Next Article in Journal
Synthesis, DFT Study, and In Vitro Evaluation of Antioxidant Properties and Cytotoxic and Cytoprotective Effects of New Hydrazones on SH-SY5Y Neuroblastoma Cell Lines
Previous Article in Journal
Efficacy of Kan Jang® in Patients with Mild COVID-19: A Randomized, Quadruple-Blind, Placebo-Controlled Trial
Previous Article in Special Issue
How to Treat Today? Oral and Facial Cancer-Associated Venous Thromboembolism
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Anti-Thrombotic Effects of PCSK9 Inhibitors

1
Department of Internal Medicine I, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 59 Martin, Slovakia
2
Department of Cardiology, Teaching Hospital Nitra, 949 01 Nitra, Slovakia
3
Oncology Centre, Teaching Hospital Martin, 036 59 Martin, Slovakia
4
Department of Hematology and Transfusiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 59 Martin, Slovakia
5
Department of Pathological Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 59 Martin, Slovakia
6
Department of Infectology and Travel Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 59 Martin, Slovakia
7
Department of Dermatovenerology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 59 Martin, Slovakia
8
Department of Medical Biology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 59 Martin, Slovakia
9
Division of Acute and Interventional Cardiology, Department of Cardiology and Angiology II, Mid-Slovakian Institute of Heart and Vessel Diseases (SÚSCCH, a.s.) in Banská Bystrica, 974 01 Banská Bystrica, Slovakia
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(9), 1197; https://doi.org/10.3390/ph16091197
Submission received: 12 July 2023 / Revised: 14 August 2023 / Accepted: 15 August 2023 / Published: 22 August 2023
(This article belongs to the Special Issue Pharmacotherapy of Thromboembolism)

Abstract

:
Atherosclerosis is the primary process that underlies cardiovascular disease. The connection between LDL cholesterol and the formation of atherosclerotic plaques is established by solid evidence. PCSK9 inhibitors have proven to be a valuable and practical resource for lowering the LDL cholesterol of many patients in recent years. Their inhibitory effect on atherosclerosis progression seems to be driven not just by lipid metabolism modification but also by LDL-independent mechanisms. We review the effect of PCSK9 inhibitors on various mechanisms involving platelet activation, inflammation, endothelial dysfunction, and the resultant clot formation. The main effectors of PCSK9 activation of platelets are CD36 receptors, lipoprotein(a), oxidised LDL particles, tissue factor, and factor VIII. Many more molecules are under investigation, and this area of research is growing rapidly.

1. Introduction

Despite new therapy options, cardiovascular events remain the most significant cause of morbidity and mortality in western countries. Atherosclerosis is the primary process that underlies cardiovascular disease. The connection between LDL cholesterol and the formation of atherosclerotic plaques has been established by years of research. Low-density lipoproteins (LDL) and high-density lipoproteins (HDL) play a central role in the transport of cholesterol [1]. The function of LDL is to transport about 60% of serum cholesterol from the liver to peripheral tissues, including the arterial walls [2]. An essential part of LDL is apolipoprotein B100 (apoB-100), which permits LDL to attach to its receptor. HDLs, in which the primary protein portion is apolipoprotein A-I, carry cholesterol from peripheral organs to the liver, enabling its elimination. The balance between HDL and LDL, and their synthesis and elimination, is essential for regulating the amount of circulating cholesterol. More than 70% of LDL is eliminated from the bloodstream by attaching to the LDL receptor in the liver [3]. Both European and American guidelines recommend a rigorous reduction in LDL cholesterol. A low LDL concentration is related to a reduction in cardiovascular events in primary and secondary prevention.
LDL cholesterol, apoB-100, and plasminogen-like apolipoprotein(a) make up the plasma protein lipoprotein(a), which is another modifiable risk factor linked to cardiovascular events [4]. LDL cholesterol and lipoprotein enter the arterial wall and are accumulated there. Lp(a) is reliant on Lp(a) plasma concentrations, Lp(a) particle size, blood pressure, and artery wall permeability. LDL cholesterol reaches the intima via the LDL receptor [5]. Both are absorbed by macrophages, which create foam cells and aid in forming atherosclerotic plaques [6].
The elimination of LDL is carried out by LDL receptors (LDLR). After binding to LDLR, the LDL part of the complex is degraded by lysosomal enzymes, and the receptor is used again on the cell membrane to bind to another LDL [7]. Lower concentrations of intracellular cholesterol result in a higher amount of surface LDL receptors on the hepatocyte membrane [8,9]. Another path for the regulation of LDL receptors is via proprotein convertase subtilisin/kexin type-9 (PCSK9). The liver is vital for the synthesis of this protein, but it is also produced in the kidney, brain, pancreas, and intestine. The role of PCSK9 is to prevent LDL receptor degradation, resulting in a reduced amount of LDLR on the cell surface [10] (Figure 1). LDL-reducing therapy is critical for a reduction in cardiovascular risk. Interest in influencing PCSK9 was increased after the finding that the excessive expression of PCSK9 in mice caused a significant increase in plasma cholesterol [11].
Inhibiting PCSK9 expression decreases serum LDL concentrations and the risk of cardiovascular disease. According to a meta-analysis of a large patient cohort, a significant positive correlation between circulating PCSK9 concentrations and the risk of major adverse cardiovascular events was found [12].
In recent years, it was shown that lipid-lowering drugs have some effects unrelated to lipid metabolism [13,14]. It appears that PCSK9 inhibitors also influence platelet function, inflammation, and endothelial dysfunction. This study aims to provide a narrative review of the possible anti-thrombotic effects of PCSK9 inhibitors (Figure 2).

2. Methods

This article is designed as a non-systematic (narrative) review. In an effort to meet these requirements, we performed a search of the three most important medical literature databases—PubMed Central, Scopus and Web of Science. We used keywords related to the topics—“PCSK9 inhibitors” or “proprotein convertase subtilisin/kexin type-9” and “thrombosis” or “venous thromboembolism” or “anti-thrombotic effect” to identify relevant articles from the recent medical literature. Afterwards, the results were reviewed by the authors and this narrative review article summarises the most recent and relevant data.

3. Proprotein Convertase Subtilis Kexin-9

Since its discovery in 2003, pro-protein convertase subtilisin/kexin type 9 (PCSK9), a soluble protease, has been the subject of a significant amount of research in the fields of cholesterol homeostasis and cardiovascular biology [15]. PCSK9, previously named Neural Apoptosis Regulated Convertase 1 (NARC1), is a member of the Pro-protein Convertases family of secretory serine proteinases [16]. PCSK9 is mainly produced in the hepatocytes of the liver and is subsequently released into circulation. The hepatocyte is not the only cell that can produce and secrete PCSK9; the cells of the intestines [17,18], adipose tissue [19], pancreas [20], brain [21] and kidneys [22] also have this ability. It is interesting to note that PCSK9 biologically fluctuates throughout the day, with late-night levels rising and late-afternoon levels falling [23]. Additionally, females have higher overall circulating PCSK9 levels than males, indicating that hormones like oestrogens are involved in the expression and secretion of PCSK9 [24]. A recent study showed that PCSK9 inhibitors are less effective at reducing LDL-c levels in women than in men, implying the possible effect of oestrogens and the importance of sex differences in the metabolism of PCSK9 [25]. It was found that multiple metabolic factors influence the levels of PCSK9, including plasma cholesterol and triglyceride levels, blood pressure, age, and body mass index [26]. The 22 kb human PCSK9 gene is found on chromosome 1p32 [16]. A 692 amino acid proteinase is encoded by its 12 exons and 11 introns [16]. PCSK9 is released in a non-active form and subsequently undergoes posttranslational modification to become a mature protein [27]. PreProPCSK9 contains five parts, but the most important is the C-terminal domain, where its M2 part is located. The M2 part plays an important role in forming the PCSK9-LDLR complex [28].

4. Platelet Activation Markers and PCSK9 Inhibitors

Recent studies have proved that PCSK9 can potentially modify platelet function. Through a variety of methods, dyslipidaemia can affect haemostasis and platelet reactivity. High LDL cholesterol concentrations relate to increased platelet reactivity and the elevated production of thromboxane. Oxidised LDL is formed because of the increased oxidative stress associated with high concentrations of circulating LDL cholesterol, significantly contributing to inflammation-driven thrombosis [29]. Oxidised LDL is the primary factor that increases platelet activation via CD36. PCSK9 binds to CD36, a negative angiogenesis regulator [30]. By identifying specific oxidised phospholipids and lipoproteins, CD36 is involved in the internalisation of apoptotic cells, bacterial and fungal infections, and LDL, triggering inflammatory reactions that alter the development of atherosclerosis. PCSK9 activates platelets after binding to CD36, and inhibitors of PCSK9 are decreasing their activity. The PCSK9—REACT study studied the relationship between PCSK9 concentrations and platelet activation. The conclusion was that increased PCSK9 is connected to platelet activation, and PCSK9 should be considered a predictor of ischemic events [31]. A recent study from Qi et al. showed that by binding to platelet receptor CD36 and activating the downstream signalling pathways, PCSK9 in the plasma directly increases platelet activation and in vivo thrombosis [32]. Platelet activation, soluble Nox2-derivated peptide, and oxidised LDL were monitored in patients with familial hypercholesterolemia before, and after, treatment with PCSK9 inhibitors. The results revealed decreased LDL, oxidised LDL, and PCSK9 concentrations [33]. Authors Cammisotto et al. proved that PCSK9 directly interacts with platelets through the CD36 receptor and activates Nox2. They showed that LDL amplifies this effect [34]. Soluble P-selectin and soluble CD40 ligands, the markers of platelet activation connected to cardiovascular risk, decreased after treatment with 10-Dehydragingerdione, which reduced PCSK9 production [35].
Recent research has demonstrated that the plasma protein PCSK9 interacts with the CD36 receptor and activates the ROS-upregulating enzymes Src kinase, mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase 5, and C-Jun amino-terminal kinase. Additionally, activating the CD36-dependent p38MAPK/cytoplasmic phospholipase A2/cysin-1/thromboxane A2 signalling pathway promoted platelet activation and thrombosis in vivo [32]. Some studies suggest that PCSK9 also plays a role in regulating CD36 and triglyceride metabolism [36]. By lowering Lp(a) concentrations, PCSK9 inhibitors may lessen the direct and indirect stimulatory effects of Lp(a) on platelets [37]. Marston et al., in their meta-analysis of the FOURIER and ODYSSEY OUTCOMES studies, demonstrated a 31% reduction in venous thromboembolism relative risk in patients treated with PCSK9 inhibitors. The reduction in this risk was associated with Lp(a) baseline concentrations, and the authors suggested Lp(a) as a marker for observation [38]. In another study, D-dimer and fibrinogen concentration changes were not observed in patients with familial hypercholesterolemia. On the other hand, a reduction in plasma PAI-1 levels was documented [39,40]. Additionally, PCSK9 inhibitors raised HDL cholesterol concentrations [41], which may prevent platelet aggregation directly or indirectly by lowering platelet membrane cholesterol [42]. Finally, PCSK9 was shown to increase apoptosis in vascular smooth muscle and endothelial cells. Cell death is known to favour thrombosis, partly through the production of procoagulant microparticles. In this situation, PCSK9 inhibitors may indirectly prevent thrombosis by reducing apoptosis [43,44,45]. Additionally, it was recently established that platelets secrete PCSK9 after activation in the presence of LDL, further increasing the platelets’ ability to aggregate and form thrombi. Furthermore, this encourages the conversion of monocytes into macrophages and foam cells, which also contributes to atherogenesis [46,47].
The impact of PCSK9 inhibitors on platelet activity highlights the interaction between PCSK9 and platelet function modification in humans. A study by Barale et al. [48] showed that platelet activation markers—sCD40L, platelet factor 4, and sP-selectin— significantly decreased after treatment with PCSK9 inhibitors. These markers correlate with PCSK9. Studies summarising the non-lipid effects of PCSK9 and PCSK9 inhibitors, mainly on the platelet activation markers, are listed in Table 1.
As mentioned above, PCSK9 levels have a positive correlation with platelet activation markers such as sCD40L, platelet factor 4, and sP-selectin. In order to predict major adverse cardiovascular events (MACEs) in coronary artery disease (CAD), circulating PCSK9 levels were proposed as a new biomarker [55]. In stable CAD patients with diabetes mellitus (DM), the studies found that baseline PCSK9 levels were independently related to the risk of MACEs. Patients with high PCSK9 levels with DM had a significantly higher risk of MACEs than those with low PCSK9 levels and non-DM [56]. These results imply that measuring plasma PCSK9 could aid in identifying diabetic people with CAD who are at higher cardiovascular risk. According to the study by authors Song et al., STEMI patients with DM undergoing primary percutaneous coronary intervention (PCI) who had high circulating PCSK9 levels were at an elevated risk of MACEs. The elevated risk of severe cardiovascular events in patients with myocardial infarction with ST segment elevation (STEMI) with high PCSK9 levels and DM may be due to the substantial association between PCSK9 and indicators of inflammation and platelet activation. The results of this study point to a possible advantage of PCSK9 inhibition in the early stages of acute coronary syndrome (ACS), particularly for patients with DM and high PCSK9 levels, by a dual mechanism on both lipid-lowering and inflammation/platelet pathways [57].

5. Endothelial Dysfunction Markers and PCSK9 Inhibitors

The endothelium plays a crucial role in maintaining vascular homeostasis [58]. One of the earliest signs of atherosclerosis, endothelial dysfunction, contributes to the development of plaques and the problems associated with atherosclerosis. The leading causes of endothelial dysfunction are cardiovascular risk factors (e.g., cholesterol concentration) and oxidative stress [59].
Several studies have shown the effect of PCSK9 inhibitors on endothelial function and the correlation between PCSK9 and endothelial dysfunction. El-Seweidy et al. studied endothelial dysfunction markers in addition to PCSK9 in a rabbit model. Their study showed a correlation between PCSK9 suppression and atherogenic and coronary risk index reduction [35]. The study from authors Maulucci et al. proved that treatment with a PCSK9 inhibitor improved endothelial function measured by mean brachial artery diameter, velocity time integral, and flow-mediated dilation (FMD) [60]. Another study used magnetic resonance imaging technology to noninvasively measure coronary endothelial function in patients living with human immunodeficiency virus infection and those with dyslipidaemia. Before PCSK9 inhibitor treatment, there was a decrease in, or no dilatation of, coronary arteries and no improvement in coronary blood flow. After treatment, the patients showed better outcomes in MRI monitoring of coronary endothelial dysfunction and increased coronary blood flow, representing an improvement in coronary artery health [61]. In vivo and ex vivo analyses proved that PCSK9 inhibitors impair endothelial dysfunction by limiting interactions among leukocytes and endothelium [54]. A possible pleiotropic effect of PCSK9 inhibitors can be found in connection to circulating endothelial progenitor cells (cEPCs). A recent study by authors Itzhaki Ben Zadok et al. on a cohort of 26 patients showed a significant increase in EPCs and VEGF after PCSK9 inhibitor treatment. Higher numbers of active cEPCs were seen in patients with CVD receiving PCSK9 inhibitor treatment, demonstrating the stimulation of endothelium repair. These results could indicate a brand-new PCSK9 inhibitor mode of action [62]. Treatment with inhibitors of PCSK9 increases cEPCs activity and differentiation into endothelial cells.
The septic state is often connected with dysfunction of the endothelium. According to a recent study, sepsis-related elevated PCSK9 expression triggered the TLR4/MyD88/NF-B and NLRP3 pathways to cause inflammation, leading to vascular endothelial dysfunction and lowered survival rates. Improved vascular endothelial function in sepsis may be achieved through the clinical therapeutic target of PCSK9 inhibition [63].
The last randomised active-controlled trial proved the cumulative benefit of adding PCSK9 inhibitors to sodium-glucose cotransporter 2 inhibitors (SGLT2i). SGLT2i are known for impairing endothelial dysfunction. Adding a PCSK9 inhibitor to SGLT2i treatment improved endothelial function assessed by FMD and the plasma concentrations of nitrate, nitrite, and isoprostane [64]. PCSK9 drugs suppress endothelial cell proinflammatory activation and reduce apoptosis in endothelial cells, smooth muscle cells, and macrophages [65]. Alirocumab, a PCSK9 inhibitor, also decreased atherosclerotic plaque vulnerability-affecting variables by a significant amount [66].
Di Minno et al. enrolled 25 patients with familial hypercholesterolemia and observed improved endothelial function after 12 weeks of PCSK9 inhibitor treatment in the administration regime every 14 days. Changes in FMD, and in the reactive hyperaemia index, were seen after 12 weeks, and the LDL score change was an independent predictor of the FMD change [67]. The ALIROCKS trial studied 24 patients before, and after, 10 weeks of treatment with a PCSK9 inhibitor. Primary outcomes of the trial were a change in carotid vessel wall fractional anisotropy, a novel magnetic resonance-based measure of vascular integrity, and a change in the carotid intima-media thickness and flow-dependent dilatation of the brachial artery measured by ultrasound. The result was that vascular endothelial growth factors and P-selectin did not alter. Although flow-dependent dilatation was somewhat improved, there were no discernible long-term benefits of PCSK9 inhibitor therapy on vascular function [68]. A specific analysis of the ODYSSEY Outcomes trial (n = 18,924 patients with acute coronary syndromes) showed a reduction in major advanced cardiovascular events (MACE). The risk of MACE after recent ACS was predicted by baseline lipoprotein(a) and corrected LDL-C concentrations and their decreases after PCSK9 inhibitor treatment. Alirocumab’s lowering of lipoprotein(a) is an independent factor in decreases in MACE, which implies that lipoprotein(a) should be a separate therapy goal following ACS [69].
In addition, compared to placebo, therapy with PSCK9 inhibitors directly decreased carotid arterial wall inflammation and reduced the volume of atherosclerotic plaques [70]. Importantly, this inflammation reduction was independent of inflammatory markers [71]. The precise mechanism through which the PCSK9 inhibitor enhances endothelial function is unknown. It is probable that a drop in LDL cholesterol concentrations mainly mediates its impact, but other pathways may also have an equal impact.
A vital component in controlling vascular function is the endothelium, which is affixed to the inside surface of blood vessels [72]. The endothelium controls the tensile strength of blood vessel walls, prevents blood from seeping into tissues, and lessens inflammation [73]. Nitric oxide (NO) is created by endothelial nitric oxide synthase (eNOS) when it is joined to its co-factor, tetrahydrobiopterin (BH4). The NO, an endothelial vasodilator, is diminished prior to the formation of atherosclerotic plaques. Endothelial dysfunction and atherosclerosis are both thought to be significantly mediated by defects in NO generation or activity. The dysfunction of eNOS is strongly associated with the pathogenesis of arteriosclerosis, hypertension, myocardial infarction, and stroke [73]. The eNOS affects endothelial cell function directly and plays a crucial role in the regulation of vasodilatation [74]. A recent study discovered that inhibiting PCSK9 reversed the decline in eNOS expression brought on by sepsis; the results supported the author’s theory that sepsis-induced endothelium dysfunction enhanced PCSK9 expression [63]. The in vitro study by authors Zulkapli et al. showed that the co-incubation of alirocumab and evolocumab at specific concentrations resulted in an upregulation of the production of the eNOS protein. Only evolocumab caused an increase, although not significant, in eNOS mRNA [75]. Treatment with a PCSK9 inhibitor given to type 2 diabetes (T2D) patients (110 individuals) along with SGLT2i treatment improved FMD and the bioavailability of NO. Increased NO may contribute towards improving endothelial dysfunction [64].

6. Inflammation Markers and PCSK9 Inhibitors

In contrast with statins, PCSK9 inhibitors do not reduce concentrations of high-sensitive CRP (hs-CRP), as demonstrated by several studies and a meta-analysis [76,77]. The same results were obtained regarding concentrations of TNF-α and IL-6 [76,78]. On the other hand, it was established that hs-CRP is positively correlated with PCSK9 concentrations in chronic coronary disease and acute coronary syndrome [58,79]. PCSK9 concentrations are also elevated in patients with systemic inflammatory response syndrome and sepsis [80]. Although hepatocytes are the primary site of PCSK9 production, vascular endothelial and smooth muscle cells express PCSK9 under the regulation of proinflammatory molecules such as IL-1β, TNF-α, and lipopolysaccharide [81]. Therefore, one could argue that, from a clinical perspective, PCSK9 plays more of a role as a by-product of inflammation than as a modulator by itself.
From experimental studies, the potential inflammatory input of PCSK9 could be mediated by the LDLR and the ensuing increased migration of monocytes into the atherosclerotic plaque [82,83]. This was partly presented by a clinical study showing a PCSK9 association with plaque formation independent of LDL concentrations and other traditional risk factors [84]. Another mechanism by which PCSK9 can modulate the migration of monocytes is the chemokine receptor type-2, which was inhibited by PCSK9 inhibitors in patients with familial hypercholesterolemia and decreased the capacity of monocytes to pass the endothelium [85].
It is theorised that PCSK9 upregulates inflammation through the increased expression of TLR-4 and LOX-1, which activate NF-kB and set off the inflammation cascade of other modulators [86]. Another study that evaluated vessel inflammation by PET/CT scanning showed a decrease in the target-to-background ratio after long-term administration of PCSK9 inhibitors [87].
Altogether, there are several theories for the positive LDL-independent effect of PCSK9 inhibition concerning inflammation and following atherothrombosis, but the actual clinical benefit of this data still needs to be evaluated.

7. Coagulation Factors and PCSK9 Inhibitors

Research on animals and humans indicates a connection between PCSK9 and thrombotic risk. The animal models showed that after ligation of the inferior vena cava and the subsequent venous thrombosis, PCSK9-/- mice had a comparatively shorter thrombus length, and demonstrated decreased neutrophil extracellular trap (NET) production (NETosis), and leucocyte attachment and accumulation [88]. Leucocyte recruitment was connected to an increased CXCL1. A potential mechanism by which PCSK9 promotes NETosis-induced thrombosis is the recruitment of myeloid cells [39,89]. The study by authors Schuster et al. demonstrated the decreased production of inflammatory molecules after PCSK9 inhibition in mice models [90]. This pathway between PCSK9 and NETosis is one of the possible connections in clot formation.
Levine et al. discovered a downregulation of PCSK9 expression in mice with either pharmacological or genetic PAI-1 inhibition when examining the potential relationship between PCSK9 and fibrinolysis. This was also shown in humans with a mutation that decreased the levels of PAI-1. Additionally, the investigators discovered a significant association between PAI-1 and PCSK9 levels in people with heart failure, pointing to a possible interaction between PCSK9 and the fibrinolytic process [38]. On the other hand, PCSK9 inhibitor treatment decreased PAI-1 levels and increased fibrinolytic activity [91,92].
A higher level of PCSK9 was found in patients at high thrombotic risk with antiphospholipid antibodies [93]. A favorable correlation between PCSK9 levels and fibrinogen levels was also found in patients with stable coronary artery disease [79]; these data show a positive correlation between PCSK9 and the coagulation system [40].
Authors Wang et al. showed a positive association between PCSK9 expression and the levels of tissue factor in patients with coronary artery disease and diabetes [94]. The tissue factor is considered a prothrombotic element, and it is regulated by LDL receptor-related protein-1 (LRP-1) [95]. LRP-1 encourages the degradation of tissue factor. LRP-1 expression is downregulated by PCSK9, which could impact circulating TF levels. Additionally, PCSK9 was shown by authors Scalise et al. to promote TF expression on monocytes, which boost procoagulant activity. The TLR4/NF-B pathway’s activation mediates this mechanism [96]. Both the extracellular vesicle-associated tissue factor and extracellular vesicle formation by procoagulant extracellular vesicles from human mononuclear cells (PBMCs) and THP-1 cells increased due to PCSK9. Pre-treatment with TLR4 (C34) and NF-B signalling (BAY 11-7082) inhibitors reduced the production of extracellular vesicles and extracellularly bound tissue factor triggered by PCSK9. An anti-PCSK9 human monoclonal antibody had a similar effect [97]. In conclusion, PCKS9 can both directly and indirectly boost TF expression (Figure 3).
Through an impact on the blood-clotting factor VIII (FVIII), PCSK9 can also modify blood coagulation [18,98]. An increased arterial and venous thrombosis risk is linked to higher FVIII levels. FVII plays a crucial role in thrombin formation and propagation of the coagulation cascade [99,100]. By facilitating its endocytosis and degradation, LRP-1 inhibits FVIII [101]. This is a potential explanation for how PCSK9 can further contribute to thrombogenesis because it has been demonstrated that it also increases FVIII levels via decreasing LRP-1 expression [102]. It was shown that anti-PCSK9 reduces FVIII levels by enhancing LRP-1 expression in mouse models [103].

8. Other Possible Implications of PCSK9 Inhibitors

Overexpression of PCSK9 was found in different cancer types, including colorectal cancer [104], breast cancer [105], lymphoblastic leukaemia [106], hepatocellular carcinoma [107] and gastric cancer [108]. Samples from the cancer regions compared to normal areas showed increased levels of PCSK9 [109]. Additionally, the increased PCSK9 levels were associated with a worse prognosis in many cancers [110]; low levels of PCSK9 are connected to reduced tumour growth and improved prognosis [111]. These results indicate that inhibiting PCSK9 could halt the spread of cancer and suggest the use of PCSK9 as a biomarker of prognosis and response to therapy. The inhibition of PCSK9 caused apoptosis in cancer models in vitro [112,113]. The pathways of the increased apoptosis due to PCSK9 inhibition are mitochondrial signalling pathways and the activation of endoplasmic reticulum stress [114]. The effect of the downregulation is not universal, in a mice model of melanoma, the inhibition of PCSK9 did not demonstrate growth reduction or a better prognosis [115]. On the other hand, in the case of hepatocellular carcinoma, in vivo and in vitro studies proved that PCSK9 has a protective influence against cancer growth and spread [116]. These data imply that, in contrast to the patterns observed with the malignancies previously mentioned, hepatocellular cancer may be an example of a cancer in which the downregulation of PCSK9 may have adverse effects. According to a study from authors Ioannou et al., while PCSK9 overexpression of LDLR on hepatocytes lowered plasma cholesterol and may suppress other malignancies, the increased exposure of the liver to cholesterol may contribute to the development of hepatocarcinogenesis [117]. It was demonstrated that in CD8+ T cells, LDL-R combines with the T-cell receptor (TCR), activating it when it binds to antigenic peptides that the major histocompatibility complex (MHC) presents to tumour cells. The activation of CD8+ T lymphocytes’ antitumor response is increased by the binding of LDL-R to TCR, which encourages cell surface recycling [109].
Viral infections (hepatitis C virus and dengue fever virus) might be associated with PCSK9. Studies identify LDL-R as a binding receptor for the entrance of hepatitis C virus (HCV) into the liver cell [118]. However, virus production is dramatically boosted following LDL-R re-expression, demonstrating that LDL-R is not associated with HCV entry but rather is directly associated with the lipid metabolism of host cells during HCV packaging. Cells lacking functional LDL-R can still become infected with HCV. Patients with HCV should take PCSK9 inhibitors cautiously since they can increase HCV packaging and infectivity [119].
During sepsis, circulating bacterial lipids are assumed to play a crucial role in causing unchecked systemic inflammatory reactions [120]. These lipids, such as LPS, are cleared by LDL-R on the liver cells. PCSK9 inhibitors reduce the density of LDL-R. Based on this knowledge, treatment with PCSK9 inhibitors can represent a new method of sepsis treatment. Indeed, a mice PCSK9-/- model was shown to have lower bacterial numbers in the blood, lungs, and peritoneal fluid than wild-type animals in sepsis models of caecal ligation and perforation, indicating that the deletion of PCSK9 is advantageous for bacterial suppression or clearance [121]. A sub-analysis of the ALBIOS trial study by the authors Vecchié et al. found that patients with septic shock and low plasma levels of PCSK9 had higher mortality rates [122]. A recent study by the authors Zhou et al. did not find a positive or negative connection among PCSK9 inhibitors, sepsis, and severe infections in populations with high cardiovascular risk and does not support the use of PCSK9 inhibitors in preventing sepsis [123].

9. Conclusions

The lipid-lowering effect of PCSK9 inhibitors is known and well-documented. PCSK9 stimulates platelets, heightens inflammation, and shifts the balance between fibrinolysis and coagulation in favour of coagulation. PCSK9 inhibitors with evidence of lowering cardiovascular morbidity and mortality decrease Lp(a) levels through unknown processes and reduce LDL cholesterol by boosting the number of LDL receptors. VTE is associated with Lp(a) levels, and the Lp(a) reduction correlates with decreased VTE incidence. It is assumed that PCSK9 alters primary and secondary haemostasis through changing platelet activity, TF, PAI-1, NETosis, and FVIII levels, and through the effect on LDL-C metabolism. Many of these above-mentioned mechanisms of action require clinical studies to be confirmed. In the future, patients could benefit from the monitoring of PCSK9 levels in serum because of the connection between platelet activation and endothelial dysfunction. The potential for expanding the indications for this new therapeutic class and clarifying their potential role in the treatment of the acute phase of ischemic cardiovascular disease may result from a thorough disentanglement of the potential pleiotropic activities of PCSK9 inhibitors, particularly of their potential anti-thrombotic effects.

Author Contributions

M.S., M.J.P. and J.B. designed the study; M.J.P. and M.P. (Monika Péčová) drafted the manuscript; M.J.P., J.J., T.H. and M.P. (Monika Péčová) performed the search of the literature, analysed, and interpreted the data; P.G., T.B., M.S. (Matej Samoš), M.S. (Marek Samec), M.M., M.P. (Martin Péč) revised the manuscript critically. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the project Operational Programme Integrated Infrastructure ITMS 2014+:313011V344 and by the research project of Comenius University in Bratislava (UK/318/2022).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Acknowledgments

This study was supported by the project Operational Programme Integrated Infrastructure ITMS 2014+:313011V344 and by the research project of Comenius University in Bratislava (UK/318/2022). Figures were created by Jakub Benko and Martin Jozef Péč.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Goldstein, J.L.; Brown, M.S. Regulation of low-density lipoprotein receptors: Implications for pathogenesis and therapy of hypercholesterolemia and atherosclerosis. Circulation 1987, 76, 504–507. [Google Scholar] [CrossRef] [PubMed]
  2. Santos-Gallego, C.G.; Badimon, J.J.; Rosenson, R.S. Beginning to understand high-density lipoproteins. Endocrinol. Metab. Clin. North Am. 2014, 43, 913–947. [Google Scholar] [CrossRef]
  3. Elshourbagy, N.A.; Meyers, H.V.; Abdel-Meguid, S.S. Cholesterol: The Good, the Bad, and the Ugly—Therapeutic Targets for the Treatment of Dyslipidemia. Med. Princ. Pract. 2013, 23, 99–111. [Google Scholar] [CrossRef] [PubMed]
  4. Borén, J.; Chapman, M.J.; Krauss, R.M.; Packard, C.J.; Bentzon, J.F.; Binder, C.J.; Daemen, M.J.; Demer, L.L.; Hegele, R.A.; Nicholls, S.J.; et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease: Pathophysiological, genetic, and therapeutic insights: A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 2020, 41, 2313–2330. [Google Scholar] [CrossRef]
  5. Cybulska, B.; Kłosiewicz-Latoszek, L.; Penson, P.E.; Banach, M. What do we know about the role of lipoprotein(a) in atherogenesis 57 years after its discovery? Prog. Cardiovasc. Dis. 2020, 63, 219–227. [Google Scholar] [CrossRef]
  6. Kiechl, S.; Willeit, J. The Mysteries of Lipoprotein(a) and Cardiovascular Disease Revisited. J. Am. Coll. Cardiol. 2010, 55, 2168–2170. [Google Scholar] [CrossRef]
  7. Segrest, J.P.; Jones, M.K.; De Loof, H.; Dashti, N. Structure of apolipoprotein B-100 in low density lipoproteins. J. Lipid Res. 2001, 42, 1346–1367. [Google Scholar] [CrossRef] [PubMed]
  8. Brown, M.S.; Anderson, R.G.; Basu, S.K.; Goldstein, J.L. Recycling of cell-surface receptors: Observations from the LDL receptor system. Cold Spring Harb. Symp. Quant. Biol. 1982, 46 Pt 2, 713–721. [Google Scholar] [CrossRef]
  9. Brown, M.S.; Goldstein, J.L. A receptor-mediated pathway for cholesterol homeostasis. Science 1986, 232, 34–47. [Google Scholar] [CrossRef]
  10. Zhang, D.W.; Garuti, R.; Tang, W.J.; Cohen, J.C.; Hobbs, H.H. Structural requirements for PCSK9-mediated degradation of the low-density lipoprotein receptor. Proc. Natl. Acad. Sci. USA 2008, 105, 13045–13050. [Google Scholar] [CrossRef]
  11. Maxwell, K.N.; Breslow, J.L. Adenoviral-mediated expression of Pcsk9 in mice results in a low-density lipoprotein receptor knockout phenotype. Proc. Natl. Acad. Sci. USA 2004, 101, 7100–7105. [Google Scholar] [CrossRef] [PubMed]
  12. Zhou, Y.; Chen, W.; Lu, M.; Wang, Y. Association Between Circulating Proprotein Convertase Subtilisin/Kexin Type 9 and Major Adverse Cardiovascular Events, Stroke, and All-Cause Mortality: Systemic Review and Meta-Analysis. Front. Cardiovasc. Med. 2021, 8, 617249. [Google Scholar] [CrossRef]
  13. Ugovšek, S.; Šebeštjen, M. Non-Lipid Effects of PCSK9 Monoclonal Antibodies on Vessel Wall. J. Clin. Med. 2022, 11, 3625. [Google Scholar] [CrossRef]
  14. Zulkapli, R.; Yusof, M.Y.P.M.; Abd Muid, S.; Wang, S.M.; Firus Khan, A.Y.; Nawawi, H. A Systematic Review on Attenuation of PCSK9 in Relation to Atherogenesis Biomarkers Associated with Natural Products or Plant Bioactive Compounds in In Vitro Studies: A Critique on the Quality and Imprecision of Studies. Int. J. Environ. Res. Public Health 2022, 19, 12878. [Google Scholar] [CrossRef]
  15. Seidah, N.; Awan, Z.; Chrétien, M.; Mbikay, M. PCSK9. Circ. Res. 2014, 114, 1022–1036. [Google Scholar] [CrossRef] [PubMed]
  16. Ragusa, R.; Basta, G.; Neglia, D.; De Caterina, R.; Del Turco, S.; Caselli, C. PCSK9 and atherosclerosis: Looking beyond LDL regulation. Eur. J. Clin. Investig. 2021, 51, e13459. [Google Scholar] [CrossRef]
  17. Leblond, F.; Seidah, N.G.; Précourt, L.-P.; Delvin, E.; Dominguez, M.; Levy, E. Regulation of the proprotein convertase subtilisin/kexin type 9 in intestinal epithelial cells. Am. J. Physiol. Liver Physiol. 2009, 296, G805–G815. [Google Scholar] [CrossRef]
  18. Luquero, A.; Badimon, L.; Borrell-Pages, M. PCSK9 Functions in Atherosclerosis Are Not Limited to Plasmatic LDL-Cholesterol Regulation. Front. Cardiovasc. Med. 2021, 8, 639727. [Google Scholar] [CrossRef]
  19. Rohrbach, S.; Li, L.; Novoyatleva, T.; Niemann, B.; Knapp, F.; Molenda, N.; Schulz, R. Impact of PCSK9 on CTRP9-Induced Metabolic Effects in Adult Rat Cardiomyocytes. Front. Physiol. 2021, 12, 593862. [Google Scholar] [CrossRef]
  20. Langhi, C.; Le May, C.; Gmyr, V.; Vandewalle, B.; Kerr-Conte, J.; Krempf, M.; Pattou, F.; Costet, P.; Cariou, B. PCSK9 is expressed in pancreatic δ-cells and does not alter insulin secretion. Biochem. Biophys. Res. Commun. 2009, 390, 1288–1293. [Google Scholar] [CrossRef]
  21. O’Connell, E.M.; Lohoff, F.W. Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) in the Brain and Relevance for Neuropsychiatric Disorders. Front. Neurosci. 2020, 14, 609. [Google Scholar] [CrossRef]
  22. Artunc, F. Kidney-derived PCSK9—A new driver of hyperlipidemia in nephrotic syndrome? Kidney Int. 2020, 98, 1393–1395. [Google Scholar] [CrossRef]
  23. Persson, L.; Cao, G.; Ståhle, L.; Sjöberg, B.G.; Troutt, J.S.; Konrad, R.J.; Gälman, C.; Wallén, H.; Eriksson, M.; Hafström, I.; et al. Circulating Proprotein Convertase Subtilisin Kexin Type 9 Has a Diurnal Rhythm Synchronous with Cholesterol Synthesis and Is Reduced by Fasting in Humans. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 2666–2672. [Google Scholar] [CrossRef] [PubMed]
  24. Ferri, N.; Ruscica, M.; Coggi, D.; Bonomi, A.; Amato, M.; Frigerio, B.; Sansaro, D.; Ravani, A.; Veglia, F.; Capra, N.; et al. Sex-specific predictors of PCSK9 levels in a European population: The IMPROVE study. Atherosclerosis 2020, 309, 39–46. [Google Scholar] [CrossRef]
  25. Galema-Boers, A.M.H.; Mulder, J.W.C.M.; Steward, K.; Roeters van Lennep, J.E. Sex differences in efficacy and safety of PCSK9 monoclonal antibodies: A real-world registry. Atherosclerosis 2023, S0021-9150(23)00115-6. [Google Scholar] [CrossRef]
  26. Cui, Q.; Ju, X.; Yang, T.; Zhang, M.; Tang, W.; Chen, Q.; Hu, Y.; Haas, J.V.; Troutt, J.S.; Pickard, R.T.; et al. Serum PCSK9 is associated with multiple metabolic factors in a large Han Chinese population. Atherosclerosis 2010, 213, 632–636. [Google Scholar] [CrossRef]
  27. Sundararaman, S.S.; Döring, Y.; van der Vorst, E.P.C. PCSK9: A Multi-Faceted Protein That Is Involved in Cardiovascular Biology. Biomedicines 2021, 9, 793. [Google Scholar] [CrossRef] [PubMed]
  28. Saavedra, Y.G.L.; Day, R.; Seidah, N.G. The M2 Module of the Cys-His-rich Domain (CHRD) of PCSK9 Protein Is Needed for the Extracellular Low-density Lipoprotein Receptor (LDLR) Degradation Pathway. J. Biol. Chem. 2012, 287, 43492–43501. [Google Scholar] [CrossRef] [PubMed]
  29. Obermayer, G.; Afonyushkin, T.; Binder, C.J. Oxidized low-density lipoprotein in inflammation-driven thrombosis. J. Thromb. Haemost. 2018, 16, 418–428. [Google Scholar] [CrossRef]
  30. Liu, C.; Chen, J.; Chen, H.; Zhang, T.; He, D.; Luo, Q.; Chi, J.; Hong, Z.; Liao, Y.; Zhang, S.; et al. PCSK9 Inhibition: From Current Advances to Evolving Future. Cells 2022, 11, 2972. [Google Scholar] [CrossRef]
  31. Navarese, E.P.; Kolodziejczak, M.; Winter, M.-P.; Alimohammadi, A.; Lang, I.M.; Buffon, A.; Lip, G.Y.; Siller-Matula, J.M. Association of PCSK9 with platelet reactivity in patients with acute coronary syndrome treated with prasugrel or ticagrelor: The PCSK9-REACT study. Int. J. Cardiol. 2017, 227, 644–649. [Google Scholar] [CrossRef]
  32. Qi, Z.; Hu, L.; Zhang, J.; Yang, W.; Liu, X.; Jia, D.; Yao, Z.; Chang, L.; Pan, G.; Zhong, H.; et al. PCSK9 (Proprotein Convertase Subtilisin/Kexin 9) Enhances Platelet Activation, Thrombosis, and Myocardial Infarct Expansion by Binding to Platelet CD36. Circulation. 2021, 143, 45–61. [Google Scholar] [CrossRef] [PubMed]
  33. Cammisotto, V.; Baratta, F.; Castellani, V.; Bartimoccia, S.; Nocella, C.; D’Erasmo, L.; Cocomello, N.; Barale, C.; Scicali, R.; Di Pino, A.; et al. Proprotein Convertase Subtilisin Kexin Type 9 Inhibitors Reduce Platelet Activation Modulating ox-LDL Pathways. Int. J. Mol. Sci. 2021, 22, 7193. [Google Scholar] [CrossRef]
  34. Cammisotto, V.; Pastori, D.; Nocella, C.; Bartimoccia, S.; Castellani, V.; Marchese, C.; Scavalli, A.S.; Ettorre, E.; Viceconte, N.; Violi, F.; et al. PCSK9 Regulates Nox2-Mediated Platelet Activation via CD36 Receptor in Patients with Atrial Fibrillation. Antioxidants 2020, 9, 296. [Google Scholar] [CrossRef]
  35. El-Seweidy, M.M.; Sarhan Amin, R.; Husseini Atteia, H.; El-Zeiky, R.R.; Al-Gabri, N.A. Dyslipidemia induced inflammatory status, platelet activation and endothelial dysfunction in rabbits: Protective role of 10-Dehydrogingerdione. Biomed. Pharmacother. 2019, 110, 456–464. [Google Scholar] [CrossRef]
  36. Demers, A.; Samami, S.; Lauzier, B.; Des Rosiers, C.; Ngo Sock, E.; Ong, H.; Mayer, G. PCSK9 Induces CD36 Degradation and Affects Long-Chain Fatty Acid Uptake and Triglyceride Metabolism in Adipocytes and in Mouse Liver. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 2517–2525. [Google Scholar] [CrossRef]
  37. Gaudet, D.; Kereiakes, D.J.; McKenney, J.M.; Roth, E.M.; Hanotin, C.; Gipe, D.; Du, Y.; Ferrand, A.-C.; Ginsberg, H.N.; Stein, E.A. Effect of alirocumab, a monoclonal proprotein convertase subtilisin/kexin 9 antibody, on lipoprotein(a) concentrations (a pooled analysis of 150 mg every two weeks dosing from phase 2 trials). Am. J. Cardiol. 2014, 114, 711–715. [Google Scholar] [CrossRef]
  38. Marston, N.A.; Gurmu, Y.; Melloni, G.E.M.; Bonaca, M.; Gencer, B.; Sever, P.S.; Pedersen, T.R.; Keech, A.C.; Roselli, C.; Lubitz, S.A.; et al. The Effect of PCSK9 (Proprotein Convertase Subtilisin/Kexin Type 9) Inhibition on the Risk of Venous Thromboembolism. Circulation 2020, 141, 1600–1607. [Google Scholar] [CrossRef] [PubMed]
  39. Levine, J.A.; Oleaga, C.; Eren, M.; Amaral, A.P.; Shang, M.; Lux, E.; Khan, S.S.; Shah, S.J.; Omura, Y.; Pamir, N.; et al. Role of PAI-1 in hepatic steatosis and dyslipidemia. Sci. Rep. 2021, 11, 430. [Google Scholar] [CrossRef]
  40. Puccini, M.; Landmesser, U.; Rauch, U. Pleiotropic Effects of PCSK9: Focus on Thrombosis and Haemostasis. Metabolites 2022, 12, 226. [Google Scholar] [CrossRef] [PubMed]
  41. Filippatos, T.D.; Kei, A.; Rizos, C.V.; Elisaf, M.S. Effects of PCSK9 Inhibitors on Other than Low-Density Lipoprotein Cholesterol Lipid Variables. J. Cardiovasc. Pharmacol. Ther. 2018, 23, 3–12. [Google Scholar] [CrossRef]
  42. Pęczek, P.; Leśniewski, M.; Mazurek, T.; Szarpak, L.; Filipiak, K.J.; Gąsecka, A. Antiplatelet Effects of PCSK9 Inhibitors in Primary Hypercholesterolemia. Life 2021, 11, 466. [Google Scholar] [CrossRef] [PubMed]
  43. Li, J.; Liang, X.; Wang, Y.; Xu, Z.; Li, G. Investigation of highly expressed PCSK9 in atherosclerotic plaques and ox-LDL-induced endothelial cell apoptosis. Mol. Med. Rep. 2017, 16, 1817–1825. [Google Scholar] [CrossRef]
  44. Paciullo, F.; Momi, S.; Gresele, P. PCSK9 in Haemostasis and Thrombosis: Possible Pleiotropic Effects of PCSK9 Inhibitors in Cardiovascular Prevention. Thromb. Haemost. 2019, 119, 359–367. [Google Scholar] [CrossRef]
  45. Barale, C.; Melchionda, E.; Morotti, A.; Russo, I. PCSK9 Biology and Its Role in Atherothrombosis. Int. J. Mol. Sci. 2021, 22, 5880. [Google Scholar] [CrossRef]
  46. Petersen-Uribe, A.; Kremser, M.; Rohlfing, A.K.; Castor, T.; Kolb, K.; Dicenta, V.; Emschermann, F.; Li, B.; Borst, O.; Rath, D.; et al. Platelet-Derived PCSK9 Is Associated with LDL Metabolism and Modulates Atherothrombotic Mechanisms in Coronary Artery Disease. Int. J. Mol. Sci. 2021, 22, 11179. [Google Scholar] [CrossRef] [PubMed]
  47. Cammisotto, V.; Baratta, F.; Simeone, P.G.; Barale, C.; Lupia, E.; Galardo, G.; Santilli, F.; Russo, I.; Pignatelli, P. Proprotein Convertase Subtilisin Kexin Type 9 (PCSK9) Beyond Lipids: The Role in Oxidative Stress and Thrombosis. Antioxidants 2022, 11, 569. [Google Scholar] [CrossRef]
  48. Barale, C.; Bonomo, K.; Frascaroli, C.; Morotti, A.; Guerrasio, A.; Cavalot, F.; Russo, I. Platelet function and activation markers in primary hypercholesterolemia treated with anti-PCSK9 monoclonal antibody: A 12-month follow-up. Nutr. Metab. Cardiovasc. Dis. 2020, 30, 282–291. [Google Scholar] [CrossRef]
  49. Li, S.; Zhu, C.-G.; Guo, Y.-L.; Xu, R.-X.; Zhang, Y.; Sun, J.; Li, J.-J. The relationship between the plasma PCSK9 levels and platelet indices in patients with stable coronary artery disease. J. Atheroscler. Thromb. 2015, 22, 76–84. [Google Scholar] [CrossRef] [PubMed]
  50. Pastori, D.; Nocella, C.; Farcomeni, A.; Bartimoccia, S.; Santulli, M.; Vasaturo, F.; Carnevale, R.; Menichelli, D.; Violi, F.; Pignatelli, P.; et al. Relationship of PCSK9 and Urinary Thromboxane Excretion to Cardiovascular Events in Patients with Atrial Fibrillation. J. Am. Coll. Cardiol. 2017, 70, 1455–1462. [Google Scholar] [CrossRef]
  51. El-Seweidy, M.M.; Amin, R.S.; Atteia, H.H.; El-Zeiky, R.R.; Al-Gabri, N.A. New Insight on a Combination of Policosanol and 10-Dehydrogingerdione Phytochemicals as Inhibitors for Platelet Activation Biomarkers and Atherogenicity Risk in Dyslipidemic Rabbits: Role of CETP and PCSK9 Inhibition. Appl. Biochem. Biotechnol. 2018, 186, 805–815. [Google Scholar] [CrossRef]
  52. Koskinas, K.C.; Windecker, S.; Buhayer, A.; Gencer, B.; Pedrazzini, G.; Mueller, C.; Cook, S.; Muller, O.; Matter, C.M.; Räber, L.; et al. Design of the randomized, placebo-controlled evolocumab for early reduction of LDL-cholesterol levels in patients with acute coronary syndromes (EVOPACS) trial. Clin. Cardiol. 2018, 41, 1513–1520. [Google Scholar] [CrossRef]
  53. Di Minno, A.; Orsini, R.C.; Chiesa, M.; Cavalca, V.; Calcaterra, I.; Tripaldella, M.; Anesi, A.; Fiorelli, S.; Eligini, S.; Colombo, G.I.; et al. Treatment with PCSK9 Inhibitors in Patients with Familial Hypercholesterolemia Lowers Plasma Levels of Platelet-Activating Factor and Its Precursors: A Combined Metabolomic and Lipidomic Approach. Biomedicines 2021, 9, 1073. [Google Scholar] [CrossRef]
  54. Marques, P.; Domingo, E.; Rubio, A.; Martinez-Hervás, S.; Ascaso, J.F.; Piqueras, L.; Real, J.T.; Sanz, M.-J. Beneficial effects of PCSK9 inhibition with alirocumab in familial hypercholesterolemia involve modulation of new immune players. Biomed. Pharmacother. 2022, 145, 112460. [Google Scholar] [CrossRef]
  55. Werner, C.; Hoffmann, M.M.; Winkler, K.; Böhm, M.; Laufs, U. Risk prediction with proprotein convertase subtilisin/kexin type 9 (PCSK9) in patients with stable coronary disease on statin treatment. Vascul Pharmacol. 2014, 62, 94–102. [Google Scholar] [CrossRef] [PubMed]
  56. Peng, J.; Liu, M.-M.; Jin, J.-L.; Cao, Y.-X.; Guo, Y.-L.; Wu, N.-Q.; Zhu, C.-G.; Dong, Q.; Sun, J.; Xu, R.-X.; et al. Association of circulating PCSK9 concentration with cardiovascular metabolic markers and outcomes in stable coronary artery disease patients with or without diabetes: A prospective, observational cohort study. Cardiovasc. Diabetol. 2020, 19, 167. [Google Scholar] [CrossRef] [PubMed]
  57. Song, L.; Zhao, X.; Chen, R.; Li, J.; Zhou, J.; Liu, C.; Zhou, P.; Wang, Y.; Chen, Y.; Zhao, H.; et al. Association of PCSK9 with inflammation and platelet activation markers and recurrent cardiovascular risks in STEMI patients undergoing primary PCI with or without diabetes. Cardiovasc. Diabetol. 2022, 21, 80. [Google Scholar] [CrossRef] [PubMed]
  58. Gencer, B.; Montecucco, F.; Nanchen, D.; Carbone, F.; Klingenberg, R.; Vuilleumier, N.; Aghlmandi, S.; Heg, D.; Räber, L.; Auer, R.; et al. Prognostic value of PCSK9 levels in patients with acute coronary syndromes. Eur. Heart J. 2016, 37, 546–553. [Google Scholar] [CrossRef] [PubMed]
  59. Heitzer, T.; Schlinzig, T.; Krohn, K.; Meinertz, T.; Münzel, T. Cardiovascular events in patients with coronary artery disease. Circulation 2001, 104, 673–2678. [Google Scholar] [CrossRef]
  60. Maulucci, G.; Cipriani, F.; Russo, D.; Casavecchia, G.; Di Staso, C.; Di Martino, L.; Ruggiero, A.; Di Biase, M.; Brunetti, N.D. Improved endothelial function after short-term therapy with evolocumab. J. Clin. Lipidol. 2018, 12, 669–673. [Google Scholar] [CrossRef]
  61. Leucker, T.M.; Gerstenblith, G.; Schär, M.; Brown, T.T.; Jones, S.R.; Afework, Y.; Weiss, R.G.; Hays, A.G. Evolocumab, a PCSK9-Monoclonal Antibody, Rapidly Reverses Coronary Artery Endothelial Dysfunction in People Living with HIV and People With Dyslipidemia. J. Am. Heart Assoc. 2020, 9, e016263. [Google Scholar] [CrossRef]
  62. Itzhaki Ben Zadok, O.; Mager, A.; Leshem-Lev, D.; Lev, E.; Kornowski, R.; Eisen, A. The Effect of Proprotein Convertase Subtilisin Kexin Type 9 Inhibitors on Circulating Endothelial Progenitor Cells in Patients with Cardiovascular Disease. Cardiovasc. Drugs Ther. 2022, 36, 85–92. [Google Scholar] [CrossRef]
  63. Huang, L.; Li, Y.; Cheng, Z.; Lv, Z.; Luo, S.; Xia, Y. PCSK9 Promotes Endothelial Dysfunction During Sepsis Via the TLR4/MyD88/NF-κB and NLRP3 Pathways. Inflammation 2022, 46, 115–128. [Google Scholar] [CrossRef]
  64. Sposito, A.C.; Breder, I.; Barreto, J.; Breder, J.; Bonilha, I.; Lima, M.; Oliveira, A.; Wolf, V.; Luchiari, B.; Carmo, H.R.D.; et al. Evolocumab on top of empagliflozin improves endothelial function of individuals with diabetes: Randomized active-controlled trial. Cardiovasc. Diabetol. 2022, 21, 147. [Google Scholar] [CrossRef]
  65. Karagiannis, A.D.; Liu, M.; Toth, P.P.; Zhao, S.; Agrawal, D.K.; Libby, P.; Chatzizisis, Y.S. Pleiotropic Anti-atherosclerotic Effects of PCSK9 Inhibitors from Molecular Biology to Clinical Translation. Curr. Atheroscler. Rep. 2018, 20, 20. [Google Scholar] [CrossRef]
  66. Kosowski, M.; Basiak, M.; Hachuła, M.; Okopień, B. Impact of Alirocumab on Release Markers of Atherosclerotic Plaque Vulnerability in Patients with Mixed Hyperlipidemia and Vulnerable Atherosclerotic Plaque. Medicina 2022, 58, 969. [Google Scholar] [CrossRef]
  67. Di Minno, A.; Gentile, M.; Iannuzzo, G.; Calcaterra, I.; Tripaldella, M.; Porro, B.; Cavalca, V.; Di Taranto, M.D.; Tremoli, E.; Fortunato, G.; et al. Endothelial Function Improvement in Patients with Familial Hypercholesterolemia Receiving PCSK-9 Inhibitors on Top of Maximally Tolerated Lipid Lowering Therapy. Thromb. Res. 2020, 194, 229–236. [Google Scholar] [CrossRef]
  68. Metzner, T.; Leitner, D.R.; Dimsity, G.; Gunzer, F.; Opriessnig, P.; Mellitzer, K.; Beck, A.; Sourij, H.; Stojakovic, T.; Deutschmann, H.; et al. Short-Term Treatment with Alirocumab, Flow-Dependent Dilatation of the Brachial Artery and Use of Magnetic Resonance Diffusion Tensor Imaging to Evaluate Vascular Structure: An Exploratory Pilot Study. Biomedicines 2022, 10, 152. [Google Scholar] [CrossRef]
  69. Bittner, V.A.; Szarek, M.; Aylward, P.E.; Bhatt, D.L.; Diaz, R.; Edelberg, J.M.; Fras, Z.; Goodman, S.G.; Halvorsen, S.; Hanotin, C.; et al. Effect of Alirocumab on Lipoprotein(a) and Cardiovascular Risk After Acute Coronary Syndrome. J. Am. Coll. Cardiol. 2020, 75, 133–144. [Google Scholar] [CrossRef]
  70. Ricci, C.; Ruscica, M.; Camera, M.; Rossetti, L.; Macchi, C.; Colciago, A.; Zanotti, I.; Lupo, M.G.; Adorni, M.P.; Cicero, A.F.G.; et al. PCSK9 induces a pro-inflammatory response in macrophages. Sci. Rep. 2018, 8, 2267. [Google Scholar] [CrossRef]
  71. Hoogeveen, R.M.; Opstal, T.S.J.; Kaiser, Y.; Stiekema, L.C.A.; Kroon, J.; Knol, R.J.J.; Bax, W.A.; Verberne, H.J.; Cornel, J.H.; Stroes, E.S.G. PCSK9 Antibody Alirocumab Attenuates Arterial Wall Inflammation Without Changes in Circulating Inflammatory Markers. JACC Cardiovasc. Imaging 2019, 12, 2571–2573. [Google Scholar] [CrossRef]
  72. Godo, S.; Shimokawa, H. Endothelial Functions. Arterioscler. Thromb. Vasc. Biol. 2017, 37, e108–e114. [Google Scholar] [CrossRef]
  73. Tran, N.; Garcia, T.; Aniqa, M.; Ali, S.; Ally, A.; Nauli, S.M. Endothelial Nitric Oxide Synthase (eNOS) and the Cardiovascular System: In Physiology and in Disease States. Am. J. Biomed. Sci. Res. 2022, 15, 153–177. [Google Scholar]
  74. Wu, W.; Geng, P.; Zhu, J.; Li, J.; Zhang, L.; Chen, W.; Zhang, D.; Lu, Y.; Xu, X. KLF2 regulates eNOS uncoupling via Nrf2/HO-1 in endothelial cells under hypoxia and reoxygenation. Chem. Biol. Interact. 2019, 305, 105–111. [Google Scholar] [CrossRef]
  75. Zulkapli, R.; Muid, S.A.; Wang, S.M.; Nawawi, H. PCSK9 Inhibitors Reduce PCSK9 and Early Atherogenic Biomarkers in Stimulated Human Coronary Artery Endothelial Cells. Int. J. Mol. Sci. 2023, 24, 5098. [Google Scholar] [CrossRef]
  76. Baruch, A.; Mosesova, S.; Davis, J.D.; Budha, N.; Vilimovskij, A.; Kahn, R.; Peng, K.; Cowan, K.J.; Harris, L.P.; Gelzleichter, T.; et al. Effects of RG7652, a Monoclonal Antibody Against PCSK9, on LDL-C, LDL-C Subfractions, and Inflammatory Biomarkers in Patients at High Risk of or With Established Coronary Heart Disease (from the Phase 2 EQUATOR Study). Am. J. Cardiol. 2017, 119, 1576–1583. [Google Scholar] [CrossRef]
  77. Pradhan, A.D.; Aday, A.W.; Rose, L.M.; Ridker, P.M. Residual Inflammatory Risk on Treatment with PCSK9 Inhibition and Statin Therapy. Circulation 2018, 138, 141–149. [Google Scholar] [CrossRef]
  78. East, C.; Bass, K.; Mehta, A.; Rahimighazikalayed, G.; Zurawski, S.; Bottiglieri, T. Alirocumab and Lipid Levels, Inflammatory Biomarkers, Metabolomics, and Safety in Patients Receiving Maintenance Dialysis: The ALIrocumab in DIALysis Study (A Phase 3 Trial to Evaluate the Efficacy and Safety of Biweekly Alirocumab in Patients on a Stable Dialysis Regimen). Kidney Med. 2022, 4, 100483. [Google Scholar] [CrossRef]
  79. Zhang, Y.; Zhu, C.-G.; Xu, R.-X.; Li, S.; Guo, Y.-L.; Sun, J.; Li, J.-J. Relation of circulating PCSK9 concentration to fibrinogen in patients with stable coronary artery disease. J. Clin. Lipidol. 2014, 8, 494–500. [Google Scholar] [CrossRef]
  80. Boyd, J.H.; Fjell, C.D.; Russell, J.A.; Sirounis, D.; Cirstea, M.S.; Walley, K.R. Increased Plasma PCSK9 Levels Are Associated with Reduced Endotoxin Clearance and the Development of Acute Organ Failures during Sepsis. J. Innate Immun. 2016, 8, 211–220. [Google Scholar] [CrossRef]
  81. Ding, Z.; Liu, S.; Wang, X.; Deng, X.; Fan, Y.; Shahanawaz, J.; Reis, R.J.S.; Varughese, K.I.; Sawamura, T.; Mehta, J.L. Cross-talk between LOX-1 and PCSK9 in vascular tissues. Cardiovasc. Res. 2015, 107, 556–567. [Google Scholar] [CrossRef] [PubMed]
  82. Ferri, N.; Tibolla, G.; Pirillo, A.; Cipollone, F.; Mezzetti, A.; Pacia, S.; Corsini, A.; Catapano, A.L. Proprotein convertase subtilisin kexin type 9 (PCSK9) secreted by cultured smooth muscle cells reduces macrophages LDLR levels. Atherosclerosis 2012, 220, 381–386. [Google Scholar] [CrossRef] [PubMed]
  83. Denis, M.; Marcinkiewicz, J.; Zaid, A.; Gauthier, D.; Poirier, S.; Lazure, C.; Seidah, N.G.; Prat, A. Gene inactivation of proprotein convertase subtilisin/kexin type 9 reduces atherosclerosis in mice. Circulation. 2012, 125, 894–901. [Google Scholar] [CrossRef] [PubMed]
  84. Xie, W.; Liu, J.; Wang, W.; Wang, M.; Qi, Y.; Zhao, F.; Sun, J.; Liu, J.; Li, Y.; Zhao, D. Association between plasma PCSK9 levels and 10-year progression of carotid atherosclerosis beyond LDL-C: A cohort study. Int. J. Cardiol. 2016, 215, 293–298. [Google Scholar] [CrossRef]
  85. Moens, S.J.B.; Neele, A.E.; Kroon, J.; van der Valk, F.M.; Bossche, J.V.D.; Hoeksema, M.A.; Hoogeveen, R.M.; Schnitzler, J.G.; Baccara-Dinet, M.T.; Manvelian, G.; et al. PCSK9 monoclonal antibodies reverse the proinflammatory profile of monocytes in familial hypercholesterolaemia. Eur. Heart J. 2017, 38, 1584–1593. [Google Scholar] [CrossRef]
  86. Wu, N.Q.; Shi, H.W.; Li, J.J. Proprotein Convertase Subtilisin/Kexin Type 9 and Inflammation: An Updated Review. Front. Cardiovasc. Med. 2022, 9, 763516. [Google Scholar] [CrossRef]
  87. Vlachopoulos, C.; Koutagiar, I.; Skoumas, I.; Terentes-Printzios, D.; Zacharis, E.; Kolovou, G.; Stamatelopoulos, K.; Rallidis, L.; Katsiki, N.; Bilianou, H.; et al. Long-Term Administration of Proprotein Convertase Subtilisin/Kexin Type 9 Inhibitors Reduces Arterial FDG Uptake. JACC Cardiovasc. Imaging 2019, 12, 2573–2574. [Google Scholar] [CrossRef]
  88. Wang, H.; Wang, Q.; Wang, J.; Guo, C.; Kleiman, K.; Meng, H.; Knight, J.S.; Eitzman, D.T. Proprotein convertase subtilisin/kexin type 9 (PCSK9) Deficiency is Protective Against Venous Thrombosis in Mice. Sci. Rep. 2017, 7, 14360. [Google Scholar] [CrossRef]
  89. Jin, L.; Batra, S.; Jeyaseelan, S. Diminished neutrophil extracellular trap (NET) formation is a novel innate immune deficiency induced by acute ethanol exposure in polymicrobial sepsis, which can be rescued by CXCL1. PLoS Pathog. 2017, 13, e1006637. [Google Scholar] [CrossRef]
  90. Schuster, S.; Rubil, S.; Endres, M.; Princen, H.M.G.; Boeckel, J.N.; Winter, K.; Werner, C.; Laufs, U. Anti-PCSK9 antibodies inhibit pro-atherogenic mechanisms in APOE*3Leiden.CETP mice. Sci. Rep. 2019, 9, 11079. [Google Scholar] [CrossRef]
  91. Park, K.-H.; Park, W.J. Endothelial Dysfunction: Clinical Implications in Cardiovascular Disease and Therapeutic Approaches. J. Korean Med. Sci. 2015, 30, 1213–1225. [Google Scholar] [CrossRef] [PubMed]
  92. Basiak, M.; Hachula, M. Effect of PCSK9 Inhibitors on Hemostasis in Patients with Isolated Hypercholesterolemia. J. Clin. Med. 2022, 11, 2542. [Google Scholar] [CrossRef] [PubMed]
  93. Ochoa, E.; Iriondo, M.; Manzano, C.; Fullaondo, A.; Villar, I.; Ruiz-Irastorza, G.; Zubiaga, A.M.; Estonba, A. LDLR and PCSK9 Are Associated with the Presence of Antiphospholipid Antibodies and the Development of Thrombosis in aPLA Carriers. PLoS ONE 2016, 11, e0146990. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, M.; Li, Y.F.; Guo, Y.G.; Chen, M.M.; Jiang, Z.L.; Song, J.Y. Positive correlation between plasma PCSK9 and tissue factors levels in patients with angiographically diagnosed coronary artery disease and diabetes mellitus. J. Geriatr. Cardiol. 2016, 13, 312–315. [Google Scholar]
  95. Hamik, A.; Setiadi, H.; Bu, G.; McEver, R.P.; Morrissey, J.H. Down-regulation of monocyte tissue factor mediated by tissue factor pathway inhibitor and the low density lipoprotein receptor-related protein. J. Biol. Chem. 1999, 274, 4962–4969. [Google Scholar] [CrossRef]
  96. Scalise, V.; Sanguinetti, C.; Neri, T.; Cianchetti, S.; Lai, M.; Carnicelli, V.; Celi, A.; Pedrinelli, R. PCSK9 Induces Tissue Factor Expression by Activation of TLR4/NFkB Signaling. Int. J. Mol. Sci. 2021, 22, 12640. [Google Scholar] [CrossRef]
  97. Scalise, V.; Lombardi, S.; Sanguinetti, C.; Nieri, D.; Pedrinelli, R.; Celi, A.; Neri, T. A novel prothrombotic role of proprotein convertase subtilisin kexin 9: The generation of procoagulant extracellular vesicles by human mononuclear cells. Mol. Biol. Rep. 2022, 49, 4129–4134. [Google Scholar] [CrossRef]
  98. Basiak, M.; Kosowski, M.; Cyrnek, M.; Bułdak, Ł.; Maligłówka, M.; Machnik, G.; Okopień, B. Pleiotropic Effects of PCSK-9 Inhibitors. Int. J. Mol. Sci. 2021, 22, 3144. [Google Scholar] [CrossRef]
  99. Jenkins, P.V.; Rawley, O.; Smith, O.P.; O’Donnell, J.S. Elevated factor VIII levels and risk of venous thrombosis. Br. J. Haematol. 2012, 157, 653–663. [Google Scholar] [CrossRef]
  100. Rietveld, I.M.; Lijfering, W.M.; le Cessie, S.; Bos, M.H.A.; Rosendaal, F.R.; Reitsma, P.H.; Cannegieter, S.C. High levels of coagulation factors and venous thrombosis risk: Strongest association for factor VIII and von Willebrand factor. J. Thromb. Haemost. 2019, 17, 99–109. [Google Scholar] [CrossRef]
  101. Bovenschen, N.; Mertens, K.; Hu, L.; Havekes, L.M.; van Vlijmen, B.J. LDL receptor cooperates with LDL receptor-related protein in regulating plasma levels of coagulation factor VIII in vivo. Blood 2005, 106, 906–912. [Google Scholar] [CrossRef] [PubMed]
  102. Canuel, M.; Sun, X.; Asselin, M.C.; Paramithiotis, E.; Prat, A.; Seidah, N.G. Proprotein convertase subtilisin/kexin type 9 (PCSK9) can mediate degradation of the low density lipoprotein receptor-related protein 1 (LRP-1). PLoS ONE 2013, 8, e64145. [Google Scholar] [CrossRef]
  103. Paciullo, F.; Petito, E.; Falcinelli, E.; Gresele, P.; Momi, S. Pleiotropic effects of PCSK9-inhibition on hemostasis: Anti-PCSK9 reduce FVIII levels by enhancing LRP1 expression. Thromb. Res. 2022, 213, 170–172. [Google Scholar] [CrossRef] [PubMed]
  104. Wong, C.C.; Wu, J.-L.; Ji, F.; Kang, W.; Bian, X.; Chen, H.; Chan, L.-S.; Luk, S.T.Y.; Tong, S.; Xu, J.; et al. The cholesterol uptake regulator PCSK9 promotes and is a therapeutic target in APC/KRAS-mutant colorectal cancer. Nat. Commun. 2022, 13, 3971. [Google Scholar] [CrossRef] [PubMed]
  105. Pitteri, S.J.; Kelly-Spratt, K.S.; Gurley, K.E.; Kennedy, J.; Buson, T.B.; Chin, A.; Wang, H.; Zhang, Q.; Wong, C.-H.; Chodosh, L.A.; et al. Tumor microenvironment-derived proteins dominate the plasma proteome response during breast cancer induction and progression. Cancer Res. 2011, 71, 5090–5100. [Google Scholar] [CrossRef]
  106. Zia, S.; Batool, S.; Shahid, R. Could PCSK9 be a new therapeutic target of Eugenol? In vitro and in silico evaluation of hypothesis. Med. Hypotheses 2020, 136, 109513. [Google Scholar] [CrossRef] [PubMed]
  107. Nagashima, S.; Morishima, K.; Okamoto, H.; Ishibashi, S. Possible involvement of PCSK9 overproduction in hyperlipoproteinemia associated with hepatocellular carcinoma: A case report. J. Clin. Lipidol. 2016, 10, 1045–1049. [Google Scholar] [CrossRef]
  108. Xu, B.; Li, S.; Fang, Y.; Zou, Y.; Song, D.; Zhang, S.; Cai, Y. Proprotein Convertase Subtilisin/Kexin Type 9 Promotes Gastric Cancer Metastasis and Suppresses Apoptosis by Facilitating MAPK Signaling Pathway Through HSP70 Up-Regulation. Front. Oncol. 2021, 10, 609663. [Google Scholar] [CrossRef]
  109. Yuan, J.; Cai, T.; Zheng, X.; Ren, Y.; Qi, J.; Lu, X.; Chen, H.; Lin, H.; Chen, Z.; Liu, M.; et al. Potentiating CD8+ T cell antitumor activity by inhibiting PCSK9 to promote LDLR-mediated TCR recycling and signaling. Protein Cell 2021, 12, 240–260. [Google Scholar] [CrossRef]
  110. Bhattacharya, A.; Chowdhury, A.; Chaudhury, K.; Shukla, P.C. Proprotein convertase subtilisin/kexin type 9 (PCSK9): A potential multifaceted player in cancer. Biochim. Biophys. Acta Rev. Cancer 2021, 1876, 188581. [Google Scholar] [CrossRef]
  111. Xie, M.; Yu, X.; Chu, X.; Xie, H.; Zhou, J.; Zhao, J.; Su, C. Low baseline plasma PCSK9 level is associated with good clinical outcomes of immune checkpoint inhibitors in advanced non-small cell lung cancer. Thorac. Cancer 2022, 13, 353–360. [Google Scholar] [CrossRef] [PubMed]
  112. Piao, M.X.; Bai, J.W.; Zhang, P.F.; Zhang, Y.Z. PCSK9 regulates apoptosis in human neuroglioma u251 cells via mitochondrial signaling pathways. Int. J. Clin. Exp. Pathol. 2015, 8, 2787–2794. [Google Scholar] [PubMed]
  113. Bai, J.; Na, H.; Hua, X.; Wei, Y.; Ye, T.; Zhang, Y.; Jian, G.; Zeng, W.; Yan, L.; Tang, Q. A retrospective study of NENs and miR-224 promotes apoptosis of BON-1 cells by targeting PCSK9 inhibition. Oncotarget 2017, 8, 6929–6939. [Google Scholar] [CrossRef] [PubMed]
  114. Oza, P.P.; Kashfi, K. The evolving landscape of PCSK9 inhibition in cancer. Eur. J. Pharmacol. 2023, 949, 175721. [Google Scholar] [CrossRef] [PubMed]
  115. Momtazi-Borojeni, A.A.; Nik, M.E.; Jaafari, M.R.; Banach, M.; Sahebkar, A. Effects of immunisation against PCSK9 in mice bearing melanoma. Arch. Med. Sci. 2019, 16, 189–199. [Google Scholar] [CrossRef]
  116. He, M.; Hu, J.; Fang, T.; Tang, W.; Lv, B.; Yang, B.; Xia, J. Protein convertase subtilisin/Kexin type 9 inhibits hepatocellular carcinoma growth by interacting with GSTP1 and suppressing the JNK signaling pathway. Cancer Biol. Med. 2021, 19, 90–103. [Google Scholar] [CrossRef]
  117. Ioannou, G.N.; Lee, S.P.; Linsley, P.S.; Gersuk, V.; Yeh, M.M.; Chen, Y.-Y.; Peng, Y.-J.; Dutta, M.; Mascarinas, G.; Molla, B.; et al. Pcsk9 Deletion Promotes Murine Nonalcoholic Steatohepatitis and Hepatic Carcinogenesis: Role of Cholesterol. Hepatol. Commun. 2022, 6, 780–794. [Google Scholar] [CrossRef]
  118. Molina, S.; Castet, V.; Fournier-Wirth, C.; Pichard-Garcia, L.; Avner, R.; Harats, D.; Roitelman, J.; Barbaras, R.; Graber, P.; Ghersa, P.; et al. The low-density lipoprotein receptor plays a role in the infection of primary human hepatocytes by hepatitis C virus. J. Hepatol. 2007, 46, 411–419. [Google Scholar] [CrossRef]
  119. Caron, J.; Pène, V.; Tolosa, L.; Villaret, M.; Luce, E.; Fourrier, A.; Heslan, J.; Saheb, S.; Bruckert, E.; Gómez-Lechón, M.; et al. Low-density lipoprotein receptor-deficient hepatocytes differentiated from induced pluripotent stem cells allow familial hypercholesterolemia modeling, CRISPR/Cas-mediated genetic correction, and productive hepatitis C virus infection. Stem Cell Res. Ther. 2019, 10, 221. [Google Scholar] [CrossRef]
  120. Grin, P.M.; Dwivedi, D.J.; Chathely, K.M.; Trigatti, B.L.; Prat, A.; Seidah, N.G.; Liaw, P.C.; Fox-Robichaud, A.E. Low-density lipoprotein (LDL)-dependent uptake of Gram-positive lipoteichoic acid and Gram-negative lipopolysaccharide occurs through LDL receptor. Sci. Rep. 2018, 8, 10496. [Google Scholar] [CrossRef]
  121. Dwivedi, D.J.; Grin, P.M.; Khan, M.; Prat, A.; Zhou, J.; Fox-Robichaud, A.E.; Seidah, N.G.; Liaw, P.C. Differential Expression of PCSK9 Modulates Infection, Inflammation, and Coagulation in a Murine Model of Sepsis. Shock 2016, 46, 672–680. [Google Scholar] [CrossRef] [PubMed]
  122. Vecchié, A.; Bonaventura, A.; Meessen, J.; Novelli, D.; Minetti, S.; Elia, E.; Ferrara, D.; Ansaldo, A.M.; Scaravilli, V.; Villa, S.; et al. PCSK9 is associated with mortality in patients with septic shock: Data from the ALBIOS study. J. Intern. Med. 2021, 289, 179–192. [Google Scholar] [CrossRef] [PubMed]
  123. Zhou, Z.; Zhang, W.; Burgner, D.; Tonkin, A.; Zhu, C.; Sun, C.; Magnussen, C.G.; Ernst, M.E.; Breslin, M.; Nicholls, S.J.; et al. The Association Between PCSK9 Inhibitor Use and Sepsis: A Systematic Review and Meta-Analysis of 20 Double-Blind, Randomized, Placebo-Controlled Trials. Am. J. Med. 2023, 136, 558–567.e20. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Figure illustrating the impact of PCSK9 inhibitors on the LDL-dependent pathway. Low-density lipoprotein receptors (LDL-R) are produced in the hepatocyte and subsequently are transported to the cell membrane. Low-density lipoprotein (LDL) particles bind to LDL-R and then they form a complex with LDL, which is endocytosed. The LDL is degraded, and LDL-R is transported back to the membrane. Moreover, PCSK9, produced in the endoplasmic reticulum (ER), binds to LDL-R and forms a PCSK9-LDL-R complex, which undergoes endocytosis and lysosomal degradation. The inhibitors of PCSK9 bind to free PCSK9 molecules and prevent them from creating a PCSK9-LDL-R complex and their subsequent degradation.
Figure 1. Figure illustrating the impact of PCSK9 inhibitors on the LDL-dependent pathway. Low-density lipoprotein receptors (LDL-R) are produced in the hepatocyte and subsequently are transported to the cell membrane. Low-density lipoprotein (LDL) particles bind to LDL-R and then they form a complex with LDL, which is endocytosed. The LDL is degraded, and LDL-R is transported back to the membrane. Moreover, PCSK9, produced in the endoplasmic reticulum (ER), binds to LDL-R and forms a PCSK9-LDL-R complex, which undergoes endocytosis and lysosomal degradation. The inhibitors of PCSK9 bind to free PCSK9 molecules and prevent them from creating a PCSK9-LDL-R complex and their subsequent degradation.
Pharmaceuticals 16 01197 g001
Figure 2. Effect of PCSK9 on thrombosis mediated through different mechanisms.
Figure 2. Effect of PCSK9 on thrombosis mediated through different mechanisms.
Pharmaceuticals 16 01197 g002
Figure 3. Effect of PCSK9 on coagulation through various mechanisms.
Figure 3. Effect of PCSK9 on coagulation through various mechanisms.
Pharmaceuticals 16 01197 g003
Table 1. Studies dealing with the non-lipid effect of PCSK9 and PCSK9 inhibitors.
Table 1. Studies dealing with the non-lipid effect of PCSK9 and PCSK9 inhibitors.
Study/AuthorsNumber of Patients and Their CharacteristicsTreatmentResult
Li et al., 2015 [49]330, with CADN/APCSK9 levels are positively associated with the platelet count and plateletcrit, while no correlation with MPV and PDW
Pastori et al., 2017 [50]907, with atrial fibrillationN/APCSK9 levels are connected to increased risk of cardiovascular events, PCSK9 levels are correlating with thromboxane B2 levels
PCSK9—REACT study, 2017 [31]178, with acute coronary syndromeN/AIncreased PCSK9 levels are associated with higher platelet reactivity
Elseweidy et al. [35]Animal modelPolicosanol, 10-dehydrogingerdioneDecreased PCSK9, platelet activation and inflammation markers (sCD40L, sP-selectin, interferon-gamma)
Elseweidy et al. [51]Animal model10-dehydrogingerdioneDecreased interferon-gamma, sCD40L, sP-selectin correlated with PCSK9 suppression
EVOPACS study, 2018 [52]308PCSK9 inhibitorsPlasma levels of PCSK9 correlate with increased platelet count and platelet activation
Barale et al., 2020 [48]24PCSK9 inhibitorsPlatelet activation markers—sCD40L, platelet factor 4, and sP-selectin, significantly decreased after treatment with PCSK9 inhibitors. These markers correlate with PCSK9.
Cammisotto et al., 2020 [34]88, with atrial fibrillationN/AMarkers of platelet activation and oxidative stress correlate, and changes were amplified by adding LDL
Cammisotto et al., 2021 [33]80, heterozygous familial hypercholesterolemiaPCSK9 inhibitorsTreatment reduces platelet activation modulating NOX2 activity and, in turn, ox-LDL formation
Qi et al., 2021 [32]N/APCSK9 inhibitorsPCSK9 in plasma enhances platelet activation and thrombosis by binding to CD36. PCSK9 inhibitors or aspirin abolish the enhancing effects of PCSK9, supporting the use of aspirin in patients with a high plasma level of PCSK9.
Di Minno et al., 2021 [53]25, with familial hypercholesterolemiaPCSK9 inhibitorsReduction in platelet-activating factors after treatment.
Marques et al., 2022 [54]14, with familial hypercholesterolemiaPCSK9 inhibitorsPCSK9 inhibition impairs systemic inflammation and endothelial dysfunction by constraining leukocyte-endothelium interactions
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

Péč, M.J.; Benko, J.; Jurica, J.; Péčová, M.; Samec, M.; Hurtová, T.; Bolek, T.; Galajda, P.; Péč, M.; Samoš, M.; et al. The Anti-Thrombotic Effects of PCSK9 Inhibitors. Pharmaceuticals 2023, 16, 1197. https://doi.org/10.3390/ph16091197

AMA Style

Péč MJ, Benko J, Jurica J, Péčová M, Samec M, Hurtová T, Bolek T, Galajda P, Péč M, Samoš M, et al. The Anti-Thrombotic Effects of PCSK9 Inhibitors. Pharmaceuticals. 2023; 16(9):1197. https://doi.org/10.3390/ph16091197

Chicago/Turabian Style

Péč, Martin Jozef, Jakub Benko, Jakub Jurica, Monika Péčová, Marek Samec, Tatiana Hurtová, Tomáš Bolek, Peter Galajda, Martin Péč, Matej Samoš, and et al. 2023. "The Anti-Thrombotic Effects of PCSK9 Inhibitors" Pharmaceuticals 16, no. 9: 1197. https://doi.org/10.3390/ph16091197

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