Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Aug 1.
Published in final edited form as: Curr Opin Lipidol. 2014 Aug;25(4):304–308. doi: 10.1097/MOL.0000000000000092

Apolipoprotein A-I Mimetics

Srinivasa T Reddy 1,2,3, Mohamad Navab 1, G M Anantharamaiah 4, Alan M Fogelman 1,5
PMCID: PMC4213064  NIHMSID: NIHMS637868  PMID: 24977978

Abstract

Purpose of Review

To summarize recent publications in the field of apolipoprotein mimetics.

Recent Findings

Apolipoprotein mimetic peptides continue to show efficacy in a number of animal models of disease and demonstrate properties that make them attractive as potential therapeutic agents. A number of new apolipoprotein mimetics have been described recently. A major site of action of apolipoprotein mimetic peptides was found to be in the small intestine where they decrease the levels of pro-inflammatory bioactive lipids. A major problem related to the use of apolipoprotein mimetic peptides is their cost, particularly those that need to be generated by solid phase synthesis with chemical addition of end blocking groups. Novel approaches to apolipoprotein mimetic therapy have emerged recently that show promise in overcoming these barriers.

Summary

Despite the recent failure of therapies designed to raise HDL-cholesterol in humans, an approach to therapy using mimetics of HDL and its components continues to show promise.

Keywords: Apolipoproteins, Apolipoprotein mimetics, HDL, Atherosclerosis, Cancer

Introduction

The failure of recent clinical trials to achieve a positive outcome using therapies that significantly raised HDL-cholesterol levels has brought into question the “HDL Hypothesis” [1**]. However, as recently reviewed, the pursuit of a strategy to use components of HDL or mimics of HDL or mimics of its components is quite different from the strategy to raise plasma HDL-cholesterol levels[2**]. In this article we will review recent advances in the field of apolipoprotein mimetics.

Efficacy of Apolipoprotein Mimetics in Models of Disease

Apolipoprotein mimetics have been shown to be efficacious in a large number of models of disease [2**, 3]. Below is a summary of recent publications on the efficacy and mechanism of action of apolipoprotein mimetics in a variety of animal models.

Bacterial lipopolysaccharide (LPS), Endotoxemia, Lung Injury, Asthma, Air Pollution, and Leukocyte Responses

LPS was administered to rats at 10mg/kg via tail vein [4*]. Ten minutes later L-4F (10 mg/kg) was administered intraperitoneally. 4F improved survival in the endotoxemic rats and attenuated histologic damage in lung tissue. The 4F-mediated improvement was accompanied by higher levels of HDL-cholesterol and lower lung myeloperoxidase (MPO) activity compared to LPS alone [4*].

It was previously shown that 4F favors the differentiation of human monocytes to an anti-inflammatory phenotype and that 4F attenuates LPS-induced inflammatory responses. White and colleagues [5*] investigated the effects of LPS on the Toll-like receptor (TLR) signaling pathway in 4F-differentiated monocyte-derived macrophages. Treatment with L-4F downregulated cell-surface TLRs (4,5 and 6) and attenuated the LPS-dependent upregulation of genes encoding TLRs 1, 2, and 6 and genes of the MyD88-dependent pathways. The L-4F-mediated changes were associated with depletion of cellular cholesterol and caveolin, components of membrane lipid rafts. [5*].

The dual (apoE and apoA-I) domain peptide Ac-hE18A-NH2 peptide with 28 amino acid residues was compared to 4F, an 18 residue apoA-I mimetic peptide for anti-endotoxin activity [6*]. The dual domain peptide inhibited endotoxin activity and disaggregated Escherichia coli 055:B5 (wild smooth serotype) better than 4F. In THP-1 cells, isolated primary leukocytes, and whole human blood, the dual domain peptide reduced responses more strongly that 4F [6*].

In a rat model of severe endotoxemia, L-4F (10 mg/kg intravenously) significantly decreased mortality and reduced lung and liver injury, even when administered 1 hour post LPS [7*]. In vitro, L-4F inhibited the activation of isolated human leukocytes and neutrophils by serum taken from patients with acute respiratory distress syndrome (ARDS)[7*].

ApoA-I null and apoE null mice were shown to have enhanced recruitment of neutrophils to the airspace in response to both inhaled LPS and direct airway inoculation with CXCL1. Conversely, treatment with L-4F or an apoE mimetic peptide (COG1410) reduced airway neutrophilia. L-4F was suspected of suppressing chemotaxis through heterologous desensitization [8*]. L-4F was found to induce chemotaxis of human neutrophils and monocytes but failed to induce calcium influx. [8*].

Air pollution is known to contribute to atherosclerosis in humans. Exposure of LDLR null mice to air borne ultrafine particles reduced HDL-cholesterol and antioxidant activity and increased atherosclerosis, which was ameliorated by an apoA-I mimetic peptide, D-4F [9*].

Using a multianalyte microphysiometer, it was demonstrated that L-4F complexed or not complexed with 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) attenuated the macrophage respiratory burst in response to oxidized LDL [10*].

Hypertension induced in mice was accompanied by increased leukocyte and platelet adhesion in cerebral venules and was attenuated by mild hypercholesterolemia [11*]. Using mice with transgenic expression of apoA-I and wild-type mice treated with 4F suggested that the beneficial effect of the mild hypercholesterolemia was HDL-mediated [11*].

Endothelial Nitric Oxide Synthase (eNOS), Tight Skin Mice, and Insulin Sensitivity

Acute hyperglycemia is known to decrease the availability of nitric oxide (NO) and impair anesthetic preconditioning (APC)-elicited protection against myocardial infarction. The authors [12*] measured myocardial infarct size in mice in the absence or presence of APC achieved with isoflurane (1.4%) with or without acute hyperglycemia. Myocardial infarct size was significantly decreased by APC in the absence, but not in the presence of acute hyperglycemia. D-4F restored the cardioprotective effect of APC during acute hyperglycemia. In vitro D-4F reduced superoxide generation and enhanced caveolin-1 and eNOS caveolar compartmentalization and posttranslational eNOS modifications restoring NO production when isoflurane was administered under hyperglycemic conditions [12*].

In vitro, reverse D-4F (D-4F sequence read from C-terminal end to the N-terminal end) improved the proliferation, migration, and tube formation of endothelial progenitor cells (EPCs) in a dose dependent manner and activated phosphor-AKT at serine residue 473 and phosphor-eNOS at serine residue 1177 [13*]. The authors concluded that reverse D-4F-mediated improvement of EPC function is dependent on the PI3K/AKT/eNOS pathway [13*].

Having previously shown that treatment with 4F of tight skin (Tsk−/+) mice, a model of systemic sclerosis decreases inflammation and restores angiogenic potential in Tsk−/+ mice, the authors [14*] hypothesized that 4F differentially modulates interferon regulating factor 5 (IRF5) in myocardium. IRF5 in heart lysates from control and Tsk−/+ mice with and without 4F treatment was determined. 4F was shown to bind IRF5. IRF5 activation was increased in Tsk−/+ hearts and 4F treatment decreased IRF5 expression and activation in the Tsk−/+ hearts by a mechanism related to 4F’s ability to bind IRF5 [14*].

Administration of L-4F to obese (B6V-Lep-ob/J) mice restored adipocyte function, increased adiponectin release and decreased levels of IL-1 and IL-6 and was associated with an increase in insulin sensitivity and decreased glucose levels [15*]. The mechanism appeared to be related to an L-4F-mediated increase in heme oxygenase 1 (HO-1) and Wnt10b activity [15*].

ApoA-I mimetic peptides in mouse models of cancer

The first evidence of the utility of apoA-I mimetic peptides in mouse models of cancer was published in 2010 [16]. In 2011 another report appeared [17]. Subsequently, 3 other manuscripts have appeared indicating the mechanism(s) of action [18*, 19*, 20*] and lending support to the possibility that apoA-I mimetic peptides may have therapeutic potential in cancer.

Properties of 4F Peptides and Comparison with ApoE Mimetic Peptides

L-4F complexed to 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) enhanced paraoxonase-1 (PON1) activity and stability compared to controls [21*].

It is known that myeloperoxidase (MPO)-derived hypochlorous acid (HOCl) induces changes in HDL function by causing oxidative modification of apoA-I [22**]. It was found that HOCl caused oxidation of the sole tryptophan (Trp) residue in L-4F. Remarkably, neither L-4F-mediated lipid binding nor ABCA1-dependent cholesterol efflux was altered as result of the oxidation of L-4F by HOCl. It was concluded that L-4F serves as a reactive substrate for HOCl, but the reaction does not influence the lipid binding and cholesterol efflux capacity of the peptide [23*].

An apoE mimetic peptide Ac-hE18A-NH2 was compared to the apoA-I mimetic peptide L-4F in apoE null mice [24*]. At equal doses, frequency and route of administration plasma cholesterol and triglyceride levels significantly decreased with the apoE mimetic peptide but not with L-4F. Both the apoE mimetic peptide and L-4F significantly decreased lesions by en face analysis, but the decrease with the apoE mimetic peptide was significantly greater than with L-4F [24*].

Other Apolipoprotein-mimetics

A nice summary of molecules that mimic apoA-I properties recently appeared [25**]. These authors developed peptide-lipid nanoparticles using branched, multivalent constructs bearing multiple 23- or 16-amino acid peptides that were combined with phospholipids to produce nanometer-scale discoidal HDL-like particles [26*]. These nanolipid particles promoted efficient cellular cholesterol efflux and were remarkably stable toward enzymatic digestion in vitro and displayed long half-lives and desirable pharmacokinetic profiles in mice and at 50 mg/kg intraperitoneally reduced plasma cholesterol by 50% [26*].

Pegylation of human holo-HDL or reconstituted phospholipid/apoA-I particles (rHDL) led to selective N-terminal monopegylation of apoA-I with full preservation of cholesterol efflux activity [27*]. The plasma clearance of PEG-rHDL was estimated after injection into apoE null mice; the half-life of PEG-rHDL was about 7-fold greater than rHDL. The PEG-rHDL at a dose of 40 mg/kg in apoE null mice led to more pronounced suppression of bone marrow myeloid progenitor cell proliferation and monocytosis. PEG-rHDL compared to rHDL also produced a more stable lesion [27*].

A 26-mer α-helical peptide (ATI-5261) was exposed to acrolein and resulted in adducts at positions 5 and 25 and led to a concentration-dependent reduction in cholesterol efflux activity [28*]. Placement of EXXK in the center of the peptide resulted in site-specific modification of lysine, which was glutamate dependent [28*].

Two tandem repeats of the 4F peptide were fused to the C terminus of the murine IgG Fc fragment [29*]. The resultant peptibody, mFC-2X4F, dose dependently promoted cholesterol efflux in vitro, and the efflux potency was superior to monomeric 4F peptide. When administered to mice, the peptibody increased both pre-β and α-1 HDL subfractions. The peptibody and apoA-I were found to coexist in the HDL particles formed in vivo [29*].

The peptide ELK-2A2K2E (EKLKAKLEELKAKLEELL-P-EKLKAKLEELKAKLEELL) at a dose of 30 mg/kg administered intraperitoneally three times per week enhanced the rate of reverse cholesterol transport in C57BL/6 mice and modestly reduced the size of atherosclerotic plaques in the aortic arch with significant reductions in CD68 positive staining, VCAM-1 positive staining and nitrotyrosine staining in the aortic arch of apoE null mice [30*].

A series of 24 amino acid residue peptides mimicking apoA-I were tested [31*]. The FAMP5 peptide was equally as effective in promoting ABCA1-mediated cholesterol efflux in vitro whether or not its end groups were chemically blocked. In contrast L-4F was no more effective than bovine serum albumin without end groups but was significantly more effective than FAMP5 when its end groups were chemically blocked. In apoE null mice fed a high-fat high-cholesterol diet for 16 weeks administration of FAMP5 at 50 mg/kg given intraperitoneally three times per week significantly reduced aortic lesion area, but a dose of 10 mg/kg was not effective [31*].

FAMP was modified so it could be radiolabeled with gallium-68 for noninvasive positron emission tomography (PET) in a rabbit model of familial hypercholesterolemia that spontaneously develops myocardial infarction (WHHL-MI) [32*]. The radiolabeled FAMP was dramatically taken up by atherosclerotic tissues in the blood vessels and aorta of WHHL-MI rabbits but not in control rabbits [32*].

Site, Mechanism of Action, and Novel Approach to ApoA-I Mimetic Peptides

ApoA-I mimetic peptides 4F, 5F, and 6F derived from the 18A sequence have been studied for their mechanism of action in detail. The small intestine was found to be a major site of action for apoA-I mimetic peptides regardless of their route of administration [33,34*]. The dose of peptide needed for efficacy in the small intestine was determined to be high (40 – 100 mg/kg) and likely explains the divergent results of studies in humans [3**, 33, 34*]. There have been three reports on the use of the 4F peptide in humans [3537]. Two of these reports showed efficacy [35, 36] and one did not [37]. The third report [37] used doses that were ineffective in the first two studies (i.e. the maximum dose tested in the third study was 1.43 mg/kg).

To overcome the problem of the high cost of production of chemically synthesized peptides such as 4F, which require chemically added end blocking groups, a search was conducted for peptides that did not need end blocking groups for efficacy. The peptide 6F was found to be such a peptide [38**, 39*]. Transgenic tomatoes expressing the 6F peptide (Tg6F) were generated. When Tg6F was freeze-dried and added to a Western diet at only 2.2% of diet by weight there was a significant reduction in aortic atherosclerosis in LDLR null mice that was significantly correlated with levels of unsaturated lysophosphatidic acid in the small intestine [38**, 39*]. Intact Tg6F was found in the small intestine but not in the plasma indicating that it was indeed working in the small intestine [38**,39*]. Addition of unsaturated (but not saturated) lysophosphatidic acid to low-fat mouse chow produced levels of unsaturated lysophosphatidic acid in the small intestine, dyslipidemia and inflammation similar to those seen on feeding LDLR null mice a high-fat high-cholesterol Western diet; and Tg6F ameliorated the changes [40**, 41*].

Conclusion

Despite the failure of clinical trials to demonstrate the value of raising HDL-cholesterol levels, there continues to be active research on strategies to use HDL, its components, or mimetics of its components as therapeutic agents. The efficacy of apolipoprotein mimetics in such a large number of models of disease suggests that if a practical method of producing and administering these agents can be found, they may still have great therapeutic potential.

Key Points.

  • Apolipoprotein mimetics have been found to be efficacious in a large number of disease models.

  • The dose of apolipoprotein mimetics required for chronic administration makes the cost of treatment prohibitive if the mimetics have to be generated by chemical synthesis.

  • At least some apolipoprotein mimetics appear to act primarily in the small intestine and thus oral administration is favored as the route of administration.

  • Novel methods for producing apolipoprotein mimetics at a reasonable cost will be required for the strategy to be tested in humans at the doses required to treat chronic conditions.

Acknowledgments

Supported in part by USPHS grant HL30568 and a Leducq Network grant.

Funding: This work was supported in part by USPHS grants HL30568 and HL34343 and a Network Grant from the Leducq Foundation.

Footnotes

Disclosures: STR, MN, GMA and AMF are principals in Bruin Pharma and AMF is an officer in Bruin Pharma.

References

  • 1**.Lüscher TF, Landmesser U, von Eckardstein A, Fogelman AM. High-density lipoprotein: Vascular protective effects, dysfunction, and potential as therapeutic target. Circ Res. 2014;114:171–182. doi: 10.1161/CIRCRESAHA.114.300935. This review puts into perspective the recent clinical trials aimed at raising HDL-cholesterol levels. [DOI] [PubMed] [Google Scholar]
  • 2**.Reddy ST, Navab M, Anantharamaiah GM, Fogelman AM. Searching for a successful HDL-based treatment strategy. Biochim Biophys Acta. 2014;1841:162–167. This review makes the case for pursuing an HDL-based treatment strategy despite the failure of the recent clinical trials aimed at raising HDL-cholesterol levels. [PubMed] [Google Scholar]
  • 3.Navab M, Reddy ST, Van Lenten BJ, et al. High-density lipoprotein and 4F peptide reduce systemic inflammation by modulating intestinal oxidized lipid metabolism: Novel hypotheses and review of literature. Arterioscler Thromb Vasc Biol. 2012;32:2553–2560. doi: 10.1161/ATVBAHA.112.300282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4*.Kwon WY, Suh Gj, Kim KS, et al. 4F, apolipoprotein AI mimetic peptide, attenuates acute lung injury and improves survival in endotoxemic rats. J Trauma Acute Care Surg. 2012;72:1576–1583. doi: 10.1097/TA.0b013e3182493ab4. This study shows that an apoA-I mimetic peptide can be effective even after endotoxemia was established. [DOI] [PubMed] [Google Scholar]
  • 5*.White CR, Smythies LE, Crossman DK, et al. Regulation of pattern recognition receptors by the apolipoprotein A-I mimetic peptide 4F. Arterioscler Thromb Vasc Biol. 2012;32:2631–2639. doi: 10.1161/ATVBAHA.112.300167. This study demonstrates that apoA-I mimetic peptides can mediate macrophage responsiveness to pro-inflammatory signals by regulating TLRs. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6*.Sharifov OF, Nayyar G, Ternovoy VV, et al. Comparison of anti-endotoxin activity of apoE and apoA mimetic derivatives of a model amphipathic peptide 18A. Innate Immunity. 2013 doi: 10.1177/1753425913514621. [Epub ahead of print]. This paper compares a dual domain peptide to a single domain peptide in ability to modulate the pro-inflammatory effects of LPS. [DOI] [PubMed] [Google Scholar]
  • 7*.Sharifov OF, Xu X, Gaggar A, et al. Anti-inflammatory mechanisms of apolipoprotein A-I mimetic peptide in acute respiratory distress syndrome secondary to sepsis. Plos One. 2013;8:e64486. doi: 10.1371/journal.pone.0064486. This manuscript provides further evidence that apoA-I mimetic peptides can be effective even after the onset of endotoxemia. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8*.Madenspacher JH, Azzam KM, Gong W, et al. Apolipoproteins and apolipoprotein mimetic peptides modulate phagocyte trafficking through chemotactic activity. J Biol Chem. 2012;287:43730–43740. doi: 10.1074/jbc.M112.377192. This manuscript demonstrates a novel property of apoA-I mimetic peptides in modulating responses to inflammatory stimuli in the lungs. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9*.Li R, Navab M, Pakbin P, et al. Ambient ultrafine particles alter lipid metabolism and HDL anti-oxidant capacity in LDLR-null mice. J Lipid Res. 2013;54:1608–1615. doi: 10.1194/jlr.M035014. This paper demonstrates the utility of an apoA-I mimetic in a mouse model of air pollution accelerated inflammation, dyslipidemia and atherosclerosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10*.Kimmel DW, Dole WP, Cliffel DE. Application of multianalyte microphysiometry to characterize metabolic responses to oxidized LDL and effects of an apoA-1 mimetic. Biochem Biophys Res Commun. 2013;431:181–185. doi: 10.1016/j.bbrc.2012.12.140. This manuscript demonstrates that apoA-I mimetic peptides alter the response of macrophages to pro-inflammatory stimuli including oxidized LDL. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11*.Rodrigues SF, Vital SA, Granger DN. Mild hypercholesterolemia blunts the proinflammatory and prothrombotic effects of hypertension on the cerebral microcirculation. Journal of Cerebral Blood Flow & Metabolism. 2013;33:483–489. doi: 10.1038/jcbfm.2012.194. This manuscript demonstrates the ability of an apoA-I mimetic peptide to alter leukocyte adhesion in cerebral venules in hypertensive animal models. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12*.Baotic I, Ge Z-D, Sedlic F, et al. Apolipoprotein A-1 mimetic peptide D-4F enhances isoflurane-induced eNOS signaling and cardioprotection during acute hyperglycemia. Am J Physiol Heart Circ Physiol. 2013;305:H219–H227. doi: 10.1152/ajpheart.00850.2012. This paper demonstrates that apoA-I mimetic peptides improve nitric oxide production. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13*.Yang N, Yao S, Wang M, et al. Apolipoprotein A-I mimetic peptide reverse D-4F improves the biological functions of mouse bone marrow-derived late EPCs via PI3K/AKT/eNOS pathway. Mol Cell Biochem. 2013;377:229–236. doi: 10.1007/s11010-013-1592-z. This manuscript provides further evidence of the importance of the endothelial nitric oxide pathway in mediating the beneficial effects of apoA-I mimetic peptides. [DOI] [PubMed] [Google Scholar]
  • 14*.Xu H, Krolikowski JG, Jones DW, et al. 4F decreases IRF5 expression and activation in hearts of tight skin mice. PLoS One. 2012;7:e52046. doi: 10.1371/journal.pone.0052046. This paper provides new insights into the mechanism of action of 4F in a mouse model of systemic sclerosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15*.Vanella L, Li M, Kim DH, et al. ApoA1 mimetic peptide reverses adipocyte dysfunction in vivo and in vitro via an increase in heme oxygenase (HO-1) and Wnt10b. Cell Cycle. 2012;11:706–714. doi: 10.4161/cc.11.4.19125. This manuscript demonstrates that apoA-I mimetic peptides can improve adipocyte dysfunction by inducing HO-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Su F, Kozak KR, Imaizumi S, et al. Apolipoprotein A-I (apoA-I) and apoA-I mimetic peptides inhibit tumor development in a mouse model of ovarian cancer. PNAS. 2010;107:19997–20002. doi: 10.1073/pnas.1009010107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gao F, Vasquez Sx, Su F, et al. L-5F, an apolipoprotein A-I mimetic, inhibits tumor angiogenesis by suppressing VEGF/basic FGF signaling pathways. Integr Biol (Camb) 2011;3:479–489. doi: 10.1039/c0ib00147c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18*.Ganapathy E, Su F, Meriwether D, et al. D-4F, an apoA-I mimetic peptide inhibits proliferation and tumorigenicity of epithelial ovarian cancer cells by upregulating the antioxidant enzyme MnSOD. In J Cancer. 2012;130:1071–1081. doi: 10.1002/ijc.26079. This manuscript suggests that induction of antioxidant enzymes such as MnSOD plays a role in the inhibition of ovarian cancer by apoA-I mimetic peptides. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19*.Su F, Grijalva V, Navab K, et al. HDL mimetics inhibit tumor development in both induced and spontaneous mouse models of colon cancer. Mol Cancer Ther. 2012:1311–1319. doi: 10.1158/1535-7163.MCT-11-0905. This paper demonstrates the efficacy of apoA-I mimetic and apoJ mimetic peptides in mouse models of colon cancer and adds further evidence of the role of lysophosphatidic acid in the mechanism of action. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20*.Gao F, Chattopadhyay A, Navab M, et al. Apolipoprotein A-I mimetic peptides inhibit expression and activity of hypoxia-inducible factor-1α in human ovarian cancer cell lines and a mouse ovarian cancer model. J Pharmcol Exp Ther. 2012;342:255–262. doi: 10.1124/jpet.112.191544. This manuscript provides evidence that the decrease in tumor burden by apoA-I mimetic peptides may be due to decreased expression and activity of HIF-1α, which induces angiogenesis through VEGF. This paper also adds further evidence that lysophosphatidic acid maybe involved in the mechanism. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21*.Mishra VK, Palgunachari MN, McPherson DT, Anantharamaiah GM. Lipid complex of apolipoprotein A-I mimetic peptide 4F is a novel platform for paraoxonase-1 binding and enhancing its activity and stability. Biochem Biophys Res Commun. 2013;430:975–980. doi: 10.1016/j.bbrc.2012.11.128. This paper provides evidence that the 4F peptide’s interaction with phospholipids provides a platform for the binding of the antioxidant enzyme PON1 and also stabilizes the enzyme. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22**.Huang Y, DiDonato JA, Levison BS, et al. An abundant dysfunctional apolipoprotein A1 in human atheroma. Nature Medicine. 2014;20:193–203. doi: 10.1038/nm.3459. This outstanding manuscript identifies MPO altered apoA-I in human atheroma and provides an antibody that may be useful to define MPO damaged apoA-I in humans. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23*.White CR, Datta G, Buck AKW, et al. Preservation of biological function despite oxidative modification of the apolipoprotein A-I mimetic peptide 4F. J Lipid Res. 2012;53:1576– 1587. doi: 10.1194/jlr.M026278. This paper demonstrates that L-4F remains functional even after modification by MPO. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24*.Nayyar G, Garber DW, Palgunachari MN, et al. Apolipoprotein E mimetic is more effective than apolipoprotein A-I mimetic in reducing lesion formation in older female apo E null mice. Atherosclerosis. 2012;224:326–331. doi: 10.1016/j.atherosclerosis.2012.05.040. This manuscript demonstrates that in mice lacking apoE an apoE mimetic peptide is superior to an apoA-I mimetic peptide. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25**.Leman LJ, Maryanoff BE, Ghadiri MR. Molecules that mimic apolipoprotein A-I: Potential agents for treating atherosclerosis. J Med Chem. 2013 doi: 10.1021/jm4005847. [Epub ahead of print]. This is a nice review of apoA-I mimetics that covers many (but not all) of the apoA-I mimetic peptides described in this review. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26*.Zhao Y, Imura T, Leman LJ, et al. Mimicry of high-density lipoprotein: functional peptide-lipid nanoparticles based on multivalent peptide constructs. J Am Chem Soc. 2013;135:13414–13424. doi: 10.1021/ja404714a. This manuscript describes the development of nanoparticles using multivalent peptide constructs that mimic HDL. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27*.Murphy AJ, Funt S, Gorman D, et al. Pegylation of high-density lipoprotein decreases plasma clearance and enhances antiatherogenic activity. Circ Res. 2013;113:e1–e9. doi: 10.1161/CIRCRESAHA.113.301112. This paper describes a method to greatly extend the plasma half-life of apoA-I. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28*.Zheng Y, Kim SH, Patel AB, et al. The positional specificity of EXXK motifs within an amphipathic a-helix dictates preferential lysine modification by acrolein: Implications for the design of high-density lipoprotein mimetic peptides. Biochemistry. 2012;51:6400–5412. doi: 10.1021/bi300626g. This manuscript details the susceptibility of lysines in apoA-I mimetic peptides to modification by reactive molecules such as acrolein based on their location in the peptide. [DOI] [PubMed] [Google Scholar]
  • 29*.Liu S-C, Atangan L, Kim KW, et al. An apoA-I mimetic peptibody generates HDL-like particles and increases alpha-1 HDL subfraction in mice. J Lipid Res. 2012;53:643–652. doi: 10.1194/jlr.M020438. This paper describes the construction of a novel apoA-I mimetic formed by tandem repeats of the 4F peptide fused to an IgGFc fragment. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30*.Ditiatkovski M, D’Souza W, Kesani R, et al. An apolipoprotein A-I mimetic peptide designed with a reductionist approach stimulates reverse cholesterol transport and reduces atherosclerosis in mice. PLoS One. 2013;8:e68802. doi: 10.1371/journal.pone.0068802. This manuscript demonstrates the efficacy of a novel apolipoprotien mimetic peptide in mice. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31*.Uehara Y, Ando S, Yahiro E, et al. FAMP, a novel apoA-I mimetic peptide, suppresses aortic plaque formation through promotion of biological HDL function in apoE-deficient mice. J Am Heart Assoc. 2013;2:e000048. doi: 10.1161/JAHA.113.000048. This manuscript describes a new apoA-I mimetic peptide and demonstrates that high doses of peptide are required which make the cost of production an issue for this and other apolipoprotein mimetics. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32*.Kawachi E, Yoshinari U, Hasegawa K, et al. Novel molecular imaging of atherosclerosis with gallium-68-labeled apolipoprotein A-I mimetic peptide and positron emission tomography. Circ J. 2013;77:1482– 1489. doi: 10.1253/circj.cj-12-0736. This paper describes a novel use of an apoA-I mimetic peptide as an imaging agent. [DOI] [PubMed] [Google Scholar]
  • 33.Navab M, Reddy ST, Anantharamaiah GM, et al. Intestine may be a major site of action for the apoA-I mimetic peptide 4F whether administered subcutaneously or orally. J Lipid Res. 2011;52:1200–1210. doi: 10.1194/jlr.M013144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34*.Navab M, Reddy ST, Anantharamaiah GM, et al. D-4F-mediated reduction in metabolites of arachidonic and linoleic acids in the small intestine is associated with decreased inflammation in low-density lipoprotein receptor-null mice. J Lipid Res. 2012;53:437–445. doi: 10.1194/jlr.M023523. This manuscript surprisingly demonstrated that the biologic activity of the apoA-I mimetic peptide D-4F was similar at similar doses whether the peptide was administered orally or subcutaneously despite differences in plasma levels of 100- to 1,000-fold. The only compartment with similar peptide levels after oral or subcutaneous administration was the small intestine. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bloedon LT, Dunbar R, Duffy D, et al. Safety, pharmacokinetics, and pharmacodynamics of oral apoA-I mimetic peptide D-4F in high-risk cardiovascular patients. J Lipid Res. 2008;49:1344–1352. doi: 10.1194/jlr.P800003-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Dunbar RL, Bloedon LT, Duff D, et al. Daily oral administration of the apolipoprotein A-I mimetic peptide D-4F in patients with coronary heart disease or equivalent risk improves high-density lipoprotein anti-inflammatory function. J Am Coll Cardiol. 2007;49(SupplA):366A. Abstract 1014–123. [Google Scholar]
  • 37.Watson CE, Weissbach N, Kjems L, et al. Treatment of patients with cardiovascular disease with L-4F, an apoA-1 mimetic, did not improve select biomarkers of HDL function. J Lipid Res. 2011;52:361–373. doi: 10.1194/jlr.M011098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38**.Chattopadhyay A, Navab M, Hough G, et al. A novel approach to oral apoA-I mimetic therapy. J Lipid Res. 2013;54:995–1010. doi: 10.1194/jlr.M033555. This paper describes a novel approach to the production and administration of apoA-I mimetic peptides and provides evidence that these peptides work in the small intestine by reducing the levels of unsaturated lysophosphatidic acid in the small intestine. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39*.Getz GS, Reardon CA. ApoA-I mimetics: tomatoes to the rescue. J Lipid Res. 2013;54:878–880. doi: 10.1194/jlr.E037572. This paper is the Commentary accompanying reference #38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40**.Navab M, Hough G, Buga GM, et al. Transgenic 6F tomatoes act on the small intestine to prevent systemic inflammation and dyslipidemia caused by Western diet and intestinally derived lysophosphatidic acid. J Lipid Res. 2013;54:3403–3418. doi: 10.1194/jlr.M042051. This manuscript provides direct evidence that increasing the levels of unsaturated (but not saturated) lysophosphatidic acid in the small intestine can cause dyslipidemia and systemic inflammation similar to that seen on feeding LDLR null mice a Western diet. Furthermore, this paper adds to the evidence that the mechanism of action of apoA-I mimetic peptides relates to their ability to reduce the levels of bioactive lipids in the small intestine. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41*.Remaley AT. Tomatoes, lysophosphatidic acid, and the small intestine: new pieces in the puzzle of apolipoprotein mimetic peptides? J Lipid Res. 2013;54:3223–3226. doi: 10.1194/jlr.E045054. This paper is the Commentary that accompanied reference #40. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES