smooth muscle cells healing atherosclerotic plaque disruptions

19
Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions Are of Local, Not Blood, Origin in Apolipoprotein E Knockout Mice Jacob F. Bentzon, MD, PhD; Claus S. Sondergaard, MSc; Moustapha Kassem, MD, PhD; Erling Falk, MD, PhD Background—Signs of preceding episodes of plaque rupture and smooth muscle cell (SMC)–mediated healing are common in atherosclerotic plaques, but the source of the healing SMCs is unknown. Recent studies suggest that activated platelets adhering to sites of injury recruit neointimal SMCs from circulating bone marrow– derived progenitor cells. Here, we analyzed the contribution of this mechanism to plaque healing after spontaneous and mechanical plaque disruption in apolipoprotein E knockout (apoE / ) mice. Methods and Results—To determine the origin of SMCs after spontaneous plaque disruption, irradiated 18-month-old apoE / mice were reconstituted with bone marrow cells from enhanced green fluorescent protein (eGFP) transgenic apoE / mice and examined when they died up to 9 months later. Plaque hemorrhage, indicating previous plaque disruption, was widely present, but no bone marrow– derived eGFP SMCs were detected. To examine the origin of healing SMCs in a model that recapitulates more features of human plaque rupture and healing, we developed a mechanical technique that produced consistent plaque disruption, superimposed thrombosis, and SMC-mediated plaque healing in apoE / mice. Mechanical plaque disruption was produced in irradiated apoE / mice reconstituted with eGFP apoE / bone marrow cells and in carotid bifurcations cross-grafted between apoE / and eGFP apoE / mice. Apart from few non– graft-derived SMCs near the anastomosis site in 1 transplanted carotid bifurcation, no SMCs originating from outside the local arterial segment were detected in healed plaques. Conclusions—Healing SMCs after atherosclerotic plaque disruption are derived entirely from the local arterial wall and not circulating progenitor cells in apoE / mice. (Circulation. 2007;116:000-000.) Key Words: atherosclerosis blood cells muscle, smooth thrombosis P laque rupture with superimposed thrombosis is the major cause of heart attack and death worldwide, but the great majority of plaque ruptures are not lethal, and most are clinically silent. 1 In nonlethal cases, plaques are healed by smooth muscle cells (SMCs) that accumulate at the rupture site, where they secrete extracellular matrix rich in glycos- aminoglycans and type III collagen. 2,3 This process restores the integrity of the plaque surface but may cause rapid plaque progression and constrictive remodeling of the arterial wall. 3–5 Serial angiography has shown that many coronary artery stenoses develop in a phasic rather than a linear manner, 6 and the frequent finding of healed rupture sites in plaques causing stenosis indicates that SMC-mediated plaque healing may be the underlying mechanism. 3,5 Clinical Perspective p ●●● Despite their clinical relevance, the mechanisms governing plaque healing, including the source of the healing SMCs, have been little explored. A longstanding but sparsely docu- mented theory has considered healing to be carried out by local plaque SMCs that are activated by growth factors (eg, platelet-derived growth factor-BB and transforming growth factor-) and mitogens (eg, thrombin) released or generated during superimposed thrombosis. 7 Curiously, however, the proliferative response of local plaque SMCs in ruptured human atherosclerotic plaques has been found to be mod- est. 5,8 Moreover, SMCs in advanced plaques show numerous features of cellular senescence both in culture and in vivo. 9 Received June 18, 2007; accepted August 28, 2007. From the Department of Cardiology, Aarhus University Hospital (Skejby), and Institute of Clinical Medicine, University of Aarhus (J.F.B., E.F.), Aarhus; Department of Molecular Biology, University of Aarhus, Aarhus (C.S.S.); and Department of Endocrinology, Odense University Hospital, Odense (M.K.), Denmark. The online-only Data Supplement, which contains an expanded Methods section, a movie, and Figures I through VI, is available at http://circ.ahajournals.org/cgi/content/full/CIRCULATIONAHA.10.1161/107.722355. Correspondence to Jacob Fog Bentzon, MD, PhD, Department of Cardiology, Research Unit, Aarhus University Hospital, Brendstrupgaardsvej, 8200 Aarhus N, Denmark. E-mail [email protected] © 2007 American Heart Association, Inc. Circulation is available at http://circ.ahajournals.org DOI: 10.1161/CIRCULATIONAHA.107.722355 1 by guest on February 18, 2018 http://circ.ahajournals.org/ Downloaded from by guest on February 18, 2018 http://circ.ahajournals.org/ Downloaded from by guest on February 18, 2018 http://circ.ahajournals.org/ Downloaded from by guest on February 18, 2018 http://circ.ahajournals.org/ Downloaded from by guest on February 18, 2018 http://circ.ahajournals.org/ Downloaded from by guest on February 18, 2018 http://circ.ahajournals.org/ Downloaded from by guest on February 18, 2018 http://circ.ahajournals.org/ Downloaded from by guest on February 18, 2018 http://circ.ahajournals.org/ Downloaded from by guest on February 18, 2018 http://circ.ahajournals.org/ Downloaded from by guest on February 18, 2018 http://circ.ahajournals.org/ Downloaded from by guest on February 18, 2018 http://circ.ahajournals.org/ Downloaded from

Upload: trinhdien

Post on 05-Jan-2017

218 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

Smooth Muscle Cells Healing Atherosclerotic PlaqueDisruptions Are of Local, Not Blood, Origin in

Apolipoprotein E Knockout MiceJacob F. Bentzon, MD, PhD; Claus S. Sondergaard, MSc;

Moustapha Kassem, MD, PhD; Erling Falk, MD, PhD

Background—Signs of preceding episodes of plaque rupture and smooth muscle cell (SMC)–mediated healing arecommon in atherosclerotic plaques, but the source of the healing SMCs is unknown. Recent studies suggest thatactivated platelets adhering to sites of injury recruit neointimal SMCs from circulating bone marrow–derived progenitorcells. Here, we analyzed the contribution of this mechanism to plaque healing after spontaneous and mechanical plaquedisruption in apolipoprotein E knockout (apoE�/�) mice.

Methods and Results—To determine the origin of SMCs after spontaneous plaque disruption, irradiated 18-month-oldapoE�/� mice were reconstituted with bone marrow cells from enhanced green fluorescent protein (eGFP) transgenicapoE�/� mice and examined when they died up to 9 months later. Plaque hemorrhage, indicating previous plaquedisruption, was widely present, but no bone marrow–derived eGFP� SMCs were detected. To examine the origin ofhealing SMCs in a model that recapitulates more features of human plaque rupture and healing, we developed amechanical technique that produced consistent plaque disruption, superimposed thrombosis, and SMC-mediated plaquehealing in apoE�/� mice. Mechanical plaque disruption was produced in irradiated apoE�/� mice reconstituted witheGFP�apoE�/� bone marrow cells and in carotid bifurcations cross-grafted between apoE�/� and eGFP�apoE�/� mice.Apart from few non–graft-derived SMCs near the anastomosis site in 1 transplanted carotid bifurcation, no SMCsoriginating from outside the local arterial segment were detected in healed plaques.

Conclusions—Healing SMCs after atherosclerotic plaque disruption are derived entirely from the local arterial wall andnot circulating progenitor cells in apoE�/� mice. (Circulation. 2007;116:000-000.)

Key Words: atherosclerosis � blood cells � muscle, smooth � thrombosis

Plaque rupture with superimposed thrombosis is the majorcause of heart attack and death worldwide, but the great

majority of plaque ruptures are not lethal, and most areclinically silent.1 In nonlethal cases, plaques are healed bysmooth muscle cells (SMCs) that accumulate at the rupturesite, where they secrete extracellular matrix rich in glycos-aminoglycans and type III collagen.2,3 This process restoresthe integrity of the plaque surface but may cause rapid plaqueprogression and constrictive remodeling of the arterialwall.3–5 Serial angiography has shown that many coronaryartery stenoses develop in a phasic rather than a linearmanner,6 and the frequent finding of healed rupture sites inplaques causing stenosis indicates that SMC-mediated plaquehealing may be the underlying mechanism.3,5

Clinical Perspective p ●●●

Despite their clinical relevance, the mechanisms governingplaque healing, including the source of the healing SMCs,have been little explored. A longstanding but sparsely docu-mented theory has considered healing to be carried out bylocal plaque SMCs that are activated by growth factors (eg,platelet-derived growth factor-BB and transforming growthfactor-�) and mitogens (eg, thrombin) released or generatedduring superimposed thrombosis.7 Curiously, however, theproliferative response of local plaque SMCs in rupturedhuman atherosclerotic plaques has been found to be mod-est.5,8 Moreover, SMCs in advanced plaques show numerousfeatures of cellular senescence both in culture and in vivo.9

Received June 18, 2007; accepted August 28, 2007.From the Department of Cardiology, Aarhus University Hospital (Skejby), and Institute of Clinical Medicine, University of Aarhus (J.F.B., E.F.),

Aarhus; Department of Molecular Biology, University of Aarhus, Aarhus (C.S.S.); and Department of Endocrinology, Odense University Hospital,Odense (M.K.), Denmark.

The online-only Data Supplement, which contains an expanded Methods section, a movie, and Figures I through VI, is available athttp://circ.ahajournals.org/cgi/content/full/CIRCULATIONAHA.10.1161/107.722355.

Correspondence to Jacob Fog Bentzon, MD, PhD, Department of Cardiology, Research Unit, Aarhus University Hospital, Brendstrupgaardsvej, 8200Aarhus N, Denmark. E-mail [email protected]

© 2007 American Heart Association, Inc.

Circulation is available at http://circ.ahajournals.org DOI: 10.1161/CIRCULATIONAHA.107.722355

1

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

Page 2: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

Observations of Zernecke et al10 recently suggested anexplanation for this paradox. They found that activatedplatelets adhering to sites of arterial injury facilitated therecruitment of neointimal SMCs from circulating bone mar-row (BM)–derived progenitor cells via the expression ofP-selectin and stromal cell–derived factor 1�. The impor-tance of activated platelets for homing of progenitor cells alsohas been supported by other experimental studies,11–13 and theidea is consistent with signs of BM-derived SMCs in humancoronary atherosclerosis, in which subclinical plaque rupturesoccur.14

Recently, we and others have shown that plaque SMCs inuncomplicated plaques in young apolipoprotein E knockout(apoE�/�) mice are recruited entirely from the local arterialwall and that the majority, if not all, descend from SMCs inthe arterial media.15–17 In the present study, we tested thehypothesis that plaque disruption with superimposed throm-bosis is the critical event that facilitates smooth muscleprogenitor cell recruitment to atherosclerotic plaques. First,we studied whether BM-derived SMCs are present in plaqueswith spontaneous plaque hemorrhage (indicating recentplaque disruption) in old apoE�/� mice. Second, we devel-oped a mechanical model of plaque disruption, superimposedthrombosis, and SMC-mediated healing in apoE�/� mice anddetermined the origin of SMCs in healed plaques.

MethodsFor an expanded Methods section, see the online-only DataSupplement.

AnimalsAll procedures were approved by the Danish Animal ExperimentsInspectorate. ApoE�/� mice (B6.129P2-Apoetm1Unc, Taconic M&B,Ry, Denmark), backcrossed �10 times to C57BL/6 mice, andenhanced green fluorescent protein (eGFP) transgenic C57BL/6 mice[C57BL/6-Tg(ACTB-EGFP)10sb/J, Jackson Laboratories, Bar Har-bor, Maine] were crossed to obtain eGFP�apoE�/� mice (hemizygousfor the eGFP transgene). All mice were fed normal chow.

BM TransplantationApoE�/� mice (male; age, 18 months; n�34) were lethally irradiatedand rescued by tail vein injection of 107 unfractionated BM cellsfrom eGFP�apoE�/� mice (male; age, 2 to 3 months; n�10).15,18

Hematopoietic chimerism was assessed after 4 weeks by flowcytometry of a peripheral blood sample as previously described andfound to be �90% in all cases (see online-only Data SupplementFigure I).15 Nine mice died before flow cytometry was performed.

Long-Term Hematopoietic ChimerasA subgroup of the eGFP�apoE�/� BM3apoE�/� transplanted mice(n�10) and age-matched apoE�/� mice (n�5) were housed untilspontaneous death (Figure 1a). Within 24 hours after death, theanimals were fixed in immersion in 4% phosphate-buffered formal-dehyde for 48 to 72 hours. Cross sections of the carotid bifurcationsand longitudinal sections of the aortic arch, including the brachio-cephalic trunk and the proximal parts of the left common carotid andsubclavian arteries, were prepared for histology (see the detaileddescription online). Sections were taken at least 50 �m apart.

Mechanical Plaque DisruptionThe other eGFP�apoE�/� BM3apoE�/� transplanted mice (n�15) aswell as age-matched apoE�/� (n�4) and eGFP�apoE�/� (n�2)control mice were subjected to mechanical plaque disruption ofcarotid bifurcation (CB) plaques (Figure 1b, top). Briefly, mice were

anesthetized, and the carotid bifurcation plaque most suitable for theprocedure was identified and isolated temporarily between loose 6-0silk ligatures. Then, a 9-0 spatula needle was inserted into thecommon carotid artery �0.5 mm proximal to the targeted plaque,advanced distally through the lumen, and inserted repeatedly into theplaque from the luminal side to produce plaque disruption (see thedetailed description and the online-only Data Supplement Movie).Immediately after the procedure, mice were randomly allocated to 1of 3 groups: killed at 30 minutes, 1 week, or 4 weeks after plaquedisruption. The control mice were all killed after 4 weeks. Serialsections taken at 100-�m intervals throughout the CB plaques wereprepared for histology.

CB TransplantationCBs containing advanced atherosclerotic plaques were transplantedfrom old apoE�/� mice into young eGFP�apoE�/� mice (n�12) andfrom old eGFP�apoE�/� mice into young apoE�/� mice (n�4)(Figure 1b, bottom). A subset of these transplantations were sexmismatched (female apoE�/� CB3male eGFP�apoE�/� mice, n�4).The others were sex matched (both male and female).

The microsurgical technique was essentially as previously de-scribed (see the detailed description online).15 The proximal anasto-mosis was prepared between recipient and donor common carotidarteries. The distal anastomosis was made between the recipientcommon carotid artery and the donor internal or external carotidartery, depending on plaque localization. The carotid branch not usedfor anastomosis was ligated. Mechanical plaque disruption of trans-planted plaques was performed immediately after reestablishment ofblood flow. Mice were killed after 30 minutes (n�2) or 4 weeks(n�14). Serial sections were collected at 100-�m steps throughoutthe disrupted CB plaques.

BMT

b Plaque SMC origin after mechanical plaque disruption

BM transplantation

eGFP+apoE-/- BM (2-3m) → apoE-/- (18m) n=4 n=6

+6w +7w +10w

n=10

+4wapoE-/- CB (12-17m) → eGFP+apoE-/- (3m)

CBT+PD Carotid bifurcation transplantation

n=5

PD

BMT

a Plaque SMC origin in long-term hematopoietic chimeras BM transplantation

eGFP+apoE-/- BM (2-3m) → apoE-/- (18m) n=10† † †† †††† ††

+6m+3m +9m

n=2

eGFP+apoE-/- CB (15 m) → apoE-/- (3m) n=4

Figure 1. Overview of experiments. a, Long-term hematopoieticchimeras. BM transplants were performed in 18-month-oldmice, and mice were housed until spontaneous death. Times ofdeath are indicated by crosses. b, Mechanical disruption ofplaques in the CB was produced in BM-transplanted mice (top)and after CB transplantation between apoE�/� andeGFP�apoE�/� mice (bottom). Age of donor and recipient miceat the time of BM or CB transplantation is indicated in parenthe-ses. BMT indicates BM transplantation; PD, mechanical plaquedisruption; and CBT, CB transplantation.

2 Circulation October 30, 2007

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

Page 3: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

Immunohistochemistry and FluorescenceIn Situ HybridizationAntibodies against smooth muscle �-actin (SM�A) and Mac2 wereused to identify SMCs and macrophages, respectively. SM�A isconsidered the most sensitive, although not specific, marker forplaque SMCs.16 Validity was confirmed by observing the expectedintracellular distribution of SM�A (subplasmalemmal) and Mac2(cytoplasmic) and by lack of staining when the specific antibody wasreplaced by an irrelevant antibody of the same isotype andconcentration.

Perls’ Prussian blue (iron) staining was performed to detect plaquehemorrhage in sections from the old hematopoietic chimeric miceand nontransplanted controls. When indicated, anti-eGFP antibodywas used to detect eGFP and Sudan Black B to stain and quench theautofluorescence of lipofuscin in these mice.19 Sequential SM�Aimmunostaining and fluorescence in situ hybridization for the Ychromosome was performed on CB plaque sections from sex-mismatched CB-transplanted mice and sex-matched controls aspreviously described.15

Fluorescence MicroscopySections were examined in an Olympus (Ballerup, Denmark) Cell-Rfluorescence microscope equipped with differential interferencecontrast (DIC) optics and motorized focus. SM�A� cell profiles inthe range of small SMC nuclear profiles or larger (from �3 �m[minor axis] �5 �m [major axis]) were analyzed for colocalizationof eGFP and SM�A by direct inspection at �400 magnification.Deconvolution of wide-field z-axis image stacks (0.2-�m opticalsection thickness) was performed on all plaque areas suspicious ofprevious needle injury using a blind 3-dimensional deconvolutionalgorithm (Autoquant Deblur 9.3, Media Cybernetics, Bethesda,Md). Nucleated SMC profiles, defined as cells in which the nucleuswas circumscribed by SM�A� staining, were analyzed for thepresence of the Y chromosome.

Statistical AnalysisThe log-rank test was used for survival analysis after BM transplan-tation. One-tailed 95% confidence Clopper-Pearson limits (CLs)were calculated for the occurrence of disruption and thrombosis afterneedle injury. CLs for the number of double-positive eGFP�SM�A�

cells were calculated by the approximation described by Hanley etal20 (95% CL, �3.0/number of observations).

The authors had full access to and take full responsibility for theintegrity of the data. All authors have read and agree to themanuscript as written.

ResultsWidespread Plaque Hemorrhage inOld ApoE�/� MicePlaque hemorrhage, assumed to be a sign of recent sponta-neous plaque disruption, has been reported in the brachioce-phalic trunk of apoE�/� mice from 30 weeks of age and in theaortic root and descending aorta from 34 weeks of age.21,22

To investigate the potential recruitment of BM-derivedSMCs to hemorrhagic plaques, we studied a cohort of oldeGFP�apoE�/� BM3apoE�/� transplanted mice (n�10) atthe time of spontaneous death (Figure 1a). Median survivaltime after BM transplantation (at 18 months of age) was 6months compared with 11 months in nonirradiated age-matched apoE�/� controls (n�5) (P�0.02, log-rank test),indicating late detrimental effects of irradiation. Times ofdeath in BM-transplanted mice are indicated in Figure 1a.Causes of death were not determined.

Plaques containing extravasated erythrocytes were presentat the postmortem examination in 7 of 10 BM-transplanted

mice (1 to 2 plaques per mouse) and in 2 of 5 age-matchednonirradiated apoE�/� controls (1 plaque per mouse) (Figure2). Intracellular iron-containing hemosiderin pigment, a signof erythrophagocytosis and thus preceding plaque hemor-rhage,23 was identified by Perls’ Prussian blue staining inplaques from 8 of 10 BM-transplanted mice (1 to 3 plaquesper animal) and in 3 of 5 nonirradiated apoE�/� controls (1plaque per animal) (Figure 2). Hemosiderin-positive cellsmost frequently coincided with intraplaque erythrocytes butwere found in other locations as well. Prussian blue stainingalso was notable in extracellular structures (see online-onlyData Supplement Figure II). There appeared to be no predi-lection site for plaque hemorrhages among the sites examined(lesser aortic arch curvature, brachiocephalic trunk, branchpoints of left common carotid and subclavian arteries, andCBs). Superimposed thrombosis was not seen.

SMCs in Hemorrhagic Plaques Are Not ofHematopoietic OriginPlaque foam cells in the old BM-transplanted mice weredouble positive for a macrophage marker (Mac2) and thetracking marker eGFP (Figure 3a), confirming the ability ofthe applied techniques to trace the progeny of hematopoieticstem cells. However, no plaque SMCs, identified by positivestaining for SM�A, were found to be eGFP� whether de-tected by the natural fluorescence of eGFP (7650 SM�A�

profiles analyzed; 95% CL, �0.04%; Figure 3b and 3c) or byan anti-eGFP antibody (4820 SM�A� cell profiles analyzed;95% CL, �0.06%; Figure 3d).

Cells exhibiting strong autofluorescence in the red andgreen color spectrum (yielding yellow in the combinedimage) often were present in plaques (pronounced examplesare shown in Figure 3c and 3d). Sudan black B stained thesecells and quenched the autofluorescence, indicating that thefluorescent pigment was lipofuscin (Figure 3e and 3f).Importantly, the autofluorescent cells could not be mistakenfor eGFP�SM�A� cells (false positives) in which the redfluorescent SM�A staining is confined to the subplasmalem-

Figure 2. Erythrocytes (E) and hemosiderin pigment (blue) pres-ent in a plaque near the branch point of the left subclavianartery in a BM-transplanted mouse. Longitudinal section. Perls’Prussian blue, Kernechtrot counterstain, and DIC. P indicatesplaque; L, lumen; and M, tunica media. Scale bar�50 �m.

Bentzon et al Source of Healing SMCs After Plaque Disruption 3

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

Page 4: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

mal space. Neither did quenching of lipofuscin autofluores-cence reveal otherwise masked eGFP�SM�A� cells (falsenegatives) whether by tracing of natural eGFP fluorescence(2430 SM�A� profiles analyzed; 95% CL, �0.1%) or byimmunofluorescent detection of eGFP (4690 SM�A� profilesanalyzed; 95% CL, �0.06%; Figure 3f).

Mechanical Plaque DisruptionIn murine plaques containing hemorrhage, the presence andextent of plaque disruption usually remain concealed, andsuperimposed thrombosis or clear signs of plaque healingwere not observed in the material presented above. Tocircumvent this limitation of atherosclerosis in apoE�/� mice,we developed a needle injury technique that allowed us tocontrol the spatial and temporal occurrence of plaque disrup-tion, superimposed thrombosis, and SMC-mediated healing(Figure 4a).

SMCs Healing Mechanical Disruptions Are Not ofHematopoietic OriginTo characterize the type of plaque injury inflicted by theneedle and to examine whether cells of hematopoietic origincontribute to plaque healing, we produced mechanical dis-ruptions in carotid plaques in eGFP�apoE�/� BM3apoE�/�

transplanted mice.Mice were killed at various time points as indicated in

Figure 1b. Acute injury to the plaque surface and thrombosiswere seen in all of the 5 mice killed after 30 minutes. Oneexample is shown in Figure 4c through 4e; the rest are shownin online-only Data Supplement Figure IV. Because this

sample was drawn at random, it can be used to estimate theconfidence interval of injured plaques in the BM-transplantedmice that were left to survive until 1 or 4 weeks aftermechanical disruption (95% CL, �55%). After 1 week, noresidual luminal thrombi were observed. After 4 weeks,distinct wedge-shaped areas of SMC accumulation, indicativeof healed disruption sites, were identified in 4 of 6 BM-transplanted mice (Figure 5a through 5c). This pattern wasnot observed in plaques removed from the contralateraluninjured CBs (4 plaques analyzed) or in age-matchedcontrols (4 mice, 6 plaques analyzed) (data not shown).

Importantly, not a single eGFP� SM�A� cell among 3560analyzed SM�A� cells was identified in serial sections of thehealed plaques in BM-transplanted mice (Figure 5a through5c). By comparison, SM�A� cells were almost uniformlyeGFP� in healed plaques in positive control eGFP�apoE�/�

mice (n�2; 469 of 478 [98%] analyzed SM�A� cell profiles;see online-only Data Supplement Figure V).

SMCs in Healed Plaques Are Derived From theLocal Artery SegmentTo evaluate whether any cell type in the circulating bloodcontributes to SMCs in healed plaques, we produced plaquedisruptions in CBs that were transplanted from apoE�/� miceinto the common carotid artery of eGFP�apoE�/� mice(Figures 1b, 4a, and 4b).

Plaque disruption with superimposed thrombosis was con-firmed in both mice (n�2) killed after 30 minutes (data notshown). Four weeks after plaque disruption, focal and distinctaccumulations of SMCs indicative of healed plaque disrup-

Figure 3. a, Foam cells (Fo) in the shoulder region of a fibrofatty plaque in the lesser curvature of the aortic arch. As expected, they areMac2� (red) and eGFP� (green), yielding a yellow overlay in the combined image. Scale bar�50 �m. See also online-only Data Supple-ment Figure III. b, Plaque from the lesser curvature of the aortic arch. Genetic tracing using eGFP fluorescence (green) reveals noBM-derived SM�A� (red) cells. Scale bar�200 �m. c, Magnification of b. Scale bar�50 �m. d, Tracing of BM-derived cells by immuno-histochemical detection of eGFP (green). No eGFP�SM�A� cells are seen. Scale bar 50 �m. e and f, Autofluorescence of intracellularpigment quenched by Sudan Black B (black) in an SM�A- and eGFP-immunostained section adjacent to d. e, DIC microscopy; f, fluo-rescence microscopy. No eGFP�SM�A� cells are unmasked by the quenching of autofluorescence. Scale bars�50 �m. Blue indicatesDAPI; gray scale, DIC; M, tunica media; F, fibrous cap; and L, lumen.

4 Circulation October 30, 2007

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

Page 5: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

tion sites were recognized in 8 of 10 mice (Figure 5d through5f). In one of these animals, the presence of multiple luminaindicated that a thrombus had been recanalized. Obliterationof the transplanted CB was found in 1 mouse.

Among 7980 SM�A� profiles analyzed in serial sections ofthe healed plaques from apoE�/� CB3eGFP�apoE�/� trans-planted mice, only 4 were eGFP� (non–graft derived). Theywere all located in the section taken closest to the distalanastomosis in 1 mouse, strongly indicating that these fewcells had migrated from the contiguous recipient vasculature.

Consistently, in the reverse experiment in whicheGFP�apoE�/� CBs were grafted into apoE�/� mice (n�4),2896 of 2982 SM�A� cellular profiles in healed plaques wereeGFP� (97%) (Figure 5g through 5i). This fraction wassimilar to that found in eGFP�apoE�/� positive control mice(see above).

As an independent tracking marker for blood-derivedSM�A� cells, we stained Y chromosomes in sections fromthe subset of mice in which CB transplantations were per-formed sex mismatched (female to male) and from sex-matched positive controls (male to male). No Y chromosomeswere detected among 432 analyzed SM�A� cell nuclei inhealed female apoE�/� CB plaques transplanted into maleeGFP�apoE�/� mice (n�4) (Figure 6). In comparison, the Ychromosome was detectable in 45 of 98 nucleated SM�A�

cell profiles in male apoE�/� CB plaques transplanted intomale eGFP�apoE�/� mice.

DiscussionThe healing process at plaque rupture sites has been describedin humans through the study of autopsy or atherectomyspecimens.2,3,5 First, the superimposed thrombus is partiallylysed by the fibrinolytic system. Then, synthetic-type SMCsaccumulate at the rupture site and secrete a fibrous extracel-lular matrix that seals the plaque defect and organizes theresidual thrombus. Finally, the plaque is re-endothelialized.Possibly, scar-like contraction of healing SMCs pulling onfibrin fibrils lead to concomitant constrictive remodeling ofthe arterial segment.4,24

Research into the cellular and molecular mechanisms thatcontrol this sequence of events, however, has been limited, atleast partly by the lack of a feasible and widely acceptedanimal model. In the present study, we describe a novelmechanical model of murine plaque disruption, superimposedthrombosis, and plaque healing and show that healing SMCsare of local arterial wall origin and not from circulatingprogenitor cells.

Models of Plaque Disruption in ApoE�/� MiceOwing to the detailed descriptions of several groups, it is wellestablished that spontaneous plaque hemorrhages occur inaging apoE�/� mice.21,22,25 Because the described hemor-rhages, at least in some anatomic locations, are found inplaques without intraplaque neovessels, the source must beluminal blood entering the plaque through a defect in the

Figure 4. a, Schematic of mechanical plaque disruption in a CB plaque (left). Same procedure after transplantation of an apoE�/� CBinto an eGFP�apoE�/� mouse (right). b, A transplanted CB 4 weeks after mechanical disruption. DA indicates distal anastomosis; PA,proximal anastomosis. *The site of needle insertion. Scale bar�1 mm. c through e, Plaque (P) disruption with a superimposed platelet-rich thrombus (T) 30 minutes after the procedure. A rupture site (R) in the fibrous cap is visible, allowing access of blood from thelumen (L) into a necrotic core. Scale bars�100 �m.

Bentzon et al Source of Healing SMCs After Plaque Disruption 5

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

Page 6: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

plaque surface (ie, by definition, spontaneous plaquedisruption).26–29

The value of these discrete disruptions for studies of plaquehealing in general and of platelet-mediated progenitor cellrecruitment in particular, however, is open to questions.28,29

First, superimposed thrombi are rarely found on spontane-ously disrupted plaques in apoE�/� mice,30 and none weredetected in the present study. It is possible, however, thattransient thrombi form over spontaneously disrupted plaquesin apoE�/� mice but that they escape detection because ofrapid lysis. This is consistent with our observation thatthrombi over mechanically disrupted plaques persisted for�1 week. Second, no definitive signs of spontaneous SMC-mediated plaque healing have been described. The presenceof layers of fibrous tissue within some murine plaques (buriedfibrous caps) has been suggested to reflect healed disruptions,but this remains to be documented.28,29

To circumvent these unanswered questions, we based mostof our analysis on a novel model in which plaque disruption

was produced by passing a microsurgical needle repeatedlyinto advanced carotid plaques from the luminal side. Thistechnique is related to but appears to be more efficient and toproduce more specific plaque injury than the model reportedpreviously by Reddick et al31 in which atherosclerotic seg-ments of apoE�/� mouse abdominal aorta were squeezedbetween forceps. Our needle injury produced plaque disrup-tion and superimposed thrombi in most, if not all, treatedmice (95% CL, �55%). Like superimposed thrombi inhumans, the thrombi were platelet rich, demonstrating thatthey had formed under rapid blood flow. Moreover, plaquedisruption was followed after 4 weeks by the appearance offocal and distinct accumulations of SMCs that resembledhealed rupture sites in humans.3

Healing SMCs Are of Local, Not Blood, OriginThe longstanding paradigm in the field has considered plaquehealing to be mediated by local proliferating plaque SMCs.7

Figure 5. Healing SMCs after mechanical plaque disruption are derived from local sources. a through c, SM�A staining of a CB plaque4 weeks after mechanical plaque disruption in an eGFP�apoE�/�3apoE�/� BM-transplanted mouse. The magnified area shows abnor-mal plaque architecture indicative of healed injury. No eGFP�SM�A� double-positive cells are seen. d through f, Healed disruption sitein an apoE�/� CB plaque transplanted into an eGFP�apoE�/� mouse recipient. A small intraplaque lumen is seen, indicating past injuryto the plaque surface. Abundant extracellular eGFP is present in the adventitia (please see note in the online-only Data Supplement).No eGFP�SM�A� cells were identified. g through i, Similar plaque in an eGFP�apoE�/� CB3apoE�/� transplanted mouse. SM�A� cellsare eGFP�. Wide-field fluorescence (a, d, g) and deconvoluted thin optical sections combined with DIC (b and c, e and f, h and i). Redindicates SM�A; green, eGFP; blue, DAPI; L, lumen; M, media; and A, adventitia. Scale bars�50 �m. See individual color channels inonline-only Data Supplement Figure VI.

6 Circulation October 30, 2007

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

Page 7: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

Recent studies in arterial injury models, however, havesuggested an alternative mechanism by which the superim-posed thrombotic response on ruptured plaques may mediaterecruitment of healing SMCs from a circulating pool ofBM-derived smooth muscle progenitor cells.10–13,32–34 Usingboth spontaneous and mechanical plaque disruptions inapoE�/� mice as model systems, we addressed this novelhypothesis, but we found that the healing SMCs were derivedentirely from the local vessel wall with no contribution fromcirculating progenitor cells.

First, not a single BM-derived SMC was detected inBM-transplanted apoE�/� mice whether in plaques withspontaneous plaque hemorrhage (median follow-up, 6months) (Figure 3) or in healed plaques 4 weeks aftermechanical plaque disruption (Figure 5a through 5c). Be-cause reconstitution of the irradiated mice with eGFP�

hematopoietic stem cells in both of these experiments wasconfirmed by flow cytometry and by the finding of eGFP�

macrophages in plaques (online-only Data Supplement Fig-ure I, Figure 3a, and data not shown), we conclude thathematopoietic stem cells do not contribute to healing SMCsin apoE�/� mice.

Second, to investigate whether healing SMCs could origi-nate from circulating cells of nonhematopoietic origin (eg,from mesenchymal stem cells in the BM or from non-BMsources), we analyzed mechanically disrupted plaques inapoE�/� CBs that were surgically inserted into the blood-stream of eGFP�apoE�/� mice. In these mice, all circulatingprogenitor cells were of eGFP� genotype regardless of theirpossible site of production. Furthermore, circulating progen-itor cells were present at physiological levels, which may notbe the case after BM transplantation. These experimentsshowed that no circulating cell type contributes to healingSMCs 4 weeks after mechanical plaque disruption (Figure 5dthrough 5f). Although 4 non–graft-derived SMCs were de-tected in 1 transplanted mouse, this small cluster of cells wasfound in the outermost serial section, strongly indicating thatthey had migrated from the contiguous recipient artery.

Finally, the use of the Y chromosome as an alternativetracking marker excluded the theoretical possibility that thefew percent of plaque SMCs that were not detectably eGFP�

in healed plaques of eGFP�apoE�/� CB3apoE�/� trans-planted mice (Figure 5g through 5i) or in eGFP�apoE�/�

positive controls (online-only Data Supplement Figure V)were derived from a special subset of circulating progenitorcells with low eGFP expression.

Candidate Sources of Healing SMCsOur findings support the classic theory that plaque healing ismediated by proliferation and matrix secretion of local plaqueor medial SMCs. The experiments, however, were not de-signed to distinguish between sources of SMCs within thearterial wall; thus, our data do not exclude the possibility thatsome SMCs may differentiate from local stem cells. Hu et al35

have described a population of Sca-1� cells in the arterialadventitia that have the potential to differentiate to SMCs invitro and to contribute to vein graft neointimal SMCs in vivo.In their experiments, however, Sca-1� cells gave rise only tosignificant numbers of neointimal SMCs when proliferation

Figure 6. a, CB plaque 4 weeks after mechanical plaque disrup-tion in a female apoE�/� CB3male eGFP�apoE�/� transplantedmouse. Multiple lumina are present, indicating recanalization ofa luminal thrombus. Scale bar�200 �m. b, Larger magnificationof a. Gray scale indicates DIC. Scale bar�50 �m. c, Detectionof Y chromosomes in the SM�A-stained section using a FITC-conjugated paint probe (green). Maximum projection of decon-voluted image stack. eGFP fluorescence is totally extinguishedby the fluorescence in situ hybridization procedure, and thedeteriorated SM�A� staining (red channel) is not shown. Thephenotype of cells with Y� nuclei (marked by arrows) can beidentified in the stored image of the SM�A immunostaining (b).No Y�SM�A� cells are seen. Red indicates SM�A; green, eGFP;blue, DAPI; L, lumen; M, tunica media; P, plaque; and A,adventitia.

Bentzon et al Source of Healing SMCs After Plaque Disruption 7

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

Page 8: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

and migration of local vein SMCs were blocked by irradiationof the vein graft. When atherosclerotic plaque disruptionsheal, large populations of SMCs, some of which are prolif-erating,5,8,31 are present in the plaque and adjacent media;hence, the contribution of adventitial stem cells in thissituation would be expected to be insignificant.

Contrasting ObservationsPrevious reports of BM-derived neointimal SMCs afterguidewire injury to nondiseased arteries in apoE�/� andwild-type mice are not easy to reconcile with the findings ofthe present study.10,33 It is possible, as first suggested by Hanand colleagues,34 that the varying degree of local SMC injuryin different models may be important. Plaque disruptioninvolves limited local SMC injury, but passing a guidewirethrough a small artery causes massive damage to medialSMCs in murine models.13 This may incapacitate localsources for repair and, in turn, by unknown mechanismsfacilitate recruitment of circulating progenitor cells.13,34 How-ever, because Li and colleagues,36 even after total allogeniceradication of local medial SMCs in their vasculopathymodel, did not observe BM-derived neointimal SMCs, thishypothesis cannot by itself explain the variable results ob-tained in the field.36

As recently pointed out by Hoofnagle and colleagues,16 itis plausible that methodological problems in immunofluores-cence analyses can explain many previous observations ofBM-derived SMCs. Most important, an absolute prerequisitefor valid analysis, which is very often not fulfilled, is clearsingle-cell resolution of the microscopic data. Also oftenoverlooked is the fact that myofilaments and hence SM�A,which is essentially the sole SMC marker used within thisfield of research, are located predominantly in the peripheryof neointimal SMCs, whereas the perinuclear region isoccupied by the organelles involved in protein synthesis(Figures 3, 5, and 6).37 If single neointimal SMCs with thischaracteristic distribution of SM�A cannot be discerned inhistological sections, it is highly questionable whether tissuepreparation, staining specificity, and microscopic resolutionare of sufficiently high quality to confirm cellular colocaliza-tion of SMCs with a BM tracking marker. Those interested inthe possibility of BM-derived SMCs are encouraged to readthe literature while bearing this in mind.

ConclusionsConsistent plaque disruption, superimposed thrombosis, andSMC-mediated healing can be induced by endovascular needleinjury to advanced carotid plaques in apoE�/� mice. Using thismodel, we found that healing SMCs are entirely derived fromthe local artery with no contribution from circulating progenitorcells. This supports the longstanding theory that plaque healingis mediated by local proliferating SMCs.

AcknowledgmentsWe thank Helle Quist, Birgitte Sahl, and Merete Dixen for excellenttechnical assistance.

Sources of FundingFunding for this work was provided by the Danish Medical ResearchCouncil, the Danish Heart Foundation, Fonden til LægevidenskabensFremme, and the University of Aarhus Research Foundation.

DisclosuresDr Falk is on the Scientific Program Board of the High-Risk PlaqueInitiative, BG Medicine, Waltham, Mass, and a consultant to BostonScientific Corporation, Natick, Mass. The other authors resport noconflicts.

References1. Davies MJ, Bland JM, Hangartner JR, Angelini A, Thomas AC. Factors

influencing the presence or absence of acute coronary artery thrombi insudden ischaemic death. Eur Heart J. 1989;10:203–208.

2. Flugelman MY, Virmani R, Correa R, Yu ZX, Farb A, Leon MB, ElamiA, Fu YM, Casscells W, Epstein SE. Smooth muscle cell abundance andfibroblast growth factors in coronary lesions of patients with nonfatalunstable angina: a clue to the mechanism of transformation from thestable to the unstable clinical state. Circulation. 1993;88:2493–2500.

3. Mann J, Davies MJ. Mechanisms of progression in native coronary arterydisease: role of healed plaque disruption. Heart. 1999;82:265–268.

4. Burke AP, Kolodgie FD, Farb A, Weber D, Virmani R. Morphologicalpredictors of arterial remodeling in coronary atherosclerosis. Circulation.2002;105:297–303.

5. Burke AP, Kolodgie FD, Farb A, Weber DK, Malcom GT, Smialek J,Virmani R. Healed plaque ruptures and sudden coronary death: evidencethat subclinical rupture has a role in plaque progression. Circulation.2001;103:934–940.

6. Libby P, Theroux P. Pathophysiology of coronary artery disease.Circulation. 2005;111:3481–3488.

7. Libby P. Inflammation in atherosclerosis. Nature. 2002;420:868–874.8. Lutgens E, de Muinck ED, Kitslaar PJ, Tordoir JH, Wellens HJ, Daemen

MJ. Biphasic pattern of cell turnover characterizes the progression fromfatty streaks to ruptured human atherosclerotic plaques. Cardiovasc Res.1999;41:473–479.

9. Gorenne I, Kavurma M, Scott S, Bennett M. Vascular smooth muscle cellsenescence in atherosclerosis. Cardiovasc Res. 2006;72:9–17.

10. Zernecke A, Schober A, Bot I, von Hundelshausen P, Liehn EA, MoppsB, Mericskay M, Gierschik P, Biessen EA, Weber C. SDF-1{alpha}/CXCR4 axis is instrumental in murine neointimal hyperplasia andrecruitment of smooth muscle progenitor cells. Circ Res. 2005;96:784–791.

11. Massberg S, Konrad I, Schurzinger K, Lorenz M, Schneider S, Zohln-hoefer D, Hoppe K, Schiemann M, Kennerknecht E, Sauer S, SchulzC, Kerstan S, Rudelius M, Seidl S, Sorge F, Langer H, Peluso M,Goyal P, Vestweber D, Emambokus NR, Busch DH, Frampton J,Gawaz M. Platelets secrete stromal cell-derived factor 1alpha andrecruit bone marrow-derived progenitor cells to arterial thrombi invivo. J Exp Med. 2006;203:1221–1233.

12. Schafer K, Schroeter MR, Dellas C, Puls M, Nitsche M, Weiss E,Hasenfuss G, Konstantinides SV. Plasminogen activator inhibitor-1 frombone marrow-derived cells suppresses neointimal formation after vascularinjury in mice. Arterioscler Thromb Vasc Biol. 2006;26:1254–1259.

13. Tanaka K, Sata M, Hirata Y, Nagai R. Diverse contribution of bonemarrow cells to neointimal hyperplasia after mechanical vascular injuries.Circ Res. 2003;93:783–790.

14. Caplice NM, Bunch TJ, Stalboerger PG, Wang S, Simper D, Miller DV,Russell SJ, Litzow MR, Edwards WD. Smooth muscle cells in humancoronary atherosclerosis can originate from cells administered at marrowtransplantation. Proc Natl Acad Sci U S A. 2003;100:4754–4759.

15. Bentzon JF, Weile C, Sondergaard CS, Hindkjaer J, Kassem M, FalkE. Smooth muscle cells in atherosclerosis originate from the localvessel wall and not circulating progenitor cells in apoE knockout mice.Arterioscler Thromb Vasc Biol. 2006;26:2696 –2702.

16. Hoofnagle MH, Thomas JA, Wamhoff BR, Owens GK. Origin of neo-intimal smooth muscle: we’ve come full circle. Arterioscler Thromb VascBiol. 2006;26:2579–2581.

17. Feil S, Hofmann F, Feil R. SM22alpha modulates vascular smooth musclecell phenotype during atherogenesis. Circ Res. 2004;94:863–865.

18. Hu Y, Davison F, Ludewig B, Erdel M, Mayr M, Url M, Dietrich H, XuQ. Smooth muscle cells in transplant atherosclerotic lesions are originatedfrom recipients, but not bone marrow progenitor cells. Circulation. 2002;106:1834–1839.

19. Schnell SA, Staines WA, Wessendorf MW. Reduction of lipofuscin-likeautofluorescence in fluorescently labeled tissue. J Histochem Cytochem.1999;47:719–730.

20. Hanley JA, Lippman-Hand A. If nothing goes wrong, is everything allright? Interpreting zero numerators. JAMA. 1983;249:1743–1745.

8 Circulation October 30, 2007

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

Page 9: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

21. Rosenfeld ME, Polinsky P, Virmani R, Kauser K, Rubanyi G, Schwartz SM.Advanced atherosclerotic lesions in the innominate artery of the ApoEknockout mouse. Arterioscler Thromb Vasc Biol. 2000;20:2587–2592.

22. Calara F, Silvestre M, Casanada F, Yuan N, Napoli C, Palinski W.Spontaneous plaque rupture and secondary thrombosis in apolipoproteinE-deficient and LDL receptor-deficient mice. J Pathol. 2001;195:257–263.

23. Kockx MM, Cromheeke KM, Knaapen MW, Bosmans JM, De MeyerGR, Herman AG, Bult H. Phagocytosis and macrophage activationassociated with hemorrhagic microvessels in human atherosclerosis.Arterioscler Thromb Vasc Biol. 2003;23:440 – 446.

24. Yee KO, Schwartz SM. Why atherosclerotic vessels narrow: the fibrinhypothesis. Thromb Haemost. 1999;82:762–771.

25. Johnson JL, Jackson CL. Atherosclerotic plaque rupture in the apoli-poprotein E knockout mouse. Atherosclerosis. 2001;154:399–406.

26. Bea F, Blessing E, Bennett B, Levitz M, Wallace EP, Rosenfeld ME.Simvastatin promotes atherosclerotic plaque stability in apoE-deficientmice independently of lipid lowering. Arterioscler Thromb Vasc Biol.2002;22:1832–1837.

27. Williams H, Johnson JL, Carson KG, Jackson CL. Characteristics ofintact and ruptured atherosclerotic plaques in brachiocephalic arteries ofapolipoprotein E knockout mice. Arterioscler Thromb Vasc Biol. 2002;22:788–792.

28. Jackson CL, Bennett MR, Biessen EA, Johnson JL, Krams R. Assessmentof unstable atherosclerosis in mice. Arterioscler Thromb Vasc Biol. 2007;27:714–720.

29. Schwartz SM, Galis ZS, Rosenfeld ME, Falk E. Plaque rupture in humansand mice. Arterioscler Thromb Vasc Biol. 2007;27:705–713.

30. Zhou J, Moller J, Danielsen CC, Bentzon J, Ravn HB, Austin RC, Falk E.Dietary supplementation with methionine and homocysteine promotesearly atherosclerosis but not plaque rupture in ApoE-deficient mice.Arterioscler Thromb Vasc Biol. 2001;21:1470–1476.

31. Reddick RL, Zhang SH, Maeda N. Aortic atherosclerotic plaque injury inapolipoprotein E deficient mice. Atherosclerosis. 1998;140:297–305.

32. Religa P, Bojakowski K, Maksymowicz M, Bojakowska M, Sirsjo A,Gaciong Z, Olszewski W, Hedin U, Thyberg J. Smooth-muscle pro-genitor cells of bone marrow origin contribute to the development ofneointimal thickenings in rat aortic allografts and injured rat carotidarteries. Transplantation. 2002;74:1310–1315.

33. Sata M, Saiura A, Kunisato A, Tojo A, Okada S, Tokuhisa T, Hirai H,Makuuchi M, Hirata Y, Nagai R. Hematopoietic stem cells differentiateinto vascular cells that participate in the pathogenesis of atherosclerosis.Nat Med. 2002;8:403–409.

34. Han CI, Campbell GR, Campbell JH. Circulating bone marrow cells cancontribute to neointimal formation. J Vasc Res. 2001;38:113–119.

35. Hu Y, Zhang Z, Torsney E, Afzal AR, Davison F, Metzler B, Xu Q.Abundant progenitor cells in the adventitia contribute to atherosclerosisof vein grafts in ApoE-deficient mice. J Clin Invest. 2004;113:1258–1265.

36. Li J, Han X, Jiang J, Zhong R, Williams GM, Pickering JG, Chow LH.Vascular smooth muscle cells of recipient origin mediate intimalexpansion after aortic allotransplantation in mice. Am J Pathol. 2001;158:1943–1947.

37. Thyberg J, Blomgren K, Hedin U, Dryjski M. Phenotypic modulation ofsmooth muscle cells during the formation of neointimal thickenings in therat carotid artery after balloon injury: an electron-microscopic and ste-reological study. Cell Tissue Res. 1995;281:421–433.

CLINICAL PERSPECTIVESmooth muscle cell (SMC)–mediated healing of atherosclerotic plaque ruptures is relevant to the full spectrum of clinicalpresentations of atherosclerosis, including the acute coronary syndromes, stable angina pectoris, ischemic stroke, andperipheral arterial disease. Plaque healing presumably protects against repeated episodes of rupture in plaques, butunfortunately, the reparative response can be so abundant with massive extracellular matrix deposition that it leads to rapidplaque growth and arterial stenosis. Given the clinical importance of plaque healing, surprisingly little is known about itsmechanisms. The longstanding paradigm has considered plaque healing to be carried out by local SMCs that are activatedby the growth factors released during superimposed thrombosis. Recent studies in arterial injury models, however, havesuggested that healing SMCs may also be recruited from a circulating pool of smooth muscle progenitor cells. In the presentpaper, we examined the origin of plaque-healing SMCs in a mouse model of atherosclerosis using a novel mechanicalplaque disruption technique. We found that the healing SMCs were entirely of local origin, presumably from local plaqueor medial SMCs. Although one should always be cautious about extrapolating from animal models to human disease, ourfindings suggest that the focus for future research on plaque healing should be on drugs and interventions that target SMCproliferation, migration, and matrix production, and not mobilization, homing, or differentiation of putative circulatingsmooth muscle progenitor cells.

Bentzon et al Source of Healing SMCs After Plaque Disruption 9

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

Page 10: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

Jacob F. Bentzon, Claus S. Sondergaard, Moustapha Kassem and Erling FalkOrigin in Apolipoprotein E Knockout Mice

Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions Are of Local, Not Blood,

Print ISSN: 0009-7322. Online ISSN: 1524-4539 Copyright © 2007 American Heart Association, Inc. All rights reserved.

is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231Circulation published online October 15, 2007;Circulation. 

http://circ.ahajournals.org/content/early/2007/10/15/CIRCULATIONAHA.107.722355.citationWorld Wide Web at:

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

http://circ.ahajournals.org/content/suppl/2007/12/10/CIRCULATIONAHA.107.722355.DC1Data Supplement (unedited) at:

  http://circ.ahajournals.org//subscriptions/

is online at: Circulation Information about subscribing to Subscriptions: 

http://www.lww.com/reprints Information about reprints can be found online at: Reprints:

  document. Permissions and Rights Question and Answer this process is available in the

click Request Permissions in the middle column of the Web page under Services. Further information aboutOffice. Once the online version of the published article for which permission is being requested is located,

can be obtained via RightsLink, a service of the Copyright Clearance Center, not the EditorialCirculationin Requests for permissions to reproduce figures, tables, or portions of articles originally publishedPermissions:

by guest on February 18, 2018http://circ.ahajournals.org/

Dow

nloaded from

Page 11: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

1

Expanded Methods

Mechanical plaque disruption Mice were anesthetized with isoflurane (induction 5%, maintenance 1.5-2.0%) and the neck was incised through the midline. Both carotid bifurcations were examined to identify the plaque most easily accessible for endovascular needle injury. The location and size of atherosclerotic plaques in the carotid bifurcation are highly variable. Where advanced plaques were present, they were located in the distal common carotid artery or in the proximal parts of the external or internal carotid artery. In some cases, no plaques suitable for the procedure were found and the mouse was excluded. Loose 6-0 silk ligatures were placed around the proximal common carotid artery and the distal external and internal carotid arteries to ensure hemostasis during the procedure. Then, a 9-0 spatula needle (Ethicon, Johnson & Johnson, Birkerød, Denmark) was inserted ~500 μm proximal to the targeted plaque, advanced distally through the lumen, and inserted repeatedly into the plaque from the luminal side (see Supplementary Video online). After removal of the needle, we allowed intermittent flow until bleeding from the puncture site had stopped. The skin was closed using 6-0 silk suture. Postoperatively, analgetics (buprenorphine, 0.1 mg/kg, repeated every 12 hours for 3 days) were administered. No perioperative deaths were recorded. Carotid bifurcation transplantation Anesthetized donor mice (pentobarbital 5 mg i.p.) were sacrificed by exsanguination, and the vascular tree was flushed with isotonic saline through the left ventricle. Both carotid bifurcations were examined and the one most suited for transplantation was excised and kept in RPMI-1640 medium (Invitrogen, Taastrup, Denmark) at room temperature. Carotid bifurcations chosen for transplantation contained isolated plaques flanked by healthy vessel segments. Recipient mice were anesthetized with isoflurane (induction 5%, maintenance 1.5-2.0%) and the right common carotid artery was dissected free of surrounding tissue. Proximal and distal microvascular clamps were applied and a segment of the artery (~1-1.5 mm) was excised. The donor carotid bifurcation was then interposed by two end-to-end anastomoses of four symmetrically placed 11-0 polyamide single sutures (Ethicon, Johnson & Johnson, Birkerød, Denmark). The proximal anastomosis was prepared between recipient and donor common carotid arteries. The distal anastomosis was prepared between the recipient common carotid artery and the donor internal or external carotid artery. The carotid branch not used for anastomosis was ligated.

After removal of the microvascular clamps and hemostatic control of the anastomosis sites, needle injury was performed as described above. The perioperative mortality was 7%. Total operation time was 60-80 minutes. Postoperatively, analgetics (buprenorphine, 0.1 mg/kg s.c. repeated every 12 hours for 3 days) were administered. Tissue processing and microscopic examination Analysis of old hematopoietic chimeras (Figure 1a in main text) For analysis of smooth muscle cell (SMC) origin in old long-term hematopoietic chimeras, we removed the aortic arch, including the brachiocephalic trunk and the proximal parts of the left carotid and subclavian arteries, and the right and left carotid bifurcations after immersion-fixation (48-72 hours) of the spontaneously dead mice. Vessels were cryoprotected in a sucrose solution (25 % w/v for 24 h + 50 % w/v for 24 h), embedded in O.C.TTM compound (Sakura Finetek, Pro-Hosp, Vaerloese, Denmark), and snap-frozen in liquid nitrogen-chilled methanol:acetone (1:1). Consecutive longitudinal sections of aortic arches and cross-sections of carotid bifurcations (4 μm thickness) were prepared. Smooth muscle α-actin (SMαA) was detected using mouse monoclonal anti-SMαA (1:50, Clone 1A4, Dako, Glostrup, Denmark) after blocking of endogenous mouse immunoglobulins with donkey anti-mouse Fab fragments (1:10, Jackson Immunoresearch, Cambridgeshire, UK) and formaldehyde fixation (4%, 10 min). This was followed by Texas-red conjugated donkey-anti mouse secondary antibody (1:400, Jackson Immunoresearch). An irrelevant monoclonal antibody (1:50, Clone DAK-GO5, DAKO) of the same isotype and concentration was used as a negative control. If not indicated otherwise, enhanced green fluorescent protein (eGFP) was detected by its natural fluorescence. To evaluate whether eGFP fluorescence was retained and detectable after spontaneous death and partial post-mortem decomposition, two eGFP+apoE-/- mice (15 and 18 months of age) were sacrificed in a pilot study, left for 24 hours at room temperature, and fixed for 72 hours. The natural fluorescence of eGFP was found to be preserved. In a subset of sections, eGFP was stained immunohistochemically using a rabbit anti-eGFP antibody (1:500, Abcam, ab290, Cambridge, UK) followed by a FITC-conjugated donkey anti-rabbit secondary antibody (1:400, Jackson Immunoresearch). The Mac2 epitope was stained as a marker for plaque macrophages using a rat anti-mouse Mac2 antibody (1:500, CL8942AP, Cedarlane Labs, Trichem Aps, Frederikssund, Denmark) followed by a Texas Red-

Page 12: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

2

conjugated goat anti-rat secondary antibody (1:400, Jackson Immunoresearch). An irrelevant rat monoclonal antibody (1:50, R2a00, Caltag, Trichem) of the same isotype and concentration was used as a negative control. Nuclei were stained with DAPI (Sigma, Broendby, Denmark) and the sections were mounted in Slowfade Light Antifade (Invitrogen, Taastrup, Denmark). In some sections, Sudan Black B (0.1 % in 70% ethanol for 5 minutes followed by brief wash in 70% ethanol) was used to stain lipofuscin and quench its autofluorescence. This step was applied after incubation with the secondary antibody. Analysis of plaques after mechanical disruption (Figure 1b in main text) Mice subjected to mechanical plaque disruption were anesthetized (pentobarbital 5 mg i.p.), killed by exsanguination, perfusion-fixed with phosphate-buffered (pH 7.2) 4% formaldehyde through the left ventricle for 5 minutes, and immersion-fixed for 6 hours at room temperature. Carotid bifurcations from mice sacrificed 30 minutes after mechanical plaque disruption were dehydrated and embedded in paraffin to best preserve the integrity of thrombi. Carotid bifurcations from the other mice were processed for cryosectioning as decribed above. Sections of mechanically disrupted plaques and non-injured control plaques were collected at levels taken 100 μm apart. To evaluate acute plaque injury, sections from each level were stained with elastin-trichrome stain using a standard protocol. To analyze SMC origin after one or four weeks, sections at each level were stained for SMαA+ as described above. Endogenous eGFP fluorescence was used to trace cells.

Page 13: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

3

Supplementary Note

Abundant extracellular eGFP was consistently present around apoE-/- carotid bifurcations grafted into eGFP+apoE-/- mouse recipients, but never in plaques or the adjacent media. The same phenomenon was not seen in positive control eGFP+apoE-/- mice (Supplementary Figure V). The origin of the extracellular eGFP was not determined. It may have originated from cells damaged during dissection of the neck which was performed before perfusion-fixation in carotid bifurcation transplanted mice to obtain photographs of the inserted grafts (Figure 4b in main text).

Page 14: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

4

Supplementary Figure I

Supplementary Figure I. Flow cytometry of peripheral blood for the analysis of hematopoietic chimerism after bone marrow (BM) transplantation. Left panel. Cells within a defined forward-side scatter gate that encompass the major leukocyte populations are analyzed for green fluorescence. Right panel. Examples of results obtained in negative control apoE-/- mice (top), positive control eGFP+apoE-/- mice (middle), and eGFP+apoE-/- BM → apoE-/- transplanted mice (bottom).

Page 15: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

5

Supplementary Figure II

Supplementary Figure II. Perls’ Prussian Blue staining of extracellular structures. (a) Staining of extracellular bodies with a membranous appearance was frequently observed. These bodies were yellow autofluorescent under blue excitation light and stained positive for Sudan Black B in adjacent sections, indicating that they were probably ceroid/lipofuscin pigments. (b) Diffuse Prussian Blue staining was also noted in extracellular matrix of chondroid metaplastic areas. Scale bars 50 μm. L, Lumen. P, plaque. M, tunica media.

Page 16: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

6

Supplementary Figure III

Supplementary Figure III. Foam cell accumulation in the shoulder region of an advanced plaque in the lesser curvature of the aortic arch. As expected, Mac-2+ macrophages are eGFP+, indicating their hematopoietic origin. The yellow color (a) is the result of colocalizing Mac-2 (red, b) and eGFP (green, c). Blue, DAPI. Greyscale, DIC. L, lumen. F, foam cells. M, tunica media. Scale bar 50 μm.

Page 17: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

7

Supplementary Figure IV

Supplementary Figure IV. Four mice sacrificed 30 minutes after mechanical plaque disruption. Plaque injury and thrombosis are seen in all mice to varying extent. The areas demarcated in images in the left column are shown in greater magnification in the right column. Elastin-trichrome stain. L, lumen. P, plaque. T, thrombus. Scale bars 100 μm.

Page 18: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

8

Supplementary Figure V

Supplementary Figure V. Carotid bifucartion plaque in a positive control eGFP+apoE-/- mouse four weeks after needle injury (at 20 months of age). Fluorescence images of SMαA (red, b) and eGFP (green, c) are shown individually and combined (a). A focal accumulation of SMαA cells (*) indicates recent plaque disruption and healing. SMαA+ cells are eGFP+. L, lumen. P, plaque. M, tunica media. Scale bar 200 μm.

Page 19: Smooth Muscle Cells Healing Atherosclerotic Plaque Disruptions

9

Supplementary Figure VI

Supplementary Figure VI. Distinct areas of SMC accumulation in carotid bifurcation plaques indicating healed disruption sites. Fluorescence images of SMαA (red, middle column) and eGFP (green, right column) are shown individually and combined (left column). (a-c) Disrupted carotid bifurcation plaque in an eGFP+apoE-/- BM → apoE-/- transplanted mouse. No eGFP+SMαA+ double-labeled cells. (d-f) Injury site in a carotid bifurcation plaque transplanted from an apoE-/- mouse donor to an eGFP+apoE-/- mouse recipient prior to mechanical plaque disruption. No eGFP+SMαA+ double-labeled cells. (g-i) Injury site in an eGFP+apoE-/- carotid bifurcation plaque grafted into an apoE-/- mouse before plaque disruption. Almost all SMαA+ cells are eGFP+. L, lumen. P, plaque. M, media. BM, bone marrow. Deconvoluted optical sections. Greyscale, DIC. Blue, DAPI. Scale bars 50 μm.