[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (402)

Search Parameters:
Keywords = PCSK9

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 5599 KiB  
Article
Effects of Cynara scolymus L. Bract Extract on Lipid Metabolism Disorders Through Modulation of HMG-CoA Reductase, Apo A-1, PCSK-9, p-AMPK, SREBP-2, and CYP2E1 Expression
by Imane Mokhtari, Abdelaaty A. Shahat, Omar M. Noman, Dragan Milenkovic, Souliman Amrani and Hicham Harnafi
Metabolites 2024, 14(12), 728; https://doi.org/10.3390/metabo14120728 - 23 Dec 2024
Viewed by 572
Abstract
Background/Objectives: Hyperlipidemia is a major contributor to metabolic complications and tissue damage, leading to conditions such as liver steatosis, atherosclerosis, and obesity. This study aimed to investigate the effects of aqueous artichoke bract extract (AE) on lipid metabolism, liver antioxidative defense, and liver [...] Read more.
Background/Objectives: Hyperlipidemia is a major contributor to metabolic complications and tissue damage, leading to conditions such as liver steatosis, atherosclerosis, and obesity. This study aimed to investigate the effects of aqueous artichoke bract extract (AE) on lipid metabolism, liver antioxidative defense, and liver steatosis in mice fed a high-fat, high-sucrose diet while elucidating the underlying mechanisms. Methods: An 8-week study used hyperlipidemic mice treated with AE at daily doses of 100 and 200 mg/kg bw, compared to fenofibrate. Plasma, liver, fecal, and biliary lipids, as well as blood glucose, were analyzed enzymatically. The liver antioxidative defense was assessed by measuring reduced glutathione, malondialdehyde (MDA), and antioxidant enzyme activities, while liver steatosis was evaluated through transaminase and alkaline phosphatase activities and histological monitoring of lipid droplets. Polyphenol profiling and quantification were performed using HPLC–DAD, and potential mechanisms were predicted by molecular docking and confirmed in HepG2 cells. Results: At 200 mg/kg, AE significantly improved plasma lipid profiles by reducing total cholesterol, triglycerides, and LDL–cholesterol while increasing HDL–cholesterol. It facilitated cholesterol reduction in the liver and its excretion, indicating activation of reverse cholesterol transport, which led to reduced body weight and liver steatosis. AE lowered MDA levels and enhanced antioxidant enzyme activities. AE was found to be safe (LD50 > 5000 mg/kg) and modulated gene expression in HepG2 cells. Conclusions: Based on our results, the artichoke bract extract could be considered a natural resource of bioactive compounds to treat hyperlipidemia and related cardiometabolic diseases. Full article
Show Figures

Figure 1

Figure 1
<p>Effect of AE on body weight (<b>A</b>), food intake (<b>B</b>), and organ relative weight in mice (<b>C</b>). AE: aqueous artichoke bract extract; NG: normal group; HG: hyperlipidemic group; AETG: AE-treated groups; FG: fenofibrate group. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.001 vs. NG. * <span class="html-italic">p</span> &lt; 0.01 and ** <span class="html-italic">p</span> &lt; 0.001 vs. HG.</p>
Full article ">Figure 1 Cont.
<p>Effect of AE on body weight (<b>A</b>), food intake (<b>B</b>), and organ relative weight in mice (<b>C</b>). AE: aqueous artichoke bract extract; NG: normal group; HG: hyperlipidemic group; AETG: AE-treated groups; FG: fenofibrate group. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.001 vs. NG. * <span class="html-italic">p</span> &lt; 0.01 and ** <span class="html-italic">p</span> &lt; 0.001 vs. HG.</p>
Full article ">Figure 2
<p>Effect of AE on plasma total cholesterol and triglyceride levels in hyperlipidemic mice. AE: aqueous artichoke bract extract; TC: total cholesterol; TG: triglycerides; NG: normal group; HG: hyperlipidemic group; AETG: AE-treated groups; FG: fenofibrate group. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.001 vs. NG. * <span class="html-italic">p</span> &lt; 0.01 and ** <span class="html-italic">p</span> &lt; 0.001 vs. HG.</p>
Full article ">Figure 3
<p>Effect of AE on plasma LDL–C and HDL–C levels in mice. AE: aqueous artichoke bract extract; HDL–C: high-density lipoprotein-cholesterol; LDL–C: low-density lipoprotein-cholesterol; NG: normal group; HG: hyperlipidemic group; AETG: AE-treated groups; FG: fenofibrate group. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>b</sup> <span class="html-italic">p</span> &lt; 0.001 vs. NG. * <span class="html-italic">p</span> &lt; 0.05, <b>**</b> <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 vs. HG.</p>
Full article ">Figure 4
<p>Effect of AE on lipid levels in the liver and adipose tissue of mice. AE: aqueous artichoke bract extract; TC: total cholesterol; TG: triglycerides; NG: normal group; HG: hyperlipidemic group; AETG: AE-treated groups; FG: fenofibrate group. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>b</sup> <span class="html-italic">p</span> &lt; 0.001 vs. NG. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.001 vs. HG.</p>
Full article ">Figure 5
<p>Effect of AE on biliary cholesterol. AE: aqueous artichoke bract extract; NG: normal group; HG: hyperlipidemic group; AETG: AE-treated groups; FG: fenofibrate group. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.05 vs. NG. * <span class="html-italic">p</span> &lt; 0.01 vs. HG.</p>
Full article ">Figure 6
<p>Effect of AE on fecal lipid excretion in mice. AE: aqueous artichoke bract extract; TC: total cholesterol; TG: triglycerides; NG: normal group; HG: hyperlipidemic group; AETG: AE-treated groups; FG: fenofibrate group. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>b</sup> <span class="html-italic">p</span> &lt; 0.001 vs. NG. * <span class="html-italic">p</span> &lt; 0.001 vs. HG.</p>
Full article ">Figure 7
<p>Effect of AE on glucose levels in mice. AE: aqueous artichoke bract extract; NG: normal group; HG: hyperlipidemic group; AETG: AE-treated groups. FG: fenofibrate group. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>b</sup> <span class="html-italic">p</span> &lt; 0.001 vs. NG. * <span class="html-italic">p</span> &lt; 0.01 and ** <span class="html-italic">p</span> &lt; 0.001 vs. HG.</p>
Full article ">Figure 8
<p>Effects of AE on mice liver histology. AE: aqueous artichoke bract extract; LD: lipid droplets; NG: normal group; HG: hyperlipidemic group; AETG: AE-treated groups; FG: fenofibrate group.</p>
Full article ">Figure 9
<p>Effect of AE on enzymatic biomarkers of hepatic injury in mice. AE: aqueous artichoke bract extract; AST: aspartate transaminase; ALT: alanine transaminase; ALP: alkaline phosphatase; NG: normal group; HG: hyperlipidemic group; AETG: AE-treated groups; FG: fenofibrate group. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.001 vs. NG. * <span class="html-italic">p</span> &lt; 0.001 vs. HG.</p>
Full article ">Figure 10
<p>Effect of AE on liver lipid oxidation in mice. AE: aqueous artichoke bract extract; NG: normal group; HG: hyperlipidemic group; AETG: AE-treated groups; FG: fenofibrate group. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.001 vs. NG. * <span class="html-italic">p</span> &lt; 0.001 vs. HG.</p>
Full article ">Figure 11
<p>Effect of AE on glutathione in hyperlipidemic mice. AE: aqueous artichoke bract extract; NG: normal control group; HG: hyperlipidemic group; AETG: AE-treated groups; FG: fenofibrate group. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.01 vs. NG. * <span class="html-italic">p</span> &lt; 0.01 and vs. HG.</p>
Full article ">Figure 12
<p>Effect of AE on superoxide dismutase and catalase activities in hyperlipidemic mice. AE: aqueous artichoke bract extract; NG: normal group; HG: hyperlipidemic group; AETG: AE-treated groups; FG: fenofibrate-treated group. <sup>a</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>b</sup> <span class="html-italic">p</span> &lt; 0.001 vs. NG. * <span class="html-italic">p</span> &lt; 0.01 and ** <span class="html-italic">p</span> &lt; 0.001 vs. HG.</p>
Full article ">Figure 13
<p>Effects of AE and its major identified polyphenols on protein expression in HepG2 cells. 1,5-dCQA: 1,5-di-<span class="html-italic">O</span>-caffeoylquinic; CQA: chlorogenic acid; AE: aqueous artichoke bract extract; PCSK-9: proprotein convertase subtilisin/kexin type 9; Apo A-1: apolipoprotein A-1; SREBP-2: sterol regulatory element-binding protein 2; CYP2E1: cytochrome P450 2E1; HMG-C-R: 3-hydroxy-3-methyl-glutaryl-CoA reductase; p-AMPK: phosphorylated AMP-activated protein kinase. * <span class="html-italic">p</span> &lt; 0.01, ** <span class="html-italic">p</span> &lt; 0.001 vs. control (0 µg/mL).</p>
Full article ">Figure 14
<p>Effects of AE and its major identified polyphenols on HepG2 cell viability. 1,5-dCQA: 1,5-di-<span class="html-italic">O</span>-caffeoylquinic; CQA: chlorogenic acid; AE: aqueous artichoke bract extract.</p>
Full article ">
35 pages, 1935 KiB  
Review
Vaccination as a Promising Approach in Cardiovascular Risk Mitigation: Are We Ready to Embrace a Vaccine Strategy?
by Georgios Tsioulos, Natalia G. Vallianou, Alexandros Skourtis, Maria Dalamaga, Evangelia Kotsi, Sofia Kargioti, Nikolaos Adamidis, Irene Karampela, Iordanis Mourouzis and Dimitris Kounatidis
Biomolecules 2024, 14(12), 1637; https://doi.org/10.3390/biom14121637 - 20 Dec 2024
Viewed by 592
Abstract
Cardiovascular disease (CVD) remains a leading global health concern, with atherosclerosis being its principal cause. Standard CVD treatments primarily focus on mitigating cardiovascular (CV) risk factors through lifestyle changes and cholesterol-lowering therapies. As atherosclerosis is marked by chronic arterial inflammation, the innate and [...] Read more.
Cardiovascular disease (CVD) remains a leading global health concern, with atherosclerosis being its principal cause. Standard CVD treatments primarily focus on mitigating cardiovascular (CV) risk factors through lifestyle changes and cholesterol-lowering therapies. As atherosclerosis is marked by chronic arterial inflammation, the innate and adaptive immune systems play vital roles in its progression, either exacerbating or alleviating disease development. This intricate interplay positions the immune system as a compelling therapeutic target. Consequently, immunomodulatory strategies have gained increasing attention, though none have yet reached widespread clinical adoption. Safety concerns, particularly the suppression of host immune defenses, remain a significant barrier to the clinical application of anti-inflammatory therapies. Recent decades have revealed the significant role of adaptive immune responses to plaque-associated autoantigens in atherogenesis, opening new perspectives for targeted immunological interventions. Preclinical models indicate that vaccines targeting specific atherosclerosis-related autoantigens can slow disease progression while preserving systemic immune function. In this context, numerous experimental studies have advanced the understanding of vaccine development by exploring diverse targeting pathways. Key strategies include passive immunization using naturally occurring immunoglobulin G (IgG) antibodies and active immunization targeting low-density lipoprotein cholesterol (LDL-C) and apolipoproteins, such as apolipoprotein B100 (ApoB100) and apolipoprotein CIII (ApoCIII). Other approaches involve vaccine formulations aimed at proteins that regulate lipoprotein metabolism, including proprotein convertase subtilisin/kexin type 9 (PCSK9), cholesteryl ester transfer protein (CETP), and angiopoietin-like protein 3 (ANGPTL3). Furthermore, the literature highlights the potential for developing non-lipid-related vaccines, with key targets including heat shock proteins (HSPs), interleukins (ILs), angiotensin III (Ang III), and a disintegrin and metalloproteinase with thrombospondin motifs 7 (ADAMTS-7). However, translating these promising findings into safe and effective clinical therapies presents substantial challenges. This review provides a critical evaluation of current anti-atherosclerotic vaccination strategies, examines their proposed mechanisms of action, and discusses key challenges that need to be overcome to enable clinical translation. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representation of adaptive immune cell involvement in the progression of atherosclerosis. In early atherosclerosis stages, Tregs accumulate in arterial walls, interacting with APCs like dendritic cells and macrophages, releasing anti-inflammatory cytokines (IL-10, TGF-β) to maintain immune tolerance and reduce inflammation. Circulating T cells also migrate to plaques, attracted by chemotactic signals (CCL5, CXCL16) from platelets and immune cells. As the disease progresses, APCs present atherosclerosis-related antigens to naïve T cells in regional lymph nodes, driving T cell polarization into pro-inflammatory Th cells and CTLs. These activated T cells return to plaques, exacerbating local inflammation. In advanced stages, Th1 cells dominate, secreting IFN-γ and TNF-α to promote plaque growth and instability. B cells modulate disease progression: B1 cells secrete protective IgM antibodies, while B2 cells release IgG antibodies, contributing to atherosclerosis progression. Overall, the adaptive immune response shifts from protective to pathogenic, driving plaque progression and vascular damage [<a href="#B14-biomolecules-14-01637" class="html-bibr">14</a>,<a href="#B15-biomolecules-14-01637" class="html-bibr">15</a>,<a href="#B16-biomolecules-14-01637" class="html-bibr">16</a>,<a href="#B17-biomolecules-14-01637" class="html-bibr">17</a>,<a href="#B18-biomolecules-14-01637" class="html-bibr">18</a>,<a href="#B19-biomolecules-14-01637" class="html-bibr">19</a>,<a href="#B20-biomolecules-14-01637" class="html-bibr">20</a>,<a href="#B21-biomolecules-14-01637" class="html-bibr">21</a>,<a href="#B22-biomolecules-14-01637" class="html-bibr">22</a>,<a href="#B23-biomolecules-14-01637" class="html-bibr">23</a>,<a href="#B24-biomolecules-14-01637" class="html-bibr">24</a>,<a href="#B25-biomolecules-14-01637" class="html-bibr">25</a>,<a href="#B26-biomolecules-14-01637" class="html-bibr">26</a>,<a href="#B27-biomolecules-14-01637" class="html-bibr">27</a>,<a href="#B28-biomolecules-14-01637" class="html-bibr">28</a>,<a href="#B29-biomolecules-14-01637" class="html-bibr">29</a>,<a href="#B30-biomolecules-14-01637" class="html-bibr">30</a>,<a href="#B31-biomolecules-14-01637" class="html-bibr">31</a>,<a href="#B32-biomolecules-14-01637" class="html-bibr">32</a>,<a href="#B33-biomolecules-14-01637" class="html-bibr">33</a>,<a href="#B34-biomolecules-14-01637" class="html-bibr">34</a>,<a href="#B35-biomolecules-14-01637" class="html-bibr">35</a>,<a href="#B36-biomolecules-14-01637" class="html-bibr">36</a>,<a href="#B37-biomolecules-14-01637" class="html-bibr">37</a>,<a href="#B38-biomolecules-14-01637" class="html-bibr">38</a>,<a href="#B39-biomolecules-14-01637" class="html-bibr">39</a>,<a href="#B40-biomolecules-14-01637" class="html-bibr">40</a>,<a href="#B41-biomolecules-14-01637" class="html-bibr">41</a>]. Abbreviations: APCs: antigen-presenting cells; B1 cells: B cell subtype 1; B2 cells: B cell subtype 2; CCL5: chemokine (C-C motif) ligand 5; CTLs: cytotoxic T lymphocytes; CXCL16: chemokine (C-X-C motif) ligand 16; DCs: dendritic cells; IFN-γ: interferon gamma; IgG: immunoglobulin G; IgM: immunoglobulin M; IL-10: interleukin-10; Mfs: macrophages; MHC: major histocompatibility complex; T cells: T lymphocytes; TGF-β: transforming growth factor beta; Th cells: T helper cells; TNF: tumor necrosis factor; Tregs: regulatory T cells. Created in BioRender. Kounatidis, D. (2024) <a href="https://BioRender.com/x00b831" target="_blank">https://BioRender.com/x00b831</a>, assessed on 24 November 2024.</p>
Full article ">Figure 2
<p>Experimental vaccine models targeting lipids to reduce inflammation in atherosclerosis. This figure depicts four distinct experimental vaccine strategies designed to mitigate the inflammation and progression of atherosclerosis by modulating immune responses. (<b>A</b>) OxLDL-C-pulsed dendritic cell transfer: LDLr−/− mice were treated with mature DCs pulsed with oxLDL-C before atherosclerosis induction via Western-type diet feeding. This approach promoted oxLDL-C-specific T cells and IgG production, diminishing foam cell recruitment and inflammation [<a href="#B84-biomolecules-14-01637" class="html-bibr">84</a>]. (<b>B</b>) PAM nanoparticle vaccine: PAM nanoparticles were used to deliver the p210 peptide in ApoE−/− mice models. This vaccine suppressed CD4<sup>+</sup> and CD8<sup>+</sup> effector T cells and shifted macrophage phenotypes, collectively reducing atherosclerotic burden and showing translational potential [<a href="#B99-biomolecules-14-01637" class="html-bibr">99</a>]. (<b>C</b>) Nanoliposome-based PCSK9-TP vaccine: A nanoliposome vaccine conjugated with PCSK9 and tetanus-derived peptides (IFPT peptide) was tested in atherosclerotic mice. The vaccine stimulated anti-inflammatory CD4<sup>+</sup> Th2 cells and IL-4 secretion, promoting atheroprotective immune responses [<a href="#B121-biomolecules-14-01637" class="html-bibr">121</a>]. (<b>D</b>) Oral TT/CETP vaccine in rabbits: A combined oral vaccine targeting TT and CETP upregulated anti-inflammatory cytokines IL-10 and TGF-β while suppressing pro-inflammatory cytokines TNF-α and IFN-γ [<a href="#B133-biomolecules-14-01637" class="html-bibr">133</a>]. Abbreviations: ApoE: apolipoprotein E; CETP: cholesteryl ester transfer protein; DC: dendritic cell; IFN-γ: interferon-gamma; IL: interleukin; LDLr: low-density lipoprotein receptor; oxLDL-C: oxidized low-density lipoprotein cholesterol; PAM: poly(amino acid)-based; PCSK9: proprotein convertase subtilisin/kexin type 9; TGF-β: transforming growth factor-beta; Th2: T-helper type 2; TNF-α: tumor necrosis factor-alpha; TP: tetanus peptide; TT: tetanus toxoid. Created in BioRender. Kounatidis, D. (2024) <a href="https://BioRender.com/f60u021" target="_blank">https://BioRender.com/f60u021</a>, assessed on 24 November 2024.</p>
Full article ">Figure 3
<p>Primary vaccination targets in atherosclerosis. Abbreviations: ADAMTS-7: A disintegrin and metalloproteinase with thrombospondin motifs 7; ANGII: angiotensin II; ANGPTL3: angiopoietin-like protein 3; ApoB100: apolipoprotein B100; ApoCIII: apolipoprotein CIII; CETP: cholesteryl ester transfer protein; HSP: heat shock protein; IgG: immunoglobulin G; LDL-C: low-density lipoprotein cholesterol; PCSK9: proprotein convertase subtilisin/kexin type 9. Created in BioRender. Kounatidis, D. (2024) <a href="https://BioRender.com/f98n410" target="_blank">https://BioRender.com/f98n410</a>, assessed on 24 November 2024.</p>
Full article ">
27 pages, 2428 KiB  
Review
The Emerging Role of PCSK9 in the Pathogenesis of Alzheimer’s Disease: A Possible Target for the Disease Treatment
by Gabriella Testa, Serena Giannelli, Erica Staurenghi, Rebecca Cecci, Lucrezia Floro, Paola Gamba, Barbara Sottero and Gabriella Leonarduzzi
Int. J. Mol. Sci. 2024, 25(24), 13637; https://doi.org/10.3390/ijms252413637 - 20 Dec 2024
Viewed by 535
Abstract
Alzheimer’s disease (AD) is a multifactorial neurodegenerative disease mainly caused by β-amyloid (Aβ) accumulation in the brain. Among the several factors that may concur to AD development, elevated cholesterol levels and brain cholesterol dyshomeostasis have been recognized to play a relevant role. Proprotein [...] Read more.
Alzheimer’s disease (AD) is a multifactorial neurodegenerative disease mainly caused by β-amyloid (Aβ) accumulation in the brain. Among the several factors that may concur to AD development, elevated cholesterol levels and brain cholesterol dyshomeostasis have been recognized to play a relevant role. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a protein primarily known to regulate plasma low-density lipoproteins (LDLs) rich in cholesterol and to be one of the main causes of familial hypercholesterolemia. In addition to that, PCSK9 is also recognized to carry out diverse important activities in the brain, including control of neuronal differentiation, apoptosis, and, importantly, LDL receptors functionality. Moreover, PCSK9 appeared to be directly involved in some of the principal processes responsible for AD development, such as inflammation, oxidative stress, and Aβ deposition. On these bases, PCSK9 management might represent a promising approach for AD treatment. The purpose of this review is to elucidate the role of PCSK9, whether or not cholesterol-related, in AD pathogenesis and to give an updated overview of the most innovative therapeutic strategies developed so far to counteract the pleiotropic activities of both humoral and brain PCSK9, focusing in particular on their potentiality for AD management. Full article
Show Figures

Figure 1

Figure 1
<p>PCSK9 involvement in brain cholesterol dyshomeostasis. Circulating PCSK9 prevents recycling of LDL receptors, inducing hypercholesterolemia, inflammation, and oxidative stress, thus leading to BBB damage that allows PCSK9 to enter into the brain. Inside the brain, PCSK9 affects receptors and transporters involved in astrocyte-synthetized cholesterol and cholesterol uptake by neurons. Abbreviations: ABC, ATP-binding cassette transporter; ApoE, Apolipoprotein E; ApoER2, Apolipoprotein E receptor 2; BBB, blood-brain barrier; CSF, cerebrospinal fluid; HDL, high-density lipoprotein; LDLR, low-density lipoprotein receptor; LRP1, lipoprotein receptor-related protein 1; PCSK9, Proprotein convertase subtilisin/kexin type 9; and VLDLR; very low-density lipoprotein receptor.</p>
Full article ">Figure 2
<p>Application of anti-PCSK9 pharmacological tools for AD treatment. Drugs developed to target circulating PCSK9 appear suitable for AD cure by counteracting amyloidogenesis and by reducing hypercholesterolemia, inflammation, and oxidative stress, thus preventing BBB damage. PCSK9-targeting drugs able to cross the BBB could be suggested to delay the neurodegenerative AD progression, exerting their activities directly inside the brain. Abbreviations: AAV, adeno-associated virus; EV, extracellular vesicle; mAb, monoclonal antibody; miRNA, microRNA; and siRNA, small interfering RNA.</p>
Full article ">Figure 3
<p>PCSK9 activities associated with Alzheimer’s disease onset and development and the relative factors involved. Abbreviations: ABCA1, ATP-binding cassette transporter A1; ApoE, Apolipoprotein E; ApoER2, Apolipoprotein E receptor 2; BACE1, beta-site amyloid precursor protein-cleaving enzyme-1; BBB, blood-brain barrier; LDLR, low-density lipoprotein receptor; LRP1, lipoprotein receptor-related protein 1; NF-κB, nuclear factor kappa B; NLRP3, NOD-like receptor protein 3; TLR4, Toll-like receptor 4; VLDLR; very low-density lipoprotein receptor.</p>
Full article ">
12 pages, 5281 KiB  
Article
PAC1 Agonist Maxadilan Reduces Atherosclerotic Lesions in Hypercholesterolemic ApoE-Deficient Mice
by Lilli Mey, Gabriel A. Bonaterra, Joy Hoffmann, Hans Schwarzbach, Anja Schwarz, Lee E. Eiden, Eberhard Weihe and Ralf Kinscherf
Int. J. Mol. Sci. 2024, 25(24), 13245; https://doi.org/10.3390/ijms252413245 - 10 Dec 2024
Viewed by 390
Abstract
A possible involvement of immune- and vasoregulatory PACAP signaling at the PAC1 receptor in atherogenesis and plaque-associated vascular inflammation has been suggested. Therefore, we tested the PAC1 receptor agonist Maxadilan and the PAC1 selective antagonist M65 on plaque development and lumen stenosis in [...] Read more.
A possible involvement of immune- and vasoregulatory PACAP signaling at the PAC1 receptor in atherogenesis and plaque-associated vascular inflammation has been suggested. Therefore, we tested the PAC1 receptor agonist Maxadilan and the PAC1 selective antagonist M65 on plaque development and lumen stenosis in the ApoE−/− atherosclerosis model for possible effects on atherogenesis. Adult male ApoE−/− mice were fed a cholesterol-enriched diet (CED) or standard chow (SC) treated with Maxadilan, M65 or Sham. Effects of treatment on atherosclerotic plaques, lumen stenosis, apoptosis and pro-inflammatory signatures were analyzed in the brachiocephalic trunk (BT). The percentage of Maxadilan treated mice exhibiting plaques under SC and CED was lower than that of Sham or M65 treatment indicating opposite effects of Maxadilan and M65. Maxadilan application inhibited lumen stenosis in SC and CED mice compared to the Sham mice. In spite of increased cholesterol levels, lumen stenosis of Maxadilan-treated mice was similar under CED and SC. In contrast, M65 under SC or CED did not reveal a significant influence on lumen stenosis. Maxadilan significantly reduced the TNF-α-immunoreactive (TNF-α+) area in the plaques under CED, but not under SC. In contrast, the IL-1β+ area was reduced after Maxadilan treatment in SC mice but remained unchanged in CED mice compared to Sham mice. Maxadilan reduced caspase-3 immunoreactive (caspase-3+) in the tunica media under both, SC and CED without affecting lipid content in plaques. Despite persistent hypercholesterolemia, Maxadilan reduces lumen stenosis, apoptosis and TNF-α driven inflammation. Our data suggest that Maxadilan provides atheroprotection by acting downstream of hypercholesterolemia-induced vascular inflammation. This implicates the potential of PAC1-specific agonist drugs against atherosclerosis even beyond statins and PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitors. Full article
(This article belongs to the Special Issue Atherosclerosis: From Molecular Basis to Therapy)
Show Figures

Figure 1

Figure 1
<p>Percentage of mice exhibiting macroscopically visible atherosclerotic plaques in the ascending aorta (aao), descending aorta (dao), aortic arch (aa), and branches of Sham and Maxadilan-treated ApoE<sup>−/−</sup> mice under standard chow (SC) or cholesterol-enriched diet (CED): Brachiocephalic trunk (BT), right/left carotid artery (rca/lca), right/left subclavian artery (rsca/lsca). Intraluminal contrast staining with methylene blue enhances the visibility of white atherosclerosis plaques, indicated by black arrows. For statistical significance, see <span class="html-italic">p</span> values in the individual micrographs. The numbers in parentheses denote the numbers of animals.</p>
Full article ">Figure 2
<p>Lumen stenosis and plaque development in the brachiocephalic trunk (BT)of ApoE<sup>−/−</sup> mice at the age of 20 weeks, fed with standard chow (SC) or cholesterol-enriched diet (CED). Treated mice received PAC1 agonist Maxadilan (20 nmol/kg) or PAC1 antagonist M65 (20 nmol/kg), dissolved in physiological saline solution i.p., Sham groups received physiological saline solution. Cross sections of areas with maximum plaque sizes were prepared and stained with hematoxylin/eosin. Lumen stenosis was measured in %; data are provided as mean ± SEM. For statistical significance, see <span class="html-italic">p</span> values in the individual micrographs.</p>
Full article ">Figure 3
<p>Immunohistochemistry and quantification of the inflammatory markers TNF-α and IL-1β in the plaque and tunica media of BT in ApoE<sup>−/−</sup> mice under SC and CED, after Maxadilan treatment or Sham (<b>A</b>). The percentages of immunohistochemically positive-stained areas per plaque or media were evaluated. Black arrowheads indicate immunoreactive positive cells. Data (<b>B</b>) TNF-α and (<b>C</b>) IL-1β are provided as mean + SEM (n = 7–9). * <span class="html-italic">p</span> ≤ 0.05 vs. Sham under SC or CED. Scale bar: 100 µm.</p>
Full article ">Figure 4
<p>Immunohistochemistry and quantification of immunoreactive areas (in %) in relation to the whole plaque or media of cross sections of BT of ApoE<sup>−/−</sup> mice under SC or CED after Maxadilan treatment or Sham (<b>A</b>). Antibodies were directed against cyclooxygenase-2 (COX-2) and cleaved caspase-3 (cl. casp.3, apoptosis). Black arrowheads indicate immunoreactive positive cells. Data (<b>B</b>) COX-2 and (<b>C</b>) cleaved caspase-3 are provided as mean + SEM (n = 7–9). ** <span class="html-italic">p</span> ≤ 0.01 vs. Sham under SC; <sup>#</sup> <span class="html-italic">p</span> ≤ 0.05 <sup>###</sup> <span class="html-italic">p</span> ≤ 0.001 vs. Sham under CED. Scale bar: 100 µm.</p>
Full article ">Figure 5
<p>ORO (on the left) and CD68 (on the right) (immuno)histochemistry of atherosclerotic BT cross-sections of ApoE<sup>−/−</sup> mice under Maxadilan treatment or Sham SC or CED. Oil red positive lipid deposits are shown in red. (<b>A</b>,<b>B</b>) Lipid-loaden MΦ in the plaque-forming foam cells (black arrowheads) are distinguishable from intimal cells, smooth muscle cells of the tunica media with small lipid droplets (arrows) and cells of perivascular fat tissue showing distinct fat vacuoles. (<b>C</b>) CD68<sup>+</sup> Mo and MΦ (right side, white arrowheads) stained with HRP-DAB in brown. Data are provided as mean + SEM (n = 7–9). Diagrams show % of lipid or Mo/MΦ positive area in relation to the whole plaque area. Intima (i), media (m) and adventitia (a). Scale bar: 100 µm.</p>
Full article ">
15 pages, 930 KiB  
Article
Prescriptive Appropriateness and Efficacy of Cholesterol-Lowering Drugs in a Secondary Prevention Setting—A Retrospective Analysis from Two Italian Cardiac Rehabilitation Centers
by Francesca Saladini, Stefania Baggio, Federica Marcato, Francesco Campisi, Roberto Verlato, Giampaolo Pasquetto, Emanuele Bertaglia, Gaetano Povolo, Paolo Buja and Nicola Ferri
J. Clin. Med. 2024, 13(24), 7505; https://doi.org/10.3390/jcm13247505 - 10 Dec 2024
Viewed by 369
Abstract
Background: Treatment of CV risk factors, such as cholesterol level, represents one of the main goals to reduce atherosclerotic burden. The aim of this study was to investigate the prescriptive appropriateness of cholesterol-lowering drugs among patients who experienced an atherosclerotic CV disease (ASCVD). [...] Read more.
Background: Treatment of CV risk factors, such as cholesterol level, represents one of the main goals to reduce atherosclerotic burden. The aim of this study was to investigate the prescriptive appropriateness of cholesterol-lowering drugs among patients who experienced an atherosclerotic CV disease (ASCVD). Methods: We investigated 155 patients who underwent cardiac rehabilitation in 2020. The European Society of Cardiology (ESC) 2021 guidelines on CV disease prevention and 2019 ESC Guidelines on dyslipidemias were followed to detect the appropriateness of prescription. SCORE2 and SCORE2-OP risk estimations were used to detect patients’ CV risk profiles. Patients were divided into three groups: 1 (n = 118) patients admitted for their first CV event, 2A (n = 18) patients who experienced a previous CV event years before, and 2B (n = 19) patients admitted for a new event with a previous CV event 2 years before. Low-density lipoprotein (LDL) cholesterol level was detected at the time of admission to the hospital, during cardiac rehabilitation, and at the first visit after rehabilitation. Results: The statistics for our study participants, with a mean age of 66.1 years, were: 72.4% overweight/obese, 63.9% diabetic, 72.5% smokers, 93.0% hypertensives, and 91.7% had dyslipidemias. In group 1, only 5.1% had a low/moderate risk, 44.1% presented a high risk, and 50.8% a very high risk according to calculators. The average LDL levels were 115.8 mg/dL (2.99 mol/L) upon admission to the hospital, 66.4 mg/dL (1.72 mmol/L) at the time of cardiac rehabilitation, and 64.8 mg/dL (1.67 mmol/L) at the subsequent medical visit. In the overall group, only 36.0% had LDL < 55 mg/dL (1.42 mmol/L). In group 1, 79.4% were treated with high-intensity statin alone or plus ezetimibe; in group 2A, the percentage increased up to 87.5%, while group 2B 33.4% was treated with high-intensity statin plus ezetimibe and 33.3% were treated with PCSK9 inhibitors. Conclusions: This retrospective study confirms the importance of properly calculating CV risk profiles. The main limitations for the efficacy of lipid-lowering drugs were: patient’s compliance, drugs side effects, lifestyle habits, and collaboration with a general practitioner. Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

Figure 1
<p>Reasons for admission to hospital among group 1 and group 2A and 2B patients considered as a whole group.</p>
Full article ">Figure 2
<p>Lipid-lowering drugs suggested at the time of hospital discharge on the left panel and at the time of discharge form cardiac rehabilitation (right panel) among group 1 patients (n = 118); patients admitted to hospital for their first cardiovascular event, divided into two subgroups: those who reached and those who did not reach the LDL target &lt; 55 mg/dL (1.42 mmol/L).</p>
Full article ">Figure 3
<p>Lipid-lowering drugs suggested at the time of hospital discharge on the left panel and at the time of discharge form cardiac rehabilitation, right panel, among group 2A patients (n = 18); patients admitted to hospital for a second cardiovascular event 2 years after the previous one, divided into two subgroups: those who reached and those who did not reach the LDL target &lt; 55 mg/dL (1.42 mmol/L).</p>
Full article ">Figure 4
<p>Lipid-lowering drugs suggested at the time of hospital discharge on the left panel and at the time of discharge form cardiac rehabilitation, right panel, among group 2B patients (n = 19); patients admitted to hospital for a second cardiovascular event within 2 years of the previous one, divided into two subgroups: those who reached and those who did not reach the LDL target &lt; 40 mg/dL (1.03 mmol/L).</p>
Full article ">
16 pages, 1942 KiB  
Perspective
Management of Hypercholesterolemia in Patients with Coronary Artery Disease: A Glimpse into the Future
by Alessandro Sciahbasi, Paola Russo, Michela Zuccanti, Laura Chiorazzo, Francesco Maria Castelli and Antonino Granatelli
J. Clin. Med. 2024, 13(23), 7420; https://doi.org/10.3390/jcm13237420 - 5 Dec 2024
Viewed by 785
Abstract
Cardio-cerebral vascular diseases due to atherosclerosis are still the leading cause of death worldwide. Low-density lipoprotein cholesterol (LDL-C) and apolipoprotein B have been identified as the primary factors responsible for the atherosclerotic process, with a causal effect. Many drugs aimed at reducing LDL-C [...] Read more.
Cardio-cerebral vascular diseases due to atherosclerosis are still the leading cause of death worldwide. Low-density lipoprotein cholesterol (LDL-C) and apolipoprotein B have been identified as the primary factors responsible for the atherosclerotic process, with a causal effect. Many drugs aimed at reducing LDL-C levels are already on the market, acting in different ways in terms of mechanism of action, efficacy, and safety. Moreover, new lipid-lowering agents and new technologies in the fields of gene editing and immunotherapy are currently under investigation. A more recent biomarker associated with an increased risk of plaque generation, progression, and subsequent ASCVD is the lipoprotein (a) and, in the next few years, it will be the new target of pharmacological therapy. The aim of this review is to present the landscape of therapies already approved to reduce LDL-C levels, evaluating their efficacy, tolerability, and indications. Moreover, we take a glimpse into the future to evaluate experimental novel therapies to lower LDL-C levels that will be approved in the next few years or are under clinical evaluation. Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

Figure 1
<p>Major discoveries in treatment of hypercholesterolemia. Since the discovery of statin in 1971 by Akira Endo, multiple improvements in low-density lipoprotein cholesterol have been obtained, and in the near future we are waiting for primary results on studies with gene editing and vaccines. EMA: European Medical Agency; FDA: Food and Drug Administration; PCSK9: Proprotein convertase subtilisin/kexin type 9.</p>
Full article ">Figure 2
<p>Schematic representation of the atherosclerotic process. LDL-C interacts with the proteoglycan matrix and accumulates in the arterial wall and is susceptible to oxidation and modification to a toxic form, generating oxidized LDL-C (oxLDL-C). OxLDL-C activates endothelial cells to produce adhesion molecules and chemokines to recruit monocytes into the arterial wall, which, after differentiation into macrophages, internalize oxLDL-C through the scavenger receptor and become foam cells. These cells induce the production of chemokines and the expression of receptors (TLR-4, TLR-6, and CD36) on endothelial cells.</p>
Full article ">Figure 3
<p>Latest European Society of Cardiology Guidelines. According to guidelines, patients are classified into 5 different risk score classes with different therapeutic low-density lipoprotein cholesterol targets. ACR: albumin to creatinine ratio; ASCVD: atherosclerotic cardiovascular disease; CKD: chronic kidney disease; DM: diabetes mellitus; eGFR: estimated glomerular filtration rate; OP-2: Older Persons-2; SCORE-2: Systematic Coronary Risk Estimation; TOD: target organ damage.</p>
Full article ">Figure 4
<p>Mechanism of action of drugs commercially available. Statins block the 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase, the enzyme that converts HMG-CoA into mevalonic acid. Ezetimibe inhibits the Niemann-Pick C1-like protein 1 at the level of the brush border of the intestine, reducing absorption of dietary and biliary cholesterol and its incorporation into chylomicrons. Bempedoic acid inhibits an ATP citrate lyase responsible for cholesterol synthesis in the same enzymatic cascade target of statins, but upstream of HMG-CoA reductase. Monoclonal antibodies are directed against circulating proprotein convertase subtilisin/kexin type 9. Small interfering RNAs are short RNA molecules that interfere with the production of different proteins through a “carrier” (viral or non-viral vectors), forming the RNA-induced silencing complex that binds to the target complementary mRNA, cleaving it. Lomitapide is an oral selective inhibitor of the microsomial triglyceride transfer protein, decreasing the production of triglycerides, chylomicrons and very low-density lipoproteins. Evinacumab is a monoclonal antibody blocking ANGPTL-3 that inhibits lipoprotein lipase, responsible for hydrolysis of triglycerides content of very low-density lipoproteins. ANGPTL: Angiopoietin-like proteins, HMG-CoA: hydroxy-3-methyl-glutaryl-coenzyme A, LDL: low-density lipoprotein, LPL: lipoprotein lipase, MTP, microsomial triglyceride transfer protein, NPC1L1: Niemann-Pick C1-like protein 1, PCSK9: proprotein convertase subtilisin/kexin type 9.</p>
Full article ">
16 pages, 1589 KiB  
Review
Pleiotropic Effects of PCSK9 Inhibitors on Cardio-Cerebrovascular Diseases
by Zhenzhen Li, Lin Zhu, Yeqiong Xu, Yiting Zhang, Yukai Liu, Huiling Sun, Shuo Li, Meng Wang, Teng Jiang, Junshan Zhou and Qiwen Deng
Biomedicines 2024, 12(12), 2729; https://doi.org/10.3390/biomedicines12122729 - 28 Nov 2024
Viewed by 437
Abstract
Cardiovascular disease (CVD) and ischemic stroke (IS) are the primary causes of mortality worldwide. Hypercholesterolemia has been recognized as an independent risk factor for CVD and IS. Numerous clinical trials have unequivocally demonstrated that reducing levels of low-density lipoprotein cholesterol (LDL-C) significantly mitigates [...] Read more.
Cardiovascular disease (CVD) and ischemic stroke (IS) are the primary causes of mortality worldwide. Hypercholesterolemia has been recognized as an independent risk factor for CVD and IS. Numerous clinical trials have unequivocally demonstrated that reducing levels of low-density lipoprotein cholesterol (LDL-C) significantly mitigates the risk of both cardiac and cerebral vascular events, thereby enhancing patient prognosis. Consequently, LDL-C reduction remains a pivotal therapeutic strategy for CVD and IS. However, despite intensive statin therapy, a significant proportion of high-risk hypercholesterolemic patients fail to achieve sufficient reductions in LDL-C levels. In response to this challenge, an inhibitor targeting proprotein convertase subtilisin-kexin type 9 (PCSK9) has been developed as a therapeutic intervention for hyperlipidemia. Numerous randomized controlled trials (RCTs) have conclusively demonstrated that the combination of PCSK9 inhibitors and statins significantly enhances prognosis not only in patients with CVD, but also in those afflicted with symptomatic intracranial artery stenosis (sICAS). PCSK9 inhibitors significantly reduce LDL-C levels by binding to the PCSK9 molecule and preventing its interaction with LDLRs. This prevents degradation of the receptor and increases uptake of LDL-C, thereby decreasing its concentration in blood. Besides significantly reducing LDL-C levels, PCSK9 inhibitors also demonstrate anti-inflammatory and anti-atherosclerotic properties while promoting plaque stabilization and inhibiting platelet aggregation and thrombosis. This article aims to provide a comprehensive review based on the relevant literature regarding the evolving understanding of pleiotropic effects associated with PCSK9 inhibitors, particularly focusing on their impact on the cardiovascular system and central nervous system. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Figure 1

Figure 1
<p><b>Timeline of research on PCSK9.</b> In 2001, PCSK9 was initially discovered. In 2007, the structure of PCSK9 was revealed binding to LDLR. In 2015, PCSK9 mAbs was first approved by FDA/EMA. Subsequently, a series of phase I, II, and III trials were conducted. In 2021, PCSK9 siRNA was approved by FDA/EMA. Abbreviations: PCSK9, proprotein convertase subtilisin-kexin type 9. LDLR, low-density lipoprotein cholesterol receptor. PCSK9 KO, PCSK9 knockout. PCSK9 mAbs, monoclonal antibodies against PCSK9. FDA, the Food and Drug Administration. EMA, the European Medicines Agency. PCSK9 siRNA, PCSK9 small interfering RNA. The figure was created with BioRender.com.</p>
Full article ">Figure 2
<p><b>PCSK9 inhibitors in lipid metabolism and anti-atherosclerotic and anti-inflammatory effects.</b> First, PCSK9 binds to the LDLR, leading to a reduction in LDLR expression. Consequently, the ability to eliminate LDL-C diminishes. PCSK9 mAbs significantly reduce LDL-C levels by binding to the PCSK9 molecule and preventing its interaction with the LDLR. This prevents degradation of the receptor and increases uptake of LDL-C, thereby decreasing its concentration in blood. Second, PCSK9 siRNA effectively inhibits endothelial cell apoptosis by reducing the expression of pro-apoptotic proteins such as Bax, Caspase3, and Caspase9. Conversely, it is observed that the level of anti-apoptotic protein Bcl-2 is increased, ultimately reducing the development of atherosclerosis. Third, PCSK9 mAbs effectively suppress monocyte adhesion to endothelial cells through reducing the expression of NF-κB, eNOS, and ICAM-1. Ultimately, this leads to a decrease in both the size of atherosclerotic plaques formed as well as an attenuation of the inflammatory response. In addition, macrophage inflammatory responses induced by ox-LDL play a pivotal role in the pathogenesis of atherosclerosis. PCSK9 may accelerate inflammation of atherosclerotic plaques through activating the TLR 4/NF-κB pathway and promoting interactions between SRs on macrophage surfaces. PCSK9 siRNA reduces macrophage numbers and attenuates inflammatory response triggered by ox-LDLs in macrophages. Consequently, there is a subsequent decrease in the expression of vascular inflammation regulators such as TNF-α, IL-1β, and MCP-1. Abbreviations: PCSK9, proprotein convertase subtilisin-kexin type 9. LDL, low-density lipoprotein cholesterol. LDLR, low-density lipoprotein cholesterol receptor. PCSK9 mAbs, monoclonal antibodies against PCSK9. siRNA, small interfering RNA. NF-κB, nuclear factor kappa-B. eNOS, endothelial nitric oxide synthase. ICAM-1, intercellular cell adhesion molecule-1. ox-LDL, oxidized low-density lipoprotein. TLR 4, Toll-like receptor 4. TNF-α, tumor necrosis factor-alpha. IL-1β, interleukin 1 beta. MCP-1, monocyte chemoattractant protein-1. The figure was created with BioRender.com.</p>
Full article ">Figure 3
<p><b>PCSK9 inhibitors in antiplatelet aggregation and antithrombosis</b>. PCSK9 activates CD36 and LOX-1 receptors in platelets, thereby enhancing platelet activation. The interaction between CD36 and ox-LDL triggers signaling pathways, leading to the expression of P-selectin and activation of integrin αIIb β3 (the receptor for fibrinogen). This promotes the formation of platelet–leukocyte complexes through P-selectin and cross-linking adjacent platelets via fibrinogen. Additionally, the binding of ox-LDL to LOX-1 triggers the activation of integrins αIIbβ3 and α2β1, subsequently inducing alterations in platelet morphology and aggregation, ultimately facilitating thrombosis. In addition, deficiency in PCSK9 is associated with protection against venous thrombosis by reducing leukocyte recruitment and NET formation at the site of thrombosis. Abbreviations: PCSK9, proprotein convertase subtilisin-kexin type 9. ox-LDL, oxidized low-density lipoprotein. CD36, platelet glycoprotein 4, a scavenger receptor (SR). LOX-1, lectin-like oxidized low-density lipoprotein receptor-1. αIIbβ3 and α2β1, the receptors for fibrinogen. The figure was created with BioRender.com.</p>
Full article ">Figure 4
<p><b>Pleiotropic effects of PCSK9 inhibitors on cardio-cerebrovascular diseases</b>. PCSK9 inhibitors not only influence lipid metabolism but also exert effects on the cardiovascular system, central nervous system, and other physiological processes. Abbreviations: PCSK9, proprotein convertase subtilisin-kexin type 9. CVD, cardiovascular disease. IS, ischemic stroke. The figure was created with BioRender.com.</p>
Full article ">
15 pages, 2547 KiB  
Article
Carotenoid Interactions with PCSK9: Exploring Novel Cholesterol-Lowering Strategies
by Alessandro Medoro, Giovanni Scapagnini, Simone Brogi, Tassadaq Hussain Jafar, Truong Tan Trung, Luciano Saso and Sergio Davinelli
Pharmaceuticals 2024, 17(12), 1597; https://doi.org/10.3390/ph17121597 - 27 Nov 2024
Viewed by 697
Abstract
Background/Objectives: This study investigated the potential of green algae-derived carotenoids as natural inhibitors of the proprotein convertase subtilisin/kexin type 9 (PCSK9), a key regulator of cholesterol metabolism. PCSK9 promotes the degradation of low-density lipoprotein receptors (LDLR), thereby increasing blood cholesterol levels and [...] Read more.
Background/Objectives: This study investigated the potential of green algae-derived carotenoids as natural inhibitors of the proprotein convertase subtilisin/kexin type 9 (PCSK9), a key regulator of cholesterol metabolism. PCSK9 promotes the degradation of low-density lipoprotein receptors (LDLR), thereby increasing blood cholesterol levels and elevating the risk of cardiovascular diseases. Methods/Results: We screened the pharmacophore fit score of 27 carotenoids with PCSK9 and identified 14 that were analyzed for binding affinity and molecular interactions. Astaxanthin, siphonaxanthin, and prasinoxanthin were identified as the top candidates, demonstrating strong binding affinity (−10.5, −10.3, and −9.4 Kcal/mol, respectively) and stable interactions with several known key residues within the active site of PCSK9, including Pro-331, Arg-357, Cys-358, Val-359, Asp-360, Ile-416, Leu-436, Thr-437, Pro-438, Leu-440, Arg-458, Val-460, Trp-461, Arg-476, Cys-477, Ala-478, Ala-649, Val-650, and Asp-651. Density functional theory analysis confirmed the stability of astaxanthin and its favorable electronic properties, suggesting its potential as an effective inhibitor. Molecular dynamics simulations of the PCSK9–astaxanthin complex revealed sustained structural stability and key interactions critical for maintaining the functional integrity of the protein. Conclusions: These findings provide evidence that specific carotenoids, particularly astaxanthin, may offer a cost-effective alternative to existing PCSK9 inhibitors, providing a potential approach for managing cholesterol levels and reducing cardiovascular risk. Pre-clinical and clinical validations are required to confirm the therapeutic potential of these compounds. Full article
(This article belongs to the Special Issue The Role of Phytochemicals in Aging and Aging-Related Diseases)
Show Figures

Figure 1

Figure 1
<p>3D and 2D molecular interaction analyses of PCSK9 (grey) with (<b>A</b>) astaxanthin (dark green), (<b>B</b>) siphonaxanthin (light green), and (<b>C</b>) prasinoxanthin (blue). All three compounds showed similar residue interaction with PCSK9 due to the high structural similarity. Astaxanthin and siphonaxanthin showed almost two hydrogen bonds with key residues.</p>
Full article ">Figure 2
<p>(<b>A</b>) Optimized geometry (<b>B</b>) HOMO, LUMO orbitals and their energy gap (ΔEg), (<b>C</b>) DOS plot diagram, and (<b>D</b>) MEP map of astaxanthin by DFT level of calculations.</p>
Full article ">Figure 3
<p>(<b>A</b>) RMSD analysis of simulation complex PCSK9–astaxanthin. The left Y-axis showed the variation of PCSK9 RMSD through time, while the right Y-axis showed the variation of astaxanthin RMSD through time. (<b>B</b>) The analysis of RMSF during simulation complex of PCSK9–astaxanthin.</p>
Full article ">Figure 4
<p>(<b>A</b>) PCSK9 SSE (%) distribution by residue index throughout the simulation complex. Red columns indicate the alpha helices, and light blue columns indicate the beta strands. (<b>B</b>) PCSK9–astaxanthin contact histogram. Hydrogen bonds are in green, hydrophobic interaction in purple, and water bridges in blue.</p>
Full article ">Figure 5
<p>Graphical representations of rGyr, IntraHB, MolSA, SASA, and PSA.</p>
Full article ">
17 pages, 572 KiB  
Review
PCSK9 Inhibitors: Focus on Evolocumab and Its Impact on Atherosclerosis Progression
by Maram H. Abduljabbar
Pharmaceuticals 2024, 17(12), 1581; https://doi.org/10.3390/ph17121581 - 25 Nov 2024
Viewed by 1267
Abstract
This paper investigates the therapeutic use of PCSK9 inhibitors, particularly Evolocumab, as monoclonal antibodies for the treatment of atherosclerosis based on recent literature reviews. PCSK9 is an outstanding example of a breakthrough in medical science, with advancements in understanding its biological function driving [...] Read more.
This paper investigates the therapeutic use of PCSK9 inhibitors, particularly Evolocumab, as monoclonal antibodies for the treatment of atherosclerosis based on recent literature reviews. PCSK9 is an outstanding example of a breakthrough in medical science, with advancements in understanding its biological function driving substantial progress in atherosclerosis treatment. Atherosclerotic cardiovascular disease (ASCVD) is a leading global cause of mortality, imposing substantial financial burdens on healthcare systems. Elevated low-density lipoprotein cholesterol (LDL-C), a modifiable risk factor, plays a pivotal role in the development of ASCVD. Emerging treatments such as PCSK9 inhibitors are now being introduced to combat this issue, with the goal of reducing ASCVD risk by directly targeting LDL-C levels. This discovery highlighted the potential of monoclonal antibodies to inhibit PCSK9, thereby enhancing LDL-C receptor activity. This breakthrough led to the development of Alirocumab and Evolocumab inhibitors, which typically reduce LDL-C levels by approximately 50%. This research underscores the importance of PCSK9 inhibitors in treating ASCVD, drawing on evidence from various randomized controlled trials such as FOURIER, ODYSSEY OUTCOMES, and VESALIUS-CV. These trials have also shown that PCSK9 inhibitors are effective and safe for the treatment of several cardiovascular disorders. PCSK9 inhibitors are therefore useful in patients who do not reach their target LDL-C levels when on the highest doses of statins or patients with very high cardiovascular risk who cannot tolerate statins at all. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The role of PCSK9 (Evolocumab) inhibitors in CVD.</p>
Full article ">
20 pages, 12941 KiB  
Article
Impact of Kiwifruit Consumption on Cholesterol Metabolism in Rat Liver: A Gene Expression Analysis in Induced Hypercholesterolemia
by Abdolvahab Ebrahimpour Gorji, Anna Ciecierska, Hanna Leontowicz, Zahra Roudbari and Tomasz Sadkowski
Nutrients 2024, 16(23), 3999; https://doi.org/10.3390/nu16233999 - 22 Nov 2024
Viewed by 741
Abstract
Background/Objectives: Cholesterol is vital in various bodily functions, such as maintaining cell membranes, producing hormones, etc. However, imbalances, like hypercholesterolemia, can lead to diseases such as cancer, kidney disease, non-alcoholic fatty liver disease, and cardiovascular conditions. This study explores the impact of kiwifruit [...] Read more.
Background/Objectives: Cholesterol is vital in various bodily functions, such as maintaining cell membranes, producing hormones, etc. However, imbalances, like hypercholesterolemia, can lead to diseases such as cancer, kidney disease, non-alcoholic fatty liver disease, and cardiovascular conditions. This study explores the impact of kiwifruit consumption, specifically Actinidia arguta cultivar Geneva and Actinidia deliciosa cultivar Hayward, on cholesterol and lipid metabolism in rat liver. Methods: Rats were divided into groups: a 1% cholesterol control group (Ch), a 5% Geneva kiwifruit-supplemented group (ChGENE), and a 5% Hayward kiwifruit-supplemented group (ChHAYW). Gene expression was analyzed using Gene Spring v.14. Gene ontology, pathway analysis, miRNA, and transcription factor prediction were performed using DAVID, Reactome, and miRNet. In addition, we used Agilent Literature Search software to gain further insights. Results: Statistical analysis identified 72 genes in ChGENE-Ch and 2 genes in ChHAYW-Ch comparison. Key genes involved in cholesterol metabolism pathways, including PCSK9, SCD1, SLC27A5, HMGCR, and DHCR24, showed lower expression in the kiwifruit-supplemented groups. The genes mentioned above showed lower expression in the kiwifruit-supplemented group, probably contributing to the liver lipid level reduction. Further analysis identified miRNA-26a, miRNA-29a/b/c, miRNA-33a/b, and miRNA-155 targeting hub genes. Conclusions: Our findings suggest that dietary supplementation with kiwifruit, particularly the Geneva cultivar, reduces fat accumulation in the liver of rats with hypercholesterolemia, likely through downregulation of critical genes involved in cholesterol metabolism. These studies highlight the potential of kiwifruit as a part of a dietary strategy to manage cholesterol levels. Full article
(This article belongs to the Special Issue Healthy Diet to Prevent Cardiovascular Disease)
Show Figures

Figure 1

Figure 1
<p>Schematic showing the main steps of the experimental setup. (<b>A</b>)—Ch control group fed with a semi-synthetic diet with the addition of 1% cholesterol, (<b>B</b>)—ChGENE group fed a semi-synthetic diet with the addition of 1% cholesterol and 5% freeze-dried fruits of <span class="html-italic">A. arguta</span> cultivar Geneva, (<b>C</b>)—ChHAYW group fed with diet semi-synthetic with the addition of 1% cholesterol and 5% freeze-dried <span class="html-italic">A. deliciosa</span> cultivar Hayward.</p>
Full article ">Figure 2
<p>The gene expression profiles of ChGENE and ChHAYW in comparison to Ch. (<b>A</b>) Heatmap depicting the gene expression of ChGENE. (<b>B</b>) Heatmap illustrating the gene expression of ChHAYW. (<b>C</b>) Presentation of the top ten upregulated and downregulated genes in ChGENE compared to Ch.</p>
Full article ">Figure 3
<p>(<b>A</b>) The circular heatmap visually conveys the proportion of shared gene expression between ChGENE and ChHAYW. (<b>B</b>) The Venn diagram illustrates the gene expression count in ChGENE and ChHAYW.</p>
Full article ">Figure 4
<p>Quantitative expression analysis of genes related to lipids and cholesterol metabolism by Real-time qPCR. Statistical significance is shown as * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>The chord plot represents the relationship between GO terms and genes exhibiting significant expression changes under the ChGENE and ChHAYW treatments.</p>
Full article ">Figure 6
<p>A Sankey plot depicting genes within individual pathways (<b>left</b>) and a dot plot where the size of the dots corresponds to the number of genes; the dot’s color signifies the <span class="html-italic">p</span>-values (<b>right</b>) for the ChGENE treatment.</p>
Full article ">Figure 7
<p>A Sankey plot depicting genes within individual pathways (<b>left</b>) and a dot plot where the size of the dots corresponds to the number of genes and the color of the dots signify the <span class="html-italic">p</span>-values (<b>right</b>) for the ChHAYW treatment.</p>
Full article ">Figure 8
<p>Agilent Literature Search network analysis between DEGs and other critical genes governing cholesterol and lipid metabolism. Green squares denote genes identified by Agilent Literature Search as engaged in lipid metabolism; red triangles signify upregulated DEGs; blue V-shaped symbols signify downregulated DEGs.</p>
Full article ">Figure 9
<p>The interaction network of predicted miRNAs and their corresponding target DEGs. MiRNAs—blue squares; TFs—green circles; DEGs—yellow circles; red line—connection between TF and DEGs; gray line—connection between miRNAs and DEGs.</p>
Full article ">
8 pages, 229 KiB  
Perspective
Therapeutic Challenges Derived from the Interaction Among Apolipoprotein E, Cholesterol, and Amyloid in Alzheimer’s Disease
by Manuel Menendez-Gonzalez
Int. J. Mol. Sci. 2024, 25(22), 12029; https://doi.org/10.3390/ijms252212029 - 8 Nov 2024
Viewed by 1390
Abstract
The isoform E4 of the Apolipoprotein E (ApoE) represents one of the strongest genetic risk factors for late-onset Alzheimer’s disease (AD). ApoE has key roles in cholesterol transport and amyloid-β (Aβ) metabolism, which are both central to AD pathogenesis. The E4 isoform has [...] Read more.
The isoform E4 of the Apolipoprotein E (ApoE) represents one of the strongest genetic risk factors for late-onset Alzheimer’s disease (AD). ApoE has key roles in cholesterol transport and amyloid-β (Aβ) metabolism, which are both central to AD pathogenesis. The E4 isoform has been implicated in reduced cholesterol homeostasis, increased Aβ aggregation, and heightened tau phosphorylation, contributing to amyloid plaques and neurodegeneration. This manuscript examines the complex interactions among ApoE isoforms, cholesterol metabolism, and amyloid pathology. Moreover, the therapeutic challenges associated with lipid-lowering agents (e.g., statins, PCSK9 inhibitors), anti-amyloid immunotherapies, and anticoagulants are described, focusing on ApoE4 carriers. Decision-making challenges are discussed by analyzing the pros and cons of these therapies. Full article
(This article belongs to the Special Issue New Advances in Research on Alzheimer’s Disease: 2nd Edition)
10 pages, 2740 KiB  
Article
Correlation of Eight (8) Polymorphisms and Their Genotypes with the Risk Factors of Cardiovascular Disease in a Black Elderly Population
by Joseph Musonda Chalwe, Christa Johanna Grobler and Wilna Hendrika Oldewage-Theron
Curr. Issues Mol. Biol. 2024, 46(11), 12694-12703; https://doi.org/10.3390/cimb46110753 - 8 Nov 2024
Viewed by 932
Abstract
Single nucleotide polymorphisms (SNPs) have been associated with the development of cardiovascular diseases (CVDs). This study correlated eight SNPs with the risk factors of CVD in a black elderly population. Genotyping was used to detect eight polymorphisms; rs675 (ApoA-IV), rs699 (Angiotensinogen (AGT)), rs247616 [...] Read more.
Single nucleotide polymorphisms (SNPs) have been associated with the development of cardiovascular diseases (CVDs). This study correlated eight SNPs with the risk factors of CVD in a black elderly population. Genotyping was used to detect eight polymorphisms; rs675 (ApoA-IV), rs699 (Angiotensinogen (AGT)), rs247616 and rs1968905 (Cholesteryl ester transfer protein (CETP)), rs1801278 (Insulin receptor substrate 1 (IRS-1)), rs1805087 (Methylenetetrahydrofolate reductase (MTHFR)) and rs28362286 and rs67608943 (Proprotein convertase subtilisin/kexin type 9 (PCSK9)), as well as their genotypes in deoxyribonucleic acid (DNA) extracted from peripheral blood. The cardiovascular risk (CVR) measurements were conducted on a Konelab 20i Thermo Scientific autoanalyzer and an enzyme-linked immunoassay (ELISA) assay. International Business Machines Corporation (IBM)® Statistical Package for the Social Sciences ® (SPSS) version 28 was used for statistical analysis. The heterozygous and homozygous genotypes of the eight polymorphisms were detected with the corresponding CVD risk factors. Subgroup analysis indicated that certain genotype carriers exhibited variations in their concentrations of CVR factors compared to others; however, these differences did not reach statistical significance. For example, carriers of the G genotype of the rs699 polymorphism showed marginally different blood pressure readings compared to the AG genotype carriers. The multiple linear regression analysis indicated that the only significant association was between PCSK9 and the rs28362286 (p = 0.029) polymorphism. The findings of our study show that single nucleotide polymorphisms are disseminated across the human genome. The heterozygous and homozygous genotypes of the SNPs require further investigation as they may have independent and possible collective roles in increasing the risk of CVDs. Full article
(This article belongs to the Special Issue A Focus on the Molecular Basis of Cardiovascular Diseases)
Show Figures

Graphical abstract

Graphical abstract
Full article ">
7 pages, 411 KiB  
Brief Report
Nonalcoholic Fatty Liver Disease Risk and Proprotein Convertase Subtilisin Kexin 9 in Familial Hypercholesterolemia Under Statin Treatment
by Masato Hamasaki, Naoki Sakane and Kazuhiko Kotani
Nutrients 2024, 16(21), 3686; https://doi.org/10.3390/nu16213686 - 29 Oct 2024
Viewed by 865
Abstract
Background/Objectives: Fatty acids are involved in some hepatic disorders. The proprotein convertase subtilisin kexin 9 (PCSK9) inhibits the uptake of low-density lipoproteins (LDLs), which contain lipids, into the liver and may thus be associated with nonalcoholic fatty liver disease (NAFLD), a cardiovascular disorder [...] Read more.
Background/Objectives: Fatty acids are involved in some hepatic disorders. The proprotein convertase subtilisin kexin 9 (PCSK9) inhibits the uptake of low-density lipoproteins (LDLs), which contain lipids, into the liver and may thus be associated with nonalcoholic fatty liver disease (NAFLD), a cardiovascular disorder (CVD) risk. Statins reduce blood LDL–cholesterol (LDL-C) levels and CVD risk and can attenuate the development of NAFLD while increasing blood PCSK9 levels. Methods: We investigated the correlation between PCSK9 and liver conditions in patients with familial hypercholesterolemia (FH), a CVD risk population with elevated blood LDL-C levels, under statin treatment. Blood tests for lipids, PCSK9, and liver function (aspartate aminotransferase [AST] and alanine aminotransferase [ALT]) were performed in patients with FH taking statins (n = 25, mean age = 57 years, 12% of males). The ALT:AST ratio was used as a marker of NAFLD risk. Results: The mean LDL-C level was 3.38 mmol/L, and the median PCSK9 level was 312 ng/mL. The median ALT:AST ratio was 0.88. A significant negative correlation was observed between the PCSK9 and ALT:AST ratio (β = −0.67, p < 0.05). Conclusions: Their negative correlation might give a hypothetical insight into the effect of statin treatment on the development of NAFLD, in relation to PCSK9 behavior, in patients with FH. Full article
Show Figures

Figure 1

Figure 1
<p>Correlation between the PCSK9 and ALT:AST ratio in patients with FH under statin treatment. ALT, alanine aminotransferase; AST, aspartate aminotransferase; PCSK9, proprotein convertase subtilisin/kexin type 9. The PCSK9 levels and ALT:AST ratio were log-transformed. Pearson’s correlation coefficient (<span class="html-italic">r</span>) was = −0.45 (<span class="html-italic">p</span> = 0.02).</p>
Full article ">
18 pages, 4142 KiB  
Article
Antarctic Krill Euphausia superba Oil Supplementation Attenuates Hypercholesterolemia, Fatty Liver, and Oxidative Stress in Diet-Induced Obese Mice
by Jun-Hui Choi, Se-Eun Park and Seung Kim
Nutrients 2024, 16(21), 3614; https://doi.org/10.3390/nu16213614 - 24 Oct 2024
Viewed by 1246
Abstract
Background: Several Previous studies indicate that consuming krill oil may aid in reducing hypercholesterolemia and improving cholesterol metabolism. Therefore, our study was designed to investigate the effectiveness of Antarctic krill oil (Euphausia superba) (ESKO) in combating obesity and lowering fat/lipid/cholesterol levels. [...] Read more.
Background: Several Previous studies indicate that consuming krill oil may aid in reducing hypercholesterolemia and improving cholesterol metabolism. Therefore, our study was designed to investigate the effectiveness of Antarctic krill oil (Euphausia superba) (ESKO) in combating obesity and lowering fat/lipid/cholesterol levels. Methods: The study aimed to investigate the molecular docking model targeting 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) using ESKO-derived eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and astaxanthin. In this study, histological alterations in the liver of the obesity model (ICR male mouse), obesity-related or antioxidant markers in both liver and serum, the molecular mechanisms in HepG2 cells and liver tissue, and HMGCR activity were analyzed. Results: Our findings revealed that a high-fat diet (HFD) significantly led to increased oxidative stress, obesity-related indicators, and cardiovascular-associated risk indices. However, ESKO effectively mitigated HFD-induced oxidative stress, fat accumulation, and the suppression of low-density lipoprotein receptor (LDLR) or activation of related molecular pathways. This was achieved through improvements in metabolic parameters, including CD36/liver X receptor α (LXRα)/sterol regulatory element-binding protein 1c (SREBP1c), proprotein convertase subtilsin/kexin type 9 (PCSK-9), and HMGCR, ultimately ameliorating HFD-induced hypercholesterolemia and obesity. Conclusions: These beneficial findings indicate that ESKO might have significant potential for preventing and treating obesity-related disorders. Full article
(This article belongs to the Special Issue Nutrition and Dietary Intake in Liver-Related Diseases)
Show Figures

Figure 1

Figure 1
<p>Effect of ESKO on cell viability (<b>A</b>,<b>B</b>), lipid (<b>C</b>), triglyceride (<b>D</b>) level, and HMGCR activity (<b>E</b>) in HepG2 cells. MTT assay showing the effect of ESKO and palmitic acid on HepG2 cells viability. Cells were incubated with ESKO at different concentrations (0–1000 μg/mL) or palmitic acid (0–1000 μM) for 24 h and cell viability was analyzed by MTT reduction assay. Each value is the mean ± SD of triplicate measurements. # <span class="html-italic">p</span> &lt; 0.01, compared with Control group, * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01, compared with palmitic acid only-treated group. ESKO, <span class="html-italic">Euphausia superba</span> krill oil; HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR).</p>
Full article ">Figure 2
<p>Effect of ESKO on CD36 (<b>A</b>,<b>B</b>), LXRα (<b>C</b>), SREBP1C (<b>D</b>), LDLR (<b>E</b>), and PCSK9 (<b>F</b>) signaling and HMGCR activity (<b>G</b>) in HepG2 cells. Each value is the mean ± SD of triplicate measurements. # <span class="html-italic">p</span> &lt; 0.01, compared with Control group, * <span class="html-italic">p</span> &lt; 0.01, compared with palmitic acid only-treated group as negative control (NC). PA, palmitic acid; ESKO, <span class="html-italic">Euphausia superba</span> krill oil; LXRα, liver X receptor α; SREBP1c, sterol regulatory element-binding protein 1c; LDLR, low-density lipoprotein receptor; PCSK9, proprotein convertase subtilsin/kexin type 9; HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase.</p>
Full article ">Figure 3
<p>Effect of ESKO on body weight (<b>A</b>,<b>C</b>,<b>D</b>) and feed intake (<b>B</b>,<b>E</b>) in DIO mouse models. Each value is the mean ± SD (<span class="html-italic">n</span> = 6). # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01, compared with Control group. Control, non-treated normal group; Model, diet-induced obesity (DIO) model group; Feno, 200 mg/kg fenofibrate-administrated DIO group, ESKO, 400 mg/kg <span class="html-italic">Euphausia superba</span> krill oil-administrated DIO group.</p>
Full article ">Figure 4
<p>Histopathological analysis of livers and epididymal fats from DIO models. Effects of ESKO and fenofibrate on hepatic (<b>A</b>,<b>B</b>) or epididymal fats (<b>C</b>,<b>D</b>) in DIO model was analyzed with H&amp;E, microscope, ImageJ, or fat weight. Each value is the mean ± SD (<span class="html-italic">n</span> = 6). # <span class="html-italic">p</span> &lt; 0.01, compared with Control group, * <span class="html-italic">p</span> &lt; 0.01, compared with DIO model group. Control, non-treated normal group; Model, diet-induced obesity (DIO) model group; Feno, 200 mg/kg fenofibrate-administrated DIO group, ESKO, 400 mg/kg <span class="html-italic">Euphausia superba</span> krill oil-administrated DIO group.</p>
Full article ">Figure 5
<p>Effect of ESKO on CD36 (<b>A</b>,<b>B</b>), LXRα (<b>C</b>), SREBP1C (<b>D</b>), LDLR (<b>E</b>), and PCSK9 (<b>F</b>) signaling and HMGCR activity (<b>G</b>) in livers from DIO model. Each value is the mean ± SD (<span class="html-italic">n</span> = 6). # <span class="html-italic">p</span> &lt; 0.01, compared with Control group, * <span class="html-italic">p</span> &lt; 0.01, compared with DIO Model group. Control, non-treated normal group; Model, diet-induced obesity (DIO) Model group; Feno, 200 mg/kg fenofibrate-administrated DIO group, ESKO, 400 mg/kg <span class="html-italic">Euphausia superba</span> krill oil-administrated DIO group. LXRα, liver X receptor α; SREBP1c, sterol regulatory element-binding protein 1c; LDLR, low-density lipoprotein receptor; PCSK9, proprotein convertase subtilsin/kexin type 9; HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase.</p>
Full article ">
9 pages, 861 KiB  
Article
Proprotein Convertase Subtilisin Kexin 9 Inhibitor in Severe Sepsis and Septic Shock Patients in a Phase II Prospective Cohort Study—Preliminary Results
by Ziv Rosman, Yasmin Maor, Iris Zohar, Gingy Ronen Balmor, Miri Schamroth Pravda, Adam Lee Goldstein, Milena Tocut and Arie Soroksky
Infect. Dis. Rep. 2024, 16(6), 1036-1044; https://doi.org/10.3390/idr16060083 - 24 Oct 2024
Viewed by 899
Abstract
Sepsis is a life-threatening organ dysfunction syndrome caused by a dysregulated host response to infection that has a high mortality rate. Proprotein convertase subtilisin kexin 9 (PCSK9) is a serine protease secreted by the liver. Its binding to the low-density lipoprotein (LDL) receptor [...] Read more.
Sepsis is a life-threatening organ dysfunction syndrome caused by a dysregulated host response to infection that has a high mortality rate. Proprotein convertase subtilisin kexin 9 (PCSK9) is a serine protease secreted by the liver. Its binding to the low-density lipoprotein (LDL) receptor enhances its degradation, causing an increase in LDL levels in the blood. Objectives: Administering a PCSK9 inhibitor leading to an increase in lipid uptake by the liver may positively affect septic patients due to the increased removal of endotoxins. Methods: This preliminary study aimed to examine the safety of PCSK9 inhibitor use in septic and septic shock patients. We treated five septic patients in the intensive care unit with 300 mg of alirocumab following serious adverse events for 28 days. Results: Four of our patients did not experience any adverse events, and all of them survived. One patient died after discharge from the intensive care unit, and this death was presumably not related to the study drug. The patients rapidly recovered from the inflammatory stage of sepsis. Conclusions: Alirocumab appears safe in severe sepsis and septic shock patients. The outcome data are promising. Only a basic safety profile can be assessed based on this pilot study. Further study with a PCSK-9 inhibitor in septic or septic shock patients is required to further determine its benefit in ICU patients. Full article
(This article belongs to the Section Bacterial Diseases)
Show Figures

Figure 1

Figure 1
<p>Patient recruitment.</p>
Full article ">Figure 2
<p>Procalcitonin levels.</p>
Full article ">Figure 3
<p>IL-6 levels. Patient V’s IL-6 samples were partially lost.</p>
Full article ">Figure 4
<p>CRP levels.</p>
Full article ">Figure 5
<p>WBC levels.</p>
Full article ">
Back to TopTop