Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Oct 19.
Published in final edited form as: Curr Opin Lipidol. 2020 Apr;31(2):71–79. doi: 10.1097/MOL.0000000000000673

NOVEL LCAT-BASED THERAPEUTIC APPROACHES

Lita A Freeman 1,+, Sotirios Karathanasis 1,2,+, Alan T Remaley 1,*
PMCID: PMC8525913  NIHMSID: NIHMS1743769  PMID: 32073411

Abstract

Purpose of review:

To review recent LCAT-based therapeutic approaches for atherosclerosis, acute coronary syndrome, and LCAT deficiency disorders.

Recent findings:

A wide variety of approaches to using LCAT as a novel therapeutic target have been proposed. Enzyme replacement therapy with recombinant human LCAT (rhLCAT) is the most clinically advanced therapy for atherosclerosis and familial LCAT deficiency (FLD), with Phase I and Phase 2A clinical trials recently completed. Liver-directed LCAT gene therapy and engineered cell therapies are also another promising approach. Peptide and small molecule activators have shown efficacy in early stage preclinical studies. Finally, lifestyle modifications, such as fat-restricted diets, cessation of cigarette smoking, and a diet rich in antioxidants may potentially suppress lipoprotein abnormalities in FLD patients and help preserve LCAT activity and renal function but have not been adequately tested.

Summary:

Preclinical and early-stage clinical trials demonstrate the promise of novel LCAT therapies as HDL-raising agents that may be used to treat not only FLD but potentially also atherosclerosis and other disorders with low or dysfunctional HDL.

Keywords: LCAT, HDL, atherosclerosis, familial LCAT deficiency, LP-X

INTRODUCTION

Low levels of high-density lipoprotein-cholesterol (HDL-C) in plasma are strongly inversely related to cardiovascular disease (CVD) risk. However, most therapies targeted at raising plasma HDL-C have not been successful to date in reducing CVD. Recent efforts have focused on not just raising HDL-C but on improving HDL function, in particular reverse cholesterol transport (RCT) (1) but also other functions as well (2). One such novel approach for improving HDL function involves agents that increase the activity of Lecithin:Cholesterol Acyltransferase (LCAT).

LCAT is a plasma enzyme produced by the liver that plays a key role in HDL metabolism (3). It mediates transfer of a fatty acid from the sn-2 position of phospholipids (PL) to free cholesterol (FC) to form cholesteryl ester (CE) and lysolecithin (3) (4) (5). This reaction takes place mainly on HDL and is an essential step in HDL maturation from small discoidal particles to mature spherical HDL particles. Importantly, this is thought to be a key step in RCT (3). Because of its ability to increase HDL-C and promote RCT, LCAT has long been considered as an attractive target for the development of potential therapeutics against atherosclerosis.

LCAT is defective in Familial LCAT Deficiency (FLD), an autosomal recessive disorder characterized by extremely low levels of plasma HDL-C, low LDL-C and plasma cholesteryl esters, corneal opacities, anemia, and proteinuria. The proteinuria typically progresses to nephrotic syndrome and end-stage renal failure usually by the fourth or fifth decade of life and is the main cause of morbidity and mortality in these patients. Due to mutations in specific locations on LCAT (6), FLD patients cannot esterify cholesterol on either HDL or apoB-containing lipoproteins (apoB-Lps). This results in the accumulation in plasma of unusual vesicle-like structures, termed LP-X, that are highly enriched in FC and PL. A less severe form of FLD is Fish-Eye Disease (FED). FED patients do not suffer from kidney disease but are at increased risk of CVD compared to FLD patients (79). They have mutations in LCAT that retain activity towards beta lipoproteins and, consequently, have low HDL-C but near-normal levels of LDL-C, likely contributing to their increased CVD risk. They also have very low or undetectable plasma LP-X levels, which may explain why they typically do not develop proteinuria and kidney disease (10, 11).

Early studies indicated that LP-X may be nephrotoxic (10) (12), providing a plausible mechanism for the increased lipid deposition observed in mesangial cells in kidneys of FLD patients (3). Recent work from our laboratory has directly demonstrated that LCAT reduces plasma and tissue accumulation of LP-X, protecting against kidney disease in mouse models of FLD nephropathy (13, 14). LP-X levels have also been shown to associate with renal disease in humans (11, 15).

These observations suggest that therapies that increase LCAT or LCAT activity will be effective not only for the treatment of kidney disease in FLD patients but also for the treatment of CVD in FED patients and possibly in patients with increased CVD risk for secondary prevention.

In this review we discuss the various approaches for LCAT therapies, including protein replacement biologics, gene and cell therapies, and peptide and small molecule LCAT activators (summarized in Table I). In particular, protein replacement therapy has recently entered clinical evaluation (1618) and the data are likely to be relevant for not only LCAT therapeutics but the broader area of HDL therapies.

Table 1:

Indications for LCAT activators

Disorder Mutation Type
Or Defect
rhLCAT, WT rhLCAT, modified Activating Antibody Gene
Therapy
Peptides Small Molecules
FLD Active Site + + +
FLD Major Truncation + + +
FLD Small Truncation + + −/+ + −/+ −/+
FLD Secretion
defects
+ + +
FLD Stability/
Structure
+ + −/+ + −/+ −/+
FLD Partial loss of activity + + −/+ + −/+ −/+
Cholestasis All types + + + + + +
Atherosclerosis Low HDL + + + + + +
ACS Endothelial NO production + + + + + +
Oxidative stress disorders Oxidative inactivation of endogenous LCAT + + + + + +

LCAT enzyme replacement therapy and gene therapy would not require the presence of functional LCAT. Activating antibodies, peptides and small molecule activators of LCAT require expression of LCAT protein, either wild-type or with a mutation permissive for enzyme activation by the activating agent.

BIOLOGICS

Several LCAT-raising biologic approaches have been developed or are currently under development as potential therapeutics for atherosclerosis, acute coronary syndrome (ACS), FLD, FED, and cholestasis.

rhLCAT as a therapeutic for atherosclerosis and ACS

Recombinant human LCAT (rhLCAT) for enzyme replacement therapy was first developed for clinical testing by AlphaCore Pharmaceuticals. In a first-in-human randomized, blinded, placebo-controlled, dose-escalation Phase I trial in subjects with stable atherosclerosis, this early formulation of rhLCAT, ACP501, raised HDL-C by as much as 50%, improved cholesterol efflux, and was found to be safe and well-tolerated (19). MedImmune developed a newer and more potent formulation of rhLCAT, MEDI6012, which was also found to raise HDL-C in stable atherosclerosis patients for at least a week after a single infusion. A Phase I and Phase 2a randomized, blinded, placebo-controlled, dose-escalation study of intravenous (IV) trials with MEDI6012 in stable atherosclerosis patients showed that MEDI6012 was safe, well-tolerated and also increased HDL-C, HDL-CE, and, interestingly, decreased apoB and small LDL particles in human subjects (1618).

Preclinical work using wild-type (WT) and LCAT-knockout (KO) mice showed that treatment with MEDI6012 dramatically increased in-vivo macrophage cholesterol efflux in LCAT-KO mice (20). More recent findings suggest that in addition to improving RCT, LCAT treatment may also improve HDL functionality. Specifically, HDL from myocardial infarction (MI) patients is impaired in its ability to stimulate NO production from endothelial cells and this impairment is reversed in HDL isolated from MI patient plasma incubated with LCAT ex-vivo (21). The ability of rhLCAT to restore HDL-mediated NO production suggests that rhLCAT may be a potential therapeutic target for restoring HDL function in not only ACS but in other cardiovascular diseases as well.

rhLCAT as a therapeutic for FLD

Preclinical studies in mice demonstrated that ACP501 injection restored a normal lipid phenotype in LCAT-KO mice and also increased cholesterol efflux (22). FLD patients have very low HDL, and it is conceivable that reduced HDL may drive kidney diseases in these patients. However, other conditions with very low HDL levels, such as FED, Tangier disease and apoA-I deficiency, also have very low HDL, yet patients do not develop kidney disease. These findings indirectly suggest that LP-X is the causative factor in FLD nephropathy. Consistent with this, we recently demonstrated that infusing LP-X into LCAT-deficient mice causes renal disease; importantly, the presence of LCAT in WT mice prevents kidney damage from injected LP-X (13). Moreover, in SREBP-Tg x LCAT-KO mice, which produce abundant quantities of LP-X on a protein-rich/carbohydrate-low (PRCL) diet, rhLCAT treatment decreases LP-X and protects against renal disease (14). Finally, a very recent publication documented that LP-X plasma levels, as determined by a new LP-X quantification assay (11), is in fact a major discriminator of the presence or absence of renal disease among two patients carrying identical LCAT mutations (15).

In a single FLD patient tested, ACP-501 treatment rapidly increased plasma HDL-C (23), with rapid disappearance of the small discoidal preβ-HDL and α-4 particles and subsequent appearance of large spherical α-HDL particles. LDL-C also increased but more slowly than HDL-C, likely due to transfer of newly formed CE to LDL by CETP. Despite the patient’s advanced renal disease, anemia improved significantly and renal function parameters remained stable or slightly improved (23). Like the Phase I study, ACP501 was safe and well-tolerated in the one FLD patient given the therapy, who was treated once or twice a week for approximately 7 months.

The data so far indicating LP-X as a causative factor in FLD nephropathy and the availability of a recently published standardized LP-X quantification assay (11) should expedite clinical development of such protein replacement therapies for FLD and possibly other disorders with highly elevated plasma LP-X.

Cholestasis

As mentioned above, patients with primary biliary cholestasis (PBC) also have LP-X in their plasma. In fact, the most common cause of increased LP-X is liver disease and in particular the type of liver disease that leads to cholestasis. Even though the LP-X in cholestatic patients appears to be of hepatic origin, in contrast to FLD where LP-X is formed in the circulation (24), recent data in our laboratory show that LCAT overexpression or MEDI6012 treatment dramatically suppresses plasma LP-X levels in a model of chemically induced cholestasis in mice (25). Although LP-X does not seem to be causative for liver damage in PBC, it was thought to be responsible for xanthoma formation in these patients (26). Whether LP-X has other negative consequences like renal disease in cholestatic patients is not known, but it can reach such high levels in these patients to cause hyperviscosity syndrome requiring plasmapheresis (27). Therefore, LCAT protein therapy may also be useful for cholestatic liver disease.

Modified rhLCAT therapeutics

As noted below, LCAT Cys31Tyr substitution, like other Cys31 modifications with large bulky hydrophobic groups, increases LCAT activity by 10-fold (28). LCAT Cys31Tyr is a natural rare mutation in humans (29). Moreover, injecting LCAT Cys31Tyr into rabbits resulted in large HDL particles, increased cholesterol efflux from peripheral tissues into plasma, increased neutral sterol excretion into feces, and decreased progression of atherosclerosis (30). Other modifications of LCAT may increase LCAT stability and plasma half-life (31). This modified, more active recombinant LCAT holds promise to deliver LCAT as a more potent enzyme replacement therapy and has been patented by Amgen (3133).

Finally, single administration of 27C3, an antibody against LCAT Cys31Tyr, into cynomolgus monkeys rapidly increased plasma LCAT enzymatic activity and led to a 35% increase in HDL-C that persisted up to 32 days. This class of antibodies that appears to stabilize LCAT represents another novel biological approach for the treatment of dyslipidemia and cardiovascular disease (28).

GENE AND CELL THERAPY

Although gene therapy for the treatment of human genetic diseases was first conceptualized in the 1970s, it is only recently that gene therapies have started to emerge as real therapies in humans. Following the early failure of gene therapy trials with retroviral vectors and highly immunogenic adenovirus vectors in the late 1990s and early 2000s, huge gaps have been closed concerning virus biology, vector dynamics, immune interaction, and vector safety with the development of a class of viral vectors called adeno-associated viruses (AAVs) as a leading platform for new gene therapies (34).

The use of recombinant viruses expressing LCAT for gene therapy for “treating or preventing dyslipoproteinaemia-related diseases” was first proposed in 2001 (35). Santamarina, Hoeg and Brewer subsequently patented methods to increase LCAT activity in plasma by either cell transfection with vectors containing LCAT, by upregulating LCAT gene expression pharmaceutically, and/or by administering LCAT itself, to prevent or treat atherosclerosis or to treat FLD or FED (36). An early study using adenovirus encoding hLCAT in non-human primates showed a sustained elevation of HDL-C after a single treatment (37).

The development of AAV vectors with tissue tropism, for example AAV8, which preferentially infects hepatocytes (38), has reinvigorated the idea of developing AAV LCAT gene delivery therapeutics for the treatment of FLD. Early optimization of such vectors regarding the use of naturally occurring LCAT variants with increased activity, codon optimization, and effective dose determination in rodent FLD models have been recently described (38, 39).

Despite these promising developments for an effective LCAT gene therapy, there are issues that need to be addressed before such therapy becomes broadly available. These issues include transfection efficiency for adequate LCAT production to sustain physiologic LCAT plasma levels, pre-existing anti-viral vector neutralizing antibodies, and immunogenicity, especially with high viral infection loads. Nevertheless, it is encouraging that clinical investigators have taken up these challenges proposing to optimize such therapies in humans (see for example: http://grantome.com/grant/NIH/P01-HL059407-16A1-8339).

Given the potential limitations with direct gene therapy, indirect gene delivery has also been considered. The basic concept is to isolate adipose tissue cells from patients, transfect them with an LCAT expressing vector, and then after in vitro expansion, return (implant) these LCAT-producing cells to the same patient (40). Although this is conceptually attractive, particularly since it is autologous cell transplantation which avoids immunogenic issues, it also has limitations. First, re-integration of the cells in the tissue and long-term survival may not be optimal, as implied by efforts to develop biomaterials (Fibrin glue) to support survival of these cells and optimal LCAT production after cell implantation (41). Second, the efficiency of LCAT transport from the implanted tissue to the circulation and the number of implanted cells will determine whether physiological LCAT plasma levels can be achieved. Finally, this is an autologous cell transplantation and, therefore, it represents a procedure rather than a drug that can be used to treat all patients. Despite these limitations, the concept of genetically engineered cell therapy for FLD is a valid approach, especially if one can develop an efficient genetically engineered hematopoietic stem cell that can engraft and proliferate leading to sustained production of LCAT (see for example reference (42)).

PEPTIDE ACTIVATORS

LCAT has very low intrinsic activity and requires apolipoproteins for full activity. The major activator of LCAT on HDL is apoA-I (43). ApoA-IV, apoC-I, and apoE, but not apoA-II, apoC-II or apoC-III, can also significantly activate LCAT (43). LCAT does have some activity on apoB-containing lipoprotein particles (LCAT beta activity) and apoE is the major physiological activator of LCAT on these lipoproteins (44). In the brain, nascent apoE-containing lipoproteins are secreted from glial cells and apoE activates LCAT on these lipoprotein particles (45).

Based on the above findings, synthetic peptides that activate LCAT were developed (Fig. 1). All of these peptides contain an amphipathic helix and are thus like apoA-I mimetic peptides, which are also being developed into a form of HDL therapy (46). One of the first such synthetic “peptides” to activate LCAT was full-length, mature apoC-I (47). Truncated versions of apoC-I showed 50–100% activation compared to full-length apoC-I (48) (Fig. 1A). Figure 1B shows other mimetic peptides, based mostly on apoA-I, that can activate LCAT (49) (50) (51, 52) (53) (54) (Table 2). The peptide farthest along in clinical development is ESP24218 (aka P-642), a 22-aa apoA-I mimetic peptide designed by Esperion Therapeutics (55) (56). P-642 complexed with phospholipids is 1.25X more efficient in activating LCAT than proteoliposomes made with human apoA-I (57). Phase I studies were completed but development of the peptide was discontinued (56). Finally, an apoA-I polypeptide or mimetic peptide that activates LCAT fused to an antibody Fc region to enhance plasma half-life has been described (58).

FIGURE 1: Synthetic peptide activators of LCAT.

FIGURE 1:

FIGURE 1:

(A) ApoC-I-based LCAT activators. The full-length, mature apoC-I amino acid sequence (amino acids 1–57) is shown on the top line, followed by truncated versions of apoC-I. The activity of full-length apoC-I is defined as 100%. Note that peptide 32–57 encompasses one of the major phospholipid-binding domains of apoC-I. (B) ApoA-I mimetic peptide/amphipathic helix-based LCAT activators. A helical grid representation of the GALA peptide demonstrates stacking of glutamic acid residues (previously published, from Reference (52)). Activities relative to full-length apoA-I are shown in Table 2.

TABLE 2.

Percent activation of synthetic peptide LCAT activators relative to apoA-I

Percent activity
(compared to full-length apoA-I)
Peptide Egg PC (Vesicular) Egg PC (Discoidal) DMPC
ApoA-I 100 100 100*
LAP-20 50
AP 18
GALA 85
18A 18 30
37pA Up to 100** 30
[Glu1,8Leu11,17]18A 34 460
ESP24218 93 – 125
*

35% as active as apo A-l in egg PC vesicular assay system

**

Biphasic; low affinity compared to apoA-I (ref. 53).

Adapted from Reference (43)

SMALL MOLECULES

A small heterocyclic amine called Compound A ([3-(5-(ethylthio)-1,3,4-thiadiazol-2-ylthio)pyrazine-2-carbonitrile]) (Fig. 2A) (59) covalently binds Cys31 in LCAT (Fig. 2B, C) (60)) and increases plasma CE and HDL-C levels in mice and hamsters (59) (61). Interestingly, in vitro, Compound A was able to rescue some naturally occurring LCAT mutations that cause FLD. These mutations are not in the active site but rather promote either the binding or orientation of its lipid to enhance catalysis (60). Site-directed mutagenesis of Cys31 demonstrated that charged residues (Glu, Arg, and Lys) at amino acid #31 decreased LCAT activity, whereas bulky hydrophobic groups (Trp, Leu, Phe, and Met) at this position increased activity (60). Consistently, the wild-type chicken LCAT sequence contains a Phe in place of Cys at position 31 (62) and chickens have very high HDL (63). Modeling studies suggested that Cys31 alterations that activate LCAT all show increased movement of the membrane-binding domain. Movements of the lid and substrate binding region were also altered and correlated with changes in LCAT activity. Large hydrophobic adducts to Cys31 appear to activate LCAT by promoting either the binding of its hydrophobic lipid substrates and/or a more favorable orientation of the lipid substrate for catalysis (60). Our studies also revealed the importance of specific functional groups of Compound A for LCAT activation (Figure 2B, C) (60).

FIGURE 2.

FIGURE 2

FIGURE 2

(A) Compound A. (B,C) Compound A plus LCAT – 3D model (B) and functional groups of Compound A important for LCAT binding (C). The α,β-unsaturated system (cyanopyrazine ring) and the presence of Lewis bases (e.g., nitrogen atoms in the cyanopyrazine ring) in Compound A were important for its ability to activate LCAT. The S-ethyl group tail of compound A also appears to be necessary for appropriately situating the molecule via hydrogen-bonding toward the Cys31 for nucleophilic attack. From Reference (60), previously published. (D) Piperidinylpyrazolopyridine and related activators. Structure of compounds 1 (patent example 95, Ref. (65)), 2 (patent example 46, Ref. (65)), and 3 (patent example 3, Reference (68)) (From Figure 1a of Reference (69)). Altering functional groups on these compounds highlighted the importance of a hydroxyl group and chirality at C4 and the pyrazine ring structure and led to a novel derivative, Compound 8, with even greater ability to activate LCAT and rescue Arg244 mutations (8: Compound 8 from Figure 7 of Reference (69) [7-(trifluoromethyl)−1-(1-(5-(trifluoromethyl)pyrazin-2-yl)piperidin-4-yl)−1H-imidazo[4,5-b]pyridin-5(4H)-one]). (E) Structure of Daiichi compound 1 bound to LCAT (From Figure 2 of Reference (69), previously published).

A novel, orally available LCAT activator, DS-8190, that increased LCAT activity, promoted reverse cholesterol transport (RCT) and prevented atherosclerosis progression in mice was developed by Daiichi (64), as well as 4 other patented additional small-molecule LCAT activators (6568). Activator Compounds 1–3 (Figure 2D) activated LCAT but did not alter LCAT’s affinity for HDL (69). Structural studies of LCAT bound to Compound 1 indicated that it binds within the membrane-binding domain of LCAT and changes the conformation of the lid, likely stabilizing the membrane-binding domain and facilitating entry of the substrate into the active site (Figure 2D, E) (69). Most importantly, Compounds 1–3 were able to rescue acyltransferase activities of some naturally occurring mutations that cause FLD. Like Compound A, these would not be active site mutations but rather mutations that alter LCAT structure or stability.

LIFESTYLE MODIFICATIONS

Fat Restricted Diets

In contrast to PBC, where LP-X appears to originate from the liver, in FLD patients LP-X is formed in the circulation, most likely from surface components shed off from the lipolysis of triglyceride-rich lipoprotein metabolism (70). Therefore, reduction or limited availability of chylomicrons will be expected to reduce LP-X precursors and, therefore, LP-X plasma levels in FLD patients. Consistently, significant improvements in renal function and proteinuria following the adoption of a fat-restricted diet (70) along with administration of an angiotensin II receptor blocker were seen in a few patients on this diet (71). More recently, treatment of FLD patients with a combination of nicotinic acid and fenofibrate reduced LP-X plasma levels and urine albumin excretion (72). Based on these data, it is reasonable to suggest that newer, more effective triglyceride reducing agents may potentially also possibly be effective in suppressing LP-X plasma levels and kidney dysfunction in FLD patients but this remains to be tested in clinical trials.

Cigarettes and oxidative stress

Cigarette smoke reduces LCAT activity (i.e. acquired LCAT deficiency) by aldehyde-induced modification of its free thiols (73) (74, 75) (76). Oxidative stress also reduces LCAT activity (77). Cessation of smoking and a diet rich in antioxidants are lifestyle modifications anticipated to preserve LCAT activity and raise HDL. LCAT modifications to resist such oxidative stress-induced damage may also be of value in raising HDL in diseases with increased oxidative stress, such as hemolytic anemia in sickle cell disease (78) or psoriasis (79) (80).

CONCLUSION

LCAT-based therapeutics have come a long way in the past few years. Phase I and Phase 2A rhLCAT enzyme replacement therapy trials for atherosclerosis have been completed and have shown safety and tolerability as well as an improved lipoprotein profile. A first-in-human trial demonstrated that rhLCAT raised HDL and stabilized or improved renal function parameters in a single FLD patient. Gene therapy trials are just over the horizon and peptide and small molecule LCAT activators show promise as well. Lifestyle modifications, such as fat-restricted diets for FLD patients, has shown positive results and cessation of cigarette smoking, which is recommended in any case, has the added benefit of preserving LCAT functionality. LCAT-based therapeutics may be of general utility for raising HDL-C and improving HDL functionality in a number of disorders. There is a critical need for LCAT-based therapeutics to treat FLD patients, who have only limited and mostly palliative treatment options available.

KEY POINTS.

  1. LCAT is a plasma enzyme that raises high-density lipoprotein (HDL) levels in plasma and may protect against atherosclerosis.

  2. LCAT is defective in patients with Familial LCAT Deficiency (FLD), who progress to nephrotic syndrome and end-stage renal failure in the fourth or fifth decade of life.

  3. Numerous LCAT-based therapies are under development and some have completed clinical trials.

  4. Enzyme replacement therapy, gene and cell-based therapy, peptide and small-molecule LCAT activators, and lifestyle modifications are appropriate for atherosclerosis, ACS, and other disorders resulting in low or dysfunctional HDL.

  5. Enzyme replacement therapy with rhLCAT and gene therapy are promising avenues for treatment of FLD.

Funding:

Research was supported by the Intramural Research Program of the NHLBI at the National Institutes of Health.

REFERENCES

  • 1.Karathanasis SK, Freeman LA, Gordon SM, Remaley AT. The Changing Face of HDL and the Best Way to Measure It. Clin Chem. 2017;63(1):196–210. [DOI] [PubMed] [Google Scholar]
  • 2.Cuchel M, Rohatgi A, Sacks FM, Guyton JR. JCL roundtable: High-density lipoprotein function and reverse cholesterol transport. J Clin Lipidol. 2018;12(5):1086–94. [DOI] [PubMed] [Google Scholar]
  • 3.Ahsan L, Ossoli AF, Freeman LA, Vaisman B, Amar MJ, Shamburek RD, Remaley AT Role of Lecithin: Cholesterol Acyltransferase in HDL Metabolism and Atherosclerosis. In: Komoda T, editor. The HDL Handbook (Second Edition): Biological Functions and Clinical Implications. 2nd ed: Academic Press; 2014. p. 159–94. [Google Scholar]
  • 4.Glomset JA. The mechanism of the plasma cholesterol esterification reaction: plasma fatty acid transferase. Biochim Biophys Acta. 1962;65:128–35. [DOI] [PubMed] [Google Scholar]
  • 5.**.Calabresi L LCAT deficiency: update for 2020. Current Opinion in Lipidology. 2020. (April). [DOI] [PubMed] [Google Scholar]; A renowned expert in the field provides an update on LCAT deficiency.
  • 6.Glukhova A, Hinkovska-Galcheva V, Kelly R, Abe A, Shayman JA, Tesmer JJ. Structure and function of lysosomal phospholipase A2 and lecithin:cholesterol acyltransferase. Nat Commun. 2015;6:6250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Duivenvoorden R, Holleboom AG, van den Bogaard B, Nederveen AJ, de Groot E, Hutten BA, et al. Carriers of lecithin cholesterol acyltransferase gene mutations have accelerated atherogenesis as assessed by carotid 3.0-T magnetic resonance imaging [corrected]. J Am Coll Cardiol. 2011;58(24):2481–7. [DOI] [PubMed] [Google Scholar]
  • 8.Fazio S, Linton MF. Low levels of high-density lipoprotein cholesterol due to lecithin:cholesterol acyltransferase mutations increase carotid atherosclerosis. J Am Coll Cardiol. 2011;58(24):2488–90. [DOI] [PubMed] [Google Scholar]
  • 9.*.Oldoni F, Baldassarre D, Castelnuovo S, Ossoli A, Amato M, van Capelleveen J, et al. Complete and Partial Lecithin:Cholesterol Acyltransferase Deficiency Is Differentially Associated With Atherosclerosis. Circulation. 2018;138(10):1000–7. [DOI] [PubMed] [Google Scholar]; In this study, heterozygous carriers of FLD mutations were found to have significantly lower carotid intima-media thickness (IMT) than heterozygous carriers of FED mutations, whose IMTs did not differ from controls. This observation suggests that decreasing the ability of LCAT to esterify cholesterol on apoB-containing lipoproteins may decrease risk for atherosclerosis.
  • 10.Lynn EG, Choy PC, Magil A, O K. Uptake and metabolism of lipoprotein-X in mesangial cells. Mol Cell Biochem. 1997;175(1–2):187–94. [DOI] [PubMed] [Google Scholar]
  • 11.*.Freeman LA, Shamburek RD, Sampson ML, Neufeld EB, Sato M, Karathanasis SK, et al. Plasma lipoprotein-X quantification on filipin-stained gels: monitoring recombinant LCAT treatment ex vivo. J Lipid Res. 2019;60(5):1050–7. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper describes a novel method to visualize LP-X in FLD, FED and cholestatic patient plasma on agarose gels using filipin, a free cholesterol (FC) stain. This sensitive, reliable, and quantitative method for measuring LP-X and other FC-enriched lipoproteins will help to elucide the role of LP-X and other abnormal lipoproteins in the pathology of FLD and cholestatic liver disease.
  • 12.O K, Ly M, Fang DZ, Frohlich J, Choy PC. Effect of lipoprotein-X on lipid metabolism in rat kidney. Mol Cell Biochem. 1997;173(1–2):17–24. [DOI] [PubMed] [Google Scholar]
  • 13.Ossoli A, Neufeld EB, Thacker SG, Vaisman B, Pryor M, Freeman LA, et al. Lipoprotein X Causes Renal Disease in LCAT Deficiency. PLoS One. 2016;11(2):e0150083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.*.Vaisman BL, Neufeld EB, Freeman LA, Gordon SM, Sampson ML, Pryor M, et al. LCAT Enzyme Replacement Therapy Reduces LP-X and Improves Kidney Function in a Mouse Model of Familial LCAT Deficiency. J Pharmacol Exp Ther. 2019;368(3):423–34. [DOI] [PMC free article] [PubMed] [Google Scholar]; SREBP-Tg x LCAT-KO mice fed a protein-rich, carbohydrate-low diet rapidly accumulate LP-X in plasma and develop kidney disease that can be markedly reduced by injections of recombinant human LCAT during the diet. This study suggests that LCAT enzyme replacement therapy may be effective in reducing LP-X and preventing renal disease in FLD patients.
  • 15.**.Lamiquiz-Moneo I, Civeira F, Gomez-Coronado D, Blanco-Vaca F, Villafuerte-Ledesma HM, Gil M, et al. Lipid Profile Rather Than the LCAT Mutation Explains Renal Disease in Familial LCAT Deficiency. J Clin Med. 2019;8(11). [DOI] [PMC free article] [PubMed] [Google Scholar]; Of two siblings with identical LCAT mutations, only the sibling with plasma LP-X developed kidney disease. This study supports a causative role for LP-X in FLD kidney disease.
  • 16.Abuhsira L, Karathanasis S, Shi D, Hirschberg B, George RT. Reduction in ApoB Levels After a Single Dose of MEDI6012 in Subjects Receiving High-Intensity Statin Therapy Suggests Enhancement of Low-Density Lipoprotein Receptor-Mediated Reverse Cholesterol Transport. Circulation 2017; 136:A14164. [Google Scholar]
  • 17.George RT, Abuhsira L, Stoughton S, She D, Buss N, Bakker-Arkema R, Koren M, Hirshberg B. Safety, Pharmacokinetics, and Pharmacodynamics of Single Ascending Doses of Recombinant Human Lecithin-Cholesterol Acyltransferase (MEDI6012) in Subjects with Stable Coronary Heart Disease. Circulation 2017;136:A14664. [Google Scholar]
  • 18.**.Bonaca M, George RT, Morrow DA, Abuhsira L, She D, Park JG, et al. Multiple ascending doses of recombinant human lecithin-cholesterol acyltransferase in patients with atherosclerosis: phase 2a primary results ESC2018; 2018: European Heart Journal 2018. p. 49–50. [DOI] [PubMed] [Google Scholar]; In a Phase 2a study, multiple ascending doses of recombinant human LCAT (MEDI6012) administered IV to patients with stable atherosclerosis was safe and well-tolerated and raised HDL-C, HDL-CE and CE. The HDL-raising effects support further study of MEDI6012 as a potential therapy for atherosclerosis.
  • 19.Shamburek RD, Bakker-Arkema R, Shamburek AM, Freeman LA, Amar MJ, Auerbach B, et al. Safety and Tolerability of ACP-501, a Recombinant Human Lecithin:Cholesterol Acyltransferase, in a Phase 1 Single-Dose Escalation Study. Circ Res. 2016;118(1):73–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Collins HL, Vaisman B, Adelman SJ, Remaley AT, Karathanasis S. Acute LCAT infusion increases whole body macrophage-specific cholesterol efflux in LCAT-deficient human ApoA-I transgenic mice. American Heart Association Scientific Sessions; November 13, 2016; New Orleans, LA. [Google Scholar]
  • 21.**.Ossoli A, Simonelli S, Varrenti M, Morici N, Oliva F, Stucchi M, et al. Recombinant LCAT (Lecithin:Cholesterol Acyltransferase) Rescues Defective HDL (High-Density Lipoprotein)-Mediated Endothelial Protection in Acute Coronary Syndrome. Arterioscler Thromb Vasc Biol. 2019;39(5):915–24. [DOI] [PubMed] [Google Scholar]; Incubating STEMI patient plasma with rhLCAT ex vivo restores HDL’s ability to promote NO production from cultured endothelial cells. This study suggests that LCAT may be a potential therapeutic target to restore HDL functionality in acute coronary syndrome.
  • 22.Rousset X, Vaisman B, Auerbach B, Krause BR, Homan R, Stonik J, et al. Effect of recombinant human lecithin cholesterol acyltransferase infusion on lipoprotein metabolism in mice. J Pharmacol Exp Ther. 2010;335(1):140–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Shamburek RD, Bakker-Arkema R, Auerbach BJ, Krause BR, Homan R, Amar MJ, et al. Familial lecithin:cholesterol acyltransferase deficiency: First-in-human treatment with enzyme replacement. J Clin Lipidol. 2016;10(2):356–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.*.Miida T, Hirayama S. Controversy over the atherogenicity of lipoprotein-X. Curr Opin Endocrinol Diabetes Obes. 2019;26(2):117–23. [DOI] [PubMed] [Google Scholar]; The authors discuss factors influencing the possible atherogenicity of different types of LP-X particles and conclude that further studies are needed.
  • 25.*.Amar M, Freeman L, Nishida T, Sampson M, Pryor M, Vaisman B, Neufeld E, Karathanasis S, Remaley AT. LCAT Protects Against Lipoprotein-X Formation in a Murine Model of Drug-Induced Intrahepatic Cholestasis. Pharmacology Research & Perspectives. 2020;8(1):e00554. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this study, mice were gavaged with alpha-napthylisothiocyanate to induce cholestasis and accumulation of LP-X in plasma. LCAT overexpression or injection of rhLCAT prevented LP-X accumulation, indicating that rhLCAT may be a potential therapeutic for lowering LP-X in patients with cholestatic liver disease.
  • 26.Suzuki L, Hirayama S, Fukui M, Sasaki M, Hiroi S, Ayaori M, et al. Lipoprotein-X in cholestatic patients causes xanthomas and promotes foam cell formation in human macrophages. J Clin Lipidol. 2017;11(1):110–8. [DOI] [PubMed] [Google Scholar]
  • 27.Rosenson RS, Baker AL, Chow MJ, Hay RV. Hyperviscosity syndrome in a hypercholesterolemic patient with primary biliary cirrhosis. Gastroenterology. 1990;98(5 Pt 1):1351–7. [DOI] [PubMed] [Google Scholar]
  • 28.Gunawardane RN, Fordstrom P, Piper DE, Masterman S, Siu S, Liu D, et al. Agonistic Human Antibodies Binding to Lecithin-Cholesterol Acyltransferase Modulate High Density Lipoprotein Metabolism. J Biol Chem. 2016;291(6):2799–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Broad Database ExAC browser [Available from: http://exac.broadinstitute.org/transcript/ENST00000600871.]
  • 30.Zhou M, Sawyer J, Kelley K, Fordstrom P, Chan J, Tonn G, Carlson T, Retter M, Meininger D, Cheng J, Gates A, Woodward A, Delaney J, Zhang R, Tang J, Liu Q, Cao P, Luchoomun J, Voogt J, Turner S, Shan B, Boone T, Rudel L, Schwarz M. Lecithin Cholesterol Acyltransferase Promotes Reverse Cholesterol Transport and Attenuates Atherosclerosis Progression in New Zealand White Rabbits. Circulation 2009;120:S1175. [Google Scholar]
  • 31.Zhou M, Shen W, Shan B, Schwarz M, Meininger DP (Seattle, WA), Boone T., inventor; Amgen Inc., assignee. Modified lecithin-cholesterol acyltransferase enzymes patent 9006408. 2015.
  • 32.Boone T, Meininger DP, Schwarz M, Shan B, Shen W, Zhou M, inventor; Amgen Inc., assignee. Modified lecithin-cholesterol acyltransferase enzymes patent 8168416. 2012.
  • 33.Zhou M, Shen W, Shan B, Schwarz M, Meininger DP, Boone T., inventor; Amgen Inc., assignee. Modified lecithin-cholesterol acyltransferase enzymes patent 8703926. 2014.
  • 34.**.Dunbar CE, High KA, Joung JK, Kohn DB, Ozawa K, Sadelain M. Gene therapy comes of age. Science. 2018;359(6372). [DOI] [PubMed] [Google Scholar]; Renowned experts in the field review key landmarks in the development of gene therapy, current status of gene therapy in the clinic, and emerging technologies.
  • 35.Denefle Patrice, Duverger Nicholas, D, Latta-Mahieu Martine, Seguret Sandrine, inventor; Aventis Pharma SA, assignee. Recombinant viruses expressing lecithin-cholesterol acyltransferase, and uses thereof in gene therapy. United States patent US20010014319A1. 2001.
  • 36.Santamarina-Fojo S, Hoeg JH, Brewer HB, inventor; DEPARTMENT OF HEALTH AND HUMAN SERVICES, GOVERNMENT OF THE United States, Secretary of US Department of Health and Human Services (HHS), assignee. Use of lecithin-cholesterol acyltransferase (LCAT) to reduce accumulation of cholesterol. United States patent US6635614B1. 2003.
  • 37.Amar MJ, Shamburek RD, Vaisman B, Knapper CL, Foger B, Hoyt RF Jr., et al. Adenoviral expression of human lecithin-cholesterol acyltransferase in nonhuman primates leads to an antiatherogenic lipoprotein phenotype by increasing high-density lipoprotein and lowering low-density lipoprotein. Metabolism. 2009;58(4):568–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lagor WR, Johnston JC, Lock M, Vandenberghe LH, Rader DJ. Adeno-associated viruses as liver-directed gene delivery vehicles: focus on lipoprotein metabolism. Methods Mol Biol. 2013;1027:273–307. [DOI] [PubMed] [Google Scholar]
  • 39.Christopher DM. Pre-Clinical Development of AAV-Mediated Gene Therapy for Familial Lecithin Cholesterol Acyltransferase Deficiency. Publicly Accessible Penn Dissertations. 2228.: University of Pennsylvania; 2017. [Google Scholar]
  • 40.Kuroda M, Bujo H, Aso M, Saito Y. Adipocytes as a vehicle for ex vivo gene therapy: Novel replacement therapy for diabetes and other metabolic diseases. J Diabetes Investig. 2011;2(5):333–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Aoyagi Y, Kuroda M, Asada S, Bujo H, Tanaka S, Konno S, et al. Fibrin glue increases the cell survival and the transduced gene product secretion of the ceiling culture-derived adipocytes transplanted in mice. Exp Mol Med. 2011;43(3):161–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ozog S, Timberlake ND, Hermann K, Garijo O, Haworth KG, Shi G, et al. Resveratrol trimer enhances gene delivery to hematopoietic stem cells by reducing antiviral restriction at endosomes. Blood. 2019. October 17;134(16):1298–1311. doi: 10.1182/blood.2019000040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Anantharamaiah GM, Venkatachalapathi YV, Brouillette CG, Segrest JP. Use of synthetic peptide analogues to localize lecithin:cholesterol acyltransferase activating domain in apolipoprotein A-I. Arteriosclerosis. 1990;10(1):95–105. [DOI] [PubMed] [Google Scholar]
  • 44.Zhao Y, Thorngate FE, Weisgraber KH, Williams DL, Parks JS. Apolipoprotein E is the major physiological activator of lecithin-cholesterol acyltransferase (LCAT) on apolipoprotein B lipoproteins. Biochemistry. 2005;44(3):1013–25. [DOI] [PubMed] [Google Scholar]
  • 45.Hirsch-Reinshagen V, Donkin J, Stukas S, Chan J, Wilkinson A, Fan J, et al. LCAT synthesized by primary astrocytes esterifies cholesterol on glia-derived lipoproteins. J Lipid Res. 2009;50(5):885–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Sviridov D, Remaley AT. High-density lipoprotein mimetics: promises and challenges. Biochem J. 2015;472(3):249–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sigler GF, Soutar AK, Smith LC, Gotto AM Jr., Sparrow JT. The solid phase synthesis of a protein activator for lecithin-cholesterol acyltransferase corresponding to human plasma apoC-I. Proc Natl Acad Sci U S A. 1976;73(5):1422–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Soutar AK, Sigler GF, Smith LC, Gotto AM Jr., Sparrow JT. Lecithin:cholesterol acyltransferase activation and lipid binding by synthetic fragments of apolipoprotein C-I. Scand J Clin Lab Invest Suppl. 1978;150:53–8. [PubMed] [Google Scholar]
  • 49.Pownall HJ, Hu A, Gotto AM Jr., Albers JJ, Sparrow JT. Activation of lecithin:cholesterol acyltransferase by a synthetic model lipid-associating peptide. Proc Natl Acad Sci U S A. 1980;77(6):3154–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Yokoyama S, Fukushima D, Kupferberg JP, Kezdy FJ, Kaiser ET. The mechanism of activation of lecithin:cholesterol acyltransferase by apolipoprotein A-I and an amphiphilic peptide. J Biol Chem. 1980;255(15):7333–9. [PubMed] [Google Scholar]
  • 51.Subbarao NK, Fielding CJ, Hamilton RL, Szoka FC Jr. Lecithin:cholesterol acyltransferase activation by synthetic amphipathic peptides. Proteins. 1988;3(3):187–98. [DOI] [PubMed] [Google Scholar]
  • 52.Subbarao NK, Parente RA, Szoka FC Jr., Nadasdi L, Pongracz K. pH-dependent bilayer destabilization by an amphipathic peptide. Biochemistry. 1987;26(11):2964–72. [DOI] [PubMed] [Google Scholar]
  • 53.Chung BH, Anatharamaiah GM, Brouillette CG, Nishida T, Segrest JP. Studies of synthetic peptide analogs of the amphipathic helix. Correlation of structure with function. J Biol Chem. 1985;260(18):10256–62. [PubMed] [Google Scholar]
  • 54.Epand RM, Surewicz WK, Hughes DW, Mantsch H, Segrest JP, Allen TM, et al. Properties of lipid complexes with amphipathic helix-forming peptides. Role of distribution of peptide charges. J Biol Chem. 1989;264(8):4628–35. [PubMed] [Google Scholar]
  • 55.Dasseux JL Sekul R, Buttner K, Cornut I, Metz G, DuFourcq J., inventor; Dasseux JL, Sekul R, Buttner K, Cornut I, Metz G, DuFourcq J., assignee. Apolipoprotein A-I Agonists And Their Use To Treat Dyslipidemic Disorders. USA patent 6004925. 1999.
  • 56.Li D, Gordon S, Schwendeman A, Remaley A. Apolipoprotein Mimetic Peptides for Stimulating Cholesterol Efflux. In: Anantharamaiah GM and Goldberg D, editors. Apolipoprotein Mimetics in the Management of Human Disease. Switzerland: ADIS / Springer; 2015. p. 29–42. [Google Scholar]
  • 57.Vaisman BL, Remaley AT. Measurement of lecithin-cholesterol acyltransferase activity with the use of a Peptide-proteoliposome substrate. In: Freeman LA, editor. Lipoproteins and Cardiovascular Disease: Methods and Protocols Methods Mol Biol. 1027. 2013/August/06 ed. New York: Springer; 2013. p. 343–52. [DOI] [PubMed] [Google Scholar]
  • 58.*.Martha S. Hayden-Ledbetter JL, inventor; Theripion Inc, assignee. ApoA-1 fusion polypeptides and related compositions and methods. United States patent US20180201664A1. 2018.; Fusing apoA-I mimetic peptides that activate LCAT to Fc may increase their half-life in plasma, thus enhancing their therapeutic effect.
  • 59.Chen Z, Wang SP, Krsmanovic ML, Castro-Perez J, Gagen K, Mendoza V, et al. Small molecule activation of lecithin cholesterol acyltransferase modulates lipoprotein metabolism in mice and hamsters. Metabolism. 2012;61(4):470–81. [DOI] [PubMed] [Google Scholar]
  • 60.Freeman LA, Demosky SJ Jr., Konaklieva M, Kuskovsky R, Aponte A, Ossoli AF, et al. Lecithin:Cholesterol Acyltransferase Activation by Sulfhydryl-Reactive Small Molecules: Role of Cysteine-31. J Pharmacol Exp Ther. 2017;362(2):306–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kayser Frank, Labelle Marc, Shan Bei, Zhang Jian, Zhou Mingyue, inventor; Amgen Inc., assignee. Methods for Treating Atherosclerosis. USA patent 8426358 2008. April/24/2008.
  • 62.Hengstschlager-Ottnad E, Kuchler K, Schneider WJ. Chicken lecithin-cholesterol acyltransferase. Molecular characterization reveals unusual structure and expression pattern. J Biol Chem. 1995;270(44):26139–45. [DOI] [PubMed] [Google Scholar]
  • 63.Hermier D, Forgez P, Williams J, Chapman MJ. Alterations in plasma lipoproteins and apolipoproteins associated with estrogen-induced hyperlipidemia in the laying hen. Eur J Biochem. 1989;184(1):109–18. [DOI] [PubMed] [Google Scholar]
  • 64.Yamada K, Delawary M, Kobayashi H, Kaneko T, Watanabe A, Shimizu S, Sugidachi A. A Novel and Orally Available Lecithin Cholesterol Acyltransferase Activator Prevents the Progression of Plaque Accumulation in an Atherosclerosis Mouse Model. Circulation. 2017;136(Suppl_1):A15584. [Google Scholar]
  • 65.Kobayashi H, Ohkawa N, Takano M, Kubota H, Onoda T, Kaneko T, Arai M, Terasaka N, inventor; Daiichi Sankyo Co Ltd, assignee. Piperidinylpyrazolopyridine derivative patent US9150575B2. 2015.
  • 66.Kobayashi H, Onoda T, Kaneko T, Ishibashi K, inventor. Condensed Pyrazole Derivative patent WO2015111545A1. 2015.
  • 67.Kobayashi H, Arai M, Kaneko T, Terasaka N, inventor; Daiichi Sankyo Co Ltd, assignee. 5-hydroxy-4-(trifluoromethyl)pyrazolopyridine derivative. patent US9796709B2. 2016.
  • 68.Onoda T, Kaneko T, Arai M, Kobayashi H, Terasaka N, inventor; Daiichi Sankyo Co., Ltd, assignee. Imidazopyridine Derivative. patent WO2015087996A1. 2015.
  • 69.**.Manthei KA, Yang SM, Baljinnyam B, Chang L, Glukhova A, Yuan W, et al. Molecular basis for activation of lecithin:cholesterol acyltransferase by a compound that increases HDL cholesterol. Elife. 2018;7. pii: e41604. doi: 10.7554/eLife.41604. [DOI] [PMC free article] [PubMed] [Google Scholar]; Structural and biochemical studies of a small-molecule LCAT activator bound to LCAT reveals a novel activation mechanism and inspires rational design of a more potent LCAT agonist.
  • 70.Glomset JA, Norum KR, Nichols AV, King WC, Mitchell CD, Applegate KR, et al. Plasma lipoproteins in familial lecithin: cholesterol acyltransferase deficiency: effects of dietary manipulation. Scand J Clin Lab Invest Suppl. 1975;142:3–30. [PubMed] [Google Scholar]
  • 71.Naito S, Kamata M, Furuya M, Hayashi M, Kuroda M, Bujo H, et al. Amelioration of circulating lipoprotein profile and proteinuria in a patient with LCAT deficiency due to a novel mutation (Cys74Tyr) in the lid region of LCAT under a fat-restricted diet and ARB treatment. Atherosclerosis. 2013;228(1):193–7. [DOI] [PubMed] [Google Scholar]
  • 72.Yee MS, Pavitt DV, Richmond W, Cook HT, McLean AG, Valabhji J, et al. Changes in lipoprotein profile and urinary albumin excretion in familial LCAT deficiency with lipid lowering therapy. Atherosclerosis. 2009;205(2):528–32. [DOI] [PubMed] [Google Scholar]
  • 73.Mulligan JJ, Hojnacki JL, Cluette JE, Kew RR, Stack DJ, Nicolosi RJ, et al. Cigarette smoke-induced depression in LCAT activity. Exp Mol Pathol. 1981;35(1):25–35. [DOI] [PubMed] [Google Scholar]
  • 74.McCall MR, van den Berg JJ, Kuypers FA, Tribble DL, Krauss RM, Knoff LJ, et al. Modification of LCAT activity and HDL structure. New links between cigarette smoke and coronary heart disease risk. Arterioscler Thromb. 1994;14(2):248–53. [DOI] [PubMed] [Google Scholar]
  • 75.Chen C, Loo G. Inhibition of lecithin: cholesterol acyltransferase activity in human blood plasma by cigarette smoke extract and reactive aldehydes. J Biochem Toxicol. 1995;10(3):121–8. [DOI] [PubMed] [Google Scholar]
  • 76.Bielicki JK, Forte TM, McCall MR. Gas-phase cigarette smoke inhibits plasma lecithin-cholesterol acyltransferase activity by modification of the enzyme’s free thiols. Biochim Biophys Acta. 1995;1258(1):35–40. [DOI] [PubMed] [Google Scholar]
  • 77.Chen C, Loo G. Effect of peroxyl radicals on lecithin/cholesterol acyltransferase activity in human plasma. Lipids. 1995;30(7):627–31. [DOI] [PubMed] [Google Scholar]
  • 78.Soupene E, Borja MS, Borda M, Larkin SK, Kuypers FA. Featured Article: Alterations of lecithin cholesterol acyltransferase activity and apolipoprotein A-I functionality in human sickle blood. Exp Biol Med (Maywood). 2016;241(17):1933–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Holzer M, Wolf P, Inzinger M, Trieb M, Curcic S, Pasterk L, et al. Anti-psoriatic therapy recovers high-density lipoprotein composition and function. J Invest Dermatol. 2014;134(3):635–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Mehta NN, Gelfand JM. High-density lipoprotein cholesterol function improves after successful treatment of psoriasis: a step forward in the right direction. J Invest Dermatol. 2014;134(3):592–5. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES