononenko, O.; Iatsyshyna, A.; Yoshitake, T.; Kehr, J.; Alkass, K.; Druid, H.; Wadensten, H.; et al. Asymmetry of the endogenous opioid system in the human anterior cingulate: A putative molecular basis for lateralization of emotions and pain. Cereb. Cortex 2015, 25, 97–108. [Google Scholar] [CrossRef] [PubMed]
  62. Gordon, V.M.; Rehemtulla, A.; Leppla, S.H. A role for PACE4 in the proteolytic activation of anthrax toxin protective antigen. Infect. Immun. 1997, 65, 3370–3375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Xiao, Y.; Chen, G.; Richard, J.; Rougeau, N.; Li, H.; Seidah, N.G.; Cohen, E.A. Cell-surface processing of extracellular human immunodeficiency virus type 1 Vpr by proprotein convertases. Virology 2008, 372, 384–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Remacle, A.G.; Shiryaev, S.A.; Oh, E.-S.; Cieplak, P.; Srinivasan, A.; Wei, G.; Liddington, R.C.; Ratnikov, B.I.; Parent, A.; Desjardins, R.; et al. Substrate cleavage analysis of furin and related proprotein convertases. A comparative study. J. Biol. Chem. 2008, 283, 20897–20906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Constam, D.B.; Robertson, E.J. SPC4/PACE4 regulates a TGFbeta signaling network during axis formation. Genes Dev. 2000, 14, 1146–1155. [Google Scholar] [CrossRef]
  66. Beck, S.; Le Good, J.A.; Guzman, M.; Ben Haim, N.; Roy, K.; Beermann, F.; Constam, D.B. Extraembryonic proteases regulate Nodal signalling during gastrulation. Nat. Cell Biol. 2002, 4, 981–985. [Google Scholar] [CrossRef]
  67. Jones, W.D.; Mullins, M.C. Cell signaling pathways controlling an axis organizing center in the zebrafish. Curr. Top. Dev. Biol. 2022, 150, 149–209. [Google Scholar]
  68. Blanchet, M.-H.; Le Good, J.A.; Mesnard, D.; Oorschot, V.; Baflast, S.; Minchiotti, G.; Klumperman, J.; Constam, D.B. Cripto recruits Furin and PACE4 and controls Nodal trafficking during proteolytic maturation. EMBO J. 2008, 27, 2580–2591. [Google Scholar] [CrossRef] [Green Version]
  69. Mujoomdar, M.L.; Hogan, L.M.; Parlow, A.F.; Nachtigal, M.W. Pcsk6 mutant mice exhibit progressive loss of ovarian function, altered gene expression, and formation of ovarian pathology. Reproduction 2011, 141, 343–355. [Google Scholar] [CrossRef] [Green Version]
  70. Akiyama, I.; Yoshino, O.; Osuga, Y.; Shi, J.; Hirota, Y.; Hirata, T.; Harada, M.; Koga, K.; Fujimoto, A.; Yano, T.; et al. The localization and regulation of proprotein convertase subtilisin/kexin (PCSK) 6 in human ovary. Am. J. Reprod. Immunol. 2012, 68, 491–498. [Google Scholar] [CrossRef]
  71. Diaz, F.J.; Sugiura, K.; Eppig, J.J. Regulation of Pcsk6 expression during the preantral to antral follicle transition in mice: Opposing roles of FSH and oocytes. Biol. Reprod. 2008, 78, 176–183. [Google Scholar] [CrossRef] [PubMed]
  72. Wang, Y.; Wang, X.H.; Fan, D.X.; Zhang, Y.; Li, M.Q.; Wu, H.X.; Jin, L.P. PCSK6 regulated by LH inhibits the apoptosis of human granulosa cells via activin A and TGFβ2. J. Endocrinol. 2014, 222, 151–160. [Google Scholar] [CrossRef]
  73. Yan, C.; Wang, P.; DeMayo, J.; DeMayo, F.J.; Elvin, J.A.; Carino, C.; Prasad, S.V.; Skinner, S.S.; Dunbar, B.S.; Dube, J.L.; et al. Synergistic roles of bone morphogenetic protein 15 and growth differentiation factor 9 in ovarian function. Mol. Endocrinol. 2001, 15, 854–866. [Google Scholar] [CrossRef] [PubMed]
  74. Dixit, H.; Rao, L.K.; Padmalatha, V.; Kanakavalli, M.; Deenadayal, M.; Gupta, N.; Chakravarty, B.; Singh, L. Mutational screening of the coding region of growth differentiation factor 9 gene in Indian women with ovarian failure. Menopause 2005, 12, 749–754. [Google Scholar] [CrossRef] [PubMed]
  75. Dixit, H.; Rao, L.K.; Padmalatha, V.V.; Kanakavalli, M.; Deenadayal, M.; Gupta, N.; Chakrabarty, B.; Singh, L. Missense mutations in the BMP15 gene are associated with ovarian failure. Hum. Genet. 2006, 119, 408–415. [Google Scholar] [CrossRef]
  76. Marinakis, N.M.; Tsoutsou, E.; Sofocleous, C.; Veltra, D.; Papaefthimiou, P.; Lytras, A.; Traeger-Synodinos, J.; Kanaka-Gantenbein, C. Ovarian insufficiency and secondary amenorrhea in a patient with a novel variant within GDF9 gene. Menopause 2022, 29, 491–495. [Google Scholar] [CrossRef]
  77. Malfait, A.-M.; Seymour, A.B.; Gao, F.; Tortorella, M.D.; Le Graverand-Gastineau, M.-P.H.; Wood, L.S.; Doherty, M.; Doherty, S.; Zhang, W.; Arden, N.K.; et al. A role for PACE4 in osteoarthritis pain: Evidence from human genetic association and null mutant phenotype. Ann. Rheum. Dis. 2012, 71, 1042–1048. [Google Scholar] [CrossRef]
  78. Roughley, P.J.; Mort, J.S. The role of aggrecan in normal and osteoarthritic cartilage. J. Exp. Orthop. 2014, 1, 8. [Google Scholar] [CrossRef] [Green Version]
  79. Hurysz, B.; Bottini, N. Emerging proteoglycans and proteoglycan-targeted therapies in rheumatoid arthritis. Am. J. Physiol. Physiol. 2022, 322, C1061–C1067. [Google Scholar] [CrossRef]
  80. Li, T.; Peng, J.; Li, Q.; Shu, Y.; Zhu, P.; Hao, L. The Mechanism and Role of ADAMTS Protein Family in Osteoarthritis. Biomolecules 2022, 12, 959. [Google Scholar] [CrossRef]
  81. Malfait, A.-M.; Arner, E.C.; Song, R.-H.; Alston, J.T.; Markosyan, S.; Staten, N.; Yang, Z.; Griggs, D.W.; Tortorella, M.D. Proprotein convertase activation of aggrecanases in cartilage in situ. Arch. Biochem. Biophys. 2008, 478, 43–51. [Google Scholar] [CrossRef]
  82. Wang, F.; Wang, L.; Jiang, H.; Chang, X.; Pan, J. Inhibition of PCSK6 may play a protective role in the development of rheumatoid arthritis. J. Rheumatol. 2015, 42, 161–169. [Google Scholar] [CrossRef]
  83. Jiang, H.; Wang, L.; Wang, F.; Pan, J. Proprotein convertase subtilisin/kexin type 6 promotes in vitro proliferation, migration and inflammatory cytokine secretion of synovial fibroblast-like cells from rheumatoid arthritis via nuclear-κB, signal transducer and activator of transcription 3 and extracellular signal regulated 1/2 pathways. Mol. Med. Rep. 2017, 16, 8477–8484. [Google Scholar] [PubMed] [Green Version]
  84. Jiang, H.; Wang, L.; Pan, J. Inhibition of pro-protein convertase subtilisin/kexin type 6 has a protective role against synovitis in a rat model of rheumatoid arthritis. Mol. Med. Rep. 2015, 12, 7681–7686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Prica, M.; Kamalathasan, S.; Gopaul, K.; Warriner, D. Adult congenital heart disease: A review of the simple lesions. Br. J. Hosp. Med. 2022, 83, 1–12. [Google Scholar] [CrossRef] [PubMed]
  86. Webb, G.; Gatzoulis, M.A. Atrial septal defects in the adult: Recent progress and overview. Circulation 2006, 114, 1645–1653. [Google Scholar] [CrossRef] [Green Version]
  87. McCulley, D.J.; Black, B.L. Transcription factor pathways and congenital heart disease. Curr. Top. Dev. Biol. 2012, 100, 253–277. [Google Scholar]
  88. Zhou, L.; Liu, J.; Olson, P.; Zhang, K.; Wynne, J.; Xie, L. Tbx5 and Osr1 interact to regulate posterior second heart field cell cycle progression for cardiac septation. J. Mol. Cell. Cardiol. 2015, 85, 1–12. [Google Scholar] [CrossRef] [Green Version]
  89. Xie, L.; Hoffmann, A.D.; Burnicka-Turek, O.; Friedland-Little, J.M.; Zhang, K.; Moskowitz, I.P. Tbx5-hedgehog molecular networks are essential in the second heart field for atrial septation. Dev. Cell 2012, 23, 280–291. [Google Scholar] [CrossRef] [Green Version]
  90. Wang, Q.; Lan, Y.; Cho, E.S.; Maltby, K.M.; Jiang, R. Odd-skipped related 1 (Odd 1) is an essential regulator of heart and urogenital development. Dev. Biol. 2005, 288, 582–594. [Google Scholar] [CrossRef] [Green Version]
  91. Zhang, K.K.; Xiang, M.; Zhou, L.; Liu, J.; Curry, N.; Heine Suner, D.; Garcia-Pavia, P.; Zhang, X.; Wang, Q.; Xie, L. Gene network and familial analyses uncover a gene network involving Tbx5/Osr1/Pcsk6 interaction in the second heart field for atrial septation. Hum. Mol. Genet. 2016, 25, 1140–1151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Flaquer, A.; Baumbach, C.; Piñero, E.; Algas, F.G.; Sanchez, M.A.D.L.F.; Rosell, J.; Toquero, J.; Alonso-Pulpon, L.; Garcia-Pavia, P.; Strauch, K.; et al. Genome-wide linkage analysis of congenital heart defects using MOD score analysis identifies two novel loci. BMC Genet. 2013, 14, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Rader, D.J.; Jaye, M. Endothelial lipase: A new member of the triglyceride lipase gene family. Curr. Opin. Lipidol. 2000, 11, 141–147. [Google Scholar] [CrossRef]
  94. Khetarpal, S.A.; Vitali, C.; Levin, M.G.; Klarin, D.; Park, J.; Pampana, A.; Millar, J.S.; Kuwano, T.; Sugasini, D.; Subbaiah, P.V.; et al. Endothelial lipase mediates efficient lipolysis of triglyceride-rich lipoproteins. PLoS Genet. 2021, 17, e1009802. [Google Scholar] [CrossRef] [PubMed]
  95. Ma, K.; Cilingiroglu, M.; Otvos, J.D.; Ballantyne, C.M.; Marian, A.J.; Chan, L. Endothelial lipase is a major genetic determinant for high-density lipoprotein concentration, structure, and metabolism. Proc. Natl. Acad. Sci. USA 2003, 100, 2748–2753. [Google Scholar] [CrossRef] [Green Version]
  96. Ishida, T.; Choi, S.; Kundu, R.K.; Hirata, K.; Rubin, E.M.; Cooper, A.D.; Quertermous, T. Endothelial lipase is a major determinant of HDL level. J. Clin. Investig. 2003, 111, 347–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Jaye, M.; Lynch, K.J.; A Krawiec, J.; Marchadier, D.; Maugeais, C.; Doan, K.; South, V.J.; Amin, D.V.; Perrone, M.H.; Rader, D.J. A novel endothelial-derived lipase that modulates HDL metabolism. Nat. Genet. 1999, 21, 424–428. [Google Scholar] [CrossRef]
  98. Edmondson, A.C.; Brown, R.J.; Kathiresan, S.; Cupples, L.A.; Demissie, S.; Manning, A.K.; Jensen, M.K.; Rimm, E.B.; Wang, J.; Rodrigues, A.; et al. Loss-of-function variants in endothelial lipase are a cause of elevated HDL cholesterol in humans. J. Clin. Investig. 2009, 119, 1042–1050. [Google Scholar] [CrossRef] [Green Version]
  99. Cole, J.; Blackhurst, D.M.; Solomon, G.A.E.; Ratanjee, B.D.; Benjamin, R.; Marais, A.D. Atherosclerotic cardiovascular disease in hyperalphalipoproteinemia due to LIPG variants. J. Clin. Lipidol. 2021, 15, 142–150.e142. [Google Scholar] [CrossRef]
  100. Pisciotta, L.; Ossoli, A.; Ronca, A.; Garuti, A.; Fresa, R.; Favari, E.; Calabresi, L.; Calandra, S.; Bertolini, S. Plasma HDL pattern, cholesterol efflux and cholesterol loading capacity of serum in carriers of a novel missense variant (Gly176Trp) of endothelial lipase. J. Clin. Lipidol. 2022, in press. [CrossRef]
  101. Wang, L.; Zhang, C.; Sun, S.; Chen, Y.; Hu, Y.; Wang, H.; Liu, M.; Dong, N.; Wu, Q. Autoactivation and calpain-1-mediated shedding of hepsin in human hepatoma cells. Biochem. J. 2019, 476, 2355–2369. [Google Scholar] [CrossRef] [PubMed]
  102. Jiang, J.; Wu, S.; Wang, W.; Chen, S.; Peng, J.; Zhang, X.; Wu, Q. Ectodomain shedding and autocleavage of the cardiac membrane protease corin. J. Biol. Chem. 2011, 286, 10066–10072. [Google Scholar] [CrossRef] [Green Version]
  103. Gauster, M.; Hrzenjak, A.; Schick, K.; Frank, S. Endothelial lipase is inactivated upon cleavage by the members of the proprotein convertase family. J. Lipid Res. 2005, 46, 977–987. [Google Scholar] [CrossRef] [PubMed]
  104. Jin, W.; Wang, X.; Millar, J.S.; Quertermous, T.; Rothblat, G.H.; Glick, J.M.; Rader, D.J. Hepatic proprotein convertases modulate HDL metabolism. Cell Metab. 2007, 6, 129–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Hato, T.; Tabata, M.; Oike, Y. The role of angiopoietin-like proteins in angiogenesis and metabolism. Trends Cardiovasc. Med. 2008, 18, 6–14. [Google Scholar] [CrossRef] [PubMed]
  106. Essalmani, R.; Susan-Resiga, D.; Chamberland, A.; Asselin, M.-C.; Canuel, M.; Constam, D.; Creemers, J.W.; Day, R.; Gauthier, D.; Prat, A.; et al. Furin is the primary in vivo convertase of angiopoietin-like 3 and endothelial lipase in hepatocytes. J. Biol. Chem. 2013, 288, 26410–26418. [Google Scholar] [CrossRef] [Green Version]
  107. Nishikimi, T.; Kato, J. Cardiac Peptides-Current Physiology, Pathophysiology, Biochemistry, Molecular Biology, and Clinical Application. Biology 2022, 11, 330. [Google Scholar] [CrossRef]
  108. Salo, P.P.; Havulinna, A.S.; Tukiainen, T.; Raitakari, O.; Lehtimäki, T.; Kähönen, M.; Kettunen, J.; Männikkö, M.; Eriksson, J.G.; Jula, A.; et al. Genome-Wide Association Study Implicates Atrial Natriuretic Peptide Rather Than B-Type Natriuretic Peptide in the Regulation of Blood Pressure in the General Population. Circ. Cardiovasc. Genet. 2017, 10, e001713. [Google Scholar] [CrossRef] [Green Version]
  109. John, S.W.; Krege, J.H.; Oliver, P.M.; Hagaman, J.R.; Hodgin, J.B.; Pang, S.C.; Flynn, T.G.; Smithies, O. Genetic decreases in atrial natriuretic peptide and salt-sensitive hypertension. Science 1995, 267, 679–681. [Google Scholar] [CrossRef]
  110. Tamura, N.; Ogawa, Y.; Chusho, H.; Nakamura, K.; Nakao, K.; Suda, M.; Kasahara, M.; Hashimoto, R.; Katsuura, G.; Mukoyama, M.; et al. Cardiac fibrosis in mice lacking brain natriuretic peptide. Proc. Natl. Acad. Sci. USA 2000, 97, 4239–4244. [Google Scholar] [CrossRef] [Green Version]
  111. Song, W.; Wang, H.; Wu, Q. Atrial natriuretic peptide in cardiovascular biology and disease (NPPA). Gene 2015, 569, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Rubattu, S.; Forte, M.; Marchitti, S.; Volpe, M. Molecular Implications of Natriuretic Peptides in the Protection from Hypertension and Target Organ Damage Development. Int. J. Mol. Sci. 2019, 20, 798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Yan, W.; Sheng, N.; Seto, M.; Morser, J.; Wu, Q. Corin, a mosaic transmembrane serine protease encoded by a novel cDNA from human heart. J. Biol. Chem. 1999, 274, 14926–14935. [Google Scholar] [CrossRef]
  114. Zhang, X.; Gu, X.; Zhang, Y.; Dong, N.; Wu, Q. Corin: A Key Mediator in Sodium Homeostasis, Vascular Remodeling, and Heart Failure. Biology 2022, 11, 717. [Google Scholar] [CrossRef]
  115. Dong, N.; Niu, Y.; Chen, Y.; Sun, S.; Wu, Q. Function and regulation of corin in physiology and disease. Biochem. Soc. Trans. 2020, 48, 1905–1916. [Google Scholar] [CrossRef] [PubMed]
  116. Yan, W.; Wu, F.; Morser, J.; Wu, Q. Corin, a transmembrane cardiac serine protease, acts as a pro-atrial natriuretic peptide-converting enzyme. Proc. Natl. Acad. Sci. USA 2000, 97, 8525–8529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Knappe, S.; Wu, F.; Masikat, M.R.; Morser, J.; Wu, Q. Functional analysis of the transmembrane domain and activation cleavage of human corin: Design and characterization of a soluble corin. J. Biol. Chem. 2003, 278, 52363–52370. [Google Scholar] [CrossRef] [Green Version]
  118. Chen, S.; Wang, H.; Li, H.; Zhang, Y.; Wu, Q. Functional analysis of corin protein domains required for PCSK6-mediated activation. Int. J. Biochem. Cell Biol. 2018, 94, 31–39. [Google Scholar] [CrossRef]
  119. Wang, W.; Shen, J.; Cui, Y.; Jiang, J.; Chen, S.; Peng, J.; Wu, Q. Impaired sodium excretion and salt-sensitive hypertension in corin-deficient mice. Kidney Int. 2012, 82, 26–33. [Google Scholar] [CrossRef] [Green Version]
  120. Zhang, Y.; Zhou, T.; Niu, Y.; He, M.; Wang, C.; Liu, M.; Yang, J.; Zhang, Y.; Zhou, J.; Fukuda, K.; et al. Identification and functional analysis of CORIN variants in hypertensive patients. Hum. Mutat. 2017, 38, 1700–1710. [Google Scholar] [CrossRef]
  121. Wakim, V.; Abi Khalil, E.; Salloum, A.K.; Khazen, G.; Ghassibe-Sabbagh, M.; Zalloua, P.A. New susceptibility alleles associated with severe coronary artery stenosis in the Lebanese population. BMC Med. Genom. 2021, 14, 90. [Google Scholar]
  122. Zhang, J.; Yin, Y.; Chen, L.; Chu, C.; Wang, Y.; Lv, Y.; He, M.; Martin, M.; Huang, P.-H.; Mu, J.-J.; et al. Short-Term High-Salt Diet Increases Corin Level to Regulate the Salt-Water Balance in Humans and Rodents. Am. J. Hypertens. 2018, 31, 253–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Chan, J.C.; Knudson, O.; Wu, F.; Morser, J.; Dole, W.P.; Wu, Q. Hypertension in mice lacking the proatrial natriuretic peptide convertase corin. Proc. Natl. Acad. Sci. USA 2005, 102, 785–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Khoury, E.E.; Fokra, A.; Kinaneh, S.; Knaney, Y.; Aronson, D.; Abassi, Z. Distribution of Cardiac and Renal Corin and Proprotein Convertase Subtilisin/Kexin-6 in the Experimental Model of Cardio-Renal Syndrome of Various Severities. Front. Physiol. 2021, 12, 673497. [Google Scholar] [CrossRef]
  125. Libby, P.; Ridker, P.M.; Hansson, G.K. Progress and challenges in translating the biology of atherosclerosis. Nature 2011, 473, 317–325. [Google Scholar] [CrossRef] [PubMed]
  126. Suh, J.H.; Yoon, J.S.; Kwon, J.B.; Kim, H.W.; Wang, Y.P. Identification of genomic aberrations by array comparative genomic hybridization in patients with aortic dissections. Korean J. Thorac. Cardiovasc. Surg. 2011, 44, 123–130. [Google Scholar] [CrossRef]
  127. Turpeinen, H.; Raitoharju, E.; Oksanen, A.; Oksala, N.; Levula, M.; Lyytikäinen, L.-P.; Järvinen, O.; Creemers, J.W.; Kähönen, M.; Laaksonen, R.; et al. Proprotein convertases in human atherosclerotic plaques: The overexpression of FURIN and its substrate cytokines BAFF and APRIL. Atherosclerosis 2011, 219, 799–806. [Google Scholar] [CrossRef]
  128. Suur, B.E.; Chemaly, M.; Liljeqvist, M.L.; Djordjevic, D.; Stenemo, M.; Bergman, O.; Karlöf, E.; Lengquist, M.; Odeberg, J.; Hurt-Camejo, E.; et al. Therapeutic potential of the Proprotein Convertase Subtilisin/Kexin family in vascular disease. Front. Pharmacol. 2022, 13, 988561. [Google Scholar] [CrossRef]
  129. Testa, G.; Staurenghi, E.; Giannelli, S.; Sottero, B.; Gargiulo, S.; Poli, G.; Gamba, P.; Leonarduzzi, G. Up-regulation of PCSK6 by lipid oxidation products: A possible role in atherosclerosis. Biochimie 2021, 181, 191–203. [Google Scholar] [CrossRef]
  130. Röhl, S.; Suur, B.E.; Lengquist, M.; Seime, T.; Caidahl, K.; Hedin, U.; Arner, A.; Matic, L.; Razuvaev, A. Lack of PCSK6 Increases Flow-Mediated Outward Arterial Remodeling in Mice. Cells 2020, 9, 1009. [Google Scholar] [CrossRef] [Green Version]
  131. Perisic, L.; Hedin, E.; Razuvaev, A.; Lengquist, M.; Osterholm, C.; Folkersen, L.; Gillgren, P.; Paulsson-Berne, G.; Ponten, F.; Odeberg, J.; et al. Profiling of atherosclerotic lesions by gene and tissue microarrays reveals PCSK6 as a novel protease in unstable carotid atherosclerosis. Arter. Thromb. Vasc. Biol. 2013, 33, 2432–2443. [Google Scholar] [CrossRef]
  132. Rykaczewska, U.; Suur, B.E.; Röhl, S.; Razuvaev, A.; Lengquist, M.; Sabater-Lleal, M.; van der Laan, S.W.; Miller, C.L.; Wirka, R.C.; Kronqvist, M.; et al. PCSK6 Is a Key Protease in the Control of Smooth Muscle Cell Function in Vascular Remodeling. Circ. Res. 2020, 126, 571–585. [Google Scholar] [CrossRef] [PubMed]
  133. Bassi, D.E.; Mahloogi, H.; Klein-Szanto, A.J. The proprotein convertases furin and PACE4 play a significant role in tumor progression. Mol. Carcinog. 2000, 28, 63–69. [Google Scholar] [CrossRef]
  134. Prabhu, S.D.; Frangogiannis, N.G. The Biological Basis for Cardiac Repair After Myocardial Infarction: From Inflammation to Fibrosis. Circ. Res. 2016, 119, 91–112. [Google Scholar] [CrossRef]
  135. Wu, Q.; Kuo, H.C.; Deng, G.G. Serine proteases and cardiac function. Biochim. Biophys. Acta 2005, 1751, 82–94. [Google Scholar] [CrossRef] [PubMed]
  136. Kuhn, T.C.; Knobel, J.; Burkert-Rettenmaier, S.; Li, X.; Meyer, I.S.; Jungmann, A.; Sicklinger, F.; Backs, J.; Lasitschka, F.; Müller, O.J.; et al. Secretome Analysis of Cardiomyocytes Identifies PCSK6 (Proprotein Convertase Subtilisin/Kexin Type 6) as a Novel Player in Cardiac Remodeling After Myocardial Infarction. Circulation 2020, 141, 1628–1644. [Google Scholar] [CrossRef]
  137. Sawada, Y.; Inoue, M.; Kanda, T.; Sakamaki, T.; Tanaka, S.; Minamino, N.; Nagai, R.; Takeuchi, T. Co-elevation of brain natriuretic peptide and proprotein-processing endoprotease furin after myocardial infarction in rats. FEBS Lett. 1997, 400, 177–182. [Google Scholar] [CrossRef] [Green Version]
  138. Yang, S.F.; Chou, R.H.; Lin, S.J.; Li, S.Y.; Huang, P.H. Serum PCSK6 and corin levels are not associated with cardiovascular outcomes in patients undergoing coronary angiography. PLoS ONE 2019, 14, e0226129. [Google Scholar] [CrossRef] [Green Version]
  139. Nakou, E.S.; Parthenakis, F.I.; Kallergis, E.M.; Marketou, M.E.; Nakos, K.S.; Vardas, P.E. Healthy aging and myocardium: A complicated process with various effects in cardiac structure and physiology. Int. J. Cardiol. 2016, 209, 167–175. [Google Scholar] [CrossRef]
  140. Bernhard, D.; Laufer, G. The aging cardiomyocyte: A mini-review. Gerontology 2008, 54, 24–31. [Google Scholar] [CrossRef]
  141. Iemitsu, M.; Maeda, S.; Otsuki, T.; Sugawara, J.; Kuno, S.; Ajisaka, R.; Matsuda, M. Arterial stiffness, physical activity, and atrial natriuretic Peptide gene polymorphism in older subjects. Hypertens. Res. 2008, 31, 767–774. [Google Scholar] [CrossRef] [Green Version]
  142. Pollack, J.A.; Skvorak, J.P.; Nazian, S.J.; Landon, C.S.; Dietz, J.R. Alterations in atrial natriuretic peptide (ANP) secretion and renal effects in aging. J. Gerontol. A Biol. Sci. Med. Sci. 1997, 52, B196–B202. [Google Scholar] [CrossRef] [PubMed]
  143. Häseli, S.; Deubel, S.; Jung, T.; Grune, T.; Ott, C. Cardiomyocyte Contractility and Autophagy in a Premature Senescence Model of Cardiac Aging. Oxidative Med. Cell. Longev. 2020, 2020, 8141307. [Google Scholar] [CrossRef] [PubMed]
  144. Zhan, W.; Chen, L.; Liu, H.; Long, C.; Liu, J.; Ding, S.; Wu, Q.; Chen, S. Pcsk6 Deficiency Promotes Cardiomyocyte Senescence by Modulating Ddit3-Mediated ER Stress. Genes 2022, 13, 711. [Google Scholar] [CrossRef] [PubMed]
  145. Marciniak, S.J.; Yun, C.Y.; Oyadomari, S.; Novoa, I.; Zhang, Y.; Jungreis, R.; Nagata, K.; Harding, H.P.; Ron, D. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev. 2004, 18, 3066–3077. [Google Scholar] [CrossRef] [Green Version]
  146. Fu, H.Y.; Okada, K.-I.; Liao, Y.; Tsukamoto, O.; Isomura, T.; Asai, M.; Sawada, T.; Okuda, K.; Asano, Y.; Sanada, S.; et al. Ablation of C/EBP homologous protein attenuates endoplasmic reticulum-mediated apoptosis and cardiac dysfunction induced by pressure overload. Circulation 2010, 122, 361–369. [Google Scholar] [CrossRef]
  147. Seidah, N.G.; Prat, A. The Multifaceted Biology of PCSK9. Endocr. Rev. 2022, 43, 558–582. [Google Scholar] [CrossRef]
Figure 1. Schematic presentation of human PCSK6 domains. Human PCSK6 consists of a signal peptide (Signal), a pro-domain, a subtilisin-like catalytic domain, a P or Homo B domain, and a C-terminal cysteine-rich domain. In the catalytic domain, the conserved active sites, Asp (D), His (H), and Ser (S), are at positions 205, 246, and 420, respectively. Three predicted N-glycosylation sites (Y shaped symbols) are at positions 259, 914, and 932, respectively. Two autoactivation cleavage sites are at Arg (R) 117 (open arrowhead) (second cleavage) and R149 (red arrowhead) (first cleavage).
Figure 1. Schematic presentation of human PCSK6 domains. Human PCSK6 consists of a signal peptide (Signal), a pro-domain, a subtilisin-like catalytic domain, a P or Homo B domain, and a C-terminal cysteine-rich domain. In the catalytic domain, the conserved active sites, Asp (D), His (H), and Ser (S), are at positions 205, 246, and 420, respectively. Three predicted N-glycosylation sites (Y shaped symbols) are at positions 259, 914, and 932, respectively. Two autoactivation cleavage sites are at Arg (R) 117 (open arrowhead) (second cleavage) and R149 (red arrowhead) (first cleavage).
Ijms 23 13429 g001
Figure 2. PCSK6 intracellular trafficking and autoactivation. PCSK6 is synthesized as a one-chain zymogen. The signal peptide is removed by signal peptidase in the ER (not shown). The pro-domain (white), the catalytic domain (blue), the P/Homo B domain (gray), and the C-terminal cysteine-rich domain (black) are shown. Within the ER, first autocleavage occurs between the pro-domain and the catalytic domain (red arrowhead). The cleaved pro-domain remains attached, acts as an inhibitor of PCSK6, and facilitates PCSK6 folding and ER exiting. Upon secretion, a second autocleavage occurs within the pro-domain (open arrowhead), which disassociates the pro-domain, converting PCSK6 to an active enzyme.
Figure 2. PCSK6 intracellular trafficking and autoactivation. PCSK6 is synthesized as a one-chain zymogen. The signal peptide is removed by signal peptidase in the ER (not shown). The pro-domain (white), the catalytic domain (blue), the P/Homo B domain (gray), and the C-terminal cysteine-rich domain (black) are shown. Within the ER, first autocleavage occurs between the pro-domain and the catalytic domain (red arrowhead). The cleaved pro-domain remains attached, acts as an inhibitor of PCSK6, and facilitates PCSK6 folding and ER exiting. Upon secretion, a second autocleavage occurs within the pro-domain (open arrowhead), which disassociates the pro-domain, converting PCSK6 to an active enzyme.
Ijms 23 13429 g002
Figure 3. Role of PCSK6 in cardiovascular development and pathophysiology. PCSK6 plays a key role in atrial septum formation during embryogenesis. In adult stages, PCSK6 regulates lipoprotein metabolism and blood pressure by inactivating endothelial lipase and activating corin, respectively. PCSK6 participates in vascular remodeling and cardiac repair under pathological conditions such as atherosclerosis and acute MI. In aged hearts, PCKS6 prevents senescence in cardiomyocytes by reducing ER stress.
Figure 3. Role of PCSK6 in cardiovascular development and pathophysiology. PCSK6 plays a key role in atrial septum formation during embryogenesis. In adult stages, PCSK6 regulates lipoprotein metabolism and blood pressure by inactivating endothelial lipase and activating corin, respectively. PCSK6 participates in vascular remodeling and cardiac repair under pathological conditions such as atherosclerosis and acute MI. In aged hearts, PCKS6 prevents senescence in cardiomyocytes by reducing ER stress.
Ijms 23 13429 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wu, Q.; Chen, S. Proprotein Convertase Subtilisin/Kexin 6 in Cardiovascular Biology and Disease. Int. J. Mol. Sci. 2022, 23, 13429. https://doi.org/10.3390/ijms232113429

AMA Style

Wu Q, Chen S. Proprotein Convertase Subtilisin/Kexin 6 in Cardiovascular Biology and Disease. International Journal of Molecular Sciences. 2022; 23(21):13429. https://doi.org/10.3390/ijms232113429

Chicago/Turabian Style

Wu, Qingyu, and Shenghan Chen. 2022. "Proprotein Convertase Subtilisin/Kexin 6 in Cardiovascular Biology and Disease" International Journal of Molecular Sciences 23, no. 21: 13429. https://doi.org/10.3390/ijms232113429

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