文档库 最新最全的文档下载
当前位置:文档库 › The neurovascular unit as a selective barrier to polymorphonuclear granulocyte

The neurovascular unit as a selective barrier to polymorphonuclear granulocyte

ORIGINAL PAPER

The neurovascular unit as a selective barrier

to polymorphonuclear granulocyte(PMN)in?ltration

into the brain after ischemic injury

Gaby Enzmann?Caroline Mysiorek?Roser Gorina?Yu-Jung Cheng?

Sharang Ghavampour?Melanie-Jane Hannocks?Vincent Prinz?

Ulrich Dirnagl?Matthias Endres?Marco Prinz?Rudi Beschorner?

Patrick N.Harter?Michel Mittelbronn?Britta Engelhardt?Lydia Sorokin

Received:23August2012/Revised:14December2012/Accepted:14December2012/Published online:27December2012óThe Author(s)2012.This article is published with open access at https://www.wendangku.net/doc/fb11487624.html,

Abstract The migration of polymorphonuclear granulo-cytes(PMN)into the brain parenchyma and release of their abundant proteases are considered the main causes of neuronal cell death and reperfusion injury following ischemia.Yet,therapies targeting PMN egress have been largely ineffective.To address this discrepancy we inves-tigated the temporo-spatial localization of PMNs early after transient ischemia in a murine transient middle cerebral artery occlusion(tMCAO)model and human stroke https://www.wendangku.net/doc/fb11487624.html,ing speci?c markers that distinguish PMN (Ly6G)from monocytes/macrophages(Ly6C)and that de?ne the cellular and basement membrane boundaries of the neurovascular unit(NVU),histology and confocal microscopy revealed that virtually no PMNs entered the infarcted CNS parenchyma.Regardless of tMCAO duration,PMNs were mainly restricted to luminal surfaces or perivascular spaces of cerebral vessels.Vascular PMN accumulation showed no spatial correlation with increased vessel permeability,enhanced expression of endothelial cell adhesion molecules,platelet aggregation or release of neu-trophil extracellular traps.Live cell imaging studies con?rmed that oxygen and glucose deprivation followed by reoxygenation fail to induce PMN migration across a brain endothelial monolayer under?ow conditions in vitro.The absence of PMN in?ltration in infarcted brain tissues was corroborated in25human stroke specimens collected at early time points after infarction.Our observations identify the NVU rather than the brain parenchyma as the site of PMN action after CNS ischemia and suggest reappraisal of targets for therapies to reduce reperfusion injury after stroke.

Electronic supplementary material The online version of this article(doi:10.1007/s00401-012-1076-3)contains supplementary material,which is available to authorized users.

G.EnzmannáR.GorinaáB.Engelhardt(&)

Theodor Kocher Institute,University of Bern, Freiestrasse1,3012Bern,Switzerland

e-mail:bengel@tki.unibe.ch

C.MysiorekáY.-J.ChengáS.Ghavampourá

M.-J.HannocksáL.Sorokin(&)

Institute of Physiological Chemistry and Pathobiochemistry, University of Mu¨nster,Waldeyerstrasse15,

48149Mu¨nster,Germany

e-mail:sorokin@uni-muenster.de

V.PrinzáU.DirnagláM.Endres

Department of Neurology,Berlin,Germany

U.DirnagláM.Endres

Center for Stroke Research Berlin,Charite′University, Berlin,Germany M.Prinz

Department of Neuropathology,University of Freiburg, Freiburg,Germany

M.Prinz

BIOSS Centre for Biological Signaling Studies,

University of Freiburg,Freiburg,Germany

R.Beschorner

Department of Neuropathology,Institute of Pathology and Neuropathology,University of Tu¨bingen,Tu¨bingen,Germany P.N.HarteráM.Mittelbronn(&)

Institute of Neurology(Edinger Institute),

University of Frankfurt,Frankfurt,Germany

e-mail:Michel.Mittelbronn@kgu.de

Acta Neuropathol(2013)125:395–412 DOI10.1007/s00401-012-1076-3

Keywords Neurovascular unitáPolymorphonuclear granulocyteáMigrationá

HumanáMouse

Introduction

Reperfusion of cerebral vessels after transient occlusion is associated with immune cell recruitment,which contributes to both damage of the vessel and the surrounding tissue. Polymorphonuclear granulocytes(PMNs)are considered to play a prominent role in microvascular responses to ischemia via protease-mediated tissue damage and neuro-nal cell death during reperfusion by direct contact with CNS tissue[2,7,19,39,49,60].This necessitates PMN extravasation across the blood–brain barrier(BBB)into the brain parenchyma at early stages after the ischemic insult, prior to terminal neuronal damage[25].

Yet,the effects of experimental neutropenia in rodent stroke models range from decreased infarct volume[12,62, 75]to little or no in?uence on lesion size[11,44,45],and PMN mobilization from the bone marrow induced by gran-ulocyte colony stimulating factor(G-CSF)does not worsen clinical outcome in murine stroke models[83]or in a recent clinical phase II trial(AXIS)[76].Furthermore,although some animal experiments targeting adhesion molecules mediating the multi-step PMN migration across in?amed microvessels have shown reduced infarct size[25],clinical trials targeting PMN adhesion to endothelial ICAM-1[9,36, 47]or aiming at preventing PMN in?ltration into the brain by neutralizing the a M b2-integrin(CD11b/CD18)[54]have failed to alleviate stroke severity,suggesting that the mode of PMN action requires reassessment.

These discrepancies may re?ect differences between the pathogenic mechanisms involved in ischemia/reperfusion in animal experiments and human stroke cases or,alter-natively,could be linked to the assumption that PMNs use ICAM-1and b2-integrins to migrate into the brain paren-chyma in reperfusion injury,as in other in?ammatory scenarios.This,however,has not been formally shown and most PMN depletion studies or adhesion molecule blocking studies after cerebral ischemia either did not assess PMN/ immune cell in?ltration[15,16,67]or employed methods that do not permit unequivocal identi?cation of PMNs[37, 52,61,78].A common problem has been the use of broad speci?city reagents such as the antibody clone,RB6-8C5, targeting the Gr-1antigen,which recognizes both Ly6G and Ly6C and therefore stains PMNs and monocytes[35]. Similarly,myeloperoxidase staining identi?es PMNs but also monocytes and activated microglial cells[13].Hence, the use of such broad speci?city reagents in?ow cytometry and/or immunohistochemistry inevitably results in an over-estimation of PMN numbers.It is therefore important to reassess the role of PMNs after cerebral ischemia using more speci?c immune cell markers that are now available, in particular in relation to their association with adhesion molecules and their precise localization within the vascu-lature and/or brain parenchyma.

That CNS vessels and neurons are functionally coupled is well illustrated by the rapid response of neurons to focal ischemia.Mechanistically,this is achieved by the neurovas-cular unit(NVU),composed of a monolayer of specialized endothelial cells(EC)interconnected by complex tight junctions,the underlying endothelial basement membrane (BM)and a second BM,known as the parenchymal BM as it marks the border to the CNS parenchyma.Together with the associated ensheathing layer of astrocyte endfeet,the parenchymal BM de?nes the glia limitans[3,64,89],which is structurally and functionally interconnected to the sur-rounding neurons via astrocytes and microglia[14,18].With the exception of capillaries,where endothelial and paren-chymal BMs fuse to form one composite BM,in all brain parenchymal vessels the endothelial and parenchymal BMs are structurally and biochemically distinct entities,which de?ne the inner and outer limits of the perivascular space.

The NVU strictly controls immune cell emigration from the blood vessel that requires a cascade of adhesive inter-actions that are well described for neuroin?ammation[32]. Due to the specialized structure of the NVU,leukocyte entry into the brain parenchyma involves two differently regulated steps:migration of leukocytes across the endo-thelium into the perivascular space and progression across the glia limitans into the brain parenchyma.In a murine model of multiple sclerosis,experimental autoimmune encephalomyelitis(EAE),induction of disease symptoms occurs only upon immune cell penetration of the glia limitans into the CNS parenchyma,whereas accumulation of in?ammatory cells within the perivascular space does not translate into clinical disease[1,8,88],highlighting the signi?cance of the glia limitans as the effective border to the CNS parenchyma.

While histological analyses of brain samples from murine models of transient ischemia and human stroke tissues have detected‘‘in?ammatory in?ltrates’’in the brain[33,68],there has been little attempt to either spe-ci?cally identify PMNs using unique molecular markers or to localize their precise position within the brain at de?ned time points after ischemic stroke.This study presents a collaborative investigation involving stroke researchers, neuropathologists,and basic scientists to determine the temporo-spatial relationship between immune cells and blood vessel micro-architecture in the mouse and human brain at early(acute)stages after ischemia using a panel of markers for different subsets of myeloid cells,endothelial cell

adhesion molecules,and the BMs of the NVU.We employ the speci?c PMN marker,Ly6G[17],in immunohisto-chemistry and in double and triple immuno?uorescence confocal microscopy to precisely localize PMNs in relation to the cellular and BM components of the NVU,and to inves-tigate correlations between PMN localization and altered vessel permeability or expression of endothelial cell adhesion molecules known to be involved in PMN rolling(P-selectin), arrest and crawling(ICAM-1,ICAM-2)or diapedesis(PE-CAM-1,CD99,JAM-A).An in vitro model for the BBB[82], where PMN migration across an endothelial monolayer is measured under physiological?ow,is employed to investi-gate the effects of transient oxygen–glucose deprivation followed by reoxygenation.Our mouse and human data highlight the need for a critical reappraisal of the precise site of PMN action after stroke and molecular targets for therapies to reduce reperfusion injury after stroke.

Materials and methods

Animals

151male C57BL/6and129Sv8–12-week-old mice were employed(Table1).Animal experiments were performed according to Swiss(56/08)and German(G0383/09)legislation.

Transient middle cerebral artery occlusion(tMCAO)

Transient focal ischemia using the intraluminal?lament model was performed[26].The left middle cerebral artery was unilaterally occluded for30,60,and90min,after which the?lament was withdrawn and the tissue was rep-erfused for varying lengths of time,resulting in the following ischemia/reperfusion(I/R)protocols:I/R:30 min/6,12,18,24,48,72h,1and2weeks;60min/6,12,18, 24,48,72h,1and2weeks;90min/3,18,48h).Motor–sensory scores of the mice analyzing gait disturbances were determined[10,27]prior to termination of the experiments. To account for experimenter variability,tMCAO was per-formed in two independent laboratories and brains were distributed to two separate laboratories for independent immunohistological and immuno?uorescence analyses.All analyses included coronal sections through the core of the lesion and adjacent penumbra(Bregma0.50mm),and caudal to the ischemic area(Bregma-2.46mm).Hence,all analyses were standardized to the Stroke Therapy Aca-demic Industry Roundtable(STAIR)criteria[34].

Flow cytometry

In?ammatory cells from ischemic ipsilateral and contra-lateral brain hemispheres were isolated and stained as described previously[28,30].Brie?y,6anaesthetized stroke mice in3independent experiments were perfused with297.4mosm/l phosphate buffered saline(PBS),pH 7.4,to remove peripheral blood,their brains dissected,and ischemic and contralateral cerebral hemispheres separated. The tissue was then mechanically dissociated,digested with collagenase VIII and DNAse I,?ltered,and cells were separated by centrifugation through a Percoll-gradient. CD45high in?ammatory cells and CD45intermediate microglia were collected from the interphase and stained for Ly6G to identify PMNs.Flow cytometry was performed using a

Table1Mouse tissues analyzed

Mouse strain Ischemia

(min)

Acute

reperfusion

Surgery

performed

Total

animals

analyzed

Animals

analyzed

by IH

Antibodies employed c Animals

analyzed

by IF

Antibodies employed c

C57Bl/6306,12,18,24,

48,72h

and

2weeks Berlin,

Bern

5912Leukocyte markers:CD45,

Gr-1,Ly6G,F4/80,CD11b,

Endothelial cell markers:

PECAM-1,VCAM-1,

ICAM-1,ICAM-2,

P-selectin

47a BM:Pan-laminin,laminin a5,

laminin a2,collagen IV

Leukocyte markers:CD45,

Ly6G,Ly6C,F4/80,CD11b,

CD41

Endothelial cell markers:

PECAM-1,VCAM-1,

ICAM-1,ICAM-2,

P-selectin.

C57Bl/6606,12,18,24,

48,72h

and

2weeks Berlin,

Bern

68860a

C57Bl/6903,18,48h Berlin936b 129Sv306,72h Berlin633 129Sv903,18,48h Berlin936 a18,24,48,72h and2week samples analyzed

b18h,48h analyzed

c See Table2

FACSCalibur;CellQuest(Becton–Dickinson)and FlowJo (Tree Star Inc.)software were employed for data analysis. Immunohistochemistry

Mice were perfused with1%paraformaldehyde(PFA)in PBS,pH7.4,their brains removed,embedded in TissueTek (OCT Compound,Haslab)and frozen in a dry ice/isopentane bath.Cryostat sections(6l m)spanning the whole lesion area were prepared,?xed in-20°C acetone and stained using a three-step immunoperoxidase staining kit(Vectastain).Pri-mary antibodies employed are listed in Table2.Secondary antibodies included biotinylated anti-rat and goat antibodies and were consecutively incubated with avidin–biotin com-plex(ABC)and peroxidase substrate solution(AEC,Vectastain).Sections were assessed using a Nikon Eclipse E600microscope equipped with a digital camera. Immuno?uorescence

Immuno?uorescence staining was performed on non-per-fused,snap frozen tissues.5-l m cryostat sections were?xed in-20°C methanol.For confocal microscopy or3D recon-structions,tissues were?xed in1.5%PFA in PBS,pH7.4, for1.5h at4°C,embedded in1%agarose in PBS,pH7.4, and100-l m sections were prepared using a Zeiss vibratome. Thick and thin sections were treated with PBS,pH7.4,plus 1%BSA before incubation at4°C with primary antibody (Table2).Secondary antibodies included goat anti-rabbit and donkey anti-rat IgG conjugated with Alexa Fluor488or

Table2Primary antibodies to extracellular matrix molecules and in?ammatory cells

Molecule Antibody name/clone Marker IF/IH a Reference/source

Mouse

Pan-Laminin455All BMs IF,IH[80]b

Laminin a2401Parenchymal BM IF[77]b

Laminin a4377Endothelial BM IF[73]b

Laminin a5405Endothelial BM IF[80]b

Collagen IV–All BMs IF[86]c

P-Selectin Rabbit anti-P-selectin Activated endothelium IH[22],BD Pharmingen ICAM-125ZC7Endothelial adhesion molecule IF,IH[72],BD Pharmingen ICAM-23C4Endothelial adhesion molecule IH BD Pharmingen VCAM-19DB3Endothelial adhesion molecule IF,IH[29],BD Pharmingen PECAM-1Mec13.3Endothelial junctions IH BD Pharmingen

Ly6G1A8PMNs IF,IH[17],BD Pharmingen Ly6C AL-21Monocytes IF[17],BD Pharmingen Gr-1RB6-8C5PMNs,monocytes IF,IH[35],BD Pharmingen F4/80A3-1Macrophages IF,IH BD Pharmingen

CD4530G12

M1-9All leukocytes IF

IH

BD Pharmingen

CD11b/Mac-1M1/70Microglia,monocytes,macrophages IF,IH BD Pharmingen

CD41MWreg30Platelets IF,IH BD Pharmingen

IgG236BA-9200Murine IgG IF,IH BD Pharmingen Antigen Antibody name/clone Marker IF/IH a Reference/source

Human

CD3Polyclonal T lymphocytes IH Dako

CD15C3D-1PMNs,monocytes IH,IF Dako

CD452B11All leukocytes IH,IF Dako

CD68PG-M1Activated macrophages,monocytes,microglia IH,IF Dako

LCA PD7/26All leukocytes IH Dako

Hif1alpha NB100-134Hypoxia IH Novus biologicals c.Caspase-3Asp175Apoptotic cells IH Cell signaling

a Employed in IF immuno?uorescence or IH immunohistochemistry

b Provided by L.Sorokin

c Kindly provide

d by K.von der Mark(Erlangen)

Alexa Fluor594(Molecular Probes).Sections were exam-ined using a Zeiss AxioImager microscope equipped with epi?uorescent optics and documented using a Hamamatsu ORCA ER camera or with a Zeiss confocal laser scanning system LSM510meta.Images were analyzed using Volocity 5.2software(ImproVision,Perkin Elmer).

To quantitate PMN numbers within vessels,in the peri-vascular space or in the brain parenchyma,100-l m coronal sections throughout the entire brain were double immuno-?uorescently stained for pan-laminin and Ly6G or CD45. Ly6G?or CD45?cells located within or outside of the pan-laminin staining were counted in optical sections throughout the thick sections and expressed per0.001mm3brain vol-ume,and normalized to the proportion of the brain volume occupied by vessels(i.e.,total cells9vessel volume/total brain volume).This calculation re?ects the relative numbers of PMNs accumulated in the different brain areas.Cells in or associated with vessels were similarly expressed per m3brain volume,and subsequently normalized to the proportion of the brain volume occupied by vessels.The cells in or associated with vessels as a percentage of the total cell number were calculated from these values.At least5?elds of view were analyzed/section,and at least3different thick sections were analyzed within ischemic lesions from2to3mice.Statistical analyses were employed to determine whether PMN numbers were signi?cantly higher in meninges,cortex or striatum (one-way ANOVA),and to test for deviations from100% Ly6G?cells in or in association with vessels(one-way ANOVA).

In vivo blood–brain barrier permeability

tMCAO/24h reperfusion mice were injected with2% Hoechst33258(Calbiochem)plus2%Evans Blue(Alfa Aesar)(total of2mice),or with a combination of3kDa Texas Red-conjugated Dextran and10kDa FITC-conjugated Dextran,100l g each(Molecular Probes)(total of2mice). The dyes were allowed to circulate for30min and15min, respectively,before sacri?cing the mice.Only dextran-injected mice were perfused with PBS,pH7.4,followed by 4%formaldehyde/PBS,pH7.4,and brains were frozen. Brains from Hoechst/Evans blue-injected mice were snap frozen in TissueTek in a dry ice/isopentane bath.Cryosec-tions were analyzed for possible extravasation of tracers from microvessels of the stroke-af?icted hemisphere,with extravasation across the fenestrated endothelium of the cho-roid plexus serving as internal positive control.

In vitro blood–brain barrier model

Mixed glial cell cultures enriched in astrocytes[41]were cocultured with primary mouse brain microvascular endothelial cells(pMBMECs)[82]and subjected to oxygen and glucose deprivation(OGD)by adding glu-cose-and serum-free medium and keeping the cells under anoxic conditions using GasPack EZ bags(Bec-ton–Dickinson)for4h.For normoxic controls,cells were exposed to serum-free DMEM containing glucose during the OGD period.4h IL-1b(20ng/ml)stimulated pMBMECs under normoxic conditions were also employed.After20h of reoxygenation,the endothelial monolayer,grown on an insert(Millicell CM,Millipore), was placed on a?ow chamber and highly puri?ed bone marrow derived PMNs were perfused over the pMB-MECs.PMNs were allowed to accumulate for4min at low shear stress(0.25dyn/cm2);the subsequent PMN interaction with pMBMECs under physiological shear stress(1.5dyn/cm2)was recorded with an inverted microscope(AxioObserver Z1,Carl Zeiss)at209 magni?cation(objective EC Plan Neo?uar910/0.3). Time-lapse videos were created by taking one image every20s over a20-min period(AxioVision,Carl Zeiss).Image J software was used for the analysis of the movies.pMBMECs were also immunostained for ICAM-1and ICAM-2and counter-stained with Hoechst dye to show the cell nuclei.

Human samples

Brain autopsy and biopsy material of twenty-?ve stroke patients(Supplementary Table1)was analyzed by H&E, chloracetate esterase,myeloperoxidase,immunohistochem-ical,or immuno?uorescent(Table2)staining in accordance with the local ethics committee.The DNA-binding dye TO-PRO-3(Invitrogen,Germany)was employed to mark all nuclei.Infarct staging was performed according to the3-stage stroke classi?cation frequently used in neuropathological diagnostics,which was?rst described in detail by Hugo Spatz in1939[81],and of which cellular reactions have been more sophisticatedly deciphered over the years(for review see [24]).Histopathologically,the infarct lesions were classi?ed into stage I(acute),stage II(subacute),and stage III(chronic). Human brains were?xed in4%phosphate-buffered form-aldehyde;pH7.4,embedded in paraf?n,and3-l m sections were analyzed.Immunohistochemical single and double stainings were performed using the Benchmark and Discov-eryXT immunohistochemistry systems(Ventana/Roche, France)and counterstained with hematoxylin.Primary anti-bodies employed are listed in Table2.Immuno?uorescence images were analyzed and recorded using a Leica TCS SP confocal microscope,and the EZ-C1software.After recording,digital images were further processed and adjusted for brightness,contrast,and colour balance with ImageJ (NIH).

Results

Since C57BL/6mice develop larger infarcts after tMCAO than Sv129mice[59,66]and Sv129mice exhibit higher levels of circulating PMNs compared with C57BL/6mice [65],we studied tMCAO in both mouse strains in parallel (Table1).

Temporal and spatial appearance of polymorphonuclear granulocytes(PMNs)in the brain after transient middle cerebral artery occlusion(tMCAO)

To assess effects of ischemia duration on immune cell recruitment,C57BL/6mice were subjected to tMCAO for 30,60,and90min,and Sv129mice to30and90min (Table1).Since the lesion matures over time,reperfusion times of6,12,18,24,48,72h,1and2weeks were examined to cover the entire acute phase of reperfusion injury in all tMCAO scenarios(Table1).Tissue damage

caused by these tMCAO times is well characterized,with 30min causing mainly selective nerve cell injury and as-trogliosis in the striatum,and60and90min causing selective nerve cell injury involving large parts of the MCA territory[27,50].Immunohistological and immuno?uo-rescence analyses for the presence of various leukocyte populations were investigated in coronal brain sections as described above using the antibodies listed in Table2.

There were no overt differences between C57BL/6and Sv129mice in the time point of?rst appearance or the dis-tribution of CD45?immune cells in the ischemic brains. Flow cytometry(Fig.1a)and immunohistochemistry (Fig.1b)revealed Ly6G?PMNs to be the?rst cell type detectable in the ipsilateral hemisphere,independent of the duration of ischemia.However,the absolute time point of their appearance and how long they were detectable varied with occlusion length.The appearance of Ly6G?PMNs was monophasic,with sparse appearance of PMNs at12h of reperfusion in30and60min tMCAO samples and at3h in 90min tMCAO,in all cases peaking at18–24h(Fig.1a).

Surprisingly,immunohistochemistry localized Ly6G? cells predominantly to the leptomeningeal space of the ischemic hemisphere either con?ned within vessel lumina or closely associated with vessel surfaces regardless of MCA occlusion time(Fig.1b).To a lesser extent,PMNs were also present in the leptomeninges of the contralateral hemisphere. Additional PMNs were detected in close association with arterioles in the motor–sensory cortex(Fig.1b),and only isolated PMNs occurred in the striatum within the lesion core, principally in association with tears in the tissue(Fig.1b). The latter was enhanced in60and90min occlusions where tissue integrity was severely compromised.

The only site where considerable numbers of PMNs were found outside of blood vessels was in the subarachnoid space covering the surface of both brain and optic nerves,remote from the ischemic area.PMNs were not observed in the adjacent cortical layers indicating their failure to penetrate the glia limitans.In accordance with previous reports[38],some PMNs were detected in the parenchyma of the lateral preoptic area(LPO)(Supple-mentary Fig.1).As the LPO is not directly irrigated by the MCA these effects may be due to an indirect occlusion of the anterior medial striate artery that originates from the MCA[23]and irrigates dorso-lateral aspects of the cau-date–putamen.The ophthalmic artery is derived from the internal carotid artery(ICA)that travels ventrally to the optic nerve within the optic canal and is enveloped within a dural sleeve of the optic nerve[40].As the ICA is occluded in the tMCAO model employed,this probably accounts for the meningeal PMN accumulation observed around the optic nerve.Importantly,both the LPO and the meningeal in?ltration around the optic nerve would skew?ow cytometry or biochemical analyses for the presence of PMNs in the brain after experimental ischemic infarct.

Ly6C?CD11b?monocytes were scarce and detectable at24–48h reperfusion in30and60min tMCAO;isolated CD45?CD11b?F4/80?macrophages were detected at 48h,peaking at72h reperfusion.In contrast to Ly6G? PMNs,isolated Ly6C?CD11b?monocytes were also found within the brain parenchyma at24h reperfusion,and at later stages also CD45?CD11b?F4/80?macrophages (Supplementary Fig.2a,b).

Polymorphonuclear granulocyte(PMN)localization

within the neurovascular unit(NVU)

Due to the limitations of immunohistochemistry and con-ventional immuno?uorescence microscopy in localizing Fig.1Localization of PMNs in the ischemic brain following60min of tMCAO and18h and24h reperfusion.a In?ammatory cells were isolated from the ipsilateral ischemic and contralateral hemisphere of 6mice following60min tMCAO and24h reperfusion by enzymatic digestion and density gradient centrifugation,and analyzed by?ow https://www.wendangku.net/doc/fb11487624.html,ing Forward(FSC)and Side Scatter(SSC)pro?les in?ammatory cells were separated according to size(FSC)and granularity(SSC),respectively,and displayed in a dot blot.PMNs are characterized by a high SSC signal due to their high content of granules.Such a population was only found in the ipsilateral and not in the contralateral hemisphere.Positive Ly6G immunoreactivity of the scatter gated population,as depicted in the histogram(right side), con?rmed their identity as PMNs.b Schematic representation of the two planes of the brain examined(X Bregma0.50mm,Y Bregma -2.46mm)and corresponding coronal sections showing areas analyzed.Immunohistochemistry of sections from60min tMCAO and24h perfusion for CD45and Ly6G reveal distribution of total leukocytes and PMNs,respectively,in the meninges(Mng),motor–sensory cortex(Ctx),and striatum(Str).The majority of cells were detected either intra-or perivascular in the meninges(arrows)and the penumbral cortex(arrowheads)and the cells comprised mainly Ly6G?PMNs.Bar is50l m

c

cells to de?ned layers within the NVU,?uorescently stained100-l m sections were analyzed by confocal microscopy to obtain optical sections and3D reconstruc-tions.Thirty and60min tMCAO at18and24h reperfusion were examined.Sections were double stained with Ly6G or CD45and pan-laminin antibody which rec-ognizes the majority of the laminin isoforms,or a speci?c marker of the endothelial BM,laminin a5,permitting localization of PMNs within vessel lumina or in the peri-vascular space,as de?ned by the endothelial and parenchymal BMs(termed vessel associated),or outside of the endothelial and parenchymal BMs and hence within the CNS parenchyma(termed intraparenchymal)(Fig.2a,b). Figure2c illustrates that most CD45?leukocytes and, speci?cally,Ly6G?PMNs localized within the lumina of vessels of30–40l m diameter,predominantly representing arterioles,in the meninges and cortex,and did not occur in the ischemic core.Total numbers of CD45?and Ly6G? cells in the meninges,cortex,and striatum were counted and expressed per mm3brain volume,and subsequently normalized to the proportion of the brain volume occupied by vessels,permitting assessment of the relative numbers of PMNs accumulated in the different brain areas,reveal-ing an overall low number of PMNs and their tendency to localize to the meninges(although this was not statistically

signi?cant)(Fig.2d).At18and24h reperfusion,numbers of CD45?and Ly6G?cells did not differ signi?cantly and data shown in Fig.2d are for Ly6G?cells.Cells associated with vessels were similarly expressed per mm3brain vol-ume,and subsequently normalized to the proportion of the brain volume occupied by vessels.The percentage of vessel associated Ly6G?cells was calculated using these nor-malized values(Fig.2d).Statistical analyses revealed the absence of signi?cant deviations from100%cells asso-ciated with vessels in the striatum(mean90%±7; P=0.866),meninges(mean94%±4;P=0.766),and cortex(mean89%±7;P=0.56)at both18and24h reperfusion,regardless of occlusion time(data for60min tMCAO,24h reperfusion is shown in Fig.2d).PMNs found were also not associated with CD41?platelets (Fig.3)or erythrocytes,indicating the absence of any hemorrhagic transformation.

Polymorphonuclear granulocyte(PMN)accumulation

is not associated with upregulated endothelial adhesion molecule expression,platelet aggregation or vascular permeability

PMN accumulation within blood vessel lumina suggests endothelial cell activation or aberrant endothelial cell physiology.To investigate potential correlations between PMN localization and local altered expression of endo-thelial cell adhesion molecules implicated in PMN adhesion to or migration across the vascular wall in in?ammation,speci?cally P-selectin,ICAM-1,ICAM-2, and PECAM-1were analyzed in60min tMCAO samples at6,12,18,24h of reperfusion using confocal immuno-?uorescence microscopy.No differences in staining patterns or intensity were observed for PECAM-1between lesioned and non-lesioned hemispheres(not shown).As previously reported,immuno?uorescence staining for vas-cular ICAM-1and VCAM-1was more extensive in the ischemic area[16,51,63],principally in vessels in the meninges and cortex(Fig.3).Staining for P-selectin, which is not constitutively expressed in the brain paren-chyma[22],was enhanced in isolated vessels in the penumbra at12h and24h reperfusion(Fig.3).Despite enhanced immuno?uorescence staining for P-selectin, ICAM-1and VCAM-1in many vessels of the lesioned hemisphere compared to the non-lesioned hemisphere, there was no strict spatial correlation of increased adhesion molecule expression with sites of vascular PMN accumu-lation and[90%of the vessels showing increased VCAM-1and ICAM-1staining were not associated with Ly6G? cells(Fig.3)nor was PMN localization associated with platelet aggregates,as determined by double immuno?uo-rescence staining for Ly6G and CD41(Fig.3).Rather, platelet aggregations were localized mainly to the lumina of dilated vessels both in the absence and presence of PMNs(Fig.3).Finally,neutrophil extracellular traps (NETs),which have been reported to trap PMNs within Fig.2Localization of neutrophils in the neurovascular unit.a Con-focal microscopy of a thick section(100l m)double stained for laminin a5,showing the inner endothelial BM,and laminin a2, showing the outer parenchymal BM,and b schematic representation of the constituents of the NVU.Relative sizes and numbers of the NVU constituents are not to scale.M?refers to macrophages and DC is dendritic cells.c Schematic representation of the plane of the brain examined(X Bregma0.50mm)and corresponding coronal section showing areas analyzed(striatum Str,meninges Mng and motor–sensory cortex Ctx).Corresponding confocal microscopy of thick sections from plane X of60min tMCAO,24h reperfusion,double stained with antibodies to pan-laminin,to mark all BMs,and CD45, or to laminin a5,as a marker of the endothelial BM,and Ly6G,reveal localization of total leukocytes and PMNs mainly in association with arteries or large arterioles(outer panels)and veins(middle panel). Images shown are from area Ctx.d Analyses of individual Z stacks permitted localization of Ly6G?PMNs within vessel lumina or between the endothelial and parenchymal BM(vessel associated),or intra-parenchymally.Staining shown is for an arteriole.Graphs to the right show total Ly6G?PMN numbers/0.001mm3brain volume normalized to the proportion of the brain volume occupied by vessels in Str striatum,Mng meninges and Ctx cortex.Normalized Ly6G? PMN numbers associated with vessels in the different areas are expressed as percentages of the normalized total Ly6G?cell numbers and show no statistically signi?cant differences from100%cells associated with vessels(P values0.56–0.86).Data shown are mean±SEM from2to3mice.NS is no statistically signi?cant difference.Bars in c and d are40l m

c

vessels lumens,were investigated by staining for DNA as previously described [85],revealing the absence of vas-cular NET formation in any of the samples analyzed.Importantly,PMN accumulation within blood vessels was not associated with local changes in vascular perme-ability as shown by triple staining for murine immunoglobulin (IgG),Ly6G and pan-laminin (Supple-mentary Fig.3a).In general,the presence of IgG in the brain parenchyma was detected in 40%of all 60min tMCAO 18h and 24h samples,mostly observed around larger vessels in the cortex,and was not correlated with sites of Ly6G ?PMN accumulation (Supplementary Fig.3a).To further address possible in vivo poststroke BBB leakage,60tMCAO/24h reperfusion mice were intravenously injected with either Evans Blue and Hoechst 33258or 3kDa Texas Red-Dextran and 10kDa FITC-Dextran prior to sacri?ce.In addition to staining of endo-thelial cell nuclei,Hoechst 33258labelled nuclei of neurons in the lateral preoptic area (LPO)of the ischemic hemisphere.Extravasation of Evans blue (Supplementary Fig.3b)or labelled dextrans (data not shown)was fur-thermore observed only around some dilated vessels within the striatum or around focal arterioles in the penumbra in the ischemic hemisphere.

Oxygen and glucose deprivation fails to induce

polymorphonuclear granulocyte (PMN)extravasation across the blood–brain barrier under physiological ?ow in vitro

Our observations suggest that in contrast to in?ammatory stimuli,ischemia/reperfusion fails to induce the signals in brain endothelial cells required to mediate the extravasation of cir-culating PMNs into the brain parenchyma.To mimic PMN interactions with the BBB under physiological ?ow (1.5dyn/cm 2)after ischemia/reperfusion in vitro,glial cells in coculture with primary mouse brain microvascular endothelial cells (pMBMECs)were exposed to normoxic conditions or 4h of oxygen and glucose deprivation (OGD)followed by 20h of reoxygenation.For live cell imaging,the endothelial mono-layer was placed in the ?ow chamber and highly puri?ed bone marrow derived PMNs were perfused over the pMBMECs.The dynamic PMN interaction with the pMBMECs was recorded under constant ?ow for https://www.wendangku.net/doc/fb11487624.html,parisons were made with cocultures treated with or without IL-1b under normoxic conditions.While PMNs were able to arrest,crawl,and diapedese across the IL-1b treated pMBMECs,pMB-MECs exposed to OGD/reoxygenation induced brief PMN arrest and crawling but did not result in PMN diapedesis

across

Fig.3Endothelial adhesion molecules or platelet accumulation are not suf?cient for Ly6G ?PMN extravasation in vivo.Data shown are for 60min tMCAO and 24h reperfusion (16l m sections).Double immuno?uorescence staining for Ly6G and intercellular adhesion molecule-1(ICAM-1)and 2(ICAM-2),vascular endothelial cell adhesion molecule (VCAM-1),or P-selectin showed some upregula-tion of adhesion molecules in cortical and meningeal microvessels of

the ischemic hemisphere but no strict spatial correlation with Ly6G ?PMNs which localize intraluminally (arrows ),perivascularly (arrow-head )or in the meningeal compartment (asterisk ).Immuno?uorescence staining for CD41?platelets and Ly6G revealed that platelet accumu-lation in vessels was not associated with the localization of PMNs.Bars are 40l m

the endothelium,thus resembling non-stimulated normoxic conditions (Fig.4a,b;Supplementary videos 1–3).Surpris-ingly,OGD/reoxygenation upregulated cell surface expression of adhesion molecules,as exempli?ed by the increased immunostaining for ICAM-1on the pMBMECs (Fig.4c),which was similar to that induced by IL-1b treatment.These data indicate that although ICAM-1is upregulated on brain endothelium under ischemic conditions in vitro and in vivo,ischemia/reperfusion fails to induce the sum of traf?c signals in brain endothelial cells required to promote PMN transmigra-tion across the endothelial monolayer.

Localization of polymorphonuclear granulocytes (PMNs)in human stroke specimens

To determine whether our ?ndings were relevant to human stroke,25specimens [24autopsy cases including 17pure acute stroke (stage I,Fig.5)and 8mixed (apart from stage I also showing stage II or III infarct regions in other CNS areas)and one biopsy specimen (Supplementary Table 1)]were examined by immunohistochemistry and immuno-?uorescence microscopy as described for the mouse tMCAO samples.As anti-human Ly6G antibodies are not available,morphology together with CD15?immuno-staining (which mainly recognizes PMNs and a subset of monocytes)and enzyme histochemistry for myeloperoxi-dase and chloracetate esterase were employed to identify PMNs.Very few PMNs were detected in both early infarct stages (stage I)and at stages of resorption (stage II)(Supplementary Fig.4),with the majority of the PMNs being localized either within the lumen of blood vessels (Fig.5d)or in the perivascular space between the endo-thelial and parenchymal BMs visualized using a pan-collagen type IV antibody (data not shown).Absence

of

Fig.4PMN interaction with endothelium in an in vitro blood–brain barrier model.PMN interaction with pMBMECs under normoxic,ischemic (OGD ?reox)or IL-1b stimulated conditions under physiological ?ow (1.5dyn/cm 2)was recorded for 20min and the dynamic behaviour of arrested PMNs was analyzed.a The number of PMNs arrested on pMBMECs counted per ?eld of view (FOV).b Proportions of PMNs that were stationary,crawling,undergoing diapedesis or detachment from pMBMECs.PMNs that remained immobile on the monolayer were de?ned as ‘Stationary’,PMNs that polarized and crawled on the monolayer but did not diapedese across the endothelial monolayer were described as ‘Crawling’,PMNs that crawled until they found a suitable site for diapedesis were de?ned as undergoing ‘Diapedsis’,and PMNs that detached during the video acquisition time were termed ‘Detachment’.Data in a and b are mean ±SD,n =3.c Immuno?uorescence staining of pMBMECs for ICAM-1and ICAM-2under normoxic,ischemic,and IL-1b stimu-lated conditions shows upregulation of ICAM-1under both,ischemic and IL-1b stimulation,ICAM-2staining remains unaffected.Endo-thelial cells are counter-stained with Hoechst dye to show the cell nuclei.Bar 50l m

granulocytic in?ltration into the CNS parenchyma was especially noted in very acute stroke lesions (\48h),even though this has been proposed to be the main time frame for PMNs to invade infarcted brain tissue after CNS ischemia.Analyses of samples of such \48h infarct lesions revealed the appearance of cells morphologically resembling PMNs not only in vessels but also in the CNS parenchyma;however,CD15staining was restricted to cells within vessel lumina (Fig.5d).Indeed,upon careful examination multiple cells showing PMN morphology in the CNS parenchyma were found to be positive for cleaved caspase-3(Fig.5e),suggesting that they represent apop-totic bodies,which morphologically are easily confused with PMNs due to their fragmented nuclei.Already at this early infarct stage,a low amount of extravasated CD68?monocytic cells was observed (Fig.5f).Using this com-bination of CD15staining,together with morphology and enzyme histochemistry for myeloperoxidase and chlorac-etate esterase (not shown)to identify PMNs in very early infarct lesions,rare in?ltration in the subarachnoid and the subpial space,in the cortical layers I and II (not shown)and the Virchow-Robin space was observed.No PMNs were

detected in the inner cortical layers or in the infarct center and border zones.Even at later stages after infarction PMNs remained con?ned to vessel lumina,despite exten-sive presence of CD45-positive leukocytes (not shown)which mainly consisted of CD163-negative (not shown)and CD68-positive macrophages and activated microglia and a moderate fraction of CD3-positive T-cells (while CD20-positive B-cells were virtually absent)and aberrant dilated appearance of vessels (Supplementary Fig.4),indicative of ischemia.

Discussion

By bringing together stroke researchers,neuropathologists,cell biologists,and neuroimmunologists specialized on the cellular and extracellular matrix components of the NVU and immune cell penetration of the NVU,we have been able to comprehensively investigate the in vivo PMN localization after ischemic stroke in mouse and human samples.Our data support an early appearance of PMNs after ischemic stroke in both mouse tMCAO and in

human

Fig.5Histopathology of human acute stroke specimens (Stage I).a H&E staining of human stage I stroke specimen demonstrating demarcation of the ischemic core from the penumbra (arrows );b H&E staining (high magni?cation of a )showing the presence of eosinophilic neurons indicating an early ischemic neuronal damage;c At the infarct border zone,eosinophilic neurons (arrowhead ,as depicted in Fig.5b)are surrounded by glial cells showing severe hypoxic changes (arrows )as indicated by the strong expression of hypoxia-inducible factor-1alpha (Hif1alpha);d CD15immunohisto-chemistry indicating that PMNs are primarily located within blood vessels in acute human stroke lesions;e characterization of cells with

histomorphological features of PMNs within the CNS parenchyma:while intravascular cells with PMN-like morphology (as seen in Fig.5d)were strongly CD15-positive,intraparenchymally located cells exhibiting PMN-like morphology and being CD15-negative are strongly positive for cleaved caspase-3(arrow )indicating that these cells undergo apoptosis;f CD68-positive cells of the monocytic lineage are mainly located in the perivascular space or within the brain parenchyma.Data shown are from a 75-year-old male patient suffering from an acute right parietal ischemic infarct (for details see Supplementary Table 1)

samples,as shown by others[21],but contrary to previous concepts our data show that PMNs are(1)limited in number,(2)associate with vessel lumina or the perivas-cular and leptomeningeal space,and(3)do not strictly correlate with either platelet aggregates,sites of increased vessel permeability,or sites of enhanced expression of endothelial adhesion molecules known to be required for PMN extravasation in in?ammation.Studies of CNS autoimmune in?ammation have shown that localization of immune cells within the leptomeningeal and perivascular space is not suf?cient to induce disease symptoms[1,8]. Rather,penetration of the parenchymal BM is required before access to CNS parenchyma and induction of path-ological processes is possible[1,84].Hence,the localization of PMNs to the vasculature early after ische-mic stroke necessitates reassessment of their role in stroke.

Although some adhesion molecules,such as VCAM-1, were upregulated on vessels in the ischemic hemisphere, the expression of adhesion molecules was heterogeneous with some vessels having low and others high expression levels,and there was no spatial correlation with PMN accumulation within vessels or in the perivascular space. While previous studies have investigated adhesion mole-cules in ischemic stroke showing results similar to those obtained here[16,51,63],no previous study has correlated in vivo adhesion molecule expression with localization of PMNs.Indeed,most studies have involved?ow cytometry or myeloperoxidase expression in excised brains to quan-tify PMNs while adhesion molecules were analyzed by immuno?uorescence microscopy on tissue sections,which led to the false conclusion that the two are correlated.This has also been the justi?cation for employing mice lacking ICAM-1,or the use of function blocking antibodies tar-geting adhesion molecules in MCAO experiments,which have produced variable results[9,36,47].Normally, extravasation of PMNs during in?ammation occurs at the level of postcapillary venules[69]and involves E-and P-selectin-mediated rolling on the endothelial cell surface, and subsequent ICAM-1mediated arrest and diapedesis across the endothelial cell monolayer[31,55].The absence of a spatial correlation between upregulated expression of endothelial P-selectin,VCAM-1and ICAM-1and vascular sites of PMN accumulation in the tMCAO samples sug-gests the absence of the complete cascade of these events and that the mode of endothelial activation that occurs after ischemic stroke is not suf?cient to trigger PMN extrava-sation into the brain parenchyma.This is supported by the in vitro studies involving pMBMECs,which demonstrated that while OGD/reoxygenation can upregulate endothelial ICAM-1this was not suf?cient to support transmigration of PMNs across the pMBMEC monolayer.In addition,the failure of those few PMNs that enter the perivascular space to penetrate into the brain parenchyma proper also re?ects the absence of the molecular signals required for their invasion into the CNS as observed in in?ammation.

The concept that the brain parenchyma is a tissue that is unique in its resistance to leukocyte diapedesis has previ-ously been suggested by others,who have shown that even direct intracerebral injection of chemotactic cytokines that are suf?cient to induce PMN extravasation into other tis-sues fail to trigger PMN extravasation into the brain parenchyma[4].Thus,PMN migration from the blood stream across the BBB and the glia limitans into the brain parenchyma requires more than presence of chemotactic factors and induction of leukocyte adhesion to cerebral endothelium.

The precise molecular mechanism involved in PMN accumulation within vessels observed in the current study is not clear and we can only speculate on the molecules involved.The fact that PMNs accumulated in larger ves-sels,mainly arterioles,further supports the hypothesis that the molecular mechanism/s involved are distinct from those described above for postcapillary venules.A recent study involving transient ligation and reperfusion of the vena cava reported a similar PMN accumulation that was not associated with expression of the classical adhesion molecules or platelets,but rather with DNA NETs released by PMNs and resulting in their aggregation[85].This was not the case here,as NETS were not detected in any of the tMCAO samples.However,it is possible that hypoxia or changes in blood?ow,resulting from vessel occlusion, alters the expression of as yet unidenti?ed adhesion mol-ecules,either upregulating molecules that promote PMN adherence but also downregulating anti-adhesive mole-cules.The endothelial glycocalyx,composed largely of glycosaminoglycans carrying highly negatively charged heparin sulfate chains,is known to repel cells in the cir-culation,and the depletion of heparan sulfate chains signi?cantly enhances immune cell adhesion[57,58].As the glycocalyx changes with blood?ow[42],it is con-ceivable that it may present part of a novel adhesion mechanism in larger vessels.This is supported by the recent identi?cation of a role for myeloperoxidase,released from PMNs,in charged interactions between PMNs and the gylcocalyx of endothelium which promotes PMN adher-ence[53].While this is a promising?eld,the role of charge molecules such as cell surface glycans has rarely been considered in immune cell recruitment,mainly due to the complex chemistry associated with their analysis.

Importantly,our data provide evidence that previous studies have overestimated PMN contribution to stroke because of the methods used for their identi?cation and shed new light on previous animal studies and clinical trials.The largest majority of previous studies have focused on permanent MCAO,rather than the temporary MCAO employed here,which may be one reason for the

comparatively low PMN numbers detected in our study.In addition,the high numbers of PMNs detected in the lep-tomeningeal areas,especially surrounding the optical nerve as detected here but also reported previously[38],suggest that previous studies solely based on?ow cytometry or enzymatic assessment of PMN in?ltration into the brain are likely to have equated these?ndings with number of PMNs present within the ischemic brain parenchyma.Myeloper-oxidase,commonly used as a PMN marker,is expressed by PMNs,monocytes and activated microglia present in the ischemic brain[13],while the Gr-1antibody(clone RB6-8C5),frequently used to identify PMNs,recognizes both Ly6C and Ly6G expressed by monocytes and PMNs, respectively[35].The use of these tools would therefore inevitably lead to an over-estimation of PMNs.Only few studies have attempted to localize PMNs in the brain parenchyma at de?ned times after MCAO,including that of Garcia and Kamijyo[38],who employed electron microscopy of permanent MCAO in rats.Consistent with our data,the majority of the PMNs were found in vessels, and while rare isolated PMNs were detected outside of the endothelial layer,the magni?cations shown are too high to de?nitively state that the PMN is in the brain parenchyma (and not in the perivascular space)nor do they provide evidence for neuronal death in the close vicinity of PMNs, rather isolated images of damaged neurons are shown.The concept that PMNs contribute to reperfusion injury at early stages after ischemic stroke has led to clinical trials tar-geting PMNs to minimize infarct volume,most of which have not been successful[25].While this may in part be attributable to factors such as patient cohort size or demography,or adverse effects of the PMN targeting strategies[36],the data presented here suggest that the role of PMNs in ischemic stroke remains unclear and needs further investigation before their consideration as a rele-vant therapeutic target.

The PMN speci?c anti-Ly6G antibody(clone1A8) employed here[17],in conjunction with immuno?uores-cence staining for de?ned markers of the vascular basement membranes and confocal analyses have permit-ted a reliable identi?cation and localization of PMNs in ischemic brain sections,and highlight the importance of supplementing quantitative analyses of ischemic tissue by in vivo localization studies.However,the choice of such in vivo imaging technologies is also crucial;while intra-vital microscopy(IVM)via a cortical window has been used to assess in vivo leukocyte–endothelial cell interac-tions in the ischemic brain[5,48],the working depth achievable is limited and in addition only meningeal postcapillary venules run parallel to the surface of the skull,whereas cortical postcapillary venules are orientated perpendicularly to the surface of the brain and the focal plane[46,71].Hence,it is impossible to discern leukocyte–endothelial interactions in cortical postcapillary venules as this requires tracing of leukocytes over a de?ned period along a certain vessel length.As meningeal and parenchymal blood vessels differ from one another in several aspects[64],including the absence of a glial en-sheathment in leptomeningeal vessels[70]and the constitutive expression of P-selectin in meningeal but not parenchymal microvessels[6],studying meningeal vessels in stroke is not a substitute for cortical microvessels.Given our detection of a high number of PMNs in the leptome-ninges,intravital imaging of such areas might overestimate PMN recruitment into the brain after stroke.Taken toge-ther,this suggests that confocal microscopic analysis of thick brain sections using de?ned markers of immune cells and the borders of the NVU is,at present,the most ef?cient mode of assessing immune cell in?ltration after ischemic stroke in experimental models.

The analysis of human autopsy samples corroborated the observation that PMNs rarely occur in the brain paren-chyma early after stroke.Clearly,it is impossible to prove that the patient samples analyzed had undergone reperfu-sion after the vessel occlusion/brain ischemia and to de?ne this histologically.However,spontaneous reperfusion is a frequent phenomenon in clinical stroke,which may also occur in parts of the ischemic area,and even in patients receiving thrombolysis often some brain tissue undergoes infarction despite reperfusion[43,74].Hence,a spectrum ranging from no reperfusion to different times and extent of reperfusion are likely in such autopsy samples,validating the comparison with the mouse tMCAO data.The question therefore arises how the concept that PMN leukocytes strongly in?ltrate the brain parenchyma within the?rst 24h after onset of brain infarction has gained access into widely accepted neurological and neuropathological text-books[33].Several studies have de?ned the time frame of PMN detection in the brain after ischemic stroke,mainly using SPECT and CT scan analyses of injected tracer immune cells[2,87].Already these studies revealed con-?icting results about when and over which time period PMNs are detectable in the brain;however,the discrep-ancies were attributed to the injection of mixed leukocytes [87]versus puri?ed PMNs prior to SPECT[2].While SPECT permits high temporal resolution of such tracer cells,it has poor spatial resolution and does not permit the distinction between PMNs within or outside the con?nes of the NVU.A second point of con?ict might be the extent of foci of fresh bleedings in mainly ischemic infarction.Even selected pictures from classical textbooks[33],employed as evidence for PMN entry into the brain parenchyma after ischemic stroke,depict fresh foci of hemorrhages and associated leakage of various cellular populations includ-ing erythrocytes and monocytes,indicative of more severe vascular disruption.Therefore,vascular disruption might

be a very logical explanation for the detection of PMNs in the CNS parenchyma in stroke cases accompanied by hemorrhages.However,these?ndings differ considerably from classical forms of purely ischemic stroke lesions. Finally,pioneering studies addressing the role of PMNs in stroke did not have access to the complex panel of immunocytochemical markers,available now,to precisely de?ne cell types and their localization in relation to the cellular and BMs layers of the NVU,but rather relied mainly on classical histomorphological examination[38, 79].These drawbacks might constitute problems if sections are assessed by histology only where PMNs may be con-fused with different stages of apoptotic?gures,which also frequently occur in hypoxic-ischemic brain lesions,due to their similar morphology.While isolated studies exist,like that of Lindsberg et al.[56],which included early human stroke samples without secondary hemorrhages showing CD15-positive cells in the lesioned CNS,even these state that granulocytes in early infarction are mostly found within the intravascular space and typically aggregate at the walls of CNS vessels and are,therefore,in accordance with our?ndings.However,Lindsberg et al.admit that a systematic evaluation of morphological details such as whether granulocytes were still surrounded by a capillary lumen was not possible in their cohort.

In conclusion,our data indicate that PMNs do not gain access to the brain parenchyma early after ischemic stroke and highlight the vascular compartment of the NVU rather than neurons,as previously suggested also by others[20], as the site of potential PMN action.

Acknowledgments The authors thank P.Ko¨ckemann for help with animals,R.Hallmann for critical reading of the manuscript,and S. Butz and D.Vestweber for kind gifts of antibodies.The work was supported by the European Union’s Seventh Framework Programme (FP7/2007–2013)grant agreements n°201024and n°202213(Euro-pean Stroke Network)to BE and LS,the Olga-Mayen?sch Foundation to BE,the German Research Council DFG(SFB-TR43)and Federal Ministry for Education and Research(BMBF Centre for Stroke Research),and the Medical Faculty of the University of Mu¨nster, Germany.ME and UD thank the Excellence Cluster‘NeuroCure’, Charite′-Universita¨tsmedizin Berlin,for support.BE thanks the Microscopy Imaging Center of the University of Bern for support. Open Access This article is distributed under the terms of the Creative Commons Attribution License which permits any use,dis-tribution,and reproduction in any medium,provided the original author(s)and the source are credited.

References

1.Agrawal S,Anderson P,Durbeej M,van Rooijen N,Ivars F,Op-

denakker G,Sorokin LM(2006)Dystroglycan is selectively cleaved at the parenchymal basement membrane at sites of leukocyte extravasation in experimental autoimmune encephalomyelitis.

J Exp Med203:1007–1019

2.Akopov SE,Simonian NA,Grigorian GS(1996)Dynamics of

polymorphonuclear leukocyte accumulation in acute cerebral infarction and their correlation with brain tissue damage.Stroke 27:1739–1743

3.Alcolado R,Weller RO,Parrish EP,Garrod D(1988)The cranial

arachnoid and pia mater in man:anatomical and ultrastructural observations.Neuropathol Appl Neurobiol14:1–17

4.Andersson PB,Perry VH,Gordon S(1992)Intracerebral injection

of proin?ammatory cytokines or leukocyte chemotaxins induces minimal myelomonocytic cell recruitment to the parenchyma of the central nervous system.J Exp Med176:255–259

5.Arumugam TV,Salter JW,Chidlow JH,Ballantyne CM,Kevil

CG,Granger DN(2004)Contributions of LFA-1and Mac-1to brain injury and microvascular dysfunction induced by transient middle cerebral artery occlusion.Am J Physiol Heart Circ Physiol287:H2555–H2560

6.Barkalow FJ,Goodman MJ,Gerritsen ME,Mayadas TN(1996)

Brain endothelium lack one of two pathways of P-selectin-med-iated neutrophil adhesion.Blood88:4585–4593

7.Barone FC,Schmidt DB,Hillegass LM,Price WJ,White RF,

Feuerstein GZ,Clark RK,Lee EV,Griswold DE,Sarau HM (1992)Reperfusion increases neutrophils and leukotriene B4 receptor binding in rat focal ischemia.Stroke23:1337–1347 8.Bartholomaus I,Kawakami N,Odoardi F,Schlager C,Miljkovic

D,Ellwart JW,Klinkert WE,Flugel-Koch C,Issekutz TB,We-kerle H,Flugel A(2009)Effector T cell interactions with meningeal vascular structures in nascent autoimmune CNS lesions.Nature462:94–98

9.Becker KJ(2002)Anti-leukocyte antibodies:LeukArrest

(Hu23F2G)and Enlimomab(R6.5)in acute stroke.Curr Med Res Opin18Suppl2:s18–s22

10.Bederson JB,Pitts LH,Tsuji M,Nishimura MC,Davis RL,

Bartkowski H(1986)Rat middle cerebral artery occlusion: evaluation of the model and development of a neurologic examination.Stroke17:472–476

11.Beray-Berthat V,Croci N,Plotkine M,Margaill I(2003)Poly-

morphonuclear neutrophils contribute to infarction and oxidative stress in the cortex but not in the striatum after ischemia-reper-fusion in rats.Brain Res987:32–38

12.Bratane BT,Bouley J,Schneider A,Bastan B,Henninger N,

Fisher M(2009)Granulocyte-colony stimulating factor delays PWI/DWI mismatch evolution and reduces?nal infarct volume in permanent-suture and embolic focal cerebral ischemia models in the rat.Stroke40:3102–3106

13.Breckwoldt MO,Chen JW,Stangenberg L,Aikawa E,Rodriguez

E,Qiu S,Moskowitz MA,Weissleder R(2008)Tracking the in?ammatory response in stroke in vivo by sensing the enzyme myeloperoxidase.Proc Natl Acad Sci USA105:18584–18589 14.Cai W,Guzman R,Hsu AR,Wang H,Chen K,Sun G,Gera A,

Choi R,Bliss T,He L,Li ZB,Maag AL,Hori N,Zhao H, Moseley M,Steinberg GK,Chen X(2009)Positron emission tomography imaging of poststroke angiogenesis.Stroke 40:270–277

15.Connolly ES Jr,Winfree CJ,Prestigiacomo CJ,Kim SC,Cho-

udhri TF,Hoh BL,Naka Y,Solomon RA,Pinsky DJ(1997) Exacerbation of cerebral injury in mice that express the P-selectin gene:identi?cation of P-selectin blockade as a new target for the treatment of stroke.Circ Res81:304–310

16.Connolly ES Jr,Winfree CJ,Springer TA,Naka Y,Liao H,Yan

SD,Stern DM,Solomon RA,Gutierrez-Ramos JC,Pinsky DJ (1996)Cerebral protection in homozygous null ICAM-1mice after middle cerebral artery occlusion.Role of neutrophil adhe-sion in the pathogenesis of stroke.J Clin Invest97:209–216 17.Daley JM,Thomay AA,Connolly MD,Reichner JS,Albina JE

(2008)Use of Ly6G-speci?c monoclonal antibody to deplete neutrophils in mice.J Leukoc Biol83:64–70

18.del Zoppo GJ(2010)Acute anti-in?ammatory approaches to

ischemic stroke.Ann N Y Acad Sci1207:143–148

19.del Zoppo GJ(2004)Chapter9:Lessons from stroke trials using

anti-in?ammatory approaches that have failed.In:Dirnagl U, Elger B(eds)Neuroin?ammation in stroke.Springer,Berlin, pp155–184

20.del Zoppo GJ(2009)In?ammation and the neurovascular unit in

the setting of focal cerebral ischemia.Neuroscience158:972–982 21.Dirnagl U,Iadecola C,Moskowitz MA(1999)Pathobiology of

ischaemic stroke:an integrated view.Trends Neurosci 22:391–397

22.Doring A,Wild M,Vestweber D,Deutsch U,Engelhardt B

(2007)E-and P-selectin are not required for the development of experimental autoimmune encephalomyelitis in C57BL/6and SJL mice.J Immunol179:8470–8479

23.Dorr A,Sled JG,Kabani N(2007)Three-dimensional cerebral

vasculature of the CBA mouse brain:a magnetic resonance imaging and micro computed tomography study.Neuroimage 35:1409–1423

24.Elison D,Love S,Chimeli L,Harding BN,Lowe J,Vinters HV

(2004)Neuropathology:a reference text of CNS pathology.

Elsevier,Mosby

25.Emerich DF,Dean RL3rd,Bartus RT(2002)The role of leu-

kocytes following cerebral ischemia:pathogenic variable or bystander reaction to emerging infarct?Exp Neurol173:168–181 26.Endres M,Fink K,Zhu J,Stagliano NE,Bondada V,Geddes JW,

Azuma T,Mattson MP,Kwiatkowski DJ,Moskowitz MA(1999) Neuroprotective effects of gelsolin during murine stroke.J Clin Invest103:347–354

27.Endres M,Namura S,Shimizu-Sasamata M,Waeber C,Zhang L,

Gomez-Isla T,Hyman BT,Moskowitz MA(1998)Attenuation of delayed neuronal death after mild focal ischemia in mice by inhibition of the caspase family.J Cereb Blood Flow Metab 18:238–247

28.Engelhardt B,Conley FK,Kilshaw PJ,Butcher EC(1995)

Lymphocytes in?ltrating the CNS during in?ammation display a distinctive phenotype and bind to VCAM-1but not to MAdCAM-

1.Int Immunol7:481–491

29.Engelhardt B,Laschinger M,Schulz M,Samulowitz U,Vest-

weber D,Hoch G(1998)The development of experimental autoimmune encephalomyelitis in the mouse requires a4-integrin but not a4b7-integrin.J Clin Invest102:2096–2105

30.Engelhardt B,Martin-Simonet MT,Rott LS,Butcher EC,Michie

SA(1998)Adhesion molecule phenotype of T lymphocytes in in?amed CNS.J Neuroimmunol84:92–104

31.Engelhardt B,Ransohoff RM(2005)The ins and outs of

T-lymphocyte traf?cking to the CNS:anatomical sites and molecular mechanisms.Trends Immunol26:485–495

32.Engelhardt B,Sorokin L(2009)The blood-brain and the blood-

cerebrospinal?uid barriers:function and dysfunction.Semin Immunopathol31:497–511

33.Ferrer I,Kaste M,Kalimo H(2008)Vascular diseases In:Love S,

Louis DN,Ellison DW(eds)Green?eld‘s neuropathology.Hod-der Arnold,London,pp121–240

34.Fisher M,Feuerstein G,Howells DW,Hurn PD,Kent TA,Savitz

SI,Lo EH(2009)Update of the stroke therapy academic industry roundtable preclinical recommendations.Stroke40:2244–2250 35.Fleming TJ,Fleming ML,Malek TR(1993)Selective expression

of Ly-6G on myeloid lineage cells in mouse bone marrow.RB6-8C5mAb to granulocyte-differentiation antigen(Gr-1)detects members of the Ly-6family.J Immunol151:2399–2408

36.Furuya K,Takeda H,Azhar S,McCarron RM,Chen Y,Ruetzler

CA,Wolcott KM,DeGraba TJ,Rothlein R,Hugli TE,del Zoppo GJ,Hallenbeck JM(2001)Examination of several potential mechanisms for the negative outcome in a clinical stroke trial of

enlimomab,a murine anti-human intercellular adhesion mole-cule-1antibody:a bedside-to-bench study.Stroke32:2665–2674 37.Garau A,Bertini R,Colotta F,Casilli F,Bigini P,Cagnotto A,

Mennini T,Ghezzi P,Villa P(2005)Neuroprotection with the CXCL8inhibitor repertaxin in transient brain ischemia.Cytokine 30:125–131

38.Garcia JH,Kamijyo Y(1974)Cerebral infarction.Evolution of

histopathological changes after occlusion of a middle cerebral artery in primates.J Neuropathol Exp Neurol33:408–421

39.Garcia JH,Liu KF,Yoshida Y,Lian J,Chen S,del Zoppo GJ

(1994)In?ux of leukocytes and platelets in an evolving brain infarct(Wistar rat).Am J Pathol144:188–199

40.Glaser JS(1999)Neuro-ophthalmology.Lippincott Williams&

Wilkins,Philadelphia

41.Gorina R,Santalucia T,Petegnief V,Ejarque-Ortiz A,Saura J,

Planas AM(2009)Astrocytes are very sensitive to develop innate immune responses to lipid-carried short interfering RNA.Glia 57:93–107

42.Gouverneur M,Berg B,Nieuwdorp M,Stroes E,Vink H(2006)

Vasculoprotective properties of the endothelial glycocalyx: effects of?uid shear stress.J Intern Med259:393–400

43.Hacke W,Albers G,Al-Rawi Y,Bogousslavsky J,Davalos A,

Eliasziw M,Fischer M,Furlan A,Kaste M,Lees KR,Soehngen M, Warach S(2005)The Desmoteplase in Acute Ischemic Stroke Trial (DIAS):a phase II MRI-based9-hour window acute stroke throm-bolysis trial with intravenous desmoteplase.Stroke36:66–73 44.Harris AK,Ergul A,Kozak A,Machado LS,Johnson MH,Fagan

SC(2005)Effect of neutrophil depletion on gelatinase expres-sion,edema formation and hemorrhagic transformation after focal ischemic stroke.BMC Neurosci6:49

45.Hayward NJ,Elliott PJ,Sawyer SD,Bronson RT,Bartus RT

(1996)Lack of evidence for neutrophil participation during infarct formation following focal cerebral ischemia in the rat.Exp Neurol139:188–202

46.Heinzer S,Krucker T,Stampanoni M,Abela R,Meyer EP,

Schuler A,Schneider P,Muller R(2006)Hierarchical microi-maging for multiscale analysis of large vascular networks.

Neuroimage32:626–636

47.Investigators EAST(2001)Use of anti ICAM-1therapy in

ischemic stroke:Results of the Enlimomab acute stroke trial.

Neurol57:1428–1434

48.Ishikawa M,Vowinkel T,Stokes KY,Arumugam TV,Yilmaz G,

Nanda A,Granger DN(2005)CD40/CD40ligand signaling in mouse cerebral microvasculature after focal ischemia/reperfu-sion.Circulation111:1690–1696

49.Jin R,Yang G,Li G(2010)In?ammatory mechanisms in

ischemic stroke:role of in?ammatory cells.J Leukoc Biol 87:779–789

50.Katchanov J,Waeber C,Gertz K,Gietz A,Winter B,Bruck W,

Dirnagl U,Veh RW,Endres M(2003)Selective neuronal vul-nerability following mild focal brain ischemia in the mouse.

Brain Pathol13:452–464

51.Kato H,Kogure K,Liu XH,Araki T,Itoyama Y(1996)Pro-

gressive expression of immunomolecules on activated microglia and invading leukocytes following focal cerebral ischemia in the rat.Brain Res734:203–212

52.Kitagawa K,Matsumoto M,Mabuchi T,Yagita Y,Ohtsuki T,

Hori M,Yanagihara T(1998)De?ciency of intercellular adhesion molecule1attenuates microcirculatory disturbance and infarction size in focal cerebral ischemia.J Cereb Blood Flow Metab 18:1336–1345

53.Klinke A,Nussbaum C,Kubala L,Friedrichs K,Rudolph TK,

Rudolph V,Paust HJ,Schroder C,Benten D,Lau D,Szocs K, Furtmuller PG,Heeringa P,Sydow K,Duchstein HJ,Ehmke H, Schumacher U,Meinertz T,Sperandio M,Baldus S(2011)

Myeloperoxidase attracts neutrophils by physical forces.Blood 117:1350–1358

54.Krams M,Lees KR,Hacke W,Grieve AP,Orgogozo JM,Ford

GA(2003)Acute Stroke Therapy by Inhibition of Neutrophils (ASTIN):an adaptive dose-response study of UK-279,276in acute ischemic stroke.Stroke34:2543–2548

55.Ley K,Laudanna C,Cybulsky MI,Nourshargh S(2007)Getting

to the site of in?ammation:the leukocyte adhesion cascade updated.Nat Rev Immunol7:678–689

56.Lindsberg PJ,Carpen O,Paetau A,Karjalainen-Lindsberg ML,

Kaste M(1996)Endothelial ICAM-1expression associated with in?ammatory cell response in human ischemic stroke.Circulation 94:939–945

57.Lipowsky HH(2012)The endothelial glycocalyx as a barrier to

leukocyte adhesion and its mediation by extracellular proteases.

Ann Biomed Eng40:840–848

58.Lipowsky HH,Gao L,Lescanic A(2011)Shedding of the

endothelial glycocalyx in arterioles,capillaries,and venules and its effect on capillary hemodynamics during in?ammation.Am J Physiol Heart Circ Physiol301:H2235–H2245

59.Maeda K,Hata R,Hossmann KA(1998)Differences in the

cerebrovascular anatomy of C57black/6and SV129mice.Neu-roReport9:1317–1319

60.Maier CM,Ahern K,Cheng ML,Lee JE,Yenari MA,Steinberg GK

(1998)Optimal depth and duration of mild hypothermia in a focal model of transient cerebral ischemia:effects on neurologic outcome, infarct size,apoptosis,and in?ammation.Stroke29:2171–2180 61.Matsuo Y,Onodera H,Shiga Y,Shozuhara H,Ninomiya M,

Kihara T,Tamatani T,Miyasaka M,Kogure K(1994)Role of cell adhesion molecules in brain injury after transient middle cerebral artery occlusion in the rat.Brain Res656:344–352 62.McColl BW,Rothwell NJ,Allan SM(2007)Systemic in?am-

matory stimulus potentiates the acute phase and CXC chemokine responses to experimental stroke and exacerbates brain damage via interleukin-1-and neutrophil-dependent mechanisms.J Neu-rosci27:4403–4412

63.Okada Y,Copeland BR,Mori E,Tung MM,Thomas WS,del

Zoppo GJ(1994)P-selectin and intercellular adhesion molecule-1 expression after focal brain ischemia and reperfusion.Stroke 25:202–211

64.Owens T,Bechmann I,Engelhardt B(2008)Perivascular spaces

and the two steps to neuroin?ammation.J Neuropathol Exp Neurol67:1113–1121

65.Petkova SB,Yuan R,Tsaih SW,Schott W,Roopenian DC,Pa-

igen B(2008)Genetic in?uence on immune phenotype revealed strain-speci?c variations in peripheral blood lineages.Physiol Genomics34:304–314

66.Pham TH,Baluk P,Xu Y,Grigorova I,Bankovich AJ,Pappu R,

Coughlin SR,McDonald DM,Schwab SR,Cyster JG(2010) Lymphatic endothelial cell sphingosine kinase activity is required for lymphocyte egress and lymphatic patterning.J Exp Med 207:17–27

67.Prestigiacomo CJ,Kim SC,Connolly ES Jr,Liao H,Yan SF,

Pinsky DJ(1999)CD18-mediated neutrophil recruitment con-tributes to the pathogenesis of reperfused but not nonreperfused stroke.Stroke30:1110–1117

68.Price CJ,Menon DK,Peters AM,Ballinger JR,Barber RW,

Balan KK,Lynch A,Xuereb JH,Fryer T,Guadagno JV,War-burton EA(2004)Cerebral neutrophil recruitment,histology,and outcome in acute ischemic stroke:an imaging-based study.

Stroke35:1659–1664

69.Raine CS,Cannella B,Duijvestijn AM,Cross AH(1990)Homing to

central nervous system vasculature by antigen-speci?c lympho-cytes.II.Lymphocyte/endothelial cell adhesion during the initial stages of autoimmune https://www.wendangku.net/doc/fb11487624.html,b Invest63:476–48970.Rascher G,Wolburg H(1997)The tight junctions of the lepto-

meningeal blood-cerebrospinal?uid barrier during development.

J Hirnforsch38:525–540

71.Reina-De La Torre F,Rodriguez-Baeza A,Sahuquillo-Barris J

(1998)Morphological characteristics and distribution pattern of the arterial vessels in human cerebral cortex:a scanning electron microscope study.Anat Rec251:87–96

72.Reiss Y,Hoch G,Deutsch U,Engelhardt B(1998)T cell inter-

action with ICAM-1-de?cient endothelium in vitro:essetial role for ICAM-1and ICAM-2in transendothelial migration of T cells.

Eur J Immunol28:3086–3099

73.Ringelmann B,Ro¨der C,Hallmann R,Maley M,Davies M,

Grounds M,Sorokin LM(1999)Expression of laminin a1,a2,a4 and a5chains,?bronectin and tenascin-C in skeletal muscle of dystrophic129ReJ dy/dy mice.Exp Cell Res246:165–182 74.Rother J,Schellinger PD,Gass A,Siebler M,Villringer A,Fie-

bach JB,Fiehler J,Jansen O,Kucinski T,Schoder V,Szabo K, Junge-Hulsing GJ,Hennerici M,Zeumer H,Sartor K,Weiller C, Hacke W(2002)Effect of intravenous thrombolysis on MRI parameters and functional outcome in acute stroke\6hours.

Stroke33:2438–2445

75.Schabitz WR,Kollmar R,Schwaninger M,Juettler E,Bardutzky

J,Scholzke MN,Sommer C,Schwab S(2003)Neuroprotective effect of granulocyte colony-stimulating factor after focal cere-bral ischemia.Stroke34:745–751

76.Schabitz WR,Laage R,Vogt G,Koch W,Kollmar R,Schwab S,

Schneider D,Hamann GF,Rosenkranz M,Veltkamp R,Fiebach JB,Hacke W,Grotta JC,Fisher M,Schneider A(2010)AXIS:a trial of intravenous granulocyte colony-stimulating factor in acute ischemic stroke.Stroke41:2545–2551

77.Schuler F,Sorokin LM(1995)Expression of laminin isoforms in

mouse myogenic cells in vitro and in vivo.J Cell Sci108: 3795-3805

78.Soriano SG,Coxon A,Wang YF,Frosch MP,Lipton SA,Hickey

PR,Mayadas TN(1999)Mice de?cient in Mac-1(CD11b/CD18) are less susceptible to cerebral ischemia/reperfusion injury.

Stroke30:134–139

79.Sornas R,Ostlund H,Muller R(1972)Cerebrospinal?uid

cytology after stroke.Arch Neurol26:489–501

80.Sorokin LM,Frieser M,Pausch F,Kro¨ger S,Ohage E,Deutz-

mann R(1997)Developmental regulation of laminin alpha5 suggests a role in epithelial and endothelial cell maturation.Dev Biol189:285–300

81.Spatz H(1939)Pathologische Anatomie der Kreislaufsto¨rungen

des Gehirns.Z Ges Neurol Psychiatr167:301–357

82.Steiner O,Coisne C,Cecchelli R,Boscacci R,Deutsch U,

Engelhardt B,Lyck R(2010)Differential roles for endothelial ICAM-1,ICAM-2,and VCAM-1in shear-resistant T cell arrest, polarization,and directed crawling on blood-brain barrier endo-thelium.J Immunol185:4846–4855

83.Strecker JK,Sevimli S,Schilling M,Klocke R,Nikol S,

Schneider A,Schabitz WR(2010)Effects of G-CSF treatment on neutrophil mobilization and neurological outcome after transient focal ischemia.Exp Neurol222:108–113

84.Toft-Hansen H,Buist R,Sun XJ,Schellenberg A,Peeling J,

Owens T(2006)Metalloproteinases control brain in?ammation induced by pertussis toxin in mice overexpressing the chemokine CCL2in the central nervous system.J Immunol177:7242–7249 85.von Bruhl ML,Stark K,Steinhart A,Chandraratne S,Konrad I,

Lorenz M,Khandoga A,Tirniceriu A,Coletti R,Kollnberger M, Byrne RA,Laitinen I,Walch A,Brill A,Pfeiler S,Manukyan D, Braun S,Lange P,Riegger J,Ware J,Eckart A,Haidari S, Rudelius M,Schulz C,Echtler K,Brinkmann V,Schwaiger M, Preissner KT,Wagner DD,Mackman N,Engelmann B,Massberg S(2012)Monocytes,neutrophils,and platelets cooperate to

initiate and propagate venous thrombosis in mice in vivo.J Exp Med209:819–835

86.von der Mark H,Oberbaumer I,Timpl R,Kemler R,Wick G

(1985)Immunochemical and autoantigenic properties of the globular domain of basement membrane collagen(type IV).Eur J Biochem146:555–562

87.Wang PY,Kao CH,Mui MY,Wang SJ(1993)Leukocyte in?l-

tration in acute hemispheric ischemic stroke.Stroke24:236–24088.Wu C,Ivars F,Anderson P,Hallmann R,Vestweber D,Nilsson P,

Robenek H,Tryggvason K,Song J,Korpos E,Loser K,Beissert S, Georges-Labouesse E,Sorokin LM(2009)Endothelial basement membrane laminin alpha5selectively inhibits T lymphocyte extravasation into the brain.Nat Med15:519–527

89.Zhang ET,Inman CB,Weller RO(1990)Interrelationships of the

pia mater and the perivascular(Virchow-Robin)spaces in the human cerebrum.J Anat170:111–123

相关文档