Top Banner
Effect of Macrophage-Derived Apolipoprotein E on Established Atherosclerosis in Apolipoprotein E–Deficient Mice Weibin Shi, Xuping Wang, Nicholas J. Wang, William H. McBride, Aldons J. Lusis Abstract—Apolipoprotein E– deficient (apoE 2/2 ) mice have hyperlipidemia and develop spontaneous atherosclerosis in a time-dependent manner. Although macrophage-derived apoE has been shown to prevent the development of atherosclerosis in apoE 2/2 mice, whether it would induce regression of established atherosclerosis is unknown. To determine this, 8-week-old apoE 2/2 mice were transplanted with apoE 1/1 bone marrow. Four weeks after transplanta- tion, when plasma cholesterol levels had reached normal levels, a group of mice (n512) were killed and their aortic lesions were measured and used as a baseline to judge regression. Twelve and 20 weeks after transplantation, aortic lesion areas of the mice were 934062184 mm 2 (mean6SEM, n58) and 12 21161433 mm 2 (n59), respectively, values not significantly different from the lesion areas of the baseline mice (12 34762487 mm 2 ;n512, P.0.05). In contrast, apoE 2/2 mice reconstituted with apoE 2/2 bone marrow developed severe atherosclerotic lesions (453 036629 767 mm 2 , n57) 20 weeks after transplantation. These data suggest that macrophage-derived apoE was insufficient to induce significant regression of established atherosclerotic lesions in apoE 2/2 mice, although it was sufficient to eliminate hypercholesterolemia and prevent progression of aortic lesions. (Arterioscler Thromb Vasc Biol. 2000;20:2261-2266.) Key Words: atherosclerosis n macrophages n apolipoprotein E n regression n bone marrow transplantation P revious studies have provided evidence that atheroscle- rosis can undergo regression in both humans and animal models, 1–4 but results have varied greatly, depending on the animal models used or the population investigated. 5–7 Mice are increasingly used in the study of atherosclerosis, and certain mice such as apoE-deficient (apoE 2/2 ) mice develop all phases of lesions found in humans. 8,9 Recently, several studies have reported that atherosclerotic lesions of mice can undergo considerable regression. 10 –16 ApoE is a 34-kD glycoprotein that plays an important role in lipoprotein metabolism. 17 It mediates uptake and degrada- tion of chylomicron and VLDL remnants by acting as a ligand for the LDL receptor and the LDL receptor–related protein. 17,18 Although the vast majority of plasma apoE is derived from the liver, 19 –22 apoE is also synthesized by macrophages in various organs. 23,24 ApoE deficiency results in severe hypercholesterolemia and diffuse atherosclerotic disease in humans 25 and in gene-targeted mice. 8,9 Targeted mice develop foam cell–rich fatty streaks in the aortic sinus and proximal aorta by the age of 3 months, and after 5 months, fibrous lesions are present. 26,27 Because apoE is synthesized by monocytes/macrophages 23,24 but not by gran- ulocytes and lymphocytes, 28 bone marrow transplantation (BMT) has been used to examine the role of macrophage-de- rived apoE in atherosclerosis in vivo. Recent BMT studies have shown that macrophage-derived apoE results in the normalization of serum cholesterol levels and prevents the development of atherosclerosis in apoE 2/2 mice. 29 –31 How- ever, it is unknown whether macrophage-derived apoE can induce regression of established atherosclerotic lesions. The purpose of the present study was to determine the effect of macrophage-derived apoE on established atherosclerosis in apoE 2/2 mice by BMT. Methods Mice ApoE 2/2 C57BL/6J mice and wild-type C57BL/6J mice were pur- chased from the Jackson Laboratory, Bar Harbor, Me. Mice were fed with a regular chow diet and maintained in a temperature-controlled room with a 12-hour light/dark cycle. All procedures were in accordance with current National Institutes of Health guidelines and were approved by the UCLA Animal Research Committee. Bone Marrow Transplantation Female recipient apoE 2/2 mice were lethally irradiated with a dose of 1000 rads from a cobalt source. Bone marrow cells were harvested by flushing the femurs and tibias of male donor mice with Dulbec- co’s modified Eagle’s medium containing 10% fetal bovine serum and 5 U/mL heparin. Red blood cells were lysed with ACK buffer (150 mmol/L NH 4 Cl, 61 mmol/L KHCO 3 , and 1 mmol/L Na 2 EDTA, pH 7.3). The remaining cells were washed and suspended in Dulbecco’s modified Eagle’s medium with 1% bovine albumin. Received June 8, 2000; revision accepted July 17, 2000. From the Department of Medicine (W.S., X.W., N.J.W., A.J.L.), Department of Microbiology and Molecular Genetics, and the Department of Radiation Oncology (W.H.M.), School of Medicine, University of California, Los Angeles. Correspondence to Aldons J. Lusis, Department of Medicine, UCLA School of Medicine, 47-123 CHS, Los Angeles, CA 90095-1679. E-mail [email protected] © 2000 American Heart Association, Inc. Arterioscler Thromb Vasc Biol. is available at http://www.atvbaha.org 2261 by guest on August 25, 2015 http://atvb.ahajournals.org/ Downloaded from
7

Effect of macrophage-derived apolipoprotein E on hyperlipidemia and atherosclerosis of LDLR-deficient mice

Feb 25, 2023

Download

Documents

Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Effect of macrophage-derived apolipoprotein E on hyperlipidemia and atherosclerosis of LDLR-deficient mice

Effect of Macrophage-Derived Apolipoprotein E onEstablished Atherosclerosis in

Apolipoprotein E–Deficient MiceWeibin Shi, Xuping Wang, Nicholas J. Wang, William H. McBride, Aldons J. Lusis

Abstract—Apolipoprotein E–deficient (apoE2/2) mice have hyperlipidemia and develop spontaneous atherosclerosis in atime-dependent manner. Although macrophage-derived apoE has been shown to prevent the development ofatherosclerosis in apoE2/2 mice, whether it would induce regression of established atherosclerosis is unknown. Todetermine this, 8-week-old apoE2/2 mice were transplanted with apoE1/1 bone marrow. Four weeks after transplanta-tion, when plasma cholesterol levels had reached normal levels, a group of mice (n512) were killed and their aorticlesions were measured and used as a baseline to judge regression. Twelve and 20 weeks after transplantation, aorticlesion areas of the mice were 934062184mm2 (mean6SEM, n58) and 12 21161433mm2 (n59), respectively, valuesnot significantly different from the lesion areas of the baseline mice (12 34762487mm2; n512, P.0.05). In contrast,apoE2/2 mice reconstituted with apoE2/2 bone marrow developed severe atherosclerotic lesions (453 036629 767mm2,n57) 20 weeks after transplantation. These data suggest that macrophage-derived apoE was insufficient to inducesignificant regression of established atherosclerotic lesions in apoE2/2 mice, although it was sufficient to eliminatehypercholesterolemia and prevent progression of aortic lesions.(Arterioscler Thromb Vasc Biol. 2000;20:2261-2266.)

Key Words: atherosclerosisn macrophagesn apolipoprotein En regressionn bone marrow transplantation

Previous studies have provided evidence that atheroscle-rosis can undergo regression in both humans and animal

models,1–4 but results have varied greatly, depending on theanimal models used or the population investigated.5–7 Miceare increasingly used in the study of atherosclerosis, andcertain mice such as apoE-deficient (apoE2/2) mice developall phases of lesions found in humans.8,9 Recently, severalstudies have reported that atherosclerotic lesions of mice canundergo considerable regression.10–16

ApoE is a 34-kD glycoprotein that plays an important rolein lipoprotein metabolism.17 It mediates uptake and degrada-tion of chylomicron and VLDL remnants by acting as aligand for the LDL receptor and the LDL receptor–relatedprotein.17,18 Although the vast majority of plasma apoE isderived from the liver,19–22 apoE is also synthesized bymacrophages in various organs.23,24 ApoE deficiency resultsin severe hypercholesterolemia and diffuse atheroscleroticdisease in humans25 and in gene-targeted mice.8,9 Targetedmice develop foam cell–rich fatty streaks in the aortic sinusand proximal aorta by the age of 3 months, and after 5months, fibrous lesions are present.26,27 Because apoE issynthesized by monocytes/macrophages23,24 but not by gran-ulocytes and lymphocytes,28 bone marrow transplantation(BMT) has been used to examine the role of macrophage-de-rived apoE in atherosclerosis in vivo. Recent BMT studies

have shown that macrophage-derived apoE results in thenormalization of serum cholesterol levels and prevents thedevelopment of atherosclerosis in apoE2/2 mice.29–31 How-ever, it is unknown whether macrophage-derived apoE caninduce regression of established atherosclerotic lesions. Thepurpose of the present study was to determine the effect ofmacrophage-derived apoE on established atherosclerosis inapoE2/2 mice by BMT.

MethodsMiceApoE2/2 C57BL/6J mice and wild-type C57BL/6J mice were pur-chased from the Jackson Laboratory, Bar Harbor, Me. Mice were fedwith a regular chow diet and maintained in a temperature-controlledroom with a 12-hour light/dark cycle. All procedures were inaccordance with current National Institutes of Health guidelines andwere approved by the UCLA Animal Research Committee.

Bone Marrow TransplantationFemale recipient apoE2/2 mice were lethally irradiated with a dose of1000 rads from a cobalt source. Bone marrow cells were harvestedby flushing the femurs and tibias of male donor mice with Dulbec-co’s modified Eagle’s medium containing 10% fetal bovine serumand 5 U/mL heparin. Red blood cells were lysed with ACK buffer(150 mmol/L NH4Cl, 61 mmol/L KHCO3, and 1 mmol/L Na2EDTA,pH 7.3). The remaining cells were washed and suspended inDulbecco’s modified Eagle’s medium with 1% bovine albumin.

Received June 8, 2000; revision accepted July 17, 2000.From the Department of Medicine (W.S., X.W., N.J.W., A.J.L.), Department of Microbiology and Molecular Genetics, and the Department of Radiation

Oncology (W.H.M.), School of Medicine, University of California, Los Angeles.Correspondence to Aldons J. Lusis, Department of Medicine, UCLA School of Medicine, 47-123 CHS, Los Angeles, CA 90095-1679. E-mail

[email protected]© 2000 American Heart Association, Inc.

Arterioscler Thromb Vasc Biol.is available at http://www.atvbaha.org

2261 by guest on August 25, 2015http://atvb.ahajournals.org/Downloaded from

Page 2: Effect of macrophage-derived apolipoprotein E on hyperlipidemia and atherosclerosis of LDLR-deficient mice

Each recipient mouse was injected with 107 bone marrow cells in 0.3mL through the tail vein.

Experimental ProtocolsAt 8 weeks of age, female apoE2/2 mice (n529) were reconstitutedwith bone marrow cells from wild-type male C57BL/6J mice(apoE1/13apoE2/2) and maintained on a chow diet. Four weeks aftertransplantation, a group of mice (n512) were killed and served as thebaseline group by which to judge regression of atheroscleroticlesions. The remaining mice were killed at 12 (n58) or 20 (n59)weeks after transplantation for lesion analyses. In addition, a groupof age-matched, female apoE2/2 mice were transplanted with bonemarrow from male apoE2/2 mice (apoE2/23apoE2/2) and main-tained on a chow diet for 20 weeks (n57).

Western Blot Analysis for ApoEThe presence of apoE in plasma was determined by Western blotanalysis. In brief, 1mL of plasma was separated by electrophoresison 12% SDS polyacrylamide gels and electrophoretically transferredto nitrocellulose membranes. The membranes were incubated with apolyclonal rabbit anti-mouse apoE antibody (BioDesign Interna-tional) for 1 hour and then incubated for 0.5 hour with a horseradishperoxidase–conjugated anti-rabbit secondary antibody. The signalswere detected by the enhanced chemiluminescent detection methodaccording to the manufacturer’s instructions (ECL Western blotting,Amersham).

DNA Preparation and PCR ofMale-Specific SequencesOvernight-fasted mice were bled from the retro-orbital vein underisoflurane anesthesia. After centrifugation, the plasma was collectedand used for lipid analyses as indicated below. The blood cell pelletwas lysed in ACK buffer to remove red blood cells. DNA wasprepared by adding polymerase chain reaction (PCR) solution(10 mmol/L Tris, pH 8.0; 2.5 mmol/L MgCl2; 1% Tween-20; and 0.4mg/mL proteinase K) to each leukocyte pellet and incubating theresulting mixture at 60°C for 2 hours followed by a 95°C incubationfor 20 minutes. PCR was performed to amplify a 250-bp sequence ofthe Y chromosome.32 The upstream primer was 59-GAG GGC CATGTC AAG CGC CCC ATG AATG-39and the downstream primerwas 59-AGA CAC TGT GAA ATC GGG AGG CT-39. The cyclingconditions were denaturing for 1 minute at 94°C, annealing for 1minute at 62°C, and extension for 1 minute at 72°C.

Aortic Lesion AnalysisMethods for the quantification of atherosclerotic lesions in the aortawere done as previously reported.33 In brief, the heart and proximalaorta were excised and embedded in OCT compound. Serial 10-mm-thick cryosections from the middle portion of the ventricle to theaortic arch were collected and mounted on poly-D-lysine–coatedslides. In the region from the appearance to the disappearance of theaortic valves, every other section was collected. In all other regions,every fifth section was collected. Sections were stained with oil redO and hematoxylin, counterstained with fast green, and examined bylight microscopy.

Immunohistochemical Analyses ofAtherosclerotic LesionsImmunohistochemical analyses of atherosclerotic lesions in theaortic root were performed as previously described.33 In brief,10-mm-thick cryosections were fixed in acetone and incubated witha rabbit polyclonal antibody to mouse apoE or a rat monoclonalantibody to mouse macrophages, MOMA-2 (Accurate Chemicals),followed by incubation with biotinylated anti-rabbit or anti-ratsecondary antibodies. Signals were detected with peroxidase chro-mogen kits (Vector Laboratories). We used an FITC-labeled poly-clonal antibody to human smooth muscle cell actin (Sigma) to detectsmooth muscle cells in the lesions.

Plasma Lipid MeasurementsEnzymatic assays for total cholesterol, HDL cholesterol, and triglyc-eride were performed in 96-well plates on a Biomek 2000 automatedlaboratory workstation (Beckman Instruments, Inc) as described.34

Measurements on plasma samples were performed in triplicate withknown control samples on each plate to ensure accuracy.

Statistical AnalysisPlasma lipid levels were expressed as mean6SEM, with n indicatingthe number of mice. Atherosclerotic lesion areas were expressed asvalues of individual mice. ANOVA was used to compare differencesin atherosclerotic lesions and lipid levels among different groups ofmice over time. Differences were considered statistically significantat P,0.05.

ResultsReconstitution of Recipient Bone MarrowApoE was detected by Western blot analysis in the plasma ofapoE2/2 mice reconstituted with wild-type bone marrow asearly as 2 weeks after transplantation (Figure 1A). In contrast,apoE was absent in apoE2/2 mice reconstituted with apoE2/2

bone marrow. Moreover, because male mice containing XYchromosomes were used as donors for the female recipients,we designed primers to genotype a segment of the Ychromosome. As shown in Figure 1B, 2 weeks after trans-plantation, the Y chromosome was detected by PCR inperipheral leukocytes of recipient mice.

Effect of BMT on Plasma LipidsReconstitution of apoE2/2 mice with wild-type bone marrowresulted in dramatic changes in plasma cholesterol andtriglyceride levels (Figure 2). Two weeks after transplanta-tion, plasma total cholesterol and triglyceride levels weresignificantly reduced and HDL cholesterol levels increased(P,0.05). By 4 weeks after transplantation, plasma totalcholesterol, triglyceride, and HDL cholesterol levels hadreached normal levels. In contrast, in apoE2/23apoE2/2

mice, plasma total cholesterol levels gradually increased,from 334611 mg/L (mean6SEM) before transplantation to556624 mg/L 20 weeks after transplantation. There was asignificant decrease in triglyceride levels 2 weeks after

Figure 1. A, Western blot analysis of plasma apoE in apoE2/2

mice 2 weeks after BMT. One microliter of undiluted plasmawas electrophoresed on 12% SDS gels, transferred to nitrocel-lulose membranes, and probed with a polyclonal antibody tomouse apoE. Lanes 1 through 6, mice transplanted with wild-type bone marrow; lanes 7 through 9, mice transplanted withapoE2/2 bone marrow; lane 10, positive control (plasma fromwild-type mice). B, PCR of DNA extracted from the peripheralblood of female apoE2/2 mice 2 weeks after transplantation toamplify a 250-bp sequence of the Y chromosome. Lane 1,100-bp ladder; lanes 2 through 8, samples from transplantedmice; lane 9, negative control (female mouse DNA); lane 10,positive control (male mouse DNA).

2262 Arterioscler Thromb Vasc Biol. October 2000

by guest on August 25, 2015http://atvb.ahajournals.org/Downloaded from

Page 3: Effect of macrophage-derived apolipoprotein E on hyperlipidemia and atherosclerosis of LDLR-deficient mice

transplantation (P,0.05). Plasma HDL cholesterol levelswere not significantly altered after transplantation.

Effect on Aortic Atherosclerotic Lesion AreasThe size of atherosclerotic lesions at the aortic root wasquantified by light microscopy. The apoE1/13apoE2/2 micethat were killed 4 weeks after transplantation had an averagelesion area per section of 12 34762487mm2 (mean6SEM,n512; Figure 3). Twelve and 20 weeks after transplantation,the average lesion areas of apoE1/13apoE2/2 mice were934062185mm2 (n58) and 12 21161433mm2 (n59), re-spectively. Compared with mice that were killed 4 weeksafter transplantation, aortic lesion areas did not show asignificant increase or decrease in mice that were killed at 12and 20 weeks after transplantation (P.0.05). In contrast,apoE2/23apoE2/2 mice developed severe atherosclerosis 20weeks after transplantation, with an average lesion area of453 036629 767mm2 per section (n57).

Effect on Morphology of Atherosclerotic LesionsIn mice that were killed 4 weeks after transplantation, aorticlesions consisted primarily of macrophage-derived foam cells(Figure 4). Smooth muscle cells were undetectable in the

lesions, and there were no fibrous caps. In mice that werekilled 12 weeks and 20 weeks after transplantation, aorticlesions were flatter and had developed thin, fibrous caps inabout half of the mice. Macrophage-derived foam cells werestill the main cellular component of the lesions. Smoothmuscle cells were observed in the fibrous caps. In contrast,mice reconstituted with apoE2/2 bone marrow developedadvanced lesions containing numerous smooth muscle cells,calcification, and necrotic areas.

ApoE Expression in Atherosclerotic LesionsImmunohistochemistry analysis showed that apoE was abun-dantly expressed in atherosclerotic lesions of the apoE2/2

mice reconstituted with wild-type bone marrow (Figure 5). Incontrast, apoE was not detected in lesions of those apoE2/2

mice reconstituted with apoE2/2 bone marrow.

DiscussionThe principal aim of the present study was to evaluatewhether macrophage-derived apoE was sufficient to inducesignificant regression of established atherosclerotic lesions inapoE-deficient mice by BMT. Wild-type and apoE-deficientmice were used as bone marrow donors for apoE-deficientmice. We found that repopulation of cells in BMT-treatedmice with normal hematopoietic cells eliminated hypercho-lesterolemia and prevented progression of the lesions but didnot induce significant regression of established atherosclero-sis in apoE-deficient mice.

BMT leads to the replacement of recipient tissue macro-phages by macrophages of donor origin.35 Bone marrow cellsaccumulate in the liver, spleen, and bone marrow severalhours after injection.36,37 By 4 weeks after transplantation,.95% of macrophages in the bone marrow were of donor

Figure 2. Effects of BMT on plasma total cholesterol (A), HDLcholesterol (B), and triglyceride (C) levels in apoE2/2 recipientmice. Plasma lipid levels were measured before and 2, 4, 12,and 20 weeks after transplantation. ApoE2/2 mice were trans-planted with bone marrow from either apoE1/1 or apoE2/2 mice.Values are mean6SE of 7 to 12 mice. *P,0.05 vsapoE2/23apoE2/2.

Figure 3. Atherosclerotic lesion areas in cross sections of aorticroot from apoE2/2 mice transplanted with apoE1/1 or apoE2/2

bone marrow. Each point represents a mean lesion area persection from 1 mouse. Mice were fed a chow diet during theexperiment and were killed at 4, 12, and 20 weeks after trans-plantation. ApoE2/232/2 represents mice transplanted withapoE2/2 bone marrow and killed 20 weeks after transplantation.

Shi et al ApoE and Atherosclerosis in Mice 2263

by guest on August 25, 2015http://atvb.ahajournals.org/Downloaded from

Page 4: Effect of macrophage-derived apolipoprotein E on hyperlipidemia and atherosclerosis of LDLR-deficient mice

origin.29 In the present study, we observed that apoE waspresent in the peripheral circulation as early as 2 weeks aftertransplantation, and by 4 weeks, macrophage-derived apoEwas sufficient to normalize plasma lipid levels of apoE-deficient recipient mice. As in previous studies,38,39 we usedmale mice as bone marrow donors for female recipients sothat engraftment of bone marrow could be verified bygenotyping male-specific makers of the Y chromosome.Because donor and recipient mice were derived from thesame inbred strain, graft-versus-host disease would not occur.Moreover, all recipient mice were healthy throughout theexperiment.

Although macrophages produce only a small percentage('10%) of plasma apoE, this amount is sufficient to reversehypercholesterolemia and elevate plasma HDL levels.29,30

Our present findings are consistent with this observation. Thefinding that BMT induced a decrease of plasma triglyceridelevels in apoE1/13apoE2/2 mice is consistent with that of aprevious study.31 The temporary decrease at 2 weeks wasprobably caused by the transplantation procedure, because itwas also observed in apoE2/23apoE2/2 mice.

Previous studies29–31 and our present study have indicatedthat after transplantation, a period of 4 weeks is required formacrophage-derived apoE to achieve its full therapeutic

effect in apoE-deficient recipients. During this period, asperipheral tissues are reconstituted with apoE-expressingmacrophages, plasma cholesterol levels gradually fall butatherosclerotic lesions continue to progress. We observed asignificant increase in atherosclerotic lesion size, from26136781 mm2 at the time of transplantation to12 34762487mm2 4 weeks after transplantation. Therefore,selecting atherosclerotic lesions at 4 weeks after transplanta-tion as the baseline seems an appropriate standard by whichto judge regression in apoE-deficient mice. In the presentexperiment, apoE-deficient mice received transplantation at 8weeks of age, and by the time their bone marrow wasreplaced with donor marrow, they were 12 weeks of age.Nakashima et al27 reported that apoE-deficient mice of thisage develop fatty streak lesions. Indeed, we observed thatatherosclerotic lesions of these mice consisted primarily ofmacrophage-derived foam cells and that smooth muscle cellsand fibrous caps were absent in the lesions.

One important finding of the present study is that 16 weeksafter plasma lipid levels were normalized, the size of theatherosclerotic lesions was not significantly reduced in apoE-deficient mice. This finding is consistent with the notion thatthe regression of atherosclerotic lesions is a slow process.Indeed, in the rhesus monkey model, Tucker et al5 did not

Figure 4. Representative light photomi-crographs of aortic atherosclerotic lesionsin apoE2/2 mice transplanted withapoE1/1 or apoE2/2 bone marrow. Sec-tions were stained with oil red O andhematoxylin (A, B, C), an anti-mousemacrophage antibody MOMA-2 (D, E, F),or an FITC-labeled antibody to smoothmuscle cell actin (G, H, I). The first andsecond columns represent lesions frommice transplanted with apoE1/1 bonemarrow and killed at 4 and 20 weeks,respectively, after transplantation. Thethird column represents lesions from micetransplanted with apoE2/2 bone marrowand killed at 20 weeks after transplanta-tion. Original magnification, 325.

Figure 5. Immunohistochemical analysis ofapoE expression in aortic atheroscleroticlesions of apoE2/2 mice transplanted withapoE2/2 (A) or wild-type (B) bone marrow.Sections were stained with a polyclonal rabbitanti-mouse apo E antibody. Original magnifi-cation, 340.

2264 Arterioscler Thromb Vasc Biol. October 2000

by guest on August 25, 2015http://atvb.ahajournals.org/Downloaded from

Page 5: Effect of macrophage-derived apolipoprotein E on hyperlipidemia and atherosclerosis of LDLR-deficient mice

find definite evidence of regression after 4 months on alow-fat diet. Kokatnur et al40 reported that experimentalatherosclerosis in rhesus monkeys showed evidence of re-gression only after treatment with a low-fat diet for 64 weeks.However, Tsukamoto et al16 recently reported that liver-directed gene transfer and hepatic expression of humanapoE3 in chow-fed, apoE-deficient mice resulted in an almostcomplete regression of fatty streaks within 6 weeks, whereasexpression of human apoE4 reduced cholesterol levels to thesame extent as apoE3 but did not induce significant regres-sion. That study suggests that effects beyond the reduction ofplasma cholesterol levels are required to induce regression.Mouse apoE is similar to human apoE3 in term of the 2polymorphic amino acids.41 The reasons for the discrepancybetween the data of Tsukamoto et al16 and ours are unclear.One possible explanation is that gene transfer resulted inmore apoE production than did BMT. Indeed, Desurmont etal11 observed that regression of fatty streak lesions in apoE2/2

mice 6 months after injection of the adenovirus encodinghuman apoE cDNA was dependent on plasma apoEconcentration.

Hyperlipidemia plays an important role in the progressionof atherosclerosis. However, elimination of hyperlipidemiaalone seems insufficient to induce regression of atheroscle-rosis. Indeed, our failure to observe a significant reduction ofatherosclerotic lesions 16 weeks after normalization ofplasma lipid levels suggests that treatments other than nor-malizing plasma lipid levels are necessary to induce signifi-cant regression of atherosclerosis.

AcknowledgmentsThis work was supported by National Institutes of Health grantHL-30568. The authors thank Yishou Shi for technical help.

References1. Armstrong ML, Warner ED, Connor WE. Regression of coronary ath-

erosclerosis in rhesus monkeys.Circ Res. 1970;27:59–67.2. Rossouw JE. Lipid-lowering interventions in angiographic trials.Am J

Cardiol. 1995;76:86C–92C.3. Sacks FM, Gibson CM, Rosner B, Pasternak RC, Stone PH. The influence

of pretreatment low density lipoprotein cholesterol concentrations on theeffect of hypocholesterolemic therapy on coronary atherosclerosis inangiographic trials: Harvard Atherosclerosis Reversibility ProjectResearch Group.Am J Cardiol.1995;76:78C–85C.

4. Wagner WD, Clarkson TB. Effect on regression potential of atheroscle-rosis produced by intermittent or continuous hypercholesterolemia.Ath-erosclerosis. 1977;27:369–381.

5. Tucker CF, Catsulis C, Strong JP, Eggen DA. Regression of early cho-lesterol-induced aortic lesions in rhesus monkeys.Am J Pathol. 1971;65:493–514.

6. Stone PH, Gibson CM, Pasternak RC, McManus K, Diaz L, BoucherT, Spears R, Sandor T, Rosner B, Sacks FM. Natural history ofcoronary atherosclerosis using quantitative angiography in men, andimplications for clinical trials of coronary regression: the HarvardAtherosclerosis Reversibility Project Study Group.Am J Cardiol.1993;71:766 –772.

7. Blankenhorn DH, Hodis HN. George Lyman Duff Memorial Lecture:arterial imaging and atherosclerosis reversal.Arterioscler Thromb. 1994;14:177–192.

8. Plump AS, Smith JD, Hayek T, Aalto-Seta¨la K, Walsh A, Verstuyft JG,Rubin EM, Breslow JL. Severe hypercholesterolemia and atherosclerosisin apolipoprotein E-deficient mice created by homologous recombinationin ES cells.Cell. 1992;71:343–353.

9. Zhang SH, Reddick RL, Piedrahita JA, Maeda N. Spontaneous hyper-cholesterolemia and arterial lesions in mice lacking apolipoprotein E.Science. 1992;258:468–471.

10. Linton MF, Babaev VR, Gleaves LA, Atkinson JB, Fazio S. Regressionof atherosclerosis in apolipoprotein E deficient mice after bone marrowtransplantation.Circulation. 1996;94(suppl I):I-519. Abstract.

11. Desurmont C, Caillaud JM, Emmanuel F, Benoit P, Fruchart JC, CastroG, Branellec D, Heard JM, Duverger N. Complete atherosclerosisregression after human apoE gene transfer in apoE-deficient/nude mice.Arterioscler Thromb Vasc Biol. 2000;20:435–442.

12. Tangirala RK, Tsukamoto K, Chun SH, Usher D, Pure E, Rader DJ.Regression of atherosclerosis induced by liver-directed gene transfer ofapolipoprotein A-I in mice.Circulation. 1999;100:1816–1822.

13. Murayama T, Yokode M, Kataoka H, Imabayashi T, Yoshida H, Sano H,Nishikawa S, Nishikawa S, Kita T. Intraperitoneal administration ofanti-c-fmsmonoclonal antibody prevents initial events of atherogenesisbut does not reduce the size of advanced lesions in apolipoproteinE-deficient mice.Circulation. 1999;99:1740–1746.

14. Kozarsky KF, Donahee MH, Glick JM, Krieger M, Rader DJ. Genetransfer and hepatic overexpression of the HDL receptor SR-BI reducesatherosclerosis in the cholesterol-fed LDL receptor–deficient mouse.Arterioscler Thromb Vasc Biol. 2000;20:721–727.

15. Gijbels MJ, van der Cammen M, van der Laan LJ, Emeis JJ, Havekes LM,Hofker MH, Kraal G. Progression and regression of atherosclerosis inAPOE3-Leiden transgenic mice: an immunohistochemical study.Athero-sclerosis. 1999;143:15–25.

16. Tsukamoto K, Rajendra Tangirala R, Chun SH, Pure E, Rader DJ. Rapidregression of atherosclerosis induced by liver-directed gene transfer ofapoE in apoE-deficient mice.Arterioscler Thromb Vasc Biol. 1999;19:2162–2170.

17. Mahley RW. Apolipoprotein E: cholesterol transport protein withexpanding role in cell biology.Science. 1988;240:622–630.

18. Beisiegel U. Receptors for triglyceride-rich lipoproteins and their role inlipoprotein metabolism.Curr Opin Lipidol. 1995;6:117–122.

19. Zannis VI, Cole FS, Jackson CL, Kurnit DM, Karathanasis SK. Dis-tribution of apolipoprotein A-I, C-II, C-III, and E mRNA in fetalhuman tissues: time-dependent induction of apolipoprotein E mRNAby cultures of human monocyte-macrophages.Biochemistry. 1985;24:4450 – 4455.

20. Williams DL, Dawson PA, Newman TC, Rudel LL. Synthesis of apoli-poprotein E by peripheral tissues: potential functions in reverse choles-terol transport and cellular cholesterol metabolism.Ann N Y Acad Sci.1985;454:222–229.

21. Driscoll DM, Getz GS. Extrahepatic synthesis of apolipoprotein E.JLipid Res. 1984;25:1368–1379.

22. Linton MF, Gish R, Hubl ST, Butler E, Esquivel C, Bry WI, BoylesJK, Wardell MR, Young SG. Phenotypes of apolipoprotein B andapolipoprotein E after liver transplantation.J Clin Invest. 1991;88:270 –281.

23. Basu SK, Brown MS, Ho YK, Havel RJ, Goldstein JL. Mouse macro-phages synthesize and secrete a protein resembling apolipoprotein E.Proc Natl Acad Sci U S A. 1981;78:7545–7549.

24. Basu SK, Ho YK, Brown MS, Bilheimer DW, Anderson RG, GoldsteinJL. Biochemical and genetic studies of the apoprotein E secreted bymouse macrophages and human monocytes. J Biol Chem. 1982;257:9788–9795.

25. Ghiselli G, Schaefer EJ, Gascon P, Brewer HB Jr. Type III hyperlipo-proteinemia associated with apolipoprotein E deficiency.Science. 1981;214:1239–1241.

26. Reddick RL, Zhang SH, Maeda N. Atherosclerosis in mice lacking apo E:evaluation of lesional development and progression.Arterioscler Thromb.1994;14:141–147.

27. Nakashima Y, Plump AS, Raines EW, Breslow JL, Ross R. ApoE-deficient mice develop lesions of all phases of atherosclerosis throughoutthe arterial tree.Arterioscler Thromb. 1994;14:133–140.

28. Lin CT, Xu YF, Wu JY, Chan L. Immunoreactive apolipoprotein E is awidely distributed cellular protein: immunohistochemical localization ofapolipoprotein E in baboon tissues.J Clin Invest. 1986;78:947–958.

29. Linton MF, Atkinson JB, Fazio S. Prevention of atherosclerosis in apo-lipoprotein E-deficient mice by bone marrow transplantation.Science.1995;267:1034–1037.

30. Boisvert WA, Spangenberg J, Curtiss LK. Treatment of severe hypercho-lesterolemia in apolipoprotein E-deficient mice by bone marrow trans-plantation.J Clin Invest. 1995;96:1118–1124.

31. Van Eck M, Herijgers N, Yates J, Pearce NJ, Hoogerbrugge PM, GrootPH, Van Berkel TJ. Bone marrow transplantation in apolipoproteinE-deficient mice: effect of apoE gene dosage on serum lipid concen-

Shi et al ApoE and Atherosclerosis in Mice 2265

by guest on August 25, 2015http://atvb.ahajournals.org/Downloaded from

Page 6: Effect of macrophage-derived apolipoprotein E on hyperlipidemia and atherosclerosis of LDLR-deficient mice

trations,b-VLDL catabolism, and atherosclerosis.Arterioscler ThrombVas Biol. 1997;17:3117–3126.

32. Gubbay J, Collignon J, Koopman P, Capel B, Economou A, MunsterbergA, Vivian N, Goodfellow P, Lovell-Badge R. A gene mapping to thesex-determining region of the mouse Y chromosome is a member of anovel family of embryonically expressed genes.Nature. 1990:346:245–250.

33. Qiao JH, Xie PZ, Fishbein MC, Kreuzer J, Drake TA, Demer LL, LusisAJ. Pathology of atheromatous lesions in inbred and genetically engi-neered mice: genetic determination of arterial calcification.ArteriosclerThromb. 1994;14:1480–1497.

34. Hedrick CC, Castellani LW, Warden CH, Puppione DL, LusisAJ. Influence of mouse apolipoprotein A-II on plasma lipoproteins intransgenic mice.J Biol Chem. 1993;268:20676–20682.

35. Longo DL, Davis ML. Early appearance of donor-type antigen-presentingcells in the thymuses of 1200 R radiation-induced bone marrow chimerascorrelates with self-recognition of donor I region gene products.J Immunol. 1983;130:2525–2527.

36. Takezawa RI, Watanabe Y, Akaike T. Direct evidence of macrophagedifferentiation from bone marrow cells in the liver: a possible origin ofKupffer cells.J Biochem. 1995;118:1175–1183.

37. Hendrikx PJ, Martens CM, Hagenbeek A, Keij JF, Visser JW. Homing offluorescently labeled murine hematopoietic stem cells.Exp Hematol.1996;24:129–140.

38. Blomberg M, Rao S, Reilly J, Tiarks C, Peters S, Kittler E, QuesenberryP. Repetitive bone marrow transplantation in nonmyeloablated recipients.Exp Hematol. 1998;26:320–324.

39. Eglitis MA, Mezey E. Hematopoietic cells differentiate into bothmicroglia and macroglia in the brains of adult mice.Proc Natl Acad SciU S A. 1997;94:4080–4085.

40. Kokatnur MG, Malcom GT, Eggen DA, Strong JP. Depletion of aorticfree and ester cholesterol by dietary means in rhesus monkeys with fattystreaks.Atherosclerosis. 1975;21:195–203.

41. Rajavashisth TB, Kaptein JS, Reue KL, Lusis AJ. Evolution of apoli-poprotein E: mouse sequence and evidence for an 11-nucleotide ancestralunit. Proc Natl Acad Sci U S A. 1985;82:8085–8089.

2266 Arterioscler Thromb Vasc Biol. October 2000

by guest on August 25, 2015http://atvb.ahajournals.org/Downloaded from

Page 7: Effect of macrophage-derived apolipoprotein E on hyperlipidemia and atherosclerosis of LDLR-deficient mice

Weibin Shi, Xuping Wang, Nicholas J. Wang, William H. McBride and Aldons J. LusisDeficient Mice−Apolipoprotein E

Effect of Macrophage-Derived Apolipoprotein E on Established Atherosclerosis in

Print ISSN: 1079-5642. Online ISSN: 1524-4636 Copyright © 2000 American Heart Association, Inc. All rights reserved.

Greenville Avenue, Dallas, TX 75231is published by the American Heart Association, 7272Arteriosclerosis, Thrombosis, and Vascular Biology

doi: 10.1161/01.ATV.20.10.22612000;20:2261-2266Arterioscler Thromb Vasc Biol. 

http://atvb.ahajournals.org/content/20/10/2261World Wide Web at:

The online version of this article, along with updated information and services, is located on the

  http://atvb.ahajournals.org//subscriptions/

at: is onlineArteriosclerosis, Thrombosis, and Vascular Biology Information about subscribing to Subscriptions:

  http://www.lww.com/reprints

Information about reprints can be found online at: Reprints: 

document. Question and AnswerPermissions and Rightspage under Services. Further information about this process is available in the

which permission is being requested is located, click Request Permissions in the middle column of the WebCopyright Clearance Center, not the Editorial Office. Once the online version of the published article for

can be obtained via RightsLink, a service of theArteriosclerosis, Thrombosis, and Vascular Biologyin Requests for permissions to reproduce figures, tables, or portions of articles originally publishedPermissions:

by guest on August 25, 2015http://atvb.ahajournals.org/Downloaded from