You are viewing a javascript disabled version of the site. Please enable Javascript for this site to function properly.
Go to headerGo to navigationGo to searchGo to contentsGo to footer
In content section. Select this link to jump to navigation

Small-interfering RNA targeting proprotein convertase subtilisin/kexin type 9 might promote fatty liver disease and hepatocellular carcinoma through upregulation of CD36

Abstract

Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to low-density lipoprotein (LDL) receptor and fatty acid translocase CD36, inducing lysosomal degradation of these two receptors in the liver cells. Both monoclonal antibody (mAb) and small-interfering RNA (siRNA) targeting PCSK9 have been designed for treatment of familial hypercholesterolemia recently, with elevating LDL receptors on the liver cell surface and increasing LDL uptake as the main beneficial mechanism. However, given that the binding domains of PCSK9 for LDL receptor and CD36 are different, and PCSK9 mAb only attacks the domain for LDL receptor, CD36 expression remains partially controlled under PCSK9 mAb treatment. In contrast, PCSK9 siRNA brings on complete loss of PCSK9, resulting in overexpression of CD36. Based on the fact that CD36 is a key factor in the pathogenesis of non-alcoholic fatty liver disease (NAFLD) and subsequent hepatocellular carcinoma (HCC), the risk of developing NAFLD and HCC on long-term use of PCSK9 siRNA is thus raised as a hypothesis. Additionally, because CD36 is also involved in the promotion of malignant diseases other than HCC, such as acute myeloid leukemia, gastric cancer, breast cancer, and colorectal cancer, the speculative danger of flourishing these malignancies by PCSK9 siRNA is discussed as well.

1Introduction

Non-alcoholic fatty liver disease (NAFLD), histologically ranging from non-alcoholic fatty liver to non-alcoholic steatohepatitis (NASH), has evolved to be the most common cause of cryptogenic cirrhosis and one of the main preceding etiologies for development of hepatocellular carcinoma (HCC) [1]. Meta-analysis studies had shown that NAFLD significantly increased the risk of HCC [2], with a 38% prevalence of HCC in non-cirrhotic NASH, much higher than that in non-cirrhotic liver diseases of other etiologies [3]. Treatment of important predisposing factors for NAFLD, such as diabetes, obesity, and metabolic syndrome, has been aggressively advocated to diminish the threat of NAFLD but survey of HCC in NAFLD still remains a challenge because HCC could develop in both cirrhotic and non-cirrhotic NAFLD, while the latter traditionally has not been a focus of HCC screening [4]. Thus, it seems crucial that any possible strategies should be adopted to avoid risky issues linked to the development of NAFLD and subsequent HCC.

Importantly, free fatty acids have been found to be closely related to progressive liver fibrosis seen in NAFLD with their ability to trigger the profibrogenic Hippo signaling transcriptional coactivator yes-associated protein 1 through p38 mitogen-activated protein kinase pathway [5]. Based on the recognition of transmembrane fatty acid translocase cluster of differentiation 36 (CD36) as a master in regulating cellular lipid metabolism by facilitating cellular uptake of long-chain free fatty acids [6], herein, the potential pathogenic roles of CD36 in NAFLD and HCC will be reviewed and two kinds of lipid-lowering agents, the monoclonal antibody (mAb) and small-interfering RNA (siRNA) against proprotein convertase subtilisin/kexin type 9 (PCSK9) which regulates CD36 expression, will be examined to see whether these modern therapeutic modalities for hypercholesterolemia might have adverse effects on liver regarding the risk of NAFLD and HCC.

2CD36 in the pathogenesis of NAFLD and consecutive HCC

CD36 is a multiligand transmembrane receptor which can bind thrombospondin-1, thrombospondin-2, long chain fatty acid, oxidized low density lipoprotein, and oxidized phospholipids, acting also as a receptor for pathogen-associated molecular patterns in activation of innate immunity and clearance of cell debris with phagocytosis besides its role in fatty acid transportation [7]. As a fatty acid translocase, the hydrophobic extracellular domains of CD36 enable fatty acids to be translocated through cell membrane into cells with a high efficiency of fatty acid utilization such as adipose tissue, skeletal muscle, heart, and liver [8].

It has been demonstrated in cultured hepatoma cells that transport of fatty acid into cells is promoted by formation of a heterotetrameric protein complex comprising CD36, caveolin-1, fatty acid-binding protein, and calcium-independent membrane phospholipase A2 [9]. Notably, fatty acid itself can increase CD36 expression via stimulating transcription factor SOX2 [10], then take advantage of CD36-mediated oxidative stress to make hepatocytes activated and involved in the process of liver fibrosis [11]. Therefore, CD36 is considered to be the key driver of fatty acid-related lipotoxicity, including elevated endoplasmic reticulum stress, reactive oxygen species, insulin resistance, tissue damage, apoptosis, and impaired autophagy, during the process of NAFLD with modulation of its expression in the liver as a major influencing factor for the progression of hepatosteatosis [12].

In addition to its pathological role in NAFLD, overexpression of CD36 was detected in human HCC, in two research works being considered to cause the development of HCC. One of those studies discovered that the highly upregulated CD36 exerted a stimulatory effect on HCC growth and metastasis through activating the Src/PI3K/AKT/mTOR signaling pathway-dependent aerobic glycolysis [13]. The other showed that the overexpressed CD36 was able to accelerate the progression of HCC by promoting the expression of aldo– keto reductases family 1 member C2 and increasing fatty acids uptake [14]. Furthermore, it has been demonstrated in murine and human NAFLD-associated HCC tissues that CD36-mediated oxidized low-density lipoprotein (LDL) uptake can activate CCAAT/enhancer-binding protein beta expression to straightly upregulate Nogo-B, a metabolic modulator which interacts with autophagy-related 5 to enhance yes-associated protein 1 oncogenic activity, escalating high-fat diet-induced metabolic disturbance and carcinogenesis in the liver [15].

Even more interestingly, epithelial-mesenchymal transition, a well-known characteristic of cancer progression, has been revealed to be induced by CD36-mediated upward free fatty acid intake in HCC cells via the TGF-β and Wnt/β-catenin signaling pathways [16]. Beside fatty acids, CD36 also cooperates with cartilage oligomeric matrix protein secreted by hepatic stellate cells to boost proliferation, invasion, and metastasis of HCC cells by activating MEK/ERK and PI3K/AKT signaling pathways [17]. Hence, it is no wonder why CD36 has been chosen as an emerging therapeutic target for HCC given its pivotal role in metabolic dysregulation involved in tumour initiation and progression [18].

3Add-on use of PCSK9 mAb and siRNA in lipid control

Hypercholesterolemia is associated with the pathogenesis of a variety of disorders, inclusive of atherosclerotic cardiovascular disease, NAFLD, obesity, diabetes, neurodegenerative diseases, cancer, osteoporosis, and virus infection. Many novel therapeutic modalities aiming at different targets to lower cholesterol levels have been innovated recently. Among those targets, PCSK9 emerges as a striking one, especially for familial hypercholesterolemia [19].

PCSK9, which belongs to the proprotein convertase family and consists of three domains (pro-domain, catalytic domain, and Cys-His-rich domain) in its mature form. It is synthesized in hepatocytes and pancreatic β-cells. PCSK9 can bind to LDL receptor intracellularly within Golgi apparatus or extracellularly on the cell surface when released into plasma. Consequently it induces lysosomal degradation of LDL receptor, reducing uptake of cholesterol to the cell, and ultimately leading to elevated cholesterol levels in the blood. The very-low density lipoprotein receptor is also a target of degradation by PCSK9. Thus, gain-of-function mutations in PCSK9 gene have been identified to be the causes of familial hypercholesterolemia while loss-of-function mutations in PCSK9 gene resulted in hypocholesterolemia [20]. Innovative strategies in development for blocking PCSK9 function and alleviating hypercholesterolemia include mAb, siRNA, vaccine, clustered regularly interspaced short palindromic repeats (CRISPR)/Cas system of gene deletion/editing, and orally active inhibitors. Anti-PCSK9 mAb blocks the interaction between PCSK9 and LDL receptor, reduces the degradation of LDL receptor, and consequently lowers LDL levels. Vaccines induce antibodies targeting PCSK9. Orally active inhibitors are small molecule LDL receptor analogs which can also interfere with the binding of PCSK9 to LDL receptor. siRNA acts by sequences-specific degradation of PCSK9 mRNA and functionally inhibits PCSK9 gene expression. CRISPR/Cas system decreases PCSK9 levels by inducing DNA double-strand breaks, resulting in error structures in PCSK9 gene [21]. So far, major beneficial cholesterol homeostasis outcomes have been detected in clinical trials using mAbs and siRNAs against PCSK9 [22]. These outcomes have received supportive confirmation from meta-analysis studies [23, 24], prompting recommendations for add-on use to achieve better lipid control and prevent cardiovascular diseases in patients already on other lipid-lowering agents, still the treatment goals remain unmet [25].

4A speculation based on different influence of PCSK9 mAb and siRNA on CD36 expression

PCSK9 not only can bind to LDL receptor but also to CD36 in adipocytes and liver cells, subsequently giving rise to degradation of CD36 and lowering cellular uptake of long-chain fatty acids [26]. Nonetheless, the PCSK9 domains which attach to LDL receptor and CD36 are not the same. Degradation of LDL receptor requires the catalytic unit of PCSK9, while the C-terminal Cys-His rich domain (CHRD) of PCSK9 is responsible for CD36 binding. In the presence of PCSK9 mAbs, which attack PCSK9 on the catalytic domain and rescue LDL receptor from endocytosis, the C-terminal CHRD of PCSK9 can still tie up CD36 and bring on partial reduction of CD36 through endosomes/lysosomes degradation [27]. On the contrary, silencing PCSK9 synthesis with siRNA decreases the whole PCSK9 product and presumably preserves the LDL receptor and CD36 on the cell surface [28].

On that account, a meta-analysis of clinical trials designed for treating hypercholesterolemia with siRNA targeting PCSK9 (inclisiran) revealed a 51% decrease of LDL level and a 24% lower major adverse cardiovascular events rate as compared with placebo [29]. Additionally, a pooled patient-level analysis of inclisiran trials for familial hypercholesterolemia disclosed a 50.7% placebo-corrected drop of LDL levels [30]. However, the risk of persistent elevated CD36 expression in hepatocytes on long-term use of PCSK9 siRNA ought to be meticulously examined.

Animal experiments demonstrated clearly that high fat-diet was able to induce hepatic endoplasmic reticulum stress, insulin resistance, NASH, and fibrosis in PCSK9 knockout mice [31]. Moreover, when injected with the hepatic carcinogen diethylnitrosamine early-in-life, PCSK9 knockout mice were found to develop liver cancer more likely than wild type mice [32]. Accordingly, based on the pathogenic roles of CD36 in liver diseases, it seems reasonable to assume that deletion of PCSK9 expression for a long duration via PCSK9 siRNA treatment might promote the development of NAFLD or even HCC due to uncontrolled CD36 expression.

5Discussion

Intriguingly, single neucleotide polymorphism of PCSK9 gene with nonsynoymous variants in the population has been detected years ago. As we know from published reports that both heterozygous and homozygous carriers of PCSK9 R46 L variant had comparatively lower lipoprotein(a) and LDL cholesterol levels than non-carriers, resulting in a significantly reduced odds ratio for myocardial infarction or aortic stenosis, presumably due to decreased atherosclerotic burden [33]. This finding was confirmed by a series of population studies and meta-analyses [34]. Whether this particular variant leads to hepatosteatosis, however, is a problem of debate, with positive and negative opinions [35, 36]. Importantly, the so-called loss-of-function of this variant probably interferes with the regulation of LDL receptor expression only. The R46 L missense mutation locates at the pro-domain, which remains associated with the catalytic domain responsible for LDL receptor binding after maturation of PCSK9 [37]. Therefore, it likely does not affect the CHRD responsible for CD36 binding and degradation. It is no wonder then that the R46 L variant, assumed here to still be capable of downregulating liver cell surface CD36, was shown to be protective against liver damage in people with NAFLD [38]. On the other hand, the missense mutation in the so-called gain-of-function PCSK9 variant (rs505151 E670 G) locating at the CHRD [37], probably increases CD36 binding to a great extent, leading to downregulation of CD36 and the consequent lipid profile abnormality [39]. In other words, let’s consider the possibility that the gain-of-function of E670 G actually targets at CD36 rather than LDL receptor. If this hypothesis was true, carriers of E670 G variant would have lower incidence of NAFLD. All these speculations, nevertheless, await evidence support from further laboratory works and epidemiologic studies.

Just like its engagement in the pathogenesis of HCC, CD36 upregulation has been detected in other malignant tumors with fatty acids and a variety of other ligands as CD36 activators in accelerating tumor initiation, proliferation, invasion, metastasis, immunosuppression, chemoresistance, and radioresistance [40]. For instance, in acute myeloid leukemia, CD36-overexpressed leukemia stem cells were peculiarly capable of proliferation [41], which was probably stimulated by apolipoprotein C2 through CD36-mediated LYN– ERK signaling activation [42]. In gastric cancer cell lines, higher CD36 expression led to more aggressive characteristics of migration, invasion, epithelial-mesenchymal transition, and fatty acid-mediated metastasis through the AKT/GSK-3β/β-catenin signaling pathway [43, 44]. Similar results of enhanced epithelial-mesenchymal transition and migration potential generated by fatty acids from cancer-associated fibroblasts via CD36 were seen in mouse models of colorectal cancer [45], and a conceivable mechanism was disclosed to be upregulation of the metallopeptidase MMP28 tightly connected to the overexpression of CD36 in colorectal cancer cells [46]. In breast cancer, CD36-mediated fatty acid uptake not only could potentiate invasiveness of cancer cells [47], but also foster the resistance to HER2-targeted therapies [48]. As a consequence, CD36 has been classified as a prospective therapeutic target for various types of cancer [49].

When taken together, the risk of developing NAFLD, HCC, and a lot of other malignant tumors on long-term eradication of PCSK9 with ensuing CD36 overexpression looks not an exaggerated warning. Although no significant difference of adverse effects was observed between siRNA targeting PCSK9 (inclisiran) and placebo in three major phase 3 clinical trials for management of familial hypercholesterolemia, the observation time is up to 540 days only [50], which is much shorter than what is presumed for a carcinogenic process. Even more worrisome is a recent proposal that people without familial hypercholesterolemia could start on lipid-lowering therapy plus inclisiran as well at thirty years of age, to reduce the cumulative LDL and atherosclerotic plaque burden afterwards [51]. In my opinion, the danger of developing NAFLD, HCC, and many other cancers on totally obliterating PCSK9 should firstly be excluded by regularly scheduled laboratory and image studies during follow-up in carefully designed clinical trials lasting for at least ten years before raising such an assertion, especially when more novel gene silencing therapies targeting PCSK9 are coming [52].

Finally, the atypical antipsychotic drug clozapine had been revealed to be capable of increasing CD36 expression in macrophages through a NADPH oxidase-reactive oxygen species-peroxisome proliferator-activated receptor γ pathway [53]. Coincidentally, this drug has been disclosed to be able to worsen glucose intolerance and NAFLD in obese mice [54], with the fact that schizophrenia patients on clozapine have a remarkable 43.2% prevalence of metabolic syndrome, of which NAFLD is a component [55]. Additionally, in two large epidemiologic researches, clozapine has recently found to be significantly, in a dose-response manner associated with hematological malignancies, that is, lymphoma and leukemia, as been compared with non-use of clozapine [56, 57]. These astonishing findings remind us again of the possible adverse effects of CD36 overexpression resulting from genetic abolishing of PCSK9. Thus, it is ardently hoped that more intensive investigations could be performed to study the safety of siRNA and other gene silencing therapies targeting PCSK9 in the near future.

Acknowledgments

None. The author received no financial support for the research, authorship, and/or publication of this article.

Conflict of interest

The author declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

References

[1] 

Seval GC , Kabacam G , Yakut M , Seven G , Savas B , Elhan A , et al. The natural course of non-alcoholic fatty liver disease. Hepatol Forum. (2020) ;1: (1):20–4. doi: 10.14744/hf.2020.0008.

[2] 

Petrelli F , Manara M , Colombo S , De Santi G , Ghidini M , Mariani M , et al. Hepatocellular carcinoma in patients with nonalcoholic fatty liver disease: A systematic review and meta-analysis: HCC and Steatosis or Steatohepatitis. Neoplasia (2022) ;30: :100809. doi: 10.1016/j.neo.2022.100809.

[3] 

Stine JG , Wentworth BJ , Zimmet A , Rinella ME , Loomba R , Caldwell SH , et al. Systematic review with meta-analysis: risk of hepatocellular carcinoma in non-alcoholic steatohepatitis without cirrhosis compared to other liver diseases. Aliment Pharmacol Ther. (2018) ;48: (48):696–703. doi: 10.1111/apt.14937.

[4] 

Massoud O , Charlton M . Nonalcoholic Fatty Liver Disease/Nonalcoholic Steatohepatitis and Hepatocellular Carcinoma. Clin Liver Dis. (2018) ;22: (22):201–11. doi: 10.1016/j.cld.2017.08.014.

[5] 

Salloum S , Jeyarajan AJ , Kruger AJ , Holmes JA , Shao T , Sojoodi M , et al. Fatty Acids Activate the Transcriptional Coactivator YAP1 to Promote Liver Fibrosis via p38 Mitogen-Activated Protein Kinase. Cell Mol Gastroenterol Hepatol. (2021) ;12: (4):1297–310. doi: 10.1016/j.jcmgh.2021.06.003.

[6] 

Glatz JFC , Nabben M , Luiken JJFP . CD36 (SR-B2) as master regulator of cellular fatty acid homeostasis. Curr Opin Lipidol. (2022) ;33: (33):103–11. doi: 10.1097/MOL.0000000000000819.

[7] 

Zaidi NE , Shazali NAH , Leow TC , Osman MA , Ibrahim K , Cheng WH , et al. CD36-Fatty Acid-Mediated Metastasis via the Bidirectional Interactions of Cancer Cells and Macrophages. Cells (2022) ;11: (22):3556. doi: 10.3390/cells11223556.

[8] 

Glatz JFC , Luiken JJFP . Time for a détente in the war on the mechanism of cellular fatty acid uptake. J Lipid Res. (2020) ;61: (9):1300–1303. doi: 10.1194/jlr.6192020LTE.

[9] 

Alves-Bezerra M , Cohen DE . Triglyceride Metabolism in the Liver. Compr Physiol. (2017) ;8: (8):1–8. doi: 10.1002/cphy.c170012.

[10] 

Shen C , Chen JH , Oh HR , Park JH . Transcription factor SOX2 contributes to nonalcoholic fatty liver disease development by regulating the expression of the fatty acid transporter CD36. FEBS Lett. (2021) ;595: (19):2493–503. doi: 10.1002/1873-3468.14193.

[11] 

Liu J , Yang P , Zuo G , He S , Tan W , Zhang X , et al. Long-chain fatty acid activates hepatocytes through CD36 mediated oxidative stress. Lipids Health Dis. (2018) ;17: (1):153. doi: 10.1186/s12944-018-0790-9.

[12] 

Rada P , González-Rodríguez Á , García-Monzón C , Valverde ÁM . Understanding lipotoxicity in NAFLD pathogenesis: is CD36 a key driver? Cell Death Dis. (2020) ;11: (9):802. doi: 10.1038/s41419-020-03003-w.

[13] 

Luo X , Zheng E , Wei L , Zeng H , Qin H , Zhang X , et al. The fatty acid receptor CD36 promotes HCC progression through activating Src/PI3K/AKT axis-dependent aerobic glycolysis. Cell Death Dis. (2021) ;12: (4):328. doi: 10.1038/s41419-021-03596-w.

[14] 

Tao L , Ding X , Yan L , Xu G , Zhang P , Ji A , et al. CD36 accelerates the progression of hepatocellular carcinoma by promoting FAs absorption. Med Oncol. (2022) ;39: (12):202. doi: 10.1007/s12032-022-01808-7.

[15] 

Tian Y , Yang B , Qiu W , Hao Y , Zhang Z , Yang B , et al. ER-residential Nogo-B accelerates NAFLD-associated HCC mediated by metabolic reprogramming of oxLDL lipophagy. Nat Commun (2019) ;10: (1):3391. doi: 10.1038/s41467-019-11274-x.

[16] 

Nath A , Li I , Roberts LR , Chan C . Elevated free fatty acid uptake via CD36 promotes epithelial-mesenchymal transition in hepatocellular carcinoma. Sci Rep. (2015) ;5: :14752. doi: 10.1038/srep14752.

[17] 

Li Q , Wang C , Wang Y , Sun L , Liu Z , Wang L , et al. HSCs-derived COMP drives hepatocellular carcinoma progression by activating MEK/ERK and PI3K/AKT signaling pathways. J Exp Clin Cancer Res. (2018) ;37: (1):231. doi: 10.1186/s13046-018-0908-y.

[18] 

Du D , Liu C , Qin M , Zhang X , Xi T , Yuan S , et al. Metabolic dysregulation and emerging therapeutical targets for hepatocellular carcinoma. Acta Pharm Sin B. (2022) ;12: (12):558–80. doi: 10.1016/j.apsb.2021.09.019.

[19] 

Duan Y , Gong K , Xu S , Zhang F , Meng X , Han J . Regulation of cholesterol homeostasis in health and diseases: from mechanisms to targeted therapeutics. Signal Transduct Target Ther. (2022) ;7: (1):265. doi: 10.1038/s41392-022-01125-5.

[20] 

Maligłówka M , Kosowski M , Hachuła M , Cyrnek M , Bułdak Ł , Basiak M , et al. Insight into the Evolving Role of PCSK9. Metabolites. (2022) ;12: (3):256. doi: 10.3390/metabo12030256.

[21] 

Liu C , Chen J , Chen H , Zhang T , He D , Luo Q , et al. PCSK9 Inhibition: From Current Advances to Evolving Future. Cells (2022) ;11: (19):2972. doi: 10.3390/cells11192972.

[22] 

Seidah NG , Prat A . The multifaceted biology of PCSK9. Endocr Rev. (2021) ;43: (43):558–82. doi: 10.1210/endrev/bnab035.

[23] 

Huang YT , Ho LT , Hsu HY , Tu YK , Chien KL . Efficacy and Safety of Proprotein Convertase Subtilisin/Kexin Type 9 Inhibitors as Adjuvant Treatments for Patients with Hypercholesterolemia Treated with Statin: A Systematic Review and Network Meta-analysis. Front Pharmacol. (2022) ;13: :832614. doi: 10.3389/fphar.2022.832614

[24] 

Toth PP , Bray S , Villa G , Palagashvili T , Sattar N , Stroes ESG , et al. Network Meta-Analysis of Randomized Trials Evaluating the Comparative Efficacy of Lipid-Lowering Therapies Added to Maximally Tolerated Statins for the Reduction of Low-Density Lipoprotein Cholesterol. J Am Heart Assoc. (2022) ;11: (18):e025551. doi: 10.1161/JAHA.122.025551.

[25] 

Coppinger C , Movahed MR , Azemawah V , Peyton L , Gregory J , Hashemzadeh M . A Comprehensive Review of PCSK9 Inhibitors. J Cardiovasc Pharmacol Ther. (2022) ;27: :10742484221100107. doi: 10.1177/10742484221100107.

[26] 

Demers A , Samami S , Lauzier B , Des Rosiers C , Ngo Sock ET , Ong H , et al. 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: (12):2517–25. doi: 10.1161/ATVBAHA.115.306032.

[27] 

Seidah NG , Garçon D . Expanding Biology of PCSK Roles in Atherosclerosis and Beyond. Curr Atheroscler Rep. (2022) ;24: (10):821–30. doi: 10.1007/s11883-022-01057-z.

[28] 

Rogula S , Błażejowska E , Gąsecka A , Szarpak Ł , Jaguszewski MJ , Mazurek T , et al, Inclisiran-Silencing the Cholesterol, Speaking up the Prognosis. J Clin Med (2021) ;10: (11):2467. doi: 10.3390/jcm10112467.

[29] 

Khan SA , Naz A , Qamar Masood M , Shah R . Meta-Analysis of Inclisiran for the Treatment of Hypercholesterolemia. Am J Cardiol. (2020) ;134: :69–73. 10.1016/j.amjcard.2020.08.018.

[30] 

Wright RS , Ray KK , Raal FJ , Kallend DG , Jaros M , Koenig W , et al. Pooled Patient-Level Analysis of Inclisiran Trials in Patients With Familial Hypercholesterolemia or Atherosclerosis. J Am Coll Cardiol. (2021) ;77: (9):1182–93. doi: 10.1016/j.jacc.2020.12.058.

[31] 

Lebeau PF , Byun JH , Platko K , Al-Hashimi AA , Lhoták Š , MacDonald ME , et al. Pcsk9 knockout exacerbates diet-induced non-alcoholic steatohepatitis, fibrosis and liver injury in mice. JHEP Rep. (2019) ;1: (6):418–29. doi: 10.1016/j.jhepr.2019.10.009.

[32] 

Ioannou GN , Lee SP , Linsley PS , Gersuk V , Yeh MM , Chen YY , et al. Pcsk9 Deletion Promotes Murine Nonalcoholic Steatohepatitis and Hepatic Carcinogenesis: Role of Cholesterol. Hepatol Commun. (2022) ;6: (6):780–94. doi: 10.1002/hep4.1858.

[33] 

Langsted A , Nordestgaard BG , Benn M , Tybjærg-Hansen A , Kamstrup PR . PCSK9 R46L Loss-of-Function Mutation Reduces Lipoprotein(a), LDL Cholesterol, and Risk of Aortic Valve Stenosis. J Clin Endocrinol Metab. (2016) ;101: (9):3281–7. doi: 10.1210/jc.2016-1206.

[34] 

Kent ST , Rosenson RS , Avery CL , Chen YI , Correa A , Cummings SR , et al. PCSK9 Loss-of-Function Variants, Low-Density Lipoprotein Cholesterol, and Risk of Coronary Heart Disease and Stroke: Data From 9 Studies of Blacks and Whites. Circ Cardiovasc Genet. (2017) ;10: (4):e001632. doi: 10.1161/CIRCGENETICS.116.001632.

[35] 

Baragetti A , Balzarotti G , Grigore L , Pellegatta F , Guerrini U , Pisano G , et al. PCSK9 deficiency results in increased ectopic fat accumulation in experimental models and in humans. Eur J Prev Cardiol. (2017) ;24: (17):1870–77. doi: 10.1177/2047487317724342.

[36] 

Rimbert A , Smati S , Dijk W , Le May C , Cariou B . Genetic Inhibition of PCSK9 and Liver Function. JAMA Cardiol. (2021) ;6: (6):353–4. doi: 10.1001/jamacardio.2020.5341.

[37] 

Lebeau PF , Platko K , Byun JH , Makda Y , Austin RC . The Emerging Roles of Intracellular PCSK9 and Their Implications in Endoplasmic Reticulum Stress and Metabolic Diseases. Metabolites. (2022) ;12: (3):215. doi: 10.3390/metabo12030215.

[38] 

Grimaudo S , Bartesaghi S , Rametta R , Marra F , Margherita Mancina R , Pihlajamäki J , et al. PCSK9 rsR46L loss-of-function variant protects against liver damage in individuals with NAFLD. Liver Int. (2021) ;41: (41):321–32. doi: 10.1111/liv.14711.

[39] 

Qiu C , Zeng P , Li X , Zhang Z , Pan B , Peng ZYF , et al. What is the impact of PCSK9 rs51 and rspolymorphisms on serum lipids level and cardiovascular risk: a meta-analysis. Lipids Health Dis. (2017) ;16: (1):111. doi: 10.1186/s12944-017-0506-6.

[40] 

Wang J , Li Y . CD36 tango in cancer: signaling pathways and functions. Theranostics. (2019) ;9: (17):4893–908. doi: 10.7150/thno.36037.

[41] 

Sachs K , Sarver AL , Noble-Orcutt KE , LaRue RS , Antony ML , Chang D , et al. Single-Cell Gene Expression Analyses Reveal Distinct Self-Renewing and Proliferating Subsets in the Leukemia Stem Cell Compartment in Acute Myeloid Leukemia. Cancer Res. (2020) ;80: (80):458–70. doi: 10.1158/0008-5472.CAN-18-2932.

[42] 

Zhang T , Yang J , Vaikari VP , Beckford JS , Wu S , Akhtari M , et al. Apolipoprotein C2 - CD36 Promotes Leukemia Growth and Presents a Targetable Axis in Acute Myeloid Leukemia. Blood Cancer Discov. (2020) ;1: (1):198–213. doi: 10.1158/2643-3230.BCD-19-0077.

[43] 

Wang J , Wen T , Li Z , Che X , Gong L , Jiao Z , et al. CD36 upregulates DEK transcription and promotes cell migration and invasion via GSK-3β/β-catenin-mediated epithelial-to-mesenchymal transition in gastric cancer. Aging (Albany NY). (2020) ;13: (2):1883–97. doi: 10.18632/aging.103985.

[44] 

Pan J , Fan Z , Wang Z , Dai Q , Xiang Z , Yuan F , et al. CD36 mediates palmitate acid-induced metastasis of gastric cancer via AKT/GSK-3β/β-catenin pathway. J Exp Clin Cancer Res. (2019) ;38: (1):52. doi: 10.1186/s13046-019-1049-7.

[45] 

Gong J , Lin Y , Zhang H , Liu C , Cheng Z , Yang X , et al. Reprogramming of lipid metabolism in cancer-associated fibroblasts potentiates migration of colorectal cancer cells. Cell Death Dis. (2020) ;11: (4):267. doi: 10.1038/s41419-020-2434-z.

[46] 

Drury J , Rychahou PG , Kelson CO , Geisen ME , Wu Y , He D , et al. Upregulation of CD36, a Fatty Acid Translocase, Promotes Colorectal Cancer Metastasis by Increasing MMP28 and Decreasing E-Cadherin Expression. Cancers (Basel). (2022) ;14: (1):252. doi: 10.3390/cancers14010252.

[47] 

Zaoui M , Morel M , Ferrand N , Fellahi S , Bastard JP , Lamazière A , et al. Breast-Associated Adipocytes Secretome Induce Fatty Acid Uptake and Invasiveness in Breast Cancer Cells via CD36 Independently of Body Mass Index, Menopausal Status and Mammary Density. Cancers (Basel) (2019) ;11: (12):2012. doi: 10.3390/cancers11122012.

[48] 

Feng WW , Wilkins O , Bang S , Ung M , Li J , An J , et al. CD36-Mediated Metabolic Rewiring of Breast Cancer Cells Promotes Resistance to HER2-Targeted Therapies. Cell Rep. (2019) ;29: (11):3405–20.e5. doi: 10.1016/j.celre2019.11.008.

[49] 

Ruan C , Meng Y , Song H . CD an emerging therapeutic target for cancer and its molecular mechanisms. J Cancer Res Clin Oncol. (2022) ;148: (7):1551–8. doi: 10.1007/s00432-022-03957-8.

[50] 

Sinning D , Landmesser U . Low-density Lipoprotein-Cholesterol Lowering Strategies for Prevention of Atherosclerotic Cardiovascular Disease: Focus on siRNA Treatment Targeting PCSK9 (Inclisiran). Curr Cardiol Rep. (2020) ;22: (12):176. doi: 10.1007/s11886-020-01427-6.

[51] 

Pirillo A , Catapano AL . Inclisiran: How Widely and When Should We Use It? Curr Atheroscler Rep. (2022) ;24: (10):803–11. doi: 10.1007/s11883-022-01056-0.

[52] 

Katzmann JL , Cupido AJ , Laufs U . Gene Therapy Targeting PCSK9. Metabolites. (2022) ;12: (1):70. doi: 10.3390/metabo12010070.

[53] 

Chen CH , Leu SJ , Hsu CP , Pan CC , Shyue SK , Lee TS . Atypical antipsychotic drugs deregulate the cholesterol metabolism of macrophage-foam cells by activating NOX-ROS-PPARγ-CD36 signaling pathway. Metabolism. (2021) ;123: :154847. doi: 10.1016/j.metabol.2021.154847.

[54] 

Chang GR , Liu HY , Yang WC , Wang CM , Wu CF , Lin JW , et al. Clozapine Worsens Glucose Intolerance, Nonalcoholic Fatty Liver Disease, Kidney Damage, and Retinal Injury and Increases Renal Reactive Oxygen Species Production and Chromium Loss in Obese Mice. Int J Mol Sci (2021) ;22: (13):6680. doi: 10.3390/ijms22136680.

[55] 

Zhang Y , Chen M , Chen J , Wu Z , Yu S , Fang Y , et al. Metabolic syndrome in patients taking clozapine: prevalence and influence of catechol-O-methyltransferase genotype. Psychopharmacology (Berl). (2014) ;231: (10):2211–8. doi: 10.1007/s00213-013-3410-4.

[56] 

Chrétien B , Lelong-Boulouard V , Chantepie S , Sassier M , Bertho M , Brazo P , et al. Haematologic malignancies associated with clozapine v. all other antipsychotic agents: a pharmacovigilance study in VigiBase®. Psychol Med. (2021) ;51: (9):1459–66. doi: 10.1017/S0033291720000161.

[57] 

Tiihonen J , Tanskanen A , Bell JS , Dawson JL , Kataja V , Taipale H . Long-term treatment with clozapine and other antipsychotic drugs and the risk of haematological malignancies in people with schizophrenia: a nationwide case-control and cohort study in Finland. Lancet Psychiatry. (2022) ;9: (9):353–62. doi: 10.1016/S2215-0366(22)00044-X.